AU2013204136B2 - Therapeutic Human Anti-IL-1R Monoclonal Antibody - Google Patents

Therapeutic Human Anti-IL-1R Monoclonal Antibody Download PDF

Info

Publication number
AU2013204136B2
AU2013204136B2 AU2013204136A AU2013204136A AU2013204136B2 AU 2013204136 B2 AU2013204136 B2 AU 2013204136B2 AU 2013204136 A AU2013204136 A AU 2013204136A AU 2013204136 A AU2013204136 A AU 2013204136A AU 2013204136 B2 AU2013204136 B2 AU 2013204136B2
Authority
AU
Australia
Prior art keywords
seq
antibody
amino acid
acid sequence
heavy chain
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.)
Expired
Application number
AU2013204136A
Other versions
AU2013204136A1 (en
Inventor
Gary S. Elliott
Haichun Huang
Frank Martin
Xueming Qian
Brian Varnum
Chris Vezina
Alison Witte
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.)
Amgen Inc
ER Squibb and Sons LLC
Original Assignee
ER Squibb and Sons LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2008207483A external-priority patent/AU2008207483B2/en
Priority claimed from AU2012202672A external-priority patent/AU2012202672B2/en
Application filed by ER Squibb and Sons LLC filed Critical ER Squibb and Sons LLC
Priority to AU2013204136A priority Critical patent/AU2013204136B2/en
Publication of AU2013204136A1 publication Critical patent/AU2013204136A1/en
Assigned to AMGEN INC., MEDAREX, L.L.C. reassignment AMGEN INC. Amend patent request/document other than specification (104) Assignors: AMGEN INC., MEDAREX, INC.
Assigned to AMGEN INC., E. R. SQUIBB & SONS, L.L.C. reassignment AMGEN INC. Alteration of Name(s) of Applicant(s) under S113 Assignors: AMGEN INC., MEDAREX, L.L.C.
Application granted granted Critical
Publication of AU2013204136B2 publication Critical patent/AU2013204136B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Landscapes

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

Abstract

Antibodies that interact with interleukin-1 receptor type 1 (IL-iRi) are described. Methods of treating IL-1 mediated diseases by administering a 5 pharmaceutically effective amount of antibodies to IL-IRI are described. Methods of detecting the amount of IL-IRI in a sample using antibodies to IL-IRI are described.

Description

2013204136 12 Apr 2013
THERAPEUTIC HUMAN ANTI-IL-1R1 MONOCLONAL ANTIBODY
This application is related to and claims priority to U.S. provisional application Serial No. 60/408,719 filed September 6,2002, the disclosure of which is incorporated by 5 reference herein.
FIELD OF THE INVENTION
The invention relates to antibodies that bind interleukin-1 receptor type 1 (IL-1R1) protein. Compositions, particularly pharmaceutical compositions and methods for 10 treating of IL-1 mediated diseases, such as rheumatoid arthritis, osteoarthritis, and other inflammatory conditions, are also provided.
BACKGROUND OF THE INVENTION
Antibody Development 15 Inflammation is the body’s response to injuries resulting from mechanical damage, infection, or antigenic stimulation. Inflammatory reactions often are expressed pathologically. Such conditions arise when the inflammation is expressed in an exaggerated manner, is inappropriately stimulated, or persists after the injurious agent is removed. 20 The inflammatory response is mediated, inter alia, by cytokines. One of the most potent inflammatory cytokines yet discovered is interleukin-1 (IL-1). An increase in IL-1 signaling causes persistent inflammation associated with several diseases, and IL-1 is thought to be a key mediator in many diseases and medical conditions. This cytokine is manufactured primarily (though not exclusively) by cells of the macrophage/monocyte 25 lineage and may be produced in two forms: IL-1 alpha (IL-la) and IL-1 beta (IL-Ιβ), IL-1 stimulates cellular responses by interacting with a heterodimeric receptor complex comprised of two transmembrane proteins, IL-1 receptor type I (EL-1R1) and IL-1 receptor accessory protein (IL-lRAcP). IL-1 first binds to IL-1R1; IL-lRAcP is then recruited to this complex (Greenfeder et ah, 1995, J. Biol. Chem. 270:13757-13765; Yoon 1 2013204136 12 Apr 2013 and Dinarello, 1998, J. Immunology 160:3170-3179; Cullinan et al., 1998, J. Immunology 161:5614-5620), followed by signal transduction resulting in the induction of a cellular response.
Cell-based binding studies suggest that IL-lRAcP stabilizes the IL-1R signaling 5 complex by slowing the ligand off-rate (Wesche et al., 1998, FEBSLetters 429:303-3061. While the interaction of the IL-1 with IL-1R lias been thoroughly characterized, the interaction of IL-lRAcP with ligand-bound receptor remains poorly defined. Since IL-lRAcP has no significant affinity for either IL-1 or IL-1R1 alone, but high affinity for the complex, it follows that novel binding sites for IL-lRAcP are created by the IL-1/IL-1R 10 binding event, which may even include contributions fiom IL-1 residues (Ettorre et al., 1997, Enr, Cytokine Netw. 8:161-171). Another molecule, IL-1 receptor antagonist (IL-lra) competes with IL-Ια and IL-Ιβ for receptor binding but foils to recruit IL-lRAcP, resulting in an occupied but non-signaling receptor. IL-1 activity can additionally be counterbalanced by IL-1R type II, a decoy receptor that binds ligand but does not 15 participate in signaling due to a truncated intracellular domain. IL-lra and IL-1R type II act to reduce the severity and duration of IL-1 mediated inflammatory events (Wesche et al, 1998, FEBS Letters 429:303-306; Dripps et al, 1991, J. Biol. Chem. 266:10331-10336; Dripps et al, 1991,/. Biol. Chem. 266:20331-203351.
Interleukin-l inhibitors may be produced fiom any protein capable of specifically 20 preventing activation of cellular receptors to IL-1, which may result fiom a number of mechanisms. Such mechanisms include down-regulating IL-1 production, binding fiee IL-1, interfering with IL-1 binding to IL-1R, interfering with formation of the IL-1R-IL-lRAcP complex, or interfering with modulation of IL-1 signaling after binding to its receptor. Classes of IL-1 inhibitors include: 25 · interleukin-1 receptor antagonists such as IL-lra, as described below; • anti-IL-lR monoclonal antibodies (e.g., as disclosed in published European Patent Application No. EP 623674, the disclosure of which is hereby incorporated by reference); • IL-1 binding proteins such as soluble IL-1 receptors (e.g., as disclosed in U. S. 30 Pat. Nos. 5,492,888; 5,488,032; 5,464,937; 5,319,071; and 5,180,812; the disclosures of which are hereby incorporated by reference); 2 2013204136 12 Apr 2013 • anti-IL-l monoclonal antibodies (e.g., as disclosed in International Patent Application Publication Nos. WO 9501997, WO 9402627, WO 9006371, U.S. Pat. No. 4,935,343, BP 364778, EP 267611 and BP 220063, the disclosures of which are hereby incorporated by reference); 5 · . IL-1 receptor accessory proteins and antibodies thereto (e.g., as disclosed in
International Patent Application Publication Nos. WO 96/23067 and WO 99/37773, the disclosure of which is hereby incorporated by reference); and
• inhibitors of interleukin-1/3 converting enzyme (ICE) or caspase I (e.g., as disclosed in International Patent Application Publication Nos. WO 99/46248, WO 10 99/47545, and WO 99/47154, the disclosures of which are hereby incorporated by reference), which can be used to inhibit IL-1 β production and secretion; • interleukin-1/3 protease inhibitors; and • other compounds and proteins that block in vivo synthesis or extracellular release ofIL-1. 15 Exemplary IL-1 inhibitors are disclosed in the following references: US Pat. Nos. 5,747,444; 5,359,032; 5,608,035; 5,843,905; 5,359,032; 5,866,576; 5,869,660; 5,869,315; 5,872,095; 5,955,480; and 5,965,564; International Patent Application Publication Nos W098/21957, WO96/09323, W091/17184, W096/40907, W098/32733, W098/42325, WO98/44940, W098/47892, W098/56377, WO99/03837, WO99/06426, W099/06042, 20 W091/17249, W098/32733, W098/17661, WO97/08174, W095/34326, W099/36426, and W099/36415; European patent applications Publication Nos. EP534978 and EP89479; and French patent application no. FR 2762514. The disclosures of all of the aforementioned references are hereby incorporated by reference.
Interleukin-1 receptor antagonist (IL-lra) is a human protein that acts as a natural 25 inhibitor of interleukin-1 and is a member of the IL-1 family, which includes IL-Ια and IL-lp. Preferred receptor antagonists (including IL-lra and variants and derivatives thereof), as well as methods of making and using thereof, are described in U.S. Patent No. 5,075,222; International Patent Application Publication Nos. WO 91/08285; WO 91/17184; W092/16221; W093/21946; WO 94/06457; WO 94/21275; WO 94/21235; 30 DE 4219626, WO 94/20517; WO 96/22793;WO 97/28828; and WO 99/36541, Australian Patent Application No. AU9173636; and French Patent Application No. FR2706772; the 3 2013204136 12 Apr 2013 disclosures of which are incorporated herein by reference. Tire proteins include glycosylated as well as non-glycosylated forms of IL-1 receptor antagonists.
Specifically, three useful forms of IL-lra and variants thereof are disclosed and described in U.S. Patent No. 5,075,222 (“the ’222 patent”). IL-lraa is characterized by 5 SDS-PAGE as a 22-23 kD molecule having an approximate isoelectric point of 4.8, eluting from a Mono Q FPLC column at around 52 mM NaCl in Tris buffer, pH 7.6. IL-lrap is characterized as a 22-23 kD protein, eluting from a Mono Q column at 48 mM NaCl. Both IL-lraa and IL-lrap are glycosylated, IL-lrax is characterized as a 20 kD protein, eluting fiOnr a Mono Q column at 48 mM NaCl, and is non-glycosylated. The 10 ‘222 patent also discloses methods for isolating the genes responsible for coding the inhibitor's, cloning the gene in suitable vectors and cell types, and expressing the gene to produce the inhibitors, While effective, IL-lra has a relatively short half-life. In current use, IL-lra is administered once a day, The art would thus benefit from an antagonist of the IL-1 receptor with an appreciably longer half-life. 15 Preventing IL-1 signaling by inhibiting IL-1 from binding the IL-1 receptor is an attractive therapeutic approach for treating IL-1 mediated diseases. There is a need in the art for clinically effective inhibitors of the IL-1 signaling pathway that may ameliorate the effects of IL-1 mediated diseases and are suitable for delivery into human patients. A human antibody that blocks IL-1 signaling would be particularly advantageous in 20 fillfilling this need and would provide a longer half-life than currently available therapy.
SUMMARY OP THE INVENTION
The invention provides monoclonal antibodies that bind to interleukin-1 receptor type I (1L-1R1). Preferably, the antibodies inhibit IL-1 signaling by competing with IL-25 IjS and IL-labinding to IL-1R1. Also provided by this invention are hybridoma cell lines that produce, and most preferably, secrete into cell culture media the monoclonal antibodies of the invention. The antibodies of die invention successfully block IL-1 signaling in human cells and are useful diereby in heating patients with IL-1 mediated diseases. The invention further provides fusion proteins comprising the sequence of an 30 antibody Fc region and one or more sequences selected from the group consisting of SEQ ID NO: 10, SEQ ID NO; 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID 4 2013204136 12 Apr 2013 NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SBQ 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. Such, molecules can be prepared using methods as described, for example, in WO 00/24782, which is incorporated by reference. Such molecules can be expressed, for 5 example, in mammalian cells (e.g Chinese Hamster Ovary cells) or bacterial cells (e.g. E. coli cells).
In certain aspects, the invention provides antibodies, preferably monoclonal antibodies, most preferably human antibodies, comprising a heavy chain and a light chain, wherein the heavy chain comprises an amino acid sequence as set forth in any of 10 SBQ ID NO: 2, SEQ ID NO: 6, or SEQ ID NO: 8, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof.
The invention also provides antibodies, preferably monoclonal antibodies, most preferably human antibodies, comprising a heavy chain and a light chain, wherein the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 4 or an antigen-15 binding or an immunologically functional immunoglobulin fragment thereof.
In certain aspects, antibodies of the invention comprise a heavy chain and a light chain, wherein the variable region of the heavy chain comprises an amino acid sequence as set forth in any of SEQ ID NO: 10, SEQ ID NO: 14, or SEQ ID NO: 16 or an antigen-binding or an immunologically functional immunoglobulin fragment thereof, hi other 20 aspects, the light chain variable region comprises an amino acid sequence as set forth in any of SEQ ID NO: 12 or SEQ ID NO: 18, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof. In additional aspects, the heavy chain comprises an amino acid sequence as set forth in any of 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, 25 SEQ ID NO: 34, or SEQ ID NO: 36, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof. In still further aspects, the light chain comprises an amino acid sequence as set forth in any of SEQ ID NO: 38 or SEQ ID NO: 40, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof. Such antibody chains are useful in preparing antibodies that bind specifically to 30 IL-1R1 and also in preparation of bispecific antibodies in which the resulting molecule binds to EL-1R1 and/or to another target molecule (e.g., TNF or a TNE receptor). 5 2013204136 12 Apr 2013
Tile invention also provides antibodies that bind specifically to IL-1R1, wherein the heavy chain comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 10, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof, and the light chain comprises a light chain 5 variable region comprising an amino acid sequence as set forth in SEQ ID NO: 12, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof.
In certain aspects, the invention also provides antibodies, comprising a heavy chain and a light chain, wherein the heavy chain comprises a first variable region, and wherein the first variable region comprises a sequence that has at least 90%, more 10 preferably at least 95%, and most preferably about 99% identity to the amino acid sequence as set forth in SEQ ID NO: 10, and wherein the light chain comprises a second variable region, and wherein the second variable region comprises a sequence that has at least 90%, more preferably at least 95%, and most preferably about 99%, identity to the amino acid sequence as set forth in SEQ ID NO: 12, wherein the antibody interacts with 15 II^lRl.
The invention further provides antibodies that specifically bind to EL-1R1, wherein the heavy chain comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 14, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof and the light chain 20 comprises a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 12, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof
In certain aspects, the invention provides antibodies, comprising a heavy chain and a light chain, wherein the heavy chain comprises a first variable region, and wherein 25 the first variable region comprises a sequence that has at least 90%, more preferably at least 95%, and most preferably about 99%, identity to the amino acid sequence as set forth in SEQ ID NO: 14, and wherein the light chain comprises a second variable region, and wherein the second variable region comprises a sequence that has at least 90%, more preferably at least 95%, and most preferably about 99%, identity to the amino acid 30 sequence as set forth in SEQ ID NO: 12, wherein the antibody interacts with IL- 1R1.
The invention also provides antibodies that bind specifically to IL-1R1, wherein the heavy chain comprises a heavy chain variable region comprising an amino acid 6 2013204136 12 Apr 2013 sequence as set forth in SEQ ID NO: 16, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof, and the light chain comprises a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 18, or an antigen-binding or an irmnunologically functional immunoglobulin fragment thereof. 5 In certain aspects, the invention provides antibodies, comprising a heavy chain and a light chain, wherein the heavy chain comprises a first variable region, and wherein the first variable region comprises a sequence that has at least 90%, more preferably at least 95%, and most preferably about 99%, identity to the amino acid sequence as set forth in. SEQ ID NO: 16, and wherein tire light chain comprises a second variable region, 10 and wherein the second variable region comprises an amino acid sequence that has at least 90%, more preferably at least 95%, and most preferably about 99%, identity to the amino acid sequence as set forth in SEQ ID NO: 18, wherein the antibody interacts with IL-1R1.
The invention also provides antibodies that bind specifically to IL-1R1, wherein 15 the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 20, SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 28, or SEQ ID NO: 30, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof. 20 The invention further provides antibodies that bind specifically to IL-1R1, wherein the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 32, SEQ ID NO: 34, or SEQ ID NO: 36, or an antigen-binding or an immunologically functional immunoglobulin fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 40, or an antigen-binding or an immunologically 25 functional immunoglobulin fragment thereof.
The invention also provides embodiments of all of the foregoing that are single chain antibodies, single chain Fv antibodies, Fab antibodies, Fab’ antibodies and (Fab’)2 antibodies.
In particular aspects, the invention provides a light chain comprising an amino 30 acid sequence as set forth in any of SEQ ID NO: 38 or SEQ ID NO: 40, or an antigenbinding or an immunologically functional immunoglobulin fragment thereof. 7 2013204136 12 Apr 2013
In addition, the invention provides a heavy chain comprising an amino acid sequence as set forth in any of 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 or SEQ ID NO: 36, or an antigen-binding or an immunologically functional immunoglobulin 5 fragment thereof.
The invention also relates to isolated human antibodies that specifically bind IL-1R1, wherein the antibody comprises: (a) human heavy chain framework regions, a human heavy chain CDR1 region, a human heavy chain CDR2 region, and a human heavy chain CDR3 region; and (b) human light chain framework regions, a human light 10 chain CDR1 region, a human light chain CDR2 region, and a hiunan light chain CDR3 region, hi certain aspects, the human heavy chain CDR1 region can be the heavy chain CDR1 region of 26F5, 27F2, or 15C4 as shown in Figure 10 and the hiunan light chain CDR1 region can be the light chain CDR1 region of 26F5, 27F2, or 15C4 as shown in Figure 11. In other aspects, the human heavy chain CDR2 region can be the heavy chain 15 CDR2 region of 26F5, 27F2, or 15C4 as shown in Figure 10 and the human light chain CDR2 region can be the light chain CDR2 region of 26F5, 27F2, or 15C4 as shown in Figure 11. In still other aspects, the human heavy chain CDR3 region is the heavy chain CDR3 region of 26F5, 27F2, or 15C4 as shown in Figure 10, and the human light chain CDR3 region is the light chain CDR3 region of 26F5,27F2, or 15C4 as shown in Figure 20 11.
In addition, the invention provides an isolated human antibody that specifically binds to interleukin-1 receptor type 1 (IL-1R1), comprising: a human heavy chain CDR1 region, wherein the heavy chain CDR1 has the amino acid sequence of SEQ ID NO: 61, SEQ ID NO: 62, or SEQ ID NO: 63; a human heavy chain CDR2 region, wherein the 25 heavy chain CDR2 has the amino acid sequence of SEQ ID NO: 64, SEQ ID NO: 65, or SEQ ID NO: 66; and/or a human heavy chain CDR3 region, wherein the heavy chain CDR3 has the amino acid sequence of SEQ ID NO: 67, SEQ ID NO: 68, or SEQ ID NO: 69.
The invention also provides an isolated human antibody that specifically binds to 30 interleukin-1 receptor type 1 (IL-1R1), comprising: a human light chain CDR1 region, wherein the light chain CDR1 has the amino acid sequence of SEQ ID NO: 70 or SEQ ID NO: 71; a human heavy chain CDR2 region, wherein the heavy chain CDR2 has the 8 2013204136 12 Apr 2013 amino acid sequence of SEQ ID NO: 72 or SEQ ID NO: 73; and/or a human heavy chain CDR3 region, wherein the heavy chain CDR3 has the amino acid sequence of SEQ ID NO: 74 or SEQ ID NO: 75.
In certain embodiments, the antibodies of the invention bind to the third domain of 5 IL-1R1, which is shown in Figure 17. Preferably, the epitope for an antibody of the invention consists of the amino acid sequence YSV, which is referred to as Epitope 4 herein and shown in Figure 24. The invention further relates to fusion proteins and other molecules capable of binding to Epitope 4 (together with the aforementioned antibodies, collectively referred to herein as “specific binding partners”), such as may be prepared 10 using methods as described, for example, in WO 00/24782, which is incorporated by reference. Such molecules can be expressed, for example, in mammalian cells (e.g. Chinese Hamster Ovary cells) or bacterial cells (e.g. E. coli cells).
Furthermore, the invention provides a method for epitope mapping of a selected antigen. In one aspect, the method comprises the steps of: (a) generating a set of fusion 15 proteins, wherein each fusion protein comprises (i) avidin and (ii) a fragment of the antigen; (b) screening the set of fusion proteins for binding to one or more specific binding partners for the antigen; (c) isolating the fusion proteins on a medium comprising biotin, whereby the avidin binds to the biotin; and (d) analyzing the fusion proteins bound by the specific binding partner or partners to determine binding sites on the antigen for 20 the specific binding partner or partners. In a particular aspect, the specific binding partners are antibodies.
In additional embodiments, the invention provides methods for treating an IL-1 mediated disease, condition or disorder, comprising the step of administering a pharmaceutically effective amount of one or a plurality of monoclonal antibodies of the 25 invention or an antigen-binding or an immunologic ally functional immunoglobulin fragment thereof to an individual in need thereof,
The invention also provides methods for detecting the level of IL-1R1 in a biological sample, comprising the step of contacting the sample with a monoclonal antibody of the invention or antigen-binding fragment thereof. The anti-IL-lR antibodies 30 of the invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, immunoprecipitation assays and enzyme-linked immunosorbent assays (ELISA) {See, Sola, 1987, Monoclonal Antibodies: 9 2013204136 12 Apr 2013 A Manual of Techniques, pp. 147-158, CRC Press, Inc.) for the detection and quantitation of IL-1R. The antibodies can bind IL-1R with an affinity that is appropriate for the assay method being employed.
Specific preferred embodiments of the invention will become evident from the 5 following more detailed description of certain preferred embodiments and the claims.
BRIEF DESCRIPTION OF THE FIGURES
Figures 1A-1B depict a cDNA sequence (Fig. 1A) encoding a human anti-IL-lRl antibody heavy chain IgGl constant region (SEQ ID NO: 1) and the amino acid sequence 10 (Fig. IB) of a human anti-IL-lRl antibody heavy chain IgGl constant region (SEQ ID NO: 2).
Figures 2A-2B depict a cDNA sequence (Fig. 2A) encoding a human anti-IL-lRl antibody kappa chain constant region (SEQ ID NO: 3) and the amino acid sequence (Fig. 2B) of a human anti-IL-lRl antibody kappa chain constant region (SEQ ID NO: 4). 15 Figures 3A-3B depict a cDNA sequence (Fig. 3A) encoding a human anti-IL-lRl antibody heavy chain IgG2 constant region (SEQ ID NO: 5) and the amino acid sequence (Fig. 3B) of a human anti-IL-lRl antibody heavy chain IgG2 constant region (SEQ ID NO: 6).
Figures 4A-4B depict a cDNA sequence (Fig. 4A) encoding a human anti-IL-lRl 20 antibody heavy chain IgG4 constant region (SEQ ID NO: 7) and the amino acid sequence (Fig. 4B) of a human anti-IL-lRl antibody heavy chain IgG4 constant region (SEQ ID .NO: 8).
Figures 5A-5B depict a cDNA sequence (Fig. 5A) encoding the 26F5 anti-IL-lRl antibody heavy chain variable region (SEQ ID NO: 9) and the amino acid sequence (Fig. 25 5B) of the 26F5 anti-IL-lRl antibody heavy chain variable region (SEQ ID NO: 10).
Figures 6A-6B depict a cDNA sequence (Fig, 6A) encoding the 26F5 anti-IL-lRl antibody kappa chain variable region (SEQ ID NO: 11) and the amino acid sequence (Fig. 6B) of the 26F5 anti-IL-lRl antibody kappa chain variable region (SEQ ID NO: 12). 10 2013204136 12 Apr 2013
Figures 7A-7B depict a cDNA sequence (Fig. 7A) encoding the 27F2 anti-IL-lRl antibody heavy chain variable region (SEQ ID NO: 13) and the amino acid sequence (Fig. 7B) of the 27F2 anti-IL-lRl antibody heavy chain variable region (SEQ ID NO: 14). 5 Figures 8A-8B depict a cDNA sequence (Fig, 8A) encoding the 15C4 anti-IL-lRl antibody heavy chain variable region (SEQ ID NO: 15) and the amino acid sequence (Fig. 8B) of the 15C4 anti-IL-lRl antibody heavy chain variable region (SEQ ID NO: 16).
Figures 9A-9B depict a cDNA sequence (Fig. 9A) encoding the 15C4 anti-IL-lRl 10 antibody kappa chain variable region (SEQ ID NO: 17) and the amino acid sequence (Fig. 9B) of the 15C4 anti-IL-lRl antibody kappa chain variable region (SEQ ID NO: 18).
Figure 10 shows an amino acid sequence alignment of heavy chains from anti-IL-1R1 antibodies designated 15C4, 27F2, and 26F5. The complementarity determining 15 regions (CDRs) are underlined. CDR1 for 26F5 is designated SEQ ID NO: 61; for 27F2 is designated SEQ ID NO: 62; for 15C4 is designated SEQ ID NO: 63. CDR2 for 26F5 is designated SEQ ID NO: 64; for 27F2 is designated SEQ ID NO: 65; for 15C4 is designated SEQ ID NO: 66. CDR1 for 26F5 is designated SEQ ID NO: 67; for 27F2 is designated SEQ ID NO: 68; for 15C4 is designated SEQ ID NO: 69. 20 Figure 11 shows an amino acid sequence alignment of light chains from anti-IL-
Rl-γ antibodies designated 15C4, 27F2, and 26F5. CDR1 for 26F5/27F2 is designated SEQ ID NO: 70; for 15C4 is designated SEQ ID NO: 71. CDR2 for 26F5/27F2 is designated SEQ ID NO: 72; for 15C4 is designated SEQ ID NO: 73, CDR1 for 26F5/27F2 is designated SEQ ID NO: 74; for 15C4 is designated SEQ ID NO: 75. 25 -- Figure 12 is a graph illustrating the inhibitory effect of anti-IL-lRl antibodies on IL- 1R/IL-1 /3/IL-1 RAcP complex formation.
Figure 13 is a graph showing the inhibitory effect of an anti-IL-lRl monoclonal antibody as described herein and designated 15C4 on IL-1R/IL-1 cx/IL-IRacP complex formation. 11 2013204136 12 Apr 2013
Fignre 14 is a graph representing the ability of anti-IL-llll antibodies to block IL-Ιβ binding while not significantly interfering with binding of IL-Ira compared with IgG control.
Figure 15A is a graph showing inhibition of IL-6 production in primary human 5 chondrocytes by anti-IL-lRl antibodies identified herein and designated 15C4,26F5, and 27F2 compared with IL-lra.
Figure 15B is a graph showing inhibition of IL-6 production in primary human chondrocytes by IL-lra and monoclonal antibodies 15C4 and 27F2 compared with the class of monoclonal antibodies represented by 10H7 and 24E12. 10 Figure 16 is a graph showing inhibition of IL-6 production in human whole blood by anti-IL-lRl monoclonal antibodies designated 15C4, 26F5, and 27F2 compared with IL-lra.
Fignre 17 depicts human amino acid (SEQ ID NO: 76) and nucleotide (SEQ ID NO: 77) and rat nucleotide (SEQ ID NO: 78) and amino acid (SEQ ID NO: 79) 3rd 15 domain IL-1R1 sequences. The numbered bars above the human sequence indicate the 15 different sites mutated to construct the 15 different mutated proteins. The rat residues introduced by mutation are listed below the rat nucleic acid sequence.
Fignre 18 shows Western blot analysis demonstrating anti-IL-lRl monoclonal antibody recognition of IL-1R1 mutants. 20 Figure 19 is a drawing representing (I) activation of the IL-1 signaling pathway, which starts with binding of IL-lj8 to IL-1R1, and recruitment of IL-lRacP, and three classes of anti-IL-lRl antibodies: (Π) 3ld domain epitope IL-1 blockers, (ΕΓ) 3rd domain epitope RAcP blockers, and (IV) lst/2nii domain epitope IL-1 blockers.
Figure 20 depicts the crystal structure of 15C4 and 27F2 with mutation 10 as 25 described herein. The gray residues indicate the 15C4 and 27F2 epitopes.
Fignre 21 depicts the 15C4 epitopes in the third domain of extracellular IL-1R1.
Figure 22 depicts 24E12 epitopes in the third domain of extracellular IL-1R1.
Fignre 23 depicts the amino acid sequence (SEQ ID NO: 59) of the avidin-human IL-1R1-FLAG chimeric protein of the invention. 12 2013204136 12 Apr 2013
Figure 24 depicts the amino acid sequence (SEQ ID NO: 60) of an avidin-cynomolgus IL-1R1-FLAG chimeric protein. The recombinant chicken avidin (italicized) is joined to the mature extracellular domain of cynomolgus IL-1R1 (underlined, with C-terminal FLAG tag in bold) by a 6 amino acid linker. Four amino 5 acids from human IL-1R1 that were introduced alone and in combination into the cynomolgus sequence are in bold under the cynomolgus sequence. Epitope 4 is bold, italicized, and underlined.
Figure 25A shows a Western blot analysis of anti-human IL1-R1 antibody (anti-huILl-Rl) binding to I1-1R1, The * indicates that antibodies were used at 5 jWg/mL, 10 whereas in the remainder antibodies were used at 1 jc/g/mL.
Figure 25B shows a summary of the densitometric analysis of a duplicate set of Western blot experiments.
Figure 26 shows graphs representing die binding of anti-huILlRl antibodies to avidin IL-1R1-FLAG proteins in a multiplexed bead-based binding assay. 15
DETAILED DESCRIPTION OF CERTAIN PREFERRED EMBODIMENTS
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All references cited in this application are expressly incorporated by reference herein for any purpose. 20 Definitions A disease or medical condition is considered to be an "interleukin-1 (IL-1) mediated disease" if the naturally-occurring or experimentally-induced disease or medical condition is associated with elevated levels of IL-1 in bodily fluids'or tissue or if cells or tissues taken from the body produce elevated levels of IL-1 in culture. Elevated levels of 25 IL-1 can include, for example, levels that exceed those normally found in a particular· cell or tissue, or can be any detectable level of IL-1 in a cell or tissue that normally does not express IL-1. In many cases, IL-1 mediated diseases are also recognized by the following additional two conditions: (1) pathological findings associated with the disease or medical condition can be mimicked experimentally in animals by administration of IL-1 or up-30 regulation of expression of IL-1; and (2) a pathology induced in experimental animal 13 2013204136 12 Apr 2013 models of the disease or medical condition can be inhibited or abolished by treatment with agents that inhibit the action of IL-1, In most IL-1 mediated diseases at least two of the three conditions are met, and in many IL-1 mediated diseases all three conditions are met. 5 A non-exclusive list of acute and chronic IL-l-mediated diseases includes but is not limited to the following: acute pancreatitis; amyelolateroschlerosis (ALS); Alzheimer's disease; cachexia/anorexia, including AIDS-induced cachexia; asthma arid other pulmonary diseases; atherosclerosis; autoimmune vasculitis; chronic fatigue syndrome; Clostridium associated illnesses, including Clostridium-associated diarrhea; 10 coronary conditions and indications, including congestive heart failure, coronary restenosis, myocardial infarction, myocardial dysfunction (e.g., related to sepsis), and coronary artery bypass graft; cancer, such as multiple myeloma and myelogenous (eg., AML or CML) and other leukemias, as well as tumor metastasis; diabetes (eg., insulin-dependent diabetes); endometriosis; fever, fibromyalgia; glomerulonephritis; graft versus 15 host disease/transplant rejection; hemorrhagic shock; hyperalgesia; inflammatory bowel disease; inflammatory conditions of a joint, including osteoarthritis, psoriatic arthritis and rheumatoid arthritis; inflammatory eye disease, as may be associated with, e.g., comeal transplant; ischemia, including cerebral ischemia (e.g., brain injury as a result of trauma, epilepsy, hemorrhage or stroke, each of which may lead to neurodegeneration); 20 Kawasaki’s disease; learning impairment; lung diseases (e.g., ARDS); multiple sclerosis; myopathies (e.g., muscle protein metabolism, especially in sepsis); neurotoxicity (e.g., as induced by HIV); osteoporosis; pain, including cancer-related pain; Parkinson’s disease; periodontal disease; pre-term labor; psoriasis; reperfhsion injury; septic shock; side effects from radiation therapy; temporal mandibular joint disease; sleep disturbance; 25 uveitis; or an inflammatory condition resulting from strain, sprain, cartilage damage, trauma, orthopedic surgery, infection or other disease processes. Methods of the invention for treating these acute and chronic IL-l-mediated diseases, as well as other IL-l-mediated conditions and diseases, are described below.
Conventional techniques may be used for preparing recombinant DNA, 30 performing oligonucleotide synthesis, and practicing tissue culture and transformation (e.g., electroporation, transfection or lipofection). Enzymatic reactions and purification techniques may be performed according to manufacturer's specifications or as commonly 14 2013204136 12 Apr 2013 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., 2001, Molecular 5 Cloning: A Laboratory Manual, 3rd cd., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., which is incorporated herein by reference for any purpose. Unless specific definitions are provided, the nomenclature utilized 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 10 known and commonly used in the art. Standard techniques may be used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients.
As utilized in accordance with the present disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings: 15 The term "isolated polynucleotide" means that the subject polynucleotide, (1) is not associated (covalently or noncovalently) with all or a portion of other, polynucleotides with which the subject polynucleotide is associated in nature, (2) is associated with a molecule with which it is not associated in nature, or (3) does not occur in nature associated with any other polynucleotides. Such an isolated polynucleotide may be 20 genomic DNA, cDNA, mRNA or other PNA, of synthetic origin, or any combination thereof.
The term “isolated protein” referred to herein means that a subject protein (1) is free of at least some other proteins with which it would normally be found, (2) is essentially free of other proteins fiom the same source, e.g., from the same species, (3) is 25 expressed by a cell fiom a different species, (4) has been separated fiom at least about 50 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is associated in nature, (5) is not associated (by covalent or noncovalent interaction) with portions of a protein with which the “isolated protein” is associated in nature, (6) is operably associated (by covalent or noncovalent interaction) with a polypeptide with 30 which it is not associated in nature, or (7) does not occur in nature. Genomic DNA, cDNA, mRNA or other RNA, of synthetic origin, or any combination thereof may encode such an isolated protein. Preferably, the isolated protein is substantially fiee fiom 15 2013204136 12 Apr 2013 proteins or polypeptides or other contaminants that are found in its natural environment that would interfere with its therapeutic, diagnostic, prophylactic, research or other use.
The terms "polypeptide" or “protein” means one or more chains of amino acids, wherein each chain comprises amino acids covalently linked by peptide bonds, and 5 wherein said polypeptide or protein can comprise a plurality of chains non-covalently and/or covalently linked together by peptide bonds, having the sequence of native proteins, that is, proteins produced by naturally-occurring and specifically nonrecombinant cells, or genetically-engineered or recombinant cells, and comprise molecules having the amino acid sequence of the native protein, or molecules having 10 deletions from, additions to, and/or substitutions of one or more amino acids of the native sequence. The terms “polypeptide” and “protein” specifically encompass anti-ILl-Rl antibodies, or sequences that have deletions from, additions to, and/or substitutions of one or more amino acid of an anti-ILR-lRl antibody. Thus, a “polypeptide” or a “protein” can comprising one (termed “a monomer”) or a plurality (termed “a multimer”) of amino 15 acid chains.
The term "polypeptide fragment" refers to a polypeptide, which can be monomeric or multimeric, that has an amino-terminal deletion, a carboxyl-terminal deletion, and/or an internal deletion or substitution of a naturally-occurring or recombinantly-produced polypeptide. In certain embodiments, a polypeptide fragment can comprise an amino acid 20 chain at least 5 to about 500 amino acids long. It will be appreciated that in certain embodiments, fragments are at least 5, 6, 8, 10, 14, 20, 50, 70, 100, 110, 150, 200, 250, 300, 350,400, or 450 amino acids long. Particularly useful polypeptide fragments include functional domains, including binding domains. In the case of an anti-ILl-Rl antibody, useful fragments include, but are not limited to: a CDR region, especially a CDR3 legion 25 of the heavy or light chain; a variable domain of a heavy or light chain; a portion of an antibody chain or just its variable region including two CDRs; and the like.
The term “immunologically functional immunoglobulin fragment” as used herein refers to a polypeptide fragment that contains at least the variable domains of the 30 immunoglobulin heavy and light chains. An immunologically functional immunoglobulin fragment of the invention is capable of binding to a ligand, preventing binding of the ligand to its receptor, interrupting the biological response resulting from 16 2013204136 12 Apr 2013 ligand binding to the receptor, or any combination thereof. Preferably, an immimologically functional immunoglobulin fragment of the invention binds specifically to IL-1R1.
The terms “naturally occurring” and “native” mean that the biological materials 5 (molecules, sequences, protein complexes, cells, and the like) to which the terms are applied can be found in nature and are not manipulated by man. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and that has not been intentionally modified by man is naturally occurring. Likewise, the terms “non-uaturally occurring” or “non-10 native” refer to a material that is not found in nature or that has been structurally modified or synthesized by man.
The term "operably linked" means that the components to which the term is applied are in a relationship that allows them to carry out their inherent functions under suitable conditions. For example, a control sequence "operably linked" to a protein 15 coding sequence is ligated thereto so that expression of the protein coding sequence is achieved under conditions compatible with the transcriptional activity of the control sequences.
The term "control sequence" means that the subject polynucleotide sequence can effect expression and processing of coding sequences to which it is ligated. The nature of 20 such control sequences may depend upon the host organism. In particular embodiments, control sequences for prokaryotes may include a promoter, ribosomal binding site, and transcription termination sequence. In other particular embodiments, control sequences for eukaryotes may include promoters comprising one or a plurality of recognition sites for transcription factors, transcription enhancer sequences, and transcription termination 25 sequence. In certain embodiments, "control sequences" can include leader sequences and/or fusion partner sequences.
The term "polynucleotide" means single-stranded or double-stranded nucleic acid polymers of at least 10 bases in length. In certain embodiments, the nucleotides comprising the polynucleotide can be ribonucleotides or deoxyribonucleotides or a 30 modified form of either type of nucleotide. Said modifications include base modifications such as bromouridine and inosine derivatives, ribose modifications such as 2%3,-dideoxyribose, and intemucleotide linkage modifications such as phosphorothioate, 17 2013204136 12 Apr 2013 pliosphorotlithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate and phosphoroamidate. The term includes single and double stranded forms of DNA.
The term "oligonucleotide" means a polynucleotide comprising a length of 200 5 bases or fewer. In preferred embodiments, oligonucleotides are 10 to 60 bases in length. In more preferred embodiments, oligonucleotides are 12, 13, 14, 15, 16, 17,18, 19, or 20 to 40 bases in length. Oligonucleotides may be single stranded or double stranded, e.g., for use in the construction of a mutant gene. Oligonucleotides of the invention may be sense or antisense oligonucleotides, 10 The term "naturally occurring nucleotides" includes deoxyribonucleotides and ribonucleotides. The term "modified nucleotides" includes nucleotides with modified or substituted sugar groups or modified or substituted bases. The team "oligonucleotide linkages" includes linkages such as phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate, 15 phosphoroamidate, and the like. See, e.g., LaPlanche et al. (1986), Nucl. Acids Res. 14:9081; Stec et al. (1984), J. Am. Chem. Soc. 106:6077: Stein et al. (19881 Nucl. Adds Res. 16:3209; Zon et al (1991), Anti-Cancer Drug Design 6:539; Zon et al. (1991), Oligonucleotides and Analogues: A Practical Approach, pp. 87-108 (F. Eckstein, ed.), Oxford University Press, Oxford England; Stec et al., U.S. Pat. No. 5,151,510; Uhlmann 20 and Peyman (1990), Chemical Reviews 20:543, the disclosures of which are hereby incorporated by reference for any purpose. An oligonucleotide of the invention can include a label, including a radiolabel, a fluorescent label, a hapten or an antigenic label, for detection assays.
The term “vector” means any molecule (e.g., nucleic acid, plasmid, or virus) used 25 to transfer coding information to a host cell.
The term “expression vector” or “expression construct” refers to a vector that is suitable for transformation of a host cell and contains nucleic acid sequences that direct and/or contr ol (in conjunction with the host cell) expression of one or more heterologous coding regions operatively linked thereto. An expression construct may include, but is not 30 limited to, sequences that affect or control transcription, translation, and RNA splicing, if introns are present, of a coding region operably linked thereto. 18 2013204136 12 Apr 2013
The term “host cell” means a cell that has been transformed, or is capable of being transformed, with a nucleic acid sequence and thereby expresses a selected gene of interest. The term includes the progeny of the parent cell, whether or not the progeny is identical in morphology or in genetic make-up to the original parent cell, so long as the 5 selected gene is present.
The term “transduction” means the transfer of genes from one bacterium to another, usually by phage, “Transduction” also refers to the acquisition and transfer of eukaryotic cellular sequences by retroviruses.
The term “transfection” means the uptake of foreign or exogenous DNA by a cell, 10 and a cell has been “transfected” when the exogenous DNA has been introduced inside the cell membrane. A number of transfection techniques am well known in the art and are disclosed herein. See, e.g., Graham et al.s 1973, Virology'$2:456-, Sambrook et al.s 2001, Molecular Cloning: A Laboratory Manual, Id.; Davis et al., 1986, Basic Methods in Molecidar Biology, Elsevier; and Chu et al., 1981, Gene 13:197. Such techniques can 15 be used to introduce one or more exogenous DNA moieties into suitable host cells.
The term “transformation” refers to a change in a cell’s genetic characteristics, and a cell has been transformed when it has been modified to contain new DNA. For example, a cell is transformed where it is genetically modified irom its native state by transfection, transduction, or other- techniques. Following transfection or transduction, 20 the transforming DNA may recombine with that of the cell by physically integrating into a chromosome of the cell, or may be maintained transiently as an episomal element without being replicated, or may replicate independently as a plasmid. A cell is considered to have been “stably transformed” when the transforming DNA is replicated with the division of the cell. 25 The term “antigen” refers to a molecule or a portion of a molecule capable of being bound by a selective binding agent, such as an antibody, and additionally capable of being used in an animal to produce antibodies capable of binding to an epitope of that antigen. An antigen may have one or more epitopes.
The term "epitope" includes any determinant, preferably a polypeptide 30 determinant, capable of specific binding to an immunoglobulin or T-cell receptor. In certain embodiments, epitope determinants include chemically active suifece groupings 19 2013204136 12 Apr 2013 of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three-dimensional 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 5 antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules. In preferred embodiments, an antibody is said to specifically, bind an antigen when the dissociation constant is less than or equal to about 10 nM, more preferably when the dissociation constant is less than or equal to about 100 pM, and most preferably when the dissociation constant is less than or equal to about 10 10 pM.
The term “identity” refers to a relationship between the sequences of two or more polypeptide molecules or two or more nucleic acid molecules, as determined by comparing the sequences thereof, In the art, “identity” also means the degree of sequence relatedness between nucleic acid molecules or polypeptides, as the case may be, as 15 determined by the match between sequences of two or more nucleotides or two or more amino acids. “Identity” measures the percentage of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer-program (/. e., “algorithms”).
The term “similarity” is used in the art with regard to a related concept; in contrast 20 to “identity,” however, “similarity” refers to a measure of relatedness that includes both identical matches and conservative substitution matches. If two polypeptide sequences have, for example, 10/20 identical amino acids, and the remainder are all nonconservative substitutions, then the percentage identity and similarity would both be 50%. If in the same example, there are five more positions where there are conservative 25 substitutions, then the percentage identity remains 50%, but the percentage similarity would be 75% (15/20). Therefore, in cases where there are conservative substitutions, the percentage similarity between two polypeptides will be higher than the percentage identity between those two polypeptides.
Identity and similarity of related nucleic acids and polypeptides can be readily 30 calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, (Lesk, A.M., ed.), 1988, Oxford University Press, New York; Biocomputing: Informatics and Genome Projects, (Smith, 20 2013204136 12 Apr 2013 D.W., ed.), 1993, Academic Press, New York; Computer Analysis of Sequence Data, Part 1, (Griffin, A.M., and Griffin, H.G., eds.), 1994, Humana Press, New Jersey; von Heinje, G., Sequence Analysis in Molecular Biology, 1987, Academic Press; Sequence Analysis Primer, (Gribskov, M. and Deverenx, J., eds.), 1991, M. Stockton Press, New York; 5 Carillo et al, 1988, SIAM J. Applied Math. 48:1073; and Durbin et al, 1998, Biological Sequence Analysis, Cambridge University Press.
Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity are described in publicly available computer programs. Preferred computer program methods to determine identity .10 between two sequences include, but are not limited to, the GCG program package, including GAP (Devereux et al., 1984, Nucl. Acid. Res. 12:387; Genetics Computer Group, University of Wisconsin, Madison, WI), BLASTP, BLASTN, and FASTA (Altschul et al., 1990, J. Mol Biol. 215:403-410). The BLASTX program is publicly available from the National Center for Biotechnology Information (NCB1) and other 15 sources {BLAST Manual, Altschul et al. NCB/NLM/NIH Bethesda, MD 20894; Altschul et al, 1990, supra). The well-known Smith Waterman algorithm may also be used to determine identity.
Certain alignment schemes for aligning two amino acid sequences may result in matching of only a short region of the two sequences, and this small aligned region may 20 have very liigh sequence identity even though there is no significant relationship between the two full-length sequences. Accordingly, in certain embodiments, the selected alignment method (GAP program) will result in an alignment that spans at least 50 contiguous amino acids of the target polypeptide.
For example, using the computer algorithm GAP (Genetics Computer Group, 25 University of Wisconsin, Madison, Wl), two polypeptides for which the percentage sequence identity is to be determined are aligned for optimal matching of their respective amino acids (the “matched span”, as determined by the algorithm). In certain embodiments, a gap opening penalty (which is calculated as three-times the average diagonal; where the “average diagonal” is the average of the diagonal of the comparison 30 matrix being used; the “diagonal” is the score or number assigned to each perfect amino acid match by the particular comparison matrix) and a gap extension penalty (which is usually one-tenth of the gap opening penalty), as well as a comparison matrix such as 21 2013204136 12 Apr 2013 PAM250 or BLOSUM 62 are used in conjunction with the algorithm. In certain embodiments, a standard comparison matrix (see DayhofF et ah, 1978, Atlas of Protein Sequence and Structure 5:345-352 for the PAM 250 comparison matrix; Henikoff et ah, 1992, Proc, Natl Acad. Sci USA 89:10915-10919 for the BLOSOM 62 comparison 5 matrix) is also used by the algorithm.
In certain embodiments, the parameters for a polypeptide sequence comparison include the following:
Algorithm: Needleman et ah (1970), J. Mol. Biol. 48:443-453:
Comparison matrix: BLOSOM 62 from Henikoff et ah (1992), supra; 10 Gap Penalty: 12
Gap Length Penalty: 4 Threshold of Similarity: 0
The GAP program may be useful with the above parameters. In certain embodiments, the aforementioned parameters are the default parameters for polypeptide comparisons (along 15 with no penalty for end gaps) using the GAP algorithm.
The term “naturally occurring,” as used to refer to amino acids, refers to the twenty conventional amino acids. See Immunology—A Synthesis, 2nd Edition, (E. S. Golub and D. R. Gren, eds.), Sinauer Associates: Sunderland, MA (1991), incorporated herein by reference for any purpose. 20 Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of nonpeptide compounds are termed "peptide mimetics" or "peptidomimetics". See Fauchere (1986) , Adv. Drug Res. 15:29: Veber & Freidinger, 1985, TINS p.392; and Evans et ah (1987) , J. Med. Client. 30:1229, which are incorporated herein by reference for any 25 ptupose. Such compormds ar e oftep developed with the aid of computerized molecular modeling, Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce a similar therapeutic or prophylactic effect. Generally, peptidomimetics are structurally similar to a paradigm peptide or polypeptide (/. e., a peptide or polypeptide that has a biochemical property or pharmacological activity), such 30 as human antibody, but have one or more peptide linkages optionally replaced by a 22 2013204136 12 Apr 2013 linkage selected fiom: -CH2-NH-, -CH2-S-, -CH2-CH2-, -CH=CH-(cis and trans), -COCH2-, -CH(OH)CH2-, and --CH2SO-, by methods well known in the art. Systematic substitution of one or more amino acids of a consensus sequence with a D-amino acid of the same type (e,g., D-Iysine in place of L-lysine) may be used in certain embodiments to 5 generate more stable peptides. In addition, constrained peptides comprising a consensus sequence or a substantially identical consensus sequence variation may be generated by methods known in the art (Rizo & Gierasch, 1992, Ann. Rev. Biochem. 61:387. incorporated herein by reference for any purpose); for example, by adding internal cysteine residues capable of forming intramolecular disulfide bridges which cyclize the 10 peptide. "Antibody" or "antibody peptide(s)" refer to an intact antibody, or a binding fragment thereof that competes with the intact antibody for specific binding and includes chimeric, humanized, fully human, and bispecific antibodies. In certain embodiments, binding fragments are produced by recombinant DNA techniques. In additional 15 embodiments, binding fragments are produced by enzymatic or chemical cleavage of intact antibodies, Binding fragments include, but are not limited to, Fab, Fab', F(ab')2, Fv, and single-chain antibodies.
The term ‘heavy chain” includes a fiill-length heavy chain and fragments thereof having sufficient variable region sequence to confer1 specificity for IL-1R1. A full-length 20 heavy chain includes a variable region domain, Vh, and three constant region domains, ChIj Ch2, and Ch3. The Vh domain is at the amino-terminus of the polypeptide, and the Ch3 domain is at the carboxyl-terminus.
The term “light chain” includes a fiill-length light chain and fragments thereof having sufficient variable region sequence to confer specificity for IL-1R1. A full-length 25 light chain includes a variable region domain, Vl, and a constant region domain, CL. Like the heavy chain, the variable region domain of the light chain is at the amino-terminus of the polypeptide. A “Fab fragment” is comprised of one light chain and the CrI and variable regions of one heavy chain. The heavy chain of a Fab molecule cannot form a disulfide 30 bond with another heavy chain molecule. 23 2013204136 12 Apr 2013 A “Fab’ fragment” contains one light chain and one heavy chain that contains more of the constant region, between the ChI and Ch2 domains, such that an interchain disulfide bond can be formed between two heavy chains to form a F(ab’)2 molecule. A “F(ab’)2 fragment” contains two light chains and two heavy chains containing a 5 portion of the constant legion between the CH1 and Ch2 domains, such that an interchain disulfide bond is formed between two heavy chains.
The “Fv region” comprises the variable regions from both the heavy and light chains, but lacks the constant regions. “Single-chain antibodies” are Fv molecules in which the heavy and light chain 10 variable regions have been connected by a flexible linker to form a single polypeptide chain, which forms an antigen-binding region. Single chain antibodies are discussed in detail in International Patent Application Publication No. WO 88/01649 and U.S. Patent Nos. 4,946,778 and 5,260,203, the disclosures of which are incorporated by reference for any purpose. 15 A “bivalent antibody” other than a "multispecific" or "multifunctional" antibody, in certain embodiments, is understood to comprise binding sites having identical antigenic specificity. A “bispecific” or “bifunctional” antibody is a hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies 20 may be produced by a variety of methods including, but not limited to, fusion of hybridomas or linking of Fab* fragments. See, e.g., Songsivilai & Lachmann (1990), Clin. Exp. Immunol. 79:315-321; Kostelny el al. (1992), J. Immunol. 148:1547-1553.
In assessing antibody binding and specificity according to the invention, an antibody “substantially inhibits” adhesion of a ligand to a receptor when an excess of 25 antibody reduces the quantity of receptor bound to counterreceptor by at least about 20%, 40%, 60%, 80%, 85%, or more (as measured in an in vitro competitive binding assay).
The term "agent" means a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials.
The terms "label" or "labeled" refers to incorporation of a detectable marker, e.g., 30 by incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotin 24 2013204136 12 Apr 2013 moieties that can be detected by marked avidin {e.g., streptavidin preferably comprising a detectable marker such as a fluorescent marker, a chemiluminescent marker or an enzymatic activity that can be detected by optical or coloiimetric methods). In certain embodiments, the label can also be therapeutic. Various methods of labeling 5 polypeptides and glycoproteins are known in the art and may be used advantageously in the methods disclosed herein. Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 15N, 35S, 99mTc, luIn, 125I, 13lI), fluorescent labels (e.g., fluorescein isothiocyanate (FITC), rhodamine, or lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, β-10 galactosidase, luciferase, alkaline phosphatase), chemiluminescent labels, hapten labels such as biotinyl groups, and predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags). In certain embodiments, labels are attached by spacer arms (such as (CEfe)^ where u < about 20) of various lengths to reduce 15 potential steric hindrance.
The term “biological sample” 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, monkeys, rats, rabbits, and other animals. Such substances include, but are not limited to, blood, serum, urine, cells, organs, tissues, bone, bone 20 marrow, lymph, lymph nodes, synovial tissue;, chondrocytes, synovial macrophages, endothelial cells, vascular tissue (particularly inflamed vascular tissue), and skin. The terms "pharmaceutical agent” and “drug" refer to a chemical compound or composition capable of inducing a desired therapeutic effect when properly administered to a patient.
The term “patient” includes human and animal subjects. 25 Unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.
Amino Acids
The twenty naturally-occurring amino acids and then· abbreviations follow conventional usage. See Immunology—A Synthesis, 2nd Edition, (E. S. Golub and D. R. 30 Gren, eds.), Sinauer Associates: Sunderland, MA (1991), incorporated herein by reference for any purpose. Stereoisomers {e.g., D-amino acids) of the twenty 25 2013204136 12 Apr 2013 conventional amino acids, unnatural amino acids such as α-, α-disubstituted amino acids, N-alkyl amino acids, and other imconventional amino acids may also be suitable components for polypeptides of the invention. Examples of unconventional amino acids include: 4-hydroxyproline, γ-carboxyglutamate, ε-Ν,Ν,Ν-trimethyllysine, ε-Ν-5 acetyllysine, O-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, σ-Ν-methylarginine, and other similar amino acids and imino acids (e.g., 4-hydroxypioline). In the polypeptide notation used herein, the left-hand direction is the aminn terminal direction and the right-hand direction is the carboxyl-terminal direction, in accordance with standard usage and conventioa 10 Similarly, unless specified otherwise, the left-hand end of single-stranded polynucleotide sequences is the 5' end; the left-hand direction of double-stranded polynucleotide sequences is referred to as the 5' direction. The direction of 5' to 3' addition of nascent RNA transcripts is referred to as the transcription direction; sequence regions on the DNA strand having the same sequence as the RNA transcript that are 5’ to 15 the 5' end of the RNA transcript are referred to as "upstream sequences"; sequence regions on the DNA strand having the same sequence as the RNA transcript that are 3' to the 3' end of the RNA transcript are referred to as "downstream sequences".
Naturally occurring amino acid residues may be divided into classes based on common side chain properties: 20 1) hydrophobic: norieucine (Nor or Nle), Met, Ala, Val, Leu, lie; 2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gin; 3) acidic: Asp, Glu; 4) basic: His, Lys, Arg; 5) residues that influence chain orientation: Gly, Pro; and 25 6) aromatic: Tip, Tyr, Phe.
Conservative amino acid substitutions may involve exchange of a member of one of these classes with another member of the same class. Conservative amino acid substitutions may encompass non-naturally occurring amino acid residues, which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological 26 2013204136 12 Apr 2013 systems. These include peptidomimetics and other reversed or inverted forms of amino acid moieties.
Non-conservative substitutions may involve the exchange of a member of one of these classes for a member from another class. Such substituted residues may be 5 introduced,,/?)/' example, into regions of a human antibody that are homologous with nonhuman antibodies, or into the non-homologous regions of the molecule.
In making such changes, according to certain embodiments, the hydropathic index of amino acids may be considered. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics. They are: isoleucine 10 (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); praline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
The importance of the hydropathic amino acid index in conferring interactive 15 biological function on a protein is understood in the art (see, for example, Kyte et ah, 1982, J. Mol. Biol. 157:105-131), It is known that certain amino acids maybe substituted for other amino acids having a similar hydropathic index or score and still retain a similar biological activity. In making clianges based upon the hydropathic index, in certain embodiments, the substitution of amino acids whose hydropathic indices are within ±2 is 20 included. In certain embodiments, those that are within ±1 are included, and in certain embodiments, those within ±0.5 are included.
It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity, particularly where the biologically functional protein or peptide thereby created is intended for use in immunological 25 embodiments, as disclosed herein. In certain embodiments, the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with its iminunogenicity and antigenicity, le., with a biological property of the protein.
The following hydrophilicity values have been assigned to these amino acid 30 residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ± 1); glutamate (+3.0 ± 1); serine (+0.3); asparagine (+0.2); glutamine (+0,2); glycine (0); threonine (-0.4); proline (-0.5 ± 27 2013204136 12 Apr 2013 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5) and tryptophan (-3.4). In making changes based upon similar hydrophilicity values, in certain embodiments, the substitution of amino acids whose hydrophilicity values are within ±2 is included, in 5 certain embodiments, those that are within ±1 are included, and in certain embodiments, those within ±0.5 are included. One may also identify epitopes from primary amino acid sequences on the basis of hydrophilicity. These regions are also referred to as "epitopic core regions."
Exemplary amino acid substitutions are set forth in Table 1. 10 Table 1
Amino Acid Substitutions
Original Residues Exemplary Substitutions Preferred Substitutions Ala Val, Leu, lie Val Arg Lys, Gin, Asn Lys Asn Gin Gin Asp Glu Glu Cys Ser, Ala Ser Gin Asn Asn Glu Asp Asp Gly Pro, Ala Ala His Asn, Gin, Lys, Aig Arg lie Leu, Val, Met, Ala, Pile, Norleucine Leu Leu Norleucine, He, Val, Met, Ala, Plie lie Lys Arg, 1,4 Diamino-butyric Acid, Gin, Asn Arg Met Leu, Phe, He Leu Phe Leu, Val, He, Ala, Tyr Leu Pro Ala Gly Scr Thr, Ala, Cys Thr Thr Ser Ser Tip Tyr, Phe Tyr Tyr Tip, Phe, Thr, Ser Phe Val lie, Met, Leu, Phe, Ala, Norleucine Leu A skilled artisan will be able to determine suitable valiants of polypeptides as set forth herein using well-known techniques. In certain embodiments, one skilled in the art 15 may identify suitable areas of the molecule that may be changed without destroying 28 2013204136 12 Apr 2013 activity by targeting regions not believed to be important for activity. In other embodiments, the skilled artisan can identify residues and portions of the molecules that are conserved among similar polypeptides. In further embodiments, even areas that may be important for biological activity or for structure may be subject to conservative amino 5 acid substitutions without destroying the biological activity or without adversely affecting the polypeptide structure.
Additionally, one skilled in the art can review structure-function studies identifying residues in similar polypeptides that are important for activity or structure. In view of such a comparison, the skilled artisan can predict the importance of amino acid 10 residues in a protein that correspond to amino acid residues important for activity or structure in similar proteins. One skilled in the art may opt for chemically similar amino acid substitutions for such predicted important amino acid residues.
One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar polypeptides. In view of such 15 information, one skilled in the art may predict the alignment of amino acid residues of an antibody with respect to its three-dimensional structure. In certain embodiments, one skilled in the art may choose to not make radical changes to amino acid residues predicted to be on the surface of the protein, since such residues may be involved in important interactions with other molecules. Moreover, one skilled in the art may generate test 20 variants containing a single amino acid substitution at each desired amino acid residue. The variants can then be screened using activity assays known to those skilled in the art. Such variants could be used to gather information about suitable variants. For example, if one discovered that a change to a particular amino acid residue resulted in destroyed, undesirably reduced, or unsuitable activity, variants with such a change can be avoided. 25 hr other words, based on information gathered from such routine experiments, one skilled in the art can readily determine the amino acids where further substitutions should be avoided either alone or in combination with other mutations. A number of scientific publications have been devoted to the prediction of secondary structure. See Moult, 1996, Curr. Op. in Biotech. 7:422-427; Chou et al., 30 1974, Biochemistry 13:222-245; Chou et al, 1974, Biochemistry 113:211-222; Chou et al., 1978, Adv. Enzymol, Relat. Areas Mol. Biol. 42:45-148; Chou et al., 1979, Ann, Rev. Biocheni. 47:251-276; and Chou et al., 1979, Biophys. J. 26:367-384. Moreover, 29 2013204136 12 Apr 2013 computer programs are currently available to assist with predicting secondary structure. One method of predicting secondary structure is based upon homology modeling. For example, two polypeptides or proteins that have a sequence identity of greater than 30%, or similarity greater than 40% often have similar structural topologies. The recent growth 5 of the protein structural database (PDB) has provided enhanced predictability of secondary structure, including the potential number of folds within a polypeptide’s or protein’s structure. See Holm et al, 1999, NucL Acid. Res. 27:244-247. It has been suggested (Brenner et al, 1997, Curr. Op. Struct. Biol. 7:369-376) that there are a limited number of folds in a given polypeptide or protein and that once a critical number of 10 structures have been resolved, stractural prediction will become dramatically more accurate.
Additional methods of predicting secondary structure include "threading" (Jones, 1997, Curr. Optn. Struct. Biol. 7:377-87; Sippl et al, 1996, Structure 4:15-19), "profile analysis" (Bowie et al., 1991, Science 253:164-170; Gribskov et al, 1990, Meth. Enzym. 15 183:146-159; Gribskov et al, 1987, Proc. Nat. Acad. Set. 84:4355-4358), and "evolutionary linkage" (See Holm, 1999, supra·, and Brenner, 1997, supra).
In certain embodiments, antibody valiants include glycosylation Variants wherein the number and/or type of glycosylation site has been altered compared to the amino acid sequences of the parent polypeptide. In certain embodiments, protein variants comprise a 20 greater or a lesser number ofN-linked glycosylation sites than the native protein. AnN-linked glycosylation site is characterized by the sequence: Asn-X-Ser or Asn-X-Thr, wherein the amino acid residue designated as X may be any amino acid residue except proline. The substitution of amino acid residues to create this sequence provides a potential new site for the addition of an N-linked carbohydrate chain. Alternatively, 25 substitutions that eliminate this sequence will remove an existing N-linked carbohydrate chain. Also provided is a rearrangement of N-linked carbohydrate chains wherein one or more N-linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more newN-linked sites are created, Additional preferred antibody variants include cysteine variants wherein one or more cysteine residues are deleted from 30 or substituted for another amino acid (e.g., serine) compared to the parent amino acid sequence, Cysteine variants may be useful when antibodies must be refolded into a biologically active conformation such as after the isolation of insoluble inclusion bodies. 30 2013204136 12 Apr 2013
Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines.
According to certain embodiments, amino acid substitutions are those that: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter 5 binding affinity for forming protein complexes, (4) alter binding affinities, and/or (5) confer or modify other physicochemical or fbnctional properties on such polypeptides. According to certain embodiments, single or multiple amino acid substitutions (in certain embodiments, conservative amino add substitutions) may be made in the naturally occurring sequence (in certain embodiments, in the portion of the polypeptide outside the 10 domain(s) forming intermolecular contacts). In preferred embodiments, a conservative amino acid substitution typically does not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence). Examples of art-recognized polypeptide 15 secondary and tertiary structures are described in Proteins, Structures and Molecular Principles, (Creighton, ed.), 1984, W. H. Freeman and Company, New York; Introduction to Protein Structure (C. Branden and J. Tooze, eds.), 1991, Garland Publishing, New York, N.Y.; and Thornton et al. (1991), Nature 354:105. each of which are incorporated herein by reference. 20 Preparation of Antibodies
Naturally occurring antibody structural units typically comprise a tetramer. Each such tetramer typically is composed of two identical pairs of polypeptide chains, each pair having one full-length "light" chain (typically having a molecular weight of about 25 kDa) and one full-length "heavy" chain (typically having a molecular weight of about 50-25 70 kDa). The amino-terminal portion of each chain typically includes a variable region of about 100 to 110 or more amino acids that typically is responsible for antigen recognition. The carboxy-terminal portion of each chain typically defines a constant region responsible for effector function. Human light chains are typically classified as kappa and lambda light chains. Heavy chains are typically classified as mu, delta, gamma, 30 alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. IgG has several subclasses, including, but not limited to, IgGl, IgG2, IgG3, and IgG4. IgM has subclasses including, but not limited to, IgMl and IgM2. IgA is 31 2013204136 12 Apr 2013 similarly subdivided into subclasses including, but not limited to, IgAl and IgA2. Within full-length light and heavy chains, typically, a “J” region of about 12 or more amino acids joins the variable region and constant regions, with the heavy chain also including a "D" region of about 10 more amino acids. See, e.g., Fundamental Immunology, Ch. 7, 2nd ed., 5 (Paul, W., ed.), 1989, Raven Press, N.Y. (incorporated by reference in its entirety for all purposes). The combination of the variable regions of each light chain/heavy chain pair typically forms the antigen-binding site.
The variable regions of each of the heavy chains and light chains typically exhibit the same general structure comprising four relatively conserved framework regions (FR) 10 joined by three hyper variable regions, also called complementarity determining regions or CDRs. The CDRs from the two chains of each pair typically are aligned by the framework regions, which alignment may enable binding to a specific epitope. From N-terminal to C-terminal, both light and heavy chain variable regions typically comprise the domains FR1, CDR1, FR2, CDR2, ER3, CDR3 and FR4. The assignment of amino acids 15 to each domain is typically in accordance with the definitions of Kabat Sequences of Proteins of Immunological Interest (1987 and 1991, National Institutes of Health, Bethesda, Md.), Chothia &amp; Lesk, 1987, J Mol. Biol 196:901-917, or Chothia et al, 1989, Nature 342:878-8831.
Antibodies became useful and of interest as pharmaceutical agents with the 20 development of monoclonal antibodies. Monoclonal antibodies are produced using any method that produces antibody molecules by continuous cell lines in culture. Examples of suitable methods for preparing monoclonal antibodies include the hybridoma methods of Kohler et al (1975, Nature 256:495-497) and the human B-cell hybridoma method (Kozbor, 1984, J. Immunol. 133:3001; and Brodeur et al., 1987, Monoclonal Antibody 25 Production Techniques and Applications, (Marcel Dekker, Inc., New York), pp. 51-63).
Monoclonal antibodies may be modified for use'as therapeutics.' One example is a “chimeric” antibody in which a portion of the heavy chain and/or light chain is identical with or homologous to a corresponding sequence in antibodies derived from a particular species or belonging to a particular- antibody class or subclass, while the remainder of the 30 chain(s) is/are identical with or homologous to a corresponding sequence in antibodies derived from another species or belonging to another antibody class or subclass. Other examples are fragments of such antibodies, so long as they exhibit the desired biological 32 2013204136 12 Apr 2013 activity. See, U.S. Patent No. 4,816,567; and Morrison et al. (1985), Proc. Natl. Acad. Sci. USA £1:6851-6855, A related development is the “CDR-grafted” antibody, in which the antibody comprises one or more complementarity determining regions (CDRs) from a particular· species or belonging to a particular antibody class or subclass, while the 5 remainder of the antibody chain(s) is/are identical with or homologous to a corresponding sequence in antibodies derived from another species or belonging to another antibody class or subclass.
Another development is the “humanized” antibody. Methods for humanizing non-human antibodies are well known in the art. (See U.S. Patent Nos. 5,585,089, and 10 5,693,762). Generally, a humanized antibody is produced by a non-human animal, and then certain amino acid residues, typically from non-antigen recognizing portions of the antibody, are modified to be homologous to said residues in a human antibody of corresponding isotype. Humanization can be performed, for example, using methods described in the art (Jones et al., 1986, Nature 321:522-525; Riechmann el al., 1988, 15 Nature 332:323-327; Verhoeyen et al., 1988, Science 239:1534-15351. by substituting at least a portion of a rodent variable region for the corresponding regions of a human antibody,
More recent and more promising is the development of human antibodies without exposure of antigen to human beings (“fully human antibodies”). Using transgenic 20 animals (e.g., mice) that are capable of producing a repertoire of human antibodies in the absence of endogenous mouse immunoglobulin production, such antibodies are produced by immunization with an antigen (typically having at least 6 contiguous amino acids), optionally conjugated to a carrier, See, for example, Jakobovits et al., 1993, Proc. Natl. Acad. Sci. USA ^0:2551-2555; Jakobovits et al., 1993, Nature 362:255-258: and 25 Bruggennann et al, 1993, Year in Immunol 2:33. In one example of these methods, transgenic animals- are produced by incapacitating the endogenous mouse immunoglobulin loci encoding the mouse heavy and light immunoglobulin chains therein, and inserting loci encoding human heavy and light chain proteins into the genome thereof, Partially modified animals, which have less than the full complement of 30 modifications, are then cross-bred to obtain an animal having all of the desired immune system modifications. When administered an immunogen, these transgenic animals produce antibodies that are immunospecific for these antigens having human (rather than 33 2013204136 12 Apr 2013 in mine) amino acid sequences, including variable regions. See PCT Publication Nos. W096/33735 and W094/02602, incorporated by reference. Additional methods are described in U.S. Patent No. 5,545,807, PCT Publication Nos. WO91/10741, W090/04036, and in EP 546073B1 and BP 546073A1, incorporated by reference. 5 Human antibodies may also be produced by the expression of recombinant DNA in host cells or by expression in hybridoma cells as described herein.
Fully human antibodies can also be produced from phage-display libraries (as disclosed in Hoogenbooin ei ah, 1991,,/. Mol. Biol. 227:381; and Marks el ah, 1991, J. Mol. Biol. 222:5811. These processes mimic immune selection through the display of 10 antibody repertoires on the surface of filamentous bacteriophage, and subsequent selection of phage by their binding to an antigen of choice. One such technique is described in PCT Publication No. WO99/10494, incorporated by reference, which describes the isolation of high affinity and functional agonistic antibodies for MPL- and msk- receptors using such an approach. 15 Once the nucleotide sequences encoding such antibodies have been determined, chimeric, CDR-grafted, humanized, and fully human antibodies also may be produced by recombinant methods. Nucleic acids encoding the antibodies are introduced into host cells and expressed using materials and procedures generally known in the art.
The invention provides one or a plurality of fully human monoclonal antibodies 20 against human IL-1R1. Preferably, the antibodies bind the third domain of IL-1R1. In preferred embodiments, the invention provides nucleotide sequences encoding, and amino acid sequences comprising, heavy and light chain immunoglobulin molecules, particularly sequences corresponding to the variable regions thereof. In preferred embodiments, sequences corresponding to complementarity determining regions (CDR's), 25 specifically from CDR1 through CDR3, are provided. In additional preferred embodiments, the invention provides hybridoma cell lines expressing such immunoglobulin molecules and monoclonal antibodies produced therefrom, most preferably purified human monoclonal antibodies against human IL-1R1.
The ability to clone and reconstruct megabase-sized human loci in yeast artificial 30 chromosomes (YACs) and to introduce them into the mouse germline provides an advantageous approach to elucidating the functional components of very large or crudely mapped loci as well as generating useful models of human disease. Furthermore, the 34 2013204136 12 Apr 2013 utilization of such technology for substitution of mouse loci with their human equivalents provides unique insights into expression and regulation of human gene products during development, their communication with other systems, and their involvement in disease induction and progression. 5 An important practical application of such a strategy is the "humanization" of the mouse humoral immune system. Introduction of human immunoglobulin (Ig) loci into mice in which the endogenous Ig genes have been inactivated offers the opportunity to study the mechanisms underlying programmed expression and assembly of antibodies as well as their role in B-cell development. Furthermore, such a strategy provides a source 10 for production of fully human monoclonal antibodies (MAbs), particularly for use as therapeutic agents. Fully human antibodies are expected to minimize the immunogenic and allergic responses intrinsic to mouse or mouse-derivatized Mabs, and to thereby increase die efficacy and safety of administered antibodies in therapeutic applications. Fully human antibodies can be used in the treatment of chronic and recurring human 15 diseases, such as osteoarthritis, rheumatoid arthritis, and other inflammatory conditions, the treatment thereof requiring repeated antibody administration.
One skilled in the art can engineer mouse strains deficient in mouse antibody production with large fragments of the human Ig loci so that such mice produce human antibodies in the absence of mouse antibodies. Large human Ig fragments may preserve 20 the large variable gene diversity as well as the proper regulation of antibody production and expression. By exploiting the mouse machinery for antibody diversification and selection and the lack of immunological tolerance to human proteins, the reproduced human antibody repertoire in these mouse strains yields high affinity antibodies against any antigen of interest, including human antigens. Using the hybridoma technology, 25 antigen-specific human MAbs with the desired specificity may be produced and selected.
In certain embodiments, the skilled artisan can use constant regions from species other than human along with the human variable region(s) in such mice to produce chimeric antibodies. The antibodies of the invention can be produced by immunizing such animals with full-length IL-1R1, soluble forms of IL-lRl, or a fragment thereof. 30 See, for example, International Patent Application, Publication WO 93/12227).
The CDRs of the light and heavy chain variable regions of anti-IL-lRl antibodies of the invention can be grafted to framework regions (FRs) from the same, or another, 35 2013204136 12 Apr 2013 species. In certain embodiments, the CDRs of the light and heavy chain variable regions of anti-IL-lRl antibody may be grafted to consensus human FRs. To create consensus human FRs, FRs from several human heavy chain or light chain amino acid sequences are aligned to identify a consensus amino acid sequence. The FRs of the anti-IL-lRl 5 antibody heavy chain or light chain can be replaced with the FRs from a different heavy chain or light chain. Rare amino acids in the FRs of the heavy and light chains of anti-IL-1R1 antibody typically are not replaced, while the rest of the FR amino acids can be replaced. Rare amino acids are specific amino acids that are in positions in which they are not usually found in FRs. The grafted variable regions from anti-IL-lRl antibodies of 10 the invention can be used with a constant region that is different from the constant region of anti-IL-lRl antibody. Alternatively, the grafted variable regions are part of a single chain Fv antibody. CDR grafting is described, e.g., in U.S. Patent Nos. 6,180,370, 5,693,762, 5,693,761, 5,585,089, and 5,530,101, which are hereby incorporated by reference for any purpose. 15 In certain embodiments, the invention provides anti-ILl-Rl antibodies that comprise a human heavy chain CDR1 region having an amino acid sequence of SEQ ID NO: 61, SEQ ID NO: 62, or SEQ ID NO: 63; a human heavy chain CDR2 region having an amino acid sequence of SEQ ID NO: 64, SEQ ID NO: 65, or SEQ ID NO: 66; and/or a human heavy chain GDR3 region having an amino acid sequence of SEQ ID NO: 67, 20 SEQ ID NO: 68, or SEQ ID NO: 69.
In other embodiments, the invention provides anti-ILl-Rl antibodies that comprise a human light chain CDR1 region having an amino acid sequence of SEQ ID NO: 70 or SEQ ID NO: 71; a human heavy chain CDR2 region having an amino acid sequence of SEQ ID NO: 72 or SEQ ID NO: 73; and/or a human heavy chain CDR3 25 region having an amino acid sequence of SEQ ID NO: 74 or SEQ ID NO: 75.
Antibodies of the invention are preferably prepared using transgenic mice that have a substantial portion of the human antibody-producing locus inserted in antibody-producing cells of the mice, and that are further engineered to be deficient in producing endogenous, murine, antibodies. Such mice are capable of producing human 30 immunoglobulin molecules arid antibodies and do not produce or produce substantially reduced amounts of murine immunoglobulin molecules and antibodies. Technologies utilized for achieving this result are disclosed in the patents, applications, and references 36 2013204136 12 Apr 2013 disclosed in the specification herein. In preferred embodiments, the skilled worker may employ methods as disclosed in International Patent Application Publication No. WO 98/24893, which is hereby incorporated by reference for any purpose. See also Mendez et ah, 1997, Nature Genetics 15:146-156, which is hereby incorporated by reference for 5 any purpose.
The monoclonal antibodies (MAbs) of the invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology, e.g., the standard somatic cell hybridization technique of Kohler and Milstein, 1975, Nature 256:495. Although somatic cell hybridization procedures are preferred, in principle, other 10 techniques for producing monoclonal antibodies can be employed, e.g., viral or oncogenic transformation of B-lymphocytes.
In a preferred embodiment, human monoclonal antibodies directed against IL-1R1 can be generated using mice referred to as <4HuMab” mice, contain a human immunoglobulin gene minilocus that encodes unrearranged human heavy (// and γ) and κ 15 light chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous μ and κ chain loci. Lonberg et al., 1994, Nature 368:856-859. Accordingly, the mice exhibit reduced expression of mouse IgM or K and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgG κ monoclonal 20 antibodies. Lonberg et al, supra·, Lonberg and Huszar, 1995, Intern. Rev. Immunol. 13:65-93; Harding and Lonberg, 1995, Ann. N.Y. Acad. Set 764:536-546. The preparation of HuMab mice is described in detail in Taylor et al, 1992, Nucleic Acids Res. 20:6287-6295; Chen et al., 1993, International Immunology 5:647-656; Tuaillon et al, 1994, J. Immunol. 152:2912-2920: Lonberg et al., 1994, Nature 368:856-859: 25 Lonberg, 1994, Handbook of Exp. Pharmacology 113:49-101: Taylor et al, 1994, International Immunology 6:579-591; Lonberg &amp; Huszar, 1995, Intern. Rev. Immunol. 13:65-93; Harding &amp; Lonberg, 1995, Ann. N.Y. Acad. Sci 764:536-546; Fishwild et al., 1996, Nature Biotechnology 14:845-851. the contents of all of which are hereby incorporated by reference in their entirety. See further U.S. Patent Nos. 5,545,806; 30 5,569,825; 5,625,126; 5,633,425; 5,789,650; 5,877,397; 5,661,016; 5,814,318; 5,874,299; and 5,770,429; all to Lonberg and Kay, as well as U.S. Patent No. 5,545,807 to Surani et al.·, International Patent Application Publication Nos. WO 93/1227, published June 24, 37 2013204136 12 Apr 2013 1993; WO 92/22646, published December 23, 1992; and WO 92/03918, published March 19, 1992, the disclosures of all of which are hereby incorporated by reference in their entirety. Alternatively, the HCo7 and HCol2 transgenic mice strains described in the Examples below can be used to generate human anti-lL-lRl antibodies. 5 Advantageously, fully human monoclonal antibodies specific for 3L-1R1 are produced as follows. Transgenic mice containing human immunoglobulin genes are immunized with the IL-lRl-related antigen of interest. Lymphatic cells (such as B-cells) from the mice that express antibodies are obtained. Such recovered cells are fused with a myeloid-type cell line to prepare immortal hybridoma cell lines, and such hybridoma cell 10 lines are screened and selected to identify hybridoma cell lines that produce antibodies specific to the antigen of interest, In certain embodiments, the production of a hybridoma cell line that produces antibodies specific to JL-1R1 is provided.
In preferred embodiments, antibodies of the invention are produced by hybridoma lines. In these embodiments, the antibodies of the invention bind to IL-1R1 with a 15 dissociation constant (3¾) of between approximately 4 pM and 100 pM. In certain embodiments of the invention, the antibodies bind to IL-1R1 with a 1¾ of less than about 20 pM, In other embodiments, the antibodies of the invention bind to the third domain of J L-1R1, The nucleotide and amino acid sequences of the third domain of human and rat IL1-R1 are shown in Figure 17. 20 In preferred embodiments, the antibodies of the invention are of the IgGl, IgG2, or IgG4 isotype, with the IgG2 isotype most preferred. In preferred embodiments of the invention, the antibodies comprise a human kappa light chain and a human IgGl, IgG2, or IgG4 heavy chain. In particular embodiments, the variable regions of the antibodies are ligated to a constant region other than the constant region for the IgGl, IgG2, or IgG4 25 isotype. In certain embodiments, the antibodies of the invention have been cloned for expression in mammalian cells.
In certain embodiments, conservative amino acid substitutions to the heavy and light chains of anti-IL-lRl antibody (and corresponding modifications to the encoding nucleotides) will produce anti-IL-lRl antibodies having functional and chemical 30 characteristics similar to those of anti-IL-lRl antibody. In contrast, substantial modifications in the junctional and/or chemical characteristics of anti-IL-lRl antibody may be accomplished by selecting substitutions in the amino acid sequence of the heavy 38 2013204136 12 Apr 2013 and light chains that differ significantly in their effect on maintaining (a) the structure of the molecular backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. 5 For example, a "conservative amino acid substitution" may involve a substitution of a native amino acid residue with a nonnative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position. Furthermore, any native residue in the polypeptide may also be substituted with alanine, as has been previously described for "alanine scanning mutagenesis" (Wells, 1991, Methods Enzymol. 10 202:390 (ed. J.J. Langone), Academic Press, London).
Desired amino acid substitutions (whether conservative or non-conservative) can be determined by those skilled in the art at the time such substitutions are desired. In certain embodiments, amino acid substitutions can be used to identify important residues of anti-IL-lRl antibody, or to increase or decrease the affinity of the anti-IL-lRl 15 antib o dies describ ed herein,
In alternative embodiments, antibodies of the invention can be expressed in cell lines other than hybridoma cell lines. In these embodiments, sequences encoding particular antibodies can be used for transformation of a suitable mammalian host cell. According to these embodiments, transformation can be achieved using any known 20 method for introducing polynucleotides into a host cell, including, for example packaging the polynucleotide in a virus (or into a viral vector) and transducing a host cell with the virus (or vector) or by transfection procedures known in the art. Such procedures are exemplified by U.S. Pat, Nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455 (all of which are hereby incorporated herein by reference for any pmpose), Generally, the 25 transformation procedure used may depend upon the host to be transformed. Methods for introducing heterologous polynucleotides into mammalian cells are well known in the art and include, but are not limited to, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct micro injection of the 30 DNA into nuclei.
According to certain embodiments of the methods of the invention, a nucleic acid molecule encoding the amino acid sequence of a heavy chain constant region, a heavy 39 2013204136 12 Apr 2013 chain variable region, a light chain constant region, or a light chain variable region of an IL-1R1 antibody of the invention is inserted into an appropriate expression vector using standard ligation techniques. In a preferred embodiment, the IL-1R1 heavy or light chain constant region is appended to the C-terminus of the appropriate variable region and is 5 ligated into an expression vector. The vector is typically selected to be functional in the particular host cell employed (i.e., the vector is compatible with the host cell machinery such that amplification of the gene and/or expression of the gene can occur). For a review of expression vectors, see, Goeddel (ed.), 1990, Math. Enzymol. Vol. 185, Academic Press. N.Y. 10 Typically, expression vectors used in any of the host cells will contain sequences for plasmid maintenance and for cloning and expression of exogenous nucleotide sequences. Such sequences, collectively referred to as “flanking sequences” in certain embodiments will typically include one or more of the following nucleotide sequences: a promoter, one or more enhancer sequences, an origin of replication, a transcriptional 15 termination sequence, a complete intron sequence containing a donor and acceptor splice site, a sequence encoding a leader sequence for polypeptide secretion, a ribosome binding site, a polyadenylation sequence, a polylinker region for inserting the nucleic acid encoding the polypeptide to be expressed, and a selectable marker element. Each of these sequences is discussed below. 20 Optionally, the vector may contain a “tag”-encoding sequence, i.e., an oligonucleotide molecule located at the 5’ or 3’ end of the IL-1R1 polypeptide coding sequence; the oligonucleotide sequence encodes polyHis (such as hexaHis), or another “tag” such as FLAG, HA (hemaglutinin influenza virus), or myc for which commercially available antibodies exist. This tag is typically fused to the polypeptide upon expression 25 of the polypeptide, and can serve as a means for affinity purification or detection of the IL-1R1 antibody from the host cell. Affinity purification can be accomplished, for example, by column chromatography using antibodies against the tag as an affinity matrix. Optionally, the tag can subsequently be removed from the purified IL-1R1 polypeptide by various means such as using certain peptidases for cleavage, 30 Flanking sequences may be homologous (i.e., from the same species and/or strain as the host cell), heterologous (i.e., from a species other than the host cell species or strain), hybrid (/. e., a combination of flanking sequences from more than one source), 40 2013204136 12 Apr 2013 synthetic or native. As such, the source of a flanking sequence may be any prokaryotic or eukaryotic organism, any vertebrate or invertebrate organism, or any plant, provided that the flanking sequence is functional in, and can be activated by, the host cell machinery.
Flanking sequences useful in the vectors of this invention maybe obtained by any 5 of several methods well known in the ait. Typically, flanking sequences useful herein will have been previously identified by mapping and/or by restriction endonuclease digestion and can thus be isolated from the proper tissue source using the appropriate restriction endonucleases. In some cases, the full nucleotide sequence of a flanking sequence may be known. Here, the flanking sequence may be synthesized using the 10 methods described herein for nucleic acid synthesis or cloning.
Where all or only a portion of the flanking sequence is known, it may be obtained using polymerase chain reaction (PCR) and/or by screening a genomic library with a suitable probe such as an oligonucleotide and/or flanking sequence fragment from the same or another species. Where the flanking sequence is not known, a fragment of DNA 15 containing a flanking sequence may be isolated from a larger piece of DNA that may contain, for example, a coding sequence or even another gene or genes. Isolation maybe accomplished by restriction endonuclease digestion to produce the proper DNA fragment followed by isolation using agarose gel purification, Qiagen® column chromatography (Chatsworth, CA), or other methods known to the skilled artisan. The selection of 20 suitable enzymes to accomplish this purpose will be readily apparent to one of ordinary skill in the art.
An origin of replication is typically a part of those prokaryotic expression vectors purchased commercially, and the origin aids in the amplification of the vector in a host cell. If the vector of choice does not contain an origin of replication site, one may be 25 chemically synthesized based on a known sequence, and ligated into the vector, For example, the origin of replication from the plasmid pBR322 (New England Biolabs, Beverly, MA) is suitable for most gram-negative bacteria and various viral origins (e.g., SV40, polyoma, adenovirus, vesicular stomatitus virus (VSV), or papillomaviruses such as HPV or BPV) are useful for cloning vectors in mammalian cells. Generally, the origin 30 of replication component is not needed for mammalian expression vectors (for example, the SV40 origin is often used only because it also contains the virus early promoter). 41 2013204136 12 Apr 2013 A transcription termination sequence is typically located 3’ to the end of a polypeptide coding region and serves to terminate transcription. Usually, a transcription termination sequence in prokaryotic cells is a G-C rich fragment followed by a poly-T sequence. While the sequence is easily cloned from a library or even purchased 5 commercially as part of a vector, it can also be readily synthesized using methods for nucleic acid synthesis such as those described herein. A selectable marker gene encodes a protein necessary for the survival and growth of a host cell grown in a selective culture medium. Typical selection marker genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, 10 tetracycline, or kanamycin for prokaryotic host cells; (b) complement auxotrophic deficiencies of the cell; or (c) supply critical nutrients not available from complex or defined media. Preferred selectable markers are the kanamycin resistance gene, the ampicillin resistance gene, and the tetracycline resistance gene. A neomycin resistance gene may also be used for selection in both prokaryotic and eukaryotic host cells. 15 Other selectable genes may be used to amplify the gene that will be expressed.
Amplification is the process wherein genes that are in greater demand for the production of a protein critical for growth or cell survival are reiterated generally in random within the chromosomes of successive generations of recombinant cells. Examples of suitable selectable markers for mammalian cells include dihydrofolate reductase (DHER) and 20 promoterless thymidine kinase. Mammalian cell transformants are placed under selection pressure wherein only the transformants are uniquely adapted to survive by virtue of the selectable gene present in the vector. Selection pressure is imposed by culturing the transformed cells under conditions in which the concentration of selection agent in the medium is successively increased, thereby leading to the amplification of both the 25 selectable gene and the DNA that encodes another gene, such as IL-1R1 polypeptide comprising the vector. As a result, increased quantities of a polypeptide such as IL-1R1 polypeptide are synthesized from the amplified DNA A ribosome-binding site is usually necessary for translation initiation of mRNA and is characterized by a Shine-Dalgarno sequence (prokaryotes) or a Kozak sequence 30 (eukaryotes). The element is typically located 3’ to the promoter and 5’ to the coding sequence of the polypeptide to be expressed. 42 2013204136 12 Apr 2013
Bi some cases, such as where glycosylation is desired in a eukaryotic host cell expression system, one may manipulate the various pre- or prosequences to improve glycosylation or yield. For example, one may alter the peptidase cleavage site of a particular signal peptide, or add pro-sequences, which also may affect glycosylation. The 5 final protein product may have, in file -1 position (relative to the first amino acid of the mature protein) one or more additional amino acids incident to expression, which may not have been totally removed. For example, the final protein product may have one or two amino acid residues found in the peptidase cleavage site, attached to the amino-terminus. Alternatively, use of some enzyme cleavage sites may result in a slightly truncated form 10 of the desired polypeptide, if the enzyme cuts at such area within the mature polypeptide.
The expression and cloning vectors of the invention will typically contain a promoter that is recognized by the host organism and operably linked to the molecule encoding the auti-IL-lRl antibody. Promoters are untranscribed sequences located upstream (i.e,, 5’) to the start codon of a structural gene (generally within about 100 to 15 1000 bp) that control the transcription of the structural gene. Promoters are conventionally grouped into one of two classes: inducible promoters and constitutive promoters. Inducible promoters initiate increased levels of transcription from DNA under their control in response to some change in culture conditions, such as the presence or absence of a nutrient or a change in temperature. Constitutive promoters, on the other 20 hand, initiate continual gene product production; that is, there is little or no control over gene expression. A large number of promoters, recognized by a variety of potential host cells, are well known. A suitable promoter is operably linked to the DNA encoding heavy chain or light chain comprising an anti-IL-lRl antibody of the invention by removing the promoter from the source DNA by restriction enzyme digestion and inserting the desired 25 promoter sequence into the vector.
Suitable promoters for use with yeast hosts are also well known in the ait. Yeast enhancers are advantageously used with yeast promoters. Suitable promoters for use with mammalian host cells are well known and include, but are not limited to, those obtained from the genomes of viruses such as polyoma vires, fowlpox virus, adenovirus (such as 30 Adenovirus 2), bovine papilloma vires, avian sarcoma vires, cytomegalovirus, retroviruses, hepatitis-B vires and most preferably Simian Virus 40 (SV40). Other 43 2013204136 12 Apr 2013 suitable mammalian promoters include heterologous mammalian promoters, for example, heat-shock promoters and the actin promoter.
Additional promoters which may be of interest include, but are not limited to; the SV40 early promoter region (Bemoist and Chambon, 1981, Nature 290:304-1 O'); the 5 CMV promoter; the promoter contained in the 3’ long terminal repeat of Rous sarcoma vims (Yamamoto et ah, 1980, Cell 22:787-97); the herpes thymidine kinase promoter (Wagner et ah, 1981, Proc. Natl. Acad. Sci. USA 78:1444-45); the regulatory sequences of the metallothionine gene (Brinster et ah, 1982, Nature 296:39-42); prokaryotic, expression vectors such as the beta-lactamase promoter (Villa-Kamaroff et ah, 1978, 10 Proc. Natl. Acad. Sci. USA 75:3727-31); or the tac promoter (DeBoer et ah, 1983, Proc. Natl Acad. Sci. USA 80:21-25), Also of interest are the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: the elastase I gene contr ol region that is active in pancreatic acinar cells (Swift et ah, 1984, Cell 38:639-46; Ornitz et ah, 1986, Cold Spring Harbor Symp. Quant. Biol. 15 50:399-409 (1986); MacDonald, 1987, Hepatology 7:425-515); the insulin gene control region that is active in pancreatic beta cells (Hanahan, 1985, Nature 315:115-221: the immunoglobulin gene control region that is active in lymphoid cells (Grosschedl et ah, 1984, Cell 38:647-58; Adames et ah, 1985, Nature 318:533-38: Alexander et ah, 1987, Mol. Cell. Biol. 2:1436-44); the mouse mammary tumor virus control region that is active 20 in testicular, breast, lymphoid and mast cells (Leder et ah, 1986, Cell 45:485-95); the albumin gene control region that is active in liver (Pinkert et ah, 1987, Genes and Devel, 1:268-76); the alpha-feto-protein gene control region that is active in liver (Krumlauf et ah, 1985, Mol. Cell. Biol. 5:1639-48; Hammer et ah, 1987, Science 235:53-58); the alpha 1-antitrypsin gene control region that is active in liver (ICelsey et ah, 1987, Genes and 25 Devel.' 1:161-71); the beta-globin gene control region that is active in myeloid cells (Mogram et ah, 1985, Nature 315:338-40; Kollias et ah, 1986, Cell 46:89-94); the myelin basic protein gene control region that is active in oligodendrocyte cells in the brain (Readhead et ah, 1987, Cell 48:703-12); the myosin light chain-2 gene control region that is active in skeletal muscle (Sani, 1985, Nature 314:283-861: and the gonadotropic 30 releasing hormone gene control region that is active in the hypothalamus (Mason et ah, 1986, Science 234:1372-78). 44 2013204136 12 Apr 2013
An enhancer sequence may be inserted into the vector to increase transcription of DNA encoding light chain or heavy chain comprising an anti-IL-lRl antibody of the invention by higher eukaryotes. Enhancers are cis-acting elements of DNA, usually about 10-300 bp in length, that act on the promoter to increase transcription. Enhancers are 5 relatively oiientation- and position-independent. They have been found 5’ and 3' to the transcription unit. Several enhancer sequences available from mammalian genes (e.g., globin, elastase, albumin, alpha-feto-protein and insulin) are known. Typically, however, an enhancer from a virus is used. The SV40 enhancer, the cytomegalovirus early promoter enhancer, the polyoma enhancer, and adenovirus enhancers known in the art are 10 exemplary enhancing elements for the activation of eukaryotic promoters. While an enhancer may be spliced into the vector at a position 5’ or 3’ to a nucleic acid molecule, it is typically located at a site 5’ from the promoter·.
Expression vectors of the invention may be constructed from a starting vector such as a commercially available vector. Such vectors may or may not contain all of the 15 desired flanking sequences. Where one or more of the flanking sequences described herein are not already present in the vector, they may be individually obtained and ligated into the vector. Methods used for obtaining each of the flanking sequences are well known to one skilled in the art.
After the vector has been constructed and a nucleic acid molecule encoding light 20 chain or heavy chain or light chain and heavy chain comprising an anti-IL-lRl antibody has been inserted into the proper site of the vector, the completed vector may be inserted into a suitable host cell for amplification and/or polypeptide expression. The transformation of an expression vector for an anti-IL-lRl antibody into a selected host cell may be accomplished by well known methods including transfection, infection, 25 calcium phosphate co-precipitation, electroporation, microinjection, lipofection, DEAE-dextran mediated transfection, or other known techniques. The method selected will in part be a function of the type of host cell to be used. These methods and other suitable methods are well known to the skilled artisan, and are set forth, for example, in Sambrook etah, supra. 30 The host cell, when cultured under appropriate conditions, synthesizes an anti-IL- 1R1 antibody that can subsequently be collected from the culture medium (if the host cell secretes it into the medium) or directly from the host cell producing it (if it is not 45 2013204136 12 Apr 2013 secreted). The selection of an appropriate host cell will depend upon various factors, such as desired expression levels, polypeptide modifications that are desirable or necessary for activity (such as glycosylation or phosphorylation) and ease of folding into a biologically active molecule. 5 Mammalian cell lines available as hosts for expression are well known in the art and include, but are not limited to, many immortalized cell lines available from the American Type Culture Collection (A.T.C.C.), including but not limited to Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), and a number 10 of other cell lines. In certain embodiments, one may select cell lines by determining which cell lines have high expression levels and produce antibodies with constitutive IL-1R1 binding properties. In another embodiment, one may select a cell line from the B cell lineage that does not make its own antibody but has a capacity to make and secrete a heterologous antibody (e.g., mouse myeloma cell fines NSO and SP2/0). 15 Antibodies of the invention are useful for detecting IL-1R1 in biological samples and identification of cells or tissues that produce IL-1R1 protein. Said antibodies that bind to IL-1R1 and block interaction with other binding compounds have therapeutic use in modulating IL-1 mediated diseases. In preferred embodiments, antibodies to IL-1R1 can block IL-1R1 binding to IL-1/3 or IL-Ια, which can result in disruption of the IL-1 20 signal transduction cascade.
Antibodies of the invention that specifically bind to IL-1R1 may be useful in treatment of IL-1 mediated diseases, as discussed below. Said antibodies can be used in binding assays to detect IL-1R1 binding and their capacity to inhibit IL-1R1 from forming a complex with IL-1/3 and IL-1R accessory protein (IL-lRAcP) or with EL-la 25 and EL-lRacP. 46 2013204136 12 Apr 2013
In. certain embodiments, the invention provides methods for treating medical disorders associated with IL-1 mediated inflammatory reactions or IL-1 mediated immunoregulatory reactions. The methods of the invention include administering an anti-IL1R1 antibody of the invention to an individual who is afflicted with an inflammatory or 5 immunoregulatory disease that is mediated by IL-1. As used herein, the terms “illness,” disease, * medical condition,” or “abnormal condition,” are used interchangeably with the term “medical disorder.”
In a particular embodiment, the methods of the invention involve administering to a patient an anti- IL-1R1 antibody of the invention, thereby preventing the binding of IL-10 1 to its cell surface receptor (TGL-1R1).
To treat a medical disorder characterized by abnormal or excess expression of IL-1 or abnormal or excess IL-1 signaling, a molecule comprising an IL-1R type I antibody of this .invention is administered to the patient in an amount and for a time sufficient to induce a sustained improvement in at least one indicator that reflects the severity of the 15 disorder. An improvement is considered “sustained” if the patient exhibits the improvement on at least two occasions separated by one to four weeks. The degree of improvement is determined based on signs or symptoms, and may also employ questionnaires that are administered to the patient, such as quality-of-life questionnaires.
Various indicators that reflect the extent of the patient’s illness may be assessed 20 for determining whether the amount and time of the treatment is sufficient. The baseline value for the chosen indicator or indicators is established by examination of the patient prior to administration of the first dose of the antibody. Preferably, the baseline examination is done within about 60 days of administering the first dose. If the IL-1R antibody is being administered to treat acute symptoms, such as, for example, to treat 25 traumatic injuries (traumatic knee injuiy, stroke, head injury, etc.) the fust dose is administered as soon as practically possible after the injury or event has occurred.
Improvement is induced by repeatedly administering a dose of antibody until the patient manifests an improvement over baseline for the chosen indicator or indicators. In treating chronic conditions, this degree of improvement is obtained by repeatedly 30 administering this medicament over a period of at least a month or more, e.g., for one, two, or three months or longer, or indefinitely. A period of one to six weeks, or even a single dose, often is sufficient for treating acute conditions. 47 2013204136 12 Apr 2013
Although the extent of the patient’s illness after treatment may appear· improved according to one or more indicators, treatment may be continued indefinitely at the same level or at a reduced dose or frequency. Once treatment has been reduced or discontinued, it later may be resumed at the original level if symptoms should reappear. 5 Any efficacious route of administration may be used to therapeutically administer the antibody. The antibody may be injected via intra-articular, intravenous, intramuscular, intralesional, intraperitoneal, intracranial, inhalation or subcutaneous routes by bolus injection or by continuous infusion. For example, pulmonary diseases can involve intranasal and inhalation methods. Other suitable means of administration 10 include sustained release from implants, aerosol inhalation, eyedrops, oral preparations, including pills, syrups, lozenges or chewing gum, and topical preparations such as lotions, gels, sprays, ointments or other suitable techniques. Administration by inhalation is particularly beneficial when treating diseases associated with pulmonary disorders.
In one embodiment of the invention, an anti-IL-lRl antibody of the invention can 15 be administered once a month. In another embodiment the antibody is administered once every two weeks or one time per week to treat the various medical disorders disclosed herein. In yet another embodiment the antibody is administered at least two times per week, and in another embodiment is administered at least once per day. An adult patient is a person who is 18 years of age or older. If injected, the effective amount, per adult 20 dose, ranges from 1-200 mg/m2, or from 1-40 mg/m2 or about 5-25 mg/m2. Alternatively, a flat dose may be administered, whose amount may range from 2-400 mg/dose, 2-100 mg/dose or from about 10-80 mg/dose. If the dose is to be administered more than one time per week, an exemplary dose range is the same as the foregoing described dose ranges or lower. In one embodiment of the invention, the various indications described 25 below are treated by administering a preparation acceptable for injection containing IL-1 receptor antibody at 80-100 mg/dose, or alternatively, containing 80 mg per dose. The dose is administered repeatedly. If a route of administration other than injection is used, the dose is appropriately adjusted in accord with standard medical practices. For example, if the route of administration is inhalation, dosing may be one to seven times per 3 0 week at dose ranges from 10 mg/dose to 50 mg per dose.
In preferred embodiments, the invention also provides pharmaceutical compositions comprising a therapeutically effective amount of one or a plurality of the 48 2013204136 12 Apr 2013 antibodies of the invention together with a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative and/or adjuvant. Preferably, acceptable formulation materials are nontoxic to recipients at the dosages and concentrations employed. Tn preferred embodiments, pharmaceutical compositions comprising a therapeutically 5 effective amount of anti-IL-lRl antibodies are provided.
In certain embodiments, the pharmaceutical composition may contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition. In such embodiments, suitable formulation 10 materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HC1, citrates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing 15 agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides; disaccharides; and other carbohydrates (such as glucose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; emulsifying agents; hydrophilic polymers (such as polyvinylpyrrolidone); low molecular weight polypeptides; 20 salt-forming counterions (such as sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid or hydrogen peroxide); solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, 25 polysorbates such as polysorbate 20, polysorbate 80, triton, trimethamine, lecithin, cholesterol, tyloxapal); stability enhancing agents (such as sucrose or sorbitol); tonicity enhancing agents (such as alkali metal halides, preferably sodium or potassium chloride, mannitol sorbitol); delivery vehicles; diluents; excipients and/or pharmaceutical adjuvants. See, Remington's Pharmaceutical Sciences, 181'1 Edition, (A.R. Gennaro, ed.), 30 1990, Mack Publishing Company.
In certain embodiments, the optimal pharmaceutical composition will be determined by one skilled in the art depending upon, for example, the intended route of 49 2013204136 12 Apr 2013 administration, delivery format and desired dosage. See, for example, Remington's Pharmaceutical Sciences, supra. In certain embodiments, such compositions may influence the physical state, stability, rate of in vivo release and rate of in vivo clearance of the antibodies of the invention. 5 In certain embodiments, the primary vehicle or carrier in a pharmaceutical composition may be either aqueous or non-aqueous in nature. For example, a suitable vehicle or carrier may be water for injection, physiological saline solution or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin 10 are further exemplary vehicles. In preferred embodiments, pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, and may further include sorbitol or a suitable substitute therefor. In certain embodiments of the invention, anti-IL-lRl antibody compositions may be prepared for storage by mixing the selected composition having the desired degree of purity with optional formulation agents 15 (Remington’s Pharmaceutical Sciences, supra) in the fonn of a lyophilized cake or an aqueous solution. Further, in certain embodiments, the anti-IL-lRl antibody product may be formulated as a lyophilizate using appropriate excipients such as sucrose.
The pharmaceutical compositions of the invention can be selected for parenteral delivery. The compositions may be selected for inhalation or for delivery through the 20 digestive tract, such as orally. Preparation of such pharmaceutically acceptable compositions is within the skill of the art.
The formulation components are present preferably in concentrations that are acceptable to the site of administration. In certain embodiments, buffers are used to maintain the composition at physiological pH or at a slightly lower pH, typically within a 25 pH range of from about 5 to about 8.
When parenteral administration is contemplated, the therapeutic compositions for use in this invention may be provided in the form of a pyrogen-fiee, parenterally acceptable aqueous solution comprising the desired anti-IL-lRl antibody in a pharmaceutically acceptable vehicle. A particularly suitable vehicle for parenteral 30 injection is sterile distilled water in which the anti-IL-lRl antibody is formulated as a sterile, isotonic solution, properly preserved. In certain embodiments, the preparation can involve the formulation of the desired molecule with an agent, such as injectable 50 2013204136 12 Apr 2013 microspheres, bio-erodible particles, polymeric compounds (such as polylactic acid or polyglycolic acid), beads or liposomes, that may provide controlled or sustained release of the product which can be delivered via depot injection. In certain embodiments, hyaluronic acid may also be used, having the effect of promoting sustained duration in the 5 circulation. In certain embodiments, implantable drug delivery devices may be used to introduce the desired antibody molecule.
Pharmaceutical compositions of the invention can be formulated for inhalation. In these embodiments, anti-IL-lRl antibodies are formulated as a dry powder for inhalation. In preferred embodiments, anti-IL-lRl antibody inhalation solutions may also be 10 formulated with a propellant for aerosol delivery. In certain embodiments, solutions may be nebulized. Pulmonary administration and formulation methods therefore are further described in International Patent Publication No. W094/20069, incorporated by reference, which describes pulmonary delivery of chemically modified proteins.
It is also contemplated that formulations can be administered orally. Anti-IL-lRl 15 antibodies that are administered in this fashion can be formulated with or without earners customarily used in the compounding of solid dosage forms such as tablets and capsules. In certain embodiments, a capsule may be designed to release the active portion of the formulation at the point in the gastrointestinal tract when bioavailability is maximized and pre-systemic degradation is minimized. Additional agents can be included to facilitate 20 absorption of the anti-IL-lRl antibody. Diluents, flavorings, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binders may also be employed. A pharmaceutical composition of the invention is preferably provided to comprise an effective quantity of one or a plurality of anti-IL-lRl antibodies in a mixture with non-25 toxic excipients that are suitable for the manufacture of tablets. By dissolving the tablets in sterile water, or another appropriate vehicle, solutions may be prepared in unit-dose form. Suitable excipients include, but are not limited to, inert diluents, such as calcium carbonate, sodium carbonate or bicarbonate, lactose, or calcium phosphate; or binding agents, such as starch, gelatin, or acacia; or lubricating agents such as magnesium 30 stearate, stearic acid, or talc.
Additional pharmaceutical compositions will be evident to those skilled in the art, including formulations involving anti-IL-lRl antibodies in sustained- or controlled- 51 2013204136 12 Apr 2013 delivery formulations. Techniques for formulating a variety of other sustained- or controlled-delivery means, such as liposome carriers, bio-erodible microparticles or porous beads and depot injections, arc also known to those skilled in the art. See for example, International Patent Publication No. W093/15722, incorporated by reference, 5 which describes controlled release of porous polymeric microparticles for delivery of pharmaceutical compositions. Sustained-release preparations may include semipermeable polymer matrices in the form of shaped articles, e.g. films, or microcapsules. Sustained release matrices may include polyesters, hydrogels, polylactides (as disclosed in U.S. Patent No. 3,773,919 and European Patent Application Publication No. EP 058481), 10 copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al,, 1983, Biopolymers 22:547-5561. poly (2-hydroxyethyl-methacrylate) (Langer et al., 1981, ./. Biomed. Mater. Res. 15:167-277 and Langer, 1982, Chetn, Tech. 12:98-105), ethylene vinyl acetate (Langer et al., supra) or poly-D(-)-3-hydroxybutyric acid (European Patent Application Publication No. EP 133,988). Sustained release compositions may also 15 include liposomes that can be prepared by any of several methods known in the art. See e.g,, Eppstein et al, 1985, Proc. Natl. Acad. Sci. USA 82:3688-3692; European Patent Application Publication Nos. EP 036,676; EP 088,046 and EP 143,949.
Pharmaceutical compositions used for in vivo administration are typically provided as sterile preparations. Sterilization can be accomplished by filtration through 20 sterile filtration membranes. When the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution. Compositions for parenteral administration can be stored in lyophilized form or in a solution. Parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable 25 by a hypodermic injection needle.
Once the pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) that is reconstituted prior to administration. 30 The invention also provides kits for producing a single-dose administration unit.
The kits of the invention may each contain both a first container having a dried protein and a second container having an aqueous formulation. In certain embodiments of this 52 2013204136 12 Apr 2013 invention, kits containing single and multi-chambered pre-filled syringes (e.g., liquid syringes and lyosyringes) are provided.
The effective amount of an anti-IL-lRl antibody-containing pharmaceutical composition to be employed therapeutically will depend, for example, upon the 5 therapeutic context and objectives. One skilled in the aid will appreciate that the appropriate dosage levels for treatment will vary depending, in part, upon the molecule delivered, the indication for which the anti-IL-lRl antibody is being used, the route of administration, and the size (body weight, body surface or organ size) and/or condition (the age and general health) of the patient. In certain embodiments, the clinician may titer 10 the dosage and modify the route of administration to obtain the optimal therapeutic effect, A typical dosage may range Rom about 0.1 to up to about 100 mg/kg or more, depending on the factors mentioned above. In preferred embodiments, the dosage may range from 0.1 μg/kg up to about 100 mg/kg; more preferably Rom 1 μg/kg up to about 100 mg/kg; or even more preferably Rom 5 μg/kg up to about 100 mg/kg.
Dosing fi*equency will depend upon the pharmacokinetic parameters of the particular anti-IL-lRl antibody in the formulation used. Typically, a clinician administers the composition until a dosage is reached that achieves the desired effect. The composition may therefore be administered as a single dose, or as two or more doses (which may or may not contain the same amount of the desired molecule) over time, or as 20 a continuous infusion via an implantation device or catheter. Further refinement of the appropriate dosage is routinely made by those of ordinary skill in the art and is within the ambit of tasks routinely performed by them. Appropriate dosages may be ascertained through use of appropriate dose-response data.
The route of administration of the pharmaceutical composition is in accord with 25 known methods, e.g. orally, through injection by intravenous, intraperitoneal, intracerebral (intra-parenchymal), intracerebro ventricular, intramuscular, intra-ocular, intraarterial, intraportal, or intralesional routes; by sustained release systems or by implantation devices. In certain embodiments, the compositions may be administered by bolus injection or continuously by infusion, or by implantation device. 30 The composition also may be administered locally via implantation of a membrane, sponge or another appropriate material onto which tire desired molecule has 53 2013204136 12 Apr 2013 been absorbed or encapsulated. In certain embodiments, where an implantation device is used, the device may be implanted into any suitable tissue or organ, and delivery of die desired molecule may be via diffusion, timed-release bolus, or continuous administration.
It also maybe desirable to use anti-IL-lRl antibody pharmaceutical compositions 5 according to the invention ex vivo. In such instances, cells, tissues or organs that have been removed from the patient are exposed to anti-IL-lRl antibody pharmaceutical compositions after which the cells, tissues and/or organs are subsequently implanted back into the patient
In particular, anti-IL-lRl antibodies can be delivered by implanting certain cells 10 that have been genetically engineered, using methods such as those described herein, to express and secrete the polypeptide. In certain embodiments, such cells may be animal or human cells, and may be autologous, heterologous, or xenogeneic. In certain embodiments, the cells may be immortalized. In other embodiments, in order to decrease the chance of an immunological response, the cells may be encapsulated to avoid 15 infiltration of surrounding tissues. In further embodiments, the encapsulation materials are typically biocompatible, semi-permeable polymeric enclosures or membranes that allow the release of the protein product(s) but prevent the destruction of the cells by the patient’s immune system or by other detrimental factors from the surrounding tissues.
In certain embodiments, the invention further encompasses the administration of 20 an anti-IL-lRl antibody or pharmaceutical composition of the invention concurrently with one or more other drugs that are administered to the same patient, each drug being administered according to a regimen suitable for that medicament. This encompasses pre-treatment, simultaneous treatment, sequential treatment and alternating regimens, Examples of such drugs include, but are not limited to, antivirals, antibiotics, analgesics, 25 corticosteroids, antagonists of inflammatory cytokines, disease-modifying anti-rheumatic drugs (DMARDs), and non-steroidal anti-inflammatories,
In other embodiments, an anti-IL-lRl antibody or pharmaceutical composition of the invention can be administered in combination with other cytokine inhibitors, including those that antagonize, for example, RANKL, TGFp, IFNy, IL-6 or IL-8 and 30 TNF, particularly TNJFct. In combination with IL-6, an antibody of this invention can be used to treat and prevent the recurrence of seizures, including seizures induced by 54 2013204136 12 Apr 2013 GABAa receptor antagonism, seizures associated with EEG ictal episodes and motor limbic seizures occurring during status epilepticus. hi combination with IFNy inhibitor, an antibody of this invention is useful in treating idiopathic pulmonary fibrosis and cystic fibrosis. The combination of an IL-1 receptor antibody and RANECL inhibitors, e.g. a 5 RAKKL antibody is useful for preventing bone destruction in various settings including but not limited to various rheumatic disorders, osteoporosis, multiple myeloma or other malignancies that cause bone degeneration, or anti-tumor therapy aimed at preventing metastasis to bone, or bone destruction associated with prosthesis wear debris or with periodontitis. In addition, antibodies of the invention may be administered in 10 combination with IL-17 inhibitors such soluble forms of an IL-17 receptor (such as IL-17R:Fc) or an IL-17 antibody or IL-17R antibody, 11^18 binding protein, soluble forms of IL-18 receptors, and IL-18 antibodies, antibodies against IL-18 receptors or antibodies against CD30~ligand or against CD4.
The invention further encompasses methods for using an anti-ILlRl antibody or 15 pharmaceutical composition of the invention in treating the herein disclosed medical disorders in combination with a TNF inhibitor, preferably TNFR:Fc (ENBREL®) and any combination of the above described cytokines or cytokine inhibitors that are active agents in combination therapies. For example, in accordance with the present invention, combination therapy methods may be used for treating rheumatoid arthritis, stroke, 20 asthma, psoriasis, etc.
Conditions effectively treated by an anti-IL-lRl antibody or pharmaceutical composition described herein include pulmonary diseases such as asthma, chronic obstructive pulmonary disease, pulmonary alveolar proteinosis, bleomycin-induced pneumopathy and fibrosis, radiation-induced pulmonary fibrosis, cystic fibrosis, collagen 25 accumulation in the lungs, and ARDS, all of which may be treated with combinations of an-antibody to IL-1R and an IL-4 inhibitor and/or IL-13 inhibitor, e.g. IL-4R antibody that inhibits IL-13 and IL-4 activity. The disclosed antibodies and pharmaceutical compositions of the invention also are useful for treating broncho-pulmonary dysplasia (BPD); chronic obstructive pulmonary diseases (e.g. emphysema and chronic bronchitis), 30 and chronic fibrotic lung disease of preterm infants, hi addition, the compounds, compositions and combination therapies of the invention are used to treat occupational lung diseases, including asbestosis, coal worker’s pneumoconiosis, silicosis or similar 55 2013204136 12 Apr 2013 conditions associated with long-term exposure to fine particles. In other aspects of the invention, the disclosed compounds, compositions and combination therapies are used to treat bronchioliterans organizing pneumonia, pulmonary fibrosis, including idiopathic pulmonary fibrosis and radiation-induced pulmonary fibrosis; pulmonary sarcoidosis; and 5 allergies, including allergic rhinitis, contact dermatitis, atopic dermatitis and asthma.
Such combinations are useful also for treating patients suffering from various skin disorders, including but not limited to dermatitis herpetiformis (Duhring’s disease), atopic dermatitis, contact dermatitis, urticaria (including chronic idiopathic urticaria), and autoimmune blistering diseases, including pemphigus vulgaris and bullous pemphigoid. 10 Other diseases treatable with the combination of an II,-1R antibody and an IL-4 and/or IL-13 inhibitor include myesthenia gravis, sarcoidosis, including pulmonary sarcoidosis, scleroderma, reactive arthritis, hyper IgE syndrome, multiple sclerosis and idiopathic hyper eosinophil syndrome. The combination is used also for treating allergic reactions to medication and as an adjuvant to allergy immunotherapy. 15 The IL-1 receptor antibodies and pharmaceutical compositions described herein are useful for treating protozoal diseases, including malaria and schistosomiasis and to treat erythema nodosum leprosiun; bacterial or viral meningitis; tuberculosis, including pulmonary tuberculosis; and pneumonitis secondary to a bacterial or viral infection including influenza infection and infectious mononucleosis. 20 Cardiovascular disorders and injuries are treatable and/or preventable with disclosed either pharmaceutical compositions or anti-lLl-Rl antibodies alone or in combination with other cytokine inhibitors. Cardiovascular disorders heatable include aortic aneurysms; including abdominal aortic aneurysms, acute coronary syndrome, arteritis; vascular occlusion, including cerebral artery occlusion; complications of 25 coronary by-pass surgery; ischemia/reperfusion injury; heart disease, including atherosclerotic heart disease, myocarditis, including chronic autoimmune myocarditis and viral myocarditis; heart failure, including chronic heart failure, congestive heart failure, cachexia of heart failure; myocardial infarction; restenosis and/or atherosclerosis after heart surgery or after carotid artery balloon angioplastic procedures; silent myocardial 30 ischemia; left ventricular pump dysfunction, post implantation complications of left ventricular assist devices; Raynaud’s phenomena; thrombophlebitis; vasculitis, including Kawasaki’s vasculitis; veno-occlnsive disease, giant cell arteritis, Wegener’s 56 2013204136 12 Apr 2013 granulomatosis; mental confusion following cardio pnlmonary by pass surgery, and Schoenlein-Henoch purpura.
In certain embodiments, anti-IL-lRl antibodies and pharmaceutical compositions of the invention can also be used to treat chronic pain conditions, such as chronic pelvic 5 pain, including chronic prostatitis/pelvic pain syndrome, and post-herpetic pain.
Disorders of the endocrine system including juvenile onset diabetes (includes autoimmune diabetes mellitus and insulin-dependent types of diabetes) and maturity onset diabetes (includes non-insulin dependent and obesity-mediated diabetes) can also be treated with anti-IL-lRl antibodies or pharmaceutical compositions of the invention. 4 10 Such treatment includes secondary conditions associated with diabetes, such as diabetic retinopathy, kidney transplant rejection in diabetic patients, obesity-mediated insulin resistance, and renal failure, which itself may be associated with proteinurea and hypertension. Other endocrine disorders also are treatable with these compounds and include polycystic ovarian disease, X-linked adrenoleukodystrophy, hypothyroidism and 15 thyroiditis, including Hashimoto’s thyroiditis (i.e., autoimmune thyroiditis), thyroid cell dysfunction, including euthyroid sick syndrome.
Conditions of the gastrointestinal system are treatable or preventable with anti-IL-1R1 antibodies or pharmaceutical compositions of the invention, alone or in combination with oilier therapeutics. These conditions include coeliac disease, Crohn’s disease; 20 ulcerative colitis; idiopathic gastroparesis; pancreatitis, including chronic pancreatitis; acute pancreatitis, inflammatory bowel disease and ulcers, including gastric and duodenal ulcers.
Disorders of the genitourinary system are also treatable or preventable with the anti-IL-lRl antibodies or pharmaceutical compositions described herein. Such disorders 25 include glomerulonephritis, including autoimmune glomerulonephritis, glomerulonephritis due to exposure to toxins or glomerulonephritis secondary to infections with haemolytic streptococci or other infectious agents. Also treatable with the compounds, compositions and combination therapies of the invention are uremic syndrome and its clinical complications (for example, renal failure, anemia, and 30 hypertrophic cardiomyopathy), including uremic syndrome associated with exposure to environmental toxins, drugs or other causes. Complications that arise from inflammation of the gallbladder wall that leads to alteration in absorptive function are treatable or 57 2013204136 12 Apr 2013 preventable with the antibodies of this invention. Included in such complications are cholelithiasis (gallstones) and choliedocholithiasis (bile duct stones) and the recurrence of cholelithiasis and choliedocholithiasis. Further conditions treatable with the compounds, compositions and combination therapies of the invention are complications of 5 hemodialysis; prostate conditions, including benign prostatic hypertrophy, nonbacterial prostatitis and chronic prostatitis; and complications of hemodialysis.
Also provided herein are methods for using anti-IL-lRl antibodies of the invention, compositions, and combination therapies to treat various hematologic and oncologic disorders. For example, anti-IL-lRl antibodies, alone or in combination with 10 other cytokine inhibitors or other active agents as described above, can be used to treat various forms of cancer, including acute myelogenous leukemia, chronic myelogenous leukemia leukemia, Epstein-Barr virus-positive nasopharyngeal carcinoma, glioma, colon, stomach, prostate, renal cell, cervical and ovarian cancers, lung cancer (SCLC and NSCLC), including cancer-associated ' cachexia, fatigue, asthenia, paraneoplastic 15 syndrome of cachexia and hypercalcemia. Solid tumors, including sarcoma, osteosarcoma, and carcinoma, such as adenocarcinoma (for example, breast cancer) and squamous cell carcinoma are also treatable. Additional treatable cancers include esophogeal cancer, gastric cancer, gall bladder carcinoma, leukemia, including acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, chronic or 20 acute lymphoblastic leukemia and hairy cell leukemia. Other· malignancies with invasive metastatic potential, including multiple myeloma, can be treated with the subject compounds, compositions and combination therapies.
In addition, the disclosed anti-IL-lRl antibodies can be used to treat anemias and hematologic disorders, including chronic idiopathic neutropenia, anemia of chronic 25 disease, aplastic anemia, including FanconPs aplastic anemia; idiopathic thrombocytopenic purpura (ITP); thrombotic thrombocytopenic purpura, myelodysplastic syndromes (including refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation); myelofibrosis/myeloid metaplasia; and sickle cell vasocclusive crisis. 30 Various lymphoproliferative disorders also are treatable with anti-IL-lRl antibodies of the invention, including autoimmune lymphoproliferative syndrome (ALPS), chronic lymphoblastic leukemia, hairy cell leukemia, chronic lymphatic 58 2013204136 12 Apr 2013 leukemia, peripheral T-cell lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, follicular lymphoma, Burkitt’s lymphoma, Epstcin-Barr virus-positive T cell lymphoma, histiocytic lymphoma, Hodgkin’s disease, diffuse aggressive lymphoma, acute lymphatic leukemias, T gamma lymphopro lifer ative disease, cutaneous B cell 5 lymphoma, cutaneous T cell lymphoma (i.e., mycosis fungoides) and Sezary syndrome.
Hereditary conditions such as Gaucher’s disease, Huntington’s disease, linear IgA disease, and muscular dystrophy are treatable with the antibodies of this invention.
Other conditions treatable or preventable by the disclosed IL-1 receptor antibodies or pharmaceutical compositions include those resulting from injuries to the head or spinal 10 cord including subdural hematoma due to trauma to the head. In connection with this therapy, the compositions and combinations described are suitable for preventing cranial neurologic damage and preventing and treating cervicogenic headache. The compositions and combinations described are further suitable for treating neurological side effects associated with brain irradiation. 15 Atiti-iL-lRl antibodies and pharmaceutical composition of the invention are also useful for treating conditions of the liver such as hepatitis, including acute alcoholic hepatitis, acute drug-induced or viral hepatitis, hepatitis A, B and C, sclerosing cholangitis, hepatic sinusoid epithelium, and inflammation of the liver due to unknown causes. 20 Disorders that involve hearing loss and that are associated with abnormal IL-1 expression are treatable with the anti-IL-lRl antibodies or pharmaceutical compositions of the invention. Such disorders include cochlear nerve-associated hearing loss that is thought to result from an autoimmune process, i.e., autoimmune hearing loss. Also treatable or preventable with the anti-IL-lRl antibodies or pharmaceutical compositions 25 of the invention is Meniere’s syndrome and cholesteatoma, a middle ear disorder often associated with hearing loss.
Non-arihritic disorders of the bones and joints and also treatable with the antibodies described herein. This encompasses osteoclast disorders that lead to bone loss, such as but not limited to osteoporosis, including post-menopausal osteoporosis, 30 osteoarthritis, periodontitis resulting in tooth loosening or loss, and prosthesis loosening after joint replacement (generally associated with an inflammatory response to wear 59 2013204136 12 Apr 2013 debris). This latter condition also is called “orthopedic implant osteolysis.” Another condition treatable with the compounds, compositions and combination therapies of the invention is temporal mandibular joint dysfunction (TMJ).
The anti-IL-lRl antibodies or pharmaceutical compositions of the invention can 5 also be used to treat rheumatic disorders including adult and juvenile rheumatoid arthritis; scleroderma; systemic lupus erythematosus; gout; osteoarthritis; polymyalgia rheiunatica; seronegative spondylarthropathics, including ankylosing spondylitis, and Reiter’s disease, psoriatic arthritis and chronic Lyme arthritis. The antibodies of this invention are also useful for treating nflammation of the voluntary muscle and other muscles, including 10 dermatomyositis, inclusion body myositis, polymyositis, and lymphangioleimyomatosis.
Another use for the antibodies and pharmaceutical compositions of the invention is the treatment and/or prevention of primary amyloidosis and the secondary amyloidosis that is characteristic of various condition including Alzheimer’s disease, secondary reactive amyloidosis; Down’s syndrome; and dialysis-associated amyloidosis. Also 15 treatable with the antibodies or pharmaceutical compositions of the invention are inherited periodic fever syndromes, including familial Mediterranean fever, hyperimmunoglobulin D and periodic fever syndrome and TNF-receptor associated periodic syndromes (TRAPS).
In other embodiments, the antibodies or pharmaceutical compositions of the 20 invention can be used to treat disorders involving the skin or mucous membranes. Such disorders include acantholytic diseases, including Darier’s disease, keratosis follicularis and pemphigus vulgaris. Additional skin disorders that can be treated using antibodies of the invention include acne, acne rosacea, alopecia areata, aphthous stomatitis, bullous pemphigoid, bums, eczema, erythema, including erythema multiforme and erythema 25 nmltifomie bullosum (Stevens-Johnson syndrome), inflammatory skin disease, lichen planus, linear IgA bullous disease (chronic bullous dermatosis of childhood), loss of skin elasticity, mucosal surface ulcers, including gastric ulcers, neutrophilic dermatitis (Sweet’s syndrome), dermatomyositis, pityriasis rubra pilaris, psoriasis, pyoderma gangrenosum, multicentric reticulohistiocytosis, and toxic epidermal necrolysis. Other 30 skin related conditions treatable by the therapies and combination therapies of the present invention include dermatitis herpetiformis. 60 2013204136 12 Apr 2013
Additional disorders that can be treated with the antibodies or pharmaceutical compositions of the invention include graft -versus-host disease, and complications resulting from solid organ transplantation, such as heart, liver, skin, kidney, lung (lung transplant airway obliteration) or other transplants, including bone marrow transplants. 5 Ocular disorders also are treatable or preventable with the disclosed anti-IL-lRl antibodies or pharmaceutical compositions, including rhegmatogenous retinal detachment, and inflammatory eye disease, including inflammatory eye disease associated with smoking and macular degeneration.
Antibodies or pharmaceutical compositions of the invention, as described herein, 10 are usefiil for treating disorders that affect the female reproductive system. Examples include, but are not limited to, multiple implant failurc/infcrtility; fetal loss syndrome or IV embryo loss (spontaneous abortion); preeclamptic pregnancies or eclampsia; endometriosis, chronic cervicitis, and pre-term labor.
In addition, the antibodies or pharmaceutical compositions of the invention are 15 useful for heating and/or preventing sciatica, symptoms of aging, severe drug reactions (for example, 11-2 toxicity or bleomycin-induced pneumopathy and fibrosis), or to suppress the inflammatory response prior, during or after the transfusion of allogeneic red blood cells in cardiac or other surgery, or in treating a traumatic injury to a limb or joint, such as traumatic knee injury. Various other medical disorders treatable with the 20 disclosed anti-IL-lRl antibodies or pharmaceutical compositions include; multiple sclerosis; Behcet’s syndrome; Sjogren’s syndrome; autoimmune hemolytic anemia; beta thalassemia; amyotrophic lateral sclerosis (Lou Gehrig’s Disease); Parkinson’s disease; and tenosynovitis of unknown cause, as well as various autoimmune disorders or diseases associated with hereditary deficiencies, including x-linked mental retardation. 25 Furthermore, the anti-IL-lRl antibodies or pharmaceutical compositions of the invention are useful for treating central nervous system (CNS) injuries, including the effects of neurotoxic neurotransmitters discharged during excitation of inflammation in the central nervous system and to inhibit or prevent the development of glial scars at sites of central nervous system injury. In connection with epilepsy and the treatment of 30 seizures, reducing the severity and number of reciuiing seizures, and reducing the severity of the deleterious effects of seizures, reducing neuronal loss, neuronal degeneration, and gliosis associated with seizures. 61 2013204136 12 Apr 2013
Additional uses for the antibodies or pharmaceutical compositions of the invention include, but are limited to, treating critical illness polyneuropathy and myopathy (CffNM) acute polyneuropathy; anorexia nervosa; Bell’s palsy; chronic fatigue syndrome; transmissible dementia, including Creutzfeld-Jacob disease; demyelinating 5 neuropathy; Guillain-Barre syndrome; vertebral disc disease; Gulf war syndrome; chronic inflammatory demyelinating polyneuropathy, myasthenia gravis; silent cerebral ischemia; sleep disorders, including narcolepsy and sleep apnea; chronic neuronal degeneration; and stroke, including cerebral ischemic diseases. Still additional uses for the antibodies of the invention are anorexia and/or anorexic conditions, peritonitis, endotoxemia and septic 10 shock, granuloma formation, heat stroke, Churg-Strauss syndrome, chronic inflammation following acute infections such as tuberculosis and leprosy, systemic sclerosis and hypertrophic scarring.
In other embodiments, avidin fusion proteins comprising an amino acid sequence of one of the IL-1R1 antibodies of the invention can be constructed for various purposes. 15 Avidin fusion proteins can be generated, for example, using a mammalian expression vector containing cDNA sequence encoding recombinant chicken avidin adjacent to a multiple cloning site for insertion of a specific target gene fusion partner. The vector can include an avidin sequence with its endogenous signal sequence to enable secretion of discrete fusion gene partners that do not naturally contain signal sequences. The fiision 20 protein expressed by the vector has an avidin protein tag at the N-terminal portion of the fiision partner. The fiision strategy as described herein has the capability of secreting proteins that are normally expressed intracellularly, such as signal transduction genes or nuclear hormone receptors.
Alternatively, a vector can be used that encodes avidin without its endogenous 25 sigual sequence, which will result in C-terminal tagging of fusion protein partners. A C-terminal avidin fusion also allows for protein secretion based on the endogenous signal sequence of the fiision partner. Such a strategy can be applied to allow for correct protein processing and folding or to determine validity of a proposed signal sequence. Additionally, the vector can comprise a short nucleotide sequence encoding an amino 30 acid sequence, which can act as a specific enzyme-cleavable substrate, between the avidin and fusion partner sequences. Such enzyme-cleavable sequences allow for separation of the fiision partner from the avidin for purification or protein release purposes. 62 2013204136 12 Apr 2013
Avidin fusion proteins of the invention can be used, for example, in antibody screening, functional characterization (determination of an antibody’s utility as an agonist or antagonist, neutralizing agent, etc.), epitope mapping, or immunization strategies. Avidin fusions of a target protein can also be utilized in pharmokinetic, efficacy or other 5 standard assay formats designed to test preclhucal samples or clinical patient samples for the presence of the therapeutic antibody in blood, urine, or other tissue samples. Avidin fusion protein partners can be prepared as hill-length or truncated sequences, specific isolated structural domains, or as chimeric sequences with other homologs of the fusion partner from other species. 10 Avidin fusion proteins can be expressed using any standard means of introducing genes into cells, as described herein and known in the art. The proteins can be expressed in, for example, 293 or CHO cells by transfecting the cells with an avidin fusion construct in a solution of lipids, such as in Lipofectamine (Invitrogen, Carlsbad, CA).
Conditioned media and/or cell lysates horn cells expressing the fusion proteins 15 can be collected and applied to an assay substrate, such as biotin-coated polystyrene beads or biotin-coated ELISA plates. Collecting the conditioned media and/or cell lysate can be conducted at a time point that allows for optimum expression of the fusion protein. The time point can be determined experimentally by those skilled in the art, but is usually about 48 horns post-transfection. Fusion proteins can also be analyzed at the cell 20 membrane or intracellularly for expression and functionality in binding known ligands, receptors, or antibodies.
Avidin fusion proteins of the invention can be analyzed by any known or previously characterized method that utilizes biotin-avidin interactions. Such methods include, but are not limited to, flow cytometry and fluorescent imaging/imicroscopy. For 25 example, avidin fusions expressed in media or cell lysates can be applied to biotin-coated beads and stained with a fluorescently tagged anti-avidin antibody to indicate expression level. Also, fluorescent antibodies can be applied that recognize the specific fusion protein partner in a multicolorimetric assay format. Additionally, unlabeled antibodies specific for the fusion protein partner can be applied simultaneously with fluorescently 30 tagged antibodies in a competition assay.
In certain embodiments, the invention provides methods for mapping epitopes using avidin fusion proteins. An example of an epitope mapping method of the invention 63 2013204136 12 Apr 2013 is provided below with respect to mapping epitopes for anti-IL-lRl antibodies. However, one of skill in the art will recognize that such methods can be readily applied to mapping epitopes for any antibody and is not limited to anti-IL-lRl antibodies. For example, cDNA encoding chicken avidin (with endogenous signal sequence) can be joined with the 5 5’ end of cDNAs encoding a protein of interest (i.e. a protein that is recognized by antibodies for which determining an epitope is desired) Jxised to a FLAG-tag sequence at the 3’ end. The FLAG-tagged fusion genes can be assembled in an expression vector using conventional molecular techniques. A panel of mutant avidin-FLAG tagged proteins in which certain amino acids have been substituted (e.g., with corresponding 10 amino acid residues from another animal species) can be generated using conventional techniques. The mutant and wild type proteins can be expressed in host cells and binding of the wild-type or mutant proteins with an antibody of interest can be detected using, for example, Western blot analysis or bead-based binding assays as described herein. Thus, an epitope can be defined by determining which substitutions in the mutant proteins 15 deshoy binding to the antibody of interest.
EXAMPLES
The following examples, including the experiments conducted and results achieved are provided for illustrative purposes only and are not to be construed as 20 limiting the invention.
Example 1
Production of Human Monoclonal Antibodies Against Interleukin-1 Receptor Type I (TL-1R11
Transgenic HuMab Mice 25 Fully human monoclonal antibodies to JL-l receptor type I (IL-1R1) were prepared using the HCo7 strain of transgenic mice, which expresses human antibody genes. In each of these mouse strains, the endogenous mouse kappa light chain gene has been liomozygously disrupted as described in Chen et at (1993, EMBO J. 12:811-820), and the endogenous mouse heavy chain gene has been homozygously disrupted as 30 described in Example 1 of International Patent Application Publication No. WO 01/09187 (incorporated by reference). Each of these mouse strains carries a human kappa light 64 2013204136 12 Apr 2013 chain, transgene, K.Co5, as described in Fishwild et al. (1996, Nature Biotechnology 14:845-851), The HCo7 strain carries the HCo7 human heavy chain transgene as described in US, Patent Nos, 5,545,806; 5,625,825; and 5,545,807 (incorporated by reference), The HCo7 strain is referred to herein as HuMab mice. 5
HuMab Immunizations
To generate folly human monoclonal antibodies to IL-1R1, HuMab mice were immunized with purified recombinant IL-1R1 derived from insect or mammalian cells (for example, CHO cells) as antigen, General immunization schemes for HuMab mice 10 are described in Lonberg et al (1994, Nature 368:856-859; Fishwild et al, supra; and International Patent Application Publication No. WO 98/24884, the teachings of each of which are incorporated by reference), Mice were 6-16 weeks of age upon the first infusion of antigen, A purified recombinant preparation (25-50 //g) of IL-1R1 antigen (e.g., purified from transfected insect or mammalian cells expressing IL-1R1) was used to 15 immunize the HuMab mice intraperitoneally (IP) or subcutaneously (Sc).
Immunizations of HuMab transgenic mice were achieved using antigen in complete Freund’s adjuvant and two injections, followed by 2-4 weeks IP immunization (up to a total of 11 immunizations) with the antigen in incomplete Freund’s adjuvant. Several dozen mice were immunized for each antigen, A total of 149 mice of the HCo7 20 strain were immunized with IL-1R1. The immune response was monitored by retroorbital bleeds.
To select HuMab mice producing antibodies that bound IL-1R1, sera from immunized mice were tested by ELISA as described by Fishwild et al, supra. Briefly, microtiter plates were coated with purified recombinant IL-1R1 from insect or 25 mammalian cells at 1-2 pg/mL in PBS and 50 //L/well incubated at 4°C overnight,· then blocked with 200 //L/well of 5% chicken serum in PBS/Tween (0.05%), Dilutions of plasma from IL-1R1-immunized mice were added to each well and incubated for 1-2 hours at ambient temperature. The plates were washed with PBS/Tween and then incubated with a goat-anti-human IgG Fc-specific polyclonal reagent conjugated to 30 horseradish peroxidase (HRP) for 1 hour- at room temperature. Plates were washed with PBS/Tween and incubated with a goat anti-human IgG Fc-specific polyclonal reagent 65 2013204136 12 Apr 2013 conjugated to horseradish peroxidase (HRP) for 1 hour at room temperature. After washing, the plates were developed with ABTS substrate (Sigma Chemical Co., St. Louis, MO, Catalog No. A-1888, 0.22 mg/mL) and analyzed spectrophotometrically at OD of 415-495. Mice with sufficient titers of anti-IL-lRl human immunoglobulin were used to 5 produce monoclonal antibodies as described below.
Generation of hvbridomas producing human monoclonal antibodies to IL-1R1
Mice were prepared for monoclonal antibody production by boosting with antigen intravenously 2 days before sacrifice, and spleens were removed thereafter. The mouse 10 splenocytes were isolated from the HuMab mice and fused with PEG to a mouse myeloma cell line using standard protocols. Typically, 20-30 fusions for each antigen were performed
Briefly, single cell suspensions of splenic lymphocytes from immunized mice were fused to one-fourth the number of P3X63-Ag8.653 nonsecreting mouse myeloma 15 cells (A.T.C.C., Accession No. CRL 1580) or SP2/0 nonsecreting mouse myeloma cells (A.T.C.C., CRL 1581) with 50% PEG (Sigma). Cells were plated at approximately lxl05/well in flat bottom microtiter plates, followed by about a two week incubation in selective medium containing 10% fetal bovine serum, 10% P388D1- (A.T.C.C., Accession No, CRL TIB-63) conditioned medium, 3-5% origen (IGEN) in DMEM 20 (Mediatech, Catalog No. CRL 10013, with liigli glucose, L-glutamine and sodium pyruvate) plus 5 mM HEPES, 0.055 mM 2-mercaptoethanol, 50 mg/mL gentamycin and lx HAT (Sigma, Catalog No. CRL P-7185). After 1-2 weeks, cells were cultured in medium in which the HAT was replaced with HT.
The resulting hybridomas were screened for the production of antigen-specific -25. antibodies. Individual wells were screened by ELISA (described above) for human anti-IL-1R1 monoclonal IgG antibodies. Once extensive hybridoma growth occurred, medium was monitored usually after 10-14 days. Antibody-secreting hybridomas were replated, screened again and, if still positive for human IgG, anti-IL-lRl monoclonal antibodies were subcloned at least twice by limiting dilution. The stable subclones were 30 then cultured in vitro to generate small amounts of antibody in tissue culture medium for characterization. 66 2013204136 12 Apr 2013
Selection of Human Monoclonal Antibodies Binding tn njpl
An ELISA assay as described above was used to screen for hybridomas that showed positive reactivity with IL-1R1 immunogen. Hybridomas secreting a monoclonal 5 antibody that bound with high avidity to IL-1R1 were subcloned and further characterized. One clone from each hybridoma, which retained the reactivity of parent cells (as determined by ELISA), was chosen for making a 5-10 vial cell bank stored in liquid nitrogen.
An isotype-specific ELISA was performed to determine the isotype of the 10 monoclonal antibodies produced as disclosed herein. In these experiments, microtiter plate wells were coated with 50 //L/well of a solution of 1 /rg/mT, 0f mouse anti-human kappa light chain in PBS and incubated at 4°C overnight. After blocking with 5% chicken serum, the plates were reacted with supernatant from each tested monoclonal antibody and a purified isotype control. Plates were incubated at ambient temperature for 15 1-2 horns. The wells were then reacted with either human IgGl, IgG2 or IgG4-specific horseradish peroxidase-conjugated goat anti-human polyclonal antisera and plates were developed and analyzed as described below.
Monoclonal antibodies purified from hybridoma supernatants that showed significant binding to IL-1R1 as detected by ELISA were further tested for biological 20 activity using in vitro binding assays and human chondrocyte and whole blood cell-based assays. The antibodies that displayed the best activity were designated 15C4, 26F5, 27E2, 24E12, and 10H7. The antibodies were subjected to a preliminary epitope sorting experiment. ELISA plates were coated with human sIL-lRl (1+2+3 domain), truncated human sIL-lRl (1+2 domain), rat sIL-lRl, human sIL-lR type Π, and ovalbumin 25 (negative control). Antibody binding was detected with a horseradish peroxidase-conjugated anti-Human Fc antibody (Pierce Chemical Co., Rockford, IL). The results are summarized in Table 2. A check mark (V) in Table 2 represents a positive result for binding; "X” represents a negative result. Antibodies 15C4, 26F5, 27F2 and 24E12 bind only the IL-1R1 protein that has all three extracellular domains, indicating that the 30 epitopes for each fall within the third domain. Antibody 10H7 binds both the fiill-length extracellular domain IL-1R1 and also a truncated protein that has only domains 1 and 2, 67 2013204136 12 Apr 2013 demonstrating that the epitope for this antibody lies within either domain 1 or 2, None of the antibodies tested has cross-reactivity with human type Π receptor or rat IL-1R1.
Table 2
OA (Negative Control) Hu slL-1R1 (1+2+3 Domain) Hu SIL-1R1 (1+2 Domain) Hu SIL-1RII (1+2+3 Domain) RatslL-1R1 (1+2+3 Domain) Antibody 1SC4 X V X X X 26FS X V X X X 27F2 X V X X X 24E12 X V X X X 10H7 X V V X X 5 Example 2
In Vitro inhibition of IL-1 Receptor Type I Complex Formation by anti-IL-lRl
Antibodies
The ability of the antibodies to inhibit the extracellular binding events required for IL-1 signaling was assessed with recombinant proteins in vitro in an assay in which IL-1 10 binding to IL-1R results in formation of a high affinity binding site for IL-lRAcP, The binding of IL-lRAcP to IL-l-bound IL-1R (refered to as “complex formation”) is measured as follows, Recombinant proteins were incubated in binding assays in microtiter plates in the absence (control) or presence of antibodies. IC5o values were derived from comparisons of control values to values obtained in the presence of antibody 15 at concentrations between 10 fM and 1 μΜ, In brief, the assay was conducted as follows. Biotinylated IL-1R1 and streptavidin-coated beads (Dynal, Dynabeads M-28) were dispensed in microtiter plates. Antibody was then added to the appropriate wells in a serial dilution covering a broad range of concentrations. IL-1/3 or IL-lct was added at a concentration of 1 nM, and ILlRAcP labeled with ruthenium (prepared with NHS-Tag 20 (IGEN) according to IGEN protocols) was added at a final concentration of 5 nM. After incubation for 1 hour at room temperature, the binding reaction was analyzed with either an ORIGEN™ 1.5 or M8 instrument (IGEN International Inc.). IL-lRAcP binding to IL- 68 2013204136 12 Apr 2013 1 bound IL-1R1 was determined by detecting the electrochemiluninescence signal associated with the IL-1R1 bound beads. The reduction of signal resulting from antibody competition of either IL-1 or JL-lRAcP binding was calculated as percentage of ECL signal for maximum binding (no competition). 5 The inhibition response curve for each antibody in these binding assays was established and ICsos were derived using PRISM™ software. The results for inhibition of EL-Ιβ induced binding events are depicted by the graph in Figure 12. The IC50 values for inhibition of complex formation are shown in Table 3 below. Antibodies 15C4, 26F5, 27F2, and 24E12 strongly inhibit complex formation. These antibodies are all IL-1R1 10 third domain binders, as described above. Antibody 10H7 belongs to a class of antibodies that binds to a construct of the IL-1 R lacking the third domain. 10H7 is a less potent inhibitor of IL-1 driven binding of IL-lRAcP than the third domain binders. Complex formation inhibition by the antibodies of the invention was compared with inhibition by IL-lra. The third domain binders demonstrated similar or slightly greater ability to 15 inhibit complex formation by comparison with IL-lra.
Figure 13 depicts the ability of antibody 15C4 to inhibit IL-lRl/IL-lo/RAcP complex formation, The IC50 for IL-lRl/IL-la/RAcP complex formation was 43 pM.
Table 3
Human anti-IL-lRl 15C4 26F5 27F2 24E12 10H7 rll-lra IC50 96 pM 160pM 333 pM 348 pM 5.3 nM 555 pM 95% Confidence Limits 71 pM to 129 pM 118pMto2l9 pM 214pMto517 pM 223 pM to 542 pM 3.6 nM to 7.5 nM 414pMto 743 pM 20
Example 3
Anti-IL-lRl Antibodies Inhibit Binding of IL-lff and IL-lra to Receptor
The ability of anti-IL-lRl antibodies to inhibit binding of either IL-Ιβ or IL-lra to 1L-1R1 was assessed in an assay with recombinant proteins. The reaction mixture 25 contained 0.1 mg/mL Dynabeads M-280 Streptavidin (Dynal) and 1 nM biotinylated IL-1R1. Antibodies were added at concentrations from 320 nM to 0.3 nM Addition of 69 2013204136 12 Apr 2013 ruthenium-tagged IL-Ιβ (5 nM) or IL-lra (1 nM) initiated binding that proceeded for 1 hour at room temperature. The reaction mixtures were measured as above using an ORIGBN 1.5 or M8 instrument (IGEN International Inc.). Competition was calculated as the percentage of ECL signal for maximum binding (no competition). Antibodies 5 15C4, 26F5, and 27F2, the most poterit antibodies, block ligand (IL-1/3) binding to receptor, but do not significantly interfere with the binding of Ilmira compared with IgG control. In contrast, antibody 24E12 binds receptor but does not block IL-1/3 or IL-lra binding to receptor. Thus, antibody 24E12 represents a unique class of third domain binders distinct from the class represented by 15C4, 26F5, and 27F2. Antibody 10H7 10 inhibits both IL-1/3 and IL-lra from binding to the receptor. The results are summarized in Figure 14.
Example 4
Chondrocyte and Human Whole Blood Assays 15 Primary human chondrocytes (Cell Applications Inc., San Diego, CA) were seeded into 96-well plates at a density of 10,000 cells/well in DMEM media containing 1% FBS and 1% Pen Strep (GIBCO). Cells were allowed to recover overnight before addition of anti-ILl-RI antibodies at concentrations ranging from 10 nM to 0.1 pM for 20 minutes. IL-1/3 was added to a concentration of 1 pM (~ECSo) and culture supernatants 20 were harvested after 16 horn's incubation at 37°C. IL-6 levels in the supernatant were measured using an ELISA (Pierce-Endogen, Rockford, IL, Cat# EH2IL-65) according to the manufacturer’s instructions. The inhibition response curve for each antibody of the invention in the cell-based assays was established and IC50 values were derived using PRISM™ software. Antibodies 15C4, 26F5, and 27F2 are potent inhibitors of IL-1 25 signaling compared with IL-lra (Figure 15A). Antibodies 24E12 and 10H7 are markedly less potent than 15C4 and 27F2 (Figure 15B), Tire ICjo values for inhibition of TT.-1 p induced IL-6 production human chondrocytes are shown in Tables 4A and 4B (corresponding to Figure 15A and 15B respectively).
Anti-IL-lRl monoclonal antibodies 15C4, 26F5, and 27F2 were pre-incubated 30 40-60 minutes with human whole blood collected from normal volunteers in sodium heparin vacutainers. The assays were run as follows: 100 pL fleshly isolated blood was 70 2013204136 12 Apr 2013
Tables Donor Analysis Parameters 15C4 26F5 27F2 IL-lra 1047 IC50 126 pM 410 pM 249 pM 241 pM 95% Confidence Limits 47pMto339pM 213pMto790pM 88 pM to 703 pM 124pMto471pM 1319 IC50 lllpM 174 pM 579 pM 381 pM 95% Confidence Limits 59 pM to 208 pM 60pMto501pM 249 pM to 1.3 nM 167pMto875pM . Composite IC50 126pM 372 pM 387 pM 264 pM (Pooled Data) 95% Confidence Limits 62 pM to 255 pM 187 pM to 739 pM 202 pM to 748 pM 134 pM to 517 pM aliquoted wells of a 96-well plate. 50 μΐ, of antibody was added in RPMI medium containing 10% human AB serum. 1\Λβ was then added at a concentration of 30 pM (EC50). Culture supernatants were harvested after 18 hours, and IL-6 levels in the supernatant were measured using an ELISA. As a control, IL-lra was pre-incubated 40-5 60 minutes with whole blood and EL-6 production was measured as above. The tluee anti-IL-lRl antibodies blocked IL-1 activity with potency comparable to that of IL-lra (Figure 16). The XC50 values for inhibition of IL-1-induced IL-6 production in human whole blood are shown in Table 5.
Table 4A
Human anti-IL-lRl Ant ibodics 15C4 271¾ 26F5 rlL-lra IC50 16 pM 32 pM 26 pM 32 pM 95% Confidence Limits 15 pMto 18 pM 21 pM to 49 pM 19 pM to 36 pM 22pMto46pM 10
Table 4B
Human anti-IL-lRl Antibodies 15C4 271¾ 10H7 24E12 rIL-ra IC50 7 pM 28 pM 7.5 pM NA 20 pM 95% Confidence Limits 5.8 pM to 7,9 pM 22 pM to 35 pM 5.6nMto 10 nM NA 17 pM to 23 _PM 15
Example 5
Mutagenesis and Epitope Mapping 71 2013204136 12 Apr 2013
Site directed mutagenesis (Altered Sites® In Vitro Mutagenesis System, Promega, Madison WT) of IL-1R1 was used to prepare a panel of mutant proteins (“muteins”) in which rat amino acid residues were substituted for the corresponding human sequence. Fifteen different mutated plasmids were constructed (see numbered bars in Figure 17). 5 Plasmids encoding these substituted proteins and the parental IL-1R1 were transiently transfected in CHO cells. Mock transfectants were generated as negative controls. Conditioned medium (CM) from these cells was concentrated ~20-fold using Centriprep 10 concentration columns (Amicon). Expression of the muteins was assessed by SDS-PAGE and Western blotting. Thirteen mutant proteins were expressed at levels that 10 allowed evaluation of antibody binding. The proteins were loaded onto a gel, electrophoresed and transferred to membranes. The membranes were blocked in 1% milk in PBS, 0.1% Tween-20 and then incubated for 1 hour at room temperature with anti-IL» 1R antibodies 15C4, 27F2, or 24E12 at 0.5 ug/mL in PBS, 0.1% Tween-20. After washing, membranes were incubated with goat anti-human IgG-Fc-HRP. Signal was 15 detected using chemiluminescence (ECL) substrate (Pierce Chemical Co., Rockford, IL). Human specific sequences critical for antibody binding were identified as those that when substituted with rat sequences reduced or eliminated ECL signal. 15C4 recognition of mutants 1, 2, 4 and 10 was impaired when compared to 24E12 (Figure 18, top panel). Similarly, 27F2 binding to mutants 1, 2 and 4 was impaired (Figure 18, middle panel). 20 24E12 had no significant binding to mutants 12,13,14 and 15 (Figure 18, bottom panel).
Isolation and characterization of human anti-IL-lRl antibodies has identified three distinct classes of competitive antibodies (Figure 19). The strongest inhibitors of IL-1 biological activity, as demonstrated by cell-based bioassays, are those antibodies that bind the third domain of IL-1R1 and prevent IL-1-β association. Epitope mapping 25 experiments using a panel of third domain mutant proteins has demonstrated that this class of antibodies, which includes 15C4, 27F2 and 26F5, shares an overlapping but not identical, confonnational epitope. Figures 20 and 21 illustrate the position of 15C4 epitopes on the third domain of the IL-1 receptor, in a ribbon diagram of IL-lra bound IL~ 1 receptor (Schreuder et al, 1997, Nature 386:194-200). The IL-1 receptor residues that 30 define binding of the most potent class of antibodies are illustrated in gray. These antibodies have demonstrated superior potency, and thus these epitopes define binding sites for antibodies of a superior class. The 15C4 and 27F2 binding sites are overlapping 72 2013204136 12 Apr 2013 but not identical, as determined, by the mutational analysis of the 15 different sites within IL-1R1 desciibed above, The sites are depicted in Figure 17 as numbered bars above the protein sequence, Critical sites of interaction appear to be within mutations at sites 1 (LSDIA; SEQ ID NO: 41), 2 (VIDE; SEQ ID NO: 42), 4 (YSV) and 10 (TCFA; SEQ ID 5 NO: 43). 15C4 and 27F2 binding sites are comprised within sites 1 and 2, since substitution of the rat residues for the human residues in either site abolishes binding, 27F2 differs horn 15C4 in that changes in site 4 completely abolish its binding, whereas 15C4 binding is reduced but not completely eliminated. Mutation 10 also reduces 15C4 binding, but 27F2 has no obvious interaction with this site. Examination of the crystal 10 structure reveals that these residues define a face of the third domain that is oriented towards the space occupied by bound ligand (Figures 20 and 21) (Vigers et al, 1997, Nature 386:190-1941.
Tlie second class of antibodies identified, represented by 10H7, does not require the third domain for binding, and unlike the preferred class, inhibits IL-lra binding, This 15 class is active in bioassays, but is less potent than the preferred class.
In contrast to the strong inhibition of IH bioassays with the preferred class of antibodies, 24E12 is an ineffective inhibitor in bioassays. Antibody 24E12 inhibits the binding of IL-lRAcP with IL-l-bound IL-1R. The epitope for this class of antibodies, defined by mutants 12, 13, 14 and 15, is proximal to the transmembrane domain of IL-20 1R1, and is in a region not directly involved in either IL-1 or IL-lra binding (Figure 22).
Example 6
Cloning the anti-IL-lRl Antibody Heavy and Light Chains
Cloning of the anti-IL-lRl 15C4 MAb Light Chain 25 The light chains for three hybridomas expressing alL-lRl binding monoclonal antibodies, 15C4,27F2, and 26F5 were cloned into the mammalian cell expression vector pDSRo.19 (see International Application, Publication No. WO 90/14363, which is herein incorporated by reference for any purpose). The construction of the plasmid encoding the 15C4 kappa light chain is explicitly described herein; cloning of the other light chain 30 species was performed using similar procedures. The ocIL-lRl kappa light chain variable 73 2013204136 12 Apr 2013 region was obtained using polymerase chain reaction (PCR) amplification methods from first strand cDNA prepared from ocIL-lRl hybridoma 15C4 total RNA prepared using TRIzol® reagent (Invitrogen). First strand cDNA was synthesized using a random primer with an extension adapter (5’- GGC CGG ATA GGC CTC CAN NNN NNT -3’; SEQ 5 ID NO: 44) and 5’ RACE (rapid amplification of cDNA ends) was performed using the GeneRacer™ Kit (Invitrogen). For the complete light chain, the forward primer was the GeneRacer™ nested primer (5’ GGA CAC TGA CAT GGA CTG AAG GAG TA -3’; SEQ ID NO: 45) and the reverse primer was 5 GGG GTC AGG CTG GAA CTG AGG -3’ (SEQ ID NO: 46). The RACE products were cloned into pCR4-TOPO (Invitrogen) 10 and the DNA sequences were determined. The 15C4 kappa chain consensus DNA sequence was used to design primers for full-length antibody chain PCR amplification. The 5’ kappa PCR primer encoded the amino terminus of the signal sequence, an Xbal restriction enzyme site, and an optimized Kozak sequence (5’- CAG CAG AAG CTT CTA GAC CAC CAT GTC GCC ATC ACA ACT CAT TGG G -3’; SEQ ID NO: 47). 15 The 3’ primer encoded the carboxyl terminus and termination codon, as well as a Sail restriction site (5’- CTT GTC GAC TCA ACA CTC TCC CCT GTT GAA GCT C -3’; SEQ ID NO: 48). 5’ alL-lRl 15C4 kappa primer (SEQ ID NO: 47):
20 5’- CAG CAG AAG CTT CTA GAC CAC CAT GTC GCC ATC ACA ACT
Xbal Kozak MS P S Q L CAT TGG G-3’ I G (SEQ ID NO: 49) 25 3’ alL-lRl 15C4 kappa primer (SEQ ID NO: 48): 5 CTT GTC GAC TCA ACA CTC TCC CCT GTT GAA GCT C -3 ’
Sail * CEGRNF S (SEQ ID NO: 50)
The frill-length alL-lRl 15C4 kappa chain clone was obtained using a pCR4: 30 15C4 kappa clone by PCR amplification with the 5’ and 3’ alL-lRl 15C4 kappa primers.
The PCR reaction generated a 733 base pair product encoding the 233 aminn acids residues (including the 19 amino acid kappa chain signal sequence) of the alL-lRl 15C4 74 2013204136 12 Apr 2013 kappa chain. The PCR product was purified using a QIAquick PCR Purification kit (Qiagen Cat. No.28104), cut with Xbal and Sail, gel isolated and purified using a QIAquick Gel Extraction kit (Qiagen Cat. No.28704). This PCR fragment containing the complete alL-lRl 15C4 kappa chain was then ligated into the mammalian expression 5 vector pDSRal9. The 15C4 kappa chain expression clone was DNA sequenced to confirm that it encoded the same peptide that was identified in the 15C4 hybridoma. The final expression vector, pDSRal9:15C4 kappa is 5468 base pairs and contains the seven functional regions described in Table 6. 10 Table 6
Plasmid Base
Pair Number:_ 2 to 881 A transcription termination/polyadenylation signal from the α-subunit of the bovine pituitary glycoprotein hormone (α-FSH) (Goodwin et al., 1983, Nucleic Acids Res. 11:6873-82: Genbank Accession Number X00004) 882 to 2027 A mouse dihydrofolate reductase (DHFR) minigene containing the endogenous mouse DHFR promoter, the cDNA coding sequences, and the DHFR transcription tennination/polyadenylation signals (Gasser et al, 1982, Proc. Natl. Acad. Sci. U. S. A. 79:6522-6; Nunberg et al, 1980, Cell 19:355-64: Setzer et al., 1982, J. Biol. Client. 257:5143-7: McGrogan et al., 1985, J. Biol, Chem. 260:2307-141 2031 to 3947 pBR322 sequences containing the ampicillin resistance marker gene and the origin for replication of the plasmid in E. coli (Genbank Accession Number J01749) 3949 to 4292 An SV40 early promoter, enhancer and origin of replication (Takebe et al., 1988, Mol. Cell Biol. 8:466-72, Genbank Accession Number 102400) 4299 to 4565 A translational enhancer element from the HTLV-1 LTR domain (Seiki et al., 1983, Proc. Natl. Acad. Sci. U. S. A. 80:3618-22. Genbank Accession Number J02029) 4574 to 4730 An intron from the SV40 16S, 19S splice donor/acceptor signals (Okayama and Berg, 1983, Mol. Cell Biol. 3:280-9, Genbank Accession Number J02400) 4755 to 5468 The 15C4 kappa fight chain cDNA between theATwl and Sail sites
15 Construction of pDSR19:liIgGlCH A pDSRal9:rat variable region/human constant region IgGl (rVh/hChl) MAb expression plasmid was constructed as the result of a three-piece ligation of Xbal and 75 2013204136 12 Apr 2013
BsmBl terminated rat antibody variable region PCR product, the human IgGl constant region (CHi, hinge, CH2 and Ch3 domains) derived by SaR cleavage and gel isolation of the BsmBl and Sail fragment from the linear plasmid pDSRal9:hIgGl CH (HimSK and BsmBl ends) and a linearized pDSRal9 with Xbal and SaR ends (see co-owned and co-5 pending U.S. Provisional Patent Application No. 60/370,407, filed April 5,2002, “Human Anti-OPGL Neutralizing Antibodies As Selective OPGL Pathway Inhibitors,” incorporated by reference). The final expression vector, pDSRal9:rat variable region/human constant region IgGl (rVh/hChl), is 6158 base pairs and contains the 7 functional regions described in Table 7. 10 Table 7
Plasmid Base Pair Number: 2 to 881 A transcription termination/polyadenylation signal from the α-subunit of the bovine pituitary glycoprotein hormone (α-FSH) (Goodwin et al., 1983, Nucleic Acids Res. 11:6873-82; Genbank Accession Number X00004) 882 to 2027 A mouse dihydrofolate reductase (DHFR) minigene containing the endogenous mouse DHFR promoter, the cDNA coding sequences, and the DHFR transcription termination/polyadenylation signals (Gasser et al., 1982, Proc. Natl. Acad. Sci. U. S. A. 79:6522-6: Nimberg et al., 1980, Cell 19:355-64; Setzer et al, 1982, J. Biol. Chem. 257:5143-7: McGrogan et al., 1985, J. Biol. Chem. 260:2307-141 2031 to 3947 pBR322 sequences containing the ampicillin resistance marker gene and the origin for replication of the plasmid in E. coli (Genbank Accession Number J01749) 3949 to 4292 An SV40 early promoter, enhancer and origin of replication (Takebe et al, 1988, Mol Cell Biol. 8:466-72, Genbank Accession Number J02400) 4299 to 4565 A translational enhancer element from the HTLV-1 LTR domain (Seiki et al., 1983, Proc. Natl. Acad. Sci. U. S. A 80:3618-22, Genbank Accession Number J02029) 4574 to .4730 An intron from the SV40 16S, 19S splice donor/acceptor signals (Okayama and Berg, 1983. Mol Cell Biol 3:280-9, Genbank Accession Number J02400) 4755 to 6158 The rVh/hChl heavy chain cDNA between the Xbal and Sail sites. This heavy chain fragment sequence is shown below (SEQ 1DN0:51) with the sequences of the restriction sites underlined: Xbal JCTAG ACCACCATGG ACATCAGGCT CAGCTTAGTT TTCCTTGTCC TTTTCATAAA AGGTGTCCAG TGTGAGGTAG AACTGGTGGA GTCTGGGGGC GGCTTAGTAC AACCTGGAAG GTCCATGACA CTCTGCTGTG CAGCCTCGGG ATTCACTTTC AGAACCTATG GCATGGCCTG GGTCCGCCAG GCCCCAACGA AGGGTCTGGA gtgggtctca 76 2013204136 12 Apr 2013
TCAATTACTG CTAGTGGTGG TACCACCTAC TATCGAGACT CCGTGAAGGG CCGCTTCACT ATTTTTAGGG ATAATGCAAA AAGTACCCTA TACCTGCAGA TGGACAGTCC GAGGTCTGAG GACACGGCCA CTTATTTCTG TACATCAATT Bsntji1 TCGGAATACT GGGGCCACGG AGTCATGGTC ACCGTCTCTA GTGCCTCCACCAAGGGCCCA TCGGTCTTCC CCCTGGCACC CTCCTCCAAG AGCACCTCTGGGGGCACAGC GGCCCTGGGC TGCCTGGTCA AGGACTACTT CCCCGAACCG GTGACGGTGT CGTGGAACTC AGGCGCCCTG ACCAGCGGCG TGCACACCTT CCCGGCTGTC CTACAGTCCT CAGGACTCTA CTCCCTCAGC AGCGTGGTGACCGTGCCCTC CAGCAGCTTG GGCACCCAGA CCTACATCTG CAACGTGAATCACAAGCCCA GCAACACCAA GGTGGACAAG AAAGTTGAGC CCAAATCTTG TGACAAAACT CACACATGCC CACCGTGCCC AGCACCTGAA CTOCTGGGGG GACCGTCAGT CTTCCTCTTC CCCCCAAAAC CCAAGGACAC CCTCATGATC TCCCGGACCC CTGAGGTCAC ATGCGTGGTG GTGGACGTGA GCCACGAAGACCCTGAGGTC AAGTTCAACT GGTACGTGGA CGGCGTGGAG GTGCATAATG CCAAGACAAA GCCGCGGGAG GAGCAGTACA ACAGCACGTA CCGTGTGGTC AGCGTCCTCA CCGTCCTGCA CCAGGACTGG CTGAATGGCA AGGAGTACAAGTGCAAGGTC TCCAACAAAG CCCTCCCAGC CCCCATCGAG AAAACCATCTCCAAAGCCAA AGGGCAGCCC CGAGAACCAC AGGTGTACAC CCTGCCCCCA TCCCGGGATG AGCTGACCAA GAACCAGGTC AGCCTGACCT GCCTGGTCAA AGGCTTCTAT CCCAGCGACA TCGCCGTGGA GTGGGAGAGC AATGGGCAGCCGGAGAACAA CTACAAGACC ACGCCTCCCG TGCTGGACTC CGACGGCTCC TTCTTCCTCT ATAGCAAGCT CACCGTGGAC AAGAGCAGGT GGCAGCAGGG GAACGTCTTC TCATGCTCCG TGATGCATGA GGCTCTGCAC AACCACTACA CGCAGAAGAG CCTCTCCCTG TCTCCGGGTA Sail AATGATAAGT CGAC
The linear plasmid pDSRal9:hIgGlCH was prepared by digesting the pDSR19:rat variable region/human constant region IgGl plasmid with the restriction enzymes Xbal and BsmBl to remove the rat variable region and purified using a QlAquick Gel 5 Extraction kit. The linear plasmid pDSRal9:hIgGlCH containing the 1 kbp human IgGl constant region domain was used to accept hybridoma derived odL-lR antibody variable regions.
Cloning of the anti-ILl-RI 15C4 MAb Heavy Chain 10 The heavy chains for ten hybridomas expressing oriLl-RI binding monoclonal antibodies, 15C4, 27F2, and 26E5 were cloned into the mammalian cell expression vector pDSRal9. The construction of the plasmid encoding the 15C4 heavy chain is explicitly described; cloning of the other heavy chain species was performed using similar procedures. The ccIL-lRl 15C4 heavy chain variable region was obtained using PCR 15 amplification methods from first strand cDNA prepared from oILl-RI hybridoma 15C4 77 2013204136 12 Apr 2013 total RNA prepared using TRIzol® reagent First strand cDNA was synthesized using a random primer with an extension adapter (5’- GGC CGG ATA GGC CTC CAN NNN NNT —3’; SEQ ID NO: 44) and a 5’ RACE (rapid amplification of cDNA ends) was performed using the GeneRacer™ Kit. For the partial length heavy chain, the forward 5 primer was the GeneRacer™ nested primer (5’ GGA CAC TGA CAT GGA CTG AAG GAG TA -3’; SEQ ID NO: 45) and the reverse primer was 5’- TGA GGA CGC TGA CCA CAC G -3’ (SEQ ID NO 52.). The RACE products were cloned into pCR4-TOPO and the DNA sequences were determined. The 15 C4 heavy chain variable region consensus DNA sequence was used to design primers for the heavy chain variable region 10 PCR amplification. The 5’ heavy chain PCR primer encoded the amino terminus of the signal sequence, an Xbal restriction enzyme site, and an optimized Kozak sequence (5’-CAG CAG AAG CTT CTA GAC CAC CAT GGG GTC AAC CGC CAT CCT COS’; SEQ ID NO: 53). The 3’ primer encoded the carboxyl end of the variable region, including a naturally occurring sense strand BsniBL site (5’-GTG GAG GCA CTA GAG 15 ACG GTG ACC AGG GTT CC-3 SEQ ID NO: 54). 5’aIL-lRl 15C4 heavy chain primer (SEQ ID NO: 53):
5’- CAG CAG AAG CTT CTA GAC CAC C ATG GGG TCA ACC GCC 20 Xbal Kozak MGS T A ATCCTCG—3’ I L (SEQ ID NO: 55) 3’aIL-lRl 15C4 heavy chain primer (SEQ ID NO: 54): 25 5’- GTG GAG GCA CTA GAG ACG GTG ACC AGG GTT CC-3’ TSASSVTVLT G (SBQ ID NO: 56)
Us/mBI
Construction of the anti-ILl-RT IaGl Heavy Chain Expression Clone 30
The full-length odL-lRl 15C4 heavy chain clone was obtained fiom a pCR4:15C4 heavy chain clone by PCR amplification with the 5’ and 3’ alL-lRl 15C4 heavy chain primers. The PCR reaction generated a 442 base pair product encoding the 137 amino acids residues (including the 19 amino acid heavy chain signal sequence) of 78 2013204136 12 Apr 2013 the aH/TRl 15C4 heavy chain variable region. The PCR product was purified using a QIAquickPCR Purification kit and then digested with Xbal and BsmBI, gel isolated and purified using a QIAquick Gel Extraction kit. This fiagment containing the complete alL-lRl 15C4 heavy chain variable region was then ligated into the mammalian 5 expression vector pDSRal9;hIgGlCH. The 15C4 heavy chain IgGl expression clone was DNA sequenced to confirm that it encoded the same heavy chain variable region peptide that was identified in the 15C4 hybridoma. The final expression vector, pDSRal9:15C4 IgGl heavy chain was 6173 base pairs and contains the seven functional regions described in Table 8.
Plasmid Base Pair Number: 2 to 881 A transcription tennination/polyadenylation signal from the α-subiinit of the bovine pituitary glycoprotein hormone (cc-FSH) (Goodwin et al.t 1983, Nucleic Acids Res. 11:6873-82; Genbank Accession Number X00004) 882 to 2027 A mouse dihydrofolate reductase (DHFR) minigene containing the endogenous mouse DHFR promoter, the cDNA coding sequences, and the DHFR transcription teimination/polyadenylation signals (Gasser et al., 1982, Proc. Nad. Acad. Sci. U, S. A, 79:6522-6; Nunberg et al., 1980, Cell 19:355-64; Setzer et al., 1982, J. Biol. Chem. 257:5143-7; McGrogan et al., 1985,./. Biol. Chem. 260:2307-141 2031 to 3947 pBR322 sequences containing the ampicillin resistance marker gene and the origin for replication of the plasmid in E, coli (Genbank Accession Number JO1749) 3949 to 4292 An SV40 early promoter, enhancer and origin of replication (Takebe et al., 1988, Mol. Cell Biol. 8:466-72, Genbank Accession Number J02400) 4299 to 4565 A translational enhancer element from the HTLV-1 LTR domain (Seiki et al„ 1983, Proc. Natl. Acad. Sci, U. S. A. 80:3618-22, Genbank Accession Number J02029) 4574 to 4730 An intron from the SV40 16S, 19S splice donor/acceptor signals (Okayama and Berg, 1983. Mol, Cell Biol. 3:280-9, Genbank Accession Number J02400) 4755 to 6173 The 15C4 heavy chain IgGl cDNA between the Xbal and Sail sites 79 2013204136 12 Apr 2013
Construction of pDSR19:hIgG2Ctj A pDSRctl9:human variable region/human constant region IgG2 (hVh/hCh2) MAb expression plasmid was constructed as the result of a three-piece ligation of Xbal and BsmBl terminated human antibody variable region PCR product, a human lgG2 5 constant region (CHi, hinge, Cm and CH3 domains) PCR product with BsmBl and Sail ends and a linearized pDSRal9 with Xbal and Sail ends. The final expression vector, pDSRori9:human variable region/human constant region IgGl (hVli/hCh2) (see coowned and co-pending U.S. Provisional Patent Application No. 60/370,407, filed April 5, 2002, 'Human Anti-OPGL Neutralizing Antibodies As Selective OPGL Pathway 10 Inhibitors”), is 6164 base pairs and contains the 7 functional regions described in Table 9.
Table 9
Plasmid Base 15 Pair Number: 2 to 881 A transcription termination/polyadenylation signal from the α-subimit of the bovine pituitary glycoprotein hormone (α-FSH) (Goodwin et al, 1983, Nucleic Acids Res. 11:6873-82; Genbank Accession Number X00004) 882 to 2027 A mouse dihydrofolate reductase (DHPR) minigene containing the endogenous mouse DHPR promoter, the cDNA coding sequences, and the DHPR transcription termination/polyadenylation signals (Gasser et al., 1982, Proa Natl. Acad. Sci. U. S. A. 79:6522-6: Nunberg et al., 1980, Cell 19:355-64; Setzer et al, 1982, J. Biol. Chem. 257:5143-7; McGi'ogan et al, 1985, J. Biol Chem. 260:2307-14) 2031 to 3947 pBR322 sequences containing the ampicillin resistance marker gene and the origin for replication of the plasmid in E. coli (Genbank Accession Number J01749) 3949 to 4292 An SV40 early promoter, enhancer and origin of replication (Takebe et al, 1988, Mol Cell Biol 8:466-72, Genbank Accession Number J02400) 4299 to 4565 A translational enhancer element from the HTLV-1 LTR domain (Seiki et al, 1983, Proc. Natl. Acad. Sci. U. S. A. 80:3618-22, Genbank Accession Number J02029) 4574 to 4730 An intron from the SV40 16S, 19S splice donor/acceptor signals (Okayama and Berg, 1983. Mol Cell Biol. 3:280-9, Genbank Accession Number J02400) 4755 to 6164 The hVh/hCh2 heavy chain cDNA between the Xbal and Sail sites. The sequence of this heavy chain fragment appears below (SEQ ID NO: 57) with the restriction sites underlined: Xbal 80 2013204136 12 Apr 2013
TCTAGA CCACCATGGA CATGAGGGTC CCCGCTCAGC TCCTGGGGCT CCTGCTATTG TGGITGAGAG GTGCCAGATG TGAGGTCCAG CTGGTGCAGTCTGGGGGAGG CTTGGTACAT CCTGGGGGGT CCCTGAGACT CTCCTGTGCAGGCTCTGGAT TCACCTTCAG TGGCCATGCT TTGCACTGGG TTCGCCAGGCTCCAGGAAAA GGTCTGGAGT GGGTATCAGG TATTGGTACT CATGGTGGGACATACTATGC AGACTCCGTG AAGGGCCGAT TCACCATCTC CAGAGACAATGCCAAGAACT CCTTGTTTCT TCAAATGAAC AGCCTGAGCG CCGAGGACATGGCTGTGTAT TACTGTACAA GAAGAAACTG BsmBI GGGACAATTT GACTACTGGGGCCAGGGAAC CCTGGTCACC GTCTCTAGTG CCTCCACCAA GGGCCCATCGGTCTTCCCCC TGGCGCCCTG CTCCAGGAGC ACCTCCGAGA GCACAGCGGCCCTGGGCTGC CTGGTCAAGG ACTACTTCCC CGAACCGGTG ACGGTGTCGTGGAACTCAGG CGCTCTGACC AGCGGCGTGC ACACCTTCCC AGCTGTCCTACAGTCCTCAG GACTCTACTC CCTCAGCAGC GTGGTGACCG TGCCCTCCAGCAACTTCGGC ACCCAGACCT ACACCTGCAA CGTAGATCAC AAGCCCAGCAACACCAAGGT GGACAAGACA GTTGAGCGCA AATGTTGTGT CGAGTGCCCACCGTGCCCAG CACCACCTGT GGCAGGACCG TCAGTCTTCC TCTTCCCCCCAAAACCCAAG GACACCCTCA TGATCTCCCG GACCCCTGAG GTCACGTGCGTGGTGGTGGA CGTGAGCCAC GAAGACCCCG AGGTCCAGTT CAACTGGTACGTGGACGGCG TGGAGGTGCA TAATGCCAAG ACAAAGCCAC GG G AGGAGC AGTTCAACAGC ACGTTCCGTG TGGTCAGCGT CCTCACCGTT GTGCACCAGGACTGGCTGAA CGGCAAGGAG TACAAGTGCA AGGTCTCCAA CAAAGGCCTCCCAGCCCCCA TCGAGAAAAC CATCTCCAAA ACCAAAGGGC AGCCCCGAGAACCACAGGTG TACACCCTGC CCCCATCCCG GGAGGAGATG ACCAAGAACCAGGTCAGCCT GACCTGCCTG GTCAAAGGCT TCTACCCCAG CGACATCGCCGTGGAGTGGG AGAGCAATGG GCAGCCGGAG AACAACTACA AGACCACACCTCCCATGCTG GACTCCGACG GCTCCTTCTT CCTCTACAGC AAGCTCACCGTGGACAAGAG CAGGTGGCAG CAGGGGAACG TCTTCTCATG CTCCGTGATGCATGAGGCTC TGCACAACCA CTACACGCAG Sail AAGAGCCTCT CCCTGTCTCC GGGTAAATGA TAAGTCGAC
The linear plasmid pDSRori9:hIgG2CH was prepared by digesting the pDSR19:human variable region/human constant region IgG2 plasmid with the restriction enzymes Xbal and BsmBI to remove the human variable region and purified using a 5 QIAquick Gel Extraction kit. The linear plasmid pDSRal9:hIgG2CH containing the 1 kbp human IgG2 constant region domain was used to accept hybridoma derived alL-lR antibody variable regions.
Construction of the anti-ILl-RI IaG2 Heavy Chain Expression Clone 10
The alL-lRl 15C4 heavy chain variable region fragment, described above, was ligated into the mammalian expression vector pDSRal9;hIgG2CH. The 15C4 heavy 81 2013204136 12 Apr 2013 10 chain IgG2 expression clone was DNA sequenced to confirm that it encoded the same heavy chain variable region peptide that was identified in the 15C4 hybridoma. The final expression vector) pDSRal9:15C4 IgG2 heavy chain was 6161 base pairs and contains the seven functional regions described in Table 10.
Table 10
Pair Number: 2 to 881 A transcription termination/polyadenylation signal from the α-subunit of the bovine pituitary glycoprotein hormone (a-FSH) (Goodwin et al, 1983, Nucleic Acids Res. 11:6873-82; Genbank Accession Number X00004) 882 to 2027 A mouse dihydrofolate reductase (DHFR) minigene containing the endogenous mouse DHFR promoter, the cDNA coding sequences, and the DHFR transcription termination/polyadenylation signals (Gasser et al., 1982, Proc. Natl, Acad. Set. U. S. A. 79:6522-6: Nunberg et al., 1980, Cell 19:355-64; Setzer et al, 1982, J. Biol. Chem. 257:5143-7: McGrogan et al, 1985, J Biol Chem. 260:2307-14) 2031 to 3947 pBR322 sequences containing the ampicillin resistance marker gene and the origin for replication of the plasmid in E. coli (Genbank Accession Number J01749) 3949 to 4292 An SV40 early promoter, enhancer and origin of replication (Takebe et al., 1988, Mol, Cell Biol. 8:466-72, Genbank Accession Number J02400) 4299 to 4565 A translational enhancer element from the HTLV-1 LTR domain (Seiki et al, 1983, Proc. Natl. Acad. Sci. U. S. A. 80:3618-22. Genbank Accession Number J02029) 4574 to 4730 An intron from the SV40 16S, 19S splice donor/acceptor signals (Okayama and Berg, 1983. Mol. Cell Biol. 3:280-9, Genbank Accession Number J02400) 4755 to 6161. The 15 C4 heavy chain IgG2 cDNA between the Xbal and Sail sites
Construction of oDSRl 9;hIgG4Cn A pDSRal9:human variable region/human constant region IgG4 (hVh/hCh4) 15 MAb expression plasmid was constructed as the result of a three-piece ligation of Xbal and SsniBl terminated human antibody variable region PCR product, a gel isolated BsmBI and Sail digested human IgG4 constant region (Chi, hinge, Ch2 and Ch3 domains) fragment and a linearized pDSRal9 with Xbal and Sail ends. The final expression vector, pDSRocl9:human variable region/human constant region IgG4 (hVh/hCh4) (see co- 82 2013204136 12 Apr 2013 owned and co-pending U.S. Provisional Patent Application No. 60/370,407, filed April 5, 2002, “Human Anti-OPGL Neutralizing Antibodies As Selective OPGL Pathway Inhibitors”), is 6167 base pairs and contains the 7 functional regions described in Table 11. 10
Table 11 . Plasmid Base Pair Number:
2 to 881 A transcription termination/polyadenylation signal from the α-subunit of the bovine pituitary glycoprotein hormone (α-FSH) (Goodwin et al., 1983, Nucleic Acids Res, 11:6873-82; Genbank Accession Number X00004) 882 to 2027 A mouse dihydrofolate reductase (DHFR) minigene containing the endogenous mouse DHFR promoter, the cDNA coding sequences, and the DHFR transcription termination/polyadenylation signals (Gasser et al, 1982, Proc. Natl. Acad. Sci. U. S. A. 79:6522-6: Nunberg et al., 1980, Cell 19:355-64; Setzer et al., 1982, J. Biol. Chem. 257:5143-7; McGrogan et al., 1985, J. Biol. Chem. 260:2307-14) 2031 to 3947 pBR322 sequences containing the ampicillin resistance marker· gene and the origin for replication of the plasmid in E. coli (Genbank Accession Number J01749) 3949 to 4292 An SV40 early promoter, enhancer and origin of replication (Talcebe et al., 1988, Mol. Cell Biol. 8:466-72, Genbank Accession Number J02400) 4299 to 4565 A translational enhancer element from the HTLV-1 LTR domain (Seiki et al., 1983, Proc. Natl. Acad. Sci. U. S. A. 80:3618-22, Genbank Accession Number J02029) 4574 to 4730 An intr'on from the SV40 16S, 19S splice donor/acceptor signals (Okayama and Berg, 1983. Mol. Cell Biol. 3:280-9, Genbank Accession Number J02400) 4755 to 6167 The hVh/hCh4 heavy chain cDNA between theXba\ and Sail sites. The sequence of this heavy chain fragment appears below (SEQ ID NO:58) with the restriction sites underlined: Xbal TCT AGACCACCAT GGACATGAGG GTCCCCGCTC A GCTCCTGOG GCTCCTGCTA TTGTGGTTGA GAGGTGCCAG ATGTGAGGTC CAGCTGGTGCAGTCTGGGGG AGGCTTGGTA CATCCTGGGG GGTCCCTGAG ACTCTCCTGTGCAGGCTCTG GATTCACCTT CAGTGGCCAT GCTTTGCACT GGGTTCGCCAGGCTCCAGGA AAAGGTCTGG AGTGGGTATC AGGTATTGGT ACTCATGGTGGGACATACTA TGCAGACTCC GTGAAGGGCC GATTCACCAT CTCCAGAGACAATGCCAAGA ACTCCTTGTT TCTTCAAATG AACAGCCTGA GCGCCGAGGACATGGCTGTG TATTACTGTA CAAGAAGAAA CTGGGGACAA TTTGACTACTGGGGCCAGGG BsntBl AACCCTGGTC ACCGTCTCTA GTGCCAGCAC CAAGGGGCCATCCGTCTTCC 83 2013204136 12 Apr 2013
CCCTGGCGCC CTGCTCCAGG AGCACCTCCG AGAGCACAGCCGCCCTGGGC TGCCTGGTCA AGGACTACTT CCCCGAACCG GTGACGGTGTCGTGGAACTC AGGCGCCCTG ACCAGCGGCG TGCACACCTT CCCGGCTGTCCTACAGTCCT CAGGACTCTA CTCCCTCAGC AGCGTGGTGA CCGTGCCCTCC AGCAG CTTG GGCACGAAGA CCTACACCTG CAACGTAGAT CACAAGCCCAGCAACACCAA GGTGGACAAG AGAGTTGAGT CCAAATATGG TCCCCCATGCCCATCATGCC CAGCACCTGA GTTCCTGGGG GGACCATCAG TCTTCCTGTTCCCCCCAAAA CCCAAGGACA CTCTCATGAT CTCCCGGACC CCTGAGGTCACGTGCGTGGT GGTGGACGTG AGCCAGGAAG ACCCCGAGGT CCAGTTCAACTGGTACGTGG ATGGCGTGGA GGTGCATAAT GCCAAGACAA AGCCGCGGGAGGAGCAGTTC AACAGCACGT ACCGTGTGGT CAGCGTCCTC ACCGTCCTGCACCAGGACTG GCTGAACGGC AAGGAGTACA AGTGCAAGGT CTCCAACAAAGGCCTCCCGT CCTCCATCGA GAAAACCATC TCCAAAGCCA AAGGGCAGCCCCGAGAGCCA CAGGTGTACA CCCTGCCCCC ATCCCAGGAG GAGATGACCAAGAACCAGGT CAGCCTGACC TGCCTGGTCA AAGGCTTCTA CCCCAGCGACATCGCCGTGG AGTGGGAGAG CAATGGGCAG CCGGAGAACA
ACTACAAG ACCACGCCT CCC GTGCTGGACT CCGACGGCTC CTTCTTCCTC TACAGCAGGCTAACC GTGGA CAAGAGCAGG TGGCAGGAGG GGAATGTCTT CTCATGCTCCGTGATGCATG AGGCTCTGCA CAACCACTAC
Sail
ACACAGAAGA GCCTCTCCCTGTCTCTGGGT AAATGATAAG TCGAC
The linear plasmid pDSRal9:hIgG4CH was prepared by digesting the pDSR19:human variable region/human constant region IgG4 plasmid with the restriction enzymes Xbai and BstriBL to remove the human variable region and purified using a 5 QIAquick Gel Extraction kit The linear plasmid pDSRal9:hIgG4CH containing the 1 kbp human IgG4 constant region domain was used to accept hybridoma derived alL-lR antibody variable regions.
Construction of the anti-ILl-RI IgG4Heavv Chain Expression Clone 10
The cdL-lRl 15C4 heavy chain variable region fragment, described above, was ligated into the mammalian expression vector pDSRal9:hIgG4CH. The 15C4 heavy chain IgG4 expression clone was DNA sequenced to confirm that it encoded the same heavy chain variable region peptide that was identified in the 15C4 hybridoma. The final 15 expression vector, pDSRral9:15C4 IgG4 heavy chain was 6164 base pairs and contains the seven functional regions described in Table 12.
Table 12 20 84 2013204136 12 Apr 2013
Plasmid Base 2 to 881 A transcription termination/polyadenylation signal from the a-subunit of the bovine pituitary glycoprotein hormone (α-FSH) (Goodwin et al, 1983, Nucleic Acids Res. 11:6873-82; Genbanfc Accession Number X00004) 882 to 2027 A mouse dihydrofolate reductase (DHFR) minigene containing the endogenous mouse DHFR promoter, the cDNA coding sequences, and the DHFR transcription termination/polyadenylation signals (Gasser et al, 1982, Proc.. Natl. Acad. Sci. U. S. A. 79:6522-6; Nunberg et al., 1980, Cell 19:355-64; Setzer et al., 1982, ,/ Biol. Chem. 257:5143-7; McGrogan et al, 1985,./ Biol Chem. 260:2307-14) 2031 to 3947 pBR322 sequences containing the ampicillin resistance marker gene and the origin for replication of the plasmid in E. coli (Genbank Accession Number JO 1749) 3949 to 4292 An SV40 early promoter, enhancer and origin of replication (Takebe et al, 1988, Mol, Cell Biol. 8:466-72, Genbank * Accession Number J02400) 4299 to 4565 A translational enhancer element from the HTLV-1 LTR domain (Seiki et al, 1983, Proc. Natl, Acad. Sci. U. S. A. 80:3618-22. Genbank Accession Number J02029) 4574 to 4730 An intron from the SV40 16S, 19S splice donor/acceptor signals (Okayama and Berg, 1983, Mol, Cell Biol. 3:280-9, Genbank Accession Number J02400) 4755 to 6164 The 15C4 heavy chain IgG4 cDNA between the .AM and Sail sites 5
Example 7
Expression of anti-IL-lRl antibodies iii Chinese Hamster Ovary (CHOI Cells
Recombinant anti-IL-lRl antibodies are generated in Chinese hamster ovary cells, specifically CHO AM-l/D, as disclosed in U.S. Patent No. 6,210,924 (incorporated by 10 reference). Briefly, the DNA sequences encoding the complete heavy or light chains of each anti-IL-lRl antibody of the invention are cloned into expression vectors. CHO AM-l/D cells are co-transfected with an expression vector capable of expressing a complete heavy chain and an expression vector expressing the complete light chain of the appropriate anti-IL-lRl antibody. For example, to generate the 26F5 antibody, cells are 15 co-transfected with a vector capable of expressing a complete light chain comprising the amino acid sequence as set forth in SEQ ID NO:38 and a vector capable of expressing a complete heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 20, SEQ ID NO: 22, or SEQ ID NO: 24. To generate the 27F2 antibody, cells are co- 85 2013204136 12 Apr 2013 transfected with a vector capable of expressing a complete light chain comprising the amino acid sequence as set forth in SEQ ID NO: 38 and a vector capable of expressing a complete heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 26, SEQ ID NO: 28, or SEQ ID NO: 30. To generate the 15C4 antibody, cells are co-5 transfected with a vector capable of expressing a complete light chain comprising the amino acid sequence as set forth in SEQ ID NO: 40 and a vector capable of expressing a complete heavy chain comprising the ammo acid sequence set forth in SEQ ID NO: 32, SEQ ID NO: 34, or SEQ ID NO: 36. Table 13 summarizes the complete heavy and complete light chains for the various IL-1R1 antibodies. The designation ".../TgG_” 10 describes the sequence of the constant region for the particular antibody.
Table 13
Antibody Heavy Chain Variable Region + Heavy Chain Constant Region Complete Heavy Chain 26F5/IgGl (nucleotide) SEQ ID NO: 9 + SEQ ID NO: 1 SEQ ID NO: 19 26F5/IgGl (amino acid) SEQ ID NO: 10 + SEQ ID NO: 2 SEQ ID NO: 20 26F5/IgG2 (nucleotide) SEQ ID NO: 9 + SEQ ID NO: 5 SEQ ID NO: 21 26F5/IgG2 (amino acid) SEQ ID NO: 10 + SEQ ID NO: 6 SEQ ID NO: 22 26F5flgG4 (nucleotide) SEQ ID NO: 9 + SEQ Π) NO: 7 SEQ ID NO: 23 26FS/IgG4 (amino acid) SEQ ID NO: 10 + SEQ ID NO: 8 SEQ ID NO: 24 27F2hgGl (nucleotide) SEQ ID NO: 13 + SEQ ID NO: 1 SEQ ID NO: 25 271f2/IgGl (amino acid) SEQ ID NO: 14 + SEQ ID NO: 2 SEQ ID NO: 26 27F2HgG2 (nucleotide) SEQ ID NO: 13 + SEQ ID NO: 5 SEQ ID NO: 27 27F2dgG2 (amino acid) SEQ ID NO: 14 + SEQ ID NO: 6 SEQ ID NO: 28 27F2«gG4 (nucleotide) SEQ ID NO: 13 + SEQ ID NO: 7 SEQ ID NO: 29 27F2/IgG4 (amino acid) SEQ ID NO: 14 + SEQ ID NO: 8 SEQ ID NO: 30 15C4/IgGl (nucleotide) SEQ ID NO: 15 + SEQ ID NO: 1 SEQ ID NO: 31 l5C4/IgGl (amino acid) SEQ ID NO: 16 + SEQ ID NO: 2 SEQ ID NO: 32 15C4flgG2 (nucleotide) SEQ ID NO: 15 + SEQ ID NO: 5 SEQ ID NO: 33 15C4/IgG2 (amino acid) SEQ ID NO: 16 + SEQ Π) NO: 6 SEQ ID NO: 34 86 2013204136 12 Apr 2013 15C4/TgG4 (nucleotide) SEQ ID NO: 15 + SEQ ID NO: 7 SEQ ID NO: 35 15C4/TgG4 (amino acid) SEQ ID NO: I6 + SEQIDNO: 8 SEQ ID NO: 36 Antibody Light Chain Variable Region + Light Chain Constant Region Complete Light Chain 26F5/27F2 (nucleotide) SEQ ID NO: 11 + SEQ ID NO: 3 SEQ ID NO: 37 26F5/27F2 (amino acid) SEQ ID NO: 12 + SEQ ID NO: 4 SEQ ID NO: 38 15C4 (nucleotide) SEQ ID NO: 17 + SEQ ID NO: 3 SEQ ID NO: 39 15C4 (amino acid) SEQ ID NO: 18 + SEQ ID NO: 4 SEQ ID NO: 40
Stable expression of anti-IL-lRl antibodies is achieved by co-transfecting dihydrofolate reductase deficient (DHFR') CHO AM-l/D cells with the expression 5 vectors. Transfections are carried out using standard techniques (calcium phosphate coprecipitation) and DHFR selection. Transfected colonies are isolated and grown to confluence in 24-well plates. Antibodies produced by transfected cells are examined for appropriate folding and neutralizing activity. Clones overproducing appropriately folded anti-IL-lRl antibodies of the IgGl, IgG2, and fgG4 isotypes are selected and antibodies 10 are purified as described below.
Example 8
Production of anti-IL-lRl Antibody
Anti-IL-lRl antibodies are produced by expression in a clonal line of CHO cells. 15 For each production run, cells from a single vial are thawed into scrum-free cell culture media. The cells are grown initially in a T-flask and are serially expanded through a series of spinner flasks until sufficient inoculum has been generated to seed a 20L bioreactor. Following growth for 5-10 days, the culture is then used to inoculate a 300L bioreactor. Following growth for an additional 5-10 days, the culture is used to inoculate 20 a 2000L bioreactor. Production is carried out in a 2000L bioreactor using a fed batch culture, in which a nutrient feed containing concentrated media components is added to maintain cell growth and culture viability. Production lasts for approximately two weeks 87 2013204136 12 Apr 2013 during which time anti-ILl-Rl antibody is constitutively produced by the cells and secreted into the cell culture medium.
The production reactor is controlled at set pH, temperature, and dissolved oxygen level: pH is controlled by carbon dioxide gas and sodium carbonate addition; dissolved 5 oxygen is controlled by air, nitrogen, and oxygen gas flows.
At the end of production, the cell broth is fed into a disk stack centrifuge and the culture supernatant is separated from the cells. The concentrate is further clarified through a depth filter followed by a 0.2 ptm filter. The clarified conditioned media is then concentrated by tangential flow ultrafiltration. The conditioned media is concentrated 15-10 to 30- fold. The resulting concentrated conditioned medium is then either processed through purification or frozen for purification at a later date.
Example 9
Epitope Mapping Using Avidiu-Fusiou Proteins 15 To generate avidin-fusion proteins, cDNA encoding chicken avidin (with endogenous signal sequence) was joined with the 5’ end of cDNAs encoding the mature extracellular domains of human- or cynomolgus IL-1RI fused to a FLAG-tag sequence at the 3’ end. The FLAG-tagged fusion genes were assembled in a pALTERMAX vector using conventional molecular techniques. The amino acid sequence of the avidin-human 20 IL-1R1 fusion protein is shown in Figure 23 (SEQ ED NO: 59). The amino acid sequence of the avidin-cynomolgus IL-1R1 fusion protein is show in Figure 24 (SEQ ED NO: 60). A panel of mutant avidin-cynoIL-lRI-FLAG proteins in which human amino acids were substituted for the corresponding cynomolgus residues was generated using the Altered Sites Π Mammalian In Vitro Mutagenesis System (Promega Corp.). The mutations are 25 illustrated in Figure 24.
Plasmids encoding the avidin-cynoIL-lR mutant and wild-type proteins as well as the avidin-huIL-1RI-FLAG protein were transiently transfected into 293T cells using Cytofectine transfection reagent (Bio-Rad Laboratories, Inc.). Mock transfectants were used as negative controls. Anti-huIL-lRI monoclonal antibody (MAb) binding to these 30 proteins was evaluated by Western blot and bead-based binding assays using conditioned medium (CM) harvested from the transfected cells. 88 2013204136 12 Apr 2013
For Western blot analysis, CM was diluted 1:3 in non-reducing SDS sample buffer, boiled for 5-10 minutes and loaded onto 10% Tris-glycine gels. Following SDS-PAGE and Western transfer, the membranes were blocked with 3%BSA/1% ovalbumin in PBS/0.1% Tween-20 (PBST) and stained with anti-huIL-lRIMAbs. A goat anti-human 5 IgG-Fc-HRP antibody (Pierce Chemical Co.) diluted 1: 15,000 in PBST was used for secondary detection, Anti-FLAG detection was used to normalize for protein loading. Image capture and densitometry were performed using a FluorChem 8000 digital imaging system (Alpha Innotech Corp.). The signal intensities for the anti-huEL-lRI MAbs were normalized against the values for the anti-FLAG antibody to account for· variation in 10 protein loading. Antibody binding was expressed as a percentage of binding to the avi din-human IL-1R1-FLAG.
The results of the Western blot are shown in Figure 25A. Figure 25B shows the densitometric analysis of a duplicate set of Western blot experiments. Human residues critical for antibody binding are those that restore signal when substituted into cynoIL-15 1RI. In general, mutations 1 and 2 (illustrated in Figure 24), alone or in combination, restored binding to many of the antibodies (15C4/IgG2, 5B8,1C2,24H2, 16E9, 26E4 and 20G1) while mutations 10.1 and 10.2 did not None of these antibodies bound to wild-type cynoIL-lRI. Two antibodies (27F2 and 19C8) bound consistently to all the mutant proteins as well as to wild-type cynoIL-lRI. This suggested that epitope 4 (residues 20 Y279-V281 of cynoIL-lRI), identified in the rat/human paralog proteins and unchanged in cynomolgus IL-1RI, was the dominant epitope for these antibodies. Epitope 4 is bold, italicized, and underlined in the amino acid sequence shown in Figure 24. hi the multiplexed bead-based binding assays, avidin fusion proteins were captur ed by incubation of the CM with biotin-coated fluorescent beads, one bead set per 25 fusion protein (Beadlyte Multi-Biotin lOplex Bead Kit; Upstate Biotechnologies). The beads were washed and pooled in PBST and aliquoted to the wells of a 96-well filter bottom plate (Millipore Corp.). Antibodies (anti-huIL-lRI MAbs or anti-FLAG MAb) were added at 25 pg/ml and incubated for 1 hour. The beads were again washed and a mixture of Phycoerythrin-conjugated anti-mouse IgG antibody and anti-human IgG 30 (Fab’)2 was used to detect antibody binding. After a 1 hour incubation, the beads were washed and resuspended in PBST. Mean fluorescence intensities (MFI) were measured using a Luminex 100 (Luminex Corp). The data were normalized using the MFI values 89 for anti-FLAG MAb binding to account for variation in protein loading. Antibody binding was expressed as a percentage of binding to the avidin-huIL-lRl-FLAG (Figure 26). The binding pattern of the anti-IL-lRl antibodies to the avidin-cynoILlRl-FLAG proteins mutated with human residues as well as to wild-type 5 cynomolgus and human IL-1R1 proteins was consistent with the immunobolt analysis shown in Figure 25. 2013204136 12 Apr 2013
It should be understood that the foregoing disclosure emphasizes certain specific embodiments of the invention and that all modifications or alternatives equivalent thereto are within the spirit and scope of the invention as set forth in the 10 appended claims.
Throughout the specification, unless the context requires otherwise, the word “comprise” or variations such as “comprises” or “comprising”, will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers. 15 Each document, reference, patent application or patent cited in this text is expressly incorporated herein in their entirely by reference, which means that it should be read and considered by the reader as part of this text. That the document, reference, patent application, or patent cited in this text is not repeated in this text is merely for reasons of conciseness. 20 Reference to cited material or information contained in the text should not be understood as a concession that the material or information was part of the common general knowledge or was known in Australia or any other country. 90

Claims (92)

1. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 10, or an antigen-binding fragment thereof.
2. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the light chain comprises a light chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 12, or an antigen-binding fragment thereof.
3. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises an amino acid sequence as set forth in any of SEQ ID NO: 20, SEQ ID NO: 22, or SEQ ID NO: 24, or an antigen-binding fragment thereof.
4. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof.
5. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region comprising an amino acid sequence as set forth in SEQ ID NO: 14, or an antigen-binding fragment thereof.
6. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises an amino acid sequence as set forth in any of SEQ ID NO: 26, SEQ ID NO: 28, or SEQ ID NO: 30, or an antigen-binding fragment thereof.
7. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises: a heavy chain variable region comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 10, or an antigen-binding fragment thereof, and a light chain variable region comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 12, or an antigen-binding fragment thereof.
8. The antibody of Claim 7, wherein the heavy chain variable region comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 10, and wherein the light chain variable region comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 12, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
9. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises: a heavy chain variable region comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 14, or an antigen-binding fragment thereof, and a light chain variable region comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 12, or an antigen-binding fragment thereof.
10. The antibody of Claim 9, wherein the heavy chain variable region comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 14, and wherein the light chain variable region comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 12, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
11. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises: (a) a light chain comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof; and (b) a heavy chain comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 20, SEQ ID NO: 22, or SEQ ID NO: 24, or an antigen-binding fragment thereof.
12. The antibody of Claim 11, wherein the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 20, or an antigen-binding fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof.
13. The antibody of Claim 11, wherein the heavy chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 20, and wherein the light chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
14. The antibody of Claim 11, wherein the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 22, or an antigen-binding fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof.
15. The antibody of Claim 11, wherein the heavy chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 22, and wherein the light chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
16. The antibody of Claim 11, wherein the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 24, or an antigen-binding fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof.
17. The antibody of Claim 11, wherein the heavy chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 24, and wherein the light chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
18. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises: (a) a light chain comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof; and (b) a heavy chain comprising an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 26, SEQ ID NO: 28, or SEQ ID NO: 30, or an antigen-binding fragment thereof.
19. The antibody of Claim 18, wherein the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 26, or an antigen-binding fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof.
20. The antibody of Claim 18, wherein the heavy chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 26, and wherein the light chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
21. The antibody of Claim 18, wherein the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 28, or an antigen-binding fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof.
22. The antibody of Claim 18, wherein the heavy chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 28, and wherein the light chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
23. The antibody of Claim 18, wherein the heavy chain comprises an amino acid sequence as set forth in SEQ ID NO: 30, or an antigen-binding fragment thereof, and the light chain comprises an amino acid sequence as set forth in SEQ ID NO: 38, or an antigen-binding fragment thereof.
24. The antibody of Claim 18, wherein the heavy chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 30, and wherein the light chain comprises an amino acid sequence that has at least 90% sequence identity to the amino acid sequence as set forth in SEQ ID NO: 38, wherein the antibody specifically binds to an interleukin-1 receptor type 1 (IL-1R1).
25. The antibody according to any one of Claims 1 to 24, wherein the heavy chain and light chain are connected by a flexible linker to form a single-chain antibody.
26. The antibody of Claim 25, which is a single-chain Fv antibody.
27. The antibody according to any one of Claims 1 to 24, which is selected from the group consisting of: an Fab antibody fragment, an Fab’ antibody fragment, or an (Fab’)2 antibody fragment.
28. The antibody according to any one of Claims 1 to 24, wherein the antibody is a fully human antibody.
29. The antibody according to any one of Claims 1 to 28, wherein the antibody inhibits binding of IL-1 to its receptor.
30. A method of treating an IL-1 mediated disease in a patient, comprising administering to a patient in need thereof a pharmaceutically effective amount of the antibody of Claim 29.
31. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of the antibody of Claim 29.
32. A method of treating an IL-1 mediated disease in a patient, comprising administering to a patient in need thereof the pharmaceutical composition of Claim 31.
33. The antibody according to any one of Claims 1 to 24, which is an IgG2 antibody.
34. The antibody according to any one of Claims 1 to 29, which binds specifically to the polypeptide of SEQ ID NO: 76.
35. The antibody according to any one of Claims 1 to 29, which binds specifically to Epitope 4 of IL-1R1, and wherein Epitope 4 comprises the amino acid sequence YSV.
36. An isolated human antibody comprising: (a) human heavy chain framework regions, a human heavy chain CDR1 region comprising SEQ ID NO: 61, a human heavy chain CDR2 region comprising SEQ ID NO: 64, and a human heavy chain CDR3 region comprising SEQ ID NO: 67; and (b) human light chain framework regions, a human light chain CDR1 region comprising SEQ ID NO: 70, a human light chain CDR2 region comprising SEQ ID NO: 72, and a human light chain CDR3 region comprising SEQ ID NO: 74.
37. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 26F5, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 61, SEQ ID NO: 64, and SEQ ID NO: 67.
38. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the light chain comprises a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 26F5, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 70, SEQ ID NO: 72, and SEQ ID NO: 74.
39. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 26F5, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 61, SEQ ID NO: 64, and SEQ ID NO: 67; and a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 26F5, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 70, SEQ ID NO: 72, and SEQ ID NO: 74.
40. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 26F5.
41. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the light chain comprises a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 26F5.
42. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 26F5, and a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 26F5.
43. An isolated human antibody comprising: (a) human heavy chain framework regions, a human heavy chain CDR1 region comprising SEQ ID NO: 62, a human heavy chain CDR2 region comprising SEQ ID NO: 65, and a human heavy chain CDR3 region comprising SEQ ID NO: 68; and (b) human light chain framework regions, a human light chain CDR1 region comprising SEQ ID NO: 70, a human light chain CDR2 region comprising SEQ ID NO: 72, and a human light chain CDR3 region comprising SEQ ID NO: 74.
44. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 27F2, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 62, SEQ ID NO: 65, and SEQ ID NO: 68.
45. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the light chain comprises a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 27F2, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 70, SEQ ID NO: 72, and SEQ ID NO: 74.
46. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 27F2, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 62, SEQ ID NO: 65, and SEQ ID NO: 68; and a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 27F2, or an antigen-binding fragment thereof, wherein the antigen-binding fragment comprises at least SEQ ID NO: 70, SEQ ID NO: 72, and SEQ ID NO: 74.
47. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 27F2.
48. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), comprising a heavy chain and a light chain, wherein the light chain comprises a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 27F2.
49. An isolated antibody that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence identified in Figure 10 as corresponding to the heavy chain variable domain of 27F2, and a light chain variable region comprising the amino acid sequence identified in Figure 11 as corresponding to the light chain variable domain of 27F2.
50. The antibody according to any one of Claims 36 to 49 wherein the heavy chain and light chain are connected by a flexible linker to form a single-chain antibody.
51. The antibody of Claim 50, which is a single-chain Fv antibody.
52. The antibody according to any one of Claims 36 to 49 which is selected from the group consisting of: an Fab antibody fragment, an Fab’ antibody fragment, or an (Fab’)2 antibody fragment.
53. The antibody according to any one of Claims 36 to 49 wherein the antibody is a fully human antibody.
54. The antibody according to any one of Claims 36 to 53 wherein the antibody inhibits binding of IL-1 to its receptor.
55. The antibody according to any one of Claims 36 to 49 which is an IgG2 antibody.
56. The antibody according to any one of Claims 36 to 55 which binds specifically to the amino acid sequence of SEQ ID NO: 76.
57. The antibody according to any one of Claims 36 to 56 wherein the antibody binds specifically to a portion of the amino acid sequence of IL-1R1, wherein the portion of the amino acid sequence to which the antibody specifically binds comprises the amino acid sequence YSV.
58. A composition comprising a pharmaceutically acceptable carrier, excipients or diluent; and the antibody of any one of Claims 36 to 57.
59. A method for treating an IL-1 mediated disease in a patient, comprising administering to a patient in need thereof a therapeutically effective amount of the antibody of any one of Claims 36 to 57.
60. A method for treating an IL-1 mediated disease in a patient, comprising administering to a patient in need thereof a therapeutically effective amount of the composition of Claim 58.
61. The antibody according to any one of Claims 33 to 35 and 55 to 57, wherein the antibody is a human antibody.
62. An antibody according to any one of Claims 33 to 61, wherein the antibody specifically binds human interleukin-1 receptor type 1 (IL-1R1).
63. An isolated antibody according to any one of Claims 1 to 29 and 36 to 49 as herein before described with reference to the examples.
64. Use of a composition according to any one of Claims 31 or 58 in the preparation of a medicament for the treatment of an IL-1 mediated disease.
65. An isolated nucleic acid molecule comprising: (a) nucleotide sequences encoding human heavy chain framework regions, a nucleotide sequence encoding a human heavy chain CDR1 region comprising SEQ ID NO: 61, a nucleotide sequence encoding a human heavy chain CDR2 region comprising SEQ ID NO: 64, and a nucleotide sequence encoding a human heavy chain CDR3 region comprising SEQ ID NO: 67; and (b) nucleotide sequences encoding human light chain framework regions, a nucleotide sequence encoding a human light chain CDR1 region comprising SEQ ID NO: 70, a nucleotide sequence encoding a human light chain CDR2 region comprising SEQ ID NO: 72, and a nucleotide sequence encoding a human light chain CDR3 region comprising SEQ ID NO: 74.
66. An isolated nucleic acid molecule comprising: (a) nucleotide sequences encoding human heavy chain framework regions, a nucleotide sequence encoding a human heavy chain CDR1 region comprising SEQ ID NO: 62, a nucleotide sequence encoding a human heavy chain CDR2 region comprising SEQ ID NO: 65, and a nucleotide sequence encoding a human heavy chain CDR3 region comprising SEQ ID NO: 68; and (b) nucleotide sequences encoding human light chain framework regions, a nucleotide sequence encoding a human light chain CDR1 region comprising SEQ ID NO: 70, a nucleotide sequence encoding a human light chain CDR2 region comprising SEQ ID NO: 72, and a nucleotide sequence encoding a human light chain CDR3 region comprising SEQ ID NO: 74.
67. An isolated nucleic acid molecule comprising a nucleotide sequence encoding a human antibody or an antigen-binding fragment thereof that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody or fragment thereof comprises: (a) a heavy chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 9, or the amino acid sequences of SEQ ID NO: 61, SEQ ID NO: 64, and SEQ ID NO: 67; and (b) a light chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 11, or the amino acid sequences of SEQ ID NO: 70, SEQ ID NO: 72 and SEQ ID NO: 74.
68. An isolated nucleic acid molecule comprising a nucleotide sequence encoding a human antibody or an antigen-binding fragment thereof that specifically binds interleukin-1 receptor type 1 (IL-1R1), wherein the antibody or fragment thereof comprises: (a) a heavy chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 13, or the amino acid sequences of SEQ ID NO: 62, SEQ ID NO: 65, and SEQ ID NO: 68; and (b) a light chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 11, or the amino acid sequences of SEQ ID NO: 70, SEQ ID NO: 72 and SEQ ID NO: 74.
69. The isolated nucleic acid molecule of any one of Claims 65 to 68, wherein the nucleic acid molecule encodes an antibody that is an IgG2 antibody.
70. The isolated nucleic acid molecule of any one of Claims 65 to 68, wherein the nucleic acid molecule encodes an antibody that is a fully human antibody.
71. An expression vector comprising the nucleic acid molecule of any of Claims 65 to 70.
72. An isolated host cell comprising the expression vector of Claim 71.
73. The host cell of Claim 72 that is a eukaryotic cell.
74. The host cell of Claim 72 that is a prokaryotic cell.
75. A cell line comprising the expression vector of Claim 71.
76. A set of nucleic acid molecules comprising: (a) a first nucleic acid molecule encoding human heavy chain framework regions, a nucleotide sequence encoding a human heavy chain CDR1 region comprising SEQ ID NO: 61, a nucleotide sequence encoding a human heavy chain CDR2 region comprising SEQ ID NO: 64, and a nucleotide sequence encoding a human heavy chain CDR3 region comprising SEQ ID NO: 67; and (b) a second nucleic acid molecule encoding human light chain framework regions, a nucleotide sequence encoding a human light chain CDR1 region comprising SEQ ID NO: 70, a nucleotide sequence encoding a human light chain CDR2 region comprising SEQ ID NO: 72, and a nucleotide sequence encoding a human light chain CDR3 region comprising SEQ ID NO: 74.
77. A set of nucleic acid molecules comprising: (a) a first nucleic acid molecule encoding human heavy chain framework regions, a nucleotide sequence encoding a human heavy chain CDR1 region comprising SEQ ID NO: 62, a nucleotide sequence encoding a human heavy chain CDR2 region comprising SEQ ID NO: 65, and a nucleotide sequence encoding a human heavy chain CDR3 region comprising SEQ ID NO: 68; and (b) a second nucleic acid molecule encoding human light chain framework regions, a nucleotide sequence encoding a human light chain CDR1 region comprising SEQ ID NO: 70, a nucleotide sequence encoding a human light chain CDR2 region comprising SEQ ID NO: 72, and a nucleotide sequence encoding a human light chain CDR3 region comprising SEQ ID NO: 74.
78. A set of nucleic acid molecules comprising: (a) a first nucleic acid molecule encoding a human antibody heavy chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 9, or the amino acid sequences of SEQ ID NO: 61, SEQ ID NO: 64, and SEQ ID NO: 67; and (b) a second nucleic acid molecule encoding a human antibody light chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 11, or the amino acid sequences of SEQ ID NO: 70, SEQ ID NO: 72 and SEQ ID NO: 74.
79. A set of nucleic acid molecules comprising: (a) a first nucleic acid molecule encoding a human antibody heavy chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 13, or the amino acid sequences of SEQ ID NO: 62, SEQ ID NO: 65, and SEQ ID NO: 68; and (b) a second nucleic acid molecule encoding a human antibody light chain variable region comprising the amino acid sequence encoded by SEQ ID NO: 11, or the amino acid sequences of SEQ ID NO: 70, SEQ ID NO: 72 and SEQ ID NO: 74.
80. A set of expression vectors comprising the nucleic acid molecules of any of Claims 76 to 79, wherein one vector comprises the first nucleic acid molecule and the other vector comprises the second nucleic acid molecule.
81. An isolated host cell comprising the set of expression vectors of Claim 80.
82. The isolated host cell of Claim 81 that is a eukaryotic cell.
83. The isolated host cell of Claim 81 that is a prokaryotic cell.
84. An isolated human antibody that specifically binds to the third domain (SEQ ID NO: 76) of interleukin-1 receptor type 1 (IL-1R1) and inhibits binding to IL-Ιβ, but not significantly to IL-lra.
85. The antibody of Claim 84, wherein the antibody specifically binds to a conformational epitope within the third domain of IL-1R1.
86. The antibody of Claim 85, wherein the conformational epitope comprises the amino acid sequence YSV.
87. The antibody of any one of Claims 84 to 86, wherein the heavy chain and light chain are connected by a flexible linker to form a single-chain antibody.
88. The antibody of Claim 87, which is a single-chain Fv antibody.
89. The antibody of any one of Claims 84 to 86, which is a Fab, a Fab’, or a (Fab’)2 antibody fragment.
90. The antibody of any one of Claims 84 to 89, wherein the antibody is a fully human antibody.
91. The antibody of any one of Claims 84 to 90, which is an IgG2 antibody.
92. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of the antibody of any one of Claims 84 to 91.
AU2013204136A 2002-09-06 2013-04-12 Therapeutic Human Anti-IL-1R Monoclonal Antibody Expired AU2013204136B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013204136A AU2013204136B2 (en) 2002-09-06 2013-04-12 Therapeutic Human Anti-IL-1R Monoclonal Antibody

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US60/408,719 2002-09-06
AU2008207483A AU2008207483B2 (en) 2002-09-06 2008-08-22 Therapeutic Human Anti-IL-1R Monoclonal Antibody
AU2012202672A AU2012202672B2 (en) 2002-09-06 2012-05-08 Therapeutic Human Anti-IL-1R Monoclonal Antibody
AU2013204136A AU2013204136B2 (en) 2002-09-06 2013-04-12 Therapeutic Human Anti-IL-1R Monoclonal Antibody

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2012202672A Division AU2012202672B2 (en) 2002-09-06 2012-05-08 Therapeutic Human Anti-IL-1R Monoclonal Antibody

Publications (2)

Publication Number Publication Date
AU2013204136A1 AU2013204136A1 (en) 2013-05-09
AU2013204136B2 true AU2013204136B2 (en) 2017-07-13

Family

ID=48231038

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013204136A Expired AU2013204136B2 (en) 2002-09-06 2013-04-12 Therapeutic Human Anti-IL-1R Monoclonal Antibody

Country Status (1)

Country Link
AU (1) AU2013204136B2 (en)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Sims, J. et al. 1993 Proc. Natl. Acad. Sci. USA, vol. 90, pp. 6155-6159 *
Vigers, G. et al. 1997 Nature, vol. 386, pp. 190-194 *

Also Published As

Publication number Publication date
AU2013204136A1 (en) 2013-05-09

Similar Documents

Publication Publication Date Title
US9534053B2 (en) Therapeutic human anti-IL-1R1 monoclonal antibody
US20130164292A1 (en) Human Anti-OPGL Neutralizing Antibodies As Selective OPGL Pathway Inhibitors
AU2013204136B2 (en) Therapeutic Human Anti-IL-1R Monoclonal Antibody
AU2012202672B2 (en) Therapeutic Human Anti-IL-1R Monoclonal Antibody

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired