+ All Categories
Home > Documents > (19) - patentimages.storage.googleapis.com BE BG CH CY CZ DE DK EE ES FI FR GB GR ... PT RO SE SI SK...

(19) - patentimages.storage.googleapis.com BE BG CH CY CZ DE DK EE ES FI FR GB GR ... PT RO SE SI SK...

Date post: 18-Mar-2018
Category:
Upload: truonghuong
View: 219 times
Download: 3 times
Share this document with a friend
52
Note: Within nine months of the publication of the mention of the grant of the European patent in the European Patent Bulletin, any person may give notice to the European Patent Office of opposition to that patent, in accordance with the Implementing Regulations. Notice of opposition shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention). Printed by Jouve, 75001 PARIS (FR) (19) EP 2 331 078 B1 & (11) EP 2 331 078 B1 (12) EUROPEAN PATENT SPECIFICATION (45) Date of publication and mention of the grant of the patent: 19.09.2012 Bulletin 2012/38 (21) Application number: 09791868.4 (22) Date of filing: 25.08.2009 (51) Int Cl.: A61K 9/19 (2006.01) A61K 47/12 (2006.01) A61K 47/26 (2006.01) C07K 16/24 (2006.01) A61K 39/395 (2006.01) (86) International application number: PCT/US2009/054842 (87) International publication number: WO 2010/027766 (11.03.2010 Gazette 2010/10) (54) LYOPHILIZED FORMULATIONS OF ENGINEERED ANTI-IL-23P19 ANTIBODIES LYOPHILISIERTE FORMULIERUNGEN VON GENTECHNISCH VERÄNDERTEN ANTI-IL-23P19- ANTIKÖRPERN FORMULATIONS LYOPHILISEES D’ANTICORPS ANTI-IL-23P19 MODIFIES (84) Designated Contracting States: AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR (30) Priority: 27.08.2008 US 92326 P (43) Date of publication of application: 15.06.2011 Bulletin 2011/24 (73) Proprietor: Merck Sharp & Dohme Corp. Rahway, NJ 07065-0907 (US) (72) Inventors: LI, Xuhong North Potomac MD 20878 (US) KASHI, Ramesh, S. Warren NJ 07059 (US) BADKAR, Aniket Morris Plains NJ 07950 (US) (74) Representative: Horgan, James Michael Frederic et al Merck & Co., Inc. European Patent Department Hertford Road Hoddesdon Hertfordshire EN11 9BU (GB) (56) References cited: WO-A2-2008/088823 US-A1- 2007 218 064
Transcript

Note: Within nine months of the publication of the mention of the grant of the European patent in the European PatentBulletin, any person may give notice to the European Patent Office of opposition to that patent, in accordance with theImplementing Regulations. Notice of opposition shall not be deemed to have been filed until the opposition fee has beenpaid. (Art. 99(1) European Patent Convention).

Printed by Jouve, 75001 PARIS (FR)

(19)E

P2

331

078

B1

��&�� ���������(11) EP 2 331 078 B1

(12) EUROPEAN PATENT SPECIFICATION

(45) Date of publication and mention of the grant of the patent: 19.09.2012 Bulletin 2012/38

(21) Application number: 09791868.4

(22) Date of filing: 25.08.2009

(51) Int Cl.:A61K 9/19 (2006.01) A61K 47/12 (2006.01)

A61K 47/26 (2006.01) C07K 16/24 (2006.01)

A61K 39/395 (2006.01)

(86) International application number: PCT/US2009/054842

(87) International publication number: WO 2010/027766 (11.03.2010 Gazette 2010/10)

(54) LYOPHILIZED FORMULATIONS OF ENGINEERED ANTI-IL-23P19 ANTIBODIES

LYOPHILISIERTE FORMULIERUNGEN VON GENTECHNISCH VERÄNDERTEN ANTI-IL-23P19-ANTIKÖRPERN

FORMULATIONS LYOPHILISEES D’ANTICORPS ANTI-IL-23P19 MODIFIES

(84) Designated Contracting States: AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

(30) Priority: 27.08.2008 US 92326 P

(43) Date of publication of application: 15.06.2011 Bulletin 2011/24

(73) Proprietor: Merck Sharp & Dohme Corp.Rahway, NJ 07065-0907 (US)

(72) Inventors: • LI, Xuhong

North PotomacMD 20878 (US)

• KASHI, Ramesh, S.WarrenNJ 07059 (US)

• BADKAR, AniketMorris PlainsNJ 07950 (US)

(74) Representative: Horgan, James Michael Frederic et alMerck & Co., Inc. European Patent Department Hertford RoadHoddesdonHertfordshire EN11 9BU (GB)

(56) References cited: WO-A2-2008/088823 US-A1- 2007 218 064

EP 2 331 078 B1

2

5

10

15

20

25

30

35

40

45

50

55

Description

FIELD OF THE INVENTION

[0001] The present invention relates generally to lyophilized formulations of therapeutic antibodies.

BACKGROUND OF THE INVENTION

[0002] Interleukin-23 (IL-23) is a heterodimeric cytokine comprised of two subunits, p19 which is unique to IL-23, andp40, which is shared with IL-12. The p19 subunit is structurally related to IL-6, granulocyte-colony stimulating factor (G-CSF), and the p35 subunit of IL-12. IL-23 mediates signaling by binding to a heterodimeric receptor, comprised of IL-23R and IL-12β1, which is shared by the IL-12 receptor. A number of early studies demonstrated that the consequencesof a genetic deficiency in p40 (p40 knockout mouse; p40KO mouse) were more severe than those found in a p35KOmouse. Some of these results were eventually explained by the discovery of IL-23, and the finding that the p40KOprevents expression of not only IL-12, but also of IL-23. See, e.g., Oppmann et al. (2000) Immunity 13:715-725; Wiekowskiet al. (2001) J. Immunol. 166:7563-7570; Parham et al. (2002) J. Immunol. 168:5699-708; Frucht (2002) Sci STKE 2002,E1-E3; Elkins et al. (2002) Infection Immunity 70:1936-1948).[0003] Recent studies, through the use of p40 KO mice, have shown that blockade of both IL-23 and IL-12 is aneffective treatment for various inflammatory and autoimmune disorders. However, the blockade of IL-12 through p40appears to have various systemic consequences such as increased susceptibility to opportunistic microbial infections.Bowman et al. (2006) Curr. Opin. Infect. Dis. 19:245. Accordingly, specific blockade of the p19 subunit of IL-23 is preferredin the treatment of human disease because it interferes with the activity of IL-23 without interfering with the activity of IL-12.[0004] Therapeutic antibodies may be used to block cytokine activity. A significant limitation in using antibodies as atherapeutic agent in vivo is the immunogenicity of the antibodies. As most monoclonal antibodies are derived from non-human species, repeated use in humans results in the generation of an immune response against the therapeuticantibody. Such an immune response results in a loss of therapeutic efficacy at a minimum, and potentially a fatalanaphylactic response. Accordingly, antibodies of reduced immunogenicity in humans, such as humanized or fully humanantibodies, are preferred for treatment of human subjects: Exemplary therapeutic antibodies to IL-23p19 are disclosedin U.S. Patent Application Publication No. 2007/0009526, and in International Patent Publication Nos. WO 2007/076524,WO 2007/024846; WO 2007/147019, and WO 2009/043933. Additional humanized anti-1L=23p19 antibodies are dis-closed in commonly assigned applications published as International Patent Publication Nos. WO 2008/103432 and WO2008/103473, and in commonly-assigned U.S. Patent Application Publication No. 2007/0048315.[0005] Antibodies for use in human subjects must be stored prior to use and transported to the point of administration.Reproducibly attaining a desired level of antibody drug in a subject requires that the drug be stored in a formulation thatmaintains the bioactivity of the drug. The need exists for formulations of anti-human IL-23p19 antibodies for use, e.g.,in treatment of inflammatory, autoimmune, and proliferative disorders. Preferably, such formulations will exhibit a longhalf-life, be stable when stored and transported, and will be amenable to admnistration at high concentration, e.g. foruse in subcutaneous administration, and low concentrations, e.g. for intravenous administration.

SUMMARY OF THE INVENTION

[0006] The present invention providers lyophilized formulations of binding compounds that bind to human IL-23p19,which binding compounds are defined as human or humanized anti-human IL-23p19 antibodies, or antigen-bindingfragments thereof.[0007] In one embodiment, the lyophilized formulation is made by lyophilizing an aqueous solution which comprises5-25mg/mL human anti-IL-23p19 antibody (or antigen-binding fragment thereof), about 2.5mM sodium citrate at pH4.4-5.2, about 0.05 mg/mL polysorbate 80 and about 50mM sucrose. In various embodiments the pH of the formulationafter reconstitution with water is 4.8 (�0.4), or 4.8 (�0.2), such as between 4.6 and 5.0, e.g. about 4.4, 4.6, 4.7, 4.8,4.9, 5.0 or 5.2.[0008] In some embodiments, the lyophilized formulation enables reconstitution of the antibody (or antigen bindingfragment thereof) at a concentration of about 25 mg/mL or higher, about 50 mg/mL or higher, about 75 mg/mL or higheror about 100 mg/mL.In one embodiment, the prelyophilization solution comprises antibody, or antigen-binding fragment thereof, at about 25mg/mL. In one embodiment, the pre-lyophlization solution is about pH 4.8.[0009] In yet other embodiments, the lyophilized formulation of anti-human IL-23p19 antibody, or antigen bindingfragment thereof, of the present invention,when reconstituted, comprises 25 - 100 mg/mL anti-human IL-23p19 antibody,or antigen binding fragment thereof; about 200 mM sucrose; about 0.2 mg/mL polysorbate 80; and about 10 mM citratebuffer at pH 4.4 to 5.2. In one embodiment, the reconstituted solutions comprises antibody, or antigen-binding fragment

EP 2 331 078 B1

3

5

10

15

20

25

30

35

40

45

50

55

thereof, at about 100 mg/mL. In one embodiment, the reconstituted solution is about pH4.8.[0010] In still further embodiments, the lyophilized formulation is provided in a glass vial. In various embodiments, theglass vial contains about 5,10,15, 20,25 ,30 ,40, 40, 50,60 67.5, 75, 100, 150, 200, 300, 400, 500 mg/vial or more.[0011] Exemplary binding compounds for use in the lyophilized formulation of the present invention comprise anantibody light chain variable domain, or antigen binding fragment thereof, having one, two or three CDRs selected fromthe group consisting of SEQ ID NOs 32-46. In one embodiment, the binding compound of the present invention comprisesa light chain variable domain comprising CDRL1 selected from the group consisting of SEQ ID NOs: 32-36, a CDRL2selected from the group consisting of SEQ ID NOs:37-41;and a CDRL3 selectedf rom the group consisting of SEQ IDNOs: 4.2-46,[0012] In one embodiment, the binding compound for use in the lyophilized formulations of the present inventioncomprises an antibody heavy chain variable domain, or antigen binding fragment thereof, having one, two or three CDRsselected from the group consisting of SEQ ID NOs:15-31. In one embodiment, the binding compound of the presentinvention comprises a heavy chain variable domain comprising a CDRH1 selected from the group consisting of SEQ IDNOs: 15-19; a CDRH2 selected from the group consisting of SEQ ID NOs: 20-26; and a CDRH3 selected from the groupconsisting of SEQ ID NOs: 27-31.[0013] In some embodiments the light chain and/or heavy chain variable domains comprise a variant of one or moreof the CDRs. In various embodiments the variant domain comprises up to 1, 2, 3, 4, 5 or more conservatively modifiedamino acid residues relative to the sequence of the respective SEQ ID NOs. Conservative amino acid substitutions areprovided at Table 1.[0014] In some embodiments the light chain variable domain comprises residues 1-108 of SEQ ID NO: 14 or a variantthereof. In some embodiments the heavy chain variable domain comprises a sequence selected from the group consistingof residues 1-116 of SEQ ID NOs: 6-8, i.e. SEQ ID NO: 6, SEQ ID NO: 7 or SEQ ID NO: 8. In various embodiments thevariant variable domain comprises up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30,40 or 50 or more conservatively modifiedamino acid residues relative to the sequence of the respective SEQ ID NOs. In yet a further embodiment, the bindingcompound comprises a light chain variable domain and a heavy chain variable domain, or the antigen binding fragmentsthereof, described in this paragraph.[0015] In one embodiment the binding compound comprises a light chain sequence of SEQ ID NO: 14 and/or a heavychain sequence selected from the group consisting of SEQ ID NOs: 6-8.[0016] In other embodiments the binding compound of the present invention comprises a light chain variable domain,or an antigen binding fragment thereof, consisting essentially of residues 1-108 of SEQ ID NO: 14, and/or a heavy chainvariable domain, or an antigen binding fragment thereof, consisting essentially of a sequence selected from the groupconsisting of residues 1-116 of SEQ ID NOs: 6-8, such as SEQ ID NO: 6, SEQ ID NO: 7 or SEQ ID NO: 8.[0017] In other embodiments the binding compound of the present invention comprises a light chain variable domain,or an antigen binding fragment thereof, having at least 75%, 90%, 95%, 98% or 99% sequence homology with residues1-108 of SEQ ID NO: 14, and/or a heavy chain variable domain, or an antigen binding fragment thereof, having at least75%, 90%, 95%, 98% or 99% sequence homology with a sequence selected from the group consisting of residues 1-116of SEQ ID NOs: 6-8, such as SEQ ID NO: 6, SEQ ID NO: 7 or SEQ ID NO: 8.[0018] In some embodiments, the binding compound of the present invention further comprises a heavy chain com-prising a γ1, γ2, γ3, or γ4 human heavy chain constant region or a variant thereof. In various embodiments the bindingcompound comprises a light chain comprising a lambda or a kappa human light chain constant region.[0019] In various embodiments the binding compound of the present invention is an antibody fragment selected fromthe group consisting of, e.g., Fab, Fab’, Fab’-SH, Fv, scFv, F(ab’)2, and a diabody.[0020] In other embodiments the invention relates to a lyophilized formulation of a human or humanized anti-IL-23p19antibody, or antigen binding fragment thereof, for use in treating disorders including, but not limited to, inflammatorydisease, autoimmune disease, cancer, infectious disease (e.g. bacterial, mycobacterial, viral or fungal infection, includingchronic infections), arthritis, psoriasis, inflammatory bowel disease, multiple sclerosis, uveitis, systemic lupus erythema-tosus and diabetes.[0021] The present invention provides a vessel (e.g., a glass vial) comprising any of the lyophilized formulations setforth herein. The present invention also provides an injection device (e.g., hypodermic needle and syringe, autoinjector,lyophilization cartridge) comprising a diluent and lyophilized formulation of a human or humanized anti-IL-23p 19 antibody,or antigen binding fragment thereof.

BRIEF DESCRIPTION OF THE DRAWINGS

[0022] FIG. 1 provides a flow diagram of a manufacturing process for the lyophilized formulation of an anti-1L-23p19antibody of the present invention. The process is described more fully at Example 1, infra.[0023] FIG. 2 shows stability data (18 months) for lyophilized formulations of a humanized anti-human IL-23p19antibody at pH 5.5 stored at 5°C, as discussed in greater detail in Example 2.

EP 2 331 078 B1

4

5

10

15

20

25

30

35

40

45

50

55

[0024] FIG. 3 shows stability data (18 months) for lyophilized formulations of a humanized anti-human IL-23p19antibody at pH 5.5 stored at 25H, as discussed in greater detail in Example 2.[0025] FIG. 4 shows stability data (9 months) for lyophilized formulations of a humanized anti-human IL-23p19 antibodyat pH 5.5 stored at RH4, as discussed in greater detail in Example 2.[0026] FIG. 5 shows stability data (12 months) for lyophilized formulations of a humanized anti-human IL-23p 19antibody at pH 4.8 stored at 5°C, as discussed in greater detail in Example 2.[0027] FIG. 6 shows stability data (12 months) for lyophilized formulations of a humanized anti-human IL-23p19antibody at pH 4.8 stored at 25H, as discussed in greater detail in Example 2.[0028] FIG. 7 shows stability data (3 months) for lyophilized formulations of a humanized anti-human IL-23pl9 antibodyat pH 4.8 stored at RH4, as discussed in greater detail in Example 2.

DETAILED DESCRIPTION

[0029] As used herein, including the appended claims, the singular forms of words such as "a," "an," and "the," includetheir corresponding plural references unless the context clearly dictates otherwise. Table 6 below provides a listing ofsequence identifiers used in this application. Unless otherwise indicated,the proteins and-subjects-referred to hereinare human proteins and subject, rather than another species.[0030] The present invention provides lyophilized formulations of engineered anti-IL-23 antibodies and uses thereofto treat inflammatory, autoimmune, and proliferative disorders. In some embodiments, the lyophilized formulations ofthe present invention comprise a humanized anti-IL-23p19 antibody, or binding fragment thereof, as disclosed in co-pending, commonly assigned International Patent Publication No. WO 2008/103432

1. Definitions

[0031] "Proliferative activity" encompasses an activity that promotes, that is necessary for, or that is specifically as-sociated with, e.g., normal cell division, as well as cancer, tumors, dysplasia, cell transformation, metastasis, and ang-iogenesis.[0032] As used herein, the term "antibody" refers to any form of antibody that exhibits the desired biological activity.Thus, it is used in the broadest sense and specifically covers monoclonal antibodies (including full length monoclonalantibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), chimeric antibodies, humanizedantibodies, fully human antibodies, etc. so long as they exhibit the desired biological activity.[0033] As used herein, the terms "IL-23p19 binding fragment," "antigen binding fragment thereof," "binding fragmentthereof or "fragment thereof encompass a fragment or a derivative of an antibody that still substantially retains itsbiological activity of binding to antigen (human IL-23p19) and inhibiting its activity. Therefore, the term "antibody fragment"or IL-23p19 binding fragment refers to a portion of a full length antibody, generally the antigen binding or variable regionthereof. Examples of antibody fragments include Fab, Fab’, F(ab’)2, and Fv fragments; diabodies; linear antibodies;single-chain antibody molecules, e.g., sc-Fv; and multispecific antibodies formed from antibody fragments. Typically, abinding fragment or derivative retains at least 10% of its IL-23p19 inhibitory activity. Preferably, a binding fragment orderivative retains at least 25%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or 100% (or more) of its IL-23p19 inhibitoryactivity, although any binding fragment with sufficient affinity to exert the desired biological effect will be useful. It is alsointended that a IL-23p19 binding fragment can include variants having conservative amino acid substitutions that do notsubstantially alter its biologic activity.[0034] A "domain antibody" is an immunologically functional immunoglobulin fragment containing only the variableregion of a heavy chain or the variable region of a light chain. In some instances, two or more VH regions are covalentlyjoined with a peptide linker to create a bivalent domain antibody. The two VH regions of a bivalent domain antibody maytarget the same or different antigens.[0035] A "bivalent antibody" comprises two antigen binding sites. In some instances, the two binding sites have thesame antigen specificities. However, bivalent antibodies may be bispecific. As used herein, the term "bispecific antibody"refers to an antibody, typically a monoclonal antibody, having binding specificities for at least two different antigenicepitopes, e.g., IL-23p19 and IL-17. In one embodiment, the epitopes are from the same antigen. In another embodiment,the epitopes are from two different antigens. Methods for making bispecific antibodies are known in the art. For example,bispecific antibodies can be produced recombinantly using the co-expression of two immunoglobulin heavy chain/lightchain pairs. See, e.g., Milstein et al. (1983) Nature 305: 537-39. Alternatively, bispecific antibodies can be preparedusing chemical linkage. See, e.g., Brennan et al. (1985) Science 229:81. Bispecific antibodies include bispecific antibodyfragments. See, e.g., Holliger et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90:6444-48, Gruber et al. (1994) J. Immunol.152:5368.[0036] As used herein, the term "single-chain Fv" or "scFv" antibody refers to antibody fragments comprising the VHand VL domains of antibody, wherein these domains are present in a single polypeptide chain. Generally, the Fv polypep-

EP 2 331 078 B1

5

5

10

15

20

25

30

35

40

45

50

55

tide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desiredstructure for antigen binding. For a review of sFv, see Pluckthun (1994) THE PHARMACOLOGY OF MONOCLONALANTIBODIES, vol. 113, Rosenburg and Moore eds. Springer-Verlag, New York, pp. 269-315.[0037] The monoclonal antibodies herein also include camelized single domain antibodies. See, e.g., Muyldermanset al. (2001) Trends Biochem. Sci. 26:230; Reichmann et al. (1999) J. Immunol. Methods 231:25; WO 94/04678; WO94/25591; U.S. Pat. No. 6,005,079). In one embodiment, the present invention provides single domain antibodies com-prising two VH domains with modifications such that single domain antibodies are formed.[0038] As used herein, the term "diabodies" refers to small antibody fragments with two antigen-binding sites, whichfragments comprise a heavy chain variable domain (VH) connected to a light chain variable domain (VL) in the samepolypeptide chain (VH-VL or VL-VH). By using a linker that is too short to allow pairing between the two domains on thesame chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, e.g., EP 404,097; WO 93/11161; and Holliger et al. (1993) Proc.Natl. Acad. Sci. USA 90: 6444-6448. For a review of engineered antibody variants generally see Holliger and Hudson(2005) Nat. Biotechnol. 23:1126-1136.[0039] As used herein, the term "humanized antibody" refers to forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived fromnon-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, andtypically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc),typically that of a human immunoglobulin. The prefix "hum", "hu" or "h" is added to antibody clone designations whennecessary to distinguish humanized antibodies (e.g. hum13B8) from parental rodent antibodies (e.g. mouse 13B8, orm13B8). The humanized forms of rodent antibodies will generally comprise the same CDR sequences of the parentalrodent antibodies, although certain amino acid substitutions may be included to increase affinity, increase stability ofthe humanized antibody, or for other reasons.[0040] The antibodies of the present invention also include antibodies with modified (or blocked) Fc regions to providealtered effector functions. See, e.g., U.S. Pat. No. 5,624,821; W02003/086310; WO2005/120571; WO2006/0057702;Presta (2006) Adv. Drug Delivery Rev. 58:640-656. Such modification can be used to enhance or suppress variousreactions of the immune system, with possible beneficial effects in diagnosis and therapy. Alterations of the Fc regioninclude amino acid changes (substitutions, deletions and insertions), glycosylation or deglycosylation, and adding multipleFc. Changes to the Fc can also alter the half-life of antibodies in therapeutic antibodies, and a longer half-life wouldresult in less frequent dosing, with the concomitant increased convenience and decreased use of material. See Presta(2005) J. Allergy Clin.Immunol.116:731 at 734-35.[0041] The term "fully human antibody" refers to an antibody that comprises human immunoglobulin protein sequencesonly. A fully human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in ahybridoma derived from a mouse cell. Similarly, "mouse antibody" refers to an antibody which comprises mouse immu-noglobulin sequences only. A fully human antibody may be generated in a human being, in a transgenic animal havinghuman immunoglobulin germline sequences, by phage display or other molecular biological methods.[0042] As used herein, the term "hypervariable region" refers to the amino acid residues of an antibody that areresponsible for antigen-binding. The hypervariable region comprises amino acid residues from a "complementaritydetermining region" or "CDR" (e.g. residues 24-34 (CDRL1), 50-56 (CDRL2) and 89-97 (CDRL3) in the light chainvariable domain and residues 31-35 (CDRH1), 50-65 (CDRH2) and 95-102 (CDRH3) in the heavy chain variable domain(Kabat et al. (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutesof Health, Bethesda, Md.) and/or those residues from a "hypervariable loop" (i.e. residues 26-32 (L1), 50-52 (L2) and91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variabledomain (Chothia and Lesk (1987) J. Mol. Biol. 196: 901-917). As used herein, the term "framework" or "FR" residuesrefers to those variable domain residues other than the hypervariable region residues defined herein as CDR residues.The residue numbering above relates to the Kabat numbering system and does not necessarily correspond in detail tothe sequence numbering in the accompanying Sequence Listing.[0043] "Binding compound," as used herein, refers to a human or humanized antibody that binds to human IL-23p 19,or any antigen-binding fragment or derivative of such antibody.[0044] "Conservatively modified variants" or "conservative substitution" refers to substitutions of amino acids areknown to those of skill in this art and may be made generally without altering the biological activity of the resultingmolecule, even in essential regions of the polypeptide. Such exemplary substitutions are preferably made in accordancewith those set forth in Table 1 as follows:

EP 2 331 078 B1

6

5

10

15

20

25

30

35

40

45

50

55

[0045] In addition, those of skill in this art recognize that, in general, single amino acid substitutions in non-essentialregions of a polypeptide do not substantially alter biological activity. See, e.g., Watson et al. (1987) Molecular Biologyof the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Edition).[0046] The phrase "consists essentially of," or variations such as "consist essentially of" or "consisting essentially of,"as used throughout the specification and claims, indicate the inclusion of any recited elements or group of elements,and the optional inclusion of other elements, of similar or different nature than the recited elements, that do not materiallychange the basic or novel properties of the specified dosage regimen, method, or composition. As a non-limiting example,a binding compound that consists essentially of a recited amino acid sequence may also include one or more aminoacids, including substitutions of one or more amino acid residues, that do not materially affect the properties of thebinding compound.[0047] "Immune condition" or "immune disorder" encompasses, e.g., pathological inflammation, an inflammatory dis-order, and an autoimmune disorder or disease. "Immune condition" also refers to infections, persistent infections, andproliferative conditions, such as cancer, tumors, and angiogenesis, including infections, tumors, and cancers that resisteradication by the immune system. "Cancerous condition" includes, e.g., cancer, cancer cells, tumors, angiogenesis,and precancerous conditions such as dysplasia.[0048] "Inflammatory disorder" means a disorder or pathological condition where the pathology results, in whole or inpart, from, e.g., a change in number, change in rate of migration, or change in activation, of cells of the immune system.Cells of the immune system include, e.g., T cells, B cells, monocytes or macrophages, antigen presenting cells (APCs),dendritic cells, microglia, NK cells, NKT cells, neutrophils, eosinophils, mast cells, or any other cell specifically associatedwith the immunology, for example, cytokine-producing endothelial or epithelial cells.[0049] The antibody, or binding composition derived from the antigen-binding site of an antibody, of the contemplatedmethod binds to its antigen with an affinity that is at least two fold greater, preferably at least ten times greater, morepreferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with unrelated

Table 1

Exemplary Conservative Amino Acid Substitutions

Original residue Conservative substitution

Ala (A) Gly; Ser

Arg (R) Lys, His

Asn (N) Gln; His

Asp (D) Glu; Asn

Cys (C) Ser; Ala

Gln (Q) Asn

Glu (E) Asp; Gln

Gly (G) Ala

His (H) Asn, Gln

Ile (I) Leu; Val

Leu (L) Ile; Val

Lys (K) Arg; His

Met (M) Leu; Ile, Tyr

Phe (F) Tyr; Met; Leu

Pro (P) Ala

Ser (S) Thr

Thr (T) Ser

Trp (W) Tyr; Phe

Tyr (Y) Trp; Phe

Val (V) Ile; Leu

EP 2 331 078 B1

7

5

10

15

20

25

30

35

40

45

50

55

antigens. In a preferred embodiment the antibody will have an affinity that is greater than about 109 liters/mol, asdetermined, e.g., by Scatchard analysis. Munsen et al. (1980) Analyt. Biochem. 107:220-239.[0050] A "reconstituted" formulation is one that has been prepared by dissolving a lyophilized protein formulation in adiluent such that the protein is dispersed in the reconstituted formulation. The reconstituted formulation is suitable foradministration, e.g. parenteral administration), and may optionally be suitable for subcutaneous administration.[0051] An "isotonic" formulation has essentially the same osmotic pressure as human blood. Isotonic formulations willgenerally have an osmotic pressure from about 250 to 350 mOsm. Isotonicity can be measured, for example, using avapor pressure or ice-freezing type osmometer.

II. Human or Humanized Anti-IL-23p19 Antibodies

[0052] The lyophilized formulation of the present invention may be used with antibodies generally, including humanor humanized anti-human IL-23p19 antibodies, such as those disclosed herein. Humanized forms of anti-human IL-23p19 antibody 13B8 are provided. A hybridoma expressing antibody 13B8 was deposited pursuant to the BudapestTreaty with American Type Culture Collection (ATCC - Manassas, Virginia, USA) on August 17, 2006 under AccessionNumber PTA-7803. Humanized forms of other antibodies disclosed herein may be constructed by substituting the humanframeworks disclosed for the humanized 13B8 antibody. Substitution with the human frameworks disclosed herein aspart of humanized antibody 13B8 is most appropriate for antibodies with CDR sequences similar to 13B8.[0053] Sequences are provided for anti-human IL-23p19 antibodies m1A11, m11C1, m5F5, m21D1, m13B8, h13B8a,h13B8b and h13B8c. CDRs are provided under separate sequence identifiers, as indicated in Table 6. When referringto the antibodies, an "m" prefix connotes a murine antibody and an "h" connotes a humanized antibody. The suffixes"a", "b" and "c" refer to sequence variants of the humanized 13B8 heavy chain variable domain, as discussed in greaterdetail below.[0054] Ordinarily, amino acid sequence variants of the humanized anti-IL-23 antibody will have an amino acid sequencehaving at least 75% amino acid sequence identity with the original humanized antibody amino acid sequences of eitherthe heavy or the light chain more preferably at least 80%, more preferably at least 85%, more preferably at least 90%,and most preferably at least 95, 98, or 99%. Identity or homology with respect to this sequence is defined herein as thepercentage of amino acid residues in the candidate sequence that are identical with the humanized anti-IL-23 residues,after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity,and not considering any conservative substitutions as part of the sequence identity. None of N-terminal, C-terminal, orinternal extensions, deletions, or insertions into the antibody sequence shall be construed as affecting sequence identityor homology.[0055] Structure-function data are provided herein for anti-IL-23p19 antibodies of the present invention as follows.One of skill in the art would recognize that alteration of the CDR sequences would be expected to have the most dramaticeffects on antigen-binding affinity. Murphy et al., JANEWAY’S IMMUNOBIOLOGY, Seventh Ed., 2008, Chapter 3. TheCDR regions for the anti-1L-23p19 antibodies of the present invention are provided in the sequence listing. In addition,comparison of the antibodies disclosed herein to each other can be used to determine which residues are most criticalto antigen binding, and thus biological activity. In addition, the invention provides for several sequence variants for the13B8 antibody, including heavy chain variants 13B8 HC-a, 13B8 HC-b and 13B8 HC-c, providing the original murineCDRH2 sequence (13B8 HC-a) and two variants thereof. See Table 2.[0056] The human or humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG,IgD, IgA, and IgE. Preferably, the antibody is an IgG antibody. Any isotype of IgG can be used, including IgG1, IgG2,IgG3, and IgG4. Different constant domains may be appended to the humanized VL and VH regions provided herein. Forexample, if a particular intended use of an antibody (or fragment) of the present invention were to call for altered effectorfunctions, a heavy chain constant domain other than IgG I may be used. Although IgG1 antibodies provide for long half-life and for effector functions, such as complement activation and antibody-dependent cellular cytotoxicity, such activitiesmay not be desirable for all uses of the antibody. In such instances an IgG4 constant domain, for example, may be used.[0057] Likewise, either class of light chain can be used in the compositions and methods herein. Specifically, kappa,lambda, or variants thereof are useful in the present compositions and methods.[0058] CDR and FR residues are determined according to the standard sequence definition of Kabat. Kabat et al.(1987) Sequences of Proteins of Immunological Interest, National Institutes of Health, Bethesda Md. SEQ ID NOs: 1-5show the heavy chain variable domain sequences of various mouse anti-human IL-23p19 antibodies, and SEQ ID NOs:9-13 depict the light chain variable domain sequences. FIGS. 1 and 2 provide sequence lineups of heavy and light chainvariable domains of the various antibodies of the present invention. CDRs are indicated in the figures, and the individualCDR sequences are each presented with unique Sequence Identifiers as indicated in Table 6.[0059] Humanized forms of antibody 13B8 are provided. The humanized light chain 13B8 sequence (with kappaconstant region) is provided at SEQ ID NO: 14, and the light chain variable domain comprises residues 1-108 of thatsequence. Three versions of the humanized heavy chain 13B8 sequence (with γl constant regions) are provided at SEQ

EP 2 331 078 B1

8

5

10

15

20

25

30

35

40

45

50

55

ID NOs: 6-8, and the heavy chain variable domain comprises residues 1-116 of those sequences. The 13B8 heavychains variants are illustrated at Table 2, with differences from the parental sequence noted in bold. The Met (M) wasmodified to Lys (K) to avoid the potential for oxidation of the residue and inactivation of the antibody. The substitutionof AQKLQ for NEMFE is a replacement of the murine CDR sequence with the human germline sequence from the humanframework selected to humanize the antibody.

[0060] Humanized forms of the other antibodies disclosed herein may be created by simply substituting the parentalrodent antibody CDRs into the light and heavy chain sequences for humanized 13B8 provided at SEQ ID NOs: 14 and6. This approach is most likely to be successful for antibody chains with CDRs having high homology with the CDRs ofantibody 13B8, e.g. clone 11C1 on the heavy chain and clones 11C1 and 21D1 on the light chain. Alternatively, themurine antibodies may be independently humanized using the approaches outlines herein, e.g. at Example 1.[0061] In one embodiment, CDRs include variants of any single sequence CDR disclosed herein (SEQ ID NOs: 15-46),in which the variant comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative amino acid substitutions relative to thedisclosed sequence, as determined using the data of Table 1.[0062] Heavy and Light chain sequences (SEQ ID NOs: 6-8 and 16) are provided without signal sequences. Exemplaryheavy and light chain signal sequences are provided at SEQ ID NOs: 51 and 52, respectively. The signal sequences,or nucleic acid sequences encoding the signal sequences, may be appended to the N-terminus of the respective antibodychains to create a precursor protein for secretion from a host cell. Alternative signal sequences may also be used, andseveral can be found at "SPdb: a Signal Peptide Database." Choo et al. (2005) BMC Bioinformatics 6:249.

III. Biological Activity of Humanized Anti-IL-23

[0063] Inflammatory diseases of the skin, joints, CNS, as well as proliferative disorders elicit similar immune responses,thus IL-23 blockade should provide inhibition of these immune mediated inflammatory disorders, without comprising thehost ability to fight systemic infections. Antagonizing IL-23 should relieve the inflammation associated with inflammatorybowel disease, Crohn’s disease, Ulcerative Colitis, rheumatoid arthritis, psoriatic arthritis, psoriasis, ankylosing spond-ylitis, and atopic dermatitis. Use of IL-23 inhibitors will also provide inhibition of proliferative disorders, e.g., cancer andautoimmune disorders, e.g., multiple sclerosis, type I diabetes, and SLE. Descriptions of IL-23 in these various disorderscan be found in the following published PCT applications: WO 04/081190; WO 04/071517; WO 00/53631; and WO01/18051. IL-23 inhibitors may also find use in treatment of infections, including chronic infections, such as bacterial,mycobacterial, viral and fungal infections.[0064] The lyophilized formulations of the present invention include antibodies and fragments thereof that are biolog-ically active when reconstituted. As used herein, the term "biologically active" refers to an antibody or antibody fragmentthat is capable of binding the desired the antigenic epitope and directly or indirectly exerting a biologic effect. Typically,these effects result from the failure of IL-23 to bind its receptor. As used herein, the term "specific" refers to the selectivebinding of the antibody to the target antigen epitope. Antibodies can be tested for specificity of binding by comparingbinding to IL-23 to binding to irrelevant antigen or antigen mixture under a given set of conditions. If the antibody bindsto IL-23 at least 10, and preferably 50 times more than to irrelevant antigen or antigen mixture then it is considered tobe specific. An antibody that binds to IL-12 is not an IL-23-specific antibody. An antibody that "specifically binds" to IL-23p19 does not bind to proteins that do not comprise the IL-23p19-derived sequences, i.e. "specificity" as used hereinrelates to IL-23p19 specificity, and not any other sequences that may be present in the protein in question. For example,as used herein, an antibody that "specifically binds" to IL-23p19 will typically bind to FLAG®-hIL-23p19, which is a fusionprotein comprising IL-23p19 and a FLAG® peptide tag, but it does not bind to the FLAG® peptide tag alone or when itis fused to a protein other than IL-23p19.[0065] IL-23-specific binding compounds of the present invention, such as inhibitory 1L-23p19 specific antibodies,can inhibit its biological activity in any manner, including but not limited to production of IL-1β and TNF by peritonealmacrophages and IL-17 by TH 17 T cells. See Langrish et al. (2004) Immunol. Rev. 202:96-105. Anti-IL-23p19 antibodieswill also be able to inhibit the gene expression of IL-17A, IL-17F, CCL7, CCL17, CCL20, CCL22, CCR1, and GM-CSF.

Table 2

Antibody 13B8 CDRH2 Variants

Antibody CDRH2 Sequence SEQ ID NO:

m13B8, h13B8-a QIFPASGSADYNEMFEG 24

h13B8-b QIFPASGSADYNEKFEG 25

h13B8-c QIFPASGSADYAQKLQG 26

EP 2 331 078 B1

9

5

10

15

20

25

30

35

40

45

50

55

See Langrish et al. (2005) J. Exp. Med. 201:233-240. IL-23-specific binding compounds of the present invention, suchas anti IL-23p19 antibodies, will also block the ability of IL-23 to enhance proliferation or survival of TH17 cells. Cua andKastelein (2006) Nat. Immunol. 7:557-559. The inhibitory activity of engineered anti-IL-23p19 will be useful in the treatmentof inflammatory, autoimmune, and proliferative disorders. Examples of such disorders are described in PCT patentapplication publications WO 04/081190; WO 04/071517; WO 00/53631; and WO 01/18051.[0066] The formulation of the present invention is useful, for example, for storage and transport of human or humanizedanti-IL-23p19 antibodies (or antigen binding fragments thereof) for use in treatment or prevention of a disorder associatedwith elevated activity of IL-23 or IL-23p19, such as Th17-mediated diseases, autoimmune or chronic inflammatorydisorders, or cancers.

IV. Lyophilized Pharmaceutical Compositions

[0067] Lyophilized formulations of therapeutic proteins provide several advantages. Lyophilized formulations in generaloffer better chemical stability than solution formulations, and thus increased half-life. A lyophilized formulation may alsobe reconstituted at different concentrations depending on clinical factors, such as route of administration or dosing. Forexample, a lyophilized formulation may be reconstituted at a high concentration (i.e. in a small volume) if necessary forsubcutaneous administration, or at a lower concentration if administered intravenously. High concentrations may alsobe necessary if high dosing is required for a particular subject, particularly if administered subcutaneously where injectionvolume must be minimized. One such lyophilized antibody formulation is disclosed at U.S. Pat. No. 6,267,958. Lyophilizedformulations of another therapeutic protein are disclosed at U.S. Pat. No. 7,247,707.[0068] Typically the lyophilized formulation is prepared in anticipation of reconstitution at high concentration of drugproduct (DP, in this case human or humanized anti-IL-23p19 antibody, or antigen binding fragment thereof), i.e. inanticipation of reconstitution in a low volume of water. Subsequent dilution with water or isotonic buffer can then readilybe used to dilute the DP to a lower concentration. Typically, excipients are included in a lyophilized formulation of thepresent invention at levels that will result in a roughly isotonic formulation when reconstituted at high DP concentration,e.g. for subcutaneous administration. Reconstitution in a larger volume of water to give a lower DP concentration willnecessarily reduce the tonicity of the reconstituted solution, but such reduction may be of little significance in non-subcutaneous, e.g. intravenous, administration. If isotonicity is desired at lower DP concentration, the lyophilized powdermay be reconstituted in the standard low volume of water and then further diluted with isotonic diluent, such as 0.9%sodium chloride.[0069] In one embodiment of the present invention, human or humanized anti-IL-23p 19 antibody (or antigen bindingfragment thereof) is formulated as a lyophilized powder for subcutaneous or intravenous administration. One suchformulation is provided at Table 3 and described at Example 1. In one embodiment, the antibody (or antigen bindingfragment thereof) is provided at about 50 mg/vial, and is reconstituted with sterile water for injection prior to use. Ifdesired, the reconstituted antibody may be aseptically diluted with water or 0.9% Sodium Chloride Injection USP in asterile IV container. The target pH of the reconstituted formulation is 4.8�0.4, or optionally 4.8�0.2. In various embod-iments, the lyophilized formulation of the present invention enables reconstitution of the human or humanized anti-IL-23p19 antibody to high concentrations, such as about 20, 25, 30, 40, 50, 60, 75, 100 or more mg/mL.[0070] The present invention provides, inter alia, a lyophilized formulation comprising a human or humanized anti-IL-23p19 antibody, a citrate buffer at about pH 4.8, or at about pH 5.5, for example about 3.5, 3.8, 4.2, 4.6, 4.7, 4.8, 4.9,5.0, 5.2, 5.5 or 5.8, more preferably about 4.6, 4.7, 4.8, 4.9 or 5.0. When a range of pH values is recited, such as "a pHbetween pH 4.4 and 5.2," the range is intended to be inclusive of the recited values. Unless otherwise indicated, the pHrefers to the pH after reconstitution of the lyophilized formulations of the present invention. The pH is measured at 25°Cusing standard glass bulb pH meter. As used herein, a solution comprising "citrate buffer at pH X" refers to a solutionat pH X and comprising the citrate buffer, i.e. the pH is intended to refer to the pH of the solution.[0071] The formulations in Tables 3 and 4 (Example 1) reflect the weight of the components in a batch formulation,as lyophilized in vials, and as reconstituted. Lyophilized formulations are by definition essentially dry, and thus theconcept of concentration is not useful in describing them. Describing a lyophilized formulation in the terms of the weightof the components in a unit dose vial is more useful, but is problematic because it varies for different doses or vial sizes.In describing the lyophilized formulations of the present invention, it is useful to express the amount of a component asthe ratio of the weight of the component compared to the weight of the drug substance (DS) in the same sample (e.g.a vial). This ratio may be expressed as a percentage. Such ratios reflect an intrinsic property of the lyophilized formulationsof the present invention, independent of vial size, dosing, and reconstitution protocol.[0072] In other embodiments, the lyophilized formulation of anti-human IL-23p19 antibody, or antigen binding fragment,is defined in terms of the pre-lyophilization solution used to make the lyophilized formulation, such as the pre-lyophilizationsolution disclosed at Table 3. Pre-lyophilization solutions may comprise antibody, or antigen-binding fragment thereof,at concentrations of about 1, 3, 5, 10, 15, 20, 25, 30, 40, 50 mg/mL or higher. Such pre-lyophilization solutions may beat pH 4.4 - 5.2, , e.g. about pH 4.8, or may be at about pH 5.5.

EP 2 331 078 B1

10

5

10

15

20

25

30

35

40

45

50

55

[0073] In yet other embodiments, the lyophilized formulation of anti-human IL-23p19 antibody, or antigen bindingfragment, is defined in terms of the reconstituted solution generated from the lyophilized formulation, such as the recon-stituted solution disclosed at Table 4. Reconstituted solutions may comprise antibody, or antigen-binding fragmentthereof, at concentrations of about 10, 15, 20, 25, 30, 40, 50, 60, 75, 80, 90 or 100 mg/mL or higher. Such reconstitutedsolutions may be at pH 4.4 - 5.2, e.g. about pH 4.8, or may be at about pH 5.5.[0074] The lyophilized formulations of the present invention are formed by lyophilization (freeze-drying) of a pre-lyophilization solution. Freeze-drying is accomplished by freezing the formulation and subsequently subliming water ata temperature suitable for primary drying. Under this condition, the product temperature is below the eutectic point orthe collapse temperature of the formulation. Typically, the shelf temperature for the primary drying will range from about-30 to 25°C (provided the product remains frozen during primary drying) at a suitable pressure, ranging typically fromabout 50 to 250 mTorr. The formulation, size and type of the container holding the sample (e.g., glass vial) and thevolume of liquid will dictate the time required for drying, which can range from a few hours to several days (e.g. 40-60hrs). A secondary drying stage may be carried out at about 0-40°C, depending primarily on the type and size of containerand the type of protein employed. The secondary drying time is dictated by the desired residual moisture level in theproduct and typically takes at least about 5 hours (e.g. 10-15 hours). Typically, the moisture content of a lyophilizedformulation is less than about 5%, and preferably less than about 3%. The pressure may be the same as that employedduring the primary drying step. Freeze-drying conditions can be varied depending on the formulation and vial size.[0075] In some instances, it may be desirable to lyophilize the protein formulation in the container in which reconstitutionof the protein is to be carried out in order to avoid a transfer step. The container in this instance may, for example, bea 3, 5, 10, 20, 50 or 100 cc vial.[0076] The lyophilized formulations of the present invention are reconstituted prior to administration. The protein maybe reconstituted at a concentration of about 10, 15, 20, 25, 30, 40, 50, 60, 75, 80, 90 or 100 mg/mL or higher. Highprotein concentrations are particularly useful where subcutaneous delivery of the reconstituted formulation is intended.However, for other routes of administration, such as intravenous administration, lower concentrations of the protein maybe desired (e.g. from about 5-50 mg/mL).[0077] Reconstitution generally takes place at a temperature of about 25°C to ensure complete hydration, althoughother temperatures may be employed as desired. The time required for reconstitution will depend, e.g., on the type ofdiluent, amount of excipient(s) and protein. Exemplary diluents include sterile water, bacteriostatic water for injection(BWFI), a pH buffered solution (e.g. phosphate-buffered saline), sterile saline solution, Ringer’s solution or dextrosesolution. The diluent optionally contains a preservative. Exemplary preservatives have been described above, witharomatic alcohols such as benzyl or phenol alcohol being the preferred preservatives. The amount of preservativeemployed is determined by assessing different preservative concentrations for compatibility with the protein and pre-servative efficacy testing. For example, if the preservative is an aromatic alcohol (such as benzyl alcohol), it can bepresent in an amount from about 0.1-2.0% and preferably from about 0.5-1.5%, but most preferably about 1.0-1.2%.[0078] Various literature references are available to facilitate selection of pharmaceutically acceptable carriers orexcipients. See, e.g., Remington’s Pharmaceutical Sciences and U.S. Pharmacopeia: National Formulary, Mack Pub-lishing Company, Easton, PA (1984); Hardman et al. (2001) Goodman and Gilman’s The Pharmacological Basis ofTherapeutics, McGraw-Hill, New York, NY; Gennaro (2000) Remington: The Science and Practice of Pharmacy, Lip-pincott, Williams, and Wilkins, New York, NY; Avis et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral Med-ications, Marcel Dekker, NY; Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage Forms: Tablets, Marcel Dekker,NY; Lieberman et al. (eds.) (1990) Pharmaceutical Dosage Forms: Disperse Systems, Marcel Dekker, NY; Weiner andKotkoskie (2000) Excipient Toxicity and Safety, Marcel Dekker, Inc., New York, NY.[0079] Toxicity is a primary consideration is selecting the proper dosing of a therapeutic agent, such as a human orhumanized anti-IL-23p19 antibody (or antigen binding fragment thereof). Toxicity and therapeutic efficacy of the antibodycompositions, administered alone or in combination with an immunosuppressive agent, can be determined by standardpharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio betweentoxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio of LD50 to ED50. Antibodiesexhibiting high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studiescan be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably withina range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this rangedepending upon the dosage form employed and the route of administration utilized.[0080] Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administra-tion; parenteral delivery, including intramuscular, intradermal, subcutaneous, intramedullary injections, as well as in-trathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections. Drugs can be admin-istered in a variety of conventional ways, such as oral ingestion, pulmonarily by inhalation, topical application or cutaneous,subcutaneous, intraperitoneal, parenteral, intraarterial or intravenous injection. Modes of administration in which thevolume of solution must be limited (e.g. subcutaneous administration) require that a lyophilized formulation enable

EP 2 331 078 B1

11

5

10

15

20

25

30

35

40

45

50

55

reconstitution at high concentration.[0081] Alternately, one may administer the antibody in a local rather than systemic manner, for example, via injectionof the antibody directly into an arthritic joint or pathogen-induced lesion characterized by immunopathology, often in adepot or sustained release formulation. Furthermore, one may administer the antibody in a targeted drug delivery system,for example, in a liposome coated with a tissue-specific antibody, targeting, for example, arthritic joint or pathogen-induced lesion characterized by immunopathology. The liposomes will be targeted to and taken up selectively by theafflicted tissue.[0082] Selecting an administration regimen for a therapeutic depends on several factors, including the serum or tissueturnover rate of the entity, the level of symptoms, the immunogenicity of the entity, and the accessibility of the targetcells in the biological matrix. Preferably, an administration regimen maximizes the amount of therapeutic delivered tothe patient consistent with an acceptable level of side effects. Accordingly, the amount of biologic delivered depends inpart on the particular entity and the severity of the condition being treated. Guidance in selecting appropriate doses ofantibodies, cytokines, and small molecules are available. See, e.g., Wawrzynczak (1996) Antibody Therapy, Bios Sci-entific Pub. Ltd, Oxfordshire, UK; Kresina (ed.) (1991) Monoclonal Antibodies, Cytokines and Arthritis, Marcel Dekker,New York, NY; Bach (ed.) (1993) Monoclonal Antibodies and Peptide Therapy in Autoimmune Diseases, Marcel Dekker,New York, NY; Baert et al. (2003) New Engl. J. Med. 348:601-608; Milgrom et al. (1999) New Engl. J. Med. 341:1966-1973;Slamon et al. (2001) New Engl. J. Med. 344:783-792; Beniaminovitz et al. (2000) New Engl. J. Med. 342:613-619; Ghoshet al. (2003) New Engl. J. Med. 348:24-32; Lipsky et al. (2000) New Engl. J. Med. 343:1594-1602; Physicians DeskReference 2003 (Physicians’ Desk Reference, 57th Ed); Medical Economics Company; ISBN: 1563634457; 57th edition(November 2002).[0083] Determination of the appropriate dose is made by the clinician, e.g., using parameters or factors known orsuspected in the art to affect treatment or predicted to affect treatment. The appropriate dosage ("therapeutically effectiveamount") of the protein will depend, for example, on the condition to be treated, the severity and course of the condition,whether the protein is administered for preventive or therapeutic purposes, previous therapy, the patient’s clinical historyand response to the protein, the type of protein used, and the discretion of the attending physician. Generally, the dosebegins with an amount somewhat less than the optimum dose and it is increased by small increments thereafter untilthe desired or optimum effect is achieved relative to any negative side effects. Important diagnostic measures includethose of symptoms of, e.g., the inflammation or level of inflammatory cytokines produced. The protein is suitably admin-istered to the patient at one time or repeatedly. The protein may be administered alone or in conjunction with other drugsor therapies.[0084] Antibodies, antibody fragments, and cytokines can be provided by continuous infusion, or by doses at intervalsof, e.g., one day, 1-7 times per week, one week, two weeks, monthly, bimonthly, etc. A preferred dose protocol is oneinvolving the maximal dose or dose frequency that avoids significant undesirable side effects. A total weekly dose isgenerally at least 0.05 Pg/kg, 0.2 Pg/kg, 0.5 Pg/kg, 1 Pg/kg, 10 Pg/kg, 100 Pg/kg, 0.2 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 10mg/kg, 25 mg/kg, 50 mg/kg body weight or more. See, e.g., Yang et al. (2003) New Engl. J. Med. 349:427-434; Heroldet al. (2002) New Engl. J. Med. 346:1692-1698; Liu et al. (1999) J. Neurol. Neurosurg. Psych. 67:451-456; Portielji etal. (20003) Cancer Immunol. Immunother. 52:133-144. The desired dose of a small molecule therapeutic, e.g., a peptidemimetic, natural product, or organic chemical, is about the same as for an antibody or polypeptide, on a moles/kg basis.[0085] Subcutaneous administration may performed by injected using a syringe, or using other injection devices (e.g.the Inject-ease® device); injector pens; or needleless devices (e.g. MediJector and BioJector®).

VII. Uses

[0086] The present invention provides lyophilized formulations of anti-IL-23 antibodies (and fragments thereof) for usein the treatment of inflammatory disorders and conditions, e.g., of the central nervous system, peripheral nervous system,and gastrointestinal tract, as well as autoimmune and proliferative disorders.[0087] The lyophilized formulations of the present invention can be used in the treatment of, e.g., multiple sclerosis(MS), including relapsing-remitting MS and primary progressive MS, Alzheimer’s disease, amyotrophic lateral sclerosis(a.k.a. ALS; Lou Gehrig’s disease), ischemic brain injury, prion diseases, and HIV-associated dementia, as well asneuropathic pain, posttraumatic neuropathies, Guillain-Barre syndrome (GBS), peripheral polyneuropathy, and nerveregeneration.[0088] The lyophilized formulations of the present invention can also be used in the treatment of inflammatory boweldisorders, e.g., Crohn’s disease, ulcerative colitis, celiac disease, and irritable bowel syndrome. They can also be usedin the treatment of inflammatory disorders such as psoriasis, atopic dermatitis, arthritis, including rheumatoid arthritis,osteoarthritis, and psoriatic arthritis, autoimmune disorders, such as systemic lupus erythematosus and type I diabetes,and proliferative disorders such as cancer. See, e.g., PCT patent application publications WO 04/081190; WO 04/071517;WO 00/53631; and WO 01/18051.[0089] The broad scope of this invention is best understood with reference to the following examples, which are not

EP 2 331 078 B1

12

5

10

15

20

25

30

35

40

45

50

55

intended to limit the inventions to the specific embodiments. The specific embodiments described herein are offered byway of example only, and the invention is to be defined by the terms of the accompanying claims.

EXAMPLES

Example 1

Lyophilized Formulations of Humanized Anti-IL-23p19 Antibodies

[0090] Lyophilized formulations of a humanized anti-human IL-23p 19 antibody are prepared as follows. A batchformula for humanized anti-IL-23p19 antibody is provided in Table 3. The final concentration of humanized anti-IL-23p19antibody is 25 mg/mL. This batch formulation may used to prepare the lyophilized 50 mg/vial units, as discussed withreference to Table 4, infra. Polysorbate 80 from a vegetable source is used. Additional citric acid or sodium hydroxidemay be added to adjust the pH to the desired value of approximately 4.8 (�0.2). A pH of 4.8 is used to reduce opalescencewhen the antibody is reconstituted from citrate buffer at pH 5.5-5.6. The components are brought to a final volume of40 L with sterile water for injection (WFI). Correspondingly smaller lots may, of course, be prepared by proportionalreduction of the amounts listed in Table 3.

[0091] The unit composition of the final lyophilized formulation of humanized anti-IL-23p19 is provided at Table 4.

[0092] The unit formulation of Table 4 comprises 1/20,000th of the batch formulation of Table 3 after lyophilization toremove the water. The 50 mg of DS is added as 2.0 mL of the 25 mg/mL batch formulation of Table 3, and concentratedfour-fold by reconstitution with sterile WFI to a final volume of 0.5 mL. Accordingly, the initial 2.5 mM citrate buffer is

Table 3

Batch Formula for Anti-IL-23p19 Antibody

Component Grade Amount per Batch (g)

Humanized Anti-IL-23p19 antibody - 1000

Trisodium Citrate Dihydrate USP 14.83

Citric Acid (anhydrous) USP 9.512

Polysorbate 80 NF 2.0

Sucrose NF 700

Sodium Hydroxide NF pH adjustment

Water for injection USP q.s. to 40.00 L

Table 4

Unit Composition of Lyophilized Powder Formulation for Solution for Injection

Component Grade Amount (mg/vial) Concentration after Reconstitution (mg/mL)

Function

Humanized anti-IL-23p 19 antibody

- 50 100 Drug Substance

Trisodium Citrate Dihydrate

USP 0.7414 1.483 Buffer salt

Citric Acid (anhydrous) USP 0.476 0.951 Buffer acid

Polysorbate 80 NF 0.10 0.20 Surfactant

Sucrose NF 35 70 Stabilizer/ Tonicity Modifier

Sodium Hydroxide NF - - pH adjustment

Sterile Water for Injection USP - q.s. to 0.5 mL Solvent

EP 2 331 078 B1

13

5

10

15

20

25

30

35

40

45

50

55

concentrated to about 10 mM citrate buffer in the reconstituted solution, and the sucrose is concentrated from about 50mM to about 200 mM. Lower final concentrations may be obtained by reconstituting in a larger volume of liquid, suchas 0.5 mL of WFI and additional amounts of 0.9% sodium chloride or WFI.[0093] In order to ensure consistent delivery of the label fill, product vials may contain an appropriate volume of overfillto compensate for residual product solution that might remain in the vial and the syringe during withdrawal of thereconstituted solution. For example, a nominal fill of 2.0 mL (50 mg) may be increased to an overfill of 2.7 mL (67.5 mg).In the event of such overfill, the final unit composition will, of course, comprise proportionally greater amounts of eachcomponent listed in Table 4. In the case of a 2.7 mL fill for a nominal 2.0 mL vial, the amount of each component wouldbe 35% higher than listed in Table 4, as illustrated in Table 5. For a 2.7 mL overfill, 0.56 mL of water is used forreconstitution to a final volume of 0.675 mL, for a final concentration of 100 mg/mL. The final concentrations afterreconstitution are, of course, the same as in Table 4.

[0094] The drug is packaged in sterile 13 mm neck, 5 mL, Type 1 glass tubing vials, closed with 13-mm gray butylrubber stoppers and sealed with aluminum crimp seals with polypropylene bonnet. Vials are stored at 2 - 8°C, andrefrigerated when shipped.[0095] FIG. 1 is a flow diagram for a manufacturing process for the lyophilized formulation of humanized anti-IL-23p19antibody of the present invention, e.g. into a 50 mg unit dose vial.[0096] Compounding involves the following steps. Charge the required amount of water for injection (WFI) into a taredcompounding vessel. Charge and dissolve with mixing, sucrose, trisodium citrate dihydrate, citric acid, and polysorbate80 from a vegetable source. Measure the pH. Equilibrate the drug substance to ambient temperature and charge thedrug substance slowly into the compounding vessel. Continue to mix gently to avoid foaming. Measure the pH againand adjust if needed to bring the pH to approximately 4.8. Charge WFI to the final weight of the bulk solution withcontinued gentle mixing.[0097] Filtration involves the following steps. Connect sterilizing filter (0.22 Pm) to the sterile receiving vessel. Collectan aliquot of the bulk solution for bioburden testing prior to sterile filtration. Perform aseptic filtration using a 0.22 Pmfilter into a sterile container. Perform filter integrity testing before and after product filtration.[0098] Filling involves the following steps. Using suitable filling equipment, aseptically fill the product solution intosterilized Type I tubing glass vials to achieve a target fill volume of 2.7 ml. Perform fill weight checks during filling. Removeappropriate number of vials at beginning of filling and pool the solution for bulk sterility and endotoxin testing. Partiallyseat sterilized lyo-shape stoppers into filled vials. Load the filled vials into a suitable freeze-dryer.[0099] Lyophilization, stoppering and capping involve the following steps. Lyophilize the filled vials using an appropriatelyophilization cycle. After lyophilization is complete, backfill the vials with 0.22 Pm filtered nitrogen and fully stopper.Unload the stoppered vials from the lyophilizer and seal them.[0100] The resulting vials are inspected for visual defects and stored at 2-8 °C. Finished unit dosage vials are shippedunder refrigerated conditions.

Table 5

2.7 mL Overfull Unit Composition

Component Grade Amount (mg/vial) Concentration after Reconstitution (mg/mL)

Function

Humanized anti-IL-23p19 antibody

- 67.5 100.0 Active Pharmaceutical Ingredient (API)

Trisodium Citrate Dihydrate

USP 1.001 1.483 Buffer salt

Citric Acid (anhydrous) USP 0.643 0.951 Buffer acid

Polysorbate 80 NF 0.135 0.200 Surfactant

Sucrose NF 47.25 70.00 Stabilizer/ Tonicity Modifier

Sodium Hydroxide NF - - pH adjustment

Sterile Water for Injection USP - 0.56 mL. Solvent

EP 2 331 078 B1

14

5

10

15

20

25

30

35

40

45

50

55

Example 2

Stability Testing of Lyophilized Formulations of Humanized Anti-IL-23p 19 Antibodies

[0101] FIGS. 2-7 provide the results of stability testing of lyophilized formulations of a humanized anti-human IL-23p19antibody under various storage conditions. Some vials were stored in both upright and inverted configurations, asindicated in the figures. As discussed in more detail below, FIGS. 2-4 show stability of at least 18 months for antibodieslyophilized at pH 5.5 (citrate buffer), and FIGS. 5-7 show stability of at least 12 months for antibodies lyophilized at pH4.8 (citrate buffer), wherein 18- and 12-months are the longest time points presented rather than an experimentallydetermined stability endpoint.[0102] Stability was assessed as follows. Samples were lyophilized in 5 mL Type I glass vials, and sealed with 13 mmbromobutyl lyo stoppers (Helvoet Rubber & Plastic Technologies BV, Hellevoetsluis, The Netherlands) and flip-off alu-minum seals. B2-Coated 13 mm gray butyl lyo-stoppers (West Pharmaceutical Services Inc., Lionville, Pennsylvania,USA) may also be used. Vials were placed on stability stations under the following storage conditions: 5C (5�3°C), 25H(25,60% relative humidity), or RH4 (40°C, 70% relative humidity). Samples were obtained at an initial time point, and at0.5, 1, 2, 3, 6, 9, 12 or 18 months, as indicated in the figures.[0103] The stability of the samples is illustrated by the various characteristics presented at FIGS 2 - 7. The lyophilizedsamples were visually inspected, reconstituted, and the reconstituted formulation was visually inspected. The pH of thesamples after reconstitution was measured, and the protein concentration determined by U.V. absorbance. The sampleswere then analyzed by denaturing sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis (PAGE), with thelevel of impurities (i.e. material other than the main product band) expressed as the percentage of the total intensity ineach lane. Purity of the sample was further assessed by high performance size exclusion chromatography (HPSEC) inwhich the percentage of monomer was determined, as well as the percentages of high molecular weight species (possiblyaggregates) and late eluting peaks (possibly degradation products).[0104] Additional sample characterization data include high performance ionexchange chromatography (HP-IEX),which is used to assess purity by revealing the presence of acidic or basic variants. Results are presented as a percentageof total observed material. The samples were further characterized for biological function using an enzyme-linked im-munosorbent assay (ELISA) for binding to human IL-23p19. Results are expressed as the EC50 for the sample, i.e. theconcentration necessary to achieve half-maximal binding. Results are also provided as a percentage potency relativeto control, calculated as the 100 times the ratio of EC50 for the samples to the EC50 for a control preparation of the sameantibody. Moisture content of the lyophilized powder was also determined.[0105] Data in FIGS. 2, 3 and 4 were obtained for samples at pH 5.5 when stored at 5C, 25H, and RH4, respectively.Data in FIGS. 5, 6 and 7 were obtained for samples at pH 4.8 when stored at 5C, 25H, and RH4, respectively.[0106] The results generally demonstrate high stability of lyophilized formulations of the present invention over 1, 3,6, 9, 12 and 18 month time periods, at both pH 5.5 and 4.8. The data reveal no trending over time that would reflectinstability for samples at refrigerated storage conditions. Based on these results, samples are projected to have a shelf-life of at least 24 months.[0107] Table 6 provides a brief description of the sequences in the sequence listing.

Table 6

Sequence Identifiers

(SEQ ID NO: Description

1 m1A11 VH

2 m11C1 VH

3 m5F5 VH

4 m21D1 VH

5 m 13B8 VH

6 hum13B8 HC-a

7 hum13B8 HC-b

8 hum 13B8 HC-c

9 m1A11 VL

10 m11C1 VL

EP 2 331 078 B1

15

5

10

15

20

25

30

35

40

45

50

55

(continued)

Sequence Identifiers

(SEQ ID NO: Description

11 m5F5 VL

12 m21D1 VL

13 m13B8 VL

14 hum13B8 LC

15 m1A11 CDRH1

16 m11C1 CDRH1

17 m5F5 CDRH 1

18 m21D1 CDR1

19 m13B8 CDRH1

20 m1A11 CDRH2

21 m11C1 CDRH2

22 m5F5 CDRH2

23 m21D1 CDRH2

24 m13B8 CDRH2-a

25 h13B8 CDRH2-b

26 h 13B8 CDRH2-c

27 m1A11 CDRH3

28 m11C1 CDRH3

29 m5F5 CDRH3

30 m21D1 CDR3

31 m13B8 CDRH3

32 m1A11 CDRL1

33 m11C1 CDRL1

34 m5F5 CDRL1

35 m21D1 CDRL1

36 m13B8 CDRL1

37 m1A11 CDRL2

38 m11C1 CDRL2

39 m5F5 CDRL2

40 m21D1 CDRL2

41 m13B8 CDRL2

42 m1A11 CDRL3

43 m11C1 CDRL3

44 m5F5 CDRL3

45 m21D1 CDRL3

46 m13B8 CDRL3

47 human IL-23p19

EP 2 331 078 B1

16

5

10

15

20

25

30

35

40

45

50

55

SEQUENCE LISTING

[0108]

<110> Schering Corporation Li, Xuhong Kashi, Ramesh Badkar, Aniket

<120> Lyophilized Formulations of Engineered Anti-IL-23p19 Antibodies

<130> UB06868

<150> US 61,092326<151> 2008-08-27

<160> 52

<170> Patent In version 3.5

<210> 1<211> 117<212> PRT<213> Mus musculus

<400> 1

(continued)

Sequence Identifiers

(SEQ ID NO: Description

48 mouse IL-23p19

49 hum13B8-b HC DNA

50 hum13B8 LC DNA

51 Heavy Chain Signal Sequence

52 Light Chain Signal Sequence

EP 2 331 078 B1

17

5

10

15

20

25

30

35

40

45

50

55

<210> 2<211> 116<212> PRT<213> Mus musculus

<400> 2

EP 2 331 078 B1

18

5

10

15

20

25

30

35

40

45

50

55

<210> 3<211> 124<212> PRT<213> Mus musculus

<400> 3

EP 2 331 078 B1

19

5

10

15

20

25

30

35

40

45

50

55

<210> 4<211> 116<212> PRT<213> Mus musculus

<400> 4

<210> 5<211> 116<212> PRT<213> Mus musculus

<400> 5

EP 2 331 078 B1

20

5

10

15

20

25

30

35

40

45

50

55

<210> 6<211> 446<212> PRT<213> Artificial Sequence

<220><223> Human frameworks, rodent CDRs

<220><221> DOMAIN<222> (1)..(116)<223> Variable Domain

<400> 6

EP 2 331 078 B1

21

5

10

15

20

25

30

35

40

45

50

55

EP 2 331 078 B1

22

5

10

15

20

25

30

35

40

45

50

55

<210> 7<211> 446<212> PRT<213> Artificial Sequence

<220><223> Human frameworks, rodent CDRs

<220><221> DOMAIN<222> (1) .. (116)<223> Variable Domain

<400> 7

EP 2 331 078 B1

23

5

10

15

20

25

30

35

40

45

50

55

EP 2 331 078 B1

24

5

10

15

20

25

30

35

40

45

50

55

<210> 8<211> 446<212> PRT<213> Artificial Sequence

<220><223> Human frameworks, rodent CDRs

<220><221> DOMAIN<222> (1)..(116)<223> Variable Domain

<400> 8

EP 2 331 078 B1

25

5

10

15

20

25

30

35

40

45

50

55

EP 2 331 078 B1

26

5

10

15

20

25

30

35

40

45

50

55

<210> 9<211> 113<212> PRT<213> Mus musculus

<400> 9

EP 2 331 078 B1

27

5

10

15

20

25

30

35

40

45

50

55

<210> 10<211> 108<212> PRT<213> Mus musculus

<400> 10

<210> 11<211> 111<212> PRT

EP 2 331 078 B1

28

5

10

15

20

25

30

35

40

45

50

55

<213> Mus musculus

<400> 11

<210> 12<211> 108<212> PRT<213> Mus musculus

<400> 12

EP 2 331 078 B1

29

5

10

15

20

25

30

35

40

45

50

55

<210> 13<211> 108<212> PRT<213> Mus musculus

<400> 13

<210> 14<211> 214<212> PRT<213> Artificial Sequence

<220><223> Human frameworks, rodent CDRs

<220><221> DOMAIN<222> (1)..(108)<223> Variable Domain

<400> 14

EP 2 331 078 B1

30

5

10

15

20

25

30

35

40

45

50

55

<210> 15<211> 10<212> PRT<213> Mus musculus

<400> 15

EP 2 331 078 B1

31

5

10

15

20

25

30

35

40

45

50

55

<210> 16<211> 10<212> PRT<213> Mus musculus

<400> 16

<210> 17<211> 10<212> PRT<213> Mus musculus

<400> 17

<210> 18<211> 10<212> PRT<213> Mus musculus

<400> 18

<210> 19<211> 10<212> PRT<213> Mus musculus

<400> 19

<210> 20<211> 17<212> PRT<213> Mus musculus

<400> 20

<210> 21

EP 2 331 078 B1

32

5

10

15

20

25

30

35

40

45

50

55

<211> 17<212> PRT<213> Mus musculus

<400> 21

<210> 22<211> 17<212> PRT<213> Mus musculus

<400> 22

<210> 23<211> 17<212> PRT<213> Mus musculus

<400> 23

<210> 24<211> 17<212> PRT<213> Mus musculus

<400> 24

<210> 25<211> 17<212> PRT<213> Artificial Sequence

<220>

EP 2 331 078 B1

33

5

10

15

20

25

30

35

40

45

50

55

<223> Rodent CDR with one amino acid substitution

<400> 25

<210> 26<211> 17<212> PRT<213> Artificial Sequence

<220><223> Rodent CDR with four amino acid substitutions

<400> 26

<210> 27<211> 8<212> PRT<213> Mus musculus

<400> 27

<210> 28<211> 7<212> PRT<213> Mus musculus

<400> 28

<210> 29<211> 15<212> PRT<213> Mus musculus

<400> 29

EP 2 331 078 B1

34

5

10

15

20

25

30

35

40

45

50

55

<210> 30<211> 7<212> PRT<213> Mus musculus

<400> 30

<210> 31<211> 7<212> PRT<213> Mus musculus

<400> 31

<210> 32<211> 16<212> PRT<213> Mus musculus

<400> 32

<210> 33<211> 11<212> PRT<213> Mus musculus

<400> 33

<210> 34<211> 14<212> PRT<213> Mus musculus

<400> 34

<210> 35<211> 11<212> PRT<213> Mus musculus

<400> 35

EP 2 331 078 B1

35

5

10

15

20

25

30

35

40

45

50

55

<210> 36<211> 11<212> PRT<213> Mus musculus

<400> 36

<210> 37<211> 7<212> PRT<213> Mus musculus

<400> 37

<210> 38<211> 7<212> PRT<213> Mus musculus

<400> 38

<210> 39<211> 7<212> PRT<213> Mus musculus

<400> 39

<210> 40<211> 7<212> PRT<213> Mus musculus

<400> 40

<210> 41<211> 7

EP 2 331 078 B1

36

5

10

15

20

25

30

35

40

45

50

55

<212> PRT<213> Mus musculus

<400> 41

<210> 42<211> 9<212> PRT<213> Mus musculus

<400> 42

<210> 43<211> 9<212> PRT<213> Mus musculus

<400> 43

<210> 44<211> 9<212> PRT<213> Mus musculus

<400> 44

<210> 45<211> 9<212> PRT<213> Mus musculus

<400> 45

<210> 46<211> 9<212> PRT<213> Mus musculus

<400> 46

EP 2 331 078 B1

37

5

10

15

20

25

30

35

40

45

50

55

<210> 47<211> 170<212> PRT<213> Homo sapiens

<400> 47

<210> 48<211> 175<212> PRT<213> Mus musculus

<400> 48

EP 2 331 078 B1

38

5

10

15

20

25

30

35

40

45

50

55

<210> 49<211> 1398<212> DNA<213> Artificial Sequence

<220><223> Human constant and framework regions, rodent CDRs

<400> 49

EP 2 331 078 B1

39

5

10

15

20

25

30

35

40

45

50

55

<210> 50<211> 702<212> DNA<213> Artificial Sequence

<220><223> Human constant and framework regions, rodent CDRs

<400> 50

EP 2 331 078 B1

40

5

10

15

20

25

30

35

40

45

50

55

<210> 51<211> 19<212> PRT<213> Mus musculus

<400> 51

<210> 52<211> 19<212> PRT<213> Rattus norvegicus

<400> 52

Claims

1. A lyophilized pharmaceutical formulation of an anti-human IL-23p19 antibody, or antigen binding fragment thereof,made by lyophilizing an aqueous solution comprising:

a) 5-25 mg/mL anti-human IL-23p19 antibody, or antigen binding fragment thereof;b) about 50 mM sucrose;

EP 2 331 078 B1

41

5

10

15

20

25

30

35

40

45

50

55

c) about 0.05 mg/mL polysorbate 80; andd) about 2.5 mM citrate buffer at pH 4.4 -5.2.

2. The lyophilized pharmaceutical formulation of Claim 1, wherein the anti-human IL-23p19 antibody, or antigen bindingfragment thereof, is present at about 25 mg/mL in the aqueous solution.

3. The lyophilized pharmaceutical formulation of Claim1 or 2, wherein the aqueous solution has a pH about 4.8.

4. A lyophilized pharmaceutical formulation of an anti-human IL-23p19 antibody, or antigen binding fragment thereof,that when reconstituted comprises:

a)25-100 mg/ mL anti-human IL-23p19 antibody, or antigen binding fragment thereof;b) about 200 mM sucrose;c) about 0.2 mg/mL polysorbate 80; andd) about 10 nM citrate buffer at pH 4.4 to 5.2

5. The lyophilized pharmaceutical formulation of Claim 4, wherein the anti-human IL-23p19 antibody,or antigen bindingfragment thereof, is present at about 100 mg/mL in the reconstituted solution

6. The lyophilized pharmaceutical formulation of Claim 4, wherein the reconstituted solution has a pH of about 4.8.

7. The lyophilized formulation of any preading Claim,wherein antibody, or antigen binding fragment thereof, comprisesa light chain comprising three CDR sequences selected from the group constiting of SEQ ID NOs: 32-46

8. The lyophilized formulation of any preading Claim, wherein antibody, or antigen binding fragment thereof, comprisesa heavy chain comprising there CDR sequences selected from the group consisting of SEQ ID NOs: 15-31

9. The lyophilized formulation of any preading Claim 7 and 8 wherein antibody, or antigen comprises:

i) a light chain comprising three CDR sequences selected from the group consisting of SEQ ID NOs: 32-46; andii)a heavy chain comprising three CDR sequences selected from the group consisting of SEQ ID NOs 15-31.

10. The lyophilized formulation of any one Claims 1 to 6, wherein antibody, or antigen binding fragment thereof, comprisesa light chain variable domain comprising residues 1-108 of SEQ of ID NO:14.

11. The lyophilized formulation of any one Claims 1 to 6 or 10, wherein antibody, or antigen binding fragment thereof,comprises a heavy chain variable domain comprising a sequence selected from the group consisting of residues1-116 of SEQ ID NOs 6-8

12. The lyophilized formulation of Claim 11, wherein antibody, or antigen binding fragment thereof, comprises:

i)a light chain comprising SEQ ID NO:14;andii)a heavy chain comprising a sequence selected from the group consisting of SEQ ID NOs: 6-8

Patentansprüche

1. Eine gefriergetrocknete pharmazeutische Formulierung eines Anti-Human-IL-23p19-Antikörpers oder eines Antigen-bindenden Fragments davon, hergestellt durch Gefriertrocknen einer wässrigen Lösung, umfassend:

a) 5 - 25 mg/ml Anti-Human-IL-23p19-Antikörper oder Antigen-bindendes Fragment davon,b) etwa 50 mM Saccharose,c) etwa 0,05 mg/ml Polysorbat 80 undd) etwa 2,5 mM Citratpuffer bei pH 4,4 - 5,2.

2. Die gefriergetrocknete pharmazeutische Formulierung nach Anspruch 1, wobei der Anti-Human-IL-23p19-Antikörperoder das Antigen-bindende Fragment davon in etwa 25 mg/ml in der wässrigen Lösung vorliegt.

EP 2 331 078 B1

42

5

10

15

20

25

30

35

40

45

50

55

3. Die gefriergetrocknete pharmazeutische Formulierung nach Anspruch 1 oder 2, wobei die wässrige Lösung einenpH-Wert von etwa 4,8 besitzt.

4. Eine gefriergetrocknete pharmazeutische Formulierung eines Anti-Human-IL-23p19-Antikörpers oder eines Antigen-bindenden Fragments davon, die, wenn rekonstituiert, umfasst:

a) 25 - 100 mg/ml Anti-Human-IL-23p19-Antikörper oder Antigen-bindendes Fragment davon,b) etwa 200 mM Saccharose,c) etwa 0,2 mg/ml Polysorbat 80 undd) etwa 10 mM Citratpuffer bei pH 4,4 bis 5,2.

5. Die gefriergetrocknete pharmazeutische Formulierung nach Anspruch 4, wobei der Anti-Human-IL-23p19-Antikörperoder das Antigen-bindende Fragment davon in etwa 100 mg/ml in der rekonstituierten Lösung vorliegt.

6. Die gefriergetrocknete pharmazeutische Formulierung nach Anspruch 4, wobei die rekonstituierte Lösung einenpH-Wert von etwa 4,8 besitzt.

7. Die gefriergetrocknete Formulierung nach einem vorhergehenden Anspruch, wobei der Antikörper oder das Antigen-bindende Fragment davon eine leichte Kette umfasst, die drei CDR-Sequenzen, ausgewählt aus der Gruppe, be-stehend aus SEQ ID NR.: 32-46, umfasst.

8. Die gefriergetrocknete Formulierung nach einem vorhergehenden Anspruch, wobei der Antikörper oder das Antigen-bindende Fragment davon eine schwere Kette umfasst, die drei CDR-Sequenzen, ausgewählt aus der Gruppe,bestehend aus SEQ ID NR.: 15-31, umfasst.

9. Die gefriergetrocknete Formulierung nach Anspruch 7 und 8, wobei der Antikörper oder das Antigen-bindendeFragment davon umfasst:

i) eine leichte Kette, die drei CDR-Sequenzen umfasst, ausgewählt aus der Gruppe, bestehend aus SEQ IDNR.: 32-46, undii) eine schwere Kette, die drei CDR-Sequenzen, ausgewählt aus der Gruppe, bestehend aus SEQ ID NR.:15-31, umfasst.

10. Die gefriergetrocknete Formulierung nach einem der Ansprüche 1 bis 6, wobei der Antikörper oder das Antigen-bindende Fragment davon eine variable Domäne der leichten Kette umfasst, die die Reste 1 - 108 von SEQ ID NR:14 umfasst.

11. Die gefriergetrocknete Formulierung nach einem der Ansprüche 1 bis 6 oder 10, wobei der Antikörper oder dasAntigen-bindende Fragment davon eine variable Domäne der schweren Kette umfasst, die eine Sequenz umfasst,ausgewählt aus der Gruppe, bestehend aus den Resten 1 - 116 der SEQ ID NR.: 6-8.

12. Die gefriergetrocknete Formulierung nach Anspruch 11, wobei der Antiköper oder das Antigen-bindende Fragmentdavon umfasst:

i) eine leichte Kette, die SEQ ID NR.: 14 umfasst, undii) eine schwere Kette, die eine Sequenz umfasst, ausgewählt aus der Gruppe, bestehend aus SEQ ID NR.: 6-8.

Revendications

1. Formulation pharmaceutique lyophilisée d’un anticorps anti-IL-23p19 humaine ou d’un fragment de liaison à l’anti-gène de celui-ci, préparée en lyophilisant une solution aqueuse comprenant:

a) 5-25 mg/ml d’anticorps anti-IL-23p19 humaine ou de fragment de liaison à l’antigène de celui-ci;b) environ 50 mM de saccharose;c) environ 0,05 mg/ml de polysorbate 80; etd) environ 2,5 mM de tampon citrate à un pH de 4,4-5,2.

EP 2 331 078 B1

43

5

10

15

20

25

30

35

40

45

50

55

2. Formulation pharmaceutique lyophilisée selon la revendication 1, dans laquelle l’anticorps anti-IL-23p19 humaineou le fragment de liaison à l’antigène de celui-ci, est présent à environ 25 mg/ml dans la solution aqueuse.

3. Formulation pharmaceutique lyophilisée selon la revendication 1 ou 2, dans laquelle la solution aqueuse a un pHd’environ 4,8.

4. Formulation pharmaceutique lyophilisée d’un anticorps anti-IL-23p19 humaine ou d’un fragment de liaison à l’anti-gène de celui-ci, qui comprend lorsque reconstituée:

a) 25-100 mg/ml d’anticorps anti-IL-23p19 humaine ou de fragment de liaison à l’antigène de celui-ci;b) environ 200 mM de saccharose;c) environ 0,2 mg/ml de polysorbate 80; etd) environ 10 mM de tampon citrate à un pH de 4,4 à 5,2.

5. Formulation pharmaceutique lyophilisée selon la revendication 4, dans laquelle l’anticorps anti-IL-23p19 humaineou le fragment de liaison à l’antigène de celui-ci, est présent à environ 100 mg/ml dans la solution reconstituée.

6. Formulation pharmaceutique lyophilisée selon la revendication 4, dans laquelle la solution reconstituée a un pHd’environ 4,8.

7. Formulation lyophilisée selon l’une quelconque des revendications précédentes, dans laquelle l’anticorps ou lefragment de liaison à l’antigène de celui-ci, comprend une chaîne légère comprenant trois séquences CDR sélec-tionnées parmi le groupe consistant en les SEQ ID NO: 32-46.

8. Formulation lyophilisée selon l’une quelconque des revendications précédentes, dans laquelle l’anticorps ou lefragment de liaison à l’antigène de celui-ci, comprend une chaîne lourde comprenant trois séquences CDR sélec-tionnées parmi le groupe consistant en les SEQ ID NO: 15-31.

9. Formulation lyophilisée selon l’une quelconque des revendications 7 et 8, dans laquelle l’anticorps ou le fragmentde liaison à l’antigène de celui-ci, comprend:

i) une chaîne légère comprenant trois séquences CDR sélectionnées parmi le groupe consistant en les SEQID NO: 32-46; etii) une chaîne lourde comprenant trois séquences CDR sélectionnées parmi le groupe consistant en les SEQID NO: 15-31.

10. Formulation lyophilisée selon l’une quelconque des revendications 1 à 6, dans laquelle l’anticorps ou le fragmentde liaison à l’antigène de celui-ci, comprend un domaine variable de chaîne légère comprenant les résidus 1-108de la SEQ ID NO: 14.

11. Formulation lyophilisée selon l’une quelconque des revendications 1 à 6 ou 10, dans laquelle l’anticorps ou lefragment de liaison à l’antigène de celui-ci, comprend un domaine variable de chaîne lourde comprenant uneséquence sélectionnée parmi le groupe consistant en les résidus 1-116 des SEQ ID NO: 6-8.

12. Formulation lyophilisée selon la revendication 11, dans laquelle l’anticorps ou le fragment de liaison à l’antigène decelui-ci, comprend:

i) une chaîne légère comprenant la SEQ ID NO: 14, etii) une chaîne lourde comprenant une séquence sélectionnée parmi le groupe consistant en les SEQ ID NO: 6-8.

EP 2 331 078 B1

44

EP 2 331 078 B1

45

EP 2 331 078 B1

46

EP 2 331 078 B1

47

EP 2 331 078 B1

48

EP 2 331 078 B1

49

EP 2 331 078 B1

50

EP 2 331 078 B1

51

REFERENCES CITED IN THE DESCRIPTION

This list of references cited by the applicant is for the reader’s convenience only. It does not form part of the Europeanpatent document. Even though great care has been taken in compiling the references, errors or omissions cannot beexcluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description

• US 20070009526 A [0004]• WO 2007076524 A [0004]• WO 2007024846 A [0004]• WO 2007147019 A [0004]• WO 2009043933 A [0004]• WO 2008103432 A [0004] [0030]• WO 2008103473 A [0004]• US 20070048315 A [0004]• WO 9404678 A [0037]• WO 9425591 A [0037]• US 6005079 A [0037]• EP 404097 A [0038]

• WO 9311161 A [0038]• US 5624821 A [0040]• WO 2003086310 A [0040]• WO 2005120571 A [0040]• WO 20060057702 A [0040]• WO 04081190 A [0063] [0065] [0088]• WO 04071517 A [0063] [0065] [0088]• WO 0053631 A [0063] [0065] [0088]• WO 0118051 A [0063] [0065] [0088]• US 6267958 B [0067]• US 7247707 B [0067]• US 61092326 B [0108]

Non-patent literature cited in the description

• Oppmann et al. Immunity, 2000, vol. 13, 715-725[0002]

• Wiekowski et al. J. Immunol., 2001, vol. 166,7563-7570 [0002]

• Parham et al. J. Immunol., 2002, vol. 168, 5699-708[0002]

• Frucht. Sci STKE 2002, 2002, E1-E3 [0002]• Elkins et al. Infection Immunity, 2002, vol. 70,

1936-1948 [0002]• Bowman et al. Curr. Opin. Infect. Dis., 2006, vol. 19,

245 [0003]• Milstein et al. Nature, 1983, vol. 305, 537-39 [0035]• Brennan et al. Science, 1985, vol. 229, 81 [0035]• Holliger et al. Proc. Natl. Acad. Sci. U.S.A., 1993,

vol. 90, 6444-48 [0035]• Gruber et al. J. Immunol., 1994, vol. 152, 5368

[0035]• Pluckthun. THE PHARMACOLOGY OF MONO-

CLONAL ANTIBODIES. Springer-Verlag, 1994, vol.113, 269-315 [0036]

• Muyldermans et al. Trends Biochem. Sci., 2001,vol. 26, 230 [0037]

• Reichmann et al. J. Immunol. Methods, 1999, vol.231, 25 [0037]

• Holliger et al. Proc. Natl. Acad. Sci. USA, 1993, vol.90, 6444-6448 [0038]

• Holliger ; Hudson. Nat. Biotechnol., 2005, vol. 23,1126-1136 [0038]

• Presta. Adv. Drug Delivery Rev., 2006, vol. 58,640-656 [0040]

• Presta. J. Allergy Clin.Immunol., 2005, vol. 116, 731,734-35 [0040]

• Kabat et al. Sequences of Proteins of ImmunologicalInterest. Public Health Service, National Institutes ofHealth, 1991 [0042]

• Chothia ; Lesk. J. Mol. Biol., 1987, vol. 196, 901-917[0042]

• Watson et al. Molecular Biology of the Gene. TheBenjamin/Cummings Pub. Co, 1987, 224 [0045]

• Munsen et al. Analyt. Biochem., 1980, vol. 107,220-239 [0049]

• Murphy et al. JANEWAY’S IMMUNOBIOLOGY.2008 [0055]

• Kabat et al. Sequences of Proteins of ImmunologicalInterest. National Institutes of Health, 1987 [0058]

• Choo et al. SPdb: a Signal Peptide Database. BMCBioinformatics, 2005, vol. 6, 249 [0062]

• Langrish et al. Immunol. Rev., 2004, vol. 202,96-105 [0065]

• Langrish et al. J. Exp. Med., 2005, vol. 201, 233-240[0065]

• Cua ; Kastelein. Nat. Immunol., 2006, vol. 7,557-559 [0065]

• Remington’s Pharmaceutical Sciences and U.S.Pharmacopeia: National Formulary. Mack PublishingCompany, 1984 [0078]

• Hardman et al. Goodman and Gilman’s The Phar-macological Basis of Therapeutics. McGraw-Hill,2001 [0078]

• Gennaro. Remington: The Science and Practice ofPharmacy. Lippincott, Williams, and Wilkins, 2000[0078]

• Pharmaceutical Dosage Forms: Parenteral Medica-tions. Marcel Dekker, 1993 [0078]

EP 2 331 078 B1

52

• Pharmaceutical Dosage Forms: Tablets. MarcelDekker, 1990 [0078]

• Pharmaceutical Dosage Forms: Disperse Systems.Marcel Dekker, 1990 [0078]

• Weiner ; Kotkoskie. Excipient Toxicity and Safety.Marcel Dekker, Inc, 2000 [0078]

• Wawrzynczak. Antibody Therapy. Bios ScientificPub. Ltd, 1996 [0082]

• Monoclonal Antibodies, Cytokines and Arthritis. Mar-cel Dekker, 1991 [0082]

• Monoclonal Antibodies and Peptide Therapy in Au-toimmune Diseases. Marcel Dekker, 1993 [0082]

• Baert et al. New Engl. J. Med., 2000, vol. 348,601-608 [0082]

• Milgrom et al. New Engl. J. Med., 1999, vol. 341,1966-1973 [0082]

• Slamon et al. New Engl. J. Med., 2001, vol. 344,783-792 [0082]

• Beniaminovitz et al. New Engl. J. Med., 2000, vol.342, 613-619 [0082]

• Ghosh et al. New Engl. J. Med., 2003, vol. 348, 24-32[0082]

• Lipsky et al. New Engl. J. Med., 2000, vol. 343,1594-1602 [0082]

• Physicians Desk Reference. Medical EconomicsCompany, November 2002 [0082]

• Yang et al. New Engl. J. Med., 2003, vol. 349,427-434 [0084]

• Herold et al. New Engl. J. Med., 2002, vol. 346,1692-1698 [0084]

• Liu et al. J. Neurol. Neurosurg. Psych., 1999, vol.67, 451-456 [0084]

• Portielji et al. Cancer Immunol. Immunother., 2003,vol. 52, 133-144 [0084]


Recommended