AU2013202866A1 - Methods for treating psoriasis - Google Patents

Methods for treating psoriasis Download PDF

Info

Publication number
AU2013202866A1
AU2013202866A1 AU2013202866A AU2013202866A AU2013202866A1 AU 2013202866 A1 AU2013202866 A1 AU 2013202866A1 AU 2013202866 A AU2013202866 A AU 2013202866A AU 2013202866 A AU2013202866 A AU 2013202866A AU 2013202866 A1 AU2013202866 A1 AU 2013202866A1
Authority
AU
Australia
Prior art keywords
antibody
population
subject
antigen
subjects
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.)
Abandoned
Application number
AU2013202866A
Inventor
Walid M. Awni
Yanjun Bao
William T. Barchuk
Elliot K. Elliot K. Valdes
William G. Glass
Kenneth B. GORDON
Yihua GU
Tom C. Harris
Martin Kaul
Parvez M. Mulani
Peter Noertersheuser
Martin M. Okun
Susan K. Paulson
Joaquin Mario Valdes
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Deutschland GmbH and Co KG
AbbVie Inc
Original Assignee
Abbott GmbH and Co KG
Abbott Laboratories
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2010291927A external-priority patent/AU2010291927A1/en
Application filed by Abbott GmbH and Co KG, Abbott Laboratories filed Critical Abbott GmbH and Co KG
Priority to AU2013202866A priority Critical patent/AU2013202866A1/en
Publication of AU2013202866A1 publication Critical patent/AU2013202866A1/en
Assigned to ABBOTT LABORATORIES, AbbVie Deutschland GmbH & Co. KG reassignment ABBOTT LABORATORIES Request for Assignment Assignors: ABBOTT GMBH & CO. KG, ABBOTT LABORATORIES
Assigned to AbbVie Deutschland GmbH & Co. KG, ABBVIE INC. reassignment AbbVie Deutschland GmbH & Co. KG Request for Assignment Assignors: ABBOTT LABORATORIES, AbbVie Deutschland GmbH & Co. KG
Abandoned legal-status Critical Current

Links

Abstract

METHODS FOR TREATING PSORIASIS The invention provides methods of treating psoriasis in a subject by administering to a subject an antibody capable of binding to the p40 subunit of IL-12 and/or IL-23. 7302524:gcc

Description

1 METHODS FOR TREATING PSORIASIS RELATED APPLICATIONS This application claims priority to U.S. Application Serial No. 61/242,288 5 entitled Methods for Treating Psoriasis , filed on September 14, 2009, U.S. Application Serial No. 61/245,967 entitled Methods for Treating Psoriasis, filed on September 25, 2009, U.S. Application Serial No. 61/297623, entitled Methods for Treating Psoriasis, filed on January 22, 2010, and U.S. Application Serial No. 61/360,299, entitled Methods for Treating Psoriasis, filed on June 30, 2010, the entire contents of each of which are 10 expressly incorporated herein by reference. BACKGROUND OF THE INVENTION Psoriasis is a T cell-mediated inflammatory disease that is considered to be one of the most common autoimmune diseases, affecting approximately 2% to 3% of adults, 15 though the global prevalence varies widely (Stem R.S., et al., J Investig Dermatol Symp Proc 2004, 9: 136-39; Davidson A and Diamond B. N Engl J Med 2001, 345: 340-50; Langley R.G.B., et al., Ann Rheum Dis 2005, 64(Suppl II): iil 8-23). Psoriasis has a major impact on quality of life (de Korte J, et al., J Investig Dermatol Symp Proc 2004, 9: 140-7; Krueger G, et al., Arch Dermatol 2001, 137: 280-4; Finlay AY and Coles EC, 20 Br J Dermatol 1995, 132: 236-44) and is associated with a number of psychological and psychosocial problems (Kimball AB, et al., Am J Clin Dermatol 2005, 6: 383-92; Russo PA, et al., Australas J Dermatol 2004, 45: 155-9). Many traditional psoriasis therapies have toxic adverse effects; therefore, their long-term use is limited (Lebwohl M. and Ali S., J Am Acad Dermatol 2001, 45: 487-98; Lebwohl M. and Ali S., J Am Acad 25 Dermatol 2001, 45: 649-61). In addition, many patients with psoriasis are dissatisfied with traditional therapies (Stem RS, et al., J Investig Dermatol Symp Proc 2004, 9: 136 39; Finlay AY and Ortonne JP, J Cutan Med Surg 2004, 8: 310-20); thus, there is a clear need for therapies that are safer and easier to use and that can be prescribed on a long term basis. 30 Interleukin-12 (IL-12) and the related cytokine IL-23 are members of the IL-12 superfamily of cytokines that share a common p40 subunit (Anderson EJR, et al., Springer Semin Immunopathol 2006, 27: 425-42). Both cytokines contribute to the development of the type IT helper cell (Thl) immune response in psoriasis, but each has 2 a unique role (Rosmarin D and Strober BE, J Drugs Dermatol 2005, 4: 318-25; Hong K, et al., J Immunol 1999, 162: 7480-91; Yawalkar N, et al., J Invest Dermatol 1998, 111: 1053-57). IL-12 primarily stimulates differentiation of ThI cells and subsequent secretion of interferon-gamma, whereas IL-23 preferentially stimulates differentiation of 5 naYve T cells into effector T helper cells (Th 17) that secrete IL-17, a proinflammatory. mediator Rosmarin D and Strober BE, J Drugs Dermatol 2005, 4: 318-25; Harrington Le, et al., Nature Immunol 2005, 6: 1123-32; Park H, et al. Nature Immunol 2005, 6: 1132-41). The overexpression of IL-12 p40 and IL-23 p40 messenger RNA in psoriatic skin lesions suggests that the inhibition of IL-12 and IL-23 with a neutralizing antibody 10 to the IL-12/23 p40 subunit protein may offer an effective therapeutic approach for the treatment of psoriasis (Yawalkar N, et al., J Invest Dermatol 1998, 111: 1053-57; Lee E, et al., J Exp Med 2004, 199: 125-30; Shaker OG, et al., Clin Biochem 2006, 39: 119-25; Piskin G, et al., J Immunol 2006, 176: 1908-15). Such therapeutic approaches for the treatment of psoriasis are clearly needed in the art. 15 SUMMARY OF THE INVENTION The present invention provides methods and compositions for treating psoriasis, e.g., chronic psoriasis, using an antibody, or antigen-binding portion thereof, that binds human IL-12 and/or human IL-23. 20 In one aspect, the invention provides methods of treating psoriasis in a subject comprising administering to the subject a first dose amount of an antibody, or antigen binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a periodicity, and administering a second dose amount of the antibody, or antigen-binding portion thereof, at the same periodicity, thereby treating 25 psoriasis in the subject. In another aspect, the invention provides methods of treating psoriasis in a subject comprising administering to the subject a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a first periodicity, and administering a second dose amount of 30 the antibody, or antigen-binding portion thereof, according to a second periodicity, thereby treating psoriasis in the subject. In various embodiments, the first dose amount of the antibody, or antigen binding portion thereof, is at least about 100 mg to about 200 mg, is at least about 100 3 mg, or is at least about 200 mg. In other embodiments, the first dose amount of the antibody, or antigen-binding portion thereof, is about 100 mg, 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg. 5 The second dose amount of the antibody, or antigen-binding portion thereof, may be the same as the first dose amount of the antibody, or antigen-binding portion thereof, or different than the first dose amount of the antibody, or antigen-binding portion thereof. In various embodiments, the second dose amount of the antibody, or antigen binding portion thereof, is at least about 100 mg to about 200 mg, is at least about 200 10 mg, or is at least about 100 mg. In other embodiment, the second dose amount of the antibody, or antigen-binding portion thereof, is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, or about 190-210% of the first dose amount of the antibody, or antigen-binding portion thereof. In other embodiments, the first dose amount of the antibody, or antigen-binding portion thereof, is about 100 mg, 15 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, 200 mg. The first and second periodicities of administration of the antibody, or antigen binding portion thereof, may be about once a week, about once every other week, about once every four weeks. In one embodiment, the second periodicity of administration of 20 the antibody, or antigen-binding portion thereof, is about once every 30-200 days. The duration of the first periodicity may be about 12 weeks, about 8 weeks, about 4 weeks, about 2 weeks, or about 1 week. The duration of the first periodicity may be at least about 12 weeks, at least about 8 weeks, at least about 4 weeks, at least about 2 weeks, or at least about 1 week. 25 The duration of the second periodicity may be about 60 weeks, about 44 weeks, about 12 weeks, about 4 weeks, about 2 weeks, or about 1 week. The duration of the second periodicity may be at least about 60 weeks, at least about 44 weeks, at least about 12 weeks, at least about 4 weeks, at least about 2 weeks, or at least about 1 week. In one embodiment, the second dose amount is administered to the subject upon 30 a flare of psoriasis. In another embodiment, the second dose amount is administered to the subject prior to a flare of psoriasis. The flare of psoriasis may be indicated by loss of a Psoriasis Area and Severity Index (PASI) 90 response, by loss of a Psoriasis Area and Severity Index (PASI) 75 4 response, by loss of a Psoriasis Area and Severity Index (PASI) 50 response, or by loss of a clear or minimal Physician's Global Assessment (PGA) rating. The loss of a PASI response may be loss of PASI response of a single body region, loss of PASI response of two body regions, loss of PASI response of three body 5 regions, or loss of PASI response of four body regions. The body region may be trunk, lower extremities, upper extremities, or head and neck. In another aspect, the invention provides a method of treating psoriasis in a subject comprising administering to the subject an antibody, or antigen-binding portion 10 thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, according to a periodicity of about once every 4 weeks, thereby treating psoriasis in the subject. In yet another aspect, the invention provides a method of treating psoriasis in a subject comprising administering to the subject an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or 1L-23, according 15 to a periodicity of about once every 12 weeks, thereby treating psoriasis in the subject. - In a related aspect, the invention provides a method of treating psoriasis in a subject comprising administering to the subject: a) a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23; and b) a second dose amount that is about 40-60% of the first dose amount 20 of the antibody, or antigen-binding portion thereof, according to a periodicity of about once every 12 weeks, thereby treating psoriasis in the subject. In one embodiment, the subject achieves at least a PGA score of 0 or 1. In one embodiment, the subject achieves at least a PASI 75 response. In one embodiment, the subject achieves at least a PASI 90 response. In one embodiment, the subject achieves 25 at least a PASI 100 response. In one embodiment, the subject maintains the PGA score of 0 or I during treatment. In one embodiment, the subject maintains the PASI 75 response during treatment. In one embodiment, the subject maintains the PASI 90 response during treatment. In one embodiment, the first dose amount is at least about 200 mg. 30 In one embodiment, the second dose amount is at least about 100 mg. In another aspect, the invention provides a method of treating psoriasis in a subject comprising administering to the subject: a) a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 5 12 and/or IL-23, according to a first periodicity of about once every 4 weeks; and b) administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks, thereby treating psoriasis in the subject. 5 In one embodiment, the first dose amount is at least about 200 mg. In one embodiment, the second dose amount is at least about 100 mg. In one embodiment, the duration of the first periodicity is at least about 8 weeks. In one embodiment, the duration of the second periodicity is at least about 4 weeks, at least about 16 weeks, or at least about 44 weeks. 10 In another aspect, the invention provides a method of.treating psoriasis in a subject comprising administering to the subject: a) a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL 12 and/or IL-23, according to a first periodicity of about once every 4 weeks; and b) a second dose amount that is about 40-60% of the first dose amount of the antibody, or 15 antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks; and c) the second dose amount of the antibody, or antigen-binding portion thereof, according to a third periodicity of about once every 12 weeks, thereby treating psoriasis in the subject. In one embodiment, the first dose amount is at least about 200 mg. 20 In one embodiment, the second dose amount is at least about 100 mg. In one embodiment, the duration of the first periodicity is at least about 8 weeks. In one embodiment, the duration of the second periodicity is at least about 4 weeks. In one embodiment, the duration of the third periodicity is at least about 12 25 weeks or at least about 36 weeks. In one embodiment, the subject achieves a PGA score of 0 or 1, e.g., by about week 12. In one embodiment, the subject achieves at least a PASI 75 response, e.g., by about week 12. In one embodiment, the subject achieves at least a PASI 90 response, e.g., by about week 12. In one embodiment, the subject achieves at least a PASI 100 30 response, e.g., by about week 12. In one embodiment, the subject maintains the PGA score of 0 or I through the duration of treatment. In one embodiment, the subject maintains the PASI 75 response 6 through the duration of treatment. In one embodiment, the subject maintains the PASI 90 response through the duration of treatment. In another aspect, the invention provides a method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an 5 antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 60% of the population of subjects achieve a PASI 75 response by about week 12. In yet another aspect, the invention provides a method of treating psoriasis in a population of subjects comprising administering to each subject in the population an 10 antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein at least 25% of the population of subjects achieve a PASI 90 response by about week 12. In still another aspect, the invention provides a method of treating psoriasis in a population of subjects comprising administering to each subject in the population an 15 antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 10% of the population of subjects achieve a PASI 100 response by about week 12. In one embodiment, the method comprises administering to each subject in the population: a) a first dose amount of the antibody, or antigen-binding portion thereof, 20 according to a first periodicity of about once every 4 weeks; and b) administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks. In one embodiment, the method comprises administering to each subject in the 25 population: a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen binding portion thereof, according to a second periodicity of about once every 4 weeks; and c) the second dose amount of the antibody, or antigen-binding portion thereof, 30 according to a third periodicity of about once every 12 weeks. In one embodiment, the antibody is administered subcutaneously. In one embodiment, the antibody is a human antibody. In a preferred embodiment, the antibody is ABT-874.
7 In one embodiment, the subject or population of subjects achieves at least a PASI 75 response by about week 24 or at least a PASI 75 response by about week 52. In another embodiment, the subject or population of subjects achieves at least a PGA score of 0 or 1 by about week 24 or at least a PGA score of 0 or 1 by about week 52. 5 In another aspect, the invention is directed to a method of treating psoriasis in a population of subjects, by administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL 12 and/or IL-23, wherein at least 41% of the population of subjects achieve at least a PASI 75 response by about week 24. 10 In yet another aspect, the invention is directed to a method of treating psoriasis in a population of subjects, by administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL 12 and/or IL-23, wherein at least 35% of the population of subjects achieve at least a PGA score of 0 or 1 by about week 24. 15 In a further aspect, the invention is directed to a method of treating psoriasis in a population of subjects, by administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL 12 and/or IL-23, wherein at least 25% of the population-of subjects achieve at least a PASI 75 response by about week 52. 20 In another aspect, the invention is directed to a method of treating psoriasis in a population of subjects, by administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL 12 and/or IL-23, wherein at least 21% of the population of subjects achieve at least a PGA score of 0 or 1 by about week 52. 25 In certain embodiments of the foregoing aspects, the subject or population of subjects achieves (i) an improvement in a Dermatology Life Quality Index (DLQI) score or mean Dermatology Life Quality Index (DLQI) score of at least about -9; (ii) an improvement in a Short Form 36 Health Survey Physical Component Summary (PCS) score or mean Physical Component Summary (PCS) score of at least about 2; (iii) an 30 improvement in a Short Form 36 Health Survey Mental Component Summary (MCS) score or mean Short Form 36 Health Survey Mental Component Summary (MCS) score of at least about 4; (iv) an improvement in a visual analog scale score or mean visual analog scale score for psoriasis-related pain (VAS-Ps) of at least about -25; (v) an 8 improvement in a visual analog scale score for psoriatic arthritis-related pain (VAS-PsA) or mean visual analog scale score for psoriatic arthritis-related pain (VAS-PsA) of at least about -32; and/or (vi) a minimum clinically important difference (MCID) response rate for psoriasis-related pain (VAS.-Ps) of at least about 60%. 5 In various aspects, the invention is directed to a method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein the population of subjects achieves (i) a minimum clinically important difference (MCID) response rate for Dermatology Life 10 Quality Index (DLQI) of at least about 70% by about week 12; (ii) a minimum clinically important difference (MCID) response rate for Dermatology Life Quality Index (DLQI) of at least about 81% by about week 52; (iii) a minimum clinically important difference (MCID) response rate for Total Activity Impairment (TAI) of at least about 45% by about week 12; and/or (iv) a minimum clinically important difference (MCID) response 15 rate for Total Activity Impairment (TAI) of at least about 57% by about week 52. In one embodiment, the antibody, or antigen-binding portion thereof, is administered once every four weeks. In another embodiment, the antibody, or antigen-binding portion thereof, is administered once every 12 weeks. In further aspects, the invention is directed to a method of treating psoriasis in a 20 population of subjects, by administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein (i) at least 65% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject was treated with a biologic prior to administration of the antibody; (ii) at least 74% of the population of subjects 25 achieve at least a PASI 75 response by about week 12, wherein each subject was treated with a biologic prior to administration of the antibody; (iii) at least 78% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein none of the subjects were treated with a biologic prior to administration of the antibody; (iv) at least 82% of the population of subjects achieve at least a PASI 75 response by 30 about week 12, wherein none of the subjects were treated with a biologic prior to administration of the antibody; (v) at least 78% of the population of subjects achieve at least a PGA 0/1 response by about week 52, wherein each subject was treated with a biologic prior to administration of the antibody; (vi) at least 79% of the population of 9 subjects achieve at least a PGA 0/1 response by about week 52, wherein none of the subjects were treated with a biologic prior to administration of the antibody; (vii) at least 7 1% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject has a prior history of psoriatic arthritis; (viii) at least 78% of the 5 population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject has a prior history of psoriatic arthritis; (ix) at least 77% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein none of the subjects has a prior history of psoriatic arthritis; (x) at least 81% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein none of the 10 subjects has a prior history of psoriatic arthritis; (xi) at least 77% of the population of subjects achieve at least a PGA 0/1 response by about week 52, wherein each subject has a prior history of psoriatic arthritis; and/or (xii) at least 79% of the population of subjects achieve at least a PGA 0/1 response by about week 52, wherein none of the subjects has a prior history of psoriatic arthritis. 15 In yet another aspect, the invention is directed to methods for decreasing the risk that a subject treated with an antibody, or antigen binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, will develop a Major Adverse Cardiovascular Event (MACE). The methods include (a) selecting a subject having less that 2 risk factors selected from the group consisting of (i) a body mass 20 index (BMI) of greater than 30, (ii) a history of diabetes mellitus, (iii) blood pressure greater than 140/90, (iv) a history of myocardial infarction, (v) a history of angina requiring hospitalization, (vi) a history of coronary artery disease requiring revascularization, (vii) a history of peripheral artery disease, (viii) a history of congestive heart failure requiring hospitalization, (ix) a history of stroke or transient 25 ischemic attack; and (b) administering the antibody, or antigen binding portion thereof to the selected subject; thereby decreasing the risk that the subject will develop a Major Adverse Cardiovascular Event. In a particular embodiment, the antibody is ABT-874 or ustekinumab. In certain embodiments, the subject has 0 or I risk factor. In certain 30 embodiments, the MACE is myocardial infarction and/or cerebrovascular stroke. In other embodiments, the antibody, or antigen binding portion thereof, is administered to the selected subject in a first dose amount of at least about 100 mg to about 200 mg. In a further embodiment, the antibody, or antigen binding portion 10 thereof, is administered to the selected subject in a second dose amount of at least about 100 mg to about 200 mg. In certain embodiments, the risk factors are re-evaluated prior to administration of the second dose amount to the selected subject. In certain embodiments of the various aspects of the invention, the subject 5 achieves at least a 50% reduction in PASI score. In one aspect the subject achieves at least a 50% reduction in PASI score by about week 4. In other embodiments of the various aspects of the invention, the subject achieves at least an 80% reduction in PASI score. In one aspect the subject achieves at least an 80% reduction in PASI score by about week 12. 10 In further aspects, the invention is directed to a method of treating psoriasis in a population of subjects, comprises administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein: (i) at least 69% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a 15 baseline PASI greater than 20 prior to administration of the antibody; (ii) at least 79% of the populatin of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline PASI less than or equal to 20 prior to administration of the antibody; (iii) at least 79% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline PASI greater than 20 20 prior to administration of the antibody; (iv) at least 81% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline PASI less than or equal to 20 prior to administration of the antibody; (v) at least 67% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline weight of greater than or equal to 100 kilograms 25 prior to administration of the antibody; (vi) at least 80% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline weight of less than 100 kilograms prior to administration of the antibody; (vii) at least 72% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline weight of greater than or equal to 100 30 kilograms prior to administration of the antibody; and/or (viii) at least 85% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline weight of less than 100 kilograms prior to administration of the antibody.
11 In still further aspects, the invention is directed to a method of treating psoriasis in a population of subjects comprises administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein: (i) at least 41% of the population of subjects 5 maintains at least a PGA 0/1 response through at least week 52 of treatment; (ii) at least 79% of the population of subjects maintains at least a PGA 0/1 response through at least week 52 of treatment; (iii) at least 45% of the population of subjects maintains at least a PASI 75 response through at least week 52 of treatment; (iv) at least 82% of the population of subjects maintains at least a PASI 75 response through at least week 52 of 10 treatment; (v) at least 23% of the population of subjects maintains at least a PASI 75 response through at least week 52 of treatment; and/or (vi) at least 63% of the population of subjects maintains at least a PASI 75 response through at least week 52 of treatment. In certain embodiments of the various aspects of the invention, the method of 15 treating psoriasis comprises administering to each subject in a population: a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks. 20 In other embodiments of the various aspects of the invention, the method of treating psoriasis comprises administering to each subject in a population: a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) a second dose amount that is about 40 60% of the first dose amount of the antibody, or antigen-binding portion thereof, 25 according to a second periodicity of about once every 4 weeks; and c) the second dose amount of the antibody, or antigen-binding portion thereof, according to a third periodicity of about once every 12 weeks. In certain embodiments of the various aspects of the invention, a subject treated for psoriasis achieves a PGA of 0 or 1 in less than about 171 days. In some 30 embodiments, a subject treated for psoriasis achieves a PGA of 0 or 1 in less than about 30, 40, 50, 60, 70, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 166, 167, 168, 169 or 170 days. In certain embodiments, a patient achieves a PGA of 0 or 1 by about 69 days.
12 In related embodiments of the various aspects of the invention, the patient achieves a PASI 75 response in less than about 140 days. In some embodiments, a subject treated for psoriasis achieves a PASI 75 in less than about 30, 40, 50, 60, 70, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 136, 137, 138 or 139 days.In certain 5 embodiments, the patient achieves a PASI 75 by about 56 days. In still other embodiments of the various aspects of the invention, the subject achieves at least a 60% improvement in PASI score and maintain at least a 60% improvement in PASI score, e.g., through at least week 52 of treatment. In another aspect, the invention is directed to a method of treating psoriasis in a 10 population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein: (i) at least 10% of the population of subjects achieves a PGA score of 0 by week 24 of treatment; (ii) at least 5% of the population of subjects achieve at least a PASI 50 response by about week 2; (iii) at least 70% of the 15 population of subjects achieve at least a PASI 50 response and maintain at least a PASI 50 response through at least week 52 of treatment; (iv) at least 5% of the population of subjects achieve at least a PASI 75 response by about week 4; (v) at least 40% of the population of subjects achieve at least a PASI 75 response and maintain at least a PASI 75 response through at least week 52 of treatment; (vi) at least 10% of the population of 20 subjects achieve at least a PASI 90 response by about week 8; (vii) at least 25% of the population of subjects achieve at least a PASI 90 response and maintain at least a PASI 90 response through at least week 52 of treatment; (viii) at least 5% of the population of subjects achieve at least a PASI 100 response by about week 8; (ix) at least 10% of the population of subjects achieve at least a PASI 100 response and maintain at least a PASI 25 100 response through at least week 52 of treatment; (x) at least 5% of the population of subjects achieve at least a PGA score of 0 or 1 by about week 4; and/or (xi) at least 35% of the population of subjects achieve- at least a PGA score of 0 or 1 and maintain at least a PGA score of 0 or 1 through at least week 52 of treatment. In certain embodiments of the various aspects of the invention, the subject 30 achieves a Nail Psoriasis Severity Index (NAPSI) score of about 2.1 or less. In certain embodiments, the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 2.1 or less by about week 24. In related embodiments of the various aspects of the invention, the subject achieves a Nail Psoriasis Severity Index (NAP$I) score of about 13 1.2 or less: In certain embodiments, the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 1.2 or less by about week 52. In other embodiments of the various aspects of the invention, the subject achieves a Dermatology Life Quality Index (DLQI) score of about 0 or 1. In certain 5 embodiments, the subject achieves a Dermatology Life Quality Index (DLQI) score of about 0 or 1 by about week 24 or by about week 52. In certain embodiments, the subject achieves a clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score. A clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score may be, e.g., a decrease of greater than 5 10 points in DLQI score. In one embodiment, the subject achieves a clinically meaningful reduction in DLQI score by about week 24. In one embodiment, the subject achieves a clinically meaningful reduction in DLQI score by about week 52. In certain embodiments, the subject or population of subjects achieves an improvement in Dermatology Life Quality Index (DLQI) score of at least about -7, e.g., 15 by week 12. In a further aspect, the invention is directed to a method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein: (i) at least 35% of the population of subjects 20 achieves a Dermatology Life Quality Index (DLQI) score of 0 or 1 by about week 24; (ii) at least 18% of the population of subjects achieves a Dermatology Life Quality Index (DLQI) score of 0 or 1 by about week 52; (iii) at least 50% of the population of subjects achieves a clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score by about week 24; and/or (iv) at least 20% of the population of subjects achieves a 25 clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score by about week 52. In several embodiments of the various aspects of the invention, the subject achieves a minimum clinically important difference (MCID) in one or more health related quality of life outcomes selected from the group consisting of Dermatology Life 30 Quality Index (DLQI), Total Activity Impairment (TAI), Ps-related (VAS-Ps) pain, psoriatic arthritis-related (VAS-PsA) pain, Short Form 36 Health Survey Mental Component Summary score (MCS) and Short Form 36 Health Survey Mental Component Summary score (PCS). In various embodiments, the subject achieves a 14 minimum clinically important difference (MCID) in two, three, four, five or all six of Dermatology Life Quality Index (DLQI), Total Activity Impairment (TAI), Ps-related (VAS-Ps) pain, psoriatic arthritis-related (VAS-PsA) pain, Short Form 36 Health Survey Mental Component Summary score (MCS) or Short Form 36 Health Survey 5 Physical Component Summary score (PCS). In related embodiments, the population of subjects achieves a minimum clinically important difference (MCID) response rate for one or more health-related quality of life outcomes selected from the group consisting of Dermatology Life Quality Index (DLQI), Total Activity Impairment (TAI), Ps-related (VAS-Ps) pain, psoriatic 10 arthritis-related (VAS-PsA) pain, Short Form 36 Health Survey Mental Component Summary score (MCS) and Short Form 36 Health Survey Mental Component Summary score (PCS). In various embodiments, the population of subjects achieves a minimum clinically important difference (MCID) response rate for two, three, four, five or all six of Dermatology Life Quality Index (DLQI), Total Activity Impairment (TAI), Ps-related 15 (VAS-Ps) pain, psoriatic arthritis-related (VAS-PsA) pain, Short Form 36 Health Survey Mental Component Summary score (MCS) or Short Form 36 Health Survey Physical Component Summary score (PCS). In one embodiment of all of the foregoing aspects of the invention, the method comprises administering to the subject or to each subject in the population: a) a first dose 20 amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks. In another embodiment of all of the foregoing aspects of the invention, the 25 method comprises administering to the subject or to each subject in the population: a) about 200 mg of ABT-874 once every four weeks for two doses; and b) about 100 mg of ABT-874 every four weeks thereafter. In still another embodiment of all of the foregoing aspects of the invention, the method comprises administering to the subject or to each subject in the population: a) 30 about 200 mg of ABT-874 at weeks 0 and 4; and b) about 100 mg of ABT-874 at week 8 and every 4 weeks thereafter. In one embodiment, the antibody is ABT-874 (i.e., briakinumabm).
15 In a further aspect, the invention provides a method of treating psoriasis in a subject comprising administering to the subject: a) about 200 mg of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, once every four weeks for two doses; and b) about 100 mg of the antibody, 5 or antigen-binding portion thereof, every four weeks thereafter, thereby treating psoriasis in the subject. In one embodiment, the antibody is ABT-874. In one embodiment, the psoriasis is plaque psoriasis, e.g., chronic plaque psoriasis, such as moderate to severe chronic plaque psoriasis. In yet a further aspect, the invention provides a method of treating psoriasis in a 10 subject comprising administering to the subject: a) about 200 mg of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, at weeks 0 and 4; and b) about 100 mg of the antibody, or antigen-binding portion thereof, at week 8 and every 4 weeks thereafter, thereby treating psoriasis. in the subject. In one embodiment, the antibody is ABT-874. In one embodiment, the psoriasis 15 is plaque psoriasis, e.g., chronic plaque psoriasis, such as moderate to severe chronic plaque psoriasis. In a still further aspect, the invention provides a method of treating psoriasis in a subject comprising administering to the subject: a) about 200 mg of ABT-874 once every four weeks for two doses; and b) about 100 mg of ABT-874 every four weeks 20 thereafter, thereby treating psoriasis in the subject. In one embodiment, the antibody is ABT-874. In one embodiment, the psoriasis is plaque psoriasis, e.g., chronic plaque psoriasis, such as moderate to severe chronic plaque psoriasis. In a still further aspect, the invention provides amethod of treating psoriasis in a subject comprising administering to the subject: a) about 200 mg of ABT-874 at weeks 0 25 and 4; and b)about 100 mg of ABT-874 at week 8 and every 4 weeks thereafter, thereby treating psoriasis in the subject. In one embodiment, the antibody is ABT-874. In one embodiment, the psoriasis is plaque psoriasis, e.g., chronic plaque psoriasis, such as moderate to severe chronic plaque psoriasis. In one embodiment, the psoriasis is chronic psoriasis. In one embodiment, the 30 psoriasis is plaque psoriasis, e.g., chronic plaque psoriasis. In another embodiment, the psoriasis is chronic psoriasis, e.g., chronic plaque psoriasis. In yet another embodiment, the psoriasis is moderate to severe psoriasis, e.g., moderate to severe plaque psoriasis, moderate to severe chronic psoriasis or moderate to severe chronic plaque psoriasis. In 16 one embodiment, the subject has had a clinical diagnosis of psoriasis for at least 6 months. In another embodiment, the subject has had stable plaque psoriasis for at least 2 months. In one embodiment, the antibody is administered via subcutaneous injection. 5 In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention is capable of binding to an epitope of the p40 subunit of IL- 12 and/or IL-23. In another embodiment, the antibody, or antigen-binding portion thereof, is capable of binding to the epitope of the p40 subunit when the p40 subunit is bound to the 10 p 3 5 subunit of IL-12. In yet another embodiment, the antibody, or antigen-binding portion thereof, is capable of binding to the epitope of the p40 subunit when the p40 subunit is bound to a p19 subunit, i.e., the p19 subunit of IL-23. In one embodiment, the antibody, or antigen-binding portion thereof, is capable of binding to the epitope of the p40 subunit when the p40 subunit is bound to the p35 subunit of IL-12 and when the 15 p 4 0 subunit is bound to a p19 subunit. In one embodiment, the antibody, or antigen binding portion thereof, binds to an epitope of the p40 subunit of IL-12 to which an antibody selected from the group consisting of Y61 and J695 binds. In another embodiment, the antibody is further capable of binding to a first 20 heterodimer and is also capable of binding to a second heterodimer, wherein the first heterodimer comprises the p40 subunit of 1-12 and the p35 subunit of 11-12, and wherein the second heterodimer comprises the p40 subunit of IL-12 and a p19 subunit, i.e., the p19 subunit of IL-23. In a further embodiment, the antibody neutralizes the activity of the first 25 heterodimer. In another embodiment, the antibody neutralizes the activity of the second heterodimer. In yet another embodiment, the antibody neutralizes the activity of the first heterodimdr and the second heterodimer. In a further embodiment, the antibody, or antigen binding portion thereof, used in the methods of the invention inhibits phytohemagglutinin blast proliferation in an in 30 vitro PHA assay with an IC 50 of 1 x 10-9 M or less, or which inhibits human IFNy production with an IC 50 of I x 1 0 -( M or less. In one embodiment, the antibody, or antigen binding portion thereof, used in the methods of the invention dissociates from the p40 subunit of IL-12 with a Kd of 1 x 0-' 17 M or less or a kort rate constant of 1 x 10- s or less, as determined by surface plasmon resonance. In one embodiment, the isolated antibody, or antigen binding portion thereof, used in the methods of the invention is a chimeric antibody, a humanized antibody or a 5 human antibody. In another embodiment, the antibody, or antigen binding portion thereof, used in the methods of the invention has a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 25 and a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 26; 10- In a further embodiment, the antibody, or antigen binding portion thereof, used in the methods of the invention has a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 27 and a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 28. In one embodiment, the antibody, or antigen binding portion thereof, used in the 15 methods of the invention has a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 29 and a light chain CDRI comprising the amino acid sequence of SEQ ID NO: 30. In another embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention is capable of binding to an interleukin comprising a p40 . 20 subunit. In one embodiment, the interleukin comprises a p40 subunit and a p35 subunit, e.g., the interleukin is IL-12. In another embodiment, the interleukin comprises a p 4 0 subunit and a p19 subunit, e.g., the interleukin is IL-23. In yet another embodiment, the antibody, or antigen binding portion thereof, neutralizes the activity of the interleukin. In one embodiment, the antibody, or antigen binding portion thereof, binds to an 25 epitope of the p40 subunit. In one embodiment, the antibody, or antigen-binding portion thereof, is administered to a subject in a pharmaceutical composition comprising the antibody, or antigen binding portion thereof, and a pharmaceutically acceptable carrier. The pharmaceutical composition may also comprise an additional agent, such as a 30 therapeutic agent, e.g., budenoside, epidermal growth factor, corticosteroids, cyclosporin, sulfasalazine, aminosalicylates, 6-mercaptopurine, azathioprine, metronidazole, lipoxygenase inhibitors, mesalamine, olsalazine, balsalazide, antioxidants, thromboxane inhibitors, IL-I receptor antagonists, anti-IL-1p monoclonal 18 antibodies, anti-IL-6 monoclonal antibodies, growth factors, elastase inhibitors, pyridinyl-imidazole compounds, antibodies or agonists of TNF, LT, IL-1, IL-2, IL-6, IL 7, IL-8, IL-15, IL-16, IL-18, EMAP-II, GM-CSF, FGF, and PDGF, antibodies of CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands, 5 methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, ibuprofen, corticosteroids, prednisolone, phosphodiesterase inhibitors, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, IRAK, NIK, IKK, p38, MAP kinase inhibitors, IL-1p converting enzyme inhibitors, TNFq converting enzyme inhibitors, T-cell signaling inhibitors, metalloproteinase 10 inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors, soluble p55 TNF receptor, soluble p75 TNF receptor, sIL- I RI, sIL- 1 RII sIL-6R, anti-inflammatory cytokines, IL-4, IL- 10, IL 11, IL-13 and TGFp. In another embodiment, the therapeutic agent in the pharmaceutical composition 15 administered to the subject may be selected from the group consisting of anti-TNF antibodies and antibody fragments thereof, TNFR-Ig constructs, TACE inhibitors, PDE4 inhibitors, corticosteroids, budenoside, dexamethasone, sulfasalazine, 5-aminosalicylic acid, olsalazine, IL-I P converting enzyme inhibitors, IL-Ira, tyrosine kinase inhibitors, 6-mercaptopurines and IL-11. 20 In another embodiment, the therapeutic agent may be selected from the group consisting of corticosteroids, prednisolone, methylprednisolone, azathioprine, cyclophosphamide, cyclosporine, methotrexate, 4-aminopyridine, tizanidine, interferon plia, interferon-p1b, Copolymer 1, hyperbaric oxygen, intravenous immunoglobulin, clabribine, antibodies or agonists of TNF, LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-15, IL-16, 25 IL-18, EMAP-II, GM-CSF, FGF, PDGF, antibodies to CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands, methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, ibuprofen, corticosteroids, prednisolone, phosphodiesterase inhibitors, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, IRAK, NIK, 30 IKK, p38 or MAP kinase inhibitors, IL-1p converting enzyme inhibitors, TACE inhibitors, T-cell signaling inhibitors, kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors, soluble p55 TNF receptor, soluble p75 TNF 19 receptor, sIL-IRI, sIL-IRII, sIL-6R, sIL-13R, anti-P7s, p-selectin glycoprotein ligand (PSGL), anti-inflammatory cytokines, IL-4, IL-10, IL-13 and TGFp. In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention binds to human IL-12 and/or human IL-23 and dissociates 5 from human IL-12 and/or human IL-23, respectively, with a Kd of 1 x 10' 0 M or less and a kof rate constant of 1 x 10- 3 s- or less, as determined by surface plasmon resonance. In one embodiment, the antibody, or antigen-binding portion thereof, dissociates from human IL-12 and/or human IL-23 with a koff rate constant of 1 'x 10~ 4 s or less. In another embodiment, the antibody, or antigen-binding portion thereof, 10 dissociates from human IL-12 and/or human IL-23 with a koff rate constant of I x 10-5s or less. In another embodiment, the antibody, or antigen-binding portion thereof, binds to human IL-12 and/or human IL-23 and dissociates from human 1L-12 and/or human I 23, respectively, with a koff rate constant of 1 x 10-2 S or less, as determined by surface 15 plasmon resonance. In yet another embodiment, the antibody, or antigen-binding portion thereof, dissociates from human IL-12 and/or human IL-23 with a koff rate constant of 1 x 10- 3 s-1 or less. In a still further another embodiment, the antibody, or antigen-binding portion thereof, dissociates from human IL-12 and/or human IL-23 with a kogf rate constant of I x 10-4 s- or less. In another embodiment, the antibody, or 20 antigen-binding portion thereof, dissociates from human IL-12 and/or human IL-23 with a koff rate constant of 1 x 10 -s- or less. In still another embodiment, the antibody, or antigen-binding portion thereof, binds to human IL-12 and/or human IL-23 and dissociates from human IL-12 and/or human IL-23, respectively, with a Kd of 1.34 x 10 1 0 M or less. In yet another 25 embodiment, the antibody, or antigen-binding portion thereof, binds to human IL-12 and/or human IL-23 and dissociates from human IL-12 and/or human IL-23, respectively, with a Kd of 9.74 x 10-" M or less. In one embodiment, the antibody, or antigen-binding portion thereof, is a recombinant antibody, or antigen-binding portion thereof. 30 In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention is a neutralizing antibody, e.g., neutralizes the activity of human IL-12 and/or human IL-23. In one embodiment, the neutralizing antibody, or antigen-binding portion thereof, inhibits phytohemagglutinin blast proliferation in an in 20 vitro PHA assay with an IC 50 of 1 x 10-9 M or less. In another embodiment, the neutralizing antibody, or antigen-binding portion thereof, inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-10~M or less. In still another embodiment, the neutralizing antibody of, or antigen-binding portion thereof, 5 inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-1M or less. In yet another embodiment, the neutralizing antibody, or antigen binding portion thereof, inhibits phytohemagglutinin blast proliferation in an in vitro phytohemagglutinin blast proliferation assay (PHA assay) with an IC 50 of 1 x 10- M or less. In still another embodiment, the neutralizing antibody, or antigen-binding portion 10 thereof, inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an
IC
5 o of 1 x 10-8 M or less. In one embodiment, the neutralizing antibody, or antigen binding portion thereof, inhibits human IFNy production with an IC 5 o of 1 x 1010 M or less. In still another embodiment, the neutralizing antibody, or antigen-binding portion thereof, inhibits human IFNy production with an IC 50 of 1 x 10 -" M or less. In yet a 15 further embodiment, the neutralizing antibody, or antigen-binding portion thereof, inhibits human IFNy production with an IC 5 o of 5 x 10-12 M or less. In one embodiment, the-antibody, or an antigen-binding portion thereof, used in the methods of the invention a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay 20 with an IC 50 of 1 x 10- 9 M or less; b) has a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 25; and c) has a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 26. In one embodiment, the antibody further has a heavy chain CDR2 comprising 25 the amino acid sequence of SEQ ID NO: 27; and a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 28. In still another embodiment, the antibody, or antigen-binding portion thereof, further has a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 29; and a light chain CDR I comprising the amino acid sequence of SEQ ID NO: 30. In still another embodiment, the antibody, or antigen 30 binding portion thereof, further inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-10 M or less. In still another embodiment, the antibody, or antigen-binding portion thereof, further inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 5 o of 1 x 1011 M or less.
21 In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention has a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 31, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 32. 5 In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention comprises a heavy chain constant region selected from the group consisting of IgGi, IgG2, IgG3, IgG4, IgM, IgA and IgE constant regions. In one embodiment, the antibody heavy chain constant region is IgG1. In another embodiment, the antibody is a Fab fragment, F(ab') 2 fragment, or a single chain Fv fragment. 10 In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention dissociates from human IL- 12 and/or human IL-23 with a Kd of 1 x 10-1 M or less and binds to an epitope on the p40 subunit of human IL-12 and/or human IL-23. In one embodiment, the antibody, or antigen-binding portion thereof, used in the 15 methods of the invention is a human antibody, or antigen-binding portion thereof, which a) dissociates from human IL- 12 with a koff rate constant of 1 x 10- 3 s-I or less, as determined by surface plasmon resonance; b) has a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 25; and 20 c) has a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 26. In another embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention dissociates from human IL- 12 with a koff rate constant of 1 x 10- 4 s-1 or less. In a further embodiment, the human antibody, or antigen-binding 25 portion thereof, dissociates from human IL-12 with a koff rate constant of I x 10-5 s-I or less. In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention is a human antibody, or antigen-binding portion thereof, that binds to human IL-12 and comprises: 30 a light chain CDR3 domain comprising the amino acid sequence of SEQ ID NO: 26; and 22 a heavy chain CDR3 domain comprising the amino acid sequence of SEQ ID NO: 25. In one embodiment, the antibody, or antigen-binding portion thereof, has a light chain variable region (LCVR) having a CDR3 domain comprising the amino acid 5 sequence of SEQ ID NO: 26, and has a heavy chain variable region (HCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 25. In another embodiment, the antibody, or antigen-binding portion thereof, comprises an LCVR further having a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 28 and an HCVR further comprising a CDR2 domain comprising the amino acid sequence 10 of SEQ ID NO: 27. In yet another embodiment, the LCVR further has CDRI domain comprising the amino acid sequence of SEQ ID NO: 30 and the HCVR has a CDR 1 domain comprising the amino acid sequence of SEQ ID NO: 29. In one embodiment, the antibody, or antigen-binding portion thereof, binds human IL-12 and/or human IL-23 and is the antibody J695 (also referred to as ABT 15 874), or an antigen binding portion thereof. In one embodiment, the antibody, or antigen-binding portion thereof, binds to human IL-12 and/or human IL-23 and dissociates from human IL-12 and/or human IL 23 with a Kd.of 1.34 x 10-0 M or less, and neutralizes human IL-12 and/or human IL-23. In one embodiment, the antibody, or antigen-binding portion thereof, dissociates from 20 human IL-12 and/or human IL-23 with a Kd of 9.74 x 1011 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 5 0 of I x 10- 7 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10 25 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 5 o of I x 10-9 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 5 0 of 1 x 10 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits 30 phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of I x 101 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits human IFNy production with an IC 50 of I x 1010 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits human IFNy production with an 23
IC
50 of 1 x 10" M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits human IFNy production with an IC 50 of 5 x 1012 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, used in the methods of the invention inhibits IL-12 and/or IL-23 binding to its receptor in an IL-12 5 or IL-23 receptor binding assay (RBA), respectively, with an IC 50 of 1 x 10- 9 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits IL-12 and/or IL-23 binding to its receptor in an IL-12 or IL-23 receptor binding assay (RBA), respectively, with an IC 50 of 1 x 10-0 M or less. In one embodiment, the antibody, or antigen-binding portion thereof, inhibits IL-12 and/or IL-23 binding to its receptor in an 10 IL-12 or IL-23 receptor binding assay (RBA), respectively, with an IC 50 of 1 x 10-" M or less. BRIEF DESCRIPTION OF THE DRAWINGS 15 Figure 1 shows the patient disposition of the trial. (The term "eow" refers to every other week dosing.) Figure 2 shows the percentage of patients with at least a 75% improvement in the psoriasis area and severity index (PASI 75) during the 12-week portion of the trial By week 8, with the exception of the 200 mg x 1 group, the percentage of patients who 20 had a PASI 75 response was statistically significantly greater (p<0.001) in each ABT 874 treatment group for each comparison with placebo based on an analysis of variance of observed data for the intention-to-treat population. (The term "eow" refers to every other week dosing.) Figure 3 shows the mean percentage improvement in psoriasis area and severity 25 index (PASI) scores from baseline. The data show that *p<0.001 for each ABT-874 treatment group compared with placebo at all time points (except 100 mg eow at week 1, p=0.023) based on an analysis of variance, of observed data for the intention-to-treat population. (The term "eow" refers to every other week dosing.) Figures 4A-C show the percentage of patients who maintained a PASI 50, PASI 30 75 and PASI 90 response, respectively, at week 24 of the trial, i.e, at 12 weeks following discontinuation of administration of the antibody. Figure 4D shows the percentage of patients maintaining a PASI 75 response over time during the 24 week period of the trial.
24 Figure 5A displays the mean percentage improvement from baseline in PASI scores from Week 4 to Week 12. Figure 5B displays the mean percentage improvement from baseline in PASI scores from Week 4 to Week 12 post retreatment. 5 Figure 6A displays the serum concentration-time curve for IV dosing of ABT 874. Figure 6B displays the serum concentration-time curve for SC dosing of ABT 874. Figure 7A displays the percentage of patients re-achieving a PASI 75 response 10 following retreatment. Figure 7B displays the median time to achieve a PASI 75 response across all ABT-874 dosage groups during retreatment. Figure 7C displays the median time to loss of a PASI 75 response following the initial 12 weeks of treatment. 15 Figure 7D displays the percentage of patients achieving a PGA score of 0 or 1 following retreatment. Figures 8A-8B show the heavy chain variable region amino acid sequence alignments of a series of human antibodies that bind human IL-12 compared to germline sequences Cos-3/JH3 and Dpl18 Lv1042. Kabat numbering is used to identify amino 20 acid positions. For the Joe 9 wild type, the full sequence is shown. For the other antibodies, only those amino acids positions that differ from Joe 9 wild type are shown. Figures 8C-8D show the light chain variable region amino acid sequence alignments of a series of human antibodies that bind human IL-12. Kabat numbering is used to identify amino acid positions. For the Joe 9 wild type, the full sequence is 25 shown. For the other antibodies, only those amino acids positions that differ from Joe 9 wild type are shown. Figures 9A-9E show the CDR positions in the heavy chain of the Y61 antibody that were mutated by site-directed mutagenesis and the respective amino acid substitutions at each position. The graphs at the right of the figures show the off-rates 30 for the substituted antibodies (black bars) as compared to unmutated Y61 (open bar). Figures 9F-9H show the CDR positions in the light chain of the Y61 antibody that were mutated by site-directed mutagenesis and the respective amino acid 25 substitutions at each position. The graphs at the right of the figures show the off-rates for the substituted antibodies (black bars) as compared to unmutated Y61 (open bar). Figure 10 shows the mean PASI score at time of retreatment. The mean (SD) PASI scores at retreatment for all 58 patients who received retreatment following loss of 5 PASI 50 response during the observation/retreatment phase. N values represent the number of patients receiving retreatment. EOW=every other week; PASI=Psoriasis Area and Severity Index. Figure 11 shows the time to loss of PASI 75 during the observation/retreatment phase. Time to loss of the response during the observation/retreatment phase is depicted; 10 this loss is following primary achievement of a PASI 75 response at the initial 12-week endpoint. Loss of response was calculated from the time of the last dose received during the first 12 weeks of the study. N values represent the number of patients losing response. EOW=every other week; PASI=Psoriasis Area and Severity Index. Figure 12 shows the time to achieve PASI 75 during the observation/retreatment 15 phase. Among patients who achieved a PASI 75 response following ABT-874 retreatment during the observation/retreatment phase, time to achieve the response is depicted for each ABT-874 treatment group. N values represent the number of patients achieving response. EOW=every other week; PASI=Psoriasis Area and Severity Index. Figure 13 shows the percentage of patients achieving PASI 75 and PASI 90 at 20 week 12. Figures 14A-14D show the percentage of patients achieving PASI 75 and PASI 90 at week 12 in four specific body regions: (A) head and neck; (B) upper extremities; (C) trunk; and (D) lower extremities. Figure 15 shows the percentage of patients achieving PASI 75 and PASI 90 at 25 week 12. Figure 16 shows the percentage of patients achieving PASI 75 at week 12 by baseline body weight. Figure 17 shows the percentage of patients achieving PASI 75 at week 12 in patients with and without a history of psoriatic arthritis. 30 Figure 18 shows the percentage of patients achieving PASI 75 response at week 12 in patients with severe or very severe baseline physician's global assessment. Figure 19 shows the percentage of patients achieving PASI 75 response at week 12 by baseline PASI.
26 Figure 20 shows the VERO study design of Example 12. Figure 21 shows subject disposition throughout the VERO study of Example 12. Figure 22 shows the response of PGA of "clear" or "minimal" at week 12 of the VERO study. 5 Figure 23 shows the PASI Response rates at week 12 of the VERO study. Figure 24 shows the maintenance of PGA 0/1 response rates from week 12 to week 52, analyzed using nonresponder imputation of the VERO study. Figure 25 shows the ppercentages of patients maintaining PASI 75 and 90 responses at week 52, analyzed using nonresponder imputation of the VERO study. 10 Figure 26 shows the timing of major adverse cardiac events in relation to study drug dosing of the VERO study. Figure 27 shows the study design of Example 13. Figure 28 shows the percentage of patients achieving PGA 0/1 ("Clear" or "Minimal") at week 12*. PGA = Physician's Global Assessment; non-responder 15 imputation; P<0.001, ABT-874 vs. placebo and ABT-874 vs. etanercept. Figure 29 shows the percentage of patients achieving a PASI 75 response at week 12*. PASI = Psoriasis Area and Severity Index; *non-responder imputation; P<0.001, ABT-874 vs. placebo and ABT-874 vs. etanercept. Figure 30 shows the percentage of patients achieving a PASI 90 response at 20 week 12*. PASI = Psoriasis Area and Severity Index; *non-responder imputation; P<0.001, ABT-874 vs. placebo and ABT-874 vs. etanercept. Figure 31 shows the percentage of patients achieving a PASI 100 response at week 12*. PASI = Psoriasis Area and Severity:Index; *non-responder imputation; P<0.001, ABT-874 vs. placebo and ABT-874 vs. etanercept. 25 Figure 32 shows the study design in Example 14. Figure 33 shows the percentage of patients achieving a PGA 0/1 ("Clear or "Minimal") at week 12. *P < 0.001, ABT-874 vs. Placebo, 'P < 0.001, ABT-874 vs. ETN, NRI. Figure 34 shows the percentage of patients achieving a PASI 75 Response at 30 week 12. *P < 0.001, ABT-874 vs. Placebo, 'P < 0.001, ABT-874 vs. ETN, NRI. Figure 35 shows the PASI 90 response rate at week 12. *P < 0.001, ABT-874 vs. Placebo, 'P < 0.001, ABT-874 vs. ETN, NRI.
27 Figure 36 shows the PASI 100 response rate at week 12. *P < 0.001, ABT-874 vs. Placebo, tP <0.001, ABT-874 vs. ETN, NRI. Figure 37 depicts the results of certain Phase III clinical studies demonstrating the efficacy of ABT-874 in moderate to sever psoriasis, as described in Example 23. 5 Figure 38 depicts the interim efficacy results and specifically, PASI 75 over time upon treatment with ABT-874, as described in Example 23. Figure 39 depicts the interim efficacy results and specifically, PASI 100 over time upon treatment with ABT-874, as described in Example 23. Figure 40 depicts the frequency of MACE upon administration with ABT-874, as 10 described in Example 23. Figure 41 shows the study design as described in Example 24. A=2:1 randomization; B*=all patients who had a physician's global assessment score of "clear" or "minimal" at week 12 were stratified by treatment received during induction phase and re-randomized 2:2:1. 15 Figure 42 shows the study flow and patient disposition as described in Example 24. *Lack of efficacy was defined as a PGA score of "mild" or higher. tLack of efficacy was defined as a PGA score of "moderate" or higher. Figure 43 shows the physician's global assessment scores of "clear" or "minimal" by treatment group during the induction phase as described in Example 24. 20 Intention-to-treat analysis: patients with missing physician's global assessment scores were considered nonresponders. p<0.001 at week 12, based on Cochran-Mantel Haenszel test adjusted by center; p=O.186 for week 1, and p<0.00I for weeks 4 to 12 based on Chi Square test. Figure 44 shows the mean improvement in psoriasis area and severity index 25 scores by treatment group during the induction phase as described in Example 24. Intention-to-treat analysis: missing psoriasis area and severity scores were imputed with last observation, excluding baseline, carried forward. p<0.001 for all time points based on ANCOVA with treatment as a factor and baseline value as a covariate. Figure 45 shows the maintenance of physician's global assessment scores of 30 "clear" or "minimal" through week 52 (A), and proportions achieving PASI 75 responses (B), and PASI 100 responses (C) through week 52, as described in Example 24. *One patient in the ABT-874 every 4 week group was re-randomized but did not receive any study drug in the Maintenance Phase. tRepresents all patients who received 28 ABT-874 in the Induction Phase, had a physician's global assessment score of "clear" or "minimal" at week 12, and were re-randomized in the Maintenance Phase. Intention-to treat analysis: patients with missing physician's global assessment scores were considered non-responders (A). Intention-to-treat analysis: patients with missing PASI 5 scores were considered non-responders (B and C). p values based on Chi Square test. Figure 46 shows the study design and dosing regimens as described in Example 25. aAt Week 24, patients who achieved PASI >75 and a PGA of 0 or 1 maintained their current weekly MTX dosage through Week 51. The dosage was adjusted only for laboratory abnormalities. Patients in either treatment group who did not achieve PASI 10 >75 or a PGA of 0 or 1 or who lost response (defined as PASI <50 and PGA >3) after Week 24 discontinued the trial and were eligible to enrol in an open-label extension study of briakinumabTM. MTX, methotrexate; PASI, Psoriasis Activity and Severity Index; PGA, Physician's Global Assessment. Figure 47 shows the patient disposition as described in Example 25. MTX, 15 methotrexate. Figure 48 shows the Psoriasis Activity and Severity Index (PASI) responses through Week 52 as described in Example 25. The percentages of patients achieving PASI 75 (A), PASI 50 (B), PASI 90 (C), and PASI 100 (D) were significantly greater in the briakinumabTM group than the methotrexate (MTX) group by Week 8 (or earlier) and 20 at all time points through Week 52. *p<0.001 vs. MTX. Nonresponder imputation was used to handle missing data. Figure 49 shows the mean percentage improvements in Psoriasis Activity and Severity Index (PASI) scores from baseline as described in Example 25. At each visit, the mean percentage improvement from baseline was significantly greater in the 25 briakinumabTM group than the methotrexate (MTX) group (p<0.00 I at all time points). Last observation carried forward was used to handle missing data. Figure 50 shows the percentages of patients achieving a Physician's Global Assessment (PGA) of 0 or 1 ("Clear" or "Minimal") through Week 52 as described in Example 25. *p<0.001 vs. methotrexate (MTX). 30 Figure 51 shows the changes from baseline in Dermatology Life Quality Index (DLQI) score by visit as described in Example 25. Patients in the briakinumabTM group had statistically significantly greater decreases from baseline in DLQI scores compared with the MTX group at all time points assessed. *p<0.001 vs. methotrexate (MTX) 29 DETAILED DESCRIPTION OF THE INVENTION In order that the present invention may be more readily understood, certain terms are first defined. 5 The term "activity enhancing amino acid residue" includes an amino acid residue which improves the activity of the antibody. It should be understood that the activity enhancing amino acid residue may replace an amino acid residue at a contact, hypermutation or preferred selective mutagenesis position and, further, more than one activity enhancing amino acid residue can be present within one or more CDRs. An 10 activity enhancing amino acid residue include, an amino acid residue that improves the binding specificity/affinity of an antibody, for example anti-human IL-12 antibody binding to human IL-12. The activity enhancing amino acid residue is also intended to include an amino acid residue that improves the neutralization potency of an antibody, for example, the human IL-12 antibody which inhibits human IL-12. 15 The term "antibody" includes an immunoglobulin molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHI, CH2 and CH3. Each light 20 chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and 25 VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FRI, CDRI, FR2, CDR2, FR3, CDR3, FR4. In one embodiment, the antibody used in the compositions and methods of the invention is the antibody described in U.S. Patent No. 6,914,128, incorporated by reference herein. In another embodiment, the antibody used in the compositions and methods of the 30 invention is the antibody ABT-874 (also referred to as J695; Abbott Laboratories). The term "antigen-binding portion" of an antibody (or "antibody portion") includes fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., hIL-12). It has been shown that the antigen-binding function of an antibody can be 30 performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH I domains; (ii) a F(ab') 2 fragment, a bivalent fragment comprising two Fab fragments linked by a 5 disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546 ), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for 10 by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883) . Such single chain antibodies are also intended to be encompassed 15 within the term "antigen-binding portion" of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another 20 chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R.J., et al. (1994) Structure 2:1121-1123). Still further, an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecules, formed by covalent or non-covalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of 25 such immunoadhesion molecules include use of the streptavidin core region to make.a tetrameric scFv molecule (Kipriyanov, S.M., etal. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S.M., et al. (1994) Mol. Immunol. 31:1047-1058). Antibody portions, such as Fab and F(ab') 2 30 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, 'antibodies, antibody portions and immunoadhesion molecules can be obtained using standard 31 recombinant DNA techniques, as described herein. Preferred antigen binding portions are complete domains or pairs of complete domains. The term "backmutation " refers to a process in which some or all of the somatically mutated amino acids of a human antibody are replaced with the 5 corresponding germline residues from a homologous germline antibody sequence. The heavy and light chain sequences of the human antibody of the invention are aligned separately with the germline sequences in the VBASE database to identify the sequences with the highest homology. Differences in the human antibody of the invention are returned to the germline sequence by mutating defined nucleotide positions encoding 10 such different amino acid. The role of each amino acid thus identified as candidate for backmutation should be investigated for a direct or indirect role in antigen binding and any amino acid found after mutation to affect any desirable characteristic of the human antibody should not be included in the final human antibody; as an example, activity enhancing amino acids identified by the selective mutagenesis approach will not be 15 subject to backmutation. To minimize the number of amino acids subject to backmutation those amino acid positions found to be different from the closest germline sequence but identical to the corresponding amino acid in a second germline sequence can remain, provided that the second germline sequence is identical and colinear to the sequence of the human antibody of the invention for at least 10, preferably 12 amino 20 acids, on both sides of the amino acid in question. Backmutation may occur at any stage of antibody optimization; preferably, backmutation occurs directly before or after the selective mutagenesis approach. More preferably, backmutation occurs directly before the selective mutagenesis approach. The phrase "human interleukin 12" (abbreviated herein as hIL-12, or IL-12), as 25 used herein, includes a human cytokine that is secreted primarily by macrophages and dendritic cells. The term includes a heterodimeric protein comprising a 35 kD subunit (p35) and a 40 kD subunit (p40) which are both linked together with a disulfide bridge. The heterodimeric protein is referred to as a "p70 subunit". The structure of human IL 12 is described further in, for example, Kobayashi, et al. (1989) J. Exp Med. 170:827 30 845; Seder, et al. (1993) Proc. Nati. Acad. Sci. 90:10188-10192; Ling, et al. (1995) J. Exp Med. 154:116-127; Podlaski, et al. (1992) Arch. Biochem. Biophys. 294:230-237. The term human IL-12 is intended to include recombinant human IL-12 (rh IL-12), which can be prepared by standard recombinant expression methods.
32 The terms "Kabat numbering", "Kabat definitions and "Kabat labeling" are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e. hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or 5 an antigen binding portion thereof (Kabat et al. (1971) Ann. NYAcad, Sci. 190:382-391 and , Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91 3242). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino 10 acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3. The Kabat numbering is used herein to indicate the positions of amino acid modifications made in antibodies of the invention. For example, the Y61 anti-IL-12 15 antibody can be mutated from serine (S) to glutamic acid (E) at position 31 of the heavy chain CDR1 (H31S -+ E), or glycine (G) can be mutated to tyrosine (Y) at position 94 of the light chain CDR3 (L94G -+ Y). The term "human antibody" includes antibodies having variable and constant regions corresponding to human germline immunoglobulin sequences as described by 20 Kabat et al. (See Kabat, et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91 3242). The human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for 25 example in the CDRs and in particular CDR3. The mutations preferably are introduced using the "selective mutagenesis approach" described herein. The human antibody can have at least one position replaced with an amino acid residue, e.g., an activity enhancing amino acid residue which is not encoded by the human germline immunoglobulin sequence. The human antibody can have up to twenty positions 30 replaced with amino acid residues which are not part of the human germline immunoglobulin sequence. In other embodiments, up to ten, up to five, up to three or up to two positions are replaced. In a preferred embodiment, these replacements are within the CDR regions as described in detail below. However, the term "human antibody", as 33 used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. The phrase "recombinant human antibody" includes human antibodies that are 5 prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell (described further in Section II, below), antibodies isolated from a recombinant, combinatorial human antibody library (described further in Section III, below), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see 10 e.g., Taylor, L.D., et al. (1992) Nucl. Acids Res. 20:6287-6295) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences (See Kabat, E.A., et al. (1991) Sequences of Proteins of 15 Immunological Interest, Fifth Edition, U.S. Department of Health, and Human Services, NIH Publication No. 91-3242). In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, 20 while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. In certain embodiments, however, such recombinant antibodies are the result of selective mutagenesis approach or backmutation or both. An "isolated antibody" includes an antibody that is substantially free of other 25 antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds hIL-12 is substantially free of antibodies that specifically bind antigens other than hIL-12). An isolated antibody that specifically binds hIL-12 may bind IL-12 molecules from other species (discussed in further detail below). Moreover, an isolated antibody may be substantially free of other cellular material and/or 30 chemicals. A "neutralizing antibody" (or an "antibody that neutralized hIL-12 activity") includes an antibody whose binding to hIL-12 results in inhibition of the biological activity of hIL-12. This inhibition of the biological activity of hIL-12 can be assessed 34 by measuring one or more indicators of hIL-12 biological activity, such as inhibition of human phytohemagglutinin blast proliferation in a phytohemagglutinin blast proliferation assay (PHA), or inhibition of receptor binding in a human IL- 12 receptor binding assay (see Example 3-Interferon-gamma Induction Assay of US Patent No. 5 6,914,128). These indicators of hIL-12 biological activity can be assessed by one or more of several standard in vitro or in vivo assays known in the art (see Example 3 of US Patent No. 6,914,128). The term "activity" includes activities such as the binding specificity/affinity of an antibody for an antigen, for example, an anti-hIL-12 antibody that binds to an IL-12 10 antigen and/or the neutralizing potency of an antibody, for example, an anti-hIL-12 antibody whose binding to hIL-12 inhibits the biological activity of hIL-12, e.g. inhibition of PHA blast proliferation or inhibition of receptor binding in a human IL-12 receptor binding assay (see Example 3 of US Patent No. 6,914,128). The phrase "surface plasmon resonance" includes an optical phenomenon that 15 allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BlAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ). For further descriptions, see Example 5 of US Patent No. 6,914,128 and J6nsson, U., et al. (1993) Ann. Biol. Clin. 51:19-26; Jonsson, U., et al. (1991) Biotechniques 11:620-627; 20 Johnsson, B., et al. (1995) J. Mol. Recognit. 8:125-131; and Johnson, B., et al. (1991) Anal. Biochem. 198:268-277. The term "Koff", as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex. The term "Kd", as used herein, is intended to refer to the dissociation constant of 25 a particular antibody-antigen interaction. The phrase "nucleic acid molecule" includes DNA molecules and RNA molecules. A nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA. The phrase "isolated nucleic acid molecule", as used herein in reference to 30 nucleic acids encoding antibodies or antibody portions (e.g., VH, VL, CDR3) that bind hIL-12 including "isolated antibodies"), includes.a nucleic acid molecule in which the nucleotide sequences encoding the antibody or antibody portion are free of other nucleotide sequences encoding antibodies or antibody portions that bind antigens other 35 than hIL-12, which other sequences may naturally flank the nucleic acid in human genomic DNA. Thus, for example, an isolated nucleic acid of the invention encoding a VH region of an anti-IL- 12 antibody contains no other sequences encoding other VH regions that bind antigens other than IL-12. The phrase "isolated nucleic acid molecule" 5 is also intended to include sequences encoding bivalent, bispecific antibodies, such as diabodies in which VH and VL regions contain no other sequences other than the sequences of the diabody. The term "vector" includes a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", 10 which refers to a circular double stranded DNA loop into which additional DNA segments may be ligated. Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other 15 vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, "expression vectors"). In general, 20 expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" may be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated 25 viruses), which serve equivalent functions. The phrase "recombinant host cell" (or simply "host cell") includes a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding 30 generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term "host cell" as used herein.
36 The term "modifying", as used herein, is intended to refer to changing one or more amino acids in the antibodies or antigen-binding portions thereof. The change can be produced by adding, substituting or deleting an amino acid at one or more positions. The change can be produced using known techniques, such as PCR mutagenesis. 5 The phrase "contact position" includes an amino acid position of in the CDRI, CDR2 or CDR3 of the heavy chain variable region or the light chain variable region of an antibody which is occupied by an amino acid that contacts antigen in one of the twenty-six known antibody-antigen structures. If a CDR amino acid in any of the 26 known solved structures of antibody-antigen complexes contacts the antigen, then that 10 amino acid can be considered to occupy a contact position. Contact positions have a higher probability of being occupied by an amino acid which contact antigen than non contact positions. Preferably a contact position is a CDR position which contains an amino acid that contacts antigen in greater than 3 of the 26 structures (>] 1.5 %). Most preferably a contact position is a CDR position which contains an amino acid that 15 contacts antigen in greater than 8 of the 25 structures (>32%). The term "hypermutation position" includes an amino acid residue that occupies position in the CDR1, CDR2 or CDR3 region of the heavy chain variable region or the light chain variable region of an antibody that is considered to have a high frequency or probability for somatic hypermutation during in vivo affinity maturation of the antibody. 20 "High frequency or probability for somatic hypermutation" includes frequencies or probabilities of a 5 to about 40% chance that the residue will undergo somatic hypermutation during in vivo affinity maturation of the antibody. It should be understood that all ranges within this stated range are also intended to be part of this invention, e.g., 5 to about 30%, e.g., 5 to about 15%, e.g., 15 to about 30%. 25 The term "preferred selective mutagenesis position" includes an amino acid residue that occupies a position in the CDR 1, CDR2 or CDR3 region of the heavy chain variable region or the light chain variable region which can be considered to be both a contact and a hypermutation position. The phrase "selective mutagenesis approach" includes a method of improving the 30 activity of an antibody by selecting and individually mutating CDR amino acids at least one preferred selective mutagenesis position, hypermutation, and/or contact position. A "selectively mutated" human antibody is an antibody which contains a mutation at a position selected using a selective mutagenesis approach. In another embodiment, the 37 selective mutagenesis approach is intended to provide a method of preferentially mutating selected individual amino acid residues in the CDR 1, CDR2 or CDR3 of the heavy chain variable region (hereinafter H I, H2, and H3, respectively), or the CDR 1, CDR2 or CDR3 of the light chain variable region (hereinafter referred to as LI, L2, and 5 L3, respectively) of an antibody. Amino acid residues may be selected from preferred selective mutagenesis positions, contact positions., or hypermutation positions. Individual amino acids are selected based on their position in the light or heavy chain variable region. It should be understood that a hypermutation position can also be a contact position. In an embodiment, the selective mutagenesis approach is a "targeted 10 approach". The language "targeted approach" is intended to include a method of preferentially mutating selected individual amino acid residues in the CDR I, CDR2 or CDR3 of the heavy chain variable region or the CDR 1, CDR2 or CDR3 of the light chain variable region of an antibody in a targeted manner, e.g., a "Group-wise targeted approach" or "CDR-wise targeted approach". In the "Group-wise targeted approach", 15 individual amino acid residues in particular groups are targeted for selective mutations including groups I (including L3 and H3), II (including H2 and LI) and III (including L2 and HI), the groups being listed in order of preference for targeting. In the "CDR wise targeted approach", individual amino acid residues in particular CDRs are targeted for selective mutations with the order of preference for targeting as follows: H3, L3, 20 H2, LI, HI and L2. The selected amino acid residue is mutated, e.g., to at least two other amino acid residues, and the effect of the mutation on the activity of the antibody is determined. Activity is measured as a change in the binding specificity/affinity of the antibody, and/or neutralization potency of the antibody. It should be understood that the selective mutagenesis approach can be used for the optimization of any antibody 25 derived from any source including phage display, transgenic animals with human IgG germline genes, human antibodies isolated from human B-cells. Preferably, the selective mutagenesis approach is used on antibodies which can not be optimized further using phage display technology. It should be understood that antibodies from any source including phage display, transgenic animals with human IgG germline genes, human 30 antibodies isolated from human B-cells can be subject to backmutation prior to or after the selective mutagenesis approach. The term "activity enhancing amino acid residue" includes an amino acid residue which improves the activity of the antibody. It should be understood that the activity 38 enhancing amino acid residue may replace an amino acid residue at a preferred selective mutagenesis position, contact position, or a hypermutation position and, further, more than one activity enhancing amino acid residue can be present within one or more CDRs. An activity enhancing amino acid residue include, an amino acid residue that improves 5 the binding specificity/affinity of an antibody, for example anti-human IL- 12 antibody binding to human IL-12. The activity enhancing amino acid residue is also intended to include an amino acid residue that improves the neutralization potency of an antibody, for example, the human IL- 12 antibody which inhibits human IL-12. The term "Cmax" refers to the maximum or peak serum or plasma concentration 10 of an agent observed in a subject after its administration. The term "Tmax" refers to the time at which Cmax occurred. The term "bioavailability" or "F%" refers to a fraction or percent of a dose which is absorbed and enters the systemic circulation after administration of a given dosage form. The dose of the agent may be administered through any route, and, preferably, via 15 intravenous or subcutaneous injection. The term "combination" as in the phrase "a first agent in combination with a second agent" includes co-administration of a first agent and a second agent, which for example may be dissolved or intermixed in the same pharmaceutically acceptable carrier, or administration of a first agent, followed by the second agent, or administration 20 of the second agent, followed by the first agent. The present invention, therefore, includes methods of combination therapeutic treatment and combination pharmaceutical compositions. The term "concomitant" as in the phrase "concomitant therapeutic treatment" includes administering an agent in the presence of a second agent. A concomitant 25 therapeutic treatment method includes methods in which the first, second, third, or additional agents are co-administered. A concomitant therapeutic treatment method also includes methods in which the first or additional agents are administered in the presence of a second or additional agents, wherein the second or additional agents, for example, may have been previously administered. A concomitant therapeutic treatment method 30 may be executed step-wise by different actors. For example, one actor may administer to a subject a first agent and a second actor may to administer to the subject a second agent, and the administering steps may be executed at the same time, or nearly the same time, or at distant times, so long as the first agent (and additional agents) are after 39 administration in the presence of the second agent (and additional agents). The actor and the subject may be the same entity (e.g., human). The term "combination therapy", as used herein, refers to the administration of two or more therapeutic substances, e.g., an anti-IL-12, anti-IL-23 antibody and another 5 drug. The other drug(s) may be administered concomitant with, prior to, or following the administration of an anti-IL- 12, anti-IL-23 antibody. The term "dosing", as used herein, refers to the administration of a substance (e.g., an anti-IL-12, anti-IL-23 antibody) to achieve a therapeutic objective (e.g., treatment of psoriasis). 10 As used herein, the term "dose amount" refers to the quantity, e.g., milligrams (mg), of the substance which is administered to the subject. In one embodiment, the dose amount is a fixed dose, e.g., is not dependent on the weight of the subject to which the substance is administered. In another embodiment, the dose amount is not a fixed dose, e.g., is dependent on the weight of the subject to which the substance is 15 administered. Exemplary dose amounts, e.g., fixed dose amounts, for use in the methods of the invention include, about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, or about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, or about 300 mg. 20 In one embodiment, the dose amount is about 100 to about 300 mg. In yet another embodiment, the dose amount is about 100 to about 200 mg. Ranges intermediate to the .above-recited ranges are also contemplated by the invention. For example, ranges having any one of these values as the upper or lower limits are also intended to be part of the invention, e.g., about 110 mg to about 170 mg, about 150 mg to about 220 mg, 25 etc. As used herein, the term "periodicity" as it relates to the administration of a substance (e.g., an antibody which binds to the p40 subunit of-IL-12 and/or-IL-23) refers to a (regular) recurring cycle of administering the substance to a subject. In one embodiment, the recurring cycle of administration of the substance to the subject 30 achieves a therapeutic objective. The periodicity of administration of the substance may be about once a week, once every other week, about once every three weeks, about once every 4 weeks, about once every 5 weeks, about once every 6 weeks, about once every 7 weeks, about once every 8 weeks, about once every 9 weeks, about once every 10 40 weeks, about once every 11 weeks, about once every 12 weeks, about once every 13 weeks, about once every 14 weeks, about once every 15 weeks, about once every 16 weeks, about once every 17 weeks, about once every 18 weeks, about once every 19 weeks, about once every 20 weeks, about once every 21 weeks, about once every 22 5 weeks, about once every 23 weeks, about once every 24 weeks, about once every 5-10 days, about once every 10-20 days, about once every 10-50 days, about once every 10 100 days, about once every 10-200 days, about once every 25-35 days, about once every 20-50 days, about once every 20-100 days, about once every 20-200 days, about once every 30-50 days, about once every 30-90 days, about once every 30-100 days, about 10 once every 30-200 days, about once every 50-150 days, about once every 50-200 days, about once every 60-180 days, or about once every 80-100 days. Periodicities intermediate to the above-recited times are also contemplated by the invention. Ranges intermediate to the above-recited ranges are also contemplated by the invention. For example, ranges having any one of these values as the upper or lower limits are also 15 intended to be part of the invention, e.g., about 110 days to about 170 days, about 160 days to about 220 days, etc. As used herein, the phrase "periodicity of about once every 4 weeks" as it relates to the administration of a substance (e.g., an antibody which binds to the p40 subunit of IL-12 and/or IL-23), refers to a (regular) recurring cycle of administering the substance 20 to a subject about once every 4 weeks, about once every 28 days, or about once every month. In one embodiment, the recurring cycle of administration of the substance to the subject achieves or maintains a therapeutic objective (e.g., treating psoriasis), either alone or in conjunction with other recurring cycles (e.g., if a first periodicity, then in conjunction with a second and/or third periodicity; if a second periodicity, then in 25 conjunction with a first and/or third periodicity; and if a third periodicity, then in conjunction with a first and second periodicity) of administering the substance. Preferably, the substance is administered once every 22-34 days, every 24-32 days, even more preferably, every 26-30 days (e.g., every 26, 27, 28, 29 or 30 days), and most preferably every 28 days. 30 As used herein, the phrase "periodicity of about once every 12 weeks" as it relates to the administration of a substance (e.g., an antibody which binds to the p40 subunit of-IL-12 and/or IL-23), refers to a (regular) recurring cycle of administering the substance to a subject about once every 12 weeks, about once every 84 days, or about 41 once every 3 months. In one embodiment, the recurring cycle of administration of the substance to the subject achieves or maintains a therapeutic objective (e.g., treating psoriasis), either alone or in conjunction with other recurring cycles (e.g., if a first periodicity, then in conjunction with a second and/or third periodicity; if a second 5 periodicity, then in conjunction with a first and/or third periodicity; and if a third periodicity, then in conjunction with a first and second periodicity) of administering the substance. Preferably, the substance is administered once every 78-90 days, every 80-88 days, even more preferably, every 82-86 days (e.g., every 82, 83, 84, 85 or 86 days), and most preferably every 84 days. 10 The "duration of a periodicity" refers to a time over which the recurring cycle of administration occurs. For example, a duration of the periodicity of administration of a substance may be may about 12 weeks during which the periodicity of administration is about once every week. For example, a duration of the periodicity may be about 6 weeks during 15 which the periodicity of administration is about once every 4 weeks, e.g., the substance is administered at week zero and at week four. The duration of periodicity may be about I week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks,-about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 15 weeks, about 20 20 weeks, about 25 weeks, about 30 weeks, about 35 weeks, about 40 weeks, about 45 weeks, about 50 weeks, about 52 weeks, about 55 weeks, about 60 weeks, about 70 weeks, about 80 weeks, about 90 weeks, or about 100 weeks, or longer. In one embodiment, the duration of periodicity is for a length of time necessary or required to achieve a therapeutic objective, e.g., treatment, maintenance of treatment, etc. e.g., 25 maintain a PASI 50, PASI 75, PASI 90, PASI 100 score or PGA of 0 or 1 score. Durations of a periodicity intermediate to the above-recited times are also contemplated by the invention. The duration of periodicity may be about 4 weeks, about 8 weeks, about 12 weeks, about 16 weeks, about 20 weeks, about 24 weeks, about 28 weeks, about 32 30 weeks, about 36 weeks, about 40 weeks, about 44 weeks, about 48 weeks, about 52 weeks, or longer. The duration of periodicity may be at least about 4 weeks, at least about 8 weeks, at least about 12 weeks, at least about 16 weeks, at least about 20 weeks, at least about 24 weeks, at least about 28 weeks, at least about 32 weeks, at least about 42 36 weeks, at least about 40. weeks, at least about 44 weeks, at least about 48 weeks, or at least about 52 weeks. Furthermore, the duration of periodicity may be at least about 1 week, at least about 2 weeks, at least about 3 weeks, at least about 4 weeks, at least about 5 weeks, at 5 least about 6 weeks, at least about 7 weeks, at least about 8 weeks, at least about 9 weeks, at least about 10 weeks, at least about I1 weeks, at least about 12 weeks, at least about 15 weeks, at least about 20 weeks, at least about 25 weeks, at least about 30 weeks, at least about 35 weeks, at least about 40 weeks, at least about 45 weeks, at least about 50 weeks, at least about 55 weeks, at least about 60 weeks, at least about 70 10 weeks, at least about 80 weeks, at least about 90 weeks, or at least about 100 weeks. The term "treated," "treating" or "treatment" includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated. For example, treatment can be diminishment of one or more symptoms of a disorder or complete eradication of a disorder. "Treatment" or "treating" (e.g., 15 treating psoriasis) means achieving or maintaining a therapeutic objective. "Treatment" or "treating" can mean maintaining a response to a prior treatment (e.g., a prior response achieved following administration of a first dose amount according to a first periodicity; or achieved following administration of a first dose amount according to a first periodicity and a second dose amount according to a second periodicity; or achieved 20 following administration of a first dose amount according to a first periodicity and a first or second dose amount according to a second periodicity, and a first, second, or third dose amount according to a third periodicity. "Treatment of' or "treating" psoriasis may mean achieving or maintaining a PGA score of 0/1 or a PASI 50, PASI 75, PASI 90, or PASI 100 response score for a period of time during or following treatment (e.g., for at 25 least 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 46, 48, 50, 52, 54, 56, 58 or 60 weeks or longer). "Treatment of' or "treating" psoriasis may also mean achieving or maintaining a health-related quality of life (HRQOL) outcome. HRQOL outcomes include Dermatology Life Quality Index (DLQI), visual analog scales for Ps-related (VAS-Ps) and psoriatic arthritis-related (VAS-PsA) pain, Short 30 Form 36 Health Survey Mental (MCS) and Physical (PCS) Component Summary scores, and Total Activity Impairment (TAI) scores.. "Treatment of' or "treating" psoriasis may also mean achieving or maintaining a minimum clinically important difference (MCID) for any of the HRQOL outcomes provided herein, e.g., any one or combination 43 of DLQI, VAS-Ps, VAS-PsA, MCS, PCS and TAI.. "Treatment of' or "treating" psoriasis may also mean achieving or maintaining a minimum clinically important difference (MCID) response rate for any of the HRQOL outcomes provided herein, e.g., any one or combination of DLQI, VAS-Ps, VAS-PsA, MCS, PCS and TAI. "Treatment 5 'of' or "treating" psoriasis may also mean achieving or maintaining a clinically meaningful reduction in any of the HRQOL outcomes provided herein, e.g., any one or combination of DLQI, VAS-Ps, VAS-PsA, MCS, PCS and TAI. "Treatment of' or "treating" psoriasis may also mean achieving or maintaining a Nail Psoriasis Severity Index (NAPSI) score for a period of time during or following treatment (e.g., for at least 10 2,4,6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40,42,46,48, 50, 52, 54, 56, 58 or 60 weeks or longer). "Treatment of' or "treating" psoriasis may also mean achieving or maintaining any of the outcomes provided herein in a certain percentage of a population of subjects (e.g., in at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of a 15 population of subjects). The term "kit" as used herein refers to a packaged product comprising components with which to administer the anti-IL- 12, anti-IL-23 antibody of the invention for treatment of a IL- 12 related disorder. The kit preferably comprises a box or container that holds the components of the kit. The box or container is affixed with a 20 label or a Food and Drug Administration approved protocol. The box or container holds components of the invention which are preferably contained within plastic, polyethylene, polypropylene, ethylene, or propylene vessels. The vessels can be capped tubes or bottles. The kit can also include instructions for administering an anti-IL-12, anti-IL-23 antibody. 25 Various aspects of the invention are described in further detail in the following subsections. I. Human Antibodies that Bind to the p40 subunit of Human IL-12/Human IL-23 This invention provides methods and compositions for using human antibodies, 30 or antigen-binding portions thereof, that bind to human IL- 12 for the treatment of psoriasis. The invention also includes methods and compositions for using an antibody which binds both IL-12 and IL-23. Preferably, the human antibodies used in the invention are recombinant, neutralizing human anti-hIL-12/IL-23 antibodies.
44 In one embodiment, the antibody used in the invention is the antibody ABT-874 (see US Patent No. 6,914,128). ABT-874 is a fully human antibody against interleukin 12 (IL-12) and IL-23. It binds with great affinity to the p40 subunit common to both IL 12 and IL-23, validated targets in the treatment of psoriasis (Ps). 5 Antibodies that bind to the p40 subunit of human IL-12/IL-23 can be selected, for example, by screening one or more human VL and VH cDNA libraries with hIL-12, such as by phage display techniques as described in Example 1 of US Patent No. 6,914,128. Screening of human VL and VH cDNA libraries initially identified a series of anti-IL-12 antibodies of which one antibody, referred to herein as "Joe 9" (or "Joe 9 10 wild type"), was selected for further development. Joe 9 is a relatively low affinity human IL-12 antibody (e.g., a Koff of about 0.1 sec-1), yet is useful for specifically binding and detecting hIL- 12. The affinity of the Joe 9 antibody was improved by conducting mutagenesis of the heavy and light chain CDRs , producing a panel of light and heavy chain variable regions that were "mixed and matched" and further mutated, 15 leading to numerous additional anti-hIL-12 antibodies with increased affinity for hIL-12 (see Example 1, table 2 of US Patent No. 6,914, 128 (see table 2 of Appendix A attached hereto)) and the sequence alignments of Figures IA-D of US Patent No. 6,914,128 (see Figure 8A-D herein). Of these antibodies, the human anti-hIL-12 antibody referred to herein as Y61 20 demonstrated a significant improvement in binding affinity (e.g., a Koff of about 2 x 10 4 sec- 1 ). The Y61 anti-hIL-12 antibody was selected for further affinity maturation by individually mutating specific amino acids residues within the heavy and light chain CDRs . Amino acids residues of Y61 were selected for-site-specific mutation (selective mutagenesis approach) based on the amino acid residue occupying a preferred selective 25 mutagenesis position, contact and/or a hypermutation position. A summary of the substitutions at selected positions in the heavy and light chain CDRs is shown in Figures 2A-2H of US Patent No. 6,914,128 (Figures 9A-H herein). A preferred recombinant neutralizing antibody of the invention, referred to herein as J695 (also referred to as ABT-874 (Abbott Laboratories), resulted from a Gly to Tyr substitution at position 50 of 30 the light chain CDR2 of Y61, and a Gly to Tyr substitution at position 94 of the light chain CDR3 of Y6 1.
45 Amino acid sequence alignments of the heavy and light chain variable regions of a panel of anti-IL- 12 antibodies used in the invention, on the lineage from Joe 9 wild type to J695, are shown in Figures IA-ID of US Patent No. 6,914,128 (Figures 8A-D herein). These sequence alignments allowed for the identification of consensus 5 sequences for preferred heavy and light chain variable regions of antibodies of the invention that bind hIL-12, as well as consensus sequences for the CDR3, CDR2, and CDR1 , on the lineage from Joe 9 to J695. Moreover, the Y61 mutagenesis analysis summarized in Figures 2A-2H of US 6,914,128 (Figures 9A-H herein) allowed for the identification of consensus sequences for heavy and light chain variable regions that 10 bind hIL-12, as well as consensus sequences for the CDR3, CDR2, and CDR1 that bind hIL-12 on the lineage from Y61 to J695 that encompasses sequences with modifications from Y61 yet that retain good hIL-12 binding characteristics. Preferred CDR, VH and VL sequences of the invention (including consensus sequences) as identified by sequence identifiers in the attached Sequence Listing, are summarized below. 15 SEQ ANTIBODY REGION SEQUENCE ID CHAIN NO: 1 Consensus CDR H3 (H/S)-G-S-(H/Y)-D Joe 9 to (N/T/Y) J695 2 Consensus CDR L3 Q-(S/T)-Y-(D/E)-(S/R/K) Joe 9 to (S/G/Y)-(L/F/T/S) J695 (R/S/T/W/H)-(G/P)
(S/T/A/L)-(R/S/M/T/L)
(V/I/T/M/L) 3 Consensus CDR H2 F-I-R-Y-D-G-S-N-K-Y-Y-A Joe 9 to D-S-V-K-G -J695 4 Consensus CDR L2 (G/Y)-N-(D/S)-(Q/N)-R-P Joe 9 to S J695 5 Consensus CDR H1 F-T-F-S-(S/E)-Y-G-M-H Joe 9 to J695 46 6 Consensus CDR Li (S/T)-G-(G/S)-(R/S)-S-N Joe 9 to I-(G/V)-(S/A)-(N/G/Y) J695 (T/D)-V-(K/H) 7 Consensu's VH (full VH sequence; see Joe 9 to sequence listing) J695 8 Consensus VL (full VL sequence; see Joe 9 to sequence listing) J695 9 Consensus CDR H3 H-(G/V/C/H)-(S/T) Y61 to J695 (H/T/V/R/I)-(D/S) (N/K/A/T/S/F/W/H) 10 Consensus CDR L3 Q-S-Y-(D/S)-(Xaa) Y61 to J695. (G/D/Q/L/F/R/H/N/Y)-T-H P-A-L-L 11 Consensus CDR H2 (F/T/Y)-I-(R/A)-Y Y61 to J695 (D/S/E/A)-(G/R)-S-(Xaa) K-(Y/E)-Y-A-D-S-V-K-G 12 Consensus CDR L2 (G/Y/S/T/N/Q)-N-D-Q-R-P Y61 to J695 S 13 Consensus CDR Hi F-T-F-(Xaa)-(Xaa)-(Y/H) Y61 to J695 (G/M/A/N/S)-M-H 14 Consensus CDR Li S-G-G-R-S-N-I-G Y61 to J695 (S/C/R/N/D/T)-(N/M/I) (T/Y/D/H/K/P)-V-K 15 Consensus VH (full.VH sequence; see Y61 to J695 sequence listing) 16 Consensus VL (full VL sequence; see Y61 to J695 sequence listing) 17 Y61 CDR H3 H-G-S-H-D-N 18 Y61 CDR L3 Q-S-Y-D-R-G-T-H-P-A-L-L 19 Y61 CDR H2 F-I-R-Y-D-G-S-N-K-Y-Y-A D-S-V-K-G 20 Y61 CDR L2 G-N-D-Q-R-P-S 21 Y61 CDR Hl F-T-F-S-S-Y-G-M-H 22 Y61 CDR Li S-G-G-R-S-N-I-G-S-N-T-V
K
47 23 Y61 VH (full VH sequence; see sequence listing) 24 Y61 VL (full VL sequence; see sequence listing) 25 J695 CDR H3 H-G-S-H-D-N 26 J695 CDR L3 Q-S-Y-D-R-Y-T-H-P-A-L-L 27 J695 CDR H2 F-I-R-Y-D-G-S-N-K-Y-Y-A D-S-V-K-G 28 J695 CDR L2 Y-N-D-Q-R-P-S 29 J695 CDR Hl F-T-F-S-S-Y-G-M-H 30 J695 CDR Li S-G-S-R-S-N-I-G-S-N-T-V K 31 J695 VH (full VH sequence; see sequence listing) 32 J695 VL (full VL sequence; see sequence listing) Antibodies produced from affinity maturation of Joe 9 wild type were functionally characterized by surface plasmon resonance analysis to determine the Kd and Kff rate. A series of antibodies were produced having a Koff rate within the range 5 of about 0.1 s-I to about 1 x 10- 5 s- 1 , and more preferably a Koff of about 1 x 10- 4 s-I to I x 10-5 s-I or less. Antibodies were also characterized in vitro for their ability to inhibit phytohemagglutinin (PHA) blast proliferation, as described in Example 3 of US Patent No. 6,914,128. A series of antibodies were produced having an IC 5 0 value in the range of about IX10-6 M to about lx 10-1 M, more preferably about Ix10-10 M to Ix10 10 M or less. Accordingly, in one aspect, the invention provides methods and compositions for using an isolated human antibody, or antigen-binding portion thereof, that binds to human IL-12 and dissociates from human IL-12 with a Koff rate constant of 0.1 s-I or less, as determined by surface plasmon resonance, or which inhibits phytohemagglutinin 15 blast proliferation in an in vitro phytohemagglutinin blast proliferation assay (PHA assay) with an IC 5 0 of I x 10-6 M or less. In preferred embodiments, the isolated human IL-12 antibody, or an antigen-binding portion thereof, dissociates from human IL-12 48 with a Koff rate constant of 1 x 10-2 s-I or less, or inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-7 M or less. In more preferred embodiments, the isolated human IL-12 antibody, or an antigen-binding portion thereof, dissociates from human IL-12 with a Koff rate constant of 1 x 10- 3 s- 1 5 or less, or inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-8 M or less. In more preferred embodiments, the isolated human IL-12 antibody, or an antigen-binding portion thereof, dissociates from human IL-12 with a Koff rate constant of I x 10- 4 s- 1 or less, or inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10- 9 M or less. In more 10 preferred embodiments, the isolated human IL-12 antibody, or an antigen-binding portion thereof, dissociates from human IL-12 with a Koff rate constant of 1 x 10- 5 s-I or less, or inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-10 M or less. In even more preferred embodiments, the isolated human IL-12 antibody, or an antigen-binding portion thereof, dissociates from human 15 IL- 12 with a Koff rate constant of 1 x 10-5 s-I or less, or inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-1 1 M or less. The dissociation rate constant (Koff ) of an IL-12 antibody can be determined by surface plasmon resonance (see Example 5 of US Patent No. 6,914,128). Generally, surface plasmon resonance analysis measures real-time binding interactions between 20 ligand (recombinant human IL-12 immobilized on a biosensor matrix) and analyte (antibodies in solution) by surface plasmon resonance (SPR) using the BlAcore system (Pharmacia Biosensor, Piscataway, NJ). Surface plasmon analysis can also be performed by immobilizing the analyte (antibodies on a biosensor matrix) and presenting the ligand (recombinant IL-12 in solution). Neutralization activity of IL-12 25 antibodies, or antigen binding portions thereof, can be assessed using one or more of several suitable in vitro assays (see Example 3 of US Patent No. 6,914,128). It is well known in the art that antibody heavy and light chain CDRs play an important role in the binding specificity/affinity of an antibody for an antigen. Accordingly, the invention encompasses human antibodies having light and heavy chain 30 CDRs of Joe 9, as well as other antibodies having CDRs that have been modified to improve the binding specificity/affinity of the antibody. As demonstrated in Example 1 49 of US Patent No. 6,914,128, a series of modifications to the light and heavy chain CDRs results in affinity maturation of human anti-hIL-12 antibodies. The heavy and light chain variable region amino acid sequence alignments of a series of human antibodies ranging from Joe 9 wild type to J695 that bind human IL-12 is shown in Figures lA-ID 5 of US Patent No. 6,914,128 (Figures 8A-D herein). Consensus sequence motifs for the CDRs of antibodies can be determined from the sequence alignment. For example, a consensus motif for the VH CDR3 of the lineage from Joe 9 to J695 comprises the amino acid sequence: (H/S)-G-S-(H/Y)-D-(N/T/Y) (SEQ ID NO: 1), which encompasses amino acids from position 95 to 102 of the consensus HCVR shown in 10 SEQ ID NO: 7. A consensus motif for the VL CDR3 comprises the amino acid sequence: Q-(S/T)-Y-(D/E)-(S/R/K)-(S/G/Y)-(L/F/T/S)-(RS/T/W/H)-(G/P)-(S/T/A/L) (RIS/M/T/L-V/I/T/M/L) (SEQ ID NO: 2), which encompasses amino acids from position 89 to 97 of the consensus LCVR shown in SEQ ID NO: 8. Accordingly, in another aspect, the invention provides methods and compositions 15 comprising an isolated human antibody, or an antigen-binding portion thereof, which has the following characteristics: a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 5 0 of 1 x 10-6 M or less; b) has a heavy chain CDR3 comprising the amino acid sequence of SEQ ID 20 NO: 1; and c) has a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 2. In a preferred embodiment, the antibody further comprises a VH CDR2 comprising the amino acid sequence: F-I-R-Y-D-G-S-N-K-Y-Y-A-D-S-V-K-G (SEQ 25 ID NO: 3) (which encompasses amino acids from position 50 to 65 of the consensus HCVR comprising the amino acid sequence SEQ ID NO: 7) and further comprises a VL CDR2 comprising the amino acid sequence: (G/Y)-N-(D/S)-(Q/N)-R-P-S (SEQ ID NO: 4) (which encompasses amino acids from position 50 to 56 of the consensus LCVR comprising the amino acid sequence SEQ ID NO: 8). 30 In another preferred embodiment, the antibody further comprises a VH CDR I comprising the amino acid sequence: F-T-F-S-(S/E)-Y-G-M-H (SEQ ID NO: 5) (which encompasses amino acids from position 27 to 35 of the consensus HCVR comprising the amino acid sequence SEQ ID NO: 7) and further comprises a VL CDR I comprising the 50 amino acid sequence: (SF)-G-(G/S)-(R/S)-S-N-I-(G/V)-(S/A)-(N/G/Y)-(T/D)-V-(K/H) (SEQ ID NO: 6) (which encompasses amino acids from position 24 to 34 of the consensus LCVR comprising the amino acid sequence SEQ ID NO: 8). In yet another preferred embodiment, the antibody used in the invention 5 comprises a HCVR comprising the amino acid sequence of SEQ ID NO: 7 and a LCVR comprising the amino acid sequence of SEQ ID NO: 8. Additional consensus motifs can be determined based on the mutational analysis performed on Y61 that led to the J695 antibody (summarized in Figures 2A-2H of US Patent No. 6,914,128; Figures 9A-H herein). As demonstrated by the graphs shown in 10 Figures 2A-2H of US Patent No. 6,914,128 (Figures 9A-H herein), certain residues of the heavy and light chain CDRs of Y61 were amenable to substitution without significantly impairing the hIL- 12 binding properties of the antibody. For example, individual substitutions at position 30 in CDR HI with twelve different amino acid residues did not significantly reduce the Koff rate of the antibody, indicating that is 15 position is amenable to substitution with a variety of different amino acid residues. Thus, based on the mutational analysis (i.e., positions within Y61 that were amenable to substitution by other amino acid residues) consensus motifs were determined. The consensus motifs for the heavy and light chain CDR3s are shown in SEQ ID NOs: 9 and 10, respectively, consensus motifs for the heavy and light chain CDR2s are shown in 20 SEQ ID NOs: 11 and 12, respectively, and consensus motifs for the heavy and light chain CDRls are shown in SEQ ID NOs: 13 and 14, respectively. Consensus motifs for the VH and VL regions are shown in SEQ ID NOs: 15 and 16, respectively. Accordingly, in one aspect, the invention includes an isolated human antibody, or an antigen-binding portion thereof, which has the following characteristics: 25 a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-9 M or less; b) has a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 9; and c) has a light chain CDR3 comprising the amino acid sequence of SEQ ID 30 NO: 10. In a preferred embodiment, the antibody further comprises a VH CDR2 comprising the amino acid sequence of SEQ ID NO: 11 and further comprises a VL CDR2 comprising the amino acid sequence of SEQ ID NO: 12.
51 In another preferred embodiment, the antibody further comprises a VH CDR1 comprising the amino acid sequence of SEQ ID NO: 13 and further comprises a VL CDR1 comprising the amino acid sequence of SEQ ID NO: 14. In yet another preferred embodiment, the antibody used in the invention 5 comprises a HCVR comprising the amino acid sequence of SEQ ID NO: 15 and a LCVR comprising the amino acid sequence of SEQ ID NO: 16. A preferred antibody used in the invention, the human anti-hIL-12 antibody Y61, can be produced by affinity maturation of Joe 9 wild type by PCR mutagenesis of the CDR3 (as described in Example 1 of US Patent No. 6,914,128). Y61 had an improved 10 specificity/binding affinity determined by surface plasmon resonance and by in vitro neutralization assays. The heavy and light chain CDR3s of Y61 are shown in SEQ ID NOs: 17 and 18, respectively, the heavy and light chain CDR2s of Y61 are shown in SEQ ID NOs: 19 and 20, respectively, and the heavy and light chain CDR Is of Y61 are shown in SEQ ID NOs: 21 and 22, respectively. The VH of Y61 has the amino acid 15 sequence of SEQ ID NO: 23 and the VL of Y61 has the amino acid sequence of SEQ ID NO: 24 (these sequences are also shown in Figures IA-lD of US Patent No. 6,914,128 (Figures 8A-D herein) aligned with Joe9). Accordingly, in another aspect, the invention features use of an isolated human antibody, or an antigen-binding portion thereof, which 20 a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 5 0 of 1 x 10-9 M or less; b) has a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 17; and c) has a light chain CDR3 comprising the amino acid sequence of 25 SEQ ID NO: 18. In a preferred embodiment, the isolated human antibody, or an antigen-binding portion thereof, used in the methods and compositions of the invention has a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 19 and a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 20. 30 In another preferred embodiment, the isolated human antibody, or an antigen binding portion thereof, used in the methods and compositions of the invention, has a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 21 and a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 22.
52 In yet another preferred embodiment, the isolated human antibody, or an antigen binding portion thereof, used in the methods and compositions of the invention comprising a the heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 23, and a light chain variable region comprising the amino acid sequence 5 of SEQ ID NO: 24. In certain embodiments, the full length antibody comprises a heavy chain constant region, such as IgGI, IgG2, IgG3, IgG4, IgM, IgA and IgE constant regions, and any allotypic variant therein as described in Kabat (Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of 10 Health and Human Services, NIH Publication No. 91-3242). Preferably, the antibody heavy chain constant region is an IgGI heavy chain constant region. Alternatively, the antibody portion can be an Fab fragment, an F(ab' 2 ) fragment or a single chain Fv fragment. Modifications of individual residues of Y61 led to the production of a panel of 15 antibodies shown in Figures 2A-2H of US Patent No. 6,914,128 (Figures 9A-H herein). The specificity/binding affinity of each antibody was determined by surface plasmon resonance and/or by in vitro neutralization assays. Accordingly, in another aspect, the invention features an isolated human antibody, or an antigen-binding portion thereof, which 20 a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10-9 M or less; b) has a heavy chain CDR3 comprising the amino acid sequence selected from the group consisting of SEQ ID NO: 404-SEQ ID NO: 469; and c) has a light chain CDR3 comprising the amino acid sequence selected 25 from the group consisting of SEQ ID NO: 534-SEQ ID NO: 579. In preferred embodiment, the isolated human antibody, or an antigen-binding portion thereof, used in the methods and compositions of the invention has a heavy chain CDR2 comprising the amino acid sequence selected from the group consisting of SEQ ID NO:335-SEQ ID NO: 403; and a light chain CDR2 comprising the amino acid 30 sequence selected from the group consisting of SEQ ID NO: 506-SEQ ID NO: 533. In another preferred embodiment, the isolated human antibody, or an antigen binding portion thereof, has a heavy chain CDR1 comprising the amino acid sequence selected from the group consisting of SEQ ID NO: 288-SEQ ID NO: 334; and a light 53 chain CDR I comprising the amino acid sequence selected from the group consisting of SEQ ID NO: 470-SEQ ID NO: 505. In yet another preferred embodiment, the isolated human antibody, or an antigen binding portion thereof, comprising a the heavy chain variable region comprising the 5 amino acid sequence of SEQ ID NO: 23, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 24. In certain embodiments, the full length antibody comprising a heavy chain constant region such as IgG1, IgG2, IgG3, IgG4, IgM, IgA and IgE constant regions and any allotypic variant therein as described in Kabat (, Kabat, E.A., et al. (1991) 10 Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). Preferably, the antibody heavy chain constant region is an IgGI heavy chain constant region. Alternatively, the antibody portion can be a Fab fragment, an F(ab' 2 ) fragment or a single chain Fv fragment. 15 A particularly preferred recombinant, neutralizing antibody, J695, which may be used in the invention was produced by site-directed mutagenesis of contact and hypermutation amino acids residues of antibody Y61 (see Example 2 of US Patent No. 6,914,128 and section III below). J695 differs from Y61 by a Gly to Tyr substitution in Y61 at position 50 of the light chain CDR2 and by a Gly to Tyr substitution at position 20 94 of the light chain CDR3 . The heavy and light chain CDR3s of J695 are shown in SEQ ID NOs: 25 and 26, respectively, the heavy and light chain CDR2s of J695 are shown in SEQ ID NOs: 27 and 28, respectively, and the heavy and light chain CDR1s of J695 are shown in SEQ ID NOs: 29 and 30, respectively. The VH of J695 has the amino acid sequence of SEQ ID NO: 31 and the VL of J695 has the amino acid 25 sequence of SEQ ID NO: 32 (these sequences are also shown in Figures l A-I D of US Patent No. 6,914,128 (Figures 8A-D herein), aligned with Joe9). Accordingly, in another aspect, the invention features an isolated human antibody, or an antigen-binding portion thereof, which a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10- 9 M or less; b) has a 30 heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 25; and c) has a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 26. In preferred embodiment, the isolated human antibody, or an antigen-binding portion thereof, used in the invention has a heavy chain CDR2 comprising the amino 54 acid sequence of SEQ ID NO: 27, and a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 28. In another preferred embodiment, the isolated human antibody, or an antigen binding portion thereof, used in the invention has a heavy chain CDRl comprising the 5 amino acid sequence of SEQ ID NO: 29, and a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 30. In yet another preferred embodiment, the isolated human antibody, or an antigen binding portion thereof, used in the invention has a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 31, and a light chain variable 10 region comprising the amino acid sequence of SEQ ID'NO: 32. In certain embodiments, the full length antibody comprises a heavy chain constant region, such as IgG1, IgG2, IgG3, IgG4, IgM, IgA and IgE constant regions and any allotypic variant therein as described in Kabat (, Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of 15 Health and Human-Services, NIH Publication No. 91-3242). Preferably, the antibody heavy chain constant region is an IgGI heavy chain constant region. Alternatively, the antibody portion can be an Fab fragment, an F(ab' 2 ) fragment or a single chain Fv fragment. Additional mutations in the preferred consensus sequences for CDR3, CDR2, 20 and CDRl of antibodies on the lineage from Joe 9 to J695, or from the lineage Y61 to J695, can be made to provide additional anti-IL-12 antibodies of the invention. Such methods of modification can be performed using standard molecular biology techniques, such as by PCR mutagenesis, targeting individual contact or hypermutation amino acid residues in the light chain and/or heavy chain CDRs-, followed by kinetic and functional 25 analysis of the modified antibodies as described herein (e.g., neutralization assays described in Example 3 of US Patent No. 6,914,128, and by BIAcore analysis, as described in Example 5 of US Patent No. 6,914,128). Accordingly, in another aspect the invention features use of an isolated human antibody, or an antigen-binding portion thereof, which 30 a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of I x 10-6 M or less; b) comprises a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 1, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID 55 NO: 3 and a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 5, or a mutant thereof having one or more amino acid substitutions at a preferred selective mutagenesis position or a hypermutation position, wherein said mutant has a koff rate no more than 10-fold higher than the antibody comprising a heavy chain CDR3 comprising 5 the amino acid sequence of SEQ ID NO: 1, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 3, and a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 5; and c) comprises a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 2, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 10 4, and a light chain CDRl comprising the amino acid sequence of SEQ ID NO: 6, or a mutant thereof having one or more amino acid substitutions at a preferred selective mutagenesis position or a hypermutation position, wherein said mutant has a koff rate no more than 10-fold higher than the antibody comprising a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 2, a light chain CDR2 comprising the amino 15 acid sequence of SEQ ID NO: 4, and a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 6. . In another aspect the invention features use of an isolated human antibody, or an antigen-binding portion thereof, which a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay 20 with an IC 5 0 of 1 x 10- 9 M or less; b) comprises a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 9, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 11 and a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 13, or a mutant thereof having one or more amino acid substitutions at a preferred 25 selective mutagenesis position, contact position or a hypermutation position, wherein said mutant has a koff rate no more than 10-fold higher than the antibody comprising a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 9, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 11, and a heavy chain CDRI comprising the amino acid sequence of SEQ ID NO: 13; and 30 c) comprises a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 10, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 12, and a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 14, or a mutant thereof having one or more amino acid substitutions at a preferred 56 selective mutagenesis position, contact position or a hypermutation position, wherein said mutant has a kff rate no more than 10-fold higher than the antibody comprising a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 10, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 12, and a light chain CDR1 5 comprising the amino acid sequence of SEQ ID NO: 14. An ordinarily skilled artisan will also appreciate that additional mutations to the CDR regions of an antibody, for example in Y61 or in J695, can be made to provide additional anti-IL-12 antibodies of the invention. Such methods of modification can be performed using standard molecular biology techniques, as described above. The 10 functional and kinetic analysis of the modified antibodies can be performed as described in Example 3 of US Patent No. 6,914,128 and Example 5 of US Patent No. 6,914,128, respectively. Modifications of individual residues of Y61 that led to the identification of J695 are shown in Figures 2A-2H of US Patent No. 6,914,128 (Figures 9A-H herein) and are described in Example 2 of US Patent No. 6,914,128. 15 Accordingly, in another aspect the invention features use of an isolated human antibody, or an antigen-binding portion thereof, which a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10- 9 M or less; b) comprises a heavy chain CDR3 comprising the amino acid sequence of 20 SEQ ID NO: 17, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 19 and a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 21, or a mutant thereof having one or more amino acid substitutions at a preferred selective mutagenesis position or a hypermutation position, wherein said mutant has a koff rate no more than 10-fold higher than the antibody comprising a heavy chain CDR3 25 comprising the amino acid sequence of SEQ ID NO: 17, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 19, and a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 21; and c) comprises a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 18, a light chain CDR2 comprising the amino acid sequence of SEQ ID 30 NO: 20, and a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 22, or a mutant thereof having one or more amino acid substitutions at a preferred selective mutagenesis position or a hypermutation position, wherein said mutant has a koff rate no more than 10-fold higher than the antibody comprising a light chain CDR3 57 comprising the amino acid sequence of SEQ ID NO: 18, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 20, and a light chain CDR I comprising the amino acid sequence of SEQ ID NO: 22. In another aspect the invention features use of an isolated human antibody, or an 5 antigen-binding portion thereof, which a) inhibits phytohemagglutinin blast proliferation in an in vitro PHA assay with an IC 50 of 1 x 10- 9 M or less; b) comprises a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 25, a heavy chain CDR2 comprising the amino acid sequence of SEQ ID 10 NO: 27 and a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 29, or a mutant thereof having one or more amino acid substitutions at a preferred selective mutagenesis position or a hypermutation position, wherein said mutant has a koff rate no more than 10-fold higher than the antibody comprising a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 25, a heavy chain CDR2 15 comprising the amino acid sequence of SEQ ID NO: 27, and a heavy chain CDR I comprising the amino acid sequence of SEQ ID NO: 29; and c) comprises a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 26, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 28, and a light chain CDR I comprising the amino acid sequence of SEQ ID NO: 20 30, or a mutant thereof having one or more amino acid substitutions at a preferred selective mutagenesis position or a hypermutation position, wherein said mutant has a koff rate no more than 10-fold higher than the antibody comprising a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 26, a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 28, and a light chain CDR I comprising the 25 amino acid sequence of SEQ ID NO: 30. In yet another embodiment, the invention provides use of an isolated human antibodies, or antigen-binding portions thereof, that neutralize the activity of human IL 12, and at least one additional primate IL-12 selected from the group consisting of baboon IL- 12, marmoset IL- 12, chimpanzee IL- 12, cynomolgus IL- 12 and rhesus IL- 12, 30 but which do not neutralize the activity of the mouse IL- 12.
58 11 Selection of Recombinant Human Antibodies Recombinant human antibodies which may be used in the invention can be isolated by screening of a recombinant combinatorial antibody library, preferably a scFv phage display library, prepared using human VL and VH cDNAs prepared from mRNA 5 derived from human lymphocytes. Methods for identifying antibodies which may be used in the methods and compositions of the invention are described in US Patent No. 6,914,128, incorporated by reference herein.. Methodologies for preparing and screening such libraries are known in the art. In addition to commercially available kits for generating phage display libraries (e.g., the Pharmacia Recombinant Phage Antibody 10 System, catalog no. 27-9400-01; and the Stratagene SurJZAPTM.phage display kit, catalog no. 240612), examples of methods and reagents particularly amenable for use in generating and screening antibody display libraries can be found in, for example, Kang et al. PCT Publication No. WO 92/18619; Winter et al. PCT Publication No. WO 92/2079 1; Breitling et al. PCT Publication No. WO 93/01288; McCafferty et al. PCT 15 Publication No. WO 92/01047; Garrard et al. PCT Publication No. WO 92/09690; Fuchs et al. (1991) BiolTechnology 9:1370-1372; Hay et al. (1992) Hum Antibod Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; McCafferty et al., Nature (1990) 348:552-554; Griffiths et al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) J Mol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) PNAS 20 89:3576-3580; Garrad et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS 88:7978-7982. The antibody libraries used in this method are preferably scFv libraries prepared from human VL and VH cDNAs. The scFv antibody libraries are preferably screened using recombinant human IL-12 as the antigen to select human heavy and light chain 25 sequences having a binding activity toward IL-12. To select for antibodies specific for the p35 subunit of IL-12 or the p70 heterodimer, screening assays were performed in the presence of excess free p40 subunit. Subunit preferences can be determined, for example by, micro-Friguet titration, as described in Example 1 of US Patent No. 6,914,128. 30 Once initial human VL and VH segments are selected, "mix and match" experiments, in which different pairs of the selected VL and VH segments are screened for IL-12 binding, are performed to select preferred VJVH pair combinations (see Example 1 of US Patent No. 6,914,128). Additionally, to further improve the affinity 59 and/or lower the off rate constant for hIL- 12 binding, the VL and VH segments of the preferred VIJVH pair(s) can be randomly mutated, preferably within the CDR3 region of VH and/or VL, in a process analogous to the in vivo somatic mutation process responsible for affinity maturation of antibodies during a natural immune response. This 5 in vitro affinity maturation can be accomplished by amplifying VH and VL regions using PCR primers complimentary to the VH CDR3 or VL CDR3, respectively, which primers have been "spiked" with a random mixture of the four nucleotide bases at certain positions such that the resultant PCR products encode VH and VL segments into which random mutations have been introduced into the VH and/or VL CDR3 regions. These 10 randomly mutated VH and VL segments can be reselected and rescreened for binding to hIL-12 and sequences that exhibit high affinity and a low off rate for IL-12 binding can be selected. Table 2 of Appendix A of US Patent No. 6,914,128 (see table 2 of Appendix A attached hereto) shows antibodies that displayed altered binding specificity/affinity produced as a result of in vitro affinity maturation. 15 Following selection, isolation and screening of an anti-hIL-12 antibody of the invention from a recombinant immunoglobulin display library, nucleic acid encoding the selected antibody can be recovered from the phage particle(s) (e.g., from the phage genome) and subcloned into other expression vectors by standard recombinant DNA techniques. If desired, the nucleic acid can be further manipulated to create other 20 antibody forms of the invention (e.g., linked to nucleic acid encoding additional immunoglobulin domains, such as additional constant regions). To express a recombinant human antibody isolated by screening of a combinatorial library, the DNA encoding the antibody is cloned into a recombinant expression vector and introduced into a mammalian host cells, as described in further detail in Section IV below. 25 Methods for selecting human IL-12 binding antibodies by phage display technology, and affinity maturation of selected antibodies by random or site-directed mutagenesis of CDR regions are described in further detail in Example 1 of US Patent No. 6,914,128. As described in Example 1 of US Patent No. 6,914,128, screening of human VL 30 and VH cDNA libraries identified a series of anti-IL-12 antibodies, of which the Joe 9 antibody was selected for further development. A comparison of the heavy chain variable region of Joe 9 with the heavy chain germline sequences selected from the 60 VBASE database, revealed that Joe 9 was similar to the COS-3 germline sequence. COS-3 belongs to the VH 3 family of germline sequences. The VH 3 family is part of the human VH germline repertoire which is grouped into seven families, VH 1-VH 7 , based on nucleotide sequence homology (Tomlinson et 5 al. (1992) J. Mol. Biol., 227, 776-798 and Cook et al. (1995) Immunology Today, 16, 237-242). The VH 3 family contains the highest number of members and makes the largest contribution to the germline repertoire. For any given human VH 3 - germline antibody sequence, the amino acid sequence identity within the entire VH 3 family is high (See e.g., Tomlinson et al. (1992) J. Mol. Biol., 227, 776-798 and Cook et al. 10 (1995) Immunology Today, 16, 237-242). The range of amino acid sequence identity between any two germline VH sequences of the VH 3 family varies from 69-98 residues out of approximately 100 VH residues, (i.e., 69-98% amino acid sequence homology between any two germline VH sequences). For most pairs of germline sequences there is at least 80 or more identical amino acid residues, (i.e., at least 80% amino acid 15 sequence homology). The high degree of amino acid sequence homology between the
VH
3 family members results in certain amino acid residues being present at key sites in the CDR and framework regions of the VH chain. These amino acid residues confer structural features upon the CDRs. Studies of antibody structures have shown that CDR conformations can be 20 grouped into families of canonical CDR structures based on the key amino acid residues that occupy certain positions in the CDR and framework regions. Consequently, there are similar local CDR conformations in different antibodies that have canonical structures with identical key amino acid residues (Chothia et al. (1987) J. Mol. Biol., 196, 901-917 and Chothia et al. (1989) Nature, 342, 877-883). Within the VH 3 family 25 there-is a conservation of amino acid residue identity at the key sites for the CDR1 and CDR2 canonical structures (Chothia et al. (1992) J. Mol. Biol., 227, 799-817). The COS-3 germline VH gene, is a member of the VH 3 family and is a variant of the 3-30 (DP-49) germline VH allele. COS-3, differs from Joe9 VH amino acid sequences at only 5 positions. The high degree of amino acid sequence homology 30 between Joe9 VH and COS-3, and between Joe9 VH and the other VH 3 family members also confers a high degree of CDR structural homology (Chothia et al. (1992) J. Mol.
61 Biol., 227, 799-817; Chothia et al. (1987) J. Mol. Biol., 196, 901-917 and Chothia et al. (1989) Nature, 342, 877-883). The skilled artisan will appreciate that based on the high amino acid sequence and canonical structural similarity to Joe 9, other VH 3 family members could also be 5 used to generate antibodies that bind to human IL-12. This can be performed, for example, by selecting an appropriate VL by chain-shuffling techniques (Winter et al. (1994) Annual Rev. Immunol., 12, 433-55), or by the grafting of CDRs from a rodent or other human antibody including CDRs from antibodies of this invention onto a VH 3 family framework. 10 The human V lambda germline repertoire is grouped into 10 families based on nucleotide sequence homology (Williams et al. (1996) J. Mol. Biol., 264, 220-232). A comparison of the light chain variable region of Joe 9 with the light chain germline sequences selected from the VBASE database, revealed that Joe 9 was similar to the DPL8 lambda germline. The Joe9 VL differs from DPL8 sequence at only four 15 framework positions, and is highly homologous to the framework sequences of the other VIl family members. Based on the high amino acid sequence homology and canonical structural similarity to Joe 9, other V;1 family members may also be used to generate antibodies that bind to human IL-12. This can be performed, for example, by selecting an appropriate VH by chain-shuffling techniques (Winter et al. Supra, or by the grafting 20 of CDRs from a rodent or other human antibody including CDRs from antibodies of this invention onto a VIl family framework. The methods of the invention are intended to include recombinant antibodies that bind to hIL-12, comprising a heavy chain variable region derived from a member of the
VH
3 family of germline sequences, and a light chain variable region derived from a 25 member of the VXI family of germline sequences. Moreover, the skilled artisan will appreciate that any member of the VH 3 family heavy chain sequence can be combined with any member of the VXI family light chain sequence. Those skilled in the art will also appreciate that DNA sequence polymorphisms that lead to changes in the amino acid sequences of the germline may exist within a 30 population (e.g., the human population). Such genetic polymorphism in the germline sequences may exist among individuals within a population due to natural allelic variation. Such natural allelic variations can typically result in 1-5 % variance in the 62 nucleotide sequence of the a gene. Any and all such nucleotide variations and resulting amino acid polymorphisms in germline sequences that are the result of natural allelic variation are intended to be within the scope of the invention. Accordingly, in one aspect, the invention features an isolated human antibody, or 5 an antigen-binding portion thereof, which has the following characteristics: a) that binds to human IL-12 and dissociates from human IL-12 with a kof rate constant of 0.1 s- or less, as determined by surface plasmon resonance, or which inhibits phytohemagglutinin blast proliferation in an in vitro phytohemagglutinin blast proliferation assay (PHA assay) with an IC 50 of I x 10- 6 M or less. 10 b) has a heavy chain variable region comprising an amino acid sequence selected from a member of the VH 3 germline family, wherein the heavy chain variable region has a mutation at a contact or hypermutation position with an activity enhancing amino acid residue. c) has a light chain variable region comprising an amino acid sequence .15 selected from a member of the VX1 germline family, wherein the light chain variable region has a mutation at a preferred selective mutagenesis position, contact or hypermutation position with an activity enhancing amino acid residue. In a preferred embodiment, the isolated human antibody, or antigen binding has mutation in the heavy chain CDR3. In another preferred embodiment, the isolated 20 human antibody, or antigen binding has mutation in the light chain CDR3. In another preferred embodiment, the isolated human antibody, or antigen binding has mutation in the heavy chain CDR2. In another preferred embodiment, the isolated human antibody, or antigen binding has mutation in the light chain CDR2. In another preferred embodiment, the isolated human antibody, or antigen binding has mutation in the heavy 25 chain CDR 1. In another preferred embodiment, the isolated human antibody, or antigen binding has mutation in the light chain CDRl. An ordinarily skilled artisan will appreciate that based on the high amino acid sequence similarity between members of the VH 3 germline family, or between members of the light chain Val germline family, that mutations to the germlines sequences can 30 provide additional antibodies that bind to human IL-1 2. Table I of US Patent No. 6,914,128 (see Table 1 of Appendix A attached hereto) shows the germline sequences of the VH 3 family members and demonstrates the significant sequence homology within the family members. Also shown in table 1 of US Patent No. 6,914,128 (see table 1 of 63 Appendix A, attached hereto) are the germline sequences for VXl family members. The heavy and light chain sequences of Joe 9 are provided as a comparison. Mutations to the germline sequences of VH 3 or VXl family members may be made, for example, at the same amino acid positions as those made in the antibodies of the invention (e.g. 5 mutations in Joe 9). The modifications can be performed using standard molecular biology techniques, such as by PCR mutagenesis, targeting individual amino acid residues in the germline sequences, followed by kinetic and functional analysis of the modified antibodies as described herein (e.g., neutralization assays described in Example 3 of US Patent No. 6,914,128, and by BLAcore analysis, as described in Example 5 of 10 US Patent No. 6,914,128). Accordingly, in one aspect, the invention features use of an isolated human antibody, or an antigen-binding portion thereof, which has the following characteristics: a) has a heavy chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 595-667, wherein the heavy chain 15 variable region has a mutation at a preferred selective mutagenesis position, contact or hypermutation position with an activity enhancing amino acid residue. b) has a light chain variable region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 669-675, wherein the light chain variable region has a mutation at a preferred selective mutagenesis position, contact or 20 hypermutation position with an activity enhancing amino acid residue. An ordinarily skilled artisan will appreciate that based on the high amino acid sequence similarity between Joe 9 and COS-3 heavy chain germline sequence, and between Joe 9 and DPL8 lambda germline sequence, that other mutations to the CDR regions of these germlines sequences can provide additional antibodies that bind to 25 human IL-12. Such methods of modification can be performed using standard molecular biology techniques as described above. Accordingly, in one aspect, the invention features use of an isolated human antibody, or an antigen-binding portion thereof, which has the following characteristics: a) that binds to human IL-12 and dissociates from human IL-12 with a 30 koff rate constant of 0. 1 s- or less, as determined by surface plasmon resonance, or which inhibits phytohemagglutinin blast proliferation in an in vitro phytohemagglutinin blast proliferation assay (PHA assay) with an IC 50 of I x-10- 6 M or less.
64 b) has a heavy chain variable region comprising the COS-3 germline amino acid sequence, wherein the heavy chain variable region has a mutation at a preferred selective mutagenesis position, contact or hypermutation position with an activity enhancing amino acid residue. 5 c) has a light chain variable region comprising the DPL8 germline amino acid sequence, wherein the light chain variable region has a mutation at a preferred selective mutagenesis position, contact or hypermutation position with an activity enhancing amino acid residue. Due to certain amino acid residues occupying key sites in the CDR and 10 framework regions in the light and heavy chain variable region, structural features are conferred at these regions. In particular, the CDR2 and CDRI regions are subject to canonical structural classifications. Since there is a high degree of amino acids sequence homology between family members, these canonical features are present between family members. The skilled artisan will appreciate that modifications at the amino acid 15 residues that confer these canonical structures would produce additional antibodies that bind to IL-12. The modifications can be performed using standard molecular biology techniques as described above. Accordingly, in another aspect, the invention features use of an isolated human antibody, or an antigen-binding portion thereof, which has the following characteristics: 20 a) that binds to human IL-12 and dissociates from human IL-12 with a kort rate constant of 0.1 s1 or less, as determined by surface plasmon resonance, or which inhibits phytohemagglutinin blast proliferation in an in vitro phytohemagglutinin blast proliferation assay (PHA assay) with an IC 50 of 1 x 10- 6 M or less. b) has a heavy chain variable region comprising an amino acid sequence 25 selected from a member of the VH 3 germline family, wherein the heavy chain variable region comprises a CDR2 that is structurally similar to CDR2s from other VH 3 germline family members, and a CDR1 that is structurally similar to CDRIs from other VH 3 germline family members, and wherein the heavy chain variable region has a mutation at a preferred selective mutagenesis position, contact or hypermutation position with an 30 activity enhancing amino acid residue; c) has a light chain variable region comprising an amino acid sequence selected from a member of the VXl germline family, wherein the light chain variable region comprises a CDR2 that is structurally similar to CDR2s from other VX1 germline 65 family members, and a CDRI that is structurally similar to CDR1s from other VXl germline family members, and wherein the light chain variable region has a mutation at a preferred selective mutagenesis position, contact or hypermutation position with an activity enhancing amino acid residue. 5 Recombinant human antibodies used in the invention have variable and constant regions which are homologous to human germline immunoglobulin sequences selected from the VBASE database. Mutations to the recombinant human antibodies (e.g., by random mutagenesis or PCR mutagenesis) result in amino acids that are not encoded by human germline immunoglobulin sequences. Also, libraries of recombinant antibodies 10 which were derived from human donors will contain antibody sequences that differ from their corresponding germline sequences due to the normal process of somatic mutation that occurs during B-cell development. It should be noted that if the "germline" sequences obtained by PCR amplification encode amino acid differences in the framework regions from the true germline configuration (i.e., differences in the 15 amplified sequence as compared to the true germline sequence), it may be desirable to change these amino acid differences back to the true germline sequences (i.e., "backmutation" of framework residues to the germline configuration). Thus, the present invention can optionally include a backmutation step. To do this, the amino acid sequences of heavy and light chain encoded by the germline (as found as example in 20 VBASE database) are first compared to the mutated immunoglobulin heavy and light chain framework amino acid sequences to identify amino acid residues in the mutated immunoglobulin framework sequence that differ from the closest germline sequences. Then, the appropriate nucleotides of the mutated immunoglobulin sequence are mutated back to correspond to the germline sequence, using the genetic code to determine which 25 nucleotide changes should be made. Mutagenesis of the mutated immunoglobulin framework sequence is carried out by standard methods, such as PCR-mediated mutagenesis (in which the mutated nucleotides are incorporated into the PCR primers such that the PCR product contains the mutations) or site-directed mutagenesis. The role of each amino acid identified as candidate for backmutation should be investigated for a 30 direct or indirect role in antigen binding and any amino acid found after mutation to affect any desirable characteristic of the human antibody should not be included in the final human antibody; as an example, activity enhancing amino acids identified by the selective mutagenesis approach will not be subject to backmutation. Assays to determine 66 the characteristics of the antibody resulting from mutagenesis can include ELISA, competitive ELISA, in vitro and in vivo neutralization assays and/or (see e.g. Example 3 of US Patent No. 6,914,128) immunohistochemistry with tissue sections from various sources (including human, primate and/or other species). 5 To minimize the number of amino acids subject to backmutation those amino acid positions found to be different from the closest germline sequence but identical to the corresponding amino acid in a second germline sequence can remain, provided that the second germline sequence is identical-and colinear to the sequence of the human antibody of the invention for at least 10, preferably 12 amino acids, on both sides of the 10 amino acid in question. This would assure that any peptide epitope presented to the immune system by professional antigen presenting cells in a subject treated with the human antibody of the invention would not be foreign but identical to a self-antigen, i.e. the immunoglobulin encoded by that second germline sequence. Backmutation may occur at any stage of antibody optimization; preferably, backmutation occurs directly 15 before or after the selective mutagenesis approach. More preferably, backmutation occurs directly before the selective mutagenesis approach. III. Modifications to Preferred Selective Mutagenesis Positions, Contact and/or Hypermutation Positions 20 Typically, selection of antibodies with improved affinities can be carried out using phage display methods, as described in section II above and in US Patent No. 6,914,128, incorporated by reference herein. This can be accomplished by randomly mutating combinations of CDR residues and generating large libraries containing antibodies of different sequences. However, for these selection methods to work, the 25 antibody-antigen reaction must tend to equilibrium to allow, over time, preferential binding of higher affinity antibodies to the antigen. Selection conditions that would allow equilibrium to be established could not be determined (presumably due to additional non-specific interactions between the antigen and phage particle) when phage display methods were used to improve the affinity of selected anti-IL- 12 antibodies, 30 upon attaining a certain level of affinity achieved (i.e., that of antibody Y61). Accordingly, antibodies with even higher affinities could not be selected by phage display methods. Thus, for at least certain antibodies or antigens, phage display methods are limiting in their ability to select antibodies with a highly improved binding 67 specificity/affinity. Accordingly, a method termed Selective Mutagenesis Approach which does not require phage display affinity maturation of antibodies, was established to overcome this limitation and is provided by the invention. Although this Selective Mutagenesis Approach was developed to overcome limitations using the phage display 5 system, it should be noted that this method can also be used with the phage display system. Moreover, the selective mutagenesis approach can be used to improve the activity of any antibody. To improve the activity (e.g., affinity or neutralizing activity) of an antibody, ideally one would like to mutate every CDR position in both the heavy and light chains 10 to every other possible amino acid residue. However, since there are, on average, 70 CDR positions within an antibody, such an approach would be very time consuming and labor intensive.. Accordingly, the method of the invention allows one to improve the activity of the antibody by mutating only certain selected residues within the heavy and/or light chain CDRs. Furthermore, the method of the invention allows improvement 15 in activity of the antibody without affecting other desirable properties of the antibody. Determining which amino acid residues of an antibody variable region are in contact with an antigen cannot be accurately predicted based on primary sequence or their positions within the variable region. Nevertheless, alignments of sequences from antibodies with different specificities conducted by Kabat et al. have identified the 20 CDRs as local regions within the variable regions which differ significantly among antibodies (Kabat et al. (1971) Ann. NYAcad, Sci. 190:382-393,, Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). Structural studies have shown that the antigen binding surface is formed by amino acid residues present in the 25 CDRs. Other amino acid residues outside the CDR are also known to play structural roles or be directly involved in antigen binding. Therefore, for each antigen-antibody pair, amino acid residues within and outside of the CDRs may be important. The sequence alignment studies by Tomlison et al identified a number of positions in the heavy and light chain CDR 1 and CDR2, and in a portion of the kappa 30 chain CDR3 which are frequent sites of somatic mutation. (Tomlison et al (1996) J. Mol. Biol. 256: 813-817). In particular, positions H31, H31B, H33, H33B, H52B, H56, H58, L30, L31, L31A, L50, L53, L91, L92, L93 and L94 were identified as frequent sites for somatic mutation. However, this analysis excludes the important heavy chain 68 CDR3 regions, and sections of the light chain CDR3 which are known to lie in the center of an antibody binding site, and potentially provide important interactions with an antigen. Furthermore, Tomlison et al. propose that somatic diversity alone does not necessarily predict a role of a specific amino acid in antigen binding, and suggest 5 conserved amino acid residues that contact the antigen, and diverse amino acid residues which do not contact the antigen. This conclusion is further supported by mutational studies on the role of somatic mutations to antibody affinity (Sharon, (1990), PNAS, 87:4814-7). Nineteen somatic mutations in a high-affinity anti-p-azophenylarsonate (Ars) antibody were simultaneously replaced with their corresponding germline 10 residues, generating a germline version of the anti-Ars antibody which had a two hundred fold loss in activity. The full affinity of the anti-Ars antibody could be recovered by restoring only three of the nineteen somatic mutations, demonstrating that many somatic mutations may be permitted that do not contribute to antigen binding activity. 15 The result can be explained in part by the nature of antibody diversity itself. Immature B-cells may produce initially low affinity antibodies that recognize a number of self or non-self antigens. Moreover, antibodies may undergo in the course of affinity maturation sequence variations that may cause self-reactivity. Hypermutation of such low affinity antibodies may serve to abolish self-reactivity ("negative selection") and 20 increase affinity for the foreign antigen. Therefore, the analysis of primary and structural data of a large number of antibodies does not provide a method of predicting either (1) the role of somatic hyper-mutation sites in the affinity maturation process versus the process of decreasing affinity towards unwanted antigens, or (2) how a given amino acid contributes to the properties of a specific antigen-antibody pair. 25 Other attempts to address the role of specific amino acid residues in antigen recognition were made by analyzing a number of crystal structures of antigen-antibody complexes (MacCallum et al. (1996) J. Mol. Biol. 262: 732-745). The potential role of positions located within and outside the CDRs was indicated. Positions in CDRs involved in antigen binding in more than 10 of 26 analyzed structures included H31, 30 H33, H50, H52, H53, H54, H56, H58, H95, H96, H97, H98 and H100 in the heavy chain and L30A, L32, L91, L92, L93, L94, L96 in the light chain. However, the authors noted that prediction of antigen contacts using these and other structural data may over 69 and under predict contact positions, leading to the speculation that a different strategy may have to be applied to different antigens. Pini et al. describe randomizing multiple residues in antibody CDR sequences in a large phage display library to rapidly increase antibody affinity (Pini et al. (1998) J. 5 Biol Chem. 273: 21769-21776). However, the high affinity antibodies discussed by Pini et al. had mutations in a total of eight positions, and a reductionary analysis of which changes are absolutely required to improve affinity of the antibody becomes impractical because of the large number of possible combinations to be tested for the smallest number of amino acids required. 10 Furthermore, randomizing multiple residues may not necessarily preserve other desired properties of the antibody. Desirable properties or characteristics of an antibody are art-recognized and include for example, preservation of non-cross reactivity, e.g., with other proteins or human tissues and preservation of antibody sequences that are close to human germline immunoglobulin sequences improvement of neutralization 15 potency. Other desirable properties or characteristics include ability to preserve species cross reactivity, ability to preserve epitope specificity and ability to preserve high expression levels of protein in mammalian cells. The desirable properties or characteristics can be observed or measured using art-recognized techniques including but not limited to ELISA, competitive ELISA, in vitro and in vivo neutralization assays 20 (see e.g. Example 3 of US Patent No. 6,914,128), immunohistochemistry with tissue sections from different sources including human, primate or other sources as.the need may be, and studies to expression in mammalian cells using transient expression or stable expression. In addition, the method of Pini et al may introduce more changes than the 25 minimal number actually required to improve affinity and may lead to the antibodies triggering anti-human-antibody (HAMA) formation in human subjects. Further, as discussed elsewhere, the phage display as demonstrated here, or other related method including ribosome display may not work appropriately upon reaching certain affinities between antibody and antigen and the conditions required to reach 30 equilibrium may not be established in a reasonable time frame because of additional interactions including interactions with other phage or ribosome components and the antigen.
70 The ordinarily skilled artisan may glean interesting scientific information on the origin of antibody diversity from the teachings of the references discussed above. The present invention, however, provides a method for increasing antibody affinity of a specific antigen-antibody pair while preserving other relevant features or desirable 5 characteristics of the antibody. This is especially important'when considering the desirability of imparting a multitude of different characteristics on a specific antibody including antigen binding. If the starting antibody has desirable properties or characteristics which need to be retained, a selective mutagenesis approach can be the best strategy for preserving 10 these desirable properties while improving the activity of the antibody. For example, in the mutagenesis of Y61, the aim was to increase affinity for hIL-12, and to improve the neutralization potency of the antibody while preserving desired properties. Desired properties of Y61 included (1) preservation of non-cross reactivity with other proteins or human tissues, (2) preservation of fine epitope specificity, i.e. recognizing a p40 epitope 15 preferably in the context of the p70 (p40/p35) heterodimer, thereby preventing binding interference from free soluble p40; and (3) generation of an antibody with heavy and light chain amino acid sequences that were as close as possible to their respective germline immunoglobulin sequences. In one embodiment, the method of the invention provides a selective 20 mutagenesis approach as a strategy for preserving the desirable properties or characteristics of the antibody while improving the affinity and/or neutralization potency. The term "selective mutagenesis approach" is as defined above and includes a method of individually mutating selected amino acid residues. The amino acid residues to be mutated may first be selected from preferrd selective mutagenesis positions, then 25 from contact positions, and then from hypermutation positions. The individual selected position can be mutated to at least two other amino acid residue and the effect of the mutation both on the desired properties of the antibody, and improvement in antibody activity is determined. The Selective Mutagenesis approach comprises the steps of: 30 selecting candidate positions in the order 1) preferred selective mutagenesis positions; 2) contact positions; 3) hypermutation positions and ranking the positions based on the location of the position within the heavy and light chain variable regions of an antibody (CDR3 preferred over CDR2 preferred over CDR 1); 71 individually mutating candidate preferred selective mutagenesis positions, hypermutation and/or contact positions in the order of ranking, to all possible other amino acid residues and analyzing the effect of the individual mutations on the activity of the antibody in order to determine activity enhancing amino acid residues; 5 if necessary, making stepwise combinations of the individual activity enhancing amino acid residues and analyzing the effect of the various combinations on the activity of the antibodies; selecting mutant antibodies with activity enhancing amino acid residues and ranking the mutant antibodies based on the location and identity of the amino acid substitutions with regard to their immunogenic potential. Highest ranking is 10 given to mutant antibodies that comprise an amino acid sequence which nearly identical to a variable region sequence that is described in a germline database, or has an amino acid sequence that is comparable to other human antibodies. Lower ranking is given to mutant antibodies containing an amino acid substitution that is rarely encountered in either germline sequences or the sequences of other human antibodies. The lowest 15 ranking is given to mutant antibodies with an amino acid substitution that has not been encountered in a germline sequence or the sequence of another human antibody. As set forth above, mutant antibodies comprising at least one activity enhancing amino acid residue located in CDR3 is preferred over CDR2 which is preferred over CDR 1. The CDRs of the heavy chain variable regions are preferred over those of the light chain 20 variable region. The mutant antibodies can also be studied for improvement in activity, e.g. when compared to their corresponding parental antibody. The improvement in activity of the mutant antibody can be determined for example, by neutralization assays, or binding specificity/affinity by surface plasmon resonance analysis (see Example 3 of US Patent 25 No. 6,914,128). Preferably, the improvement in activity can be at least 2-20 fold higher than the parental antibody. The improvement in activity can be at least "x i " to "x2" fold higher than the parental antibody wherein "x i" and "x2" are integers between and including 2 to 20, including ranges within the state range, e.g. 2-15, e.g. 5-10. The mutant antibodies with the activity enhancing amino acid residue also can be 30 studied to determine whether at least one other desirable property has been retained after mutation. For example, with anti-hIL- 12 antibodies testing for, (1) preservation of non cross reactivity with other proteins or human tissues, (2) preservation of epitope recognition, i.e. recognizing a p40 epitope preferably in the context of the p70 (p4O/p35) 72 heterodimer, thereby preventing binding interference from free soluble p40; and (3) generation of antibodies with heavy and light chain amino acid sequences that were as close as possible to their respective germline immunoglobulin sequences, and determining which would be least likely to elicit a human immune response based on the 5 number of differences from the germline sequence. The same observations can be made on an antibody having.more than one activity enhancing amino acid residues, e.g. at least two or at least three activity enhancing amino acid residues, to determine whether retention of the desirable property or characteristic has occurred. An example of the use of a "selective mutagenesis approach", in the mutagenesis 10 of Y61 is described below. The individual mutations H31S->E, L50->Y, or L94G->Y each improved neutralization activity of the antibody. However, when combination clones were tested, the activity of the combined clone H31S->E + L50->Y + L94G->Y was no better than L50->Y + L94G->Y (J695). Therefore, changing the germline amino acid residue Ser to Glu at position 31 of CDR1 was unnecessary for the improved 15 activity of J695 over Y61. The selective mutagenesis approach therefore, identified the minimal number of changes that contributed to the final activity, thereby reducing the immunogenic potential of the final antibody and preserving other desired properties of the antibody. Isolated DNA encoding the VH and VL produced by the selected mutagenesis 20 approach can be converted into full length antibody chain genes, to Fab fragment genes as to a scFV gene, as described in section IV. For expression of VH and VL regions produced by the selected mutagenesis approach, expression vectors encoding the heavy and light chain can be transfected into variety host cells as described in detail in section IV. Preferred host cells include either prokaryotic host cells, for example, E coli, or 25 eukaryotic host cells, for example, yeast cells, e.g., S. cerevisae. Most preferred eukaryotic host cells are mammalian host cells, described in detail in section IV. The selective mutagenesis approach provides a method of producing antibodies with improved activities without prior affinity maturation of the antibody by other means. The selective mutagenesis approach provides a method of-producing antibodies 30 with improved affinities which have been subject to back mutations. The selective mutagenesis approach also provides a method of improving the activity of affinity matured antibodies.
73 The skilled artisan will recognize that the selective mutagenesis approach can be used in standard antibody manipulation techniques known in the art. Examples include, but are not limited to, CDR grafted antibodies, chimeric antibodies, scFV fragments, Fab fragments of a full length antibodies and human antibodies from other sources, e.g., 5 transgenic mice. Rapid large scale mutational analysis of antibodies include in vitro transcription and translation using ribosome display technology (see e.g., Hanes et al., (1997) Proc. Natl. A cad. Sci. 94: 4937-4942; Dall Acqua et al.,(1998) Cu-r. Opin. Struc. Biol. 8: 443-450; He et al., (1997) Nucleic Acid Res. 25: 5132-5134), and U.S. Patent Nos. 10 5,643,768 and 5,658,754 issued to Kawasaki. The selective mutagenesis approach also provides a method of producing antibodies with improved activities that can be selected using ribosomal display techniques. In the methods of the invention, antibodies or antigen binding portions thereof are further modified by altering individual positions in the CDRs of the HCVR and/or 15 LCVR. Although these modifications can be made in phage-displayed antibodies, the method is advantageous in that it can be performed with antibodies that are expressed in other types of host systems, such as bacterial, yeast or mammalian cell expression systems. The individual positions within the CDRs selected for modification are based on the positions being a contact and/or hypermutation position. 20 Preferred contact positions and hypermutation positions as defined herein are shown in Table 3 of US Patent No. 6,914,128 (see Appendix A of US Patent No. 6,914,128 and Table 3 of Appendix A attached hereto) and their modification in accordance with the method of the invention is described in detail in Example 2 of US Patent No. 6,914,128. Preferred contact positions are selected from the group consisting 25 of H30, H31, H3IB, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H101, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96. Preferred hypermutation positions are selected from the group consisting of H30, H31, H3IB, H32, H52, H56, H58, L30, L31, L32, L53 and L93. More preferred amino acid residues (referred to as "preferred selective mutagenesis positions") are both contact 30 and hypermutation positions and are selected from the group consisting of H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94. Particularly preferred contact positions are selected from the group consisting of L50 and L94.
74 Preferred activity enhancing amino acid residues replace amino acid residues located at positions selected from the group consisting of H30, H31, H31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIO, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94, and L96. More preferred activity 5 enhancing amino acid residues replace amino acid residues located at positions H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94. Particularly, preferred activity enhancing amino acid residues replace amino acid residues located at positions selected from the group consisting of L50 and L94. In general, the method of the invention involves selecting a particular preferred 10 selective mutagenesis position, contact and/or hypermutation position within a CDR of the heavy or light chain of a parent antibody of interest, or antigen binding portion thereof, randomly mutagenizing that individual position (e.g., by genetic means using a mutagenic oligonucleotide to generate a "mini-library" of modified antibodies), or mutating a position to specific desired amino acids, to identify activity enhancing amino 15 acid residues expressing, and purifying the modified antibodies (e.g., in a non-phage display host system), measuring the activity of the modified antibodies for antigen (e.g., by measuring koT rates by BlAcore analysis), repeating these steps for other CDR positions, as necessary, and combining individual mutations shown to have improved activity and testing whether the combinations) generate an antibody with even greater 20 activity (e.g., affinity or neutralizing potency) than the parent antibody, or antigen binding portion thereof. Accordingly, in one embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof; 25 b) selecting in order a 1) preferred selective mutagenesis position, 2) contact position, or 3) hypermutation position within a complementarity determining region (CDR) for mutation, thereby identifying a selected preferred selective mutagenesis position, contact or hypermutation position; c) individually mutating said selected preferred selective mutagenesis position, 30 contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof; 75 e) optionally, repeating steps a) through d) for at least one other preferred selective mutagenesis position, contact or hypermutation position; f) combining, in the parent antibody, or antigen-binding portion thereof, individual mutations shown to have improved activity, to form combination antibodies, 5 or antigen-binding portions thereof; and g) evaluating the activity of the combination antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof; until an antibody, or antigen-binding portion thereof, with an improved activity, relative to the parent antibody, or antigen-binding portion thereof, is obtained. Preferably, the 10 selected antibody or antibodies have an improved activity without loss or with retention of at least one desirable characteristic or property of the parental antibody as described above. The desirable characteristic or property can be measured or observed by the ordinarily skilled artisan using art-recognized techniques. Preferred contact positions are selected from the group consisting of H30, H3 1, 15 1-31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIO, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96. Preferred hypermutation positions are selected from the group consisting of H30, H31, H3IB, H32, H52, H56, H58, L30, L31, L32, L53 and L93. More preferred selective mutagenesis positions are selected from the group consisting of H30, H31, H3IB, H32, 20 H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93 and L94. Particularly preferred contact positions are selected from the group consisting of L50 and L94. In another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof; 25 b) selecting a preferred selective mutagenesis position, contact or hypermutation position within a complementarity determining region (CDR) for mutation; c) individually mutating said selected preferred selective mutagenesis position, contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof; 30 d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, thereby identifying an activity enhancing amino acid residue; 76 e) optionally, repeating steps a) through d) for at least one other preferred selective mutagenesis position, contact or hypermutation position; f) combining, in the parent antibody, or antigen-binding portion thereof, two individual activity enhancing amino acid residues shown to have improved activity, to 5 form combination antibodies, or antigen-binding portions thereof; and g) evaluating the activity of the combination antibodies, or antigen-binding portions thereof with two activity enhancing amino acid residues, relative to the parent antibody or antigen-binding portion thereof; until an antibody, or antigen-binding portion thereof, with an improved activity, relative 10 to the parent antibody, or antigen-binding portion thereof, is obtained. Preferred contact positions are selected from the group consisting of H30, H3 1, H3IB, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIOI, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96. Preferred hypermutation positions are selected from the group consisting of H30, H31, H31B, 15 H32, H52, H56, H58, L30, L31, L32, L53 and L93. More preferred selective mutagenesis positions are selected from the group consisting of H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93 and L94. Particularly preferred contact positions are selected from the group consisting of L50 and L94. In another embodiment, the invention provides a method for improving the 20 activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof; b) selecting a preferred selective mutagenesis position, contact or hypermutation position within a complementarity determining region (CDR) for mutation; c) individually mutating said selected preferred selective mutagenesis position, 25 contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, thereby identifying an activity enhancing amino acid residue; 30 e) optionally, repeating steps a) through d) for at least one other preferred selective mutagenesis position, contact or hypermutation position; 77 f) combining, in the parent antibody, or antigen-binding portion thereof, three individual activity enhancing amino acid residues shown to have improved activity, to form combination antibodies, or antigen-binding portions thereof; and g) evaluating the activity of the combination antibodies, or antigen-binding 5 portions thereof with two activity enhancing amino acid residues, relative to the parent antibody or antigen-binding portion thereof; until an antibody, or antigen-binding portion thereof, with an improved activity, relative to the parent antibody, or antigen-binding portion thereof, is obtained. Preferably, the activity enhancing amino acid residue replaces amino acid 10 residues located at positions selected from the group consisting of H30, H31, H31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H101, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96. Following mutagenesis of individual selected positions, mutated clones can be sequenced to identify which amino acid residues have been introduced into the selected 15 position in each clone. A small number of clones (e.g., about 24) can be selected for sequencing, which statistically should yield 10-15 unique antibodies, whereas larger numbers of clones (e.g., greater than 60) can be sequenced to ensure that antibodies with every possible substitution at the selected position are identified. In one embodiment, contact and/or hypermutation positions within the CDR3 20 regions of the heavy and/or light chains are first selected for mutagenesis. However, for antibodies that have already been affinity matured in vitro by random mutagenesis of the CDR3 regions via phage display selection, it may be preferably to first select contact and/or hypermutation positions within CDR1 or CDR2 of the heavy and/or light chain. In a more preferred embodiment, preferred selective mutagenesis positions 25 within the CDR3 regions of the heavy and/or light chains are first selected for mutagenesis. However, for antibodies that have already been affinity matured in vitro by random mutagenesis of the CDR3 regions via phage display selection, it may be preferably to first select preferred selective mutagenesis positions within CDR1 or CDR2 of the heavy and/or light chain. 30 In another preferred embodiment, the optimization of a selected antibody by the selective mutagenesis approach is done sequentially as follows: preferred selective mutagenesis positions selected from the group consisting of H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94 are mutated first to at 78 least 2 other amino acids each (preferably 5-14 other amino acids) and the resulting antibodies are characterized for increased affinity, neutralization potency (and possibly also for at least one other retained characteristic or property discussed elsewhere). If a mutation of a single preferred selective mutagenesis position does not increase the 5 affinity or neutralization potency at all or sufficiently and if even the combination of multiple activity enhancing amino acids replacing amino acids in preferred selective mutagenesis positions does not result in an combination antibody which meets the target activity (including affinity and/or neutralization potency), additional amino acid residues will be selected for selective mutagenesis from the group consisting of H35, H50, H53, 10 H54, H95, H96, H97, H98, L30A and L96 are mutated to at least 2 other amino acids each (preferably 5-14 other amino acids) and the resulting antibodies are characterized for increased affinity, neutralization potency (and possibly also for at least one other retained characteristic or property discussed elsewhere). If a mutation of a single amino acid residue selected from the group consisting of 15 H35, H50, H53, H54, H95, H96, H97, H98, L30A and L96 does not increase the activity (including affinity and/or neutralization potency) at all or not sufficiently and if even the combination of multiple activity enhancing amino acids replacing amino acids in those positions does not result in an combination antibody which meets the targeted activity (including affinity and/or target neutralization potency), additional amino acid residues 20 will be selected for selective mutagenesis from the group consisting of H33B, H52B, L31A and are mutated to at least 2 other amino acids each (preferably 5-14 other amino acids) and the resulting antibodies are characterized for increased affinity, neutralization potency (and possibly also for at least one other retained characteristic or property discussed elsewhere). 25 It should be understood that the sequential selective mutagenesis approach may end at any of the steps outline above as soon as an antibody with the desired activity (including affinity and neutralization potency) has been identified. If mutagenesis of the preselected positions has identified activity enhancing amino acids residues but the combination antibody still do not meet the targets set for activity (including affinity and 30 neutralization potency) and/or if the identified activity enhancing amino acids also affect other desired characteristics and are therefore not acceptable, the remaining CDR residues may be subjected to mutagenesis (see section IV).
79 The method of the invention can be used to improve activity of an antibody, or antigen binding portion thereof, to reach a predetermined target activity (e.g. a predetermined affinity and/or neutralization potency, and/or a desired property or characteristic). 5 Accordingly, the invention provides a method of improving the activity of an antibody, or antigen-binding portion thereof, to attain a predetermined target activity, comprising: a) providing a parent antibody a antigen-binding portion thereof; b) selecting a preferred selective mutagenesis position selected from group 10 consisting of H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94. c) individually mutating the selected preferred selective mutagenesis position to at least two other amino acid residues to hereby create a first panel of mutated antibodies, or antigen binding portions thereof; 15 d) evaluating the activity of the first panel of mutated antibodies, or antigen binding portions thereof to determined if mutation of a single selective mutagenesis position produces an antibody or antigen binding portion thereof with the predetermined target activity or a partial target activity; e) combining in a stepwise fashion, in the parent antibody, or antigen binding 20 portion thereof, individual mutations shown to have an improved activity, to form combination antibodies, or antigen binding portions thereof. f) evaluating the activity of the combination antibodies, or antigen binding portions thereof to determined if the combination antibodies, or antigen binding portions thereof have the predetermined target activity or a partial target activity. 25 g) if steps d) or f) do not result in an antibody or antigen binding portion thereof having the predetermined target activity, or result an antibody with only a partial activity, additional amino acid residues selected from the group consisting of H35, H50, H53, H54, H95, H96, H97, H98, L30A and L96 are mutated to at least two other amino acid residues to thereby create a second panel of mutated antibodies or antigen-binding 30 portions thereof; h) evaluating the activity of the second panel of mutated antibodies or antigen binding portions thereof, to determined if mutation of a single amino acid residue selected from the group consisting of H35, H50, H53, H54, H95, H96, H97, H98, 80 L30A and L96 results an antibody or antigen binding portion thereof, having the predetermined target activity or a partial activity; i) combining in stepwise fashion in the parent antibody, or antigen-binding portion thereof, individual mutations of step g) shown to have an improved activity, to 5 form combination antibodies, or antigen binding portions thereof; j) evaluating the activity of the combination antibodies or antigen binding portions thereof, to determined if the combination antibodies, or antigen binding portions thereof have the predetermined target activity or a partial target activity; k) if steps h) or j) do not result in an antibody or antigen binding portion thereof 10 having the predetermined target activity, or result in an antibody with only a partial activity, additional amino acid residues selected from the group consisting of H33B, H52B and L3 IA are mutated to at least two other amino acid residues to thereby create a third panel of mutated antibodies or antigen binding portions thereof; 1) evaluating the activity of the third panel of mutated antibodies or antigen binding 15 portions thereof, to determine if a mutation of a single amino acid residue selected from the group consisting of H33B, H52B and L3IA resulted in an antibody or antigen binding portion thereof, having the predetermined target activity or a partial activity; m) combining in a stepwise fashion in the parent antibody, or antigen binding 20 portion thereof, individual mutation of step k) shown to have an improved activity, to form combination antibodies, or antigen binding portions, thereof; n) evaluating the activity of the combination antibodies or antigen-binding portions thereof, to determine if the combination antibodies, or antigen binding portions thereof have the predetermined target activity to thereby produce an antibody or antigen 25 binding portion thereof with a predetermined target activity. A number of mutagenesis methods can be used, including PCR assembly, Kunkel (dut-ung-) and thiophosphate (Amersham Sculptor kit) oligonucleotide-directed mutagenesis. A wide variety of host expression systems can be used to express the mutated 30 antibodies, including bacterial, yeast, baculoviral and mammalian expression systems (as well as phage display expression systems). An example of a suitable bacterial expression vector is pUC1 19(Sfi). Other antibody expression systems are known in the art and/or are described below in section V.
81 The modified antibodies, or antigen binding portions thereof, produced by the method of the invention can be identified without the reliance on phage display methods for selection. Accordingly, the method of the invention is particularly advantageous for improving the activity of a recombinant parent antibody or antigen-binding portion 5 thereof, that was obtained by selection in a phage-display system but whose activity cannot be further improved by mutagenesis in the phage-display system. Accordingly, in another embodiment, the invention provides a method for improving the affinity of an antibody, or antigen-binding portion thereof, comprising: a) providing a recombinant parent antibody or antigen-binding portion thereof; 10 that was obtained by selection in a phage-display system but whose activity cannot be further improved by mutagenesis in said phage-display system; b) selecting a preferred selective mutagenesis position, contact or hypermutation position within a complementarity determining region (CDR) for mutation, thereby identifying a selected contact or hypermutation position; 15 c) individually mutating said selected preferred selective mutagenesis position, contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof, and expressing said panel in a non-phage display system; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding 20 portions thereof, relative to the parent antibody or antigen-binding portion thereof; e) optionally repeating steps b) through d) for at least one other preferred selective mutagenesis position, contact or hypermutation position; f) combining, in the parent antibody, or antigen-binding portion thereof, individual mutations shown to have improved activity, to form combination antibodies, 25 or antigen-binding portions thereof; and g) evaluating the activity of the combination antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof; until an antibody, or antigen-binding portion thereof, with an improved activity, relative to the parent antibody, or antigen-binding portion thereof, is obtained. 30 Preferred contact positions are selected from the group consisting of H30, H3 1, H31 B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIM0, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96. Preferred hypermutation positions are selected from the group consisting of H30, H31, H3IB, 82 H32, H52, H56, H58, L30, L31, L32, L53 and L93. More preferred selective mutagenesis positions are selected from the group consisting of H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93 and L94. Particularly preferred contact positions are selected from the group consisting of L50 and L94. 5 With available methods it is not possible or it is extremely laborious to derive an antibody with increased binding affinity and neutralization potency while retaining other properties or characteristics of the antibodies as discussed above. The method of this invention, however, can readily identify such antibodies. The antibodies subjected to the method of this invention can come from any source. 10 Therefore, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a recombinant parent antibody or antigen-binding portion thereof; b) selecting a preferred selective mutagenesis position, contact or hypermutation position within a complementarity determining region (CDR) for mutation, thereby 15 identifying a selected preferred selective mutagenesis position, contact or hypermutation position; c) individually mutating said selected preferred selective mutagenesis position, contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof and expressing 20 said panel in an appropriate expression system; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, thereby identifying an activity enhancing amino acid residue; e) evaluating the panel of mutated antibodies, or antigen-binding portions 25 thereof, relative to the parent antibody or antigen-binding portion thereof for at least one other property or characteristics, wherein the property or characteristic is one that needs to be retained in the antibody; until an antibody, or antigen-binding portion thereof, with an improved activity and at least one retained property or characteristic, relative to the parent antibody, or antigen 30 binding portion thereof, is obtained. In a preferred embodiment, the contact positions are selected from the group consisting of H30, H31, H31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H 101, L30, L31, L32, L34, L50, L52, L53, L55, L9 1, L92, L93, 83 L94 and L96 and the other characteristic is selected from 1) preservation of non crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p4O/p35 heterodimer preventing binding interference from free, soluble p 4 0 and/or 3) to produce 5 an antibody with a close to germline immunoglobulin sequence. In another preferred embodiment, the hypermutation positions are selected from the group consisting of H30, H3 1, H3 1 B, H32, H52, H56, H58, L30, L3 1, L32, L53 and L93 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. 10 recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In a more preferred embodiment the residues for selective mutagenesis are selected from the preferred selective mutagenesis positions from the group consisting of 15 H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to 20 germline immunoglobulin sequence. In a more preferred embodiment, the contact positions are selected from the group consisting of L50 and L94 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 25 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. If therefore, the affinity of an antibody for a specific antigen should be improved, but where the phage display (or related system including ribosome display) method is no longer applicable, and other desirable properties or characteristics should be retained, 30 the method of the invention can be used. Accordingly, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen binding portion thereof, comprising: 84 a) providing a recombinant parent antibody or antigen-binding portion thereof; that was obtained by selection in a phage-display system but whose activity cannot be further improved by mutagenesis in .said phage-display system; b) selecting a preferred selective mutagenesis position, contact or hypermutation 5 position within a complementarity determining region (CDR) for mutation, thereby identifying a selected preferred selective mutagenesis position, contact or hypermutation position; c) individually mutating said selected preferred selective mutagenesis position, contact or hypermutation position to at least two other amino acid residues to thereby 10 create a panel of mutated antibodies, or antigen-binding portions thereof, and expressing said panel in a non-phage display system; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof thereby identifying an activity enhancing amino acid residue; 15 e) evaluating the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof for at least one other property or characteristic, wherein the property or characteristic is one that needs to be retained, until an antibody, or antigen-binding portion thereof, with an improved activity and at least one retained property or characteristic, relative to the parent 20 antibody, or antigen-binding portion thereof, is obtained. f) optionally, repeating steps a) through e) for at least one other preferred selective mutagenesis position, contact or hypermutation position; g) combining, in the parent antibody, or antigen-binding portion thereof, at least two individual activity enhancing amino acid residues shown to have improved activity 25 and at least one retained property or characteristic, to form combination antibodies, or antigen-binding portions thereof; and h) evaluating the activity of the combination antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof; until an antibody, or antigen-binding portion thereof, with an improved activity and at 30 least one retained other property or characteristic, relative to the parent antibody, or antigen-binding portion thereof, is obtained. In a preferred embodiment, the contact positions are selected from the group consisting of H30, H31, H31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, 85 H95, H96, H97, H98, HIM0, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96 and the other characteristic is selected from 1) preservation of non crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 5 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In another preferred embodiment, the hypermutation positions are selected from the group consisting of H30, H3 1, H3 1 B, H32, H52, H56, H58, L30, L3 1, L32, L53 and L93 and the other characteristic is selected from 1) preservation of non-crossreactivity 10 with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitop'e preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In a more preferred embodiment the residues for selective mutagenesis are 15 selected from the preferred selective mutagenesis positions from the group consisting of H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding 20 interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In a more preferred embodiment, the contact positions are selected from the group consisting of L50 and L94 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation 25 of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: 30 a) providing a recombinant parent antibody or antigen-binding portion thereof; that was obtained by selection in a phage-display system but whose activity cannot be further improved by mutagenesis in said phage-display system; 86 b) selecting a preferred selective mutagenesis position, contact or hypermutation position within a complementarity determining region (CDR) for mutation, thereby identifying a selected contact or hypermutation position; c) individually mutating said selected preferred selective mutagenesis position, 5 contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof, and expressing said panel in a non-phage display system; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof 10 thereby identifying an activity enhancing amino acid residue; e) evaluating the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof for at least one other property or characteristic, wherein the property or characteristic is one that needs to be retained, until an antibody, or antigen-binding portion thereof, with an improved 15 activity and at least one retained property or characteristic, relative to the parent antibody, or antigen-binding portion thereof, is obtained. In a preferred embodiment, the contact positions are selected from the group consisting of H30, H31, H31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIO, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, 20 L94 and L96 and the other characteristic is selected from 1) preservation of non crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p4O/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. 25 In another preferred embodiment, the hypermutation positions are selected from the group consisting of H30, H3 1, H3 I B, H32, H52, H56, H58, L30, L3 1, L32, L53 and L93 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer 30 preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In a more preferred embodiment the residues for selective mutagenesis are selected from the preferred selective mutagenesis positions from the group consisting of 87 H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding 5 interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In a more preferred embodiment, the contact positions are selected from the group consisting of L50 and L94 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation 10 of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to -produce an antibody with a close to germline immunoglobulin sequence. In another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: 15 a) providing a recombinant parent antibody or antigen-binding portion thereof; that was obtained by selection in a phage-display system but whose.activity cannot be further improved by mutagenesis in said phage-display system; b) selecting a preferred selective mutagenesis position, contact or hypermutation position within a complementarity determining region (CDR) for mutation, thereby 20 identifying a selected contact or hypermutation position; c) individually mutating said selected preferred selective mutagenesis positions, contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof, and expressing said panel in a non-phage display system; 25 d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof thereby identifying tn activity enhancing amino acid residue; e) evaluating the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof for at least one 30 other property or characteristic, wherein the property or characteristic is one that needs to be retained, until an antibody, or antigen-binding portion thereof, with an improved activity and at least one retained characteristic, relative to the parent antibody, or antigen-binding portion thereof, is obtained.
88 f) optionally, repeating steps a) through e) for at least one other preferred selective mutagenesis position, contact or hypermutation position; g) combining, in the parent antibody, or antigen-binding portion thereof, at least two individual activity enhancing amino acid residues shown to have improved activity 5 and at least on retained other characteristic, to form combination antibodies, or antigen binding portions thereof; and h) evaluating the activity of the combination antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof; until an antibody, or antigen-binding portion thereof, with an improved activity and at 10 least one retained property or characteristic, relative to the parent antibody, or antigen binding portion thereof, is obtained. In a preferred embodiment, the contact positions are selected from the group consisting of H30, H31, H31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H101, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, 15 [94 and L96 and the other characteristic is selected from 1) preservation of non crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. 20 In another preferred embodiment, the hypermutation positions are selected from the group consisting of H30, H31, H3IB, H32, H52, H56, H58, L30, [31, L32, L53 and L93 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer 25 preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In a more preferred embodiment the residues for selective mutagenesis are selected from the preferred selective mutagenesis positions from the group consisting of H30, H31, H31B, H32, H33, H52, H56, H58, L30, L31, L32, L50, L91, L92, L93, L94 30 and the other characteristic is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding 89 interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. In a more preferred embodiment, the contact positions are selected from the group consisting of L50 and L94 and the other characteristic is selected from 1) 5 preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence. 10 IV. Modifications of other CDR residues Ultimately, all CDR residues in a given antibody-antigen pair identified by any means to be required as activity enhancing amino acid residues and/or required directly or indirectly for binding to the antigen and/or for retaining other desirable properties or characteristics of the antibody. Such CDR residues are referred to as "preferred 15 selective mutagenesis positions". It should be noted that in specific circumstances that preferred selective mutagenesis residues can be identified also'by other means including co-crystallization of antibody and antigen and molecular modeling. If the preferred attempts to identify activity enhancing amino acids focusing on the preferred selective mutagenesis positions, contact or hypermutation positions 20 described above are exhausted, or if additional improvements are required, the remaining CDR residues may be modified as described below. It should be understood that the antibody could already be modified in any one or more contact or hypermutation positions according to the embodiments discussed above but may require further improvements. Therefore, in another embodiment, the invention provides a method for 25 improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof; b) selecting an amino acid residue within a complementarity determining region (CDR) for mutation other than H30, H3 1, H3 1 B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIO, L30, L31, L32, L34, L50, L52, L53, 30 L55, L91, L92, L93, L94 and L96; c) individually mutating said selected position e.g., to at least two other amino acid residues to thereby create a mutated antibody or a panel of mutated antibodies, or antigen-binding portions thereof; 90 d) evaluating the activity of the mutated antibody or the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof thereby identifying an activity enhancing amino acid residue; 5 e) evaluating the mutated antibody or the panel of mutated antibodies, or antigen binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, for changes in at least one other property or characteristic until an antibody, or antigen-binding portion thereof, with an improved activity, relative to the parent antibody, or antigen-binding portion thereof, is obtained. 10 Preferably, the other characteristic or property is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence 15 If mutagenesis of a single residue is not sufficient other residues can be included; therefore, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof; b) selecting an amino acid residue within a complementarity determining region 20 (CDR) for mutation other than H30, H31, H3!B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H10 1, L30, L3 1, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96; c) individually mutating said selected position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions 25 thereof; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, thereby identifying an activity enhancing amino acid residue; e) repeating steps b) through d) for at least one other CDR position which is 30 neither the position selected under b) nor a position at H30, H3 1, H3 I B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H 101, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96; 91 f) combining, in the parent antibody, or antigen-binding portion thereof, at least two individual activity enhancing amino acid residues shown to have improved activity, to form combination antibodies, or antigen-binding portions thereof; and g) evaluating the activity of the combination antibodies, or antigen-binding 5 portions thereof with two activity enhancing amino acid residues, relative to the parent antibody or antigen-binding portion thereof until an antibody, or antigen-binding portion thereof, with an improved activity, relative to the parent antibody, or antigen-binding portion thereof, is obtained. If the preferred attempts to identify activity enhancing amino acids focusing on 10 the contact or hypermutation positions described above are exhausted, or if additional improvements are required, and the antibody in question can not further be optimized by mutagenesis and phage display (or related ribosome display) methods the remaining CDR residues may be modified as described below. It should be understood that the antibody could already be modified in any one or more preferred selective mutagenesis 15 position, contact or hypermutation positions according to the embodiments discussed above but may require further improvements. Therefore, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a recombinant parent antibody or antigen-binding portion thereof; 20 that was obtained by selection in a phage-display system but whose activity cannot be further improved by mutagenesis in said phage-display system; b) selecting a selecting an amino acid residue within a complementarity determining region (CDR) for mutation other than H30, H31, H31B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIM0, L30, L31, L32, 25 L34, L50, L52, L53, L55, L91, L92, L93, L94 and; c) individually mutating said selected contact or hypermutation position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof, and expressing said panel in a non-phage display system; 30 d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof thereby identifying an activity enhancing amino acid residue; 92 e) evaluating the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, for changes in at least one other property or characteristic, until an antibody, or antigen-binding portion thereof, with an improved activity, relative to the parent antibody, or antigen-binding 5 portion thereof, is obtained. Preferably, the other characteristic or property is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p4 0 epitope preferably in the context of the p70 p4O/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce 10 an antibody with a close to germline immunoglobulin sequence. If a single mutagenesis is not sufficient to increase the affinity of the antibody other residues may be included in the mutagenesis. Therefore, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen binding portion thereof, comprising: 15 a) providing a parent antibody or antigen-binding portion thereof that was obtained by selection in a phage-display system but Whose activity cannot be further improved by mutagenesis in said phage-display system; b) selecting an amino acid residue within a complementarity determining region (CDR) for mutation other than H30, H31, H31B, H32, H33, H35, H50, H52, H52A, 20 H53, H54, H56, H58, H95, H96, H97, H98, H101, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96; c) individually mutating said selected position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof and expression in a non-phage display system; 25 d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof thereby identifying an activity enhancing amino acid residue; e) repeating steps b) through d) for at least one other position which is neither the position selected under b) nor a position at H30, H31, H31B, H32, H33, H35, H50, H52, 30 H52A, H53, H54, H56, H58, H95, H96, H97, H98, H101, L30, L31, L32, L34, L50, L52,L53,L55,L91,L92,L93,L94; 93 g) combining, in the parent antibody, or antigen-binding portion thereof, at least two individual activity enhancing amino acid residues shown to have improved activity, to form combination antibodies, or antigen-binding portions thereof; and h) evaluating the activity and other property or characteristic of the combination 5 antibodies, or antigen-binding portions thereof with two activity enhancing amino acid residues, relative to the parent antibody or antigen-binding portion thereof; until an antibody, or antigen-binding portion thereof, with an improved activity, relative to the parent antibody, or antigen-binding portion thereof, is obtained. Preferably, the other characteristic or property is selected from 1) preservation of 10 non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p4O/p35 heterodimer preventing binding interference from free, soluble p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence The preferred attempts to identify activity enhancing amino acids focusing on the 15 preferred selective mutagenesis positions, contact or hypermutation positions described may be exhausted, or additional improvements may be required, and it is important to retain other properties or characteristics of the antibody. Therefore, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, without 20 affecting other characteristics, comprising: a) providing a parent antibody or antigen-binding portion thereof; b) selecting an amino acid residue within a complementarity determining region (CDR) for mutation other than H30, H31, H3IB, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H101, L30, L31, L32, L34, L50, L52, L53, 25 L55, L91, L92, L93, L94 and L96; c) individually mutating said selected position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding 30 portions thereof, relative to the parent antibody or antigen-binding portion thereof thereby identifying an activity enhancing amino acid residue; e) evaluating the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, for changes in 94 at least one other property or characteristic until an antibody, or antigen-binding portion thereof, with an improved activity and retained other property or characteristic, relative to the parent antibody, or antigen-binding portion thereof, is obtained. Preferably, the other characteristic or property is selected from 1) preservation of 5 non-crossreactivity with other proteins or human tissues, 2) preservation of epitope recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p40/p35 heterodimer preventing binding interference from free, soluble.p40 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence If mutagenesis of a single residue is not sufficient other residues can be included; 10 therefore, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof; b) selecting an amino acid residue within a complementarity determining region (CDR) for mutation other than H30, H31, H31B, H32, H33, H35, H50, H52, H52A, 15 H53, H54, H56, H58, H95, H96, H97, H98, HIO, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96; c) individually mutating said selected position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof; 20 d) evaluating the activity of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, thereby identifying an activity enhancing amino acid residue; e.) evaluating the panel of mutated antibodies or antigen-binding portions thereof, relative to the parent antibody or antigen-portion thereof, for changes in at least 25 one other characteristic or property; e) repeating steps b) through e) for at least one other CDR position which is neither the position selected under b) nor a position at H30, H31, H3IB, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H1OI, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96; 30 f) combining, in the parent antibody, or antigen-binding portion thereof, at least two individual activity enhancing amino acid residues shown to have improved activity and not affecting at least one other property or characteristic, to form combination antibodies, or antigen-binding portions thereof; and 95 g) evaluating the activity and the retention of at least one other property or characteristic of the combination antibodies, or antigen-binding portions thereof with two activity enhancing amino acid residues, relative to the parent antibody or antigen binding portion thereof until an antibody, or antigen-binding portion thereof, with an 5 improved activity and at least one retained other property or characteristic, relative to the parent antibody, or antigen-binding portion thereof, is obtained. Mutagenesis of the preferred selective mutagenesis position, contact and hypermutation residues may not have increased the affinity of the antibody sufficiently, and mutagenesis and the phage display method (or related ribosome display method) 10 may no longer be useful and at least one other characteristic or property of the antibody should be retained. Therefore, in another embodiment the invention provides a method to improve the affinity of an antibody or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof that was 15 obtained by selection in a phage-display system but whose activity cannot be further improved by mutagenesis in said phage-display system; b) selecting an amino acid residue within a complementarity determining region (CDR) for mutation other than H30, H3 1, H3 1 B, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIO, L30, L31, L32, L34, L50, L52, L53, 20 L55, L91, L92, L93, L94 and L96; c) individually mutating said selected position to at least two other amino acid residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof and expression in a non-phage display system; d) evaluating the activity of the panel of mutated antibodies, or antigen-binding 25 portions thereof, relative to the parent antibody or antigen-binding portion thereof thereby identifying an activity enhancing amino acid residue; e) evaluating the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, for changes in at least one other property or characteristic until an antibody, or antigen-binding portion 30 thereof, with an improved activity, relative to the parent antibody, or antigen-binding portion thereof, is obtained. Preferably, the other characteristic or property is selected from 1) preservation of non-crossreactivity with other proteins or human tissues, 2) preservation of epitope 96 recognition, i.e. recognizing p40 epitope preferably in the context of the p70 p 4 0 /p 3 5 heterodimer preventing binding interference from free, soluble p4 0 and/or 3) to produce an antibody with a close to germline immunoglobulin sequence If mutagenesis of a single residue is not sufficient other residues can be included; 5 therefore, in another embodiment, the invention provides a method for improving the activity of an antibody, or antigen-binding portion thereof, comprising: a) providing a parent antibody or antigen-binding portion thereof that was obtained by selection in a phage-display system but whose activity cannot be further improved by mutagenesis in said phage-display system; 10 b) selecting an amino acid residue within a complementarity determining region (CDR) for mutation other than H30, H31, H3IB, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, HIM0, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96; c) individually mutating said selected position to at least two other amino acid 15 residues to thereby create a panel of mutated antibodies, or antigen-binding portions thereof and expression in a non-phage display system; d) evaluating the activity and retention of at least one other property or characteristic of the panel of mutated antibodies, or antigen-binding portions thereof, relative to the parent antibody or antigen-binding portion thereof, thereby identifying an 20 activity enhancing amino acid residue; e) repeating steps b) through d) for at least one other CDR position which is neither the position selected under b) nor a position at H30, H3 1, H3IB, H32, H33, H35, H50, H52, H52A, H53, H54, H56, H58, H95, H96, H97, H98, H10M, L30, L31, L32, L34, L50, L52, L53, L55, L91, L92, L93, L94 and L96; 25 f) combining, in the parent antibody, or antigen-binding portion thereof, at least two individual activity enhancing amino acid residues shown to have improved activity and not to affect at least one other property or characteristic, to form combination antibodies, or antigen-binding portions thereof; and g) evaluating the activity and retention of at least one property or characteristic 30 of the combination antibodies, or antigen-binding portions thereof with two activity enhancing amino acid residues, relative to the parent antibody or antigen-binding portion thereof until an antibody, or antigen-binding portion thereof, with an improved activity 97 and at least one other retained characteristic or property, relative to the parent antibody, or antigen-binding portion thereof, is obtained. V. Expression of Antibodies 5 An antibody, or antibody portion, of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell. To express an antibody recombinantly, a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody such that the light and heavy chains are expressed 10 in the host cell and, preferably, secreted into the medium in which the host cells are cultured, from which medium the antibodies can be recovered. Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Fritsch and Maniatis (eds), 15 Molecular Cloning; A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Ausubel, F.M. et al. (eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, (1989) and in U.S. Patent No. 4,816,397 by Boss et al. To obtain a DNA fragment encoding the heavy chain variable region of Joe 9 wt or a Joe 9 wt-related antibody, antibodies specific for human IL- 12 were screened from 20 human libraries and mutated, as described in section II. Once DNA fragments encoding Joe 9 wt or Joe 9 wt-related VH and VL segments are obtained, mutagenesis of these sequences is carried out by standard methods, such as PCR site directed mutagenesis (PCR-mediated mutagenesis in which the mutated nucleotides are incorporated into the PCR primers such that the PCR product contains the mutations) or other site-directed 25 mutagenesis methods. Human IL-12 antibodies that displayed a level of activity and binding specificity/affinity that was desirable, for example J695, were further manipulated by standard recombinant DNA techniques, for example to convert the variable region genes to full-length antibody chain genes, to Fab fragment genes or to a scFv gene. In these manipulations, a VL- or VH-encoding DNA fragment is operatively 30 linked to another DNA fragment encoding another protein, such as an antibody constant region or a flexible linker. The term "operatively linked", as used in this context, is intended to mean that the two DNA fragments are joined such that the amino acid sequences encoded by the two DNA fragments remain in-frame.
98 The isolated DNA encoding the VH region can be converted to a full-length heavy chain gene by operatively linking the VH-encoding DNA to another DNA molecule encoding heavy chain constant regions (CH1, CH2 and CH3). The sequences of human heavy chain constant region genes are known in the art (see e.g., Kabat, E.A., 5 et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification. The heavy chain constant region can be an IgGI, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region and any allotypic variant therein as described in Kabat (, Kabat, 10 E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242), but most preferably is an IgG 1 or IgG4 constant region. For a Fab fragment heavy chain gene, the VH-encoding DNA can be operatively linked to another DNA molecule encoding only the heavy chain CHI constant region. 15 The isolated DNA encoding the VL region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the VL-encoding DNA to another DNA molecule encoding the light chain constant region, CL. The sequences of human light chain constant region genes are known in the art (see e.g., Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth 20 Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) and DNA fragments encompassing these regions can be obtained by standard PCR amplification. The light chain constant region can be a kappa or lambda constant region, but most preferably is a lambda constant region. To create a scFv gene, the VH- and VL-encoding DNA fragments are operatively 25 linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4-Ser) 3 , such that the VH and VL sequences can be expressed as a contiguous single-chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al. (1988) Science 242:423-426; Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883; McCafferty et al., Nature (1990) 348:552-554). 30 To express the antibodies, or antibody portions of the invention, DNAs encoding partial or full-length light and heavy chains, obtained as described above, are inserted into expression vectors such that the genes are operatively linked to transcriptional and translational control sequences. In this context, the term "operatively linked" is intended 99 to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene. The expression vector and expression control sequences are chosen to be compatible with the expression host 5 cell used. The antibody light chain gene and the antibody heavy chain gene can be inserted into separate vector or, more typically, both genes are inserted into the same expression vector. The antibody genes are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present). Prior to 10 insertion of the J695 or J695-related light or heavy chain sequences, the expression vector may already carry antibody constant region sequences. For example, one approach to converting the J695 or J695-related VH and VL sequences to full-length antibody genes is to insert them into expression vectors already encoding heavy chain constant and light chain constant regions, respectively, such that the VH segment is 15 operatively linked to the CH segment(s) within the vector and the VL segment is operatively linked to the CL segment within the vector. Additionally or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell. The antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the 20 antibody chain gene. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein). In addition to the antibody chain genes, the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the antibody chain genes in a host cell. The term "regulatory sequence" is intended to include 25 promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes. Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). It will,be appreciated by those skilled in the art that the design of the expression vector, 30 including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Preferred regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as 100 promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma. For further description of viral regulatory elements, and sequences thereof, see e.g., U.S. 5 Patent No. 5,168,062 by Stinski, U.S. Patent No. 4,510,245 by Bell et al. and U.S. Patent No. 4,968,615 by Schaffner et al., U.S. Patent No.5,464,758 by Bujard et al. and U.S. Patent No. 5,654,168 by Bujard et al. In addition to the antibody chain genes and regulatory sequences, the recombinant expression vectors of the invention may carry additional sequences, such as 10 sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Patents Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al.). For example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on 15 a host cell into which the vector has been introduced. Preferred selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr- host cells with methotrexate selection/amplification) and the neo gene (for G418 selection). For expression of the light and heavy chains, the expression vector(s) encoding the heavy and light chains is transfected into a host cell by standard techniques. The 20 various forms of the term "transfection" are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE dextran transfection and the like. Although it is theoretically possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of 25 antibodies in eukaryotic cells, and most preferably mammalian host cells, is the most preferred because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody. Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) 30 (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R.J. Kaufman and P.A. Sharp (1982) Mol. Biol. 159:601-621), NSO myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding antibody genes are 101 introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using 5 standard protein purification methods. Host cells can also be used to produce portions of intact antibodies, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are within the scope of the present invention. For example, it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy 10 chain (but not both) of an antibody of this invention. Recombinant DNA technology may also be used to remove some or all of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to hIL-12 The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and 15 one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than hIL-12 by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods. In a preferred system for recombinant expression of an antibody, or antigen binding portion thereof, of the invention, a recombinant expression vector encoding both 20 the antibody heavy chain and the antibody light chain is introduced into dhfr- CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to enhancer/promoter regulatory elements (e.g., derived from SV40, CMV, adenovirus and the like, such as a CMV enhancer/AdMLP promoter regulatory element or an SV40 25 enhancer/AdMLP promoter regulatory element) to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are culture to allow for expression of the antibody heavy and light chains and intact antibody is recovered from 30 the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover tlie antibody from the culture medium. Antibodies or antigen binding portions thereof of the invention can be expressed in an animal (e.g., a mouse) 102 that is transgenic for human immunoglobulin genes (see e.g., Taylor, L.D. et al. (1992) Nucl. Acids Res. 20: 6287-6295). Plant cells can also be modified to create transgenic plants that express the antibody or antigen binding portion thereof, of the invention. In view of the foregoing, another aspect of the invention pertains to nucleic acid, 5 vector and host cell compositions that can be used for recombinant expression of the antibodies and antibody portions of the invention. Preferably, the invention features isolated nucleic acids that encode CDRs of J695, or the full heavy and/or light chain variable region of J695. Accordingly, in one embodiment, the invention features an isolated nucleic acid encoding an antibody heavy chain variable region that encodes the 10 J695 heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 25. Preferably, the nucleic acid encoding the antibody heavy chain variable region further encodes a J695 heavy chain CDR2 which comprises the amino acid sequence of SEQ ID NO: 27. More preferably, the nucleic acid encoding the antibody heavy chain variable region further encodes a J695 heavy chain CDR] which comprises the amino 15 acid sequence of SEQ ID NO: 29. Even more preferably, the isolated nucleic acid encodes an antibody heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 31 (the full VH region of J695). In other embodiments, the invention features an isolated nucleic acid encoding an antibody light chain variable region that encodes the J695 light chain CDR3 20 comprising the amino acid sequence of SEQ ID NO: 26. Preferably, the nucleic acid encoding the antibody light chain variable region further encodes a J695 light chain CDR2 which comprises the amino acid sequence of SEQ ID NO: 28. More preferably, the nucleic acid encoding the antibody light chain variable region further encodes a J695 light chain CDR1 which comprises the amino acid sequence of SEQ ID NO: 30. Even 25 more preferably, the isolated nucleic acid encodes an antibody light chain variable region comprising the amino acid sequence of SEQ ID NO: 32 (the full VL region of J695). The invention also provides recombinant expression vectors encoding both an antibody heavy chain and an antibody light chain. For example, in one embodiment, the 30 invention provides a recombinant expression vector encoding: a) an antibody heavy chain having a variable region comprising the amino acid sequence of SEQ ID NO: 31; and 103 b) an antibody light chain having a variable region comprising the amino acid sequence of SEQ ID NO: 32. The invention also provides host cells into which one or more of the recombinant expression vectors of the invention have been introduced. Preferably, the host cell is a 5 mammalian host cell, more preferably the host cell is a CHO cell, an NSO cell or a COS cell. Still further the invention provides a method of synthesizing a recombinant human antibody of the invention by culturing a host cell of the invention in a suitable culture medium until a recombinant human antibody of the invention is synthesized. The method can further comprise isolating the recombinant human antibody from the culture 10 medium. VI. Pharmaceutical Compositions and Pharmaceutical Administration The antibodies and antibody-portions of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the 15 pharmaceutical composition comprises an antibody or antibody portion of the invention and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include 20 one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, 25 preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion. The antibodies and antibody-portions of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration. Preferably, the antibody or antibody-portions will be prepared as an injectable solution containing 0.1- 30 250 mg/ml antibody. The injectable solution can be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule or pre-filled syringe. The buffer can be L-histidine (1-50 mM), optimally 5-10 mM, at pH 5.0 to 7.0 (optimally pH 6.0). Other suitable buffers include but are not limited to, sodium succinate, sodium citrate, 104 sodium phosphate or potassium phosphate. Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form). Cryoprotectants can be included for a lyophilized dosage form, principally 0-10% sucrose (optimally 0.5-1.0%). Other suitable cryoprotectants include 5 trehalose and lactose. Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 2-4%). Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L-Methionine (optimally 5-10 mM). Other suitable bulking agents include glycine, arginine, can be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%). Additional surfactants include but are not 10 limited to polysorbate 20 and BRU surfactants. In one embodiment, the invention provides a formulation comprising the antibody in combination with a polyol, a surfactant, a stabilizer, and a buffer system with a pH of about 5 to 5. In one embodiment said formulation is free of metal. In a preferred embodiment, the formulation comprises the antibody and mannitol, histidine, 15 methionine, polysorbate 80, hydrochloric acid, and water. In one embodiment, an aqueous formulation is prepared comprising the antibody in a pH-buffered solution. The buffer of this invention has a pH ranging from about 4 to about 8, preferably from about 4.5 to about 7.5, more preferably from about 5 to about 7, more preferably from about 5.5 to about 6.5, and most preferably has a pH of about 20 6.0 to about 6.2. In a particularly preferred embodiment, the buffer has a pH of about 6. Ranges intermediate to the above recited pH's are also intended to be part of this invention. For example, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included. Examples of buffers that will control the pH within this range include acetate (e.g. sodium acetate), 25 succinate (such as sodium succinate), gluconate, histidine, citrate, phosphate and other organic acid buffers. In a preferred embodiment of the invention, the formulation contains a buffer system comprising histidine. In a preferred embodiment of the invention, the buffer is histidine, e.g., L-histidine. In preferred embodiments, the formulation of the invention comprises a buffer system comprising about 1-100 mM 30 histidine, preferably about 5-50 mM histidine, and most preferably 10 mM histidine. One of skill in the art will recognize that sodium chloride can be used to modify the toxicity of the solution, e.g., at a concentration of 1-300 mM, and optimally 150 mM for a liquid dosage form.
105 A polyol, which acts as a tonicifier and may stabilize the antibody, is also included in the formulation. The polyol is added to the formulation in an amount that may vary with respect to the desired isotonicity of the formulation. Preferably the aqueous formulation is isotonic. The amount of polyol added may also vary with respect 5 to the molecular weight of the polyol. For example, a lower amount of a monosaccharide (e.g., mannitol) may be added, compared to a disaccharide (such as trehalose). In a preferred embodiment of the invention, the polyol that is used in the formulation as a tonicity agent is mannitol. In a preferred embodiment, the composition comprises about 1 Oto about 100 mg/ml, or about 20 to about 80, about 20 to about 70, 10 about 30 to about 60, about 30 to about 50 mg/ml of mannitol, for example, about 10, about 20, about 30, about 40, about 50, about 60, about 70, about 80, about 90, and about 100 mg/ml of mannitol In a preferred embodiment, the formulation comprises about 40 mg/ml of mannitol (corresponding to about 4% mannitol). In a preferred embodiment, the composition comprises between about 1% to about 10% mannitol, 15 more preferably between about 2% to about 6% mannitol, and most preferably about 4% mannitol. In another embodiment of the invention, the polyol sorbitol is included in the formulation. A stabilizer or antioxidant is also added to the antibody formulation. A stabilizer can be used in both liquid and lyophilized dosage forms. Formulations of the invention 20 preferably comprise the stabilizer methionine, e.g., L-Methionine. Other stabilizers useful in formulations of the invention are known to those of skill in the art and include, but are not limited to, glycine and arginine. Cryoprotectants can be included for a lyophilized dosage form, principally sucrose (e.g., 1-10% sucrose, and optimally 0.5 1.0% sucrose). Other suitable cyroprotectants include trehalose and lactose. 25 A detergent or surfactant is also added to the antibody formulation. Exemplary detergents include nonionic detergents such as polysorbates (e.g., polysorbates 20, 80 etc.) or poloxamers (e.g., poloxamer 188). The amount of detergent added is such that it reduces aggregation of the formulated antibody and/or minimizes the formation of particulates in the formulation and/or reduces adsorption. In a preferred embodiment of 30 the invention, the formulation includes a surfactant that is a polysorbate. In another preferred embodiment of the invention, the formulation contains the detergent polysorbate 80 or Tween 80. Tween 80 is a term used to describe polyoxyethylene (20) sorbitanmonooleate (see Fiedler, Lexikon der Hifsstoffe, Editio Cantor Verlag 106 Aulendorf, 4th ed., 1996). In one preferred embodiment, the formulation contains between 0.001 to about 0.1% polysorbate 80, or between about 0.005 and 0.05% polysorbate 80, for example, about 0.001, about 0.005, about 0.01, about 0.05, or about 0.1% polysorbate 80. In a preferred embodiment, about 0.01% polysorbate 80 is found 5 in the formulation of the invention. In a preferred embodiment of the invention, the formulation is a 1.0 mL solution in a container containing the ingredients shown below in Table 1. In another embodiment, the formulation is a 0.8 mL solution in a container. Table 1: A 1.0 mL Solution" of J695 Formulation for Injection 10 Name of Ingredient Quantity Function Active substance: Antibody (J695) 2 ) 50.0 or 100.0 mg Active substance Excipients: Mannitol 40 mg Tonicity agent Polysorbate 80 0.10 mg Detergent/Surfactant Histidine 1.55 mg Buffer Methionine 1.49 mg Stabilizer Water for injection To one Iml Solvent Hydrochloric Acid q.s. pH adjustment to 6.0 'Density of the solution: 1.0398 g/mL 2) Is used as concentrate In one embodiment, the formulation is a formulation described in U.S. 15 Application Serial No. 12/625,057, which published as U.S. 2010/0172862 A1, the entire contents of which are hereby expressly incorporated by reference. In one embodiment, the formulation contains the above-identified agents (i.e., antibody, polyol, surfactant, stabilizer and buffer) and is essentially free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and 20 benzethonium Cl. In another embodiment, a preservative may be included in the formulation, particularly where the formulation is a multidose formulation. One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those 107 described in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980) may be included in the formulation provided that they do not significantly adversely affect the desired characteristics of the formulation. Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed and 5 include; additional buffering agents; co-solvents; antioxidants such as ascorbic acid; chelating agents such as EDTA; metal complexes (e.g. Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium. In one embodiment, the formulations of the invention have improved properties 10 as compared to art-recognized formulations. For example, the formulations of the invention have an improved shelf life and/or stability as compared to art recognized formulations. In one embodiment, the formulations of the invention have a shelf life of at least 18 months, e.g., in a liquid state or in a solid state. In another embodiment, the formulations of the invention have a shelf life of at least 24 months, e.g., in a liquid state 15 or in a solid state. In a preferred embodiment, the formulations of the invention have a shelf life of at least 24 months at a temperature of 2-8*C. In a preferred embodiment, the formulations of the invention have a shelf life of at least 18 months or of at least 24 months at a temperature of between about -20 and -80*C. In another embodiment, the formulations of the invention maintain stability following at least 5 freeze/thaw cycles of 20 the formulation. In a preferred aspect, the formulations of the invention comprise, e.g., an antibody, comprising at least a portion of a lambda light chain, e.g., J695, wherein the formulation provides enhanced resistance to fragmentation of the lambda light chain, e.g., reduced cleavage of the lambda light chain, as compared to art recognized formulations. 25 In one embodiment, the formulations of the invention are substantially free of metal. In one embodiment, the formulations of the invention are substantially free of a metal selected from the group consisting of Fe2+, Fe3+, Ca2+ and Cul+. In.one embodiment, the formulations of the invention comprise an amount of metal that is sufficiently low to reduce or prevent cleavage of the lambda chain in the presence of 30 histidine, e.g., the metal is present at a concentration of less than about 5,060 ppb, less than about 1,060 ppb, less than about 560 ppb, less than about 500 ppb, less than about 450 ppb, less than about 400 ppb, less than about 350 ppb, less than about 310 ppb, less than about 300 ppb, less than about 250 ppb, less than about 200 ppb, less than about 108 160 ppb, less than about 150 ppb, less than about 140 ppb, less than about 130 ppb, less than about 120 ppb, less than about 110 ppb, less than about 100 ppb, less than about 90 ppb, less than about 80 ppb, less than about 70 ppb, less than about 60 ppb, less than about 50 ppb, less than about 40 ppb, less than about 30 ppb, less than about 20 ppb, less 5 than about 10 ppb, or less than about 1 ppb. In one embodiment, the metal is present at a concentration of less than about 160 ppb. In one embodiment, the metal is present at a concentration of less than about 110 ppb. In one embodiment, the metal is present at a concentration of less than about 70 ppb, e.g., a concentration of about 60 ppb. Maximum concentrations intermediate to the above recited concentrations, e.g., less than 10 about 65 ppb, are also intended to be part of this invention. Further, ranges of values using a combination of any of the above recited values as upper and/or lower limits, e.g., concentrations between about 50 ppb and about 70 ppb, are also intended to be included. In one embodiment, the formulations of the invention are substantially free of metal following subjection to at least one procedure that removes metal, such as 15 filtration, buffer exchange, chromatography or resin exchange. Procedures useful to remove metal from formulations of the invention are known to one of skill in the art and are further described herein. In one embodiment, the formulations of the invention comprise a metal chelator, e.g., such that the molecule is not cleaved within the hinge region or is cleaved within the hinge region at a level which is less than the level of 20 cleavage observed in the absence of the metal chelator. In the formulations of the invention, the metal chelator may be, for example, a siderophore, calixerenes, an aminopolycarboxylic acid, a hydroxyaminocarboxylic acid, an N-substituted glycine, a 2-(2-amino-2-oxoethyl)aminoethane sulfonic acid (BES), a bidentate, tridentate or hexadentate iron chelator, a copper chelator, and derivatives, analogues, and 25 combinations thereof. Metal chelators useful in formulations of the invention are known to one of skill in the art, and are further described below. Particular siderophores useful in formulations of the invention include, but are not limited to, aerobactin, agrobactin, azotobactin, bacillibactin, N-(5-C3-L (5 aminopentyl) hydroxycarbamoyl)-propionamido)pentyl)-3(5-(N-hydroxyacetoamido) 30 pentyl)carbamoyl)- proprionhydroxamic acid (deferoxamine, desferrioxamine or DFO or DEF), desferrithiocin, enterobactin, erythrobactin, ferrichrome, ferrioxamine B, ferrioxamine E, fluviabactin, fusarinine C, mycobactin, parabactin, pseudobactin, 109 vibriobactin, vulnibactin, yersiniabactin, ornibactin, and derivatives, analogues, and combinations thereof. Aminopolycarboxylic acids useful in formulations of the invention include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), nitriloacetic acid (NTA), 5 trans-diaminocyclohexane tetraacetic acid (DCTA), diethylenetriamine pentaacetic acid (DTPA), N-2-acetamido-2-iminodiacetic acid (ADA), aspartic acid, bis(aminoethyl)glycolether N,N,N'N'-tetraacetic acid (EGTA), glutamic acid, and N,N' bis (2-hydroxybenzyl)ethylenediamine-N,N'-diacetic acid (HBED), and derivatives, analogues, and combinations thereof. 10 Hydroxyaminocarboxylic acids useful in formulations of the invention include, but are not limited to, N*-hydroxyethyliminodiacetic acid (HIMDA), N,N bishydroxyethylglycine (bicine), and N-(trishydroxymethylmethyl) glycine (tricine), and derivatives, analogues, and combinations thereof. N-substituted glycines, e.g., glycylglycine, as well as derivatives, analogues, or combinations thereof, are also useful 15 as metal chelators in formulations of the invention. The metal chelator 2-(2-amino-2 oxoethyl)aminoethane sulfonic acid (BES), and derivatives, analogues, and combinations thereof, can also be used. Particular calixarenes useful in formulations of the invention include, but are not limited to, a macrocycle or cyclic oligomer based on a hydroxyalkylation product of a 20 phenol and an aldehyde, and derivatives, analogues, and combinations thereof. Particular copper chelators useful in the invention include triethylenetetramine (trientine), etraethylenepentamine, D-penicillamine, ethylenediamine, bispyridine, phenantroline, bathophenanthroline, neocuproine, bathocuproine sulphonate, cuprizone, cis,cis-l,3,5,-triaminocyclohexane (TACH), tachpyr, and derivatives, analogues, and 25 combinations thereof. Additional metal chelators that can be employed in formulations of the invention include a hydroxypyridine-derivate, a hydrazone-derivate, and hydroxyphenyl-derivate, or a nicotinyl-derivate, such as l,2-dimethyl-3-hydroxypyridin-4-one (Deferiprone, DFP or Ferriprox); 2-deoxy-2-(N-carbamoylmethyl-[N'-2'-methyl-3'-hydroxypyridin-4'-onel) 30 D- glucopyranose (Feralex-G), pyridoxal isonicotinyl hydrazone (PIH); 4,5-dihydro-2 (2,4- dihydroxyphenyl)-4-methylthiazole-4-carboxylic acid (GT56-252), 4-[3,5- bis(2 hydroxyphenyl)-[l,2,4]triazol-1-yl]benzoic acid (ICL-670); N,N'-bis(o- 110 hydroxybenzyl)ethylenediamine-N,N'-diacetic acid (HBED), 5-chloro-7-iodo-quinolin 8-ol (clioquinol), and derivatives, analogues, and combinations thereof. It will be recognized that combinations of two or more of any of the foregoing metal chelators can be used in combination in the formulations of the invention. For 5 example, in a particular embodiment of the invention, the formulation comprises a combination of DTPA and DEF. In another embodiment, the formulation comprises a combination of EDTA, EGTA and DEF. The amount of antibody present in the formulation is determined, for example, by taking into account the desired dose volumes and mode(s) of administration. In one 10 embodiment of the invention, the concentration of the antibody in the formulation is between about 0.1 to about 250 mg of antibody per ml of liquid formulation. In one embodiment of the invention, the concentration of the antibody in the formulation is between about 1 to about 200 mg of antibody per ml of liquid formulation. In various embodiments, the concentration of the antibody in the formulation is between about 30 15 to about 140 mg per ml, between about 40 to about 120 mg/ml, between about 50 to about 110 mg/ml, or between about 60 to about 100 mg/ml. The formulation is especially suitable for large antibody dosages of more than 15 mg/ml. In various embodiments, the concentration of the antibody in the formulation is about 1, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 20 230, 240 or 250 mg/ml. In a preferred embodiment, the concentration of the antibody is 50 mg/ml. In another preferred embodiment, the concentration of the antibody is 100 mg/ml. In a preferred embodiment, the concentration of the antibody is at least about 100 mg/ml, at least about 110 mg/ml or at least about 120 mg/ml. In various embodiments of the invention, the concentration of the antibody in the 25 formulation is about 0.1-250 mg/ml, 0.5-220 mg/ml, 1-2 10 mg/ml, about 5-200 mg/ml, about 10-195 mg/ml, about 15-190 mg/ml, about 20-185 mg/ml, about 25-180 mg/ml, about 30-175 mg/ml, about 35-170 mg/ml, about 40-165 mg/ml, about 45-160 mg/ml, about 50-155 mg/ml, about 55-150 mg/ml, about 60-145 mg/ml, about 65-140 mg/ml, about 70-135 mg/ml, about 75-130 mg/ml, about 80-125 mg/ml, about 85-120 mg/ml, 30 about 90-115 mg/ml, about 95-110 mg/ml, about 95-105 mg/ml, or about 100 mg/ml. Ranges intermediate to the above recited concentrations, e.g., about 31-174 mg/ml, are also intended to be part of this invention. For example, ranges of values using a 111 combination of any of the above recited values as upper and/or lower limits are intended to be included. In one, the formulation provides an effective dose of 40 mg, 50 mg, 80 mg, 100 mg , or 200 mg per injection of the active ingredient, the antibody. In another 5 embodiment, the formulation provides an effective dose which ranges from about 0.1 to 250 mg of antibody. If desired, the effective daily dose of the pharmaceutical formulation may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In an embodiment of the invention, the dosage of the antibody in the 10 formulation is between about 1 to about 200 mg. In an embodiment, the dosage of the antibody in the formulation is between about 30 and about 140 mg, between about 40 and about 120 mg, between about 50 and about 110 mg, between about 60 and about 100 mg, or between about 70 and about 90 mg. In one embodiment, the pharmaceutical composition includes the antibody at a dose of about 100 to about 200 mg. In a further 15 embodiment, the composition includes the antibody at about 1, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240 or 250 mg. Ranges intermediate to the above recited dosages, e.g., about 2-139 mg, are also intended to be part of this invention. For example, ranges of values using a combination 20 of any of the above recited values as upper and/or lower limits are intended to be included. The compositions of this invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, 25 liposomes and suppositories. The preferred form depends on the intended mode of administration and therapeutic application. Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. The preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, 30 intramuscular). In a preferred embodiment, the antibody, or antigen-binding fragment thereof, is administered by subcutaneous injection. Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, 112 microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed 5 by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile, lyophilized powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any 10 additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, 15 monostearate salts and gelatin. The antibodies and antibody-portions of the present invention can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route/mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by the skilled artisan, the route and/or mode of 20 administration will vary depending upon the desired results. In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, 25 collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. In certain embodiments, an antibody or antibody portion of the invention may be 30 orally administered, for example, with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the compounds may be incorporated with 113 excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its 5 inactivation. Supplementary active compounds can also be incorporated into the compositions. In certain embodiments, an antibody or antibody portion of the invention is coformulated with and/or coadministered with one or more additional therapeutic agents that are useful for treating disorders in which IL-12 activity is detrimental. For 10 example, an anti-hIL-12 antibody or antibody portion of the invention may be coformulated and/or coadministered with one or more additional antibodies that bind other targets (e.g., antibodies that bind other cytokines or that bind cell surface molecules). Furthermore, one or more antibodies of the invention may be used in combination with two or more of the foregoing therapeutic agents. Such combination 15 therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. It will be appreciated by the skilled practitioner that when the antibodies of the invention are used as part of a combination therapy, a lower dosage of antibody may be desirable than when the antibody alone is administered to a subject (e.g., a 20 synergistic therapeutic effect may be achieved through the use of combination therapy which, in turn, permits use of a lower dose of the antibody to achieve the desired therapuetic effect). Interleukin 12 plays a critical role in the pathology associated with a variety of diseases involving immune and inflammatory elements. These diseases include, but are 25 not limited to, rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, Lyme arthritis, psoriatic arthritis, reactive arthritis, spondyloarthropathy, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, inflammatory bowel disease, insulin dependent diabetes mellitus, thyroiditis, asthma, allergic diseases, psoriasis, dermatitis scleroderma, atopic dermatitis, graft versus host disease, organ transplant rejection, 30 acute or chronic immune disease associated with organ transplantation, sarcoidosis, atherosclerosis, disseminated intravascular coagulation, Kawasaki's disease, Grave's disease, nephrotic syndrome, chronic fatigue syndrome, Wegener's granulomatosis, Henoch-Schoenlein purpurea, microscopic vasculitis of the kidneys, chronic active 114 hepatitis, uveitis, septic shock, toxic shock syndrome, sepsis syndrome, cachexia, infectious diseases, parasitic diseases, acquired immunodeficiency syndrome, acute transverse myelitis, Huntington's chorea, Parkinson's disease, Alzheimer's disease, stroke, primary biliary cirrhosis, hemolytic anemia, malignancies, heart failure, 5 myocardial infarction, Addison's disease, sporadic, polyglandular deficiency type I and polyglandular deficiency type II, Schmidt's syndrome, adult (acute) respiratory distress syndrome, alopecia, alopecia areata, seronegative arthopathy, arthropathy, Reiter's disease, psoriatic arthropathy, ulcerative colitic arthropathy, enteropathic synovitis, chlamydia, yersinia and salmonella associated arthropathy, spondyloarthopathy, 10 atheromatous disease/arteriosclerosis, atopic allergy, autoimmune bullous disease, pemphigus vulgaris, pemphigus foliaceus, pemphigoid, linear IgA disease, autoimmune haemolytic anaemia, Coombs positive haemolytic anaemia, acquired pernicious anaemia, juvenile pernicious anaemia, myalgic encephalitis/Royal Free Disease, chronic mucocutaneous candidiasis, giant cell arteritis, primary sclerosing hepatitis, cryptogenic 15 autoimmune hepatitis, Acquired Immunodeficiency Disease Syndrome, Acquired Immunodeficiency Related Diseases, Hepatitis C, common varied immunodeficiency (common variable hypogammaglobulinaemia), dilated cardiomyopathy, female infertility, ovarian failure, premature ovarian failure, fibrotic lung disease, cryptogenic fibrosing alveolitis, post-inflammatory interstitial lung disease, interstitial pneumonitis, 20 connective tissue disease associated interstitial lung disease, mixed connective tissue disease associated lung disease, systemic sclerosis associated interstitial lung disease, rheumatoid arthritis associated interstitial lung disease, systemic lupus erythematosus associated lung disease, dermatomyositis/polymyositis associated lung disease, Sjbgren's disease associated lung disease, ankylosing spondylitis associated lung disease, 25 vasculitic diffuse lung disease, haemosiderosis associated lung disease, drug-induced interstitial lung disease, radiation fibrosis, bronchiolitis obliterans, chronic eosinophilic pneumonia, lymphocytic infiltrative lung disease, postinfectious interstitial lung disease, gouty arthritis, autoimmune hepatitis, type-l autoimmune hepatitis (classical autoimmune or lupoid hepatitis), type-2 autoimmune hepatitis (anti-LKM antibody 30 hepatitis), autoimmune mediated hypoglycemia, type B insulin resistance with acanthosis nigricans, hypoparathyroidism, acute immune disease associated with organ transplantation, chronic immune disease associated with organ transplantation, osteoarthrosis, primary sclerosing cholangitis, idiopathic leucopenia, autoimmune 115 neutropenia, renal disease NOS, glomerulonephritides, microscopic vasulitis of the kidneys, lyme disease, discoid lupus erythematosus, male infertility idiopathic or NOS, sperm autoimmunity, multiple sclerosis (all subtypes), insulin-dependent diabetes mellitus, sympathetic ophthalmia, pulmonary hypertension secondary to connective 5 tissue disease, Goodpasture's syndrome, pulmonary manifestation of polyarteritis nodosa, acute rheumatic fever, rheumatoid spondylitis, Still's disease, systemic sclerosis, Takayasu's disease/arteritis, autoimmune thrombocytopenia, idiopathic thrombocytopenia, autoimmune thyroid disease, hyperthyroidism, goitrous autoimmune hypothyroidism (Hashimoto's disease), atrophic autoimmune hypothyroidism, primary 10 myxoedema, phacogenic uveitis, primary vasculitis and vitiligo. The human antibodies, and antibody portions of the invention can be used to treat autoimmune diseases, in particular those associated with inflammation, including, rheumatoid spondylitis, allergy, autoimmune diabetes, autoimmune uveitis. Preferably, the antibodies of the invention or antigen-binding portions thereof, 15 are used to treat rheumatoid arthritis, Crohn's disease, multiple sclerosis, insulin dependent diabetes mellitus and psoriasis, as described in more detail in section VII. A human antibody, or antibody portion, of the invention also can be administered with one or more additional therapeutic agents useful in the treatment of autoimmune and inflammatory diseases. 20 Antibodies of the invention, or antigen binding portions thereof can be used alone or in combination to treat such diseases. It should be understood that the IL- 12 antibodies of the invention or antigen binding portion thereof can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the 25 additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the present invention. The additional agent also can be an agent which imparts a beneficial attribute to the therapeutic composition e.g., an agent which effects the viscosity of the composition. It should further be understood that the combinations which are to be included 30 within this invention are those combinations useful for their intended purpose. The agents set forth below are illustrative for purposes and not intended to be limited. The combinations which are part of this invention can be the antibodies of the present invention and at least one additional agent selected from the lists below. The 116 combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function. Furthermore, additional agents described herein used in combination with an IL-12 antibody, are not limited to the disorder to which they are attributed for 5 treatment. Preferred combinations are non-steroidal anti-inflammatory drug(s) also referred to as NSAIDS which include drugs like ibuprofen. Other preferred combinations are corticosteroids including prednisolone; the well known side-effects of steroid use can be reduced or even eliminated by tapering the steroid dose required when treating patients 10 in combination with the anti-IL-12 antibodies of this invention. Non-limiting examples of therapeutic agents for rheumatoid arthritis with which an antibody, or antibody portion, of the invention can be combined include the following: cytokine suppressive anti-inflammatory drug(s) (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, TNF (including adalimumab / HUMIRA), LT, 15 IL-1, IL-2, IL-6, IL-7, IL-8, IL-15, IL-16, IL-18, EMAP-II, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, or their ligands including CD154 (gp39 or CD40L). 20 Preferred combinations of therapeutic agents may interfere at different points in the autoimmune and subsequent inflammatory cascade; preferred examples include TNF antagonists like chimeric, humanized or human TNF antibodies, D2E7, (U.S. application serial number 08/599,226 filed February 9, 1996), cA2 (Remicade T), CDP 571, anti TNF antibody fragments (e.g., CDP870), and soluble p55 or p75 TNF receptors, 25 derivatives thereof, (p75TNFR 1 gG (Enbrellm) or p55TNFR I gG (Lenercept), soluble IL 13 receptor (sIL-13), and also TNFot converting enzyme (TACE) inhibitors; similarly IL-I inhibitors (e.g., Interleukin-l-converting enzyme inhibitors, such as Vx740, orIL IRA etc.) may be effective for the same reason. Other preferred combinations include Interleukin 11, anti-P7s and p-selectin glycoprotein ligand (PSGL). Yet another 30 preferred combination are other key players of the autoimmune response which may act parallel to, dependent on or in concert with IL-12 function; especially preferred are IL 18 antagonists including IL-18 antibodies or soluble IL-18 receptors, or IL-18 binding proteins. It has been shown that IL-12 and IL-18 have overlapping but distinct functions 117 and a combination of antagonists to both may be most effective. Yet another preferred combination are non-depleting anti-CD4 inhibitors. Yet other preferred combinations include antagonists of the co-stimulatory pathway CD80 (B7. 1) or CD86 (B7.2) including antibodies, soluble receptors or antagonistic ligands. 5 Anti-IL12 antibodies, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, 6-MP, azathioprine sulphasalazine, mesalazine, olsalazine chloroquinine/hydroxychloroquine, pencillamine, aurothiomalate (intramuscular and oral), azathioprine, cochicine, corticosteroids (oral, inhaled and local injection), beta-2 adrenoreceptor agonists (salbutamol, terbutaline, salmeteral), xanthines 10 (theophylline, aminophylline), cromoglycate, nedocromil, ketotifen, ipratropium and oxitropium, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signaling by proinflammatory 15 cytokines such as TNFa or IL-I (e.g. IRAK, NIK, IKK , p38 or MAP kinase inhibitors), IL- IP converting enzyme inhibitors (e.g., Vx740), anti-P7s, p-selectin glycoprotein ligand (PSGL), TNFa converting enzyme (TACE) inhibitors, T-cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6 mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors 20 and derivatives thereof (e.g. soluble p55 or p75 TNF receptors and the derivatives p75TNFRIgG (Enbrel
T
m)and p55TNFRIgG (Lenercept), sIL-IRI, sIL-IRII, sIL-6R, soluble ILIl 3 receptor (sIL-1 3)) and anti-inflammatory cytokines (e.g. IL-4, IL-10, IL 11, IL- 13 and TGFP). Preferred combinations include methotrexate or leflunomide and in moderate or severe rheumatoid arthritis cases, cyclosporine. 25 Non-limiting examples of therapeutic agents for inflammatory bowel disease with which an anti-IL- 12 antibody, or antibody portion, can be combined include the following: budenoside; epidermal growth factor; corticosteroids; cyclosporin, sulfasalazine; aminosalicylates; 6-mercaptopurine; azathioprine; metronidazole; lipoxygenase inhibitors; mesalamine; olsalazine; balsalazide; antioxidants; thromboxane 30 inhibitors; IL-I receptor antagonists; anti-IL-i I monoclonal antibodies; anti-IL-6 monoclonal antibodies; growth factors; elastase inhibitors; pyridinyl-imidazole compounds; antibodies to or antagonists of other human cytokines or growth factors, for example, TNF (including adalimumab / IUMIRA), LT, IL-I, IL-2, IL-6, IL-7, IL-8, IL- 118 15, IL-16, IL-18, EMAP-II, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 or their ligands. The antibodies of the invention, or antigen binding portions 5 thereof, may also be combined with agents, such as methotrexate, cyclosporin, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adenosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signaling by proinflammatory cytokines such as TNFx or IL-I (e.g. IRAK, NIK, 10 IKK, p38 or MAP kinase inhibitors), IL-1 converting enzyme inhibitors (e.g., Vx740), anti-P7s, p-selectin glycoprotein ligand (PSGL), TNFx converting enzyme inhibitors, T cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives thereof (e.g. soluble p55 or p75 15 TNF receptors, sIL-IRI, sIL-IRII, sIL-6R, soluble IL-13 receptor (sIL-13)) and anti inflammatory cytokines (e.g. IL-4, IL-10, IL-11, IL-13 and TGFP). Preferred examples of therapeutic agents for Crohn's disease in which an antibody or an antigen binding portion can be combined include the following: TNF antagonists, for example, anti-TNF antibodies, D2E7 (adalimumab / HUMIRA), cA2 20 (Remicade T M ), CDP 571, anti-TNF antibody fragments (e.g., CDP870), TNFR-Ig constructs(p75TNFRIgG (Enbrell" and p55TNFRIgG (Lenercept)) , anti-P7s, p selectin glycoprotein ligand (PSGL), soluble IL-13 receptor (sIL-13), and PDE4 inhibitors. Antibodies of the invention or antigen binding portions thereof, can be combined with corticosteroids, for example, budenoside and dexamethasone. 25 Antibodies may also be combined with agents such as sulfasalazine, 5-aminosalicylic acid and olsalazine, and agents which interfere with synthesis or action of proinflammatory cytokines such as IL-1, for example, IL- 1 P converting enzyme inhibitors (e.g., Vx740) and IL-lra. Antibodies or antigen binding portion thereof may also be used with T cell signaling inhibitors, for example, tyrosine kinase inhibitors 6 30 mercaptopurines. Antibodies or antigen binding portions thereof, can be combined with IL- 11. Non-limiting examples of therapeutic agents for multiple sclerosis with which an antibody, or antibody portion, can be combined include the following: corticosteroids; 119 prednisolone; methylprednisolone; azathioprine; cyclophosphamide; cyclosporine; methotrexate; 4-aminopyridine; tizanidine; interferon- la (Avonex; Biogen); interferon j 1b (Betaseron; Chiron/Berlex); Copolymer 1 (Cop-1; Copaxone; Teva Pharmaceutical Industries, Inc.); hyperbaric oxygen; intravenous immunoglobulin; clabribine; 5 antibodies to or antagonists of other human cytokines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-6, IL-7, IL-8, IL-15, IL-16, IL-18, EMAP-II, GM-CSF, FGF, and PDGF. Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25,.CD28, CD30, CD40, CD45, CD69, CD80, CD86, CD90 or their ligands. The 10 antibodies of the invention, or antigen binding portions thereof, may also be combined with agents, such as methotrexate, cyclosporine, FK506, rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, for example, ibuprofen, corticosteroids such as prednisolone, phosphodiesterase inhibitors, adensosine agonists, antithrombotic agents, complement inhibitors, adrenergic agents, agents which interfere with signaling by 15 proinflammatory cytokines such as TNFoa or IL-I (e.g. IRAK, NIK, IKK, p38 or MAP kinase inhibitors), IL-1 converting enzyme inhibitors (e.g., Vx740), anti-P7s, p-selectin glycoprotein ligand (PSGL), TACE inhibitors, T-cell signaling inhibitors such as kinase inhibitors, metalloproteinase inhibitors, sulfasalazine, azathioprine, 6-mercaptopurines, angiotensin converting enzyme inhibitors, soluble cytokine receptors and derivatives 20 thereof (e.g. soluble p55 or p75 TNF receptors, sIL- 1 RI, sIL- RII, sIL-6R, soluble IL- 13 receptor (sIL-13)) and anti-inflammatory cytokines (e.g. IL-4, IL-10, IL-13 and TGF). Preferred examples of therapeutic agents for multiple sclerosis in which the antibody or antigen binding portion thereof can be combined to include interferon-, for example, IFN la and IFNO lb; copaxone, corticosteroids, IL-I inhibitors, TNF 25 inhibitors, and antibodies to CD40 ligand and CD80. An antibody, antibody portion, may be used in combination with other agents to treat skin conditions. For example, an antibody, antibody portion, or other IL-12 inhibitor of the invention is combined with PUVA therapy. PUVA is a combination of psoralen (P) and long-wave ultraviolet radiation (UVA) that is used to treat many 30 different skin conditions. The antibodies, antibody portions, or other IL-12 inhibitors of the invention can also be combined with pimecrolimus. In another embodiment, the antibodies of the invention are used to treat psoriasis, wherein the antibodies are administered in combination with tacrolimus. In a further embodiment, tacrolimus and 120 IL-12 inhibitors are administered in combination with methotrexate and/or cyclosporine. In still another embodiment, the IL-12 inhibitor of the invention is administered with excimer laser treatment for treating psoriasis. The pharmaceutical compositions of the invention may include a "therapeutically 5 effective amount" or a "prophylactically effective amount" of an antibody or antibody portion of the invention. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may vary according to factors such as the disease state, age, sex, and weight of the individual, and 10 the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired 15 prophylactic result. Typically,'since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount. Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be 20 administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used 25 herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active 30 compound and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
121 Treatment of psoriasis may be achieved by administration of a single dose amount (or more than one sub-doses totaling the dose amount) of a substance according to a single periodicity. In one embodiment, a method of treating psoriasis in a subject comprises 5 administering to the subject an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, according to a periodicity of about once every 4 weeks, thereby treating psoriasis in the subject. In another embodiment, a method of treating psoriasis in a subject comprises administering to the subject an antibody, or antigen-binding portion thereof, which is 10 capable of binding to the p40 subunit of IL-12 and/or IL-23, according to a periodicity of about once every 12 weeks, thereby treating psoriasis in the subject. Thus, a single periodicity may be employed in a single treatment regimen. Alternatively, multiple periodicities may be employed in a single treatment regimen. For example, a first dose amount may be administered according to a first periodicity, 15 and then the first dose amount or a second dose amount may be administered according to a second periodicity. Furthermore, the first dose amount or second dose amount administered according to a second periodicity may optionally be followed by a first, second, or third dose amount administered according to a third periodicity. In one embodiment, an antibody, or antigen-binding portion thereof, which is 20 capable of binding to the p40 subunit of IL-12 and/or IL-23 is administered to a subject as a first dose.amount according to a periodicity, and is further administered to the subject as a second dose amount at the same periodicity. In another embodiment, an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23 is administered to a subject 25 as a first dose amount according to a periodicity, and is further administered to the subject as a seconddose amount according to a second periodicity. In one embodiment, an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23 is administered to a subject as a first dose amount according to a periodicity, and is further administered to the 30 subject as a second dose amount according to a second periodicity, and is further administered to the subject as a first, second, or third dose amount according to a third periodicity.
122 The first dose amount of the antibody, or antigen-binding portion thereof, may be at least about 100 mg to about 200 mg, is at least about 100 mg, or is at least about 200 mg. The first dose amount of the antibody, or antigen-binding portion thereof, may be about 100 mg, about 110 mg, about 120 mg, about 130 mg, about 140 mg, about 150 5 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, or about 200 mg. In one embodiment, the first dose amount is about 180-220 mg, 185-215 mg, 190-210 mg, or 195-205 mg. In one embodiment, the first dose amount is 200 mg. In one embodiment, the first dose amount is about 80-120 mg, 85-115 mg, 90-110 mg or 95 105 mg. In one embodiment, the first dose amount is 100 mg. It should be noted that 10 doses intermediate to the above specified doses are also included herein, e.g., 105 mg, 127 mg, etc. The second dose amount of the antibody, or antigen-binding portion thereof, may be the same as the first dose amount of the antibody, or antigen-binding portion thereof, or different than the first dose amount of the antibody, or antigen-binding portion 15 thereof. The second dose amount of the antibody, or antigen-binding portion thereof, may be at least about 100 mg to about 200 mg, is at least about 200 mg, or is at least about 100 mg. Alternatively, the second dose amount of the antibody, or antigen binding portion thereof, is about 40-60% (e.g., 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59 or 60%), e.g., about 50%, of the first dose amount of 20 the antibody, or antigen-binding portion thereof, or antigen-binding portion thereof, or about 190-210% (e.g., 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210%), e.g., about 200%, of the first dose amount of the antibody, or antigen-binding portion thereof. The second dose amount of the antibody, or antigen-binding portion thereof, may be about 100 mg, about I10 mg, about 25 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, or about 200 mg. In one embodiment, the second dose amount is about 80-120 mg, 85-115 mg, 90-110 mg or 95-105 mg. In one embodiment, the second dose amount is 100 mg. In another embodiment, the second dose amount is about 180-220 mg, 185-215 mg, 190-210 mg, or 195-205 mg. In one embodiment, the 30 second dose amount is 200 mg. It should be noted that doses intermediate to the above specified doses are also included herein, e.g., 105 mg, 127 mg, etc. The first and second periodicities of administration of the antibody, or antigen binding portion thereof, may be about once a week, about once every other week, about 123 once every four weeks. The second periodicity of administration of the antibody, or antigen-binding portion thereof, may be about once every 30-200 days. The duration of the first periodicity may be about 12 weeks, about 8 weeks, about 4 weeks, about 2 weeks, or about I week. 5 The duration of the second periodicity may be about 60 weeks, about 44 weeks, about 12 weeks, about 4 weeks, about 2 weeks, or about I week. The duration of a third periodicity may be, for example, about 4 weeks, about 12 weeks, about 24 weeks, about 36 weeks, about 48 weeks or about 60 weeks.. Thus, in one aspect, a method of treating psoriasis in a subject comprises 10 administering to the subject a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23; and a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a periodicity of about once every 12 weeks, thereby treating psoriasis in the subject. 15 In another aspect, a method of treating psoriasis in a subject comprises administering to the subject a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a first periodicity of about once every 4 weeks; and administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen 20 binding portion thereof, according to a second periodicity of about once every 4 weeks, thereby treating psoriasis in the subject. In another aspect, a method of treating psoriasis in a subject comprises administering to the subject a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, 25 according to a first periodicity of about once every 4 weeks; and a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks; and the second dose amount of the antibody, or antigen-binding portion thereof, according to a third periodicity of about once every 12 weeks, thereby treating psoriasis in the subject. 30 In one embodiment, the second dose amount is administered to the subject upon a flare of psoriasis. In another embodiment, the second dose amount is administered to the subject prior to a flare of psoriasis.
124 The flare of psoriasis may be monitored by determining a subject's Psoriasis Area and Severity Index (PASI), e.g., PASI 100 response, PASI 90 response, PASI 75 response, PASI 50 response, the PASI response of a single body region, two body regions, three body regions, or four body regions, e.g., trunk, lower extremities, upper 5 extremities, or head and neck. Alternatively, the flare of psoriasis may be monitored by determining a subject's Physician's Global Assessment (PGA) rating. In one embodiment, the subject achieves or maintains a specific response to treatment. In one embodiment, the subject achieves or maintains at least a PASI 50 response. In one embodiment, the subject achieves or maintains at least a PASI 75 10 response. In one embodiment, the subject achieves or maintains at least a PASI 90 response. In one embodiment, the subject achieves or maintains at least a PASI 100 response. In one embodiment the PASI 50, 75, 90, or 100 response is achieved by about (e.g., at least about) week 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 15 45, 46, 47, 48, 49, 50, 51, or 52 following treatment (e.g., following initial treatment, e.g., at week 0). In one embodiment, the PASI 50, 75, 90, or 100 response is maintained for about (e.g., at least about) 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41,42,43, 44, 45, 46, 47, 48, 49, 50, 51, 52 weeks, e.g., following administration of a first dose 20 amount at a first periodicity, or following administration of a first or second dose amount at a second periodicity, or following administration of a first, second or third dose amount according to a third periodicity. In one embodiment, the PASI 50, 75, 90 or 100 response is maintained, once achieved, throughout the duration of treatment. In one embodiment, the subject achieves a PGA score of 0 or 1. In one 25 embodiment the PGA score of 0 or 1 is achieved by about (e.g., at least about) week 4, 5,6,7,8,9, 10, 11, 12, 13, 14, 15,16,17, 18, 19,20,21,22,23,24,25,26,27,28,29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, or 52 following treatment (e.g., following initial treatment, e.g., at week 0). In one embodiment, the PGA score of 0 or 1 is maintained for about (e.g., at least about) 4, 5, 6, 30 7, 8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31,32,33,34,35,36,37,38,39, 40,41,42,43,44,45,46,47,48,49,50,51,52 weeks, e.g., following administration of a first dose amount at a first periodicity, or following administration of a first or second dose amount at a second periodicity, or following 125 administration of a first, second or third dose amount according to a third periodicity. In one embodiment, the PGA score of 0 or 1 is maintained, once achieved, throughout the duration of treatment. In one embodiment, the subject achieves a PGA score of 0, i.e., total clearance. 5 In one embodiment the PGA score of 0 is achieved by about (e.g., at least about) week 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29,30,31, 32, 33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,or 52 following treatment (e.g., following initial treatment, e.g., at week 0). In one embodiment, the PGA score of 0 is maintained for about (e.g., at least about) 4, 5, 6, 7, 10 8,9, 10, 11, 12, 13, 14,15, 16, 17,18,19,20,21,22,23,24,25,26,27,28,29,30,31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52 weeks, e.g., following administration of a first dose amount at a first periodicity, or following administration of a first or second dose amount at a second periodicity, or following administration of a first, second or third dose amount according to a third periodicity. In 15 one embodiment, the PGA score of 0 is maintained, once achieved, throughout the duration of treatment. A method of treating psoriasis in a population of subjects may comprise administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein at 20 least 60% of the population of subjects achieve a PASI 75 response, e.g., by about week 12. A method of treating psoriasis in a population of subjects may comprise administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 25 least 25% of the population of subjects achieve a PASI 90 response, e.g., by about week 12. A method of treating psoriasis in a population of subjects may comprise administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein at 30 least 10% of the population of subjects achieve a PASI 100 response, e.g., by about week 12. A method of treating psoriasis in a subject or a population of subjects may comprise administering to the subject or each subject in the population an antibody, or 126 antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein the subject or a percentage of the population of subjects (e.g., at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of a population of subjects) achieves at least 5 a PASI 50 response by about week 12, 24, 36, 48, 52, or 60. A method of treating psoriasis in a subject or a population of subjects may comprise administering to the subject or each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein the subject or a percentage of the population of subjects (e.g., at 10 least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of a population of subjects) achieves at least a PASI 75 response by about week 12, 24, 36, 48, 52, or 60. A method of treating psoriasis in a subject or a population of subjects may comprise administering to the subject or each subject in the population an antibody, or 15 antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL-12 and/or IL-23, wherein the subject or a percentage of the population of subjects (e.g., at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of a population of subjects) achieves at least a PASI 90 response by about week 12, 24, 36, 48, 52, or 60. 20 A method of treating psoriasis in a subject or a population of subjects may comprise administering to the subject or each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein the subject or a percentage of the population of subjects (e.g., at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 25 75%, 80%, 85%, 90%, 95%, 99%, or 100% of a population of subjects) achieves at least a PASI 100 response by about week 12, 24, 36, 48, 52, or 60. A method of treating psoriasis in a subject or a population of subjects may comprise administering to the subject or each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 30 and/or IL-23, wherein the subject or a percentage of the population of subjects (e.g., at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of a population of subjects) achieves at least a PGA score of 0 or 1 by about week 12, 24, 36, 48, 52, or 60.
127 In one aspect, the subject or population of subjects treated achieves an improvement in a Dermatology Life Quality Index (DLQI) score or mean Dermatology Life Quality Index (DLQI) score of at least about -6.8, -6.9, -7.0, -8.0, -8.5, -9, -10, 10.5, -11, -12, -13, -14, -15, -16, -17, -18, -19, -20 or lower. An improvement in DLQI 5 is a reduction in DLQI score, e.g., a reduction by at least about 6.8, 6.9, 7.0, 8.0, 8.5, 9, 10, 10.5, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more. Dermatology Life Quality Index (DLQI) is a patient-reported measure of the extent to which psoriasis impacts health related quality of life. The DLQI yields a score ranging from 0 to 30, with a lower score indicating lower impact. 10 In certain embodiments, the subject achieves a clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score. A clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score may be, e.g., a decrease of greater than 5 points in DLQI score. In another aspect, the subject or population of subjects treated achieves an '15 improvement in a Short Form 36 Health Survey Physical Component Summary (PCS) score or mean Physical Component Summary (PCS) score of at least about 2, 3, 4, 5, 6, or more. An improvement in PCS is an increase in PCS score, e.g., an increase by at . least about 2, 3, 4, 5, 6, or more. In another aspect, the subject or population of subjects treated achieves an 20 improvement in a Short Form 36 Health Survey Mental Component Summary (MCS) score or mean Mental Component Summary (MCS) score of at least about 3.5, 4, 4.5, 6, 6.5, 7, or more. An improvement in PCS is an increase in MCS score, e.g., an increase by at least about 3.5, 4, 4.5, 6, 6.5, 7, or more. In another aspect, the subject or population of subjects treated achieves an 25 improvement in a visual analog scale score or a mean visual analog scale score for psoriasis-related pain (VAS-Ps) of at least about -25, -26, -27, -28, -29, -30, -31, -32, 33, -34, -35, -40, -45, -50, or less. An improvement in VAS-Ps is a reduction in VAS-Ps score, e.g., a reduction by at least about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 40, 45, 50, or more. 30 In another aspect, the subject or population of subjects treated achieves an imprbvement in a visual analog scale score for psoriatic arthritis-related pain (VAS-PsA) or a mean visual analog scale score for psoriatic arthritis-related pain (VAS-PsA) of at least about -25, -26, -27, -28, -29, -30, -31, -32, -33, -34, -35, -40, -45, -50, or less. An 128 improvement in VAS-PsA is a reduction in VAS-Ps score, e.g., a reduction by at least about 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 40, 45, 50, or more. In another aspect, the population of subjects treated achieves a minimum clinically important difference (MCID) response rate in any one or more HRQOL 5 outcomes including, e.g., DLQI, TAI, VAS-Ps, Vas-PsA, MCS and PCS of at least about 60%, 65%, 70%, 75%, 80%,-85%, 90%. In another aspect, the population of subjects treated achieves a minimum clinically important difference (MCID) response rate for psoriasis-related pain (VAS-Ps) of at least about 60%, 65%, 70%, 75%, or more, e.g., by about week 12 or by about 10 week 52. In another aspect, the population of subjects treated achieves a minimum clinically important difference (MCID) response rate for Dermatology Life Quality Index (DLQI) of at least about 70%, 75%, 80% or more by about week 12. In another aspect, the population of subjects treated achieves a minimum 15 clinically important difference (MCID) response rate for Dermatology Life Quality Index (DLQI) of at least about 75%, 80%, 85%, 90%, or more by about week 52. In another aspect, the population of subjects treated achieves a minimum clinically important difference (MCID) response rate for Total Activity Impairment (TAI) of at least about 45%, 50%, 55%, 60%, 70%, or more by about week 12. 20 In another aspect, the population of subjects treated achieves a minimum clinically important difference (MCID) response rate for Total Activity Impairment (TAI) of at least about 50%, 55%, 57%, 60%, 65% or more by about week 52. In another aspect, efficacy may be assessed by Nail Psoriasis Severity Index (NAPSI) scores, which range from 0 (no nail psoriasis) to 80 (psoriasis in all 10 25 fingernails). In certain embodiments, the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 40, 35, 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1 or less. In certain embodiments, the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 2.1 or less. In certain embodiments, the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 2.1 or less by about week 24. In certain 30 embodiments, the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 1.2 or less. In certain embodiments, the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 1.2 or less by about week 52.
129 In another aspect at least 40%, 45%, 50%, 55%, 60%, 65%, or more of the population of subjects treated achieve at least a PGA 0/1 response by about week 12, wherein each subject was treated with a biologic prior to administration of the antibody. In another aspect, at least 50%, 55%, 60%, 65%, 70%, 75% of the population of 5 subjects treated achieve at least a PASI 75 response by about week 12, wherein each subject was treated with a biologic prior to administration of the antibody. In another aspect, at least 60%, 65%, 70%, 75%, 78%, or more of the population of subjects treated achieve at least a PGA 0/1 response by about week 12, wherein none of the subjects were treated with a biologic prior to administration of the antibody. 10 In another aspect, at least 60%, 65%, 70%, 75%, 80%, 82% or more of the population of subjects achieve at least a PASI 75 response by about week 12, wherein none of the subjects were treated with a biologic prior to administration of the antibody. In another aspect, at least 60%, 65%, 70%, 75%, 78%, or more of the population of subjects -treated achieve at least a PGA 0/1 response by about week 52, wherein each 15 subject was treated with a biologic prior to administration of the antibody. In another aspect at least 60%, 65%, 70%, 75%, 79%, 80%, 82% or more of the population of subjects treated achieve at least a PGA 0/1 response by about week 52, wherein none of the subjects were treated with a biologic prior to administration of the antibody. 20 In another aspect, at least 50%, 55%, 60%, 65%, 70%, 71%, or more of the population of subjects treated achieve at least a PGA 0/1 -response by about week 12, wherein each subject treated has a prior history of psoriatic arthritis. In another aspect, at least 60%, 65%, 70%, 75%, 78%, or more of the population of subjects treated achieve at least a PASI 75 response by about week 12, wherein each 25 subject treated has a prior history of psoriatic arthritis. In another aspect, at least 60%, 65%, 70%, 75%, 77%, or more of the population of subjects treated achieve at least a PGA 0/1 response by about week 12, wherein none of the subjects treated has a prior history of psoriatic arthritis. In another aspect, at least 60%, 65%, 70%, 75%, 81%, or more of the population 30 of subjects treated achieve at least a PASI 75 response by about week 12, wherein none of the subjects treated has a prior history of psoriatic arthritis.
130 In another aspect, at least 60%, 65%, 70%, 75%, 77%, or more of the population of subjects treated achieve at least a PGA 0/1 response by about week 52, wherein each subject treated has a prior history of psoriatic arthritis. In another aspect, at least 60%, 65%, 70%, 75%, 79%, or more of the population 5 of subjects treated achieve at least a PGA 0/1 response by about week 52, wherein none of the subjects treated has a prior history of psoriatic arthritis. In another aspect, at least 50%, 55%, 60%, 65%, 69%, or more of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline PASI greater than 20 prior to administration of the antibody. 10 In another aspect, at least 60%, 65%, 70%, 75%, 79%, or more of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline PASI less than or equal to 20 prior to administration of the antibody. In another aspect, at least 60%, 65%, 70%, 75%, 79%, or more of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject 15 had a baseline PASI greater than 20 prior to administration of the antibody. In another aspect, at least 600b, 65%, 70%, 75%, 80%, 81%, or more of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline PASI less than or equal to 20 prior to administration of the antibody. 20 In another aspect, at least 50%, 55%, 60%, 65%, 67%, or more of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline weight of greater than or equal to 100 kilograms prior to administration of the antibody. In another aspect, at least 60%, 65%, 70%, 75%, 80%, or more of the 25 population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline weight of less than 100 kilograms prior to administration of the antibody. In another aspect, at least 50%, 55%, 60%, 65%, 70%, 72% or more of the population of subjects achieve at least a PASI 75 response by about week 12, wherein 30 each subject had a baseline weight of greater than or equal to 100 kilograms prior to administration of the antibody. In another aspect, at least 60%, 65%, 70%, 75%, 80%, 85%, or more of the population of subjects achieve at least a PASI 75 response by about week 12, wherein 131 each subject had a baseline weight of less than 100 kilograms prior to administration of the antibody. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, 5 specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition. 10 VII. Uses of the Invention The invention provides methods for inhibiting IL-12 activity in a subject suffering from a disorder in which IL-12 activity is detrimental. IL- 12 has been implicated in the pathophysiology of a wide variety of disorders 15 (Windhagen et al., (1995) J. Exp. Med. 182: 1985-1996; Morita et al. (1998) Arthritis and Rheumatism. 41: 306-314; Bucht et al., (1996) Clin. Exp. Immunol. 103: 347-367; Fais et al. (1994) J. Interferon Res. 14:235-238; Parronchi et al., (1997) Am. J. Path. 150:823-832; Monteleone et al., (1997) Gastroenterology. 112:1169-1178, and Berrebi et al., (1998) Am. J. Path 152:667-672; Parronchi et al (1997) Am. J. Path. 150:823 20 832). The invention provides methods for inhibiting IL-12 activity in a subject suffering from such a disorder, which method comprises administering to the subject an antibody or antibody portion of the invention such that IL- 12 activity in the subject is inhibited. Preferably, the IL-12 is human IL-12 and the subject is a human subject. Alternatively, the subject can be a mammal expressing a IL- 12 with which an antibody of the invention 25 cross-reacts. Still further the subject can be a mammal into which has been introduced hIL-12 (e.g., by administration of hIL-12 or by expression of an hIL-12 transgene). An antibody of the invention can be administered to a human subject for therapeutic purposes (discussed further below). Moreover, an antibody of the invention can be administered to a non-human mammal expressing a IL-12 with which the antibody 30 cross-reacts for veterinary purposes or as an animal model of human disease. Regarding the latter, such animal models may be useful for evaluating the therapeutic efficacy of antibodies of the invention (e.g., testing of dosages and time courses of administration). As used herein, the phrase "a disorder in which IL-12 activity is detrimental" is 132 intended to include diseases and other disorders in which the presence of IL-12 in a subject suffering from the disorder has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to a worsening of the disorder. Accordingly, a disorder in which IL-12 activity is 5 detrimental is a disorder in which inhibition of IL-12 activity is expected to alleviate the symptoms and/or progression of the disorder. Such disorders may be evidenced, for example, by an increase in the concentration of IL-12 in a biological fluid of a subject suffering from the disorder (e.g., an increase in the concentration of IL-12 in serum, plasma, synovial fluid, etc. of the subject), which can be detected, for example, using an 10 anti-IL-12 antibody as described above. There are numerous examples of disorders in which IL-12 activity is detrimental. In one embodiment, the antibodies or antigen binding portions thereof, can be used in therapy to treat the diseases or disorders described herein. In another embodiment, the antibodies or antigen binding portions thereof, can be used for the manufacture of a medicine for treating the diseases or 15 disorders described herein. The use of the antibodies and antibody portions of the invention in the treatment of a few non-limiting specific disorders is discussed further below: A. Rheumatoid Arthritis: 20 Interleukin-12 has been implicated in playing a role in inflammatory diseases such as rheumatoid arthritis. Inducible IL-12p40 message has been detected in synovia from rheumatoid arthritis patients and IL-12 has been shown to be present in the synovial fluids'from patients with rheumatoid arthritis (see e.g., Morita et al., (1998) Arthritis and Rheumatism 41: 306-314). IL-12 positive cells have been found to be 25 present in the sublining layer of the rheumatoid arthritis synovium. The human antibodies, and antibody portions of the invention can be used to treat, for example, rheumatoid arthritis, juvenile rheumatoid arthritis, Lyme arthritis, rheumatoid spondylitis, osteoarthritis and gouty arthritis. Typically, the antibody, or antibody portion, is administered systemically, although for certain disorders, local administration 30 of the antibody or antibody portion may be beneficial. An antibody, or antibody portion, of the invention also can be administered with one or more additional therapeutic agents useful in the treatment of autoimmune diseases.
133 In the collagen induced arthritis (CIA) murine model for rheumatoid arthritis, treatment of mice with an anti-IL-12 mAb (rat anti-mouse 1L-12 monoclonal antibody, C 17.15) prior to arthritis profoundly suppressed the onset, and reduced the incidence and severity of disease. Treatment with the anti-IL- 12 mAb early after onset of arthritis 5 reduced severity, but later treatment of the mice with the anti-IL- 12 mAb after the onset of disease had minimal effect on disease severity. B. Crohn's Disease Interleukin-12 also plays a role in the inflammatory bowel disease, Crohn's 10 disease. Increased expression of IFN-y and IL- 12 occurs in the intestinal mucosa of patients with Crohn's disease (see e.g., Fais et al., (1994) J. Interferon Res. 14: 235-238; Parronchi et al., (1997) Amer. J. Pathol. 150: 823-832; Monteleone et al., (1997) Gastroenterology 112: 1169-1178; Berrebi et al., (1998) Amer. J. Pathol. 152: 667-672). Anti-IL-12 antibodies have been shown to suppress disease in mouse models of colitis, 15 e.g., TNBS induced colitis 1L-2 knockout mice, and recently in IL-10 knock-out mice. Accordingly, the antibodies, and antibody portions, of the invention, can be used in the treatment of inflammatory bowel diseases. C. Multiple Sclerosis 20 Interleukin-12 has been implicated as a key mediator of multiple sclerosis. Expression of the inducible IL-12 p40 message or IL-12 itself can be demonstrated in lesions of patients with multiple sclerosis (Windhagen et al., (1995) J. Exp. Med. 182: 1985-1996, Drulovic et al., (1997) J. Neurol. Sci. 147: 145-150). Chronic progressive patients with multiple sclerosis have elevated circulating levels of IL-12. Investigations 25 with T-cells and antigen presenting cells (APCs) from patients with multiple sclerosis revealed a self-perpetuating series of immune interactions as the basis of progressive multiple sclerosis leading to a Thl-type immune response. Increased secretion of IFN-y from the T cells led to increased IL-12 production by APCs, which perpetuated the cycle leading to a chronic state of a Thl-type immune activation and disease (Balashov et al., 30 (1997) Proc. Natl. Acad. Sci. 94: 599-603). The role of IL-12 in multiple sclerosis has been investigated using mouse and rat experimental allergic encephalomyelitis (EAE) models of multiple sclerosis. In a relapsing-remitting EAE model of multiple sclerosis in mice, pretreatment with anti-IL-12 mAb delayed paralysis and reduced clinical scores.
134 Treatment with anti-IL-12 mAb at the peak of paralysis or during the subsequent remission period reduced clinical scores. Accordingly, the antibodies or antigen binding portions thereof of the invention may serve to alleviate symptoms associated with multiple sclerosis in humans. 5 D. Insulin-Dependent Diabetes Mellitus Interleukin-12 has been implicated as an important mediator of insulin-dependent diabetes mellitus (IDDM). IDDM was induced in NOD mice by administration of IL 12, and anti-IL-12 antibodies were protective in an adoptive transfer model of IDDM. 10- Early onset IDDM patients often experience a so-called "honeymoon period" during which some residual islet cell function is maintained. These residual islet cells produce insulin and regulate blood glucose levels better than administered insulin. Treatment of these early onset patients with an anti-IL-12 antibody may prevent further. destruction of islet cells, thereby maintaining an endogenous source of insulin.
15 E. Psoriasis Interleukin-12 (IL-12) and the related cytokine IL-23 have been implicated as key mediators in psoriasis. Psoriasis involves acute and chronic skin lesions that are associated with a THl-type cytokine expression profile (Hamid et al. (1996) J. Allergy 20 Clin. Immunol. 1:225-231; Turka et al. (1995) Mol. Med. 1:690-699). Both IL-12 and IL-23 contribute to the development of the type IT helper cell (Th 1) immune response in psoriasis. Moreover, the IL-12 p40 and IL-23 p40.messenger RNA is overexpressed in psoriatic skin lesions. Accordingly, the antibodies or antigen binding portions thereof of the invention may serve to alleviate chronic skin disorders such psoriasis. 25 In one embodiment, the invention provides a method for treating psoriasis. Treatment for psoriasis often includes a topical corticosteroids, vitamin D analogs, and topical or oral retinoids, or combinations thereof. In one embodiment, an IL- 12 and/or IL-23 antibody is administered in combination with or the presence of one of these common treatments. Additional therapeutic agents which can be combined with the IL 30 12 and/or IL-23 antibody for treatment of psoriasis are described in more detail below. The diagnosis of psoriasis is usually based on the appearance of the skin. Additionally a skin biopsy, or scraping and culture of skin patches may be needed to rule 135 out other skin disorders. An x-ray may be used to check for psoriatic arthritis if joint pain is present and persistent. Improvements in psoriasis in a subject can be monitored by the subject's Psoriasis Area and Severity Index Score (PASI). The method for determining the PASI 5 has been described in Fredriksson and Pettersson (1978) Dermatologica 157:238 and Marks et al. (1989) Arch Dermatol 125:235. Briefly, the index is based on evaluation of four anatomic sites, including the head, upper extremities, trunk, and lower extremities, for erythema, induration, and desquamation using a 5 point scale (0= no symptoms; 1 =slight; 2= moderate; 3=marked; 4=very marked). Based on the extent of lesions in a 10 given anatomic site, the area affected is assigned a numerical value (0=0; 1 = < 10%; 2= 10-29%; 3 = 30-49%; 4 = 50-69%; 5 = 70=89%; 6 = 90-100%). The PASI score is then calculated, wherein the possible range of PASI score is 0.0 to 72.0 with the highest score representing complete erythroderma of the severest degree. In one embodiment of the invention, an IL- 12 and/or IL-23 antibody is used for 15 the treatment of psoriasis, including plaque psoriasis, e.g., chronic plaque psoriasis, moderate plaque psoriasis, and severe plaque psoriasis, guttate psoriasis, inverse psoriasis, pustular psoriasis, pemphigus vulgaris, erythrodermic psoriasis, psoriasis associated with inflammatory bowel disease (IBD), and psoriasis associated with rheumatoid arthritis (RA). In another embodiment, an 1L-12 and/or IL-23 antibody, 20 such as J695 / ABT-874, is used to treat subjects who have psoriasis in combination with PsA. In one embodiment of the invention, an IL- 12 and/or IL-23 antibody is used for the treatment of nail psoriasis. In one aspect, the invention provides methods for treating psoriasis in difficult to treat subjects by administering antibodies, and antigen binding portions thereof, of the 25 invention, for example, ABT-874. Difficult to treat subjects may include, for example, subjects who have been previously administered biologics for the treatment of psoriasis, subjects who have had a history of psoriatic arthritis, subjects who have psoriasis and weigh greater than 100 kg, and subjects who have a baseline PASI greater than 20. Accordingly, in one aspect, the invention provides methods for treating subjects who 30 have been previously administered biologics for the treatment of psoriasis by administering antibodies, and antigen binding portions thereof, of the invention, for example, ABT-874. Specifically, the methods involve selecting subjects who have received prior biologic treatment and administering antibodies of the invention. As set 136 forth in Example 19, the data demonstrates efficacy of ABT-874 in the treatment of psoriasis in this subgroup of subjects. In another aspect, the invention provides methods for treating subjects who have had a history of psoriatic arthritis by administering antibodies, and antigen binding portions thereof, of the invention, for example, ABT 5 874. Specifically, the methods involve selecting subjects who have had a history of psoriatic arthritis and administering antibodies of the invention. In another aspect, the invention provides methods for treating subjects who weigh greater than 100 kg by administering antibodies, and antigen binding portions thereof, of the invention, for example, ABT-874. Specifically, the methods involve selecting subjects who weigh 10 greater than 100 kg and administering antibodies of the invention. In yet another aspect, the invention provides methods for treating subjects who had a baseline PASI greater than 20 by administering antibodies, and antigen binding portions thereof, of the invention, for example, ABT-874. Specifically, the methods involve selecting subjects who have a baseline PASI greater than 20 prior to administration of the antibody and 15 administering antibodies of the invention. Specific types of psoriasis included in the treatment methods of the invention are described in detail below: 20 a. Chronic plaque psoriasis Chronic plaque psoriasis (also referred to as psoriasis vulgaris) is the most common form of psoriasis. Chronic plaque psoriasis is characterized by raised reddened patches of skin, ranging from coin-sized to much larger. In chronic plaque psoriasis, the plaques may be single or multiple, they may vary in size from a few millimeters to 25 several centimeters. The plaques are usually red with a scaly surface, and reflect light when gently scratched, creating a "silvery" effect. Lesions (which are often symmetrical) from chronic plaque psoriasis occur all over body, but with predilection for extensor surfaces, including the knees, elbows, lumbosacral regions, scalp, and nails. Occasionally chronic plaque psoriasis can occur on the penis, vulva and flexures, but 30 scaling is usually absent. Diagnosis of patients with chronic plaque psoriasis is usually based on the clinical features described above. In particular, the distribution, color and typical silvery scaling of the lesion in chronic plaque psoriasis are characteristic of chronic plaque psoriasis.
137 b. Guttate psoriasis Guttate psoriasis refers to a form of psoriasis with characteristic water drop shaped scaly plaques. Flares of guttate psoriasis generally follow an infection, most notably a streptococcal throat infection. Diagnosis of guttate psoriasis is usually based 5 on the appearance of the skin, and the fact that there is often a history of recent sore throat. c. Inverse psoriasis Inverse psoriasis is a form of psoriasis in which the patient has smooth, usually moist areas of skin that are red and inflammed, which is unlike the scaling associated 10 with plaque psoriasis. Inverse psoriasis is also referred to as intertiginous psoriasis or flexural psoriasis. Inverse psoriasis occurs mostly in the armpits, groin, under the breasts and in other skin folds around the genitals and buttocks, and, as a result of the locations of presentation, rubbing and sweating can irriate the affected areas. d. Pustular psoriasis 15 Pustular psoriasis, also referred to as palmar plantar psoriasis, is a form of psoriasis that causes pus-filled blisters that vary in size and location, but often occur on the hands and feet. The blisters may be localized, or spread over large areas of the body. Pustular psoriasis can be both tender and painful, can cause fevers. e. Other psoriasis disorders 20 Other examples of psoriatic disorders which can be treated with the IL- 12 and/or IL-23 antibody include erythrodermic psoriasis, vulgaris, psoriasis associated with IBD, and psoriasis associated with arthritis, including rheumatoid arthritis. The present invention is further illustrated by the following examples which 25 should not be construed as limiting in any way. The contents of all cited references, including literature references, issued patents, and published patent applications, as cited throughout this application are hereby expressly incorporated herein by reference. It should further be understood that the contents of all the tables attached hereto (see Appendix A attached hereto and Appendix A of US Patent No. 6,914,128) as well as the 30 entire contents of U.S.. Patent No. 6,914,128 are incorporated herein by reference.
138 Coo..U -s5-s5S00eo - Ododode 0 t 00000000000000000000 C-C - -C C -C C -C-C -C-C C C -C - V w -C C -C -C C -C C C -C -C C C -C 0 00000 00000w 0 0 0 0 0 w I Cww w wo 0 w 0 0 0 w A 0 . 0 0 L0 C-C -C CC -CC -C-C tCCC noo a- a- a- a' C- C- C- C - - 0000 00000000-00 a0000000000 00 C, 0.000C oC oCoCOC o o00 C '0' 00 000 '0 '00 '000'0 00'050'0 05050 '0' '05050509505 '05C, 05 '" 'M000'00 toS .0 '0 -0 v) v) v) v) v) v) 0 v) v) v) v v) v) v) v vS v) .)00C 0 0 v v) v Cv 0 0CS S 5 0C) 000 CoCoCo00 ao ao ao 0000 aoC oC o 0C S00 0 0 - 0 0 0 0 0 0 w w w w w Co Cot oC oC oC oC oC oC oC C oC oC oC oC oC oC oC o Co wo C C D Co D toC o C. Ca 41E o Oo oo o 00 n0 0 ( 0 00 000 (5 n n C ( 0(50 'SC' ( 0 (S( 0 00 0 o n 00 (5 5 l 0000 000 000 0000 000 00000000 0 0 0 0 0 00 S' -n - -n -n ''1 n-9 m.'1S'n'm m M9S' M." m""S m' -9 "1 -n -9 -s5SmSm9N"S"S9 mS"S sn -n*' -n5" '19n 2" "2" "29999999 m~ 99 " 99 9 9 "2 999 mS "2m N '--N USCUSACASUUSUCASCAAUUSCUSAUSSUCAUCASUSAUCACCASUSSCUSUCOAOOUS5USAU S USACUUSSCUUSAUUUSASUCAUSAUCUSSZZZZC~oo o00OOO"00S 0 .(..<..(4'4<.4(..4..('.44.44.44. . . . . . . . . ~ ooo S'. .%z 4zz . . " . . . .. p pC X~~~~oooo~~~~ooooZooo~~~~o~Zo~.C C C 'roC X-~~oooo~~~~ Ci I, % w m x oM 1ZZ1Z XZA X ooCAZ X o 0oZA %A ZCAUSCACO0 0ow=ACA 0moUS 101'4sCA COSIs g1 g 11 IC 0g Ig g g ggg 'T g g 1- le C < Ce < < < C4 . < < < C C< C <. e < < < Co < p < < < < < < < < 5- '- < < < < C C C w " w w w w w w " w w :0 w CY :0 " w w %, w w w w w ;~o~oo~o~ C CY~~~~~~~~~~5 w. w vw"ww< 0I 09000,0000 00 0 0 100 0 00 00 0 1 so 00 0 0 00s0000 00004000 5 2 C-uo a '0~~~~~~~~~~~~~~ .4 'Q 00 '0 '0 'Q '0 '0 'a C, 'a 'a' Q00' 'a a00' '00'.5 000''' 0' 0' '' 0 '05 (P tv 000000000000,000000 a0coca00 00 0000'5 rCCC-' v-'C "-C -r . v t, t C" v -v'r-CCICCCCC vCv-C-"tCt-C-vC-Cvv --- Cv- " -- Ct-C--V S5-' V-' Co C w 0 Co Co-- W 'A CA 'A 0 0 C C oC oC oC o C 0 0 W- -SoC C oC oC ' A C- 0 l0 C- C-C Co.' 0 0. < < <- < <ssss- - sssss--ss - - ssss - - ssss<- - C < C U <.- a CSSS5- - - - 04* 55 5 -S' S .5 . . . . . .. C . . . .USUSSUSUSUS gg ~ ~ ~'IT (I-' t -i USUSSUCAAVUSSCCAAUCOAUCAAU0S WCACA'A CA C o No N NoUSV. w - moo oll;. US'S' ~ ~ ~ ~ ~ ~ ~ ~ x xa~ H4S A S((-S..(.((-.O S O O O .4E o04-- - -4 ", . .. : - Co;oo .oIs m-.0 S aS z z 'A .40 :0 = zz C C3 a . .= 0GA aOCO. .. ..44CS 0 cocoa.4 ' .4 4 4 ~ . ao o Z o0Co0Co o..00 0 -C'SC' CA "S 0C pi 05 w4-' *w( . a.4 )4 V3.V3'En4En.En4.4.4 a4a4.4 a4 - . a.a. CA4.4.4 o4*4 a4o4a4 .4. a .4 to.4. a.4 )4a.C.4.4 <4 < '0u0. < CC CC C CC CC C CC CC C < -4 C -4 - - - < . C C -C 0. 44 0a OO O ) OO O O OO O O O )0 0C00 0 0 0 000000000 5 / 000 0 -- C <oo o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o o C ~ o o o o < .0C 0 0 W USA A SC C CA A USSCA CACACV3W W 3 CA 0 3wC1 A 31U U CA W 1C)rU) CA COA(1) SUSW AUSUSUSACP CO CACAUSIA 0 z 0 Co C o Co o Co o Co Co C Co Co Co Co Co Co Co Co Co Co Co Co Co Co 0 0 Q0 00 Co Co Co0t C Co 00goC Co Co CA CA CA US CA CA US M US X CA US US CA CA X CA US US Co Co Co Co X CA CA Co X M US X M X US M Co Co X Co Co CA X M M S .4~~~~~~~~~~~~~~~~ X4. 4. 4. 4. 4. 4. 4. 4 4. 4. 4. 4. 4. 4. 4. .4 .4 4, .l 4 .1 " 4 1., .4 .4 "4. .4 .4 " 4 v 4 " 4 Oon0o0n nn o o n nn n n o 00000 000 0 0 0 0n0nnn0nnn0 0000 0 00 0 0 0nn0' Cw Co1 C, 3Co Co )-I <o C1 ;1 Co1 Co .o Co :$1 Co Col p Co Co Co Co Co Co Co Co Co < 4 Co Co Co Co 3Co po ).I ;I Co Co CoC oC o o 1C Co o C CoCo o C CoCo o C CoCo S.C CoCo CACA o C CoCo o C CoCo o C CoCo o C CoCoNo:C CoCo o wC wSCo 0A CW o 139 00.w a 00 00 00 0 a 0000000000 N 00000000000 10 (0 (0 (0 0 go oA oA X0 m X0 go o0 o. o. 0 (0 (0 ((A0 0 (o0 o oA 0 ( .1 0 0 n00000: 400000000000000000, (I ca WA (oW A CA ( (A W to W0 WA (0 W W 0 W 0 (A t CA (A 0 . to 0 (A 1 (.1 :4 nn "n on o n o .0 nn 0 :4 o 0 Oct (0 (A w. ( W (A W (0 (A W0 W 0 ( W (A ( C(0 .A (A CW A to 0 (0 w . (A W A to W W 9 ( 00 0000 0 0000 a 0 00000000 o 0. m tl - "I,, m444::: m4 "I4: "1 :4: -4444 4 -1 m I'll :4 4 4 4 : :., :4:4:4:4 4 4 4 : : : :4:4:4:4 4 4 4 4 .
. W W 0(A( 0(0 . 0(0 . 0( (.0 (0WW W W (0 W O (A Co . (A W W W. A( 0C . 10 AD 2 C2 C2 a w A (A (A (.0 (0 b Gb Gb s 0 0 Gb Gb Gb 10b 0b 10 10 0b GGb0' 000 0 0000 000 0 0 0 0000000 0 0 6Caa t 0 0.9) a Q a a a 0 a, a an a 00 a0 0 a a 0 a aaa o a" L.Vt- .t - rrrt.t , ,t t ,tvvv l . l . .S tq ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ 6 -c"<- nt 3-' I "tIMP t . qt 3L x :VxXxxx.I ,3 < ~ ~ ~ ~ ~ ~ ~ ~~ n .4<<- 4. c 4Io1,19C oI 411 4-e e C1,4 : to ) W Wto W W W W 'aN, w 3p,)I-, )-I 1.,0 (PG * - 0 -9 Mz w.4 I .4-c - .4.4 . 0-. w 222 (040 Z ;q Z : WW0( r, WW .x V M W V (A E AW. W A(06( aaoaa.40(ow A WA(Aa a aa Ula cla Qa a 0 tWWWW'A wwWtwU)WtWto W W W ((A0( aaa~a A Wa Waaao W1 0 li 0 XA(( (A PCO a (A A (A (A (A (A (A (A ( 0A (A ( XA (A (( (A (.C( N . A PC Ls m 0 -< (0 CO W.0 (A <6 < <4 -< . <4 - <2 <2 <2 <2 3' 3< <2 -< #9 32 P4 Z OF 14 :4.4 .< -) c K c O < - -<-c < < - -<-< 4 L k 10t 3< ;4. 4. . 43 4 4 < .4 .4 .4 4 .4 .4 .4 .4 .4 . <4 .4 .4 .4 .4 .46 -. t A C a~ ~ ~~~~. Q I Q0a09E :4:44324:4~:4::4:44 ~ m Ha a g a a a a aa a a a a a b - - -------- W' a a a a4 :4:V a4 a4 :4 4 4:0a4 :4a4 :4 a4 :4 : :4 a16 .baa a 46 P.6 to :P W WO6 V V W W W En W WW (A WO16rC W 9 m mA(X(X " N(A(Aw0m0(A(0(A(:qAw(A( R(pt0( K(:nA:n(;RA:R ;R(;ft9 :4 4:44 : :4:4 4 ::4 4:4:4::4 4*::4 0 f z 0' ( O~aa aaa aav t-9 v t- aaoa at-at-(t 'I (A -1 (0 .1 .4 A( 0( ( . 4 . 4 -1 . -1 -1 -14 1 4 -1- .4 14 1 W.4 C 323232z 2 )1,2 3-1 w z 31. 32 3 3-1 3 2 3. 321 3. 303 3-3 D33232p2r to. C. 0 3.' ), C ' o I' )'1' 3.1 ), )., 1' ), :' )' ), ), C' )I' ), p' )I' )I' w'1 'C wwwwwwww I (6~www- 1-40 00% 0 G 0%1 0 X~ - -0 -. 0% -0% C4 to 9 op 0% tow. ,8- 000 0CC) 00' U CO 10 4I 5 1I ON. 14 -1 ' -4 FA to 4 to n f CD o C03 FA CCI Co CA It C A CA3 4 CO CA CA fAVL 01 C> 14 . -4 - P -I Is.41 =C <) z) A A CK)4 Li) C.~~1COZC C .0-. -3 1 0 0 .0 )0 '0 p .0 30 0 t -0 00 fu 0 0 G C0 0 aO QO cl < PO to COM ~ 0 9' Is v irt C a ; UP a w a 41 0 t C t 0. a 0)C)C)CC)QC) 0 t cOcC W 'A ACA 'A to q: 0 CA 0CIAC~1 N 4. -4CO)V 4 lO E4 C4 O ( O 0 30 0 :0 0 Q 0 L' 00) cl 9C C) C) L 101 ..gu x to. 10 VI0 C9.0 ))~~ 1 141 L 0C4 m, 0 01 (" Q atC a > C : .i L" 0.c 0cot ~ W I..J a L~ La -N N) -i - - I-j -j -i- J 10 (/0 Q/ U' 0' 10U ' 00 0 0 00 0 G)0C0C) US ~ ~ ~ ~ ~ ~ ~ ~ ~ ~ 0 cc,: J .(16I.I-L a~I' a~. / ) ) :-00 0 00 Z U ID O C 0000 O C 00ID 0 (D M o C'
------------------------------------
tn~''0UU tot ( o n ACAWcUt WC '0 ~ . La 0~o~ 00 1 0 0 500L %wO ," DMp "( o( . 0 00000 00 00 L 0 .1""a g ?ll I I w I 00 -') C) 0 a -0 M 00 0 0n L 0 00CD0 0 00C 0o 0) 0UU 0 0 of 0 0 0 0 0 0- n . 0 .40C M VI) Mt1lI U'l ('I Vj M I0' U' I 1 1 I I I gI M En M I . 1 00000 00 0 01110 000m' %D og D -J CD COU' - j -J - .. I- -J -J O 0-4 02 Ln
-------------
142 Ln V, W (g k, w. w - - - age O~a~o~~ ~ W~D~D wO 'o . . . . . . . . . . . -n z (A En . 0 '-3. UIA o0 0 0 0 C. . C> C) CD, 4.) N)i )W W cp 0 (Dx CD L o xp 00 s- 0 0~ 00 1- 11 1 1- , 0 0 .00 0l, 151 51 111'251L' 143 -i -j j -j -j - -j j _j t W JN N N)N ~~ i. . o ~ .Ot~U '0 no -0 ' a c. r- 0 to o~ w, U a .- ) t. U, x x- 0) x x 0~ tF XXl mX 0 0 00mm0m 0000000 0 0 00 0 0 C> a a .b I I ?I C00 00 0Ol ~ 0 C) : C0 C0 0)C)0 00 0 C>C>c to l Ij "I III m M C)iw c0 0 - - - - - - - - - - - - - - 144 j i j j - - -1-1li-J -1 -J Ji -j A co 1 - -1 -4 1 - -4 -4 CA Wo MW EA CO) Cw w l w~ Cl w o o w o w ) U) o Coh to Wl (A 8o ) t 0% W' M~o L* w* Q0DW_ c w . o . . tq ? :Co <n. .3 Coto C0
I-.,--,-
-~~ - - - (i 145 0 t wD -j aw L" wD ta C w w i I. m -t w w !? U in o Z) ? vi a~ in C:l !D f- U) n in L) W) W) I ) U ) U n U) U ) U )U ) U .~ ~ .o f,! ' Z Z ZZ Z Z ZZZD -j ZZZ w co- i mc o . t .C co m -j - w I- w -- , Ln -- m L- I- co-j w to' (I a a a In a11 MC)4=1Q 'D 0 aC C Op0 4-3.1 W'-3.)t ). > l )tow wt ~ 0 4-n 400 -1- 1 4 4-1 4 146 w Ln w 0%co wm 1 0 En C)~ ~ Ql C) 0) Q) C) G)C ) ) ) C) C) G)C )C )C ) 0C C) Q) G) Q) Q) Q) wf U1) to(n (4) Ea C4) 0l ) En En) En) (n) (A) co) U) Cl) 0l Cl n ) l cII to) C ) cd) Wl En) (l) C/ C) ZZZZZZZZZZmZM z zzzzzzz Li %D,, M ) N) N) tp :ft w ) W) cc 4 - j at~ w- 4- w C>. w- m- I m L 00 0 w 0 0 ko N) E -1 1 O. En En '0 0 . o V ( l 2'U 10C/ W 4 ~ ~ L Vi w. 0% W4 md wd -dl UC (dl d. Hi wi ri LiL W LiN)N P3 w) N j -3 (3 4~ -4 W w m m pi~ t. -' 04- wLi vj W4'0 W4.WJ. M.Ji( 0 00 0000 ;0 0 00 00 0 0.0 0a 0 0 0 00 0 00 0 LiLWiL i iL iL LO wi L wi UP w Li wiL L L wLiL Li Li Li Li Li L L i (Ln V A P
L<
1-47 06) 0 1<0 W,. w 7.( LI' 0' tn C'.i 'Dn 00 w~~ w c ~ lcn c n w nE , 'to-I- P. IO0 3r tC. C4 Hnc t" v z c zzn 0n 0 00 _3 c l ii -j1 c Iw % IL M0 0n N 0 0 0 0 0 0 0 0 0 0 0o a, C> C3 I3 C3 0 t w I 0 0..JIDI 000000 00 000 00 0~ 0'.0 0 cm, t " -11 0 1 0 1 r, C> 00 0 000; 0 a'. "c C. 01 0 0 01w0P 0 m-------------m-----I------------------------- 148 U)WWU W W.L. U)Ww QN -JWA~ CD W ( Ono 0~ ~~0 N ' Ic i1I 0 0I 0 CA.n~ w Z n- o- ow( c; o CAD 0 nWc 'pR :E WW W 3L ww m w WJ DWL.WW ~ L, uz 0 C a0 a0 V300 a 0 0 ~ 0 00 .o . 0 0 0t b- coa L 0~0 ~ ~ Cfl 00o (3 ~L I ~1~U1 .~U~u ' ~ ~ 0 149 C-, ID co 0D %0w oto a..i co 0 0 0 0 00 10 lb D 1 z Cf 00 w) U) I000I 0j al m z tnZ*~ 01 0 0 so 0 10 00 QPC0K ) to 0) to000A0C0( -3 -3 -3 6-3- 3-3 -3 03 I I I I (' n (n In b 0 000 U) 0 D >C 00003 tI tn ti I-.- IV0' 0 0 a ?0 00 O 0 0> 03 0 0 0000a tnl Ali tn M' tnl mlCV1m U> OD N U )N 00 0D 0 0 0 0 0 0 00 150 c0 rt 01 rt rt 0 0 0 0 c t At :3 ft O 0 0 t 0, f (A CA 0 0 E ct ct C% ctrt 0 0 0 0 LZ r r rr rt eTz a CL 0 0 rt ri T 068 O9 00 w0 x 6 o1 I- S 00 x to 96 tx Ix Oe _ _ _ _ * - L -- - O -4 9-6 151 < t-. *L ~ ~ ~ k %D ID Ch PI - W L' LI I I a) z rt 00 'Ch n0O .
fu t 0C L<'L~ VI C" 0l C o 0 0 10 LII 0o C0 0) 10 0 > C 0p 0C 0 L.< Cl C"C ~ 40 0 0I If C> 40 %D L 40 152 EXAMPLES Example 1: Efficacy of the Fully Human IL-12/IL-23 Monoclonal Antibody, ABT 874, In the Treatment of Moderate to Severe Plaque Psoriasis 5 ABT-874 is a fully human antibody against interleukin-12 (IL-12) and IL-23. It binds with great affinity to the p40 subunit common to both IL-12 and IL-23, both validated targets in the treatment of psoriasis (Ps). The objective of the following study was to evaluate the efficacy of subcutaneous injections of ABT-874 in the treatment of patients with moderate to severe 10 plaque Ps. Adult patients with Ps affecting >10% body surface area (BSA) and a Psoriasis Area and Severity Index (PASI) score 12 at baseline were eligible for this 12-week, double-blind, placebo-controlled study. Patients were randomized to I of 6 arms: 1) 100 mg ABT-874 every other week (eow) for 12 weeks; 2) one 200-mg ABT-874 dose at 15 Week 0; 3) 200-mg ABT-874 every week for 4 weeks; 4) 200-mg ABT-874 eow for 12 weeks; 5) 200-mg ABT-874 every week for 12 weeks; or 6) placebo. Primary endpoint was a >PAS175 response at Week 12. Other efficacy assessments included the PASI50 and Physician's Global Assessment (PGA). Patients who met the primary endpoint entered a 36-week blinded/retreatment phase and were monitored for time to loss of 20 response. A total of 180 patients enrolled in the study, 30 in each arm. Baseline characteristics were similar between arms and indicative of moderate to severe Ps (all mean values except % male): age, 46 yrs, 74% male; 21 yrs duration of Ps; PASI 19; and 25% BSA affected. At Week 12, the percentages of patients achieving >PASI75 were 25 statistically significantly greater for patients in each of the 5 ABT-874 arms vs. placebo (93%, 63%, 90%, 93%, 90%, vs. 3%, respectively, p<0.001, ITT). In addition, the percentages of patients achieving >_PASI50 were statistically significantly greater for patients in each of the 5 ABT-874 arms vs. placebo (100%, 77%, 97%, 97%, and 100%, vs. 17%, p<0.001). The mean percentage decreases (improvements) in PASI at Week 12 30 were 90%, 70%, 92%, 92%, and 90%, respectively, in the ABT-874 arms, and 26% for placebo. Similarly, the percentages of patients with a PGA of Clear/Minimal were 83%, 50%, 73%, 87% and 87%, respectively, in the ABT-874 arms, and 3% for placebo.
153 In conclusion, ABT-874 was significantly more efficacious than placebo in the treatment of moderate to severe plaque psoriasis. Example 2: Safety and Efficacy of the Fully Human IL-12/-23 Monoclonal 5 Antibody, ABT-874, in the Treatment of Moderate to Severe Plaque Psoriasis ABT-874 is a fully human antibody against interleukin 12 (IL-12) and IL-23. It binds with great affinity to the p40 subunit common to both IL-12 and IL-23, validated targets in the treatment of psoriasis (Ps). The objective of this Phase II study was to investigate the efficacy and safety of subcutaneous injections of ABT-874 in the 10 treatment of moderate to severe plaque Ps. Adults with Ps affecting 210% body surface area (BSA) and a PASI score >12 were eligible for this 12-wk, double-blind, placebo-controlled study. Patients were randomized to 1 of 6 arms: 1) 100-mg ABT-874 every other week (eow) for 12 wks; 2) one 200-mg ABT-874 dose at Wk 0; 3) 200-mg ABT-874 every wk for 4 wks; 4) 200 15 mg ABT-874 eow for 12 wks; 5) 200-mg ABT-874 every wk for 12 wks; or 6) placebo. The primary endpoint was a >PASI75 response at Wk 12. Patients who met the primary endpoint entered a 36-wk blinded/retreatment phase and were monitored for time to loss of response. All patients were evaluated for safety through Wk 54. 180 patients enrolled, 30 in each arm. Baseline characteristics were similar 20 between arms (mean values presented except % male): age, 46 yrs, 74% male; 21 yrs duration of Ps; PASI=19; and 25% BSA affected. At Wk 12, the %s of patients with >PAS175 were statistically significantly greater in each of the 5 ABT-874 arms vs. placebo (93%, 63%, 90%, 93%, 90%, vs. 3%, respectively, p<0.001, ITT). During the 12-wk, DB phase, infectious AEs for the ABT-874 groups ranged from 23-43% and for 25 the placebo group was 23%, with the most common being nasopharyngitis (7-17% for ABT-874; 3% for placebo). There were no statistically significant differences between arms. No serious infectious AEs were reported, and no deaths occurred. In conclusion, ABT-874 was significantly more efficacious than placebo in the treatment of moderate to severe plaque Ps, and appears to have a favorable safety 30 profile.
154 Example 3: Maintenance of Response with the Fully Human IL-12/-23 Monoclonal Antibody, ABT-874, in the Treatment of Moderate to Severe Plaque Psoriasis The efficacy and safety of ABT-874 was evaluated in a 12-week, Phase II, 5 randomized controlled trial and 36-week follow-up phase. The objective of the following example was to analyze maintenance of response following discontinuation of therapy during the second 12 weeks of this Phase II study of subcutaneous injections of ABT-874 in the treatment of moderate to severe plaque Ps. Adults with Ps affecting 210% body surface area (BSA) and a PASI score >12 10 were eligible for this 12-week, double-blind, placebo-controlled study. Patients were randomized to 1 of 6 arms: 1) 100-mg ABT-874 every other week (eow) for 12 wks; 2) one 200-mg ABT-874 dose at Wk 0; 3) 200-mg ABT-874 every wk for 4 wks; 15 4) 200-mg ABT-874 eow for 12 wks; / 5) 200-mg ABT-874 every wk for 12 wks; or 6) placebo. The primary endpoint was a >PASI75 response at Week 12. Patients who met the primary endpoint entered a 36-week blinded/retreatment phase. Treatment with study 20 drug was discontinued, and patients were monitored for time to loss of response (a decrease in PASI score, any time during the 36-week follow-up period, to <PASI 50). Maintenance of PASI response was evaluated through Week 24. A total of 180 patients enrolled, 30 in each arm. Baseline characteristics were similar between arms (mean values presented except % male): age, 46 years, 74% male; 25 21 years duration of Ps; PASI=19; and 25% BSA affected. At Week 12, the percentages of patients with 2PASI75 were statistically significantly greater in each of the 5 ABT-874 arms vs. placebo (Table 1). At Week 24, substantial percentages of PASI 75 responders in the active treatments arms had maintained at least a PASI 50 response. 30 155 Table 1: 24-Week Efficacy of ABT-874 Maintenance of PASI Response: >PAS175 at Wk 12 Wk 24 vs. Wk 12 100 mg eow for 28/30 (93%)* 24/28 (86%) 12 wks 200 mg, one dose 19/30 (63%)* 15/19 (79%) 200-mg every wk for 27/30 (90%)* 23/27 (85%) 4 wks 200-mg eow for 28/30 (93%)* 26/28 (93%) 12 wks 200-mg every wk for 27/30 (90%)* 26/27 (96%) 12 wks Placebo 1/30 (3%) *p<0.001 vs. placebo, NRI. 5 In conclusion, ABT-874 was significantly more efficacious than placebo in the treatment of moderate to severe plaque Ps. Substantial percentages of PASI 75 responders maintained these responses at Week 24, following discontinuation of active therapy. 10 Example 4: Safety and Efficacy of ABT-874, a Fully Human IL-12/-23 Monoclonal Antibody, in the Treatment of Moderate to Severe Chronic Plaque Psoriasis 15 The objective of the following example was to demonstrate the efficacy and safety of a range of doses of a human IL- 12/23 monoclonal antibody (ABT-874) compared with placebo in the treatment of patients with clinically stable moderate to severe chronic plaque psoriasis. 20 156 I. Materials and Methods A. Study design: The following study was a 12-week, multicentre, randomised, double-blind, phase II, placebo-controlled trial that was conducted at 24 centres in the United States (16 sites) and Canada (8 sites). ABT-874 (Abbott Laboratories, Abbott 5 Park, IL) is a human monoclonal antibody with genetically engineered complementarity determining regions that have high affinity for the IL-12/23 p40 subunit protein. Patients were randomised in a 1:1:1:1:1:1 ratio to receive 1 of 6 treatments: 200 mg of ABT-874, 1 dose at week 0 (200-mg x 1); 100 mg of ABT-874 every other week (eow) for 12 weeks (100 mg eow); 200 mg of ABT-874 weekly for the first 4 weeks (200 mg x 4); 10 200 mg of ABT-874 eow for 12 weeks (200 mg eow); 200 mg of ABT-874 weekly for 12 weeks (200 mg weekly); or placebo. After week 12, all patients who achieved at least a 75% reduction in psoriasis area and severity index (PASI 75) response continued into a 36-week blinded observation/retreatment phase. 15 B. Patients: Patients were 218 years of age and had a clinical diagnosis of psoriasis for at least 6 months (determined by patient interview and confirmation of diagnosis through physical examination by the investigator), stable plaque psoriasis for at least 2 months before screening and at baseline visits as determined by subject interview, moderate to severe plaque psoriasis defined by 10% body surface area 20 (BSA) involvement at the baseline visit, a PASI score of 212 at the baseline visit, and a physician's global assessment (PGA) of at least moderate disease at the baseline visit. Patients were ineligible if they had previous exposure to systemic or biologic anti-IL-12 therapy; nonplaque psoriasis; inability to discontinue the following therapies before the baseline visit: topical psoriasis therapies at least 2 weeks before, ultraviolet B 25 light phototherapy at least 2 weeks before, psoralen-ultraviolet-light phototherapy at least 4 weeks before, systemic therapies at least 4 weeks before, and biologic therapies at least 12 weeks before; required intake of oral or injectable corticosteroids during the study (inhaled corticosteroids for stable medical conditions were allowed); an exacerbation of asthma requiring hospitalization in the 10 years prior to screening; an 30 infection or risk factors for severe infection; a history of malignancies other than successfully treated basal cell carcinoma (patients with a history of squamous cell carcinoma were excluded) or cervical carcinoma in situ; or a history of major immunologic reaction (e.g., serum sickness or anaphylactoid reaction) to an 157 immunoglobulin G-containing agent (e.g., intravenous gamma globulin, a fusion protein, or monoclonal antibody). Patients were allowed to continue treatment with medicated shampoos that did not contain corticosteroids, bland (without beta- or alpha-hydroxy acids) emollients, or 5 Class VI or VII low-potency topical corticosteroids on their palms, soles, face, inframammary area, and groin area during the course of the study. Application of these topical psoriasis therapies was not to occur within 24 hours of a study visit. Vaccination with a live viral agent was not allowed within 1 month prior to dosing with ABT-874, during the study, or for 1 month after the last dose of study drug was administered. 10 Occurrence of any of the following clinically significant abnormal laboratory results led to immediate withdrawal of a patient from the study: aspartate transaminase or alanine transaminase >5 times the upper limit of normal; serum total bilirubin >3 times the upper limit of normal; serum creatinine >3 times the upper limit of normal; creatine phosphokinase >5 times the upper limit of normal; hemoglobin <8 g/dL; white 15 blood cell count <2 x 10 9 /L; or platelet count <75 x 10 9 /L. C. Efficacy assessments: The primary efficacy assessment was the percentage of patients achieving a PASI 75 response at week 12, defined as at least a 75% reduction in PASI score relative to the baseline score. PASI is a measure of the severity of 20 psoriatic lesions (in terms of erythema, induration, and desquamation) and the extent of BSA involvement. The PASI score ranges from 0 (no psoriasis) to 72 (severe disease) (Fredriksson T, Pettersson U. Dermatologica 1978; 157: 238-44). Other efficacy measures included the percentage of patients who achieved at least PASI 75 at weeks 1, 2, 4, and 8; the percentage of patients who achieved at least PASI 50 or PASI 90 at 25 weeks 1, 2, 4, 8, and 12; and the percentage of patients who attained a PGA of clear or minimal at week 12 and at weeks 1, 2, 4, and 8. The PGA measures the severity of disease on a 6-point scale, which ranges from 0 (no disease, or clear) to 5 (very severe) (Ko H-S. Clinical trial design in psoriasis. Presented at: 49th Meeting of the Dermatologic and Ophthalmologic Advisory Committee; March 20, 1998; Bethesda, 30 MD). D. Safety assessments: Adverse events, laboratory data, and vital signs were assessed throughout the study. Patients were closely monitored for signs of infection, 158 malignancy, and immunologic reaction. Treatment-emergent AEs were defined as those events that occurred between week 0 and the earlier of 45 days after the last nonmissing study drug dose or 1 day prior to the first retreatment dose (for those patients continuing on to the 36-week trial). 5 E. Statistical analysis: The sample size was calculated-using nQuery Advisor® 4.0 (Statistical Solutions, Saugus, MA). With the assumption that 15% of the patients in the placebo group would achieve a PASI 75 response at week 12, the study designers determined that a sample size of 26 in each dosage group would be adequate to detect at 10 least a 45% difference from a treated group using the Fisher exact test with 90% power at a 0.05 2-sided significance level. The study was designed to enroll approximately 180 patients, with 30 patients in each group. The intention-to-treat population included all patients who were randomised at week 0 and received at least 1 injection of study drug; this population was used for the 15 efficacy analyses. All tests were performed at a=0.05. Nonresponder imputation was used for all efficacy analyses; any patient with a missing PASI or PGA score at any visit was considered a nonresponder at that visit. To assess the impact of the missing data, sensitivity analyses of week-12 data were completed using the last-observation-carried forward method. The primary analysis of PASI 75 response at week 12 was performed 20 using the following sequential order to adjust for multiplicity: 200 mg weekly versus placebo, 200 mg eow versus placebo, 100 mg eow versus placebo, 200 mg x 4 versus placebo, and 200 mg x 1 versus placebo. The treatment difference between each ABT 874 treatment group and the placebo group for mean percentage change in PASI score was assessed using analysis of variance, with baseline PASI score and treatment group 25 as factors. The safety analyses were conducted using the safety population, which included all patients who received at least 1 injection of study drug. II. Results A. Patients: A total of 180 patients were enrolled and randomised to 1 of the 6 30 treatment groups (Figure 1). The majority of patients (76.7% of placebo-treated patients and 98% of all ABT-874 treatment group patients) completed the 12-week portion of the study.
159 Patients were well balanced across treatment groups with respect to demographic characteristics and disease activity (table 1). Patients were predominantly male (74.4%) and white (92.2%). Mean BSA involvement was 25% and mean PASI score was 18.8. 5 B. Efficacy: The percentage of patients achieving the primary endpoint of PASI 75 response at week 12 was statistically significantly greater (p<0.001) in all of the ABT-874 treatment groups (200 mg x 1: 63.3%, 19 of 30; 100 mg eow: 93.3%, 28 of 30; 200 mg x 4: 90.0%, 27 of 30; 200 mg eow: 93.3%, 28 of 30; 200 mg weekly: 90.0%, 27 of 30) compared with placebo (3.3%, 1 of 30). For the relatively short duration of this 10 trial, PASI 75 responses in all ABT-874 treatment groups were similar with the exception of the 200 mg x I treatment group (Figure 2). A subgroup analysis by demographics (gender, age, race, and weight), baseline disease characteristics (history of psoriatic arthritis, BSA, and PASI score), and baseline therapy for psoriasis within 12 months of receiving study treatment (systemic biologic 15 and nonbiologic, topical, and phototherapy) demonstrated that ABT-874-treated patients within the various subgroups consistently achieved high levels of PASI 75 response at week 12. Nearly 100% of the higher ABT-874 dosage groups attained at least a PASI 50 response by week 12 (200 ng x 1: 76.7%, 23 of 30; 100 mg eow: 100.0%, 30 of 30; 200 20 mg x 4: 96.7%, 29 of 30; 200 mg eow: 96.7%, 29 of 30; 200 mg weekly: 100.0%, 30 of 30; placebo: 16.7%, 5 of 30; p<0.001 for each comparison with placebo). The percentage of patients achieving at least a PASI 90 response at week 12 was statistically significantly greater (p<0.001) in all but 1 (200 mg x 1) of the ABT-874 treatment groups when compared with placebo, as follows: 200 mg x 1: 16.7%, 5 of 30; 100 mg 25 eow: 53.3%, 16 of 30; 200 mg x 4: 63.3%, 19 of 30; 200 mg eow: 76.6%, 23 of 30; 200 mg weekly: 53.3%, 16 of 30; and placebo: 0%, 0 of 30. In addition, by week 12, significantly more (p<0.001) patients in all ABT-874 treatment groups had attained a clear or minimal PGA rating compared with patients in the placebo group, as follows: 200 mg x 1: 50.0%, 15 of 30; 100 mg eow: 83.3%, 25 of 30; 200 mg x 4: 73.3%, 22 of 30 30; 200 mg eow: 86.7%, 26 of 30; 200 mg weekly: 86.7%, 26 of 30; versus placebo: 3.3%, 1 of 30. The percentage of patients achieving the primary endpoint of PASI 100 response at week 12 was statistically significantly greater (p<0.001) in the following ABT-874 160 treatment groups (200 mg eow: 46.7%, 14 of 30; 200 mg weekly: 36.7%, 1 l'of 30) compared with placebo (0%, 0 of 30). Response to ABT-874 was rapid. The mean percentage improvement in PASI scores from baseline increased over time for all ABT-874 treatment groups (Figure 3) 5 and were statistically significantly greater for each ABT-874 treatment group compared with placebo at each time point (p<0.001, except for the 100 mg eow group at week 1, p=0.023). C. Safety: ABT-874 therapy was generally well tolerated (table 2). One (0.7%) 10 patient treated with ABT-874 discontinued the study owing to a localised skin discoloration; 2 (6.7%) patients treated with placebo discontinued the study, 1 for psoriatic arthropathy and 1 for ovarian cancer. Two (1 .1%) patients experienced serious adverse effects (AEs); 1 placebo-treated patient was diagnosed with ovarian cancer on day 37, and 1 ABT-874-treated patient (200 mg x 1) was diagnosed with 15 costochondritis on day 10. No patients experienced myocardial or cerebral infarctions, and there were no deaths. Patients receiving any dose of ABT-874 were significantly (p=0.033) more likely than patients receiving placebo to experience an AE at least possibly related to study drug (ABT-874: 36.0%, 54 of 150; placebo: 10.0%, 3 of 30; table 2); most of these AEs 20 were related to the injection site (injection-site reaction, erythema, pruritus, or irritation). Most AEs were mild (mild AEs occurred in 46.0% [69 of 1501 of ABT-874 treated patients and 30.0% [9 of 30] placebo-treated patients). The most common AE was injection-site reaction, occurring in 16.7% (25 of 150) of patients treated with any dose of ABT-874 (no reported injection-site reactions for placebo-treated patients; 25 p=0.028; table 3). There were no statistically significant differences between the incidences of other AEs in the ABT-874-treated patients compared with placebo-treated patients. The next most frequently reported AEs were nasopharyngitis and upper respiratory tract infection. Infectious AEs were reported by 32.8% (59 of 180) of all patients (placebo: 30 23.3%, 7 of 30; all ABT-874-treated patients: 34.7%, 52 of 150). The most common infectious AEs reported for any ABT-874 treatment group were nasopharyngitis (12.0%, 18 of 150), upper respiratory tract infection (10.7%, 16 of 150), and bronchitis and viral infection (both 2.7%, 4 of 150). No serious infectious AEs were reported.
161 Two patients reported malignancies during the study. One placebo-treated patient was diagnosed with ovarian cancer, which was ongoing as of day 129. One ABT-874 treated patient (200 mg x 4) was diagnosed with a non-melanoma skin cancer (squamous cell carcinoma) that was removed on day 133. The medical history for this patient 5 included removal of a benign skin growth in March 2005. There were no clinically significant hematology, chemistry (including blood glucose concentrations), or vital sign changes compared with placebo.
162 rg 0 C> r r- - > 0 00 tn 4 o6 6 o - 1 -f -H +1 0011 C rg44~ C> - ~ o~ - "a I 0 > 0 41 + - - Q~~o oo&Soo~C>\0 ~ ~ & C1 E Zc 6 M k ne C14 cn (7 r4 r4 S C03 bo0C'C 0nr-C -(7 ' 0 C'4 C' r4 Wr) "o 0 - '-e'";-"-1 C 0 .40 *R o 4 C 3 bo~~' iZ- u- m m -0 M- 0 C'i ~ ~ ~ ~ m 1 1 1 * 1 *- 00r C 00 -03 q*C-4~~ r4 00 (O 0 r. ~~~~~~ 0, 0,,Z~ -- 0 0 c 0~0 *.* o40033 -. >,~ a - u < 0 E 163 u 0 C). W) C. CA) ~ ~ , 0 rC~ 00 o >~ CD- \.o -o C) C) en) .0 C.) -)C CA) b m C.) v Z ii 0.~~ 0r~ 0r 05 ItZ -nC) \o CA) -- It C) 0) C) C.) C .0 M 00 00 000 F- -o - 2 o-C _ _ C) 14 L ~ ~ 0 m0 m, r,0 0 0 0-0 22 o) 0 0 0 U 0-1 . C CA~ Cj 4!0 E 00
C)
164 0 ------- r~roD 0 CC C4 o 6RN.9RR cR rRN 0 -- -i E
~
4 z m:~~C~ m N N Cq e0 x 0N N NN
N.
E m CD0 0 cc 0 0\C0 C) -- o .2 x o 0 o z -r, - - q- - N E7 ogoooooodoog C CC4 CC (A cc E oc -- m C 0 C. . - ( E2 0 E 0 0 PC "F-L * cvi - .E 'n u y ~~ ~ ~ E "- .= a.o.- g .. . c e o E .2 a U E e: S x- z .
I'-)
165 III. Conclusion The phase II, multicentre, randomised, double-blind, placebo-controlled trial described in this Example demonstrated statistically and clinically significant efficacy of ABT-874 5 in the treatment of moderate to severe chronic plaque psoriasis. With the exception of the ABT-874 200 mg x 1 treatment group, 90% or more of patients in all ABT-874 treatment groups achieved PASI 75 or greater by week 12, compared with 3.3% of placebo-treated patients. Even in the group that received only I dose of study drug (200 mg x 1), a majority (63.3%) of patients had achieved at least PASI 75 by week 12. In 10 addition, almost 100% of patients treated with ABT-874 reached PASI 50 or greater, which is considered to be a clinically significant improvement (Carlin CS, Feldman SR, Krueger JG, Menter A, Krueger GG. J Am Acad Dermatol 2004; 50: 859-66) by week 12. The results for other secondary endpoints, such as PASI 90 and PGA of clear or minimal, were consistent with and supported the primary efficacy analysis. 15 Response to ABT-874 was rapid. Statistically significant separation between placebo- and ABT-874-treated patients occurred as early as week 1 for the mean percentage improvement in PASI scores. Improvement was sustained for the 12-week duration of the trial, even for patients in the ABT-874 200 mg x I and 200 mg x 4 dosage groups. 20 ABT-874 was well tolerated, and most AEs were mild. Although ABT-874 treated patients were significantly more likely to experience an AE at least possibly related to study drug, most of these were injection site-related AEs (injection-site reaction, erythema, pruritus, or irritation). There was no apparent association between an increased dose of ABT-874 and an increased incidence of AEs. Of note, there were no 25 myocardial or cerebral infarctions. Immunologic-related events are of particular interest for patients receiving anti ILl-12/23 antibodies. The most frequently reported infectious AEs were nasopharyngitis, upper respiratory tract infection, bronchitis, and viral infection. There were no serious infectious AEs reported for the duration of this trial. Of the 2 malignancies diagnosed 30 during the study, ovarian cancer was diagnosed in a placebo-treated patient, and non melanoma skin cancer was diagnosed in an ABT-874-treated patient who had a history of a benign skin growth.
166 In summary, ABT-874 demonstrated statistically and clinically significant benefit for the treatment of patients with moderate to severe chronic plaque psoriasis, and was well tolerated. 5 Example 5: Maintenance of Response with the Fully Human IL-12/-23 Monoclonal Antibody, ABT-874, in the Treatment of Moderate to Severe Plaque Psoriasis The efficacy and safety of ABT-874 was evaluated in a 12-week, Phase II, randomized controlled trial and 36-week follow-up phase. The objective of the 10 following example was to analyze maintenance of response following discontinuation of therapy during the second 12 weeks of this Phase II study of subcutaneous injections of ABT-874 in the treatment of moderate to severe plaque Ps. Adults with Ps affecting 210% body surface area (BSA) and a PASI score 212 were eligible for this 12-week, double-blind, placebo-controlled study. Patients were 15 randomized to 1 of 6 arms: 1) 100-mg ABT-874 every other week (eow) for 12 wks; 2) one 200-mg ABT-874 dose at Wk 0; 3) 200-mg ABT-874 every wk for 4 wks; 4) 200-mg ABT-874 eow for 12 wks; 20 5) 200-mg ABT-874 every wk for 12 wks; or 6) placebo. The primary endpoint was a ?PASI 75 response at Week 12. Patients who met the primary endpoint entered a 36-week blinded/retreatment phase. Treatment with study drug was discontinued, and patients were monitored for PASI score at various 25 times during the 36-week follow-up period, including PASI 50, PASI 75 and PASI 90 responses. Maintenance of PASI response was evaluated through Week 24. A total of 180 patients enrolled, 30 in each arm. Baseline characteristics were similar between arms (mean values presented except % male): age, 46 years, 74% male; 21 years duration of Ps; PASI=19; and 25% BSA affected. 30 At Week 12, the percentages of patients with >PASI 75 were statistically significantly greater in each of the 5 ABT-874 arms vs. placebo (Table 4). At Week 24, substantial percentages of PASI 75 responders in the active treatments arms had maintained at least a PASI score of >PASI 50. Further, substantial percentages of PASI 167 75 responders in the active treatments arms had also maintained at least a PASI score of >PASI 75, as well as a PASI score of >PASI 90 (Table 4 and Figures 4A-C). The percentage of patients maintaining a PASI 75 response over time during the 24 week period is depicted in Figure 4D. 5 Table 4: 24-Week Efficacy of ABT-874 Maintenance of Maintenance of Maintenance of >PASI 50 >PASI 75 >PASI 90 PASI 75 at Response: Response: Response: Wk 12 Wk 24 vs. Wk 12 Wk 24 vs. Wk 12 Wk 24 vs. Wk 12 100 mg eow for 93%* 71% 60% 33% 12 wks 200 mg, one dose 63%* 68% 23% 7% 200-mg every wk 90%* 82% 60% 23% for 4 wks 200-mg eow for 93%* 89% 73% 53% 12 wks 200-mg every wk 90%* 85% 83% 57% for 12 wks Placebo 3% - 7% 7% *p<0.001 vs. placebo, NRI. In conclusion, ABT-874 was significantly more efficacious than placebo in the 10 treatment of moderate to severe plaque Ps. Substantial percentages of PASI 75 responders maintained a response of > PASI 50, > PASI 75, and > PASI 90 at Week 24, following discontinuation of active therapy. Example 6: Maintenance of Re-treatment Response with the Fully Human 15 IL-12/-23 Monoclonal Antibody, ABT-874, in the Treatment of Moderate to Severe Plaque Psoriasis The efficacy and safety of ABT-874 was evaluated in a 48-week, Phase H, randomized controlled trial that included a 12-week initial treatment phase and a 36 week re-treatment phase of patients responding to initial treatment. The initial 12-week 20 efficacy results and maintenance of response results are described in the above examples. The objective of the following example was to examine the re-treatment 168 response during the 36-week re-treatment/follow-up phase in patients who lost their initial responses of this Phase II study of subcutaneous injections of ABT-874 in the treatment of moderate to severe plaque Ps. The further objective of the following example was to examine safety of subcutaneous injections of ABT-874 in the treatment 5 of moderate to severe plaque Ps through 48 weeks. At baseline, demographics and clinical characteristics were similar across treatment groups (summarized in Table 5 below). Table 5: Baseline Demographics and Clinical Characteristics Treatment Group" PCabo 200 mg I 1om w c200mg.4 200 rn ow 200 mg Weekly All Charactritic (-31)) 1-30) n-130) (n-301 (n 30) (n-30) )N150) Age, yr 49(14.4) 52(12.0) 45(13.8) 43 (13.6) 44(16.0) 46(14.0) 46114.1) St., n %} nale 22 (73) Z3 (77) 22 (73) 21 (70) 23 (77) 23 (77) 112 (75) Race, n 1%) white 28(93) 25(83) 28(93) 27 (90) 30(100) 28 (93) 138(92) Weight, kg 89(17.6) 94 (21.2) 4 17.9) 92 427.0) 93(24.1) 95 (18.0) 94(21.9) Duration of psoriasis, yrs 21 (12.4) 20(13.2) 24(14.6) 22(14.2) 18 (11.5) 11(10.91 21(13.0) PASI score Mean (SD) 18(2.9) 16(8.7) 20(6.3) 20 (7.6) 20 (6.2) 19(6.3) 1946.6) MedIan, 10 16.1, 3.8 15.0, 7.5 18.7, 7.4 17.0,10.2 18.0,10.0 16. 6,5.8 17.3, 8.0 BSA affected, % Mean (SD) 21 (9.2) 24 (13.6) 28 (15.7) 24 (13.0) 29(15.8) 23(12.6) 26(14.5) Median, IQ 17.5,13.0 17.5.16.0 22.5,19.5 20.3,17.0 22.0. 24.5 19.5. 17D 20.0.21.0 PGA, n (%)' Mild 1(3) 0 0 0 0 0 0 Moderate 20(67) 19(63) 17(57) 13(43) 15(50) 17(57) 81(54) Siere 9(30) 11 (37) 12(40) 14(47) 13 (43) 11 (37) 61 (41) HistoryofPsA, n(%) 9(30) 7(23) 12(40) 9(30) 6(20) 9(30) 43(29) Previous psoriasis treatmeontS n Topical therapy 19(63) 21(70) 26(87) 15(50) 21 (70) 23 (77) 106(71) Photolthaupy 1(3) 0(20) 4(13) 4(13) 3(10) 5(17) 22(15) Systsmic nonbiologic 6(20) 4(13) 7)23) 517) 6 (20) 8V() 30(20) S Icblologic 3(10) 3(10) 7)23) 6(20) 4(13) 7(23) 27(18) 10 1 BSA, body surface area; PsA, psoriatic arthritis. *Values are mean (SD) unless otherwise noted. tData are presented for only 3 of 5 possible categories and therefore do not sum up to 30 for each group. tWithin the 12 months before study treatment. 15 Adults with psoriasis affecting >10% body surface area and a Psoriasis Area and Severity Index (PASI) score >12 were randomized to I of 6 arms: 1) one 200-mg dose ABT-874 at Week 0; 2) 100 mg of ABT-874 every other wk (eow) for 12 weeks; 3) 200 mg of ABT-874 weekly for 4 weeks; 4) 200 mg of ABT-874 eow for 12 weeks; 5) 200 20 mg of ABT-874 weekly for 12 weeks; or 6) placebo. The primary endpoint was a >PASI 75 response at Week 12. Patients who met the primary endpoint entered a 36-week re treatment phase. Treatment with study drug was discontinued, and patients who lost response (SPASI 50) during weeks 12-36 received re-treatment with the same dosing regimen assigned during the initial 12-week period. Re-treatment lasted for 12 weeks. 25 Regardless of disposition, all patients were monitored for the entire duration of the study, or until discontinuation.
169 Of the 180 patients initially enrolled, 130 (1 placebo) entered the retreatment phase and 58 (all ABT-874) were re-treated. The percentages of patients who achieved PASI 75 at week 12 and then again at 12 weeks after re-treatment were as follows for each group: one 200-mg dose, 63% vs. 55%; 100 mg eow, 93% vs 94%; 200 mg weekly 5 4 wks, 90% vs. 69%; 200 mg eow, 93% vs. 75%; and 200 mg weekly, 90% vs. 83%, respectively. Of the total 58 patients who were retreated, 76% achieved >PASI 75 at 12 weeks after re-treatment. The improvement in PASI scores over time for the re-treated patients is depicted in Figures 5A-B. Specifically, Figure 5A displays the mean percentage improvement 10 from baseline in PASI scores from weeks 4 to week 12 in PASI responders, and Figure 5B displays the mean percentage improvement from baseline in PASI scores from weeks 4 to week 12 post retreatment in PASI 75 responders. The percentages of patients who achieved PASI 50 at 12 weeks after re treatment were as follows for each group: one 200-mg dose, 82%; 100 mg eow, 100%; 15 200 mg weekly 4 wks, 77%; 200 mg eow, 83%; and 200 mg weekly, 100%. Of the total 58 patients who were retreated, 88% achieved 2PASI 50 at 12 weeks after re-treatment. The percentages of patients who achieved a PGA of "clear" or "minimal" at 12 weeks after re-treatment were as follows for each group: one 200-mg dose, 36%; 100 mg eow, 75%; 200 mg weekly 4 wks, 62%; 200 mg eow, 67%; and 200 mg weekly, 20 83%. Of the total 58 patients who were retreated, 64% achieved a PGA of "clear" or "minimal" at 12 weeks after re-treatment. Adverse events (AEs) occurring 25% in at least 1 treatment group in descending order through week 48 were: nasopharyngitis, injection-site reaction, upper respiratory tract infection, headache, hypertension, and arthralgia. An overview of treatment 25 emergent adverse events through Week 48 is displayed in Table 6 below. An overview of treatment-emergent adverse events with an incidence 25% in any treatment group is displayed in Table 7 below. 30, 170 Table 6: Overview of Treatment-Emergent Adverse Events Through Week 48* Plcbo 200 ngx1 x n100mg ow 200ng 4 200 mg oo 200 m 9 Weekly All ABT n=30 n-30 n=30 n.3 n=30 n=30 N=150 Evet n (%) n (%) n (%) n (%) n (%) n (%) n (% Any AE 18(60.0) 20(66.7) 25(83.3) 25(83.3) 25(03.3) 21(70.0) 116 (77.3) Any AE at least possibly drug-related' 4(13.3) a (30.0) 16(53.3) 16 (53.3) 13 (43.3) 10 (33.3) 64(42.7) Any savers AE 4(13.3) 1 (3.3) 0 2 (6.7) 1 (3.3) 1 (3.3) 5(3.3) Any r AE 1(3.3) 1 (3.3) 0 1 (3.3) 2 (6.7) 0 4 (2.7) Any AE leading to discontinuation of study drug 2 (6.7) 1(3.3) 0 0 0 0 1 (0.7) Any AE at least possibly drug.-rlated and serious' 0 0 0 0 1 43.3) 0 1(0.7) Any infectius AE 7 (23.) 10(33.3) 12(40.0) 14(46.7) 1s(5.3) 10(33.3) 52(41.3) Any serious Infectious AE 0 0 0 0 1(3.3) 0 I(0.7) Any nalignant AE 1 (3.3) 0 0 1(3.3) 0 0 1(0.7) Any lymphons 0 0 0 0 0 0 0 Any nonmelarnoma skin cancer 0 0 0 1(3.3) 0 0 1 0.7) Any injectIon-ite eaction-rlatd AE 0 4 (13,3) 11 (38.7) 12 (40.0) 11 (36.7) 6(20.0) 44 (29.3) Death. 0 0 0 0 *Placebo data are only for the first 12 weeks of the study; all 12-week data previously reported. tAs assessed by investigator. 5 AE, adverse event. Table 7: Treatment-Emergent Adverse Events With an Incidence of 5% or More in any Treatment Group Through Week 48* Plaeb* 200mg. 1 loongow 200ng . 4 200 ngow 20O g WekIy All ABT 30 n- n3 n=30 n=30 n30 N150 Event n (%) n (% ) n I %) n () n (%) n(% Injction-site action 0 2(6.7) 7(23.3) B (26.7) 8(26.7) 4113.3) 29(19.3) Nasopharyngiti. 1(3.3) 5(16.7) 6(20.0) 3(10.0) 4(13.3) 5(16.7) 23(15.3) Uppr respiratorytractinfection 2(6.7) 2(6.7) 5(15.7) 3(10.0) 5(16.7) 2(6.7) 17(11.3) Headache 2(6.7) 5(16.7) 1(3.3) 1(3.3) 3(10.0) 2(6.7) 12(8.0) Injection-ite erythems 0 0 1(3.3) 14(13.3) 2(6.7) 1(3.3) a (5.31 In)tionitspruritus 0 0 1(3,3) 2(6.7) 2(6.7) 2(6.7) 7(4.7) Injction-siteirritation 0 1(3.3) 3(10.0) 2(6.7) 0 0 6(4.0 Arthralgla 1 (3.3) 2(8.7) 1 (3.3) 0 0 2 (J.7) 5(3.3) Virali nfction 0 0 2 (6.7) 2(6.7) 1 (3.3) 5(3.3) Gastrenteritis viral 0 1)(3.3) 0 2 (6.7) 1 3.3) 1 (3.33) 5(3.3) Fatigue 0 2(5,7) 2)6.7) 0 0 1 (3.3) 5(3.3) Hypesttriglycei . 0 1 (3.3) 2(6.7) 2(5.7) 0 0 5(3.3) PaIn in xtrenity 0 1(3.3) 0 0 1(3.3) 2 (.7) 4(2.7) Bronchitis 0 1 (3.3) 0 1(3.3) 2(0.7) 0 4(2.7) Pharyngoaryngeal pain 0 2(6.7) 0 0 0 1(3.3) 3(2.0) Influenza 1 (3.3) 0 1(3.3) 0 2 (6.7) 0 (2.03 Back pain 0 1(3.3) 0 2(6.7) 0 3(2.0) Blood r increased 1 (3.3) 0 2 (6.7) 1 (3.3) 0 3(2.03 Urinary ract inftlon 2 (6.7) 1(3.3) 0 1 (3.3) 1 (3.3) 0 3 (2.0) In nia 1(3.3) 2(6.7) 0 1(3.3) 1(3.3) 0 3(2.0) Nausea 2(6.7) 0 3(10.0) 0 0 0 3(2.0) Cy.: 0 1(3.3) 2(6.7) 0 0 0 3(2.0) Gastroenteriti. 0 0 0 2(8.7) 2(1.3) Rhinorrh.. 0 0 0 0 0 2 '(.7) 2(1.3) Otitise.tern 0 0 0 0 2(6.7) 0 2(1.3) Vomitinj 1(3.3) 0 0 2(6.7) 0 0 2(1.3) Hyperhoiesterolenia 0 0 2(6.7) 0 0 2(1.3} Bood pr . Increased 0 0 2(6.7) 0 0 0 2(1.3) Procedural pain 0 0 2(6.7) 0 0 0 2(1.3) Limnb injury 0 2(8.7) 0 0 0 0 2(1.3) Pruriti. 2(6.7) 0 0 0 0 1 (3.3) 1 (0.7) P0N~.1 rathrfl~opath, 2(6.7) 1(3.3) 0 00 0 1(0.7) *Placebo data are only for the first 12 weeks of the study; all 12-week data previously reported. The foregoing data demonstrate that ABT-874 was highly efficacious in the treatment of moderate to severe psoriasis. Upon loss of response and re-treatment, a 15 majority of patients were able to re-achieve a PASI 75 response. Moreover, ABT-874 appears to have a favorable safety profile in the long term.
171 Example 7: Pharmacokinetics of a Fully Human IL-12/-23 Monoclonal Antibody, ABT-874, in Normal Healthy Volunteers The tolerability, safety, and pharmacokinetics (PK) of a range of doses of ABT 874 were evaluated in a randomized, double-blind, placebo-controlled dose-ranging 5 study. The objective of the following example was to investigate the pharmacokinetics of intravenous (IV) and subcutaneous (SC) injections of ABT-874 in healthy volunteers. The main inclusion criteria were: (i) healthy male volunteers between 18 and 45 years of age; (ii) no clinically relevant abnormalities in any of the investigations of the screening examination (physical exam, vital signs, electrocardiogram, biochemistry, 10 hematology, urinalysis, serology); and (iii) chest x-rays normal within 12 months prior to entering the study. The main exclusion criteria were: (i) smoking more than 10 cigarettes per day; (ii) drinking more than 30 g of alcohol per day; (iii) positive urine drug screen; (iv) chronic infections, especially by intracellular bacterial pathogens such as Mycobacterium tuberculosis; and (v) major infections requiring hospitalization or IV 15 antibiotics within the previous 2 years. Young (18-45 years of age), healthy male volunteers received 2 equal doses (1 IV and 1 SC administered 8 weeks apart) of 0.1, 0.3, 1.0, or 5.0 mg/kg ABT-874 in a 2 period crossover (2 x 2 Latin square) design. Blood samples for the determination of ABT-874 concentrations were collected before the first dose (0) and at 0.5, 1, 1.5, 2, 4, 20 8, 12, 24, 48, 72, 120, 168, 336, 504 and 672 hours after dosing. Serum concentrations of ABT-874 were measured by an enzyme-linked immunosorbent assay. ABT-874 serum concentrations were tabulated individually, described by statistical characteristics (including geometric mean and geometric standard deviation) and displayed as individual as well as mean, median, and geometric mean concentration 25 vs. time curves for IV and SC treatment and each treatment group. The following PK parameters were estimated using noncompartmental methods: e Cmax maximum serum concentration (pg/mL) e Tmax time to reach Cmax (hr) e AUC area under the serum concentration-time curve (ugxhr/mL) 30 * t/ half-life (hr) " CL clearance (mUhr) (for IV administration) " Vz volume of distribution (mL) (for IV administration) 172 e CL/F apparent CL (mL/hr) (for SC administration) e V/F apparent Vz (mL) (for SC administration) A total of 64 patients were randomized; 12 received ABT-874 and 4 received placebo for each dose group. ABT-874 appeared to follow bi-exponential kinetics 5 following N administration, entering the terminal phase approximately 7 days after administration. The mean ± SD terminal half-lives for the 0.1-, 0.3-, 1.0-, and 3.0-mg IV doses were 81.2±55.6, 147 ± 73.2, 208 ± 79.2, and 196 ± 55.4 hours, respectively. The mean ± SD terminal half-lives for the 0.1-, 0.3-, 1.0-, and 3.0-mg SC doses were 221 103, 161 ± 92.6, 210 ± 90.9, and 208 ± 79.2 hours, respectively. The mean terminal 10 half-life for IV administration ranged from 81.2 ± 55.6 hours to 208 ± 79.2 hours. The mean terminal half-life for SC administration ranged from 161 ± 92.6 hours to 221 103 hours. The overall mean terminal half-life was 8-9 days. The pharmacokinetics of ABT-874 (maximum concentration of drug [Cmax] or area under the curve [AUC]) increased proportionally to dose after both IV and SC 15 administrations. The serum concentration-time curve for IV and SC dosing is displayed in Figures 6A and 6B, respectively. The volume of distribution ranged from approximately 8-10 L after IV administration to 24-67 L after SC administration. After SC administration, the time to reach Cmax was approximately 3-4 days. Bioavailability after SC administration ranged between 42% and 62% for the doses evaluated. The 20 pharmacokinetic parameters following IV or SC administration at each dose, including Cmax (the maximum serum concentration in pg/mL), AUC (area under the serum concentration-time curve in pgxhr/mL), tmax (time to reach Cmax in hrs), t 1 /2 (half-life in hrs), CL (clearance in ml/hr) and Vz (volume of distribution (mL)), are displayed in Table 8 below. 25 173 Table 8: PK Parameters (Mean ± SD) in Healthy Human Volunteers Following IV or SC Administration of ABT-874 C- AUC, t CL* VZt Cohort Route (ptg/mL) (hr) ( 1 igxhr/mL) (hr) (mLlhr) (mL) 0.1 mg/kg IV 1.99±0.931 - 146±78.8 81.2±55.6 596±1,850 8,010±7,600 SC 0.245±0.100 66.7±10.6 84.4±40.6 221±103 183±248 66,500±135,000 0.3 mg/kg IV 7.99±3.08 - 5621202 147173.2 50.4±32.7 8,512±3,746 SC 1.09±1.12 90.0±43.6 244t150 161±92.6 183±196 24,800±7,430 5 The foregoing data demonstrate that ABT-874 administered IV and SC in single doses between 0.1 and 5.0 mg/kg was well-tolerated by young healthy male individuals. The pharmacokinetic properties of ABT-874, with its half-life of 8-9 days, are as would be expected for an IgGi antibody. 10 Example 8: Maintenance of Re-treatment Response with the Fully Human IL-12/ 23 Monoclonal Antibody, ABT-874, in the Treatment of Moderate to Severe Plaque Psoriasis The efficacy and safety of ABT-874 was evaluated in a 48-week, Phase II, 15 randomized controlled trial that included a 12-week initial treatment phase and a 36 week re-treatment phase of patients responding to initial treatment. The initial 12-week efficacy results and maintenance of response results are described in examples 1-5 above. The objective of the following example was to examine the re-treatment response during the 36-week re-treatment/follow-up phase in patients who lost their 20 initial responses of this Phase II study of subcutaneous injections of ABT-874 in the treatment of moderate to severe plaque Ps. The further objective of the following example was to examine safety of subcutaneous injections of ABT-874 in the treatment of moderate to severe plaque Ps through 48 weeks.
174 The main inclusion criteria for the trial were: (i) adults with clinical diagnosis of psoriasis for at least 6 months and stable plaque psoriasis for at least 2 months prior to screening; and (ii) moderate to severe plaque psoriasis ( 10% body surface area involvement, Psoriasis Area and Severity Index [PASI] score 212 and a Physician's 5 Global Assessment [PGA] of at least moderate disease) at the baseline visit. A first exclusion criteria for the trial was previous exposure to systemic or biologic anti-IL- 12 therapy. A second exclusion criteria was inability to discontinue the following therapies before the baseline visit: topical psoriasis therapies 22 weeks prior; ultraviolet (UV)-B light phototherapy :2 weeks prior; psoralen-UV light phototherapy 10 >4 weeks prior; systemic therapies >4 weeks prior; and biologic therapies >12 weeks prior. At baseline, demographics and clinical characteristics were similar across treatment groups (summarized in Table 5 of Example 6, above). Adults with psoriasis affecting 10% body surface area and a Psoriasis Area and 15 Severity Index (PASI) score 212 were randomized to 1 of 6 arms: 1) one 200-mg dose ABT-874 at Week 0; 2) 100 mg of ABT-874 every other wk (eow) for 12 weeks; 3) 200 mg of ABT-874 weekly for 4 weeks; 4) 200 mg of ABT-874 eow for 12 weeks; 5) 200 mg of ABT-874 weekly for 12 weeks; or 6) placebo. The primary endpoint was a >PASI 75 response at Week 12. Patients who met the primary endpoint entered a 36-week re 20 treatment phase. Treatment with study drug was discontinued, and patients who lost response (:PASI 50) during weeks 12-36 received re-treatment with the same dosing regimen assigned during the initial 12-week period. Re-treatment lasted for 12 weeks. Regardless of disposition, all patients were monitored for the entire duration of the study, or until discontinuation. 25 Outcome measurements included the following: (i) percentage of patients achieving PASI 75; (i) median time to achieve PASI 75 response after retreatment; (iii) median time to lose PASI 75 response (iii) percentage of patients with a PGA score of "Clear" or "Minimal" after retreatment. Statistical analysis was carried out as follows. Intention-to-treat (ITT) analyses 30 were performed by randomized treatment group. For PASI assessments obtained after retreatment with ABT-874, the assessments were assigned to study visits according to the number of days after the first dose of the retreatment. The proportion of patients 175 achieving PASI response (yes/no) are presented according to the derived study visit. All statistical tests were 2-tailed with a significance value of 0.05 Of the 180 patients initially enrolled (30 patients per treatment group), 130 (1 placebo) entered the retreatment phase and 58 (all ABT-874) were re-treated. The 5 percentages of patients who achieved 2PASI 75 at week 12 and then again at 12 weeks after re-treatment were as follows for each group: one 200-mg dose, 63% vs. 55%; 100 mg eow, 93% vs 94%; 200 mg weekly 4 wks, 90% vs. 69%; 200 mg eow, 93% vs. 75%; and 200 mg weekly, 90% vs. 83%, respectively. Of the total 58 patients who were retreated, 76% achieved 2PASI 75 at 12 weeks after re-treatment. A majority of 10 patients were able to re-achieve a PASI 75 response (Figure 7A). The median time (in days) to achieve PASI 75 during the retreatment phase across all ABT-874 dosage groups is depicted in Figure 7B. The median time to achieve ?PASI 75 during retreatment were as follows for each group: one 200-mg dose, between 60 and 65 days; 100 mg eow, between 55 and 60 days; 200 mg weekly 4 wks, between 15 55 and 60 days; 200 mg eow, between 25 and 35 days; and 200 mg weekly, between 55 and 60 days, respectively. The median time (in days) to lose PASI 75 following the initial 12 weeks of treatment is depicted in figure 7C. The median time to lose PASI 75 following the initial 12 weeks of treatment were as follows for each group: one 200-mg dose, between 20 55 and 60 days; 100 mg eow, between 110 and 120 days; 200 mg weekly 4 wks, between 110 and 120 days; 200 mg eow, between 160 and 180 days; and 200 mg weekly, between 180 and 190 days, respectively. The percentages of patients who achieved a PGA of "clear" or "minimal" (e.g., PGA of 0 or 1) at 12 weeks after re-treatment are depicted in Figure 7D. The 25 percentages of patients who achieved a PGA of 0 or 1 during re-treatment were as follows for each group: one 200-mg dose, between 35% and 40%; 100 mg eow, between 70% and 80%; 200 mg weekly 4 wks, between 60% and 65%; 200 mg eow, between 60% and 70%; and 200 mg weekly, between 80% and 90%, respectively. Of the total patients who were retreated, between 60 and 65% achieved a PGA of 0 or I after re 30 treatment. Adverse events (AEs) occurring 25% in at least 1 treatment group in descending order through week 48 were: nasopharyngitis, injection-site reaction, upper respiratory tract infection, headache, hypertension, and arthralgia. An overview of treatment- 176 emergent adverse events through Week 48 is displayed in Table 6 of Example 6, above. An overview of treatment-emergent adverse events with an incidence >5% in any treatment group is displayed in Table 7 of Example 6, above. The foregoing data demonstrate that ABT-874 was highly efficacious in the 5 treatment of moderate to severe psoriasis. Upon loss of response and re-treatment, a majority of patients were able to re-achieve a PASI 75 response. Moreover, ABT-874 appears to have a favorable safety profile in the long term. Example 9: Efficacy and Safety of a Fully Human Interleukin 12/23 Monoclonal 10 Antibody for the Treatment of Chronic Plaque Psoriasis: 36-Week Observation/Retreatment and 60-Week Open-Label Extension Phases I. Materials and Methods A. Study Design The following was a phase 2, multicenter study that consisted of 3 phases: a 12 15 week, double-blind, placebo-controlled phase; a 36-week observation/retreatment phase; and a 60-week, open-label extension phase. Data from the initial 12-week phase of the current study is described above in Examples 1, 2, 4, and 7. Data from the 36-week observation/retreatment phase of the current study is described above in Examples 3, 5, 6, and 8. The study methods for the previous studies are briefly reviewed in this 20 Example with an emphasis on the 36-week observation/retreatment phase and the 60 week, open-label, extension phase. 1. Treatment During Initial 12-Week and Retreatment/Observation Phases Patients were randomized equally to 1 of 6 groups: one ABT-874 200-mg dose at week 0 (200 mg x 1); ABT-874 100 mg every other week for 12 weeks (100 mg EOW); 25 ABT-874 200 mg weekly for 4 weeks (200 mg x 4); ABT-874 200 mg EOW for 12 weeks (200 mg EOW); ABT-874 200 mg weekly for 12 weeks (200 mg weekly); or placebo. Responding patients (i.e., patients who achieved a PASI 75 response at week 12 entered the 36-week single-blinded observation/retreatment phase in which they were only eligible for ABT-874 retreatment if they lost response; nonresponders at 12 weeks 30 were monitored, but ineligible for retreatment. During the 36-week observation/retreatment phase, patients who lost PASI 50 response compared with baseline were eligible for 12 weeks of ABT-874 retreatment if the loss of response occurred after week 12 and through week 36. Retreated patients received the same 177 regimen of ABT-874 assigned during the 12-week double-blind phase, with the exception of patients assigned to placebo who received ABT-874 200 mg every other week (EOW). Regardless of which study arm they entered, all patients were monitored until week 54 or study discontinuation. 5 2. Treatment During Open-Label Extension Phase Patients who achieved at least a PASI 75 response at week 12 of initial treatment and were actively participating in the observation/retreatment phase were allowed to enter the open-label extension phase. During this phase of the study, patients who lost PASI 50 response compared with baseline were retreated with either 200 mg ABT-874 10 x 1 dose or x 2 doses (1 dose/wk for 2 wk) based on their dosing regimen during the initial 12-week phase of the study. Patients receiving 200 mg ABT-874 x 1 dose included those who were initially randomized to receive 200 mg x 1, 100 mg EOW, or 200 mg x 4. Patients receiving ABT-874 x 2 doses included those who were initially randomized to receive 200 mg EOW, 200 mg weekly, or placebo. Patients who 15 experienced subsequent loss of PASI 50 response that occurred within less than 12 weeks of the previous retreatment were retreated again with the same dosing regimen (1 or 2 doses of ABT-874); retreatment could be repeated until week 56 of the open-label extension phase. 3. Participants 20 Key inclusion criteria were: > 18 years of age; clinical diagnosis of psoriasis for 2 6 months; stable plaque psoriasis for 2 2 months before screening and at baseline; moderate to severe plaque psoriasis (i.e., psoriasis on > 10% body surface area at baseline); a PASI score 12 at baseline; and a PGA score of moderate or higher at baseline. 25 Key exclusion criteria included the following: previous anti-IL-12 therapy; diagnosis of nonplaque psoriasis; inability to discontinue various treatments for psoriasis within predefined time frames, including topical therapies, ultraviolet light phototherapy, and systemic therapies; select comorbid disease or risk factors; and abnormal laboratory values. 30 4. Efficacy and Safety Assessments The primary study outcome of PASI 75 response at week 12 relative to baseline was described above in Example 3-8. Adverse events, vital signs, and laboratory parameters were monitored throughout all phases of the study. Secondary endpoints 178 examined during the 36-week observation/retreatment phase, and evaluated in the current analysis, include the proportion of retreated patients who achieved PASI 50, 75, and 90 scores after 12 weeks of retreatment; median time to loss of PASI 75 response; median time to regain PASI 75 response among retreated patients; PGA scores; and 5 safety. Secondary endpoints assessed during the 60-week open-label extension phase, and evaluated in the current analysis, include the proportion of patients who achieved PASI 50, 75, and 90 scores 12 weeks after retreatment; PGA scores; and safety. A further assessment of the 12-week data examined the impact of baseline covariates (e.g., weight, PASI or PGA score, psoriatic arthritis history, or prior therapy) on subsequent 10 achievement of PASI 75 and is presented here. PASI scores ranged from 0 = "no psoriasis" to 72 = "severe disease" and were calculated as previously described. Severity of disease was measured using PGA scores, which ranged from 0 = "clear" to 5 = "very severe" as previously described. 5. Statistical Analyses 15 Efficacy analyses were conducted using the intent-to-treat population (N = 180), which included all patients who were randomly assigned at week 0 and received at least 1 study drug injection during the first 12-week, double-blind phase of the study. Efficacy analyses were conducted for the observation/retreatment period on those patients who were randomized to ABT-874 at week 0 and who achieved t PASI 75 20 response at week 12, n = 130. For the open-label extension phase, only data for patients who continued in this phase were analyzed. The efficacy variables in the observation/retreatment period and open-label retreatment period were summarized. Patients with missing PASI or PGA scores were counted as nonresponders: Categorical variables were summarized using patient count and percentage. Continuous variables 25 were summarized using descriptive statistics (n, mean, and standard derivation). Safety analyses were conducted using the safety population, which included all patients who received at least 1 study drug injection. II. Results 30 A. Patient Demographics Demographic data for these patients were described above in Examples 3-8 and are summarized below. In the ABT-874 treatment arms, patients, on average, were similar in terms of age (mean, 46 years), were predominantly male (75%) and primarily 179 of white race (92%), and averaged 94 kg in weight. Average duration of psoriasis was 21 years, with mean PASI scores of 19. Patients receiving ABT-874 were divided between moderate (54%) and severe (41%) psoriasis, as determined by PGA scores, with a mean 25.6% of their body surface area affected. Prior therapies consisted of 5 topical therapy (71%), followed by systemic nonbiologic (20%) or biologic (18%) treatment or phototherapy (15%). B. Baseline Covariates in Responders At the 12-week evaluation, a majority of all ABT-874 treated patients achieved a PASI 75 score regardless of differences in baseline clinical and demographic covariate 10 characteristics (e.g., weight, PASI or PGA score, psoriatic arthritis history, or prior therapy, Table 9).
180 ~? 00 00 000 0 0 O 00 ~00 00 00 r-~ ~ 00 00 N n n Cu4~ 00~ 00 I-N o o a eoo o o 00 - o o o 0 S tr; c q' r o o r 0nr0ooo - o'r- 00 00 000 000 o - r0-so ww 0 C141 \ C14 - -4 o-C. or- - or- - so c'1 - CN - - 00 en 0\ 0\ 0\ (N 00 - .
L
6 00 00 aW .m oo00 -) 0 n o o - - - - o .r. - -\ Cu ~-- - -o - - o~6 14 C04 C- 0 00 00O V 00 (O (O 00 cn 0 o , o - - n -.- o os ... 0o N - Nr0 - o - o-o4 C4 2 '4 00 W 000 N -o - os-cro a Cu 0 o) Z 00 V-) 00(-) eq -0 00W)0 4 C00 Cu C14 -q - - t e a aco o o i o oo . 0 z O N N SoeVI A Q n >4 z O V Al LEm r/ VI A rJ : CL 0. < r 181 r) ~ V CN.. r W)C C 00 c 00 0 C,0"' N r00 - 0 O 0 00 00 00 00 0, - - 0co ~Cr o o oo oo0o oo 0N Os 0 No Nr cq _nr en 4 In N-N N N " E '1 -- -~ o o ~C 0oooo o - 0 S6) (O 000 006 0 000 oeoo ano 00oa 00o oo 00 O~ 00 0000 en Ow -o 0 0 00 I - - tri ri - trl\O - - c 000i0 CIO ,e4 C- W_ en 0) 0 N C ( W) - ' 0 (ON N N (N 0 )(N c C-4 00 C1 - 0 C-4 -- Cl dlC- 7 en- NIt It 0 rON onN r0 00 rO 00 - 00 00 It r d2 enO\1 <00 ~ (N00 ~ cn en0 cq 14C, 0o o o oZ -- - ; 0. - - -0- - O0 4 C1 C -- En 'I - -.. s-C'4 O o o 62 OO 0 n C4 oo o [-- c- ~ 0 W) 00 W) ) 182 C. Observation/Retreatment Phase (36 Weeks) Patients who responded with a PASI 75 score at the end of the initial 12-week study were eligible for retreatment during the 36-week observation/retreatment phase (n=130); nonresponders (n=40) were monitored, but not eligible for retreatment. 5 Of the 130 patients who were eligible for and entered the retreatment phase at week 12, 72 patients maintained a response, whereas 58 patients lost their PASI 50 response and were retreated with ABT-874 in the subsequent 12 weeks. The mean PASI score for all 58 retreated patients at the time of retreatment was 13.85, with a range of 13.2 to 14.9 across treatment groups (Figure 10). In patients who lost their PASI 75 10 response during the entire observation/retreatment phase (n=120/130), the median time to loss of the response (calculated from the last dose received during the initial 12 weeks of the study) ranged from 57 to 184 days (Figure 11). Among retreated patients, the percentage of patients who achieved PASI 75 scores 12 weeks after reinitiation of ABT 874 therapy was 54.5% for the 200 mg x 1 treatment group, 93.8% for 100 mg EOW, 15 69.2% for 200 mg x 4, 75.0% for 200 mg EOW, and 83.3% for 200 mg weekly. PASI 50 response 12 weeks after reinitiation of therapy ranged from 76.9% to 100%; PAST 90 response ranged from 9.1% to 83.3% (Table 10). The single-dose treatment group (ABT-874 200 mg x 1) generally had the lowest proportion of patients achieving a given level of response. Patients who achieved a PASI 75 response during 12 weeks of 20 retreatment showed median times to achieve a response ranging from 30 to 62 days (Figure 12). Table 10. PASI Responses After 12 Weeks of Retreatment ABT-874 Treatment Group PASI 50, n(%) PASI 75, n(%) PASI 90, n(%) 200 mg xl (n= 11) 9 (81.8) 6 (54.5) 1 (9.1) 100 mg EOW (n=16) 16(100) 15 (93.8) 12(75.0) 200 mg x4 (n= 13) 10 (76.9) 9 (69.2) 6 (46.2) 200 mg EOW (n= 12) 10 (83.3) 9 (75.0) 6 (50.0) 183 200 mg weekly (n=6) 6 (100) 5 (83.3) 5 (83.3) EOW=every other week; PASI=Psoriasis Area and Severity Index. The majority of retreated patients in each treatment group achieved a PGA score of "clear" or "minimal" after 12 weeks of retreatment (100 mg EOW, 75.0%; 200 mg 5 x4, 61.5%; 200 mg EOW, 66.7%; 200 mg weekly, 83.3%), with the exception of the ABT-874 200 mg x 1 group (200 mg x 1, 36.4%). Median time to achieve a PGA score of "clear" or "minimal" after commencing retreatment ranged from 45 to 85 days (200 mg x 1, 85 d; 100 mg EOW, 58 d; 200 mg x 4, 58 d; 200 mg EOW, 57 d; 200 mg weekly, 45 d). 10 Among patients who received ABT-874 during the first 48 weeks of the study (initial 12-week and 36-week observation/retreatment phases), there were no deaths and 4 patients with serious adverse events; 1 patient discontinued ABT-874 treatment due to an adverse event (Table 11). Among all patients who received ABT-874, injection site reactions, nasopharyngitis, and upper respiratory tract infections were the only adverse 15 events that occurred with an incidence > 10% (Table 12).
184 n _l en _- _ - - 0 00 0 < C )-C o o "* c Z2C cq 1 0) 1 >e 00 00 Go r. 0,- .e en %- C)en mC)C q- 0 0~I~0 mrOe 0 u~o n - - U ~ c o :3 0 0 7 ~ 0 0S W) CD D C 0 U) C)C )CD05> ( E~~ O0Re 0~ 00 Z CD Cl CD ~ EE C,, - C _ en Ln u0 0 en en 00 "0W0 c 00~~c r- 0 0 < C) E~~ Qo- e 4- O > .r- c V) Eo U u.. r U cz~.~o ca. -< <~ -e 185 '4oo '4 ae . a) o .x N14 N - 14 N N Oe EO (C,4 0N Nt Nn (4 - - N N eNex m r-0 0 )r- o f 00 mo . m . 0-00 rN N N) - -~ - Cf en NR c- ena)ne -j o -q C 14 4 - - - .c=.C.4 en0 w x6 c~cl All *it a)~ 0 -. U c.' - u 8 E- c-N N E r-. o E 40. e n o r- l ... - .- c . = , " z 5 x F - .C m c E- . E - A te 4-23o 4 - e Co. 2 a) - . .8 e .- ". E o o - . oa e --. o <a) 2 U . .
186 D. Open-Label Extension Phase (60 Weeks) Patients who achieved a PASI 75 at week 12 and were actively participating in the study (i.e., eligible for retreatment) during the 36-week observation/retreatment 5 phase were allowed to enter the open-label extension phase (n=105). There were no appreciable shifts in overall demographic characteristics in the open-label phase, although there was a statistically significant difference between the ABT-874 200 mg x 1 and x 2 dose groups in the distribution of subjects in each age category (P = 0.010). After week 48, patients received retreatment upon loss of response (< PASI 50) 10 with 200 mg ABT-874 x 1 or x 2 based on their initial randomized dosing group. Of the 105 subjects who entered the open-label extension phase at week 48, 96 (n = 52 and n = 44 for ABT-874 200 mg x 1 and x 2 dose groups, respectively) were retreated at least once. Patients were 'retreated up to 5 times, with the majority of retreated patients being retreated 3 times. Nine patients were not retreated with ABT-874 in the open-label 15 extension phase because they never lost PASI 50 response, but were monitored through the end of the study. With each cycle of retreatment, the proportion of patients who achieved PASI 50, 75, and 90 responses generally decreased, and response rates after retreatment were lower than those seen after initial treatment (Table 13). However, the proportion of 20 patients who achieved a PASI 50 response was similar in the ABT-874 200 mg x 2 dose group for the first 3 retreatments, and the overall PASI 75 response rates were similar for the first and second retreatments. The majority of.all retreated patients achieved a PASI 50 response with the first 3 retreatments. 25 30 187 Table 13. PASI Responses 12 Weeks After ABT-874 Retreatment in Open-Label Extension Phasea ABT-874 200 mg ABT-874 200 mg x 1 x 2 All ABT-874 PASI Response n (%) n (%) n (%) First Retreatment N = 52 N = 44 N = 96 PASI 50 38 (77.6) 37 (92.5) - 75 (84.3) PASI 75 23(46.9) 23(57.5) 46(51.7) PASI 90 13 (26.5) 16 (40.0) 29 (32.6) Second Retreatment N = 42 N = 36 N = 78 PASI 50 25 (67.6) 30 (90.9) 55 (78.6) PASI 75 16 (43.2) 21(63.6) 37 (52.9) PASI 90 5(13.5) 10(30.3) 15(21.4) Third Retreatment N = 31 N = 20 N = 51 PASI 50 10 (47.6) 13 (92.9) 23 (65.7) PASI 75 4 (19.0) 5 (35.7) 9 (25.7) PASI 90 0 0 0 5 PGA scores also declined with each cycle of open-label retreatment and were lower than observed during the initial phase of the study. During the first cycle of retreatment, 30.6% of patients receiving ABT-874 200 mg x I dose and 41.5% of patients receiving ABT-874 200 mg x 2 doses achieved a PGA score of "clear" or "minimal" 12 weeks after retreatment. The proportion of patients achieving PGA scores 10 of "clear" or "minimal" declined somewhat after the second cycle of open-label retreatment (200 mg xl, 23.7%; 200 mg x 2, 39.4% after 12 weeks) but was substantially lower following the third retreatment (200 mg x1, 4.8%; 200 mg x 2, 7.1% after 12 weeks).
188 Sixteen patients (16.7%) in the open-label extension phase experienced a treatment-emergent adverse event (occurring on or after each open-label ABT-874 dose up until 45 days after the date of stopping each dose) deemed possibly study drug related (Table 14). While no deaths or serious infections were reported during the open 5 label extension phase, 3 patients experienced serious adverse events. The only adverse events that occurred with a prevalence > 5%, but less than 10%, during the open-label extension phase were nasopharyngitis, upper respiratory tract infection, injection site reactions, and hypertension (Table 14). 10 Table 14. Overview of Treatment-Emergent AEs During Open-Label Extension Phasea ABT-874 Treatment Group 200 mg x1 200 mg x2 All ABT-874 Adverse Event, n (%) (n=52) (n=44) (n=96) Any AE 32 (61.5) 22 (50.0) 54 (56.3) Any AE at least possibly drug-related 11 (21.2) 5 (11.4) 16 (16.7) Any severe AE 2(3.8) 1 (2.3) 3(3.1) Any serious AEb 3 (5.8) 0 3 (3.1) Any AE leading to DC of study drug 2(3.8) 0 2(2.1) Any AE at least possibly drug-related and serious 0 0 0 Any infectious AE 15 (28.8) 13 (29.5) 28 (29.2) Any serious infectious AE 0 0 0 Any malignant or nonmelanoma skin cancer AE 0 0 0 Death 0 0 0 Treatment-emergent-AE with an incidence > 5% in any treatment group Nasopharyngitis 5 (9.6) 3 (6.8) 8 (8.3) Upper respiratory tract infection 4 (7.7) 4 (9.1) 8 (8.3) Injection site reaction 5 (9.6) 1 (2.3) 6 (6.3) Hypertension 2 (3.8) 3 (6.8) 5 (5.2) Headache 1 (1.9) 3(6.8) 4(4.2) Influenza 0 3(6.8) 3(3.1) AE=adverse event; DC=discontinuation. aAdverse events are reported for patients who received ABT-874 during open-label extension phase; 96 of 105 patients who entered this phase received retreatment.
189 bPatients experiencing more than I serious adverse event are counted only once in the serious adverse event total. In the 200 mg x I group, one patient experienced chest discomfort, chest pain, and dyspnea, one patient experienced obesity, and one patient experienced meningioma. 5 III. Conclusion The preliminary efficacy and safety profile of ABT-874 in patients being retreated for moderate to severe chronic plaque psoriasis was assessed. Current study data show that anti-IL-12/23 therapy has significant promise for treatment and 10 retreatment of patients with moderate to severe plaque psoriasis. Among all patients who received any ABT-874 therapy, a large majority of patients achieved PASI 75 responses following 12 weeks of treatment, regardless of baseline weight, PGA and PASI scores, psoriatic arthritis history, or prior psoriasis treatment. 15 After achieving a PASI 75 response following 12 weeks of treatment in the initial double-blind period and discontinuing treatment in the observation/retreatment period, many patients were able to maintain a PASI 75 response for some time without retreatment. The median time until loss of response was greater in patients receiving higher doses of ABT-874, with the median time to loss of PASI 75 response ranging up 20 to 184 days in the weekly dosing arm. Of particular note, the time required to regain PASI 75 responsiveness among the ABT-874 retreated patients was relatively short, ranging from 30 to 62 days. This is likely dose related, as was time to relapse. In the initial 12-week study phase, a PASI 75 response was achieved by the majority of patients in each ABT-874 treatment group 25 after 8 to 12 weeks, with a median of 57 to 85 days. Results from the open-label extension phase show a diminishing response to subsequent rounds of ABT-874 retreatment. Possible explanations for this reduced response include the abbreviated dosing regimen used during the open-label extension (ABT-874 200 mg x 1 or x 2), as well as the amount of drug received and corresponding pharmacokinetic factors such as 30 ABT-874 concentration. In conclusion, these data show that ABT-874 is an efficacious and safe treatment option for treatment and retreatment of moderate to severe chronic plaque psoriasis.
190 Example 10: Efficacy of Treatment With ABT-874, an Interleukin-12/23 Monoclonal Antibody, Across Body Regions of Patients with Moderate to Severe Psoriasis I. Materials and Methods 5 A. Study Design The following was a twelve-week, randomized, double-blind, placebo-controlled, multi-center study. One hundred and eighty adult patients with psoriasis affecting 10% body surface area and a Psoriasis Area and Severity Index (PASI) score >12 were randomized to: one 200-mg dose ABT-874 at Week 0, 100 mg ABT-874 every other 10 week (eow) for 12 weeks, 200 mg ABT-874 weekly for 4 weeks, 200 mg ABT-874 eow for 12 weeks, 200 mg ABT-874 weekly for 12 weeks, or placebo. The primary endpoint was > PASI 75 response by week 12. Individual PASI scores in the four body regions utilized for PASI evaluation were also assessed. B. Main Inclusion and Exclusion Criteria 15 The main inclusion criteria for the study were: Adult patients with chronic moderate to severe plaque psoriasis for at least 6 months; Stable for at least 2 months; 2 10% BSA; 20 PASI> 12; and Moderate or severe Physician's Global Assessment (PGA) The main exclusion criteria for the study were: Previous exposure to any systemic anti-IL-12 therapy, including ABT-874; 25 Diagnosis of erythrodermic psoriasis, pustular psoriasis, medication-induced or medication-exacerbated psoriasis, or new onset guttate psoriasis; Diagnosis of other active skin diseases or skin infections that might interfere with evaluation of psoriasis; Inability to discontinue prior medication; 30 Topicals and phototherapy for 2 weeks; Nonbiologic systemic therapies for 4 weeks; and 191 Biologics for 12 weeks C. Efficacy Measures The following efficacy measures were used in the present study: 5 The percentage of patients in each group achieving PASI 75 and 2PASI 90 response at Week 12; and The percentage of patients in each group achieving PASI 75 and PASI 90 response at week 12 in specific regions: Head and neck; 10 Upper extremities; Trunk; and Lower extremities D. Safety Measures 15 Laboratory results, vital signs, and adverse events (AEs) were used to determine the safety of the treatment. E. Statistical Methods The following statistical methods were used to determine efficacy of the 20 treatment: Comparison of the proportion of subjects achieving PASI 75 at Week 12 between ABT-874 dose group and placebo group using Fisher's exact test, with 2-sided 95% confidence interval for the difference in proportion; Patients for whom data were missing were assumed to be non-responders (non 25 responder imputation); and P < 0.05 indicated statistical significance II. Results A. Baseline Demographics 30 Baseline demographic and clinical characteristics were similar across treatment groups. See Table 15.
192 00 0-. 00 - 04 +1 +1 H 00 O lo ON 0 00 W 6' r-)~ 0 0 00 0 ' 9 0 0 0 r- 00 W)-Z: _A m~ 00 tn 0' oq r--. C? I
I
O a -H 1 r- 00 00 0 'o -4 o6 10 04 01 4 r4 r -~ - n +0I CZ 0 0 on o' 00 00 t On0 N 4 C- 0000 C, 06 06 r i r- r- 4N m -S 0' -H r- *n -H 000 0 00 S a, -- q o C' e k r) r 0 a' o e~- r4 000 -t 0- 000w ene t') (2 tn z 00 r4 (700c'n _ N ' 06 00 00 -HC~ - ~ *- 0 0 1 00 r- 00 C41 0 r -7oq 00 C) rn - g 193 00 ~ 00 w0 ~ ~00 +1 a 0 0 0 r 0 N ,I 0 ~4 6 o o--: 4 C-i 0 00 i 0 00~ ~ 00 E +1 ~ ~ en ~ " 0 0000 M* o, C(0 00 00 C &&, -E .31- s &C 06 ~ 0 oo 3 n r- 0 0 en 6 6 -46 2--o 3.
00 O ii 004 0 ~ 0 000 08 - Cj n ~0 C~ N .- Nto 9~6 0 ~ -~~ ~ > - . ' O O 0 000 VI A En > r6 co) 194 B. Efficacy A greater percentage of patients treated with any dose of ABT-874 achieved PASI 75 or PASI 90 at week 12 compared with placebo (Figure 13). In the specific body regions examined, a greater percentage of patients treated 5 with ABT-874 achieved PASI 75 and 2PASI 90 at week 12 compared with placebo (Figures 14A-14D). The percentage of patients achieving PASI 75 or 2PASI 90 at Week 12 was greater for all ABT-874 dosage groups combined vs. the placebo group (86.0% vs. 3.3% and 52.7% vs. 0.0%, respectively). For all ABT-874 dosage groups combined, a greater 10 percentage of patients achieved >75% and > 90% improvements in PASI scores as compared to placebo patients for all body regions examined: head and neck (PASI 75, 83.3% vs. 13.3%; PASI 90, 72.0% vs. 13.3%), upper extremities (PASI 75, 81.3% vs. 3.3%; PASI 90, 57.3% vs. 3.3%), trunk (PASI 75, 82.0% vs. 3.3%; PASI 90, 68.0% vs. 3.3%), lower extremities (PASI 75, 78.0% vs. 3.3%; PASI 90, 48.0% vs. 0.0%). 15 C. Safety An overview of the number and percentage of subjects with treatment-emergent adverse events is presented in Table 16. Most reported AEs were mild or moderate in severity. The percentage of patients experiencing serious AEs was low. The most frequently reported treatment-emergent adverse events occurring in 25% of patients are 20 listed in Table 17.
195 00 m oo 00 C - -) - - -- - C o Zl C) m2 mc 00 ON~ Ln. V- m ~ en C c) c) C) en 0c C) 0CD en 11 S0 0 C) 0 c li 0 0O0O C)O0 0 0 0) CC4 LI~C ) C) 0 C) -) 0 4 4 0U z u 0 u cl ~- 0 - ls 7 42z -1 00 a -s a C r o< < ~ < z<i 196 ocli en 0 u 0 tn 0 0 00 Cu 00 Ci o CIS :1 0 U I-Z (U m 197 2n rr- rc oo r - - - i - r--i tn In 'I 'IT m -C4 c eq tn en C- CC C- r4C 0 C C E 0o ene ~ O 0 Al eq z CC 040* r o ~ ~ ' '00 000 000~ ~O 00o co e r - 'S LLu U j 4( 0 aE D. 6U C ( LLu t w 1 G cl. 4 >% c ) o c z D ,x 198 C4 C4 C4 C4 N 'n ) 0 0 0 0 0 0 -~0 000000 ~ - 0 cz N1 -0 R > r . ~00 00C e u = ; W W cz~~- 0 DCDC 2U >I co m -0 U0 .~ 0 (iole Z 199 III. Conclusion In this study, treatment with ABT-874 resulted in clinically meaningful improvements in all four body regions comprising the PASI score, including traditionally resistant areas such as the head and neck and the lower extremities, in patients with moderate to severe psoriasis - Regional as well as overall benefits were demonstrated with all doses of ABT-874 compared with placebo - The results of this study support the utility of IL-12/23 as a therapeutic target in chronic plaque psoriasis. Example 11: Effect of Baseline Characteristics on the Efficacy of ABT-874 for the Treatment of Moderate to Severe Psoriasis I. Methods A. Study Design The following was a twelve-week, randomized, double-blind, placebo-controlled, multi-center study. One hundred and eighty adult patients with psoriasis affecting >10% body surface area and a Psoriasis Area and Severity Index (PASI) score >12 were randomized to: one 200-mg dose ABT-874 at Week 0, 100 mg ABT-874 every other week (eow) for 12 weeks, 200 mg ABT-874 weekly for 4 weeks, 200 mg ABT-874 eow for 12 weeks, 200 mg ABT-874 weekly for 12 weeks, or placebo. The effects of the following baseline characteristics on week 12 PASI 75 response rates were examined: weight, PsA history, PASI score, previous psoriasis treatments and PGA score (severe/very severe). PGA score was a post-hoc analysis. Results are presented for all ABT-874 dosage groups combined following 12 weeks of treatment. B. Main Inclusion and Exclusion Criteria The main inclusion criteria for the study were: Adult patients with chronic moderate to severe plaque psoriasis for at least 6 months; Stable for at least 2 months; 200 > 10% BSA; PASI > 12; and Moderate or severe Physician's Global Assessment (PGA) The main exclusion criteria for the study were: Previous exposure to any systemic anti-IL-12 therapy, including ABT-874; Diagnosis of erythrodermic psoriasis, pustular psoriasis, medication-induced or medication-exacerbated psoriasis, or new onset guttate psoriasis; Diagnosis of other active skin diseases or skin infections that might interfere with evaluation of psoriasis; Inability to discontinue prior medication; Topicals and phototherapy for 2 weeks; Nonbiologic systemic therapies for 4 weeks; and Biologics for 12 weeks C. Efficacy Measures The following efficacy measures were used in the present study: The percentage of patients in each group achieving PASI 75 and ?PASI 90 response at Week 12; and Efficacy Analysis Subgroups (Baseline): Weight (5100 kg or >100 kg) Psoriatic arthritis (PsA) history Previous psoriasis treatments PGA score D. Safety Measures Laboratory results, vital signs, and adverse events (AEs) were used to determine the safety of the treatment. E. Statistical Methods 201 The following statistical methods were used to determine efficacy of the treatment: Comparison of the proportion of subjects achieving >_PASI 75 at Week 12 between ABT-874 dose group and placebo group using Fisher's exact test, with 2-sided 95% confidence interval for the difference in proportion; Patients for whom data were missing were assumed to be non-responders (non responder imputation); and P < 0.05 indicated statistical significance II. Results A. Baseline Demographics Baseline demographic and clinical characteristics were similar across treatment groups. See Table 18.
202 r- -o r- 0w r- m 5 In a , 0 00 0' -- 0 - *.
00 0 0 eirz ONq 0 0 en en -H ei * ~C .
enm0- ~N
C
0 - II ~~~~ 'I H 0 6 ~ 0+0 3 (' LO - - O - a mq -- m cq cne ' o f 0 C> +N 4 1 " I Oq - -H~ . ' o~ +1~ m (' c-;a - c'4 -0 .0 ' 00 06 COO - - 0 - ~ -0~ O0V r,~~ g1 -1C ('4 c'J - .e ('4 mCO0 = _ F40 e~i (' ei - - - 0 0' W) 0' C;N NH 02 CO(' -H- 1c W) 0) sj0 0 z 000c - ~ 2q 0'c ('4 c'J 000 " " en wl r 203 06 't 06+1 00 ~ 0 00 00 000 ii ~c 0000~*e' D C 6- i C E 00 +I'00 o ~00 e CC 1 6 2 20 > E 0 t" "~ =- "a cc' t '4= 0: 204 B. Efficacy A greater percentage of patients treated with any dose of ABT-874 achieved >PASI 75 or >:PASI 90 at week 12 compared with placebo (Figure 15). The effect of baseline characteristics on efficacy was examined. Similar percentages of patients in two weight categories (<100 kg or >100 kg) treated with any dose of ABT-874 achieved PASI 75 at Week 12 (Figure 16). In particular, at week 12, the percentage of patients achieving PASI 75 in the 5100 kg or >100 kg groups was 88.7% vs. 81.1% respectively. Similar percentages of patients with or without a history of PsA achieved PASI 75 at Week 12 (Figure 17). In particular, PASI 75 responses for patients with and without previous PsA were 83.7% vs. 86.9% respectively. A majority of patients (85.5%) with severe or very severe baseline PGA score achieved PASI 75 at Week 12 (Figure 18). Similar percentages of patients in two baseline PASI categories (:20 or >20) achieved PASI 75 at Week 12 (Figure 19). For patients with baseline PASI scores of 520 or >20, 87.0% vs. 84.0% achieved PASI 75 responses at week 12, respectively. Finally, prior psoriasis treatments such as systemic or biologic agents, topicals or phototherapy did not appear to effect PASI 75 response rates, and thus had minimal effect on the efficacy of ABT-874 (Table 19). Table 19. PASI 75 Response at Week 12 by Prior Psoriasis Treatment. Systemic Biologic Topical Phototherapy ABT- n 30 120 30 123 106 44 22 128 874 % achieving PASI 75 90% 85% 85.2% 86.2% 88.7% 79.5% 90.9% 85.2% n 6 24 3 27 19 11 1 29 Placebo % achieving PASI 75 0.0% 4.2% 0.0%. 3.7% 0.0% 9.1% 0.0% 3.4% C. Safety An overview of the number and percentage of subjects with treatment-emergent adverse events is presented in Table 20. Most reported AEs were mild or moderate in severity. The percentage of patients experiencing serious AEs was low. The most . frequently reported treatment-emergent adverse events occurring in >5% of patients are listed in Table 21.
205 < tn c i C ' 0p~00 0 ~ 0 0' m 0 0 E zr 06. ( - . GEO eni C-~ ~ ~ 0 0 0 ~ 0 en- c- E0r 0 0m 0m 0 C-004O O 0 o o z 00 -- - 00 gor 0*j-) CPo o en Soo -Z 00 - 4 1In >~ tuS z 00 00 "D .ti _' U "a m 00 ' 2 E a ~ C tU > U ~ -c g~~~a 0- ~~0 -l 21 V 2Q- 206 0 E o 000 op r t: .t.. 0 2 ~ \ 0 0 0 ~ 0 ' 0 0 0 \ Zzl >- m- , -L z u- 'u >- D 207 U 6o 0. CO 00 ~0 0) Os.. 0 CO 0. us 00 ~ 2 00 .0 0)0 0 A U ~ 0 0 0 ) LL. ~-0 U ~ CO U 00~ 0 0 U '~ U CO CO 0 U .0 00. -C) > .5 0)0) U 4, 4, U r- r "0 "0 CO~ CO~ -~ U U UU~ 5- 4, 0. U 5 U II COUO 4,0 U C 0 -~ U -~ ~ 00 CN C~ a ~ 909 ~ O *~ 0 0 0 U 5- 0 II II II S -~ >~ U U U ~ >~ ~ ~ ~ 4, ~ > CO CO CO E-.~O U>>> 2 ii o,, 0.. 0~ .~ .~ 0.. 0.. U CO.0 C)Z 208 III. Conclusion In this study, a majority of patients with moderate to severe psoriasis treated with ABT-874 achieved a PASI 75 response at Week 12. The percentage of patients achieving PASI 75 at Week 12 was similar, regardless of baseline characteristics, including weight, physician's global assessment, PASI scores, history of psoriatic arthritis, or prior psoriasis treatments. Example 12: Efficacy and Safety Results from a Phase III, Randomized Controlled Trial Comparing Two Dosing Regimens of ABT-874 to Placebo in Patients with Moderate to Severe Psoriasis: The Vero Study I. METHODS Design e Phase III, 52-week, double-blind, placebo-controlled, multi-center clinical trial with two-phases: Induction and Maintenance (Figure 20) (NCT00570986) " Induction Phase: - Patients were randomized 2:1 and received 1 of 2 treatments: - ABT-.874, 200 mg at Weeks 0 and 4, followed by 100 mg at Week 8 - Placebo e Maintenance Phase: - Patients who achieved a Physicians Global Assessment score of "clear" or "minimal" (PGA 0/1) at Week 12 in the Induction Phase were re randomized 2:2:1 (stratified by treatment received in Induction Phase) to 1 of 3 treatment arms: - ABT-874, 100 mg every 4 weeks (q 4 wk) - ABT-874, 100 mg every 12 weeks (q 12 wk) " Placebo q 4 wk 209 Efficacy and Safety Measures " Efficacy was measured using a 6-point Physician's Global Assessment (PGA) scale and Psoriasis Area and Severity Index at Weeks 0, 1, 4, and 8 in the Induction Phase, and every month during the Maintenance Phase (Weeks 12 to 52) " Patients were assessed for adverse events throughout the study, and up to 45 days following the last dose of study medication Patients e Key inclusion criteria: - Adult patients with chronic plaque psoriasis for at least 6 months (and stable for at least 2 months) prior to baseline - Moderate to severe psoriasis defined by the following at baseline: - Affected body surface area (BSA) 210% - PGA at least "moderate" (defined as ?3) " PASI>12 e Key exclusion criteria: - Previous exposure to anti-interleukin 12 therapy, including ABT-874 - Other forms of psoriasis (other than plaque psoriasis) Treatment with any of the following: - Topical treatments (i.e., corticosteroids, vitamin D analogs, or retinoids) or UVB phototherapy within 2 weeks of baseline - PUVA phototherapy or systemic treatments for psoriasis within 4 weeks of baseline . Biologic treatments within 12 weeks of baseline Statistical Methods e Proportions of patients who achieved the following 3 primary endpoints were compared between treatment groups: 210 - PGA 0/1 at Week 12 - 75% improvement from baseline in PASI (PASI 75) at Week 12 - Maintenance of PGA 0/1 at Week 52 " Efficacy analyses were conducted in the intent-to-treat population, and missing values were dealt with using non-responder imputation (NRI) " All statistical tests were two-tailed. The primary analysis at week 12 was conducted using the Cochran-Mantel-Haenszel test.* In all other cases, one-way ANOVA and Chi Square test/Fisher's Exact test were used for continuous and discrete variables, respectively. " Statistical significance level was set at P<.05 *Adjusted for pooled centers, where the smallest center was pooled with the next smallest center to achieve at least 100 patients per pooled center. RESULTS Efficacy * 1465 patients were enrolled into the Vero study (Figure 21) - Induction Phase: = ABT-874, N = 981 - Placebo, N = 484 - Maintenance Phase: (for patients receiving ABT-874 during Induction) - ABT-874 q4 wk, N = 298 - ABT-874q12wk,N=298 " Placebo, N = 149 * Baseline demographics and clinical characteristics were similar between treatment groups (Table 22) 211 Table 22. Baseline Demographics and Clinical Characteristics Induction Phase Maintenance Phase ABT-874 Placebo ABT-874 q4 ABT-874 q12 Placebo Total Baseline Characteristics (N = 981) (N = 484) (N = 298)" (N = 298) (N = 149) (N = 1465) Mean age, yrsh 45.7 ± 13.2 45.1 ± 13.5 44.6 ±13.3 45.5 ±12.6 45.0 ± 13.4 45.5 ± 13.3 Gende (f/m),n(%) :3l5/666' - ,4/343, ~ 92/206 < 97/0 49/100 456/1009 (32'679). ' (29.1/70.9) __ _ _ (309/69. 1) '.(32.6167.4) (32,9/67.1) (31.1/68. 9) Caucasian, n (%) 888 (90.5) 432 (89.3) 275 (92.3) 278 (93.3) 127 (85.2) 1320(90.1) Weight, kg 93.8 ± 23.6 93 ±123 0 91.3 ±213 93.7 ± 23.4 89:3 ±24,6 935 ± 23.41 BSA, % i SD 24.8 ± 16.3 25.7 ± 16.9 23.7 ± 14.8 22.3 ± 13.5 25.2 17.1 25.1 ± 16.5 PASI 19.1 7.5 19.3± 7.3 18.4 ± 6.5 18.3 ± 6.2 18.9± 8.2 * 19.2 i 7.4 Psoriasis duration, yrs 18.9± 12.3 19.2 11.9 19.1 ± 12.1 18.5± 12.1 18.9± 12.4 19.0 ± 12.2 PGA, n(%)- i___-jj Moderate 514 (52.4) 242 (50.0) 170 (57.0) 164 (55.0) 85 (57.0) 756 (51.6) [Sevee ... 408(4)____ 218(45_.0) I13 (37) 124 (41.6) 53 (36) 626 (42.7) Ve-y Severe 59 (6.0) 24(5.0) 15(5.0) 10 (3.4)_ 1(7.4) 83 (5.7) Pavious Medical History, n (%) Any CVD .395 (40.3) 178 (36.8) 108 (36.2) -117 (39.3) 51 (34.2) 573 (39.1) I ypripdei __ __ 145 (4) 57 (L.8) 3(1.4) 51( 17.1) 24(6] 02(13.8)] DM 98(10.0) 40(8.3) 15(5.0) 29 (9.7) 18(12.1) 138 (9.4) q4 = every 4 weeks; q12= every 12 reeks; BSA = Affected body surfacearea; PASI = Psoriasis Area and Severity Index; PGA = Physician's Global Assessment; CVD = cardiovascular disease; DM = diabetes nellitus. One patient in the ABT-874 q4 group was re-randonizd but did nol receive any tudy dog in the Maintenance Phase. Mean value ± SD. * Significantly greater percentages of patients treated with ABT-874 vs. placebo achieved the endpoints of PGA 0/1 and PASI 75 at Week 12 (P<.001, NRI for each analysis) (Figures 22 and 23) e Continued dosing with ABT-874 in subjects who achieved a PGA 0/1 response at week 12 resulted in better maintenance of a PGA 0/1 response at week 52 compared with treatment withdrawal (Figure 24) e Dosing ABT-874 every 4 weeks resulted in better maintenance of PASI 75 and PASI 90 responses than dosing every 12 weeks (Figure 25) Safety * The most common adverse events reported among patients randomized to ABT 874 in the Induction Phase and receiving any ABT-874 through 52 weeks were: nasopharyngitis, headache, and upper respiratory tract infection (Table 23) * 16 patients in the ABT-874 q4 wk treatment group experienced back pain as compared to 6 patients in the q12 wk group, and 3 patients in the placebo group, suggesting a possible dose response relationship for this event. The reason for any potential association is unknown.
212 Table 23. Treatment-Emergent Adverse Events Occurring in >5% of Patients Induction Phase Maintenance Phase ABT-874 Placebo ABT-874 q4 ABT-874 q12 Placebo All ABT-874 (N = 981) (N = 484) (N=297) (N = 298) (N = 149) (N = 998) n (%) Nasopharyngitis 63 (6.4) 20(4.I) 39 (13.1) 35 (117) 9(6.0) 106 (10.6) Headache 53 (5.4) 9 (1.9) 0 0 0 66 (&6) Upper Respiratory Tract Infection 49(5.0) 20(5.0) 48(16.2) 24(8.1) 8 (5.4) 104 (10.4) ei - 7 00~ 16(54 6~2 0) 3_(2.0) <5% 1 "All patients randomized to ABT-874 in the induction Phase and re-randomized to ABT-874 in the Maintenance Phase. " Serious adverse events occurred in 20 (2.0%) ABT-874 and 6 (1.2%) placebo treated patients during the Induction Phase (Table 24) e Of the serious adverse events, 5 (0.5%) in the ABT-874 group and 1 (0.2%) in the placebo group involved an event of infection (Table 24) " During the placebo-controlled Induction Phase, 6 events of malignancy were reported in the ABT-874 group, while none were reported in the placebo group " Of the malignant events observed in patients treated with ABT-874, most were either squamous cell or basal cell skin carcinomas (Table 24) " A total of 7 major adverse cardiac events (MACE) defined as myocardial infarction, stroke or cardiovascular death, occurred in patients receiving ABT 874. There were no MACE events observed in patients receiving placebo. (Table 24) 213 Table 24. Summary of Treatment-Emergent Adverse Events and Adverse Events of Interest Induction Phase Maintenance Phase ABT-874 Placebo ABT-874 q4 ABT-874 q12 Placebo (N = 981) (N = 484) (N = 297) (N = 298) (N = 149) n (%) Any AE 517 (52.7) 229 (47.3) 215 (72.4) 183 (61.4) 86(57.7) AnyAE leading to 17 (1.7) 4 (0.8) 3 (1.0) 6(2.0) 1(0.7 discontinuation of study drug An seridsaE 20 (2.0)> 6'12) 4 ( 32 9R.0) 2 (1 3 Deaths 0 0 0b 0 fAEs off spec al interest: _______________________ Any Infection 219 (22.3) 96(19.8) 132 (44.4) 107(35.9) 41 (27.5) Serious Infections 5 (0.5) (2) 0 2(0.7) 1(0.7) Malignancies 6(0.6) 0 3 5 0 SCC 4 (0.4) 0 2(0.7) na BCC 0 na 2(0.7) 2(0.7) na Cardiovascular 5 (0.5)' 0 1 (0.3)' I ( 0
.
3 ) 0 AE = adverse event; SCC = squamous cell carcinoma; BCC = basal cell carcinoma; na = not applicable. 'One patient experienced cardiac arrest resulting in death (event also listed in AEs of special interest, cardiovascular). BOne event of death occurred >45 days after study discontinuation in a patient who had a cardiovascular event listed in AEs of special interest. 'One patient diagnosed with lung cancer on study day 43. and I with nasopharyngeal cancer on study day 15. dOne patient diagnosed with colon cancer on study day 285. *One patient diagnosed with tonsil cancer on study day 266. rOne patient had cardiac arrest, 3 had myocardial infarction, and I had stroke. 'One patient had an event of acute coronary syndrome. hOne patient had myocardial infarction. e Five of 7 MACE events occurred within the first 2 months of study treatment (Figure 26) e All MACE events occurred in patients who had underlying risk factors for coronary heart disease (CHD) 2 CONCLUSIONS e ABT-874 induced rapid and significantly higher efficacy responses in patients with moderate to severe psoriasis compared to placebo e Dosing ABT-874 every 4 weeks resulted in better maintenance of response than every 12 weeks e A higher incidence of infection and malignancy adverse events were observed in ABT-874 vs. placebo treated patients. Considering the immunomodulating mechanism of ABT-874, these findings are not unexpected and support the need for monitoring and surveillance for these events. e A numerical imbalance was observed for MACE events, with 7 cases reported in the ABT-874 group compared with no events in the placebo group. While 214 cardiovascular events are not unexpected in the psoriasis patient population, further evaluation will determine if an increase in the number of MACE events is a reproducible phenomenon in patients treated with ABT-874. Example 13. Efficacy and Safety Results from a Phase III, Randomized Controlled Trial Comparing the Safety and Efficacy of ABT-874 to Etanercept (ETN) and Placebo in Patients with Moderate to Severe Chronic Plaque Psoriasis OBJECTIVE The primary objective of study M10-315 was to compare, in a Phase III trial, the efficacy and safety of ABT-874 to etanercept and placebo in subjects with moderate to severe chronic plaque psoriasis over 12 weeks METHODS Main Inclusion Criteria e Males and females >18 years with a clinical diagnosis of chronic plaque psoriasis for at least 6 months " Stable plaque Ps for at least 2 months before Screening and at Baseline (Week 0) visits " Affected body surface area (BSA) >10% " Physician's Global Assessment (PGA) of at least moderate (> 3) and PASI score of >12 at the Baseline (Week 0) visit Main Exclusion Criteria e Previous exposure to systemic anti-IL-12/23p40 therapy, including ABT-874 * Previous exposure to ETN or known hypersensitivity to ETN e Inability to discontinue topical therapies, phototherapies, or systemic therapies Study Design (Figure 27) " 12-week double-blind, double dummy, multicenter, randomized study e Patients were randomized to I of the 3 following treatment arms (2:2:1); "a" indicates the second dose of the week when doses were administered twice weekly: 215 * 200 mg ABT-874 SC at Weeks 0 and 4 followed by 100 mg ABT-874 SC at Week 8 * 50 mg of ETN SC twice weekly 3-4 days apart at Weeks 0, Oa, 1, la, 2, 2a, 3, 3a, 4, 4a, 5, 5a, 6, 6a, 7, 7a, 8, 8a, 9, 9a, 10, 10a, 11, and 1 a * Placebo SC at Week 0 and 4 followed by placebo SC at Week 8 to match ABT-874 dosing, or placebo SC twice weekly 3-4 days apart at Weeks 0, Oa, 1, la, 2, 2a, 3, 3a, 4, 4a, 5, 5a, 6, 6a, 7, 7a, 8, 8a, 9, 9a, 10, 10a, 11, and 1 la to match ETN dosing Efficacy Measures e Proportion of patients achieving a PGA score of "Clear" or "Minimal" (0/1) at Week 12 " Proportion of patients achieving a PASI 75/90/100 response at Week 12 Safety Measures e Adverse events and serious adverse events were assessed throughout the study Statistical Methods e The efficacy analyses were conducted in the intent-to-treat (ITT) population; non responder imputation was used to handle missing data e Cochran-Mantel-Haenszel tests stratified by pooled center were used to compare the proportion of patients in each treatment group achieving PGA 0/1 or PASI 75 at Week 12 in the primary analysis e A Chi-Square test, or Fisher's exact test as appropriate, was used to compare proportions of patients in each treatment group achieving PASI 90 or 100 at Week 12 e All statistical tests were two-sided with the significance level of 0.05 e The safety analyses were conducted in the safety population; safety variables were summarized by treatment group RESULTS 350 patients were enrolled and analyzed 216 = ABT-874, N = 139 = Etanercept, N=139 " Placebo, N = 72 * Baseline demographics and clinical characteristics were similar across treatment groups (Table 25) 217 Table 25. Baseline Demographics and Clinical Characteristics Placebo Etanercept ABT-874 Total (N=72) (N=139) (N = 139) (N=350) Age (yrs)a 45.0 (13.6) 45.2 (14.8) 44.9 (12.9) 45.1 (13.8) Male, n (%) 46 (63.9) 85 (61.2) 93 (66.9) 224 (64.0) Caucasian, n (%) 67(93.1) 127 (91.4) 122 (87.8) 316 (90.3) Duration of psoriasis 15.5 (11.7) 15.2 (12.1) 16.3 (12.0) 15.7 (11.9) Body weight (kg)a 92.9 (25.2) 96.9 (24.9) 96.1 (24.5) 95.8 (24.8) % BSAa 22.1 (13.4) 24.7 (13.9) 24.9 (17.8) 24.2 (15.5) PGA, n (%) Moderate 34 (47.2) 69 (49.6) 63 (45.3) 166 (47.4) Severe 35 (48.6) 63 (45.3) 67 (48.2) 165 (47.1) Very Severe 3 (4.2) 7 (5.0) 9 (6.5) 19 (5.4) PASI score 18.3 (6.4) 18.5 (6.0) 19.4 (7.9) 18.8 (6.9) Previous medical history, n (%) Any CVD 26(36.1) 56 (40.3) 52(37.4) 134 (38.3) Hyperlipidemia 6 (8.3) 8 (5.8) 15 (10.8) 29 (8.3) Diabetes mellitus 7 (9.7) 13 (9.4) 9 (6.5) 29 (8.3) PGA = Physician's Global Assessment PASI = Psoriasis Area and Severity Index BSA = Body Surface Area aMean (SD) bAny patient who reported 2 or more diagnoses in the same body system were only counted once in body system total Efficacy e A statistically significant greater percentage of patients in the ABT-874 treatment group (72.7%) achieved a PGA of 0/1 at Week 12 as compared with patients receiving placebo (4.2%) or etanercept (29.5%), P<0.001 (Figure 28) " A statistically significantly greater percentage of patients in the ABT-874 treatment group (80.6%) achieved a PASI 75 response at Week 12 as compared 218 with patients receiving placebo (6.9%) or etanercept (39.6%) [P<0.001] [Figure 29] At Week 12, PASI 90 and PASI 100 responses were achieved by a statistically significantly greater percentage of patients in the ABT-874 treatment group (55.4% and 28.8%, respectively) as compared with patients receiving placebo (4.2% and 0, respectively) or etanercept (13.7% and 5.8%, respectively) [Figures 30 and 3.1] Safety e A higher percentage of patients in the ABT-874 and etanercept treatment groups experienced adverse events than patients in the placebo treatment group, respectively (50.4%, 49.6% and 44.4%, respectively) [Table 26] " Treatment discontinuation was low in both etanercept and ABT-874 treatment groups with 2.9% (4/139) discontinuing in each group (Table 26) e Serious adverse events were reported in 2 (2.8%) patients receiving placebo (coronary artery disease, psoriasis), 1 (0.7%) patient receiving etanercept (breast cancer), and 2 (1.4%) patients receiving ABT-874 (colon cancer, convulsion) (Table 26) e No deaths occurred during the study (Table 26) 219 Table 26. Overview of Treatment-Emergent Adverse Events (AEs) Treatment Group, n (%) Placebo Etanercept ABT-874 N=72 N=139 N=139 Any AE 32 (44.4) 69 (49.6) 70 (50.4) Any severe AE 2 (2.8) 1(0.7) 2 (1.4) Any serious AE 2 (2.8) 1(0.7) 2 (1.4) Any AE leading to discontinuation of study drug 2(2.8) 4(2.9) 4(2.9) Deaths 0 0 0 * The incidence of adverse events of infection including serious adverse events of infection was comparable between the ABT-874 treatment group and the etanercept treatment group (Table 27) * Eight patients experienced malignancies: three in the ABT-874 treatment group (colon, basal cell skin, squamous cell skin), four in the etanercept treatment group (2 basal cell skin, I squamous cell skin, breast) and I melanoma in the placebo treatment group (Table 27) e Eight patients reported ischaemic heart disease related events. Seven of these events were increased creatine phosphokinase and one event of coronary heart disease in a placebo patient * No major adverse cardiac events (MACE), defined as myocardial infarction, stroke, or cardiovascular death, were reported in any of the treatment groups (Table 27) 220 Table 27. Treatment-Emergent Adverse Events of Interest Treatment Group Placebo Etanercept ABT-874 N=72 N=139 N=139 n (%) n (%) n (%) Any infection 10 (13.9) 39 (28.1) 34 (24.5) Any serious infection 0 0 0 Any opportunistic infection 0 0 0 Any malignancy 1 (1.4) 4 (2.9) 3 (2.2) Squamous cell carcinoma 0 1 (0.7) 1 (0.7)a Basal cell carcinoma 0 2 1 (0.7) Other 1 ( 1
.
4 )' 1 (0.7)c 1 ( 0
.
7 )d Any injection site reaction 3 (4.2) 12 (8.6) 5 (3.6) Any nervous system disorder 0 0 1 (0.7)e Any MACEf 0 0 0 aOne patient diagnosed with lip neoplasm, malignant stage unspecified, on study day 92 bOne patient diagnosed with melanoma on study day 30 cOne patient diagnosed with breast cancer in situ on study day 110 dOne patient diagnosed with colon cancer on study day 66 "One patient diagnosed with convulsion on study day 73 Major Adverse Cardiac Event, defined as myocardial infarction, stroke, or cardiovascular death e The most frequently reported treatment-emergent adverse events occurring in at least 5% of patients in both the ABT-874 and etanercept treatment groups were upper respiratory tract infection (7.2% and 11.5%, respectively) and nasopharyngitis (7.2% and 7.9%, respectively) (Table 28) 221 Table 28. Treatment-Emergent Adverse Events Occurring in >5% of Patients in Any Treatment Group Treatment. Group Placebo Etanercept ABT-874 N=72 N=139 N=139 n (%) n (%) n (%) Upper respiratory tract infection 0 16 (11.5) 10 (7.2) Nasopharyngitis 6 (8.3) 11(7.9) 10 (7.2) 'P = 0.017, ABT-874 vs. placebo CONCLUSIONS " ABT-874 was superior to both placebo and etanercept as demonstrated by the statistically significant result of the two co-primary endpoints: PGA 0/1 (P<0.00l) and PASI 75 (P<0.001) at 12 weeks " In this study population, no deaths, no MACE, and a comparably low incidence of serious infections and malignancies were reported across treatment groups Example 14. Efficacy and Safety of ABT-874 Versus Etanercept and Placebo in Patients With Moderate to Severe Psoriasis: The VERTO Study Objective The aim of the VERTO study was to assess the efficacy, safety, and tolerability of ABT 874 compared with etanercept and placebo for the treatment of moderate to severe chronic plaque psoriasis over 12 weeks Methods Main Inclusion Criteria e Males and females >18 years with a clinical diagnosis of chronic plaque psoriasis for at least 6 months 222 " Stable plaque Ps for at least 2 months before Screening and at Baseline (Week 0) visits e Affected body surface area (BSA) >10% " Physician's Global Assessment (PGA) of at least moderate (? 3) and Psoriasis Area and Severity Index (PASI) score of > 12 at the Baseline (Week 0) visit Main Exclusion Criteria e Previous exposure to systemic anti-IL-12/23p40 therapy, including ABT-874 " Previous exposure to ETN or known hypersensitivity to ETN " Inability to discontinue topical therapies, phototherapies, or systemic therapies Study Design (Figure 32) " 12-week double-blind, double dummy, multicenter, randomized study " Patients were randomized to I of the 3 following treatment arms (2:2:1); "a" indicates the second dose of the week when doses were administered twice weekly: * 200 mg ABT-874 SC at Weeks 0 and 4 followed by 100 mg ABT-874 SC at Week 8 * 50 mg of ETN SC twice weekly 3-4 days apart at Weeks 0, Oa, 1, Ia, 2, 2a, 3, 3a, 4, 4a, 5, 5a, 6, 6a, 7, 7a, 8, 8a, 9, 9a, 10, 10a, 11, and 11 a * Placebo SC at Week 0 and 4 followed by placebo SC at Week 8 to match ABT-874 dosing, or placebo SC twice weekly 3-4 days apart at Weeks 0, Oa, 1, Ia, 2, 2a, 3, 3a, 4, 4a, 5, 5a, 6, 6a, 7, 7a, 8, 8a, 9, 9a, 10, 10a, 11, and 1 la to match ETN dosing Efficacy Measures e Proportion of patients achieving a PGA score of "Clear" or "Minimal" (0/1) at Week 12 * Proportion of patients achieving a PASI 75/90/100 response at Week 12 Safety Measures e Adverse events and serious adverse events (SAE) were assessed throughout the study 223 Statistical Methods " The efficacy analyses were conducted in the intent-to-treat (ITT) population; non responder imputation (NRI) was used to handle missing data e Cochran-Mantel-Haenszel tests stratified by pooled center were used to compare the proportion of patients in each treatment group achieving PGA 0/1 or PASI 75 at Week 12 " A Chi-Square test, or Fisher's exact test as appropriate, was used to compare proportions of patients in each treatment group achieving PASI 90 or 100 at Week 12 " All statistical tests were two-sided with the significance level of 0.05 e The safety analyses were conducted in the safety population; safety variables were summarized by treatment group Results A total of 347 patients were enrolled in VERTO. Baseline demographic and clinical characteristics were similar across treatment groups (Table 29).
224 E C6, co c) 00 ~ Lo o6 't - omtC - . j.C \J co 0) a, ( - C.) c 0 LOCOC aCiC o4N C I Z C~ 0) .- CJC r-- nct 0C0 OC 00 CDC)V r-u -O 0 :3 T-C)- iz - O E;u \ I -U r-. D- Cd) CE 0 r (D r, cjo 0 r- co aj - r c- CD ) E a)Cj0 o (D-CJ--- D~\J ~ 'L 0C.CJ ('J C'J ccLOO c U c ocq~ - - LO- 0) 70 E C 0 C~j 4- C_ 2 - * a c: -a co .( 0- Z V ~ ~ . E ~0 ~< 0a 0. (n a -0 CD co ~ ~ ~ ~ .0C D D - 225 Efficacy e A statistically significantly greater percentage of patients in the ABT-874 treatment group (71.0%) achieved a PGA of 0/1 at Week 12 as compared with patients receiving placebo (2.9%) or ETN (39.7%; Figure 33) " A statistically significantly greater percentage of patients in the ABT-874 treatment group (81.9%) achieved a PASI 75 response at Week 12 as-compared with patients receiving placebo (7.4%) or ETN (56.0%; Figure 34) e At Week 12, PASI 90 and PASI 100 responses were achieved by a statistically significantly greater percentage of patients in the ABT-874 treatment group (59.4% and 37.0%. respectively) as compared with patients receiving placebo (1.5% and 0%, respectively) or ETN (23.4% and 7.1%, respectively; Figures 35 and 36) Safety e A slightly higher percentage of patients receiving ETN or ABT-874 experienced adverse events (AE) as compared with patients receiving placebo; however, the safety profile for the two active treatments was similar (Table 30) e Serious adverse events were reported in 4 (2.9%) patients receiving ABT-874 (viral infection, melanoma in situ, anxiety/pain, lumbar vertebral fracture), 1 (0.7%),patient receiving ETN ( skin infection), and 1 (1.5%) placebo patient (hip fracture; Table 30) e No deaths occurred during the study (Table 30) Table 30. Overview of Treatment-Emergent Adverse Events Treatment Group, n (%) Placebo ETN ABT-874 (N=68) (N=1 41) (N=1 38) Any AE 31 (45.6) 76 (53.9) 68 (49.3) Any severe AE 1 (1.5) 3(2.1) 6(4.3) Any serious AE 1 (1.5) 1 (0.7) 4(2.9) Any AE leading to discontinuation of study drug 0 4 (2.8) 4 (2.9) Deaths 0 0 0 * The percentage of patients with adverse events of infection including serious adverse events of infection was comparable across treatment groups (Table 31) 226 e One malignancy each was reported in the ETN and ABT-874 groups; one patient receiving ETN was diagnosed with a basal cell carcinoma on study day 84, and one ABT-874 treated patient was diagnosed with malignant melanoma in situ on study day 29 (Table 31) e Three incidences of ischemic heart disease AE were reported in the ETN group; 4 incidences were reported for ABT-874 treated patients e No major adverse cardiac events (MACE), defined as myocardial infarction, stroke, or cardiovascular death, were reported in any of the treatment groups (Table 31) Table 31. Treatment-Emergent Adverse Events of Interest Treatment Group, n (%) Placebo ETN ABT-874 (N=68) (N=141) (N=138) Any infection 13 (19.1) 34 (24.1) 31 (22.5) Any serious infection 0 1 (0.7)a 1 ( 0
.
7 )b Any opportunistic infection 0 0 0 Tuberculosis 0 0 0 Any malignancy 0 1 (0.7)c 1 (0.
7 )d Non-melanoma skin cancer 0 1 (0.7) 0 MACE 0 0 0 aSkin infection. bViral infection. cPatient was diagnosed with basal cell carcinoma on study day 84. dPatient was diagnosed with malignant melanoma in situ on study day 29. e The most common adverse events occurring in patients receiving ABT-874 or ETN were nasopharyngitis, upper respiratory tract infection, injection site reaction, and headache; the most frequently reported adverse event for placebo patients was upper respiratory tract infection (Table 32) 227 Table 32. Treatment-Emergent Adverse Events Occurring in >5% of Patients in Any Treatment Group Treatment Group, n (%) Placebo ETN ABT-874 (N=68) (N=141) (N=138) Nasopharyngitis 2 (2.9) 11 (7.8) 10 (7.2) Upper respiratory tract infection 6 (8.8) 8.(5.7) 9 (6.5) Injection site reaction 3 (4.4) 13 (9.2) 8 (5.8) Headache 2(2.9) 7(5.0) 2(1.4) Conclusions e In patients with moderate to severe psoriasis, ABT-874 was superior to both placebo and etanercept as demonstrated by the statistically significant result of the 2 co primary endpoints: PGA 0/1 (P<0.001) and PASI 75 (P<0.001) following 12 weeks of treatment " No deaths or MACE events occurred during this study. In addition, no differences were seen across treatment groups for percentage of patients experiencing serious infections or malignancies. Example 15. Efficacy and Safety of ABT-874 Versus Methotrexate in Patients with Moderate to Severe Psoriasis Aims: To compare the efficacy and safety of ABT-874 with that of methotrexate for treatment of moderate to severe psoriasis. Methods: Phase III, 52-week, double-blind, active-controlled trial (NCT00679731). Patients were randomized 1:1 to: ABT-874 (200 mg at Weeks 0 and 4, followed by 100 mg ABT-874 every 4 weeks for Weeks 8-48) or methotrexate (5-25 mg weekly). Non responding patients (patients with psoriasis area and severity index (PASI) <75 and Physician's Global Assessment (PGA) >0/1 at Week 24 or PASI <50 and PGA >3 after Week 24) were discontinued from the study. The four primary endpoints were percentage of patients with: PASI 75 at Week 24, PGA of 0 or 1 at Week 24, PASI 75 at 228 Week 52, and PGA score of 0 or 1 at Week 52. Safety assessments were made throughout the study. Non-responder imputation (NRI) was used to handle missing data. Results: 317 patients were enrolled in the study; 68.8% of ABT-874-treated patients completed the study compared with 27.6% of methotrexate-treated patients. At Week 24, PASI 75 was achieved by 81.8% of ABT-874-treated patients vs. 39.9% of methotrexate treated patients (P<0.001) and a PGA of 0 or 1 was achieved by 80.5% of ABT-874 patients vs. 34.4% of methotrexate patients (P<0.001). At Week 52, 66.2% of ABT-874 patients achieved PASI 75 vs. 23.9% of methotrexate patients (P<0.001), and 63.0% of ABT-874 patients vs. 20.2% of methotrexate patients achieved a PGA of 0 or 1 (P<0.001). Numbers of patients with serious adverse events or discontinuing due to adverse events were similar for both treatment groups. Conclusions: At 24 and 52 weeks, ABT-874 was superior to methotrexate with respect to the primary endpoints of PASI 75 and PGA 0/1 in patients with moderate to severe psoriasis. Example 16. ABT-874 Versus Etanercept or Placebo Treatment for Moderate to Severe Psoriasis: Health-Related Quality of Life Outcomes Aims: Assess effects of treatment with ABT-874 vs. etanercept or placebo on health related quality of life (HRQOL) in patients with psoriasis. Methods: In this 12-week double-blind comparative study, patients were randomized to treatment with ABT-874 (200 mg at Weeks 0 and 4, 100 mg at Week 8), etanercept (50 mg twice weekly), or placebo. HRQOL outcomes included the Dermatology Life Quality Index (DLQI), visual analog scales for Ps-related (VAS-Ps) and psoriatic arthritis-related (VAS-PsA) pain, and Short Form 36 Health Survey Mental (MCS) and Physical (PCS) Component Summary scores. Lower DLQI and VAS scores and higher MCS and PCS scores indicated better outcomes. The last-observation-carried-forward method was used for missing values. Treatments were compared using analyses of covariance for mean 229 changes from baseline to Week 12. Percentages of patients with improvement at or above the minimum clinically important difference (MCID response) were compared with chi squared tests. Results: Treatment with ABT-874 (N=138) was associated with significantly (p < 0.05) greater mean improvements than etanercept (N=141) or placebo (N=68) in DLQI (-10.3 vs. -8.1 or -3.0), VAS-Ps (-29.1 vs.-24.0 or -6.1) and MCS (5.4 vs. 3.2 or 1.0), and vs. placebo only in PCS (4.6 vs. -0.3) and VAS-PsA (-23.5 vs. -7.2). Significantly greater MCID response rates were observed with ABT-874 vs. etanercept or placebo for MCS (43.4 vs. 30.6 or 20.6%) and vs. placebo only for DLQI (81.5 vs. 38.1%), PCS (45.0 vs. 28.6%), VAS-PsA (69.2 vs. 28.6%), and VAS-Ps (56.6 vs. 25.8%). Conclusions: ABT-874 demonstrated significantly greater improvements in all Health Related Quality of Life outcome measurements vs. placebo and in DLQI, VAS-Ps and MCS vs. etanercept. Significantly more patients achieved clinically meaningful improvement with ABT-874 in all outcome measurements vs. placebo and in MCS vs. etanercept. Example 17. Effects of ABT-874 Versus Etanercept or Placebo on Health-Related Quality of Life in Patients With Moderate to Severe Psoriasis Aims: Assess effects of ABT-874 vs. etanercept or placebo on health-related quality of life (HRQOL) in psoriasis patients. Methods: Patients were randomized to 12-week double-blind treatment with ABT-874 (200 mg at Weeks 0 and 4, 100 mg at Week 8), etanercept (50 mg twice weekly) or placebo. Assessments included the Dermatology Life Quality Index (DLQI), visual analog scales for psoriasis-related (VAS-Ps) and psoriatic arthritis-related (VAS-PsA) pain, and the Short Form 36 Health Survey Mental (MCS) and Physical (PCS) Component Summary scores. The last-observation-carried-forward method was used for missing values. Analyses of covariance compared treatments in -mean changes at Week 230 12 from baseline. Chi-square tests compared treatments in percentages of patients with improvement at or above the minimum clinically important difference (MCID response). Results: ABT-874 treatment (N=139) had significantly (p<0.05) greater improvement than placebo (N=72) for all outcomes (DLQI, 11.1 vs. 3.0; PCS, 4.6 vs. 1.1; MCS, 6.3 vs. 2.1; VAS-Ps, 36.0 vs. 7.5; VAS-PsA, 38.6 vs. 3.2). ABT-874 was associated with significantly greater improvement than etanercept (N= 139) for DLQI (11.1 vs. 9.0), VAS-Ps (36.0 vs. 29.4), and MCS (6.3 vs. 3.9). MCID response rates for VAS-Ps were significantly greater for ABT-874 than etanercept or placebo (73.9% vs. 58.0% vs. 29.6%). MCID response rates for DLQI, MCS, PCS, and VAS-PsA were greater with ABT-874 vs. etanercept or placebo, but the differences were significant only vs. placebo (DLQI, 81.8% vs. 32.9%; MCS, 45.6% vs. 30.4%; PCS, 55.9% vs. 34.8%; VAS-PsA, 83.9% vs. 20.0%). Conclusions: Compared with placebo, ABT-874 was associated with significantly greater mean improvements and MCID response rates in all HRQOL measurements. Compared with etanercept, ABT-874 had significantly greater improvement in DLQI, VAS-Ps, and MCS and greater MCID response rates for VAS-Ps. Example 18. Psoriasis Treatment With ABT-874: Effects on Health-Related Quality of Life and Work Productivity and Activity Impairment Aims: Evaluate effects of ABT-874 on health-related quality of life (HRQOL), work productivity and activity impairment (WPAI) in psoriasis patients. Methods: Patients were randomized to receive induction-phase ABT-874 (200 mg at Weeks 0 and 4, 100 mg at Week 8) or placebo. Patients achieving a Week-12 Physician's Global Assessment of "clear/minimal" were re-randomized to maintenance-phase ABT 874 (100 mg every 4 [q4] or 12 [ql2] weeks) or placebo up to Week 52. HRQOL outcomes included the Dermatology Life Quality Index (DLQI), visual analog scales for plaque psoriasis- and psoriatic arthritis-related pain, and Short Form 36 Health Survey 231 Mental and Physical Component Summary scores. Total activity impairment (TAI) was assessed by WPAI-SHP: PSO questionnaire. Last-observation-carried-forward method was used for missing values. Analysis of covariance compared mean improvements from baseline to Weeks 12 and 52 between treatments. Chi-squared tests compared percentages of patients with improvement at or above the minimum clinically important difference (MCID response). Results: ABT-874 treatment (N=98 1) had significantly (p<0.05) greater Week- 12 MCID response rates than placebo (N=484) for all outcomes (e.g., DLQI, 78.1 % vs. 19.4%; TAI, 51.4% vs. 15.3%). After re-randomization, Week-52 MCID response rates were significantly greater for ABT-874 q4 (N=297) than for q12 (N=298) in all outcomes except for psoriatic arthritis-related pain. Both ABT-874 groups had significantly greater MCID response rates than placebo (N=149) for all outcomes (e.g., DLQI, 81.1% vs. 71.1% vs. 49.3%; TAI, 56.9% vs. 45.1% vs. 31.0%). Mean improvements in all outcomes were significantly greater for ABT-874 than placebo at Week 12 and for ABT-874 q4 than ABT-874 q 12 or placebo at Week 52. Conclusion: ABT-874 treatment for psoriasis versus placebo was associated with significantly greater improvements in HRQOL and reduced TAI in both induction and maintenance phases. Example 19. Responses to ABT-874 Across Subgroups of Patients with Moderate to Severe Psoriasis Aims: To evaluate the efficacy of ABT-874 in subgroups of patients with moderate to severe psoriasis. Methods: In a 52-week, double-blind study patients were randomized to ABT-874 (200 mg, weeks-0 and 4; 100 mg, week-8) or placebo (PCB); then, if PGA "clear/minimal" (PGA 0/1) was achieved at week 12, re-randomized to ABT-874 100 mg every 4 weeks (q4wk), 100 mg every 12 weeks (ql2wk), or PCB q4wk (NCT00570986). Primary 232 endpoints were PGA 0/1 (weeks-12 and 52) and PASI 75 (week-12). NRI analysis of endpoints was conducted by baseline characteristics. Safety was assessed throughout the study. Results: 1,465 patients were randomized at baseline (mean PASI: 19.2 ±7.4; ABT-874, N=98 1; PCB, N=484). Responses at week-12 in patients with/without prior biologics, for ABT-874 (n=173/808) vs PCB (n=76/408) were, PGA 0/1: 65.9%/78.2% vs 1.3%/4.9%; and PASI 75: 74.6%/82.1% vs 1.3/5.1%; responses at week-52 for ABT-874-q4wk (n=46/252) vs -ql2wk (n=49/249) were, PGA 0/1: 78.3%/79.4% vs 34.7%/43.0%. For patients with/without a history of PsA, week- 12 responses for ABT-874 (n=290/69 1) vs PCB (n=150/334) were, PGA 0/1: 71.7%/77.9% vs 0.7%/6.0%; and PASI 75: 78.6%/81.6% vs 1.3%/6.0%; responses at week-52 for ABT-874-q4wk (n=86/212) vs -q12wk (n=79/219) were, PGA 0/1: 77.9%/79.7% vs 30.4%/45.7%. More infections and malignancies occurred in patients receiving ABT-874 vs PCB up to week-12 (22.3% vs. 19.8% and 0.6% vs. 0, respectively). Major adverse cardiac events (MACE) occurred in 7 ABT-874-treated patients (none had prior biologics; 1 had PsA; mean baseline PASI: 15.8 ±3.2). No MACE occurred in PCB-treated patients. Conclusions: High percentages of ABT-874-treated patients achieved PGA 0/1 (weeks-12 and 52) and PASI 75 (week-12), regardless of prior biologic treatment or history of PsA. More infections, malignancies and MACE occurred in ABT-874 vs PCB treated patients, indicating the importance of close surveillance for these events. Example 20. Long-term Safety of ABT-874 for the Treatment of Moderate to Severe Psoriasis- Interim Analysis from an Open-label Extension Study Introduction: Interim results from an ongoing, open-label extension study (OLE) of the anti-IL-12/23 agent, ABT-874, provide preliminary, long-term safety data. Methods: Patients randomized to ABT-874 in a prior phase 2 or 3 psoriasis trial are eligible for this 160-week, multi-center OLE upon completion or loss of response in the respective prior study. Patients receive 100 mg ABT-874 every 4 weeks, starting at OLE.
233 baseline. This analysis includes all patients with >1 dose of ABT-874 during the OLE (safety set). Adverse events (AEs) from the first dose of ABT-874, whether from OLE or preceding study, and ;45 days following the last dose of study drug are recorded. This interim analysis provides data available through November 26, 2009. Results: As of the cutoff date, the safety set consisted of 2,298 patients (2904.0 PY drug exposure, 421.7 ±186.8 mean days on treatment; 68.8% male; mean age, 45.2 ±13.2 years). 2.7% have withdrawn due to AEs; 89.1% are still enrolled. AEs occurring in >5% of patients were: URI (13.6%), nasopharyngitis (13.4%), headache (6.7%), arthralgia (5.8%), and hypertension (5.2%). Infectious AEs occurred in 45.5%; serious infections in 1.0% (most common: cellulitis, n=8; pneumonia, n=5; sepsis, n=4; and diverticulitis, n=3); and opportunistic infections in 0.3% (candidiasis, n=3; oral candidiasis, n=1; esophageal candidiasis, n=1; and coccidiomycosis, n=1). 1.6% of patients had malignancies; 1.2% had NMSCs (BCC, n=15; SCC, n=16); 1 melanoma in-situ occurred. No lymphomas were observed. 18 major adverse cardiovascular events (MACE) occurred (7 during preceding study, 11 during OLE; myocardial infarction, n=1 1; stroke, n=3; and cardiovascular death, n=4; 72.2% male; 88.9% age ?45 years; all had :2 CAD risks; mean time to OLE events 31.5 weeks). Conclusions: An OLE interim analysis supports the need to closely monitor AEs of infection, NMSC, and MACE in patients receiving ABT-874 for the treatment of moderate to severe psoriasis. Example 21. A Pooled Analysis of Phase III, Randomized, Placebo-Controlled Clinical Trials of the Anti-Interleukin 12/23 Monoclonal Antibody, ABT-874 Objective: To determine the efficacy and safety experience in moderate to severe psoriasis across ABT-874 phase III clinical trials. Methods: Data from 3 randomized, placebo-controlled studies were pooled through 12 weeks. In all studies, patients with moderate to severe psoriasis were randomized 2:1 to ABT-874 (200 mg at weeks 0 and 4, followed by 100 mg at week 8) or matching 234 placebo. The 3 trials measured common primary endpoints of physician's global assessment "clear" or "minimal" scores (PGA 0/1), and 75% improvement from baseline psoriasis area and severity index (PASI 75) at week 12 (each analyzed using nonresponder imputation). Secondary endpoints included PASI 90/100, and-DLQI. Adverse events were collected during each study and up to 45 days from the last dose of study drug. Results: Data were pooled for 1882 patients (ABT-874, N=1258 [67.4% male, 90.3% white, mean [SD] age 45.3 [13.3] yrs; placebo, N=624, with similar demographics). Baseline PASI was similar between groups (19.0 [7.5] and 19.1 [7.2] for ABT-874 and placebo, respectively). 75.1% (945/1258) and 80.8% (1017/1258) of ABT-874-treated patients achieved a PGA 0/1 and PASI 75, respectively, at week 12, compared with 4.2% (26/624) and 5.1% (32/624) of those receiving placebo (p<0.001 for both endpoints). PASI 90 responses at week 12 were 60.7% (763/1258) vs. 1.8% (11/624) in the ABT-874 vs. placebo groups, respectively; PASI 100 response was 32.4% (407/1258) with ABT 874, and was not achieved in the placebo group (p<0.001 for both endpoints). Baseline DLQI scores were 12.9 [7.0] and 12.8 [6.9] for ABT-874- and placebo-treated patients; at week 12, mean percent change in DLQI for each group was -79.1 [28.9] vs. 1.4 [69.8], respectively. Serious adverse event (AE) rates were 2.1% and 1.4% for ABT-874 vs. placebo. Incidence rates for infectious and serious infectious AEs were 22.6% and 0.5% for ABT-874, and 19.1% and 0.2% for placebo. NMSCs were observed in 6 patients receiving ABT-874; none were observed with placebo. 5 ABT-874-treated patients had major adverse cardiovascular events (MACE); no MACE were observed with placebo. Conclusions: Efficacy results pooled from 3 phase III, placebo-controlled trials in moderate to severe psoriasis demonstrated superior PGA and PASI responses with ABT 874 vs. placebo. Higher rates of infection, NMSC, and MACE occurred with ABT-874 vs. placebo, highlighting the need to monitor for these events in a larger psoriasis population.
235 Example 22. Pooled Safety and Efficacy Results from Two Phase III Trials Comparing BriakinumabTM With Etanercept and Placebo for the Treatment of Moderate to Severe Psoriasis Aims: To provide an integrated analysis of efficacy and safety results from two independent trials comparing briakinumabTM versus etanercept (ETN) and placebo (pbo) for moderate to severe psoriasis treatment. Methods: Efficacy and safety data were pooled from two 12-week, phase III, double dummy, randomized trials (NCT00710580, NCT00691964). Patients were randomized 2:2:1 to receive briakinumabTM (200 mg at Weeks 0 and 4, followed by 100 mg at Week 8), ETN (50 mg twice weekly), or matching pbo. At Week 12, primary efficacy endpoints were percentage of patients achieving a PGA score of "Clear" or "Minimal" (0/1) and percentage of patients achieving a PASI 75 response. Secondary endpoints included Week 12 PASI 90 and PASI 100 response rates. Adverse events (AE) were assessed throughout the study. Non-responder imputation (NRI) was used to handle missing data. Results: Individual trial results demonstrated superior efficacy of briakinumabTM vs. ETN and pbo (P<0.001, briakinumabTM vs. ETN or pbo, both primary endpoints for both studies). For this pooled analysis, data from 697 patients were analyzed: briakinumabTM, N=277; ETN, N=280; pbo, N=140. At Week 12, 71.8%, 34.6%, and 3.6% of briakinumabTM-, ETN-, and pbo-treated patients, respectively, achieved a PGA 0/1 (P<0.001; briakinumabTM vs. ETN or pbo). Week 12 PASI 75 response rates for briakinumabTM-, ETN-, and pbo-treated patients were 81.2%, 47.9%, and 7.1%, respectively (P<0.001, briakinumabTM vs. ETN or pbo). Week 12 PASI 90 and PASI 100 response rates were 57.4%/18.6%/2.9% and 32.9%/6.4%/0.0% for patients treated with briakinumabTM/ETN/pbo, respectively (P<0.001, briakinumabTM vs. ETN or pbo for both PASI 90 and 100). Serious AE rates were: 2.2%/0.7%/2.1% for briakinumabTM/ETN/pbo patients. Serious infection rates were: 0:4%/0.4%/0% for briakinumab T M /ETN/pbo patients. There were no deaths or major adverse cardiovascular events (MACE) in any treatment group.
236 Conclusions: Pooled efficacy results from two independent trials demonstrate that briakinumabTM was superior to ETN and pbo for moderate to severe psoriasis treatment. Serious adverse events were low across all groups and no MACE were observed. Example 23. Long term Safety and Efficacy of ABT-874 for the Treatment of Moderate to Severe Psoriasis - Interim Analysis from an Open Label Extension Study The safety and efficacy of ABT-874 in the treatment of psoriasis were assessed on an interim basis in view of results obtained as of November 26, 2009. Patients were eligible for enrollment following completion or loss of response in a preceding ABT-874 psoriasis phase II or phase III trial. The duration of the study was planned for 160 weeks with treatment comprising 100 mg ABT every 4 weeks beginning at week 0. Preceding phase II or phase III clinical trials include the studies set forth in Table 33: Table 33: Phase II and III studies Study Design Study N OLE N Phase II, 3-period, M05-736 dose-ranging 180 84 study; 12-wk DB, 6-wk obs./re reatment, followed by 60-wk OL re-treatment M06-890 Phase III, 2-period 1465 1346 study; 12-wk Induction Phase, 40-wk Maintenance Phase_ 237 M10-114 Phase III, 12-wk, 347 308 DB, placebo and active comparator (ETN) M10-315 Phase III, 12-wk, 350 314 DB, placebo and active comparator (ETN) M10-255 Phase III, 52-wk, 317 246 DB, active :omparator (MTX) The efficacy of ABT-874 in treatment of moderate to serious psoriasis was demonstrated in these previous studies, as depicted in Figure 37. The data presented in this example represents an interim analysis of results. Maintenance of Efficacy: Patients evaluated included those (i) with at least one dose of ABT-874 during the preceding phase II or III study and in the current Open Lable Extension (OLE) study and (ii) with a physician's global assessment score of "clear" or "minimal" (PGA 0/1) at the last evaluation on or before the first dose in OLE. PGA scores and psoriasis area and severity index (PASI) responses were determined every 12 weeks during the OLE. (Note that patients enrolled from study M06-890 must have received at least I dose of ABT-874 during the Induction Phase). 2298 patients had received at least I dose of ABT-874 in the OLE as of this interim analysis. Specifically, 251 (10.9%) patients had discontinued from the OLE and 61 (2.7%) withdrew due to adverse events. Ultimately, 625 patients met the criteria for the Maintenance of Efficacy population (i.e., patients who had received loading dose in the initial study (200-200-100 mg) and had a PGA-Score of "clear" or "minimal" at entry in the OLE study). The demographics and clinical characteristics of the population were as follows: Table 34: Demographics and Clinical Characteristics of Population at Preceding Study Baseline 238 All briakinumabTM (N = 2298) Male, n (%) 1581 (68.8) White, n (%) 2101 (91.4) Age, yrs, mean (SD) 45.2 (13.2) Weight, kg, mean (SD) 93.1 (23.0) Duration of psoriasis, yrs, mean (SD) 18.6 (12.3) PASI* prior 1st dose, mean (SD) 16.6 (8.8) SA* prior 1st dose, %, mean (SD) 24.8 (16.1) * From baseline prior to I" dose of ABT-874 (whether in preceding study or OLE). Results of the Maintenance of Efficacy interim analysis are depicted in Figures 38 and 39. Specifically, Figure 38 depicts the percentage of patients who have maintained PASI 75 over time while Figure 39 depicts the percentage of patients who have maintained PASI 100 over time. These results suggest that high levels of PASI and PGA response as achieved in the preceding phase II and III studies were generally maintained upon further treatment with 100 mg every four weeks. Safety Analysis: Patients evaluated included those (i) with at least one dose of ABT-874 in the current Open Lable Extension (OLE) study and (ii) with adverse effects from the first dose of ABT-874 received either in the preceding studies or the OLE. Safety was assessed throughout the studies and up to 45 days from the last dose study drug. The following table summarizes the occurrences of particular adverse effects: Table 35: Interim Safety Results BriakinumabTM Patients with events (%) Events (E/100 (N=2298) PYs) (PYs=2904) Any AE 1673(72.8) 7177 (247.1) Most common AEs* Upper resp. tract 313 (13.6) 426 (14.7) 239 infections _____________ ________ Nasopharyngitis 309 (13.4) 456 (15.7) Headache 154 (6.7) 220 (7.6) Arthralgia 133 (5.8) 146 (5.0) Hypertension 120 (5.2) 126 (4.3) Any infection 1045 (45.5) 2002 (68.9) Any serious infection 24 (1.0) 30(1.0) Opportunistic infections 6 (0.3) 6 (0.2) Malignancy 37 (1.6) 42 (1.4) NMSC 28(1.2) 33(1.1) Lymphoma 0 0 Accordingly, patients receiving ABT-874 for treatment of moderate to severe psoriasis should be monitored for certain adverse events, in particular, infection, non-melanoma skin cancer (NMSC) and cardiovascular events. MACE Analysis: Patients evaluated included those with at least one dose of ABT-874 in the preceding clinical studies or in the current Open Lable Extension (OLE) study (N=2520; 3010.6 Patient Years). A total of 18 MACE (Major Adverse Cardiovascular Events) were identified. 7 MACE were identified in one randomized controlled preliminary clinical trial including 5 during the initial 12 week placebo controlled treatment period and 2 between week 12 and week 52. In addition 11 MACE were identified in the OLE study. Of the 18 MACE cases, 11 were non-fatal myocardial infarction, 3 were non-fatal cerebrovascular strokes and 4 were cardiovascular deaths. The MACE rates were as depicted in Table 36: Table 36: MACE Rates MACE Rates: MACE Patient MACE/ 95% CI (n) Years 100PYs (PYs) 12-week Pl.-contr. period 5 376.7 1.33 (0.43, 3.10) Al briakinumabTM treatment 18 3010.6 0.60 (0.35, 0.94) eriods* 240 * Cut-Off: November 26, 2009 The frequency of MACE appeared to be equally distributed over time as depicted in Figure 40. Accordingly, no association with the duration of exposure to ABT-874 was noted. A further risk analysis was performed whereby the standard cardiovascular risk factors were analyzed by univariate analyses in patients with at least one dose of ABT 874 in a prior phase III or phase II study or in the OLE. The standard cardiovascular risk factors include body mass index (BMI), HDL-cholesterol, LDL-cholesterol, triglycerides, systolic/ diastolic blood pressure, history of hypertension, history of diabetes, history of cardiovascular disease (CVD), current cigarette smoking and age. As set forth in Table 37, the 18 subjects experiencing MACE had the following risk factors: Table 37: Percentages of Patients with MACE Having Cardiovascular Risk Factors Cardiovascular n (%) Risk BMI>30kg/m 2 16 (88.9) BL HDL<40mg/dL 8 (44.4) BL LDL>I00mg/dL 8 (44.4) BL TG>200mg/dL 7 (38.9) BL Glc>126 mg/dL 8 (44.4) Current Smoker 4 (22.2) Male Sex 13 (72.2) M>45yrs/F 55yrs 16 (88.9) Hypertension 12 (66.7) Diabetes Mellitus 4 (22.2) 241 Four specific cardiovascular risk factors were identified to be predictive for MACE including (1) history of diabetes; (2) a body mass index of at least 30 or higher; (3) inadequate blood pressure control (blood pressure of at least 140/90 or higher; and (4) a history of cardiovascular disease defined as at least one of (i) myocardial infarction; (ii) angina requiring hospitalization; (iii) coronary artery disease requiring revascularization; (iv) peripheral artery disease; (v) congestive heart failure requiring hospitalization; (vi) stroke or (vii) transient ischemic attack. In conclusion, as set forth in Table 38, the analysis showed that the rate of MACE was higher in patients with at least 2 cardiovascular risk factors as compared to patients having only 0 or 1 cardiovascular risk factors. Table 38: Risk of MACE by Number of Risk Factors Number MACE N Proportion MACE PY E/1OOPY of Risk (points) (Event) Factors* 0 or 1 3 1937 0.15% 3 2314.5 0.13 2 to 4 15 583 2.57% 15 696.1 2.15 Overall 18 2520 0.71% 18 3010.6 0.60 * Hx Diabetes, Hx CVD, Uncontrolled BP (?140/90) at Baseline, BMI ? 30 The data confirms that, in patients with one or no cardiovascular risk factors, the rate of MACE may be significantly reduced upon administration of ABT-874. Example 24. A phase III, randomized, placebo-controlled trial evaluating the efficacy and safety of two dosing regimens of the fully human interleukin-12/23 monoclonal antibody, briakinumabTM, for the treatment of moderate to severe psoriasis Introduction A previous phase II study demonstrated that briakinumabTM, a fully human anti IL12/23 monoclonal antibody, was efficacious for the treatment of moderate to severe psoriasis (see, e.g., Examples 1-11 above) (Kimball AB, et al., Arch Dermatol 2008; 144: 242 200-7. The current study further evaluated the efficacy and safety of briakinumabTM, and compared two maintenance therapy dosing regimens against placebo. Findings from this study offer expanded insights on the benefit-risk profile for anti-IL-12/23 treatment in moderate to severe psoriasis. Methods Study design, ethics, and participants . A 52-week, randomized, double-blind, placebo-controlled trial was conducted in two phases: a 12-week induction phase and a maintenance phase, during which treatment continued through 52 weeks (Figure 41). At baseline, patients were randomized 2:1 to receive briakinumabTM or placebo. Treatment began at week 0 with a 200 mg subcutaneous dose of briakinumabTM, followed by 200 mg at week 4, and 100 mg at week 8; or matched placebo at weeks 0, 4 and 8. At week 12, patients achieving a physician's global assessment (PGA) score of "clear" or "minimal" were re-randomized 2:2:1 to receive one of three maintenance regimens: briakinumabTM 100 mg every 4 weeks, 100 mg briakinumabTM every 12 weeks, or placebo every 4 weeks. The study protocol and patient informed consent were approved by an Independent Ethics Committee or Institutional Review Board at each participating study site, and informed consent was signed by all patients prior to initiating any study-related procedures. Patients were recruited from 116 clinical sites within the United States and Canada. Adults, aged 18 and over, were eligible to participate if they had'moderate to severe plaque-type psoriasis (defined as >10% affected body surface area, PGA score of at least "moderate", and Psoriasis Area and Severity Index (PASI) of 212) for at least six months, and were candidates for systemic therapy or phototherapy. Women of childbearing potential were required to use at least two methods of contraception throughout the study and for 60 days following the last dose of study drug; women who were pregnant or breastfeeding were excluded from participation. Other reasons for exclusion included previous exposure to an IL- 12 inhibitor, or any active skin condition that would interfere with the evaluation of psoriasis. Patients were not enrolled if they had any poorly controlled medical condition, or any hepatic, renal, or hematologic disease. Patients with infections, or risk for severe infections, were also excluded from the study. Tuberculosis (TB) screening was conducted on all patients prior to enrollment.
243 Those with a purified protein derivative test result of >5 mm induration (regardless of prior BCG vaccination), and/or chest x-ray findings suggestive of latent TB were not enrolled, unless prophylactic treatment as recommended by local guidelines was instituted prior to initiating study treatment. Patients with a history of malignancy, other than a successfully treated basal cell or non-metastatic squamous cell skin cancer, or cervical carcinoma in situ, were not enrolled. Use of the following therapies was not permitted during the study, and must have been discontinued within the respective timeframes prior to baseline: topical treatments, including corticosteroids, vitamin D analogs, or retinoids within 2 weeks (other than low potency topical corticosteroids on the palms, soles, face, inframammary area, or groin); UVB phototherapy within 2 weeks; psoralen and UVA phototherapy within 4 weeks; systemic nonbiologic therapies within 4 weeks; or biologic therapies within 12 weeks. Any use of oral or injectable corticosteroids was not permitted; however, inhaled corticosteroids could be continued for stable medical conditions. Study objectives and outcomes The study was conducted to evaluate the efficacy and safety of briakinumabTM for the treatment of moderate to severe psoriasis, and to compare two maintenance dosing regimens against placebo. Three primary endpoints were measured: PGA score of "clear" or "minimal" at weeks 12 and 52, and at least 75% improvement from baseline PASI ( PASI 75) at week 12. Statistical comparisons were ranked in the following order: 1) week 12 PGA "clear" or "minimal" response rates for briakinumabTM vs placebo; followed by 2) comparison of week 12 PASI 75 response rates between the two treatment groups; and then 3) week 52 PGA "clear" or "minimal" maintenance rates for the briakinumabTM every 4 week group vs placebo; followed by 4) week 52 PGA "clear" or "minimal" maintenance rates for the briakinumabTM every 12 week group vs placebo. Secondary efficacy analyses compared the proportions of patients achieving PASI 75, and 90% and 100% improvement from baseline PASI (PASI 90 and PASI 100) over time, change in PASI relative to baseline, and Dermatology Life Quality Index (DLQI) scores over time. PASI 100, PASI 90, and change from baseline DLQI at week 12 were ranked secondary endpoints; while mean PASI scores and PASI response rates over time 244 were non-ranked. Efficacy outcomes were compared between each briakinumabTM dosing group vs placebo, and between the two briakinumabTM dosing groups. An additional analysis evaluated PGA "clear" or "minimal" scores, and PASI 75 responses by the following subgroups: with or without prior biologic treatment; with or without psoriatic arthritis (PsA); baseline PASI score of >20 or 20; and baseline weight >100 kg or <100 kg. The prevalence of any cardiovascular medical history or underlying cardiovascular risk factors was determined post hoc, by treatment group. Patients were assessed for adverse events throughout the study and for 45 days following the last dose of study treatment. Statistical analyses It was estimated a priori that a sample size of 1350 subjects (900 receiving briakinumabTM, and 450 receiving placebo) would provide >90% power to demonstrate the superiority of briakinumabTM over placebo (with superiority defined as a PGA score of "clear" or "minimal" in 70% vs. 4% of briakinumabTM vs. placebo treated patients at week 12, respectively). The sample sizes needed in each treatment group after week 12 were determined assuming that approximately 630 patients would be re-randomized, and that 40% of patients receiving placebo would maintain a PGA score of "clear" or "minimal" up to week 52. It was established that re-randomization would occur at week 12 in a 2:2:1 fashion, and that 250 patients allocated to each briakinumabTM arm and 125 allocated to placebo would provide 90% power to detect an 18% difference in PGA response rates. A randomization schedule was prepared and maintained by the study sponsor's department of statistics, and administered via an interactive voice response system. Randomization at week 0 was performed by center; and re-randomization at week 12 was stratified by the treatment assigned at week 0. All doses of study drug were provided in a blinded fashion, and assigned treatment groups were concealed from study investigators and site personnel, subjects, and the sponsor's clinical team responsible for conduct of the study. All statistical tests were two-tailed with alpha level set at 0.05. Primary and ranked secondary variables were analyzed in a hierarchical fashion, where each successive comparison was made only if statistical significance was observed with the 245 next highest ranking variable. Efficacy analyses were conducted in the intention-to-treat population, with missing values imputed using nonresponder imputation for categorical variables and last observation carried forward for continuous variables. The primary analyses were conducted using a Cochran-Mantel-Haenszel test, adjusted by center. Chi-square test was used for pair-wise comparisons. Treatment differences for mean change scores were analyzed using ANCOVA, with treatment as a factor and baseline value as a covariate. One subject randomized at week 12 to the every 4 week briakinumabTM arm did not receive study drug during the Maintenance Phase and was therefore not included in the safety analysis. Results Induction phase 1465 patients were randomized (briakinumabTM, n=98 1; placebo, n=484; figure 42). Baseline demographics and clinical characteristics were comparable between the two treatment groups (Table 39), and similar to those observed in other phase III clinical trials for moderate to severe psoriasis (Reich K, et al. Lancet 2005; 366: 1367-74; Menter A, et al. J Am Acad Dermatol 2008; 58: 106-15; Saurat JH, et al. Br J Dermatol 2008; 158: 558-66; Papp KA, et al. Lancet 2008; 371: 1675-84). The percentages of patients with a cardiovascular-related medical history, or risk factors for coronary heart disease (CHD) (Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). Jama 2001; 285: 2486-97) were generally similar between groups (Table 40). Compared with placebo, the percentage of patients achieving a PGA score of "clear" or "minimal" at week 12 was significantly higher with briakinumabTM (76.0% in the briakinumabTM group vs 4.3% in the placebo group, p<0.001; Figure 43). PASI 75 responses were observed in 792 (80.7%) patients receiving briakinumabTM compared to 22 (4.5%) placebo-treated patients (p<0.001; table 41). 61.6% of patients treated with briakinumabTM achieved PASI 90, and 32.2% achieved PASI 100. The response was rapid: a 51.7% reduction in PASI was observed with briakinumabTM by week 4, and 246 improvement continued through week 12, when the mean reduction in PASI was 85.4% (Figure 44). A large reduction in mean DLQI score was also observed in briakinumabTM treated patients, indicating significant benefits in quality of life within 12 weeks. Mean DLQI score for the briakinumabTM group declined from 12.8 (SD 7.02) at baseline to 2.7 (SD 4.12) at week 12, while DLQI scores in the placebo group remained relatively stable over this period (Table 4 1). A subgroup analysis demonstrated that greater than 65% to 70% of patients receiving briakinumabTM achieved a PGA score of "clear" or "minimal" and PASI 75 response at week 12, regardless of prior biologic treatment, baseline PASI score, history of psoriatic arthritis, or baseline weight (Table 41). Maintenance phase After 12 weeks of treatment, 766 patients had attained a PGA of "clear" or "minimal" and entered the Maintenance Phase, a majority of whom were from the briakinumabTM group (n=745; figure 42). For these patients, 596 were re-randomized to continue receiving briakinumabTM at either 4 week (n=298) or 12 week (n=298) intervals, while 149 patients were allocated to placebo and were thus withdrawn from briakinumabTM treatment (withdrawal group). Demographic and clinical characteristics at baseline for these 3 treatment groups were generally similar, and comparable to those observed in the Induction Phase treatment groups (Table 42). This report excludes efficacy results for patients who initially received placebo and were re-randomized in the Maintenance Phase, as the number in this group was very small (n=21). The percentages of patients maintaining a PGA score of "clear" or "minimal" through week 52 were greater for the two briakinumabTM treatment groups than for the treatment withdrawal group, and highest in the every 4 week dosing arm (figure 45 [A]). Of 149 patients in the withdrawal group, 6.0% had a PGA score of "clear" or "minimal" at week 52 compared with 41.6% and 79.2% of 298 patients each in the every 12 week and every 4 week treatment groups, respectively (p<0.001). However, it is notable that high response levels achieved during induction lingered over a prolonged period in the withdrawal group: at week 24, during the Maintenance Phase and 12 weeks following 247 treatment withdrawal, 45.6% of patients had maintained a PGA score of "clear" or "minimal". PASI 75 response rates during the Maintenance Phase were highest among .patients who continued briakinumabTM treatment; 82.6% and 45.6% of patients in the every 4 week and every 12 week dosing groups, respectively, vs 8.7% in the withdrawal group maintained this efficacy response at week 52 (Figure 45B). PASI 100 responses, achieved by a large proportion of patients who had a PGA score of "clear" or "minimal" at re-randomization, generally continued to increase with ongoing treatment through week 32 and, in the high dose arm, did not deteriorate for the remainder of the maintenance phase (Figure 45C). 63.4% and 23.5% in the every 4 week and every 12 week briakinumabTM groups, respectively, had a PASI 100 response at week 52, compared with 4.0% of patients withdrawn from treatment (p<0.001). PASI response rates in patients withdrawn from therapy mirrored PGA results, with a protracted return toward baseline levels of disease. Safety results The most commonly observed adverse events in patients receiving briakinumabTM were nasopharyngitis, headache, upper respiratory tract infection (URI), and back pain (table 43). While back pain was observed in all treatment groups, it was reported more frequently in patients receiving briakinumabTM every 4 weeks vs every 12 weeks, or placebo (5.4% vs 2.0% and 2.0%, respectively; Table 41). Serious adverse events (SAE) were observed in 3.3% of all patients who received briakinumabTM during the Induction and/or Maintenance Phases. SAE rates during the Induction Phase were similar for briakinumabTM vs placebo (2.0% vs 1.2%, respectively). During maintenance treatment, the SAE rate was higher for the every 12 week dosing arm compared with the every 4 week or placebo arms (3.0% vs 1.3% and 1.3%, respectively). The overall rate of infections in patients exposed to briakinumabTM was higher than the infection rate for placebo during the Induction Phase (39.8% of 998 vs 19.8% of 484). Rates of infection during maintenance treatment were higher than those observed during the placebo-controlled period, and highest in the every 4 week dosing group (table 43). The total number of infections per 100 patient-years of briakinumabTM treatment was 248 100.1 (663 events, and 662.2 patient-years of briakinumabTM exposure). Serious infections-were relatively infrequent for all treatment groups, however were observed more often in briakinumabTM- vs placebo-treated patients during the first 12 weeks (0.5% vs 0.2%, respectively). Cellulitis, which was observed in 0.4% of briakinumabTM-treated patients, was the most frequently reported serious infection. There were no serious infections observed beyond week 12 in patients who received every 4 week dosing, and low rates were observed in the every 12 week and treatment withdrawal arms (0.7% and 0.7%, respectively). Opportunistic infections were limited to two nonserious events of oral candidiasis (one patient each in the Induction Phase placebo arm, and the Maintenance Phase every 4 week arm); and one serious event of worsening cytomegalovirus, localized bilaterally in the eyes, in a patient receiving placebo during the Induction Phase. 69 patients initiated treatment for latent TB (briakinumabTM, n=52; placebo, n= 17) prior to beginning study drug. No events of TB infection were observed during the study. Combining the placebo-controlled and maintenance treatment periods, nonmelanoma skin cancers were observed in a total of 10 patients treated with briakinumabTM (six patients with squamous cell carcinoma, and four with basal cell carcinoma; table 43), none were reported in the placebo arm. Four of the events of squamous cell carcinoma were diagnosed within the first 12 weeks of treatment. Major adverse cardiovascular events (MACE), defined as cardiac arrest, myocardial infarction, stroke, or acute coronary syndrome, were observed in seven patients treated with briakinumabTM. No MACE were observed in placebo-treated patients. Five of the seven events occurred during the induction phase, with onset ranging from 21 to 55 days following initiation of study treatment. The two remaining events occurred on days 131 and 225, during maintenance treatment. Of the five events observed during the Induction Phase, one was a cardiac arrest resulting in death, which occurred on day 38 in a 50 year old male patient with several underlying risk factors for CHD (including body mass index of 30 kg/m 2 , baseline triglyceride level >200 mg/dL, and blood glucose of >126 mg/dL). All other events also occurred in patients with 3 or more pre-existing risk factors for CHD. Discussion 249 This large, randomized, placebo-controlled trial in patients with moderate to severe psoriasis demonstrated robust efficacy responses to a treatment modality targeting the IL-12/23 pathways, further validating previous work which has linked these cytokines to the psoriasis disease process. Following 12 weeks of treatment, the primary endpoints of PGA "clear" or "minimal" and PASI 75 were achieved by 76% and 81% of briakinumabTM-treated patients, respectively. Moreover, a 52% mean improvement in PASI was observed by week 4 suggesting a rapid onset of efficacy. The percentage of patients achieving a PASI 100 response continued to rise through 20 weeks of treatment, and this degree of improvement was sustained remarkably well through 52 weeks. For each endpoint, response was maintained best with the every 4 week dosing regimen. The high degree of clinical response which was observed in various subgroups of patients is notable. Because patients who have failed to respond to prior biologic therapy and/or have severe psoriasis are generally more difficult to treat, the fact that most patients meeting these criteria in this study achieved a PGA score of "clear" or "minimal" and PASI 75 response highlights the considerable efficacy benefits observed with briakinumabTM. Likewise, equally high response levels were seen in patients having arthritic involvement, and in those with higher weight, further underscoring the particularly robust effects of this anti-IL- 12/23 agent. Safety findings in this phase III study were generally consistent with preliminary results in an earlier, dose-ranging study (Kimball AB, et al. Arch Dermatol 2008; 144: 200-7), particularly with regard to the observed rates of infection, which were comparable to those reported for ustekinumab, another anti-IL-12/23 agent (Papp KA, et al. Lancet 2008; 371: 1675-84; Leonardi CL, et al. Lancet 2008; 371: 1665-74). In this study, infection rates appeared higher during maintenance treatment; however, this may have been related to the large numbers of patients lost at week 12 as a result of not meeting the efficacy criteria of PGA "clear" or "minimal". Still, it is important to note that a potential increased risk for infection with an immunomodulating treatment is not unexpected, and monitoring for these events during treatment with an anti-IL- 12/23 agent is warranted. Rates of serious infection were low, with the most commonly observed event being cellulitis. It is noteworthy that no TB or serious opportunistic fungal infections 250 occurred in this large study; only a single case of candidiasis, in a patient receiving briakinumabTM every 4 weeks during maintenance treatment was observed. Among more common adverse events reported, there appeared to be a potential dose-response relationship for back pain, as the percentage of patients reporting this event was higher for the every 4 week dosing group vs the every 12 week or placebo groups, although an explanation for this is unclear. Seven major adverse cardiovascular events occurred in this study, all in patients receiving briakinumabTM. Recent evidence suggests that patients with psoriasis may be more likely to have major risk factors for cardiac events (Ludwig RJ, et al. Br J Dermatol 2007; 156: 271-6; Gisondi P, et al. Br J Dermatol 2007; 157: 68-73) and may be at higher risk for myocardial infarction (Gelfand JM, et al. Jama 2006; 296: 1735-41). Compelling evidence from a large, prospective, population-based study in a broadly representative cohort of patients with psoriasis, identified from the UK's General Practice Research Database, found that younger age and severe disease independently raised the relative risk of myocardial infarction by 3 times. Cytokines within atherosclerotic lesions have been reported to promote a Thl response leading to production of interferon y, and a cascade of other cytokines (including tumor necrosis factor, IL- 1, and IL-6), creating an inflammatory milieu that promotes atherosclerosis (Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005; 352: 1685-95). Considering the role of Thl-related cytokines in psoriasis, current evidence suggests an association between the inflammatory processes involved in psoriasis and those inherent in coronary artery disease. However, there is a lack of data distinguishing whether other variables potentially associated with psoriasis, such as poor management of lifestyle risk factors or stress, may be contributing to the risk of heart disease. No major adverse cardiovascular events were observed among the placebo-treated patients in this study. This finding is consistent with studies in psoriasis patients treated with the IL-12/23 blocker, ustekinumab. Four of 252 patients receiving ustekinumab in a phase II, placebo-controlled trial, experienced cardiac events that required hospitalization, including two myocardial infarctions and one event of cardiac failure; while no events occurred in the placebo group (Krueger GG, et al. N Engl J Med 2007; 356: 580-92). In a larger, phase III study of ustekinumab, major adverse cardiovascular 251 events were observed in 4 of 753 ustekinumab-treated patients, while a single event was observed in a placebo-treated patient after crossing over to receive ustekinumab (Leonardi CL, et al. Lancet 2008; 371: 1665-74). In a comparator trial of ustekinumab and etanercept in patients with moderate to severe psoriasis, 3 major adverse cardiovascular events were observed with ustekinumab, while none occurred with etanercept (Griffiths CE, et al. N Engl J Med; 362: 118-28). Conversely, no major adverse cardiovascular events were observed in a phase II investigation of briakinumabTM (Kimball AB, et al. Arch Dermatol 2008; 144: 200-7), or in a second phase III trial of ustekinumab (Papp KA, et al. Lancet 2008; 371: 1675-84). Ourpost hoc analysis of underlying cardiac risk factors revealed a slight imbalance between groups; however, these marginal differences may not account for the disproportion in observed cardiac events. Because the MACE observed in the current study and those observed in previous .studies of ustekinumab have been mostly isolated to anti-IL- 12/23 treatment exposed patients, more evidence will be crucial to determine whether treatment with this new therapy, the potential risk posed by psoriasis itself, or a combination of the two, is contributing to their occurrence. In light of the weight of evidence thus far, identification of cardiac risk factors prior to commencing anti IL 12/23 treatment in patients with psoriasis, and close monitoring during therapy, will be important precautionary measures. In summary, efficacy results from this 52-week, multicentre, randomized, placebo-controlled trial demonstrate that briakinumabTM rapidly induces and maintains high efficacy responses in patients with moderate to severe psoriasis, even among those generally considered to have more complicated treatment issues, such as prior failure of biologic therapy, arthritic involvement or higher weight. Efficacy responses were optimally maintained with a monthly dosing regimen. Safety results were mostly consistent with those observed in phase II testing of briakinumabTM, however the number of infections, nonmelanoma skin cancers, and MACE warrant further investigation in a larger patient population, and highlight the need to monitor for these events with the use of anti-IL-12/23 agents.
252 Table 39 Demographic and clinical characteristics at baseline by induction phase treatment group ABT-874 Placebo Total (N=981) (N=484) (N=1465) Demographicharacterics _ Age (years) 45.7 (13.2) 45.1 (13.5) 45.5 (13.3) Sex, male 666 (67.9%) 343 (70.9%) 1009 (68.9%) Weight (kg) 93.8 (23.6) 93.1 (23.0) 93.5 (23.4) Race Asian 44 (4.5%) 18 (3.7%) 62 (4.2%) Black 31(3.2%) 15 (3.1%) 46(3.1%) White 888 (90.5%) 432 (89.3%) 1320 (90.1%) Other 18 (1.8%) 19 (3.9%) 37 (2.5%) Clinical characteristics Body surface area affected (%) 24.8 (16.3) 25.7 (16.9) 25.1 (16.5) Psoriasis area and severity index 19.1 (7.5) 19.3 (7.3) 19.2 (7.4) Dermatology life quality index 12.8 (7.0) 12.7 (6.9) 12.8 (7.0) Duration of psoriasis (years) 18.9 (12.3) 19.2 (11.9) 19.0 (12.2) Patients with psoriatic arthritis' 290(29.6%) 150 (31.0%) 440(30.0%) Physician's global assessment Moderate 514(52.4%) 242 (50.0%) 756(51.6%) Severe 408 (41.6%) 218 (45.0%) 626 (42.7%) Very Severe 59 (6.0%) 24 (5.0%) 83 (5.7%) lData are mean (SD) or n (%).
253 Table 40: Risk factors for cardiovascular disease at baseline by induction phase treatment group ABT-874 Placebo Total (N=981) (N=484)~ (N=1465) CarIdiovascular disea-se meiclitry_______________ Any cardiovascular disease 395 (40.3%) 178 (36.8%) 573 (39.1%) Angina 6(0.6%) 2 (0.4%) 8 (0.5%) Cardiac arrhythmia 8(0.8%) 5(1.0%) 13 (0.9%) Congestive heart failure 4 (0.4%) 1 (0.2%) 5 (0.3%) Coronary artery disease 16(1.6%) 5(1.0%) 21(1.4%) Hypertension 302(30.8%) 139(28.7%) 441 (30.1%) Myocardial Infarction 19(1.9%) 6(1.2%) 25 (1.7%) Syncope 2 (0.2%) 0 2 (0.1%) Other 171 (17.4%) 80 (16.5%) 251 (17.1%) Cardiovascular and metabolicsyndrome 1s1 fatrs 1 Hypertension (blood pressure 2140/90 mm/Hg, or taking antihypertensive drug) 426(43.4%) 199 (41.1%) 625 (42.7%) Cigarette smoking 304 (31.0%) 148 (30.6%) 452 (30.9%) Diabetes mellitus 98 (10.0%) 40(8.3) 138 (9.4%) Obesity (BMI 230 kg/m 2 ) 508 (51.8%) 245 (50.6%) 753 (51.4%) Male a45 years 374(38.1%) 175 (36.2%) 549 (37.5%) Female 255 years 78(8.0%) 40(8.3%) 118(8.1%) LDL cholesterol >100 mg/dL 542(58.1%) 253 (55.0%) 795 (57.1%) Triglycerides >200 mg/dL 289 (29.6%) 132(27.4%) 421 (28.9%) HDL cholesterol <40 mg/dL 294(30.1%) 145 (30.1%) 439(30.1%) Multiple cardiovascular risk factors M1 risk factor 928 (94.6%) 456 (94.2%). 1384 (94.5%) 22 risk factors 804 (82.0%) 386 (79.8%) 1190 (81.2%) 23 risk factors 600 (61.2%) 268 (55.4%) 868 (59.2%) 24 risk factors 361 (36.8%) 166(34.3%) 527 (36.0%) Data are mean (SD) or ni 254 i r4 n m~ ! 4 in cn i 4) r- 00 r- 00 rZW0r- 0 C ) 0 0 C0 Nm C 0 0 N O O C04 CCOc k C. V V V CN00 eCo fn oc, "o C',~ le CN~ InN 0~~ u_0 o - c P 00 c) Co 00 'It' 0.00\ 00(N(\0 0 0 zt 0 C, c-q 4)4)) C0 3 C- 00 tn ~ ~ 0oj40. _ _ (0\_ L- W% _ 255 Table 42 Demographic and clinical characteristics at baseline by maintenance phase treatment group, for patients randomized to ABT-874 during the induction phase Withdrawal ABT-874 every ABT-874 every from ABT-874, 4 weeks 12 weeks (placebo) (N=298) (N=298) (N=149) {Demographic characteristics Age (years) 44.6 (13.3) 45.5 (12.6) 45.0 (13.4) Sex, male 206(69.1%) 201 (67.4%) 100(67.1%) Weight (kg) 91.3 (21.3) 93.7 (23.4) 89.3 (24.6) Race Asian 13 (4.4%) 7 (2.3%) 14 (9.4%) Black 7 (2.3%) 7 (2.3%) 5 (3.4%) White 275 (92.3%) 278 (93.3%) 127 (85.2%) Other 3 (1.0%) 6 (2.0%) 3 (2.0%) PCinical characteristics Body surface area affected (%) 23.7 (14.8) 22.3 (13.5) 25.2 (17.1) Psoriasis area and severity index 18.4 (6.5) 18.3 (6.2) 18.9 (8.2) Dermatology life quality index 12.7 (7.1) 12.6 (6.8) 11.9 (7.0) Duration of psoriasis (years) 19.1 (12.1) 18.5 (12.1) 18.9 (12.4) Patients with psoriatic arthritis 86 (28.9%) 79 (26.5%) 42 (28.2%) Physician's global assessment Moderate 170 (57.0%) 164 (55.0%) 85 (57.0%) Severe 113(37.9%) 124 (41.6%) 53(35.6%) Very Severe 15 (5.0%) 10 (3.4%) 11 (7.4%) Daaare mean (SD) or n () 256 0000 0..0 0 00 7-77 F z~ cl r, r,:Q CD - DCDI =rl4- CO C Q Qo 0a,60 0 IQI 0000 . o Q6 %r -- - n ~ 0 0 000 > > LI La C ~ 0 V 't CL 2 0 CL w !% Ir c uC c < In < < < 257 Example 25. Efficacy and Safety of BriakinumabTM, a Fully Human Interleukin 12/23 Monoclonal Antibody, Versus Methotrexate in Patients With Moderate to Severe Chronic Plaque Psoriasis: 52-Week Results From a Phase III, Randomised, Double-Blind Trial Although there is strong evidence supporting the use of biologic therapy for the treatment of psoriasis (Schmitt J, et al. Efficacy and tolerability of biologic and nonbiologic systemic treatments for moderate-to-severe psoriasis: meta-analysis of randomized controlled trials. Br J Dermatol 2008; 159: 513-26), there is a need to establish the optimal use of biologics, particularly as alternatives to traditional systemic therapies such as methotrexate (MTX). MTX is the most commonly prescribed systemic therapy for psoriasis worldwide (Menter A, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 4. Guidelines of care for the management and treatment of psoriasis with traditional systemic agents. J Am-Acad Dermatol 2009; 61: 451-85). Common adverse effects associated with MTX include nausea, anorexia, stomatitis, and fatigue, and cumulative or idiosyncratic toxicities of concern include hepatotoxicity, myelosuppression, and pulmonary fibrosis (Menter A, et al., ibid). Although clinical experience with MTX is abundant, large comparator trials of biologic therapy vs. MTX are sparse. Superior efficacy with adalimumab vs. MTX was reported in a 16-week, Phase III randomised trial (Saurat JH, et al, Efficacy and safety results from the randomized controlled comparative study of adalimumab vs. methotrexate vs. placebo in patients with psoriasis (CHAMPION). Br J Dermatol 2008; 158: 558-66.), and preliminary results of a 26-week trial demonstrated superior efficacy with infliximab vs. MTX (Reich K, et al., Infliximab is associated with greater improvement in health-related quality of life versus methotrexate for moderate-to-severe plaque-type psoriasis-the RESTORE 1 trial. Abstract P1187), but data are limited on the long-term use of MTX. In this Example the results of the first trial to evaluate the efficacy and safety of 1 year of treatment with MTX in comparison with briakinumabTM in patients with moderate to severe plaque psoriasis are reported. METHODS Patients 258 This Phase III, multicentre, randomised, double-blind trial was conducted at 43 sites in Europe and Canada. Patients aged 18 years were eligible to participate if they had a clinical diagnosis of psoriasis for ?6 months; had stable plaque psoriasis for >2 months; were candidates for systemic therapy or phototherapy; and had 10% body surface area involvement, a Physician's Global Assessment (PGA) score >3, and a Psoriasis Activity and Severity Index (PASI) score >12 at baseline. Patients were ineligible if they had nonplaque forms of psoriasis or if they had previously received IL- 12/-23 p40-targeting therapy or MTX. Patients also were ineligible if they had received treatment with biologics or investigational agents within the previous 12 weeks or 5 drug half-lives, conventional systemic psoriasis treatment or phototherapy within the previous 4 weeks, or topical therapy within 2 weeks of the study baseline. Patients were excluded if they had severe infections; a history of clinically significant hematologic, renal, or liver disease; a history of malignancy (except successfully treated basal cell carcinoma, nonmetastatic cutaneous squamous cell carcinoma, or cervical carcinoma in situ); or a history of active tuberculosis or evidence of latent tuberculosis (unless prophylactic treatment was received previously or initiated prior to administration of study drug). The study protocol was approved by an independent ethics committee or institutional review board at each study site, and each patient provided written informed consent. Procedures At baseline (Week 0), patients were randomised 1:1 to receive briakinumabTM (200 mg subcutaneously at Weeks 0 and 4 and 100 mg every 4 weeks from Weeks 8 to 48) or oral MTX (5 to 25 mg weekly from Weeks 0 to 51) plus oral folate (5 mg weekly from Weeks 0 to 51) (Figure 46). Patients in the MTX group received MTX 5 mg at Week 0, 10 mg at Week 1, and 15 mg/wk from Weeks 2 to 9. At Weeks 10 and 16, MTX dosage was increased by 5 mg (to 20 mg/wk at Week 10 and 25 mg/wk at Week 16) for patients who did not achieve PASI 75 or a PGA of 0 or 1. To maintain the blind, patients in the briakinumabTM group also received placebo capsules to match MTX and 259 placebo tablets to match folate, and patients in the MTX group also received subcutaneous injections of placebo to match briakinumabTM. Treatment success was defined as having achieved both PASI >75 and a PGA of 0 or I at Week 24. Patients in. the MTX group who achieved treatment success maintained their current weekly MTX dose for the remainder of the study duration. At any time during the study, the safety assessor could reduce or withhold the MTX dose as a result of patient reported symptoms, physical examination, adverse events, or laboratory abnormalities (aspartate aminotransferase or alanine aminotransferase 21.5 x upper limit of normal, platelet count <100,000/mm 3 , total white blood cell count <3,000/mm , creatinine >2 x upper limit of normal). Patients in either treatment group who did not achieve treatment success or who lost response (defined as PASI <50 and PGA 3) after Week 24 discontinued the trial and were eligible to enroll in an open-label extension study of briakinumabTM. The largest decrease in the percentage of patients remaining in the study occurred between Weeks 24 and 28, with more patients remaining in the MTX group than in the briakinumabTM group discontinuing owing to lack of efficacy. PASI and PGA responses were calculated in a conservative fashion by imputing discontinued patients as non-responders. The primary efficacy endpoints were the percentages of patients achieving PASI 75 at Week 24, a PGA of 0 or I at Week 24, PASI 75 at Week 52, and a PGA of 0 or 1 at Week 52. The PASI is a measure of the severity of skin symptoms of psoriasis, with scores ranging from 0 to 72 (Fredriksson and Pettersson, 1978). A PASI greater than 10 is considered to represent moderate to severe skin symptoms of psoriasis (Pathirana et al, European S3-guidelines on the systemic treatment of psoriasis vulgaris. J Eur Acad Dermatol Venereol 2009; 23(Suppl 2): 1-70; Smith et al, British Association of Dermatologists' guidelines for biologic interventions for psoriasis 2009. Br J Dermatol 2009; 161: 987-1019). The PGA used was a 6-point ordinal scale with 0 equalling "Clear"; 1, "Minimal"; 2, "Mild"; 3, "Moderate"; 4, "Severe"; and 5, "Very Severe" (Ko, 1998). Secondary efficacy variables included median time to achieve PASI 75 response, PASI 50/75/90/100 response rates through Week 52, proportion of patients with a PGA 0 260 or I by visit over 52 weeks, and mean percentage improvements in PASI scores from baseline. Efficacy assessments also included the change from baseline in Nail Psoriasis Severity Index (NAPSI) scores, which range from 0 (no nail psoriasis) to 80 (psoriasis in all 10 fingernails) (Rich and Scher, 2003). Patients with nonzero baseline NAPSI scores had NAPSI assessments at subsequent visits, using a single target fingernail (the nail most affected by psoriasis at baseline), with a NAPSI score range from 0 to 8. An additional secondary efficacy parameter was the Dermatology Life Quality Index (DLQI), a patient-reported measure of the extent to which psoriasis impacts health related quality of life. The DLQI yields a score ranging from 0 to 30, with a lower score indicating lower impact (Finlay and Khan, 1994). Assessments included the percentages of patients with a DLQI score of 0 or 1 (no effect of psoriasis) (Hongbo et al, Translating the science of quality of life into practice: What do dermatology life quality index scores mean? J Invest Dermatol 2005; 125: 659-64), percentages of patients with a decrease of >5 points (a clinically meaningful reduction) (Khilji et al, Clinical meaning of change in Dermatology Life Quality Index scores. Br J Dermatol 2002; 147(Suppl 62): 50), and change from baseline DLQI scores. Adverse events, laboratory data, and vital signs were assessed throughout the study. Patients were closely monitored for signs of infection, malignancy, and immunologic reaction. Treatment-emergent adverse events were defined as those occurring on or after the first dose of study drug and up to 45 days after the last dose of study drug. Statistical analysis According to the original study plan, approximately 250 patients were to be randomised. Assuming that the PASI 75 response rates at Week 24 were 70% in the briakinumabTM group and 50% in the MTX group, this sample size would provide 90% power to demonstrate the superiority of briakinumabTM over MTX (2-sided chi-square test at level of significance 5%). However, owing to difficulties in obtaining accurate enrolment numbers in a timely manner, more than 250 patients were enrolled. Given an actual enrolment number of 317 patients, the power increased from 90% to 95%. This 261 change in power had little impact because of the large differences in.endpoints between treatment groups observed in the study. The intention-to-treat population included all patients who were randomised at Week 0. The intention-to-treat population was used for the efficacy analyses. For the primary efficacy assessments, all comparisons were performed using a 2-sided Cochran Mantel-Haenszel test adjusted for country at an alpha level of 0.05. The type I error rate was controlled at 0.05 by adhering to the a priori defined order of statistical hypotheses. Nonresponder imputation was used to handle missing data. Any patient with a missing PASI or PGA score at a visit was considered a nonresponder at that visit. Last observation carried forward (LOCF) was used as a sensitivity analysis. For categorical variables, the chi-square test or Fisher's exact test (if the expected cell size was <5) was used to evaluate the superiority of briakinumabTM vs. MTX. Nonresponder imputation was used to handle missing data. LOCF was used as a sensitivity analysis. The median times to achieve PASI 75 response and PGA score of 0 or 1 were calculated using the Kaplan-Meier method. The treatment group difference was tested using the log-rank test. Patients who did not achieve a response on or before Week 52 were censored at the date of the last PASI/PGA evaluation. Differences between treatment groups in the change in PASI, DLQI, and other continuous variables were analysed using analysis of covariance with baseline value and treatment group in the model. LOCF was used to handle missing data. Analysis as observed was done as sensitivity analysis. To calculate the percentage change in NAPSI score, only patients with nonzero baseline NAPSI scores were included in the analysis. Wilcoxon 2-sample tests were used to compare the treatment difference in the change. LOCF was used to handle missing data. Analysis as observed was done as sensitivity analysis. The safety analyses were conducted using the safety population, which included all patients who received at least 1 injection of study drug. RESULTS A total of 317 patients met the inclusion criteria and were randomised to briakinumabTM (N=154) or MTX (N=l63) (Figure 47). No randomised patients were 262 excluded from the efficacy analysis (intention-to-treat population). A total of 106 (68.8%) patients in the briakinumabTM group and 45 (27.6%) patients in the MTX group completed the study through Week 52. Reasons for discontinuing the study are shown in Figure 47. Baseline demographics, clinical characteristics, and disease severity were similar between treatment groups (Table 44). Mean duration of psoriasis was 18.9 years, mean PASI score was 18.1, and mean body surface area involvement was 26.1%. Approximately 52% of patients had previously received systemic nonbiologic treatment (other than MTX) and 18% had previously received biologic therapy. Significantly more patients in the briakinumabTM group than the MTX group achieved the primary endpoints of PASI 75 response at Week 24 (81.8% vs. 39.9%; p<0.001) and at Week 52 (66.2% vs. 23.9%; p<0.001). Median time to achieve PASI 75 response was 56 days for the briakinumabTM group vs. 140 days for the MTX group (p<0.001). The percentage of patients who achieved a PASI 75 response was significantly greater in the briakinumabTM group by Week 4 and at all time points through Week 52 (Figure 48A). Similarly, significant differences between treatment groups were apparent beginning at Week 2 for PASI 50 and at Week 8 for PASI 90 and PASI 100; greater response rates in the briakinumabTM group were maintained through Week 52 (Figures 3B-3D). The mean percentage improvement in PASI scores from baseline increased over time in both treatment groups but was significant greater in the briakinumabTM group at all time points (Figure 49). PASI 75/90/100 response rates at Week 16 for MTX-treated patients were similar to those seen at Week 16 of CHAMPION. (Saurat JH, Stingl G, Dubertret L, et al; CHAMPION Study Investigators. Efficacy and safety results from the randomized controlled comparative study of adalimumab vs. methotrexate vs. placebo in patients with psoriasis (CHAMPION). Br J Dermatol 2008; 158: 558-66.) Significantly more patients in the briakinumabTM group than the MTX group achieved the primary endpoints of PGA of 0 or 1 at Week 24 (80.5% vs. 34.4%; p<0.001) and at Week 52 (63.0% vs. 20.2%; p<0.001). In addition, cleared disease as assessed by a PGA of 0 was evident in significantly more patients in the briakinumabTM group than the MTX group at Week 24 (46.1% vs. 9.2%; p<0.001) and at Week 52 (45.5% vs. 9.8%; p<0.001). Median time to achieve a PGA of 0 or 1 was 69 days for the briakinumabTM 263 group vs. 171 days for the MTX group (p<0.001). Significantly greater percentages of patients in the briakinumabTM group attained a PGA of 0 or 1 at each time point from Week 4 to Week 52 (Figure 50). Baseline NAPSI scores were similar between treatment groups (Table 44). In the briakinumabTM group, the mean NAPSI score for the target fingernail decreased from 4.8. at baseline to 2.1 at Week 24 and 1.2 at Week 52. In the MTX group, the mean NAPSI score for the target fingernail decreased from 4.8 at baseline to 3.0 at Week 24 and was also 3.0 at Week 52. The change from baseline in mean NAPSI score for the target fingernail was significantly greater in the briakinumabTM group than in the MTX group at Week 24 and Week 52 (p<0.001 for both comparisons). Significantly more patients in the briakinumabTM group than the MTX group had a DLQI score of 0 or 1 at Week 24 (70.8% vs. 34.4%; p<0.001) and at Week 52 (61.7% vs. 17.8%; p<0.001). Similarly, significantly more patients in the briakinumabTM group than the MTX group achieved a clinically meaningful reduction in DLQI score (decrease of >5 points) at Week 24 (66.2% vs. 47.9%; p<0.001) and at Week 52 (56.5% vs. 18.4%; p<0.001). Patients in the briakinumabTM group had significantly greater decreases from baseline in DLQI scores compared with the MTX group at all time points assessed (Figure 51). At Week 24, the mean DLQI score had decreased from 11.0 at baseline to 1.6 in the briakinumabTM group, vs. a decrease from 11.2 at baseline to 4.6 in the MTX group (difference between groups in change from baseline: -3.0; 95% confidence interval [CI]: -3.9, -2.1; p<0.001). DLQI scores at Week 52 (1.5 in the briakinumabTM group vs. 4.6 in the MTX group) reflected a similarly greater change from baseline with briakinumabTM vs. MTX (-3.1; 95% CI: -4.0, -2.2; p<0.00 l). For all efficacy variables, sensitivity analyses performed for missing data yielded similar results. Adverse event profiles were similar between briakinumabTM and MTX (Table 45). The most common treatment-emergent adverse events were nasopharyngitis, headache, diarrhoea, arthralgia, and upper respiratory tract infection. Diarrhoea (9.7% vs. 3.7%, p=0.04) and injection-site-related adverse events (8.4% vs. 1.8%; p=0.009) were more frequent in the briakinumabTM group than the MTX group.
264 Of the 317 patients randomised, 22 patients discontinued the study owing to I or more adverse events. Twelve (7.8%) patients in the briakinumabTM group discontinued; adverse events for 5 patients were serious (gastrointestinal hypomotility and legionella infection, breast cancer, breast neoplasm, prostate cancer, and herpes zoster). Ten (6.1%) patients in the MTX group discontinued; adverse events for 5 patients were serious (hepatic enzyme increases and hepatitis, sacroiliitis, divertictilitis, erythrodermic psoriasis, angioedema and urticaria). Fourteen (9.1%) patients in the briakinumabTM group and 10 (6.1%) patients in the MTX group reported treatment-emergent serious adverse events. Seven patients experienced serious infections, including 4 patients in the briakinumabTM group (1 case of legionella infection with candidaemia, and septic shock, I case of osteomyelitis, I case of herpes zoster, and 1 case of tonsillitis) and 3 patients in the MTX group (2 cases of diverticulitis and 1 case of hepatitis). The incidence rates of serious infectious adverse events were 4.1 and 2.7 per 100-patient-years in the briakinumabTM and MTX groups, respectively. Fourteen patients in each treatment group had positive purified protein derivative tests at baseline; however, no patient had active tuberculosis at screening or reported tuberculosis-related adverse events during the study. Three patients in the briakinumabTM group experienced malignancies (1 patient had breast cancer, I patient had breast neoplasm, and 1 patient had prostate cancer). One death was reported (oesophageal rupture in a patient in the MTX group). There were no reports of major cardiovascular events such as sudden cardiac death, myocardial infarction, or stroke. Six patients in the briakinumabTM group and 4 patients in the MTX group had ischemic heart disease adverse events (2 patients in the briakinumabTM group experienced angina pectoris and all other patients experienced increased creatinine phosphokinase), none of which were serious. DISCUSSION In this 52-week, Phase III, multicentre, randomised, double-blind study, briakinumabTM was superior to MTX in reducing the signs and symptoms of moderate to severe plaque psoriasis, improving nail psoriasis, and favourably influencing patient- 265 reported health-related quality of life. Treatment with briakinumabTM produced a rapid clinical response that was maintained through 52 weeks when compared to MTX. No clinically important safety concerns were identified in the study. The present study is the first comparator trial to evaluate the efficacy and safety of MTX versus an IL-12/-23 p40-neutralizing antibody. Compared with MTX, briakinumabTM demonstrated faster onset of action and superior efficacy through Week 52, with a generally similar safety profile. In the briakinumabTM group, a PASI 75 response was achieved by 81.8% of patients at Week 24 and by 66.2% of patients at Week 52. It is difficult to compare maintenance of clinical response over 1 year with briakinumabTM in this trial vs. ustekinumab in previous trials because the present study of briakinumabTM was a straightforward intention-to-treat analysis, whereas ustekinumab trials included dose escalation (Papp KA, et al., Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 2008; 371: 1675-84) and several selection steps of responding patients (Leonardi CL, et al., Efficacy and safety of ustekinumab, a human interleukin- 12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placeb6 controlled trial (PHOENIX 1). Lancet 2008; 371: 1665-74.). This first-ever I-year study of MTX provides insight into the maintenance of response with MTX. After Week 24, there was a relatively sharp drop in the percentage of MTX-treated patients with a PASI 75 response (39.9% at Week 24 compared with 30.7% at Week 28) and a smaller drop in the percentage of patients with a PGA of 0 or 1 (34.4% at Week 24 compared with 28.8% at Week 28). It should be noted that at Week 24, patients who did not achieve both PASI ;75 and a PGA of 0 or I were discontinued per protocol. Previous MTX comparator trials have been short term. In a 16-week study of MTX vs. cyclosporine, no differences in clinical responses were observed between groups (Heydendael VM, et al., Methotrexate versus cyclosporine in moderate-to-severe chronic plaque psoriasis. N Engl J Med 2003; 349: 658-65). More recent trials have demonstrated better efficacy with biologic therapy vs. MTX. In a 16-week trial of MTX vs. adalimumab, significantly more patients treated with adalimumab achieved PASI 75 response at Week 16 (80% vs. 36%; p<0.001) (Saurat JH, et al., Efficacy and safety 266 results from the randomized controlled comparative study of adalimumab vs. methotrexate vs. placebo in patients with psoriasis (CHAMPION). Br J Dermatol 2008; 158: 558-66). Moreover, similar to the findings of the present study, clinical response occurred more rapidly with biologic therapy than with MTX. Preliminary results of a 26 week trial of MTX vs. infliximab demonstrated that significantly more patients treated with infliximab achieved PASI 75 at Week 26 (77% vs. 31%; p<0.001) (Reich K, et al., Infliximab is associated with greater improvement in health-related quality of life versus methotrexate for moderate-to-severe plaque-type psoriasis-the RESTORE 1 trial. Abstract P1.187). In addition, greater improvement in DLQI scores from baseline to Week 26 was evident with infliximab than with MTX (-11.3 vs. -9.1; p<0.004) (Reich K, et al. Infliximab is associated with greater improvement in health-related quality of life versus methotrexate for moderate-to-severe plaque-type psoriasis-the RESTORE 1 trial. Abstract P1187). With respect to the long-term safety of MTX, previous 16-week trials reported hepatic-related adverse events leading to discontinuation in 12 (28%) of 43 patients who received 15 mg weekly (Heydendael VM, et al. Methotrexate versus cyclosporine in moderate-to-severe chronic plaque psoriasis. N Engl J Med 2003; 349: 658-65) and in 4 (4%) of 110 patients who received a starting MTX dosage of 7.5 mg weekly that was increased as needed and tolerated to 25 mg weekly (Saurat JH, et al. Efficacy and safety results from the randomized controlled comparative study of adalimumab vs. methotrexate vs. placebo in patients with psoriasis (CHAMPION). Br J Dermatol 2008; 158: 558-66). In the present study of 52 weeks of MTX treatment (5 to 25 mg weekly, per titration schedule), 16 (10%) patients in the MTX group experienced hepatic-related adverse events, but only 2 (1%) patients discontinued the study. These results suggest that initiating MTX with a low dosage that is titrated up as indicated and tolerated may be a safe strategy for long-term use of MTX. With respect to safety of briakinumabTM, 52 weeks of treatment was associated with a low incidence of serious adverse events. The incidence of serious infectious adverse events was 4.1 events per 100-patient-years with briakinumabTM and 2.7 events per 100-patient-years with MTX. Previous studies have reported a rate of 1.3 serious infectious events per 100-patient-years with adalimumab or infliximab (Menter A, et al.
267 Adalimumab therapy for moderate to severe psoriasis: A randomized, controlled phase III trial. J Am Acad Dermatol 2008; 58: 106-15; Burmester GR, et al. Adalimumab safety and mortality rates from global clinical trials of six immune-mediated inflammatory diseases. Ann Rheum Dis 2009; 68: 1863-69). In the present study of briakinumabTM, no serious cardiovascular events (eg, myocardial infarction, stroke) were reported, in contrast with low frequencies of such events in another trial of briakinumabTM (M06-890 reference) and in trials of ustekinumab (Krueger GG, et al. A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N Engl J Med 2007; 356: 580-92; Leonardi CL, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 2008; 371: 1665-74; Papp KA, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet 2008; 371: 1675-84). CONCLUSIONS In this head-to-head trial of briakinumabTM vs. MTX in patients with moderate to severe plaque psoriasis, briakinumabTM demonstrated superior efficacy compared with MTX on all primary and secondary endpoints, including clinical response measures and patient reported health-related quality of life, through 52 weeks of treatment. Based on the superior efficacy and a safety profile without clinically significant findings compared with MTX, these results demonstrate a favourable benefit-risk ratio for briakinumabTM vs. MTX as a therapeutic option for the treatment of psoriasis.
268 Table 44. Baseline Demographic and Clinical Characteristics Characteristic BriakinumabTM MTX (N=154) (N=163) Age (years), mean (SD) 45.0 (13.1) 43.1 (12.9) Male 111(72.1) 111 (68.1) White 149 (96.8) 158 (96.9) Weight (kg), mean (SD) 85.1 (17.5) 82.0 (18.6) Duration of psoriasis (years), mean 18.6 (11.9) 19.1 (11.3) (SD) BSA affected by psoriasis (%), mean 26.1 (16.7) 26.1 (16.3) (SD) History of psoriatic arthritis 25(16.2) 28(17.2) PASI score, mean (SD) 18.4 (6.7) 17-8 (6.1) NAPSI score,* mean (SD) 27.7 (16.4) 279 (19.1) NAPSI score for target fingernail,* 4.8 (2.0) 4.8 (2.1) mean (SD) PGA "Clear," "Minimal," or "Mild" 0 0 "Moderate" 75 (48.7) 87 (53.4) "Severe" 65 (42.2) 72 (44.2) "Very Severe" 14 (9.1) 4 (2.5) DLQI score,t mean (SD) 11.1 (6.6) 11.3 (7.5) Previous psoriasis treatment Topical therapy 141 (91.6) 148 (90.8) Phototherapy 100 (64.9) 105 (64.4) Systemic nonbiologic treatment 77 (50.0) 89 (54.6) Systemic biologic treatment 24 (15.6) 34 (20.9) Etanercept 14 (9.1) 12 (7.4) Infliximab 9 (5.8) 11 (6.7) Adalimumab 5 (3.2) 7 (4.3) Alefacept 2(1.3) 2(1.2) Efalizumab 6(3.9) 3(1.8) Other 3(1.9) 7(4.3) Values are n (%) unless otherwise noted. . *N=1 15 (briakinumabTM), N=108 (MTX). tN=154 (briakinumabTM), N= 162 (MTX). BSA, body surface area; DLQI, Dermatology Life Quality Index; MTX, methotrexate; NAPSI, Nail Psoriasis Severity Index; PASI, Psoriasis Area and Severity Index; PGA, Physician's Global Assessment.
269 Table 45. Adverse Events by Treatment Group Event* BriakinumabTM MTX (N=154) (N=163) Any adverse event 131 (85.1) 145 (89.0) Serious adverse events 14 (9.1) 10 (6.1) Adverse events leading to discontinuation 12 (7.8) 10 (6.1) Common adverse events Nasopharyngitis 44 (28.6) 45 (27.6) Headache 18(11.7) 22(13.5) Diarrhoea 15 (9.7)' 6 (3.7) Arthralgia 12 (7.8) 11 (6.7) Upper respiratory tract infection 11 (7.1) 12 (7.4) Back pain 10 (6.5) 9 (5.5) Fatigue 10(6.5) 10(6.1) Gastroenteritis 10(6.5) 11(6.7) Cough 9(5.8) 12(7.4) Nausea 9(5.8) 19(11.7) Influenza 8 (5.2) 9 (5.5) Rhinitis 8 (5.2) 6 (3.7) Adverse events of special interest Infections 89 (57.8) 102 (62.6) Serious infections 4 (2.6) 3 (1.8) Opportunistic infections 1 (0.6) 0 Malignancies 3(1.9) 0 Cardiovascular events 0 0 Ischemic heart disease 6 (3.9) 4 (2.5) Deaths|| 0 1 (0.6) Values are n (%). *Occurred on or after the first dose of study drug and up to 45 days after the last dose of study drug. tOccurred in >5% of patients in either treatment group. tp=0-04 vs. MTX group. §Any serious adverse event of sudden cardiac death, myocardial infarction, or stroke. Includes non-treatment-emergent deaths. MTX, methotrexate.
270 EQUIVALENTS Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following 5 claims.

Claims (187)

1. A method of treating psoriasis in a subject comprising administering to the subject a first dose amount of an antibody, or antigen-binding portion thereof, which 5 is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a periodicity, and administering a second dose amount of the antibody, or antigen-binding portion thereof, at the same periodicity, thereby treating psoriasis in the subject.
2. A method of treating psoriasis in a subject comprising administering to 10 the subject a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a first periodicity, and administering a second dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity, thereby treating psoriasis in the subject. 15
3. The method of claim 1 or 2, wherein the first dose amount of the antibody, or antigen-binding portion thereof, is at least about 100 mg to about 200 mg.
4. The method of claim 1 or 2, wherein the first dose amount of the 20 antibody, or antigen-binding portion thereof, is at least about 100 mg.
5. The method of claim 1 or 2, wherein the first dose amount of the antibody, or antigen-binding portion thereof, is at least about 200 mg. 25
6. The method of claim 1 or 2, wherein the second dose amount of the antibody, or antigen-binding portion thereof, is at least about 200 mg.
7. The method of claim 1 or 2, wherein the second dose amount of the antibody, or antigen-binding portion thereof, is at least about 100 mg. 30
8. The method of claim 1 or 2, wherein the second dose amount of the antibody, or antigen-binding portion thereof, is the same as the first dose amount of the antibody, or antigen-binding portion thereof. 35
9. The method of claim 1 or 2, wherein the second dose amount of the antibody, or antigen-binding portion thereof, is different than the first dose amount of the antibody, or antigen-binding portion thereof. 272
10. The method of claim 9, wherein the second dose amount of the antibody, or antigen-binding portion thereof, is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof. 5
11. The method of claim 9, wherein the second dose amount of the antibody, or antigen-binding portion thereof, is about 190-210% of the first dose amount of the antibody, or antigen-binding portion thereof.
12. The method of claim 1, wherein the periodicity of administration of the 10 antibody, or antigen-binding portion thereof, is about once a week.
13. The method of claim 1, wherein the periodicity of administration of the antibody, or antigen-binding portion thereof, is about once every other week. 15
14. The method of claim 1, wherein the periodicity of administration of the antibody, or antigen-binding portion thereof, is about once every four weeks.
15. The method of claim 2, wherein the first periodicity of administration of the antibody, or antigen-binding portion thereof, is about once a week. 20
16. The method of claim 2, wherein the first periodicity of administration of the antibody, or antigen-binding portion thereof, is about once every other week.
17. The method of claim 2, wherein the first periodicity of administration of 25 the antibody, or antigen-binding portion thereof, is about once every four weeks.
18. The method of claim 2, wherein the second periodicity of administration of the antibody, or antigen-binding portion thereof, is about once a week. 30
19. The method of claim 2, wherein the second periodicity of administration of the antibody, or antigen-binding portion thereof, is about once every other week.
20. The method of claim 2, wherein the second periodicity of administration of the antibody, or antigen-binding portion thereof, is about once every four weeks. 35
21. The method of claim 2, wherein the second periodicity of administration of the antibody, or antigen-binding portion thereof, is about once every 30-200 days. 273
22. The method of claim 1 or 2, wherein the duration of the first periodicity is about 12 weeks.
23. The method of claim 1 or 2, wherein the duration of the first periodicity 5 is about 8 weeks.
24. The method of claim 1 or 2, wherein the duration of the first periodicity is about 4 weeks. 10
25. The method of claim 1 or 2, wherein the duration of the first periodicity is about 2 weeks.
26. The method of claim 1 or 2, wherein the duration of the first periodicity is about 1 week. 15
27. The method of claim 1 or 2, wherein the duration of the second periodicity is about 60 weeks.
28. The method of claim 1 or 2, wherein the duration of the second 20 periodicity is about 44 weeks.
29. The method of claim 1 or 2, wherein the duration of the second periodicity is about 12 weeks. 25
30. The method of claim 1 or 2, wherein the duration of the second periodicity is about 4 weeks.
31. The method of claim 1 or 2, wherein the duration of the second periodicity is about 2 weeks. 30
32. The method of claim 1 or 2, wherein the duration of the second periodicity is about 1 week. 35
33. The method of claim 1 or 2, wherein the duration of the second periodicity is at least about 12 weeks. 274
34. The method of claim 1 or 2, wherein the duration of the second periodicity is at least about 44 weeks.
35. The method of claim 1 or 2, wherein the duration of the second 5 periodicity is at least about 60 weeks.
36. The method of claim 1 or 2, wherein the second dose amount is administered to the subject upon a flare of psoriasis. 10
37. The method of claim 1 or 2, wherein the second dose amount is administered to the subject prior to a flare of psoriasis.
38. The method of claim 36 or 37, wherein the flare of psoriasis is indicated by loss of a Psoriasis Area and Severity Index (PASI) 90 response. 15
39. The method of claim 36 or 37, wherein the flare of psoriasis is loss of a Psoriasis Area and Severity Index (PASI) 75 response.
40. The method of claim 36 or 37, wherein the flare of psoriasis is loss of a 20 Psoriasis Area and Severity Index (PASI) 50 response.
41. The method of any one of claims 38-40, wherein the loss of PASI response is loss of PASI response of a single body region. 25
42. The method of any one of claims 38-40, wherein the loss of PASI response is loss of PASI response of two body regions.
43. The method of any one of claims 38-40, wherein the loss of PASI response is loss of PASI response of three body regions. 30
44. The method of any one of claims 38-40, wherein the loss of PASI response is loss of PASI response of four body regions.
45. The method of any one of claims 41-44, wherein the body region is 35 selected from the group consisting of trunk, lower extremities, upper extremities, and head and neck. 275
46. The method of claim 36 or 37, wherein the flare of psoriasis is loss of a clear or minimal Physician's Global Assessment (PGA) rating.
47. The method of claim 1 or 2, wherein the psoriasis is chronic psoriasis. 5
48. The method of claim 1 or 2, wherein the psoriasis is chronic psoriasis affecting about >20% body surface area of the subject.
49. The method of claim 1 or 2, wherein the psoriasis is plaque psoriasis. 10
50. The method of claim 49, wherein the plaque psoriasis is moderate to severe plaque psoriasis.
51. The method of any one of claims 1-50, wherein the antibody is 15 administered subcutaneously.
52. A method of treating psoriasis in a subject comprising administering to the subject an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a periodicity of about once every 20 4 weeks, thereby treating psoriasis in the subject.
53. A method of treating psoriasis in a subject comprising administering to the subject an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a periodicity of about once every 25 12 weeks, thereby treating psoriasis in the subject.
54. A method of treating psoriasis in a subject comprising administering to the subject: a) a first dose amount of an antibody, or antigen-binding portion thereof, 30 which is capable of binding to the p40 subunit of IL- 12 and/or IL-23; and b) a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a periodicity of about once every 12 weeks, thereby treating psoriasis in the subject. 35
55. The method of any one of claims 52-54, wherein the subject achieves at least a PGA score of 0 or 1. 276
56. The method of any one of claims 52-54, wherein the subject achieves at least a PASI 75 response.
57. The method of any one of claims 52-54, wherein the subject achieves at 5 least a PASI 90 response.
58. The method of any one of claims 52-54, wherein the subject achieves at least a PASI 100 response. 10
59. The method of claim 55, wherein the subject maintains the PGA score of 0 or 1 for at least 4 weeks.
60. The method of claim 56, wherein the subject maintains the PASI 75 response for at least 4 weeks. 15
61. The method of claim 57, wherein the subject maintains the PASI 90 response for at least 4 weeks.
62. The method of any one of claims 52-61, wherein the first dose amount is 20 at least about 200 mg.
63. The method of any one of claims 52-61, wherein the second dose amount is at least about 100 mg. 25
64. A method of treating psoriasis in a subject comprising administering to the subject: a) a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a first periodicity of about once every 4 weeks; and 30 b) administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks, thereby treating psoriasis in the subject. 35
65. The method of claim 64, wherein the first dose amount is at least about 200 mg. 277
66. The method of claim 64, wherein the second dose amount is at least about 100 mg.
67. The method of claim 64, wherein the duration of the first periodicity is at 5 least about 8 weeks.
68. The method of claim 64, wherein the duration of the second periodicity is at least about 4 weeks. 10
69. The method of claim 64, wherein the duration of the second periodicity is at least about 16 weeks.
70. The method of claim 62, wherein the duration of the second periodicity is at least about 44 weeks. 15
71. A method of treating psoriasis in a subject comprising administering to the subject: a) a first dose amount of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, according to a first 20 periodicity of about once every 4 weeks; and b) a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks; and c) the second dose amount of the antibody, or antigen-binding portion 25 thereof, according to a third periodicity of about once every 12 weeks, thereby treating psoriasis in the subject.
72. The method of claim 71, wherein the first dose amount is at least about 200 mg. 30
73. The method of claim 71, wherein the second dose amount is at least about 100 mg.
74. The method of claim 71, wherein the duration of the first periodicity is at 35 least about 8 weeks.
75. The method of claim 71, wherein the duration of the second periodicity is at least about 4 weeks. 278
76. The method of claim 71, wherein the duration of the third periodicity is at least about 12 weeks. 5
77. The method of claim 71, wherein the duration of the third periodicity is at least about 36 weeks.
78. The method of any one of claims 64-77, wherein the subject achieves a PGA score of 0 or 1. 10
79. The method of any one of claims 64-77, wherein the subject achieves at least a PASI 75 response.
80. The method of any one of claims 64-77, wherein the subject achieves at 15 least a PASI 90 response.
81. The method of any one of claims 64-77, wherein the subject achieves at least a PASI 100 response. 20
82. The method of claim 78, wherein the subject maintains a PGA score of 0 or 1 for at least 4 weeks.
83. The method of claim 79, wherein the subject maintains a PASI 75 response for at least 4 weeks. 25
84. The method of any claim 80, wherein the subject maintains a PASI 90 response for at least 4 weeks.
85. A method of treating psoriasis in a population of subjects, comprising 30 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 60% of the population of subjects achieve a PASI 75 response by about week 12.
86. A method of treating psoriasis in a population of subjects comprising 35 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 25% of the population of subjects achieve a PASI 90 response by about week 12. 279
87. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 10% of the population of subjects achieve a PASI 100 response by about week 12. 5
88. The method of any one of claims 85-87, comprising administering to each subject in the population: a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and 10 b) administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks.
89. The method of any one of claims 85-87, comprising administering to 15 each subject in the population: a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of 20 about once every 4 weeks; and c) the second dose amount of the antibody, or antigen-binding portion thereof, according to a third periodicity of about once every 12 weeks.
90. The method of any of claims 52-89, wherein the psoriasis is moderate to 25 severe or chronic psoriasis.
91. The method of any of claims 52-89, wherein the psoriasis is plaque psoriasis. 30
92. The method of any one of claims 52-89, wherein the antibody is administered subcutaneously.
93. The method of any one of claims 52-89, wherein the antibody is a human antibody. 35
94. The method of any one of claims 52-87, wherein the antibody is ABT RZ'7zt 280
95. The method of any one of the preceding claims, wherein the subject or population of subjects achieves at least a PASI 75 response by about week 24.
96. The method of any one of the preceding claims, wherein the subject or 5 population of subjects achieves at least a PASI 75 response by about week 52.
97. The method of any one of the preceding claims, wherein the subject or population of subjects achieves at least a PGA score of 0 or 1 by about week 24. 10
98. The method of any of the preceding claims, wherein the subject or population of subjects achieves at least a PGA score of 0 or 1 by about week 52.
99. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion 15 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 41% of the population of subjects achieve at least a PASI 75 response by about week 24.
100. A method of treating psoriasis in a population of subjects, comprising 20 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 35% of the population of subjects achieve at least a PGA score of 0 or 1 by about week 24. 25
101. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 25% of the population of subjects achieve at least a PASI 75 response by about week 52. 30
102. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 21% of the population of subjects achieve at least a PGA score of 0 or 1 by about 35 week 52.
103. The method of any one of the preceding claims, wherein the subject or population of subjects achieves an improvement in a Dermatology Life Quality Index 281 (DLQI) score or mean Dermatology Life Quality Index (DLQI) score of at least about 9.
104. The method of any one of the preceding claims, wherein the subject or 5 population of subjects achieves an improvement in a Short Form 36 Health Survey Physical Component Summary (PCS) score or mean Short Form 36 Health Survey Physical Component Summary (PCS) score of at least about 2.
105. The method of any one of the preceding claims, wherein the subject or 10 population of subjects achieves an improvement in a Short Form 36 Health Survey Mental Component Summary (MCS) score or mean Short Form 36 Health Survey Mental Component Summary (MCS) score of at least about 4.
106. The method of any one of the preceding claims, wherein the subject or 15 population of subjects achieves an improvement in a visual analog scale score or mean visual analog scale score for psoriasis-related pain (VAS-Ps) of at least about -25.
107. The method of any one of the preceding claims, wherein the subject or population of subjects achieves an improvement in a visual analog scale score for 20 psoriatic arthritis-related pain (VAS-PsA) or mean visual analog scale score for psoriatic arthritis-related pain (VAS-PsA) of at least about -32.
108. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion 25 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein the population of subjects achieves a minimum clinically important difference (MCID) response rate for psoriasis-related pain (VAS-Ps) of at least about 60%.
109. A method of treating psoriasis in a population of subjects comprising 30 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein the population of subjects achieves a minimum clinically important difference (MCID) response rate for Dermatology Life Quality Index (DLQI) of at least about 70% by about week 12. 35
110. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein 282 the population of subjects achieves a minimum clinically important difference (MCID) response rate for Dermatology Life Quality Index (DLQI) of at least about 81% by about week 52. 5
111. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein the population of subjects achieves a minimum clinically important difference (MCID) response rate for Total Activity Impairment (TAI) of at least about 45% by about week 10 12.
112. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein 15 the population of subjects achieves a minimum clinically important difference (MCID) response rate for Total Activity Impairment (TAI) of at least about 57% by about week 52.
113. A method of treating psoriasis in a population of subjects, comprising 20 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 65% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject was treated with a biologic prior to administration of the antibody. 25
114. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 74% of the population of subjects achieve at least a PASI 75 response by about 30 week 12, wherein each subject was treated with a biologic prior to administration of the antibody.
115. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion 35 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 78% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein none of the subjects were treated with a biologic prior to administration of the antibody. 283
116. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 5 least 82% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein none of the subjects were treated with a biologic prior to administration of the antibody.
117. A method of treating psoriasis in a population of subjects, comprising 10 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 78% of the population of subjects achieve at least a PGA 0/1 response by about week 52, wherein each subject was treated with a biologic prior to administration of the antibody. 15
118. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 79% of the population of subjects achieve at least a PGA 0/1 response by about 20 week 52, wherein none of the subjects were treated with a biologic prior to administration of the antibody.
119. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion 25 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 71% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject has a prior history of psoriatic arthritis.
120. A method of treating psoriasis in a population of subjects, comprising 30 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 78% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject has a prior history of psoriatic arthritis. 35
121. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 284 least 77% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein none of the subjects has a prior history of psoriatic arthritis.
122. A method of treating psoriasis in a population of subjects, comprising 5 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 81% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein none of the subjects has a prior history of psoriatic arthritis. 10
123. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 77% of the population of subjects achieve at least a PGA 0/1 response by about week 52, wherein each subject has a prior history of psoriatic arthritis. 15
124. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 79% of the population of subjects achieve at least a PGA 0/1 response by about 20 week 52, wherein none of the subjects has a prior history of psoriatic arthritis.
125. A method for decreasing the risk that a subject treated with an antibody, or antigen binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, will develop a Major Adverse Cardiovascular Event (MACE), comprising: 25 (a) selecting a subject having less than 2 risk factors selected from the group consisting of (i) a body mass index (BMI) of greater than 30, (ii) a history of diabetes mellitus, (iii) blood pressure greater than 140/90, (iv) a history of myocardial infarction, (v) a history of angina requiring hospitalization, (vi) a history of coronary artery disease requiring revascularization, (vii) a history of peripheral artery disease, (viii) a history of 30 congestive heart failure requiring hospitalization, (ix) a history of stroke or transient ischemic attack; and (b) administering the antibody, or antigen binding portion thereof to the selected subject, thereby decreasing the risk that the subject will develop a Major Adverse 35 Cardiovascular Events. 285
126. The method of claim 125 wherein the antibody is ABT-874.
127. The method of claim 125 or 126 wherein the subject has 1 risk factor. 5
128. The method of claim 125 or 126 wherein the subject has 0 risk factors.
129. The method of any one of the preceding claims wherein the MACE is myocardial infarction. 10
130. The method of any one of claims 125-128 wherein the MACE is cerebrovascular stroke.
131. The method of any one of the preceding claims wherein the antibody, or antigen binding portion thereof, is administered in a first dose amount of at least about 15 100 mg to about 200 mg.
132. The method of any one of the preceding claims wherein the antibody, or antigen binding portion thereof, is administered in a second dose amount of at least about 100 mg to about 200 mg. 20
133. The method of claim 132 wherein the risk factors are reevaluated prior to administration of the second dose amount to the subject.
134. The method of any one of claims 52-54 and 64-77, wherein the subject 25 achieves at least a 50% reduction in PASI score.
135. The method of any one of claims 52-54 and 64-77, wherein the subject achieves at least an 80% reduction in PASI score. 30
136. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion 286 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 69% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline PASI greater than 20 prior to administration of the antibody. 5
137. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 79% of the population of subjects achieve at least a PGA 0/1 response by about 10 week 12, wherein each subject had a baseline PASI less than or equal to 20 prior to administration of the antibody.
138. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion 15 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 79% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline PASI greater than 20 prior to administration of the antibody. 20
139. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 81% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline PASI less than or equal to 20 prior to 25 administration of the antibody.
140. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 30 least 67% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline weight of greater than or equal to 100 kilograms prior to administration of the antibody.
141. A method of treating psoriasis in a population of subjects, comprising 35 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 287 least 80% of the population of subjects achieve at least a PGA 0/1 response by about week 12, wherein each subject had a baseline weight of less than 100 kilograms prior to administration of the antibody. 5
142. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 72% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline weight of greater than or equal to 100 10 kilograms prior to administration of the antibody.
143. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 15 least 85% of the population of subjects achieve at least a PASI 75 response by about week 12, wherein each subject had a baseline weight of less than 100 kilograms prior to administration of the antibody.
144. A method of treating psoriasis in a population of subjects, comprising 20 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 41% of the population of subjects maintains at least a PGA 0/1 response through at least week 52 of treatment. 25
145. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 79% of the population of subjects maintains at least a PGA 0/1 response through at least week 52 of treatment. 30
146. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 45% of the population of subjects maintains at least a PASI 75 response through at 35 least week 52 of treatment. 288
147. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 5 least 82% of the population of subjects maintains at least a PASI 75 response through at least week 52 of treatment.
148. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion 10 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 23% of the population of subjects maintains at least a PASI 75 response through at least week 52 of treatment.
149. A method of treating psoriasis in a population of subjects, comprising 15 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 63% of the population of subjects maintains at least a PASI 75 response through at least week 52 of treatment. 20
150. The method of any one of claims 144, 146 and 148, comprising administering to each subject in the population: a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) administering a second dose amount that is about 40-60% of the first 25 dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks.
151. The method of any one of claims 145, 147 and 149, comprising administering to each subject in the population: 30 a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second periodicity of about once every 4 weeks; and 289 c) the second dose amount of the antibody, or antigen-binding portion thereof, according to a third periodicity of about once every 12 weeks.
152. The method of any one of claims 52-54 and 64-67, wherein the subject 5 achieves a PGA of 0 or 1 in less than about 171 days.
153. The method of claim 152, wherein the patient achieves a PGA of 0 or 1 by about 69 days. 10
154. The method of any one of claims 52-54 and 64-67, wherein the patient achieves a PASI 75 response in less than about 140 days.
155. The method of claim 154, wherein the patient achieves a PASI 75 by about 56 days. 15
156. The method of any one of claims 52-54 and 64-67, wherein the subject achieves at least a 60% improvement in PASI score and maintain at least a 60% improvement in PASI score through at least week 52 of treatment. 20
157. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 10% of the population of subjects achieves a PGA score of 0 by week 24 of treatment. 25
158. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 5% of the population of subjects achieve at least a PASI 50 response by about 30 week 2.
159. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 290 least 70% of the population of subjects achieve at least a PASI 50 response and maintain at least a PASI 50 response through at least week 52 of treatment.
160. A method of treating psoriasis in a population of subjects, comprising 5 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 5% of the population of subjects achieve at least a PASI 75 response by about week 4. 10
161. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 40% of the population of subjects achieve at least a PASI 75 response and maintain at least a PASI 75 response through at least week 52 of treatment. 15
162. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 10% of the population of subjects achieve at least a PASI 90 response by about 20 week 8.
163. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 25 least 25% of the population of subjects achieve at least a PASI 90 response and maintain at least a PASI 90 response through at least week 52 of treatment.
164. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion 30 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 5% of the population of subjects achieve at least a PASI 100 response by about week 8.
165. A method of treating psoriasis in a population of subjects, comprising 35 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 291 least 10% of the population of subjects achieve at least a PASI 100 response and maintain at least a PASI 100 response through at least week 52 of treatment.
166. A method of treating psoriasis in a population of subjects, comprising 5 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 5% of the population of subjects achieve at least a PGA score of 0 or 1 by about week 4. 10
167. A method of treating psoriasis in a population of subjects, comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 35% of the population of subjects achieve at least a PGA score of 0 or 1 and maintain at least a PGA score of 0 or 1 through at least week 52 of treatment. 15
168. The method of any one of claims 52-54 and 64-67, wherein the subject achieves a Nail Psoriasis Severity Index (NAPSI) score of about 2.1 or less.
169. The method of any one of claims 52-54 and 64-67, wherein the subject 20 achieves a Nail Psoriasis Severity Index (NAPSI) score of about 1.2 or less.
170. The method of any one of claims 52-54 and 64-67, wherein the subject achieves a Dermatology Life Quality Index (DLQI) score of about 0 or 1. 25
171. The method of any one of claims 52-54 and 64-67, wherein the subject achieves a clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score.
172. The method of claim 171, wherein the subject achieves a clinically 30 meaningful reduction in DLQI score by about week 24.
173. The method of claim 171, wherein the subject achieves a clinically meaningful reduction in DLQI score by about week 52. 292
174. The method of any one of claims 52-54 and 64-67, wherein the subject or population of subjects achieves an improvement in Dermatology Life Quality Index (DLQI) score of at least about -7 by week 12. 5
175. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 35% of the population of subjects achieves a Dermatology Life Quality Index 10 (DLQI) score of 0 or 1 by about week 24.
176. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at 15 least 18% of the population of subjects achieves a Dermatology Life Quality Index (DLQI) score of 0 or 1 by about week 52.
177. A method of treating psoriasis in a population of subjects comprising administering to each subject in the population an antibody, or antigen-binding portion 20 thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 50% of the population of subjects achieves a clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score by about week 24.
178. A method of treating psoriasis in a population of subjects comprising 25 administering to each subject in the population an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, wherein at least 20% of the population of subjects achieves a clinically meaningful reduction in Dermatology Life Quality Index (DLQI) score by about week 52. 30
179. The method of any one of claims 52-54 and 64-77, wherein the subject achieves a minimum clinically important difference (MCID) in any one or more health related quality of life outcomes selected from the group consisting of Dermatology Life Quality Index (DLQI), Total Activity Impairment (TAI), Ps-related (VAS-Ps) pain, psoriatic arthritis-related (VAS-PsA) pain, Short Form 36 Health Survey Mental 293 Component Summary score (MCS) and Short Form 36 Health Survey Mental Component Summary score (PCS).
180. The method of any one of the preceding claims, wherein the method 5 comprises administering to the subject or to each subject in the population: a) a first dose amount of the antibody, or antigen-binding portion thereof, according to a first periodicity of about once every 4 weeks; and b) administering a second dose amount that is about 40-60% of the first dose amount of the antibody, or antigen-binding portion thereof, according to a second 10 periodicity of about once every 4 weeks.
181. The method of method of any one of the preceding claims, wherein the method comprises administering to the subject or to each subject in the population: a) about 200 mg of ABT-874 once every four weeks for two doses; and 15 b) about 100 mg of ABT-874 every four weeks thereafter.
182. The method of method of any one of the preceding claims, wherein the method comprises administering to the subject or to each subject in the population: a) about 200 mg of ABT-874 at weeks 0 and 4; and 20 b) about 100 mg of ABT-874 at week 8 and every 4 weeks thereafter.
183. A method of treating psoriasis in a subject comprising administering to the subject: a) about 200 mg of an antibody, or antigen-binding portion thereof, which is 25 capable of binding to the p40 subunit of IL- 12 and/or IL-23, once every four weeks for two doses; and b) about 100 mg of the antibody, or antigen-binding portion thereof, every four weeks thereafter, thereby treating psoriasis in the subject. 30
184. A method of treating psoriasis in a subject comprising administering to the subject: a) about 200 mg of an antibody, or antigen-binding portion thereof, which is capable of binding to the p40 subunit of IL- 12 and/or IL-23, at weeks 0 and 4; and 35 b) about 100 mg of the antibody, or antigen-binding portion thereof, at week 8 and every 4 weeks thereafter, thereby treating psoriasis in the subject. 294
185. The method of claim 183 or 184, wherein the antibody is ABT-874.
186. A method of treating psoriasis in a subject comprising administering to 5 the subject: a) about 200 mg of ABT-874 once every four weeks for two doses; and b) about 100 mg of ABT-874 every four weeks thereafter, thereby treating psoriasis in the subject. 10
187. A method of treating psoriasis in a subject comprising administering to the subject: a) about 200 mg of ABT-874 at weeks 0 and 4; and b) about 100 mg of ABT-874 at week 8 and every 4 weeks thereafter, thereby treating psoriasis in the subject. 15 20
AU2013202866A 2009-09-14 2013-04-08 Methods for treating psoriasis Abandoned AU2013202866A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013202866A AU2013202866A1 (en) 2009-09-14 2013-04-08 Methods for treating psoriasis

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US61/242,288 2009-09-14
US61/245,967 2009-09-25
US61/297,623 2010-01-22
US61/360,299 2010-06-30
AU2010291927A AU2010291927A1 (en) 2009-09-14 2010-09-14 Methods for treating psoriasis
AU2013202866A AU2013202866A1 (en) 2009-09-14 2013-04-08 Methods for treating psoriasis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2010291927A Division AU2010291927A1 (en) 2009-09-14 2010-09-14 Methods for treating psoriasis

Publications (1)

Publication Number Publication Date
AU2013202866A1 true AU2013202866A1 (en) 2013-05-02

Family

ID=48414292

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013202866A Abandoned AU2013202866A1 (en) 2009-09-14 2013-04-08 Methods for treating psoriasis

Country Status (1)

Country Link
AU (1) AU2013202866A1 (en)

Similar Documents

Publication Publication Date Title
US8178092B2 (en) Methods of treating psoriasis by administration of antibodies to the p40 subunit of IL-12 and/or IL-23
US8557239B2 (en) Methods for treating psoriasis using antibodies that bind to the P40 subunit of IL-12 and/or IL-23
US7776331B1 (en) Methods of treating plaque psoriasis
US20120189637A1 (en) Methods for treating psoriasis
US20140093516A1 (en) Human antibodies that bind the p40 subunit of human il-12/il-23 and uses therefor
AU2013202866A1 (en) Methods for treating psoriasis
AU2013202860A1 (en) Methods for treating psoriasis
AU2013202849A1 (en) Methods for treating psoriasis
AU2013203142A1 (en) Methods for treating psoriasis

Legal Events

Date Code Title Description
PC1 Assignment before grant (sect. 113)

Owner name: ABBOTT LABORATORIES; ABBVIE DEUTSCHLAND GMBH & CO.

Free format text: FORMER APPLICANT(S): ABBOTT LABORATORIES; ABBOTT GMBH & CO. KG

PC1 Assignment before grant (sect. 113)

Owner name: ABBVIE INC.; ABBVIE DEUTSCHLAND GMBH & CO. KG

Free format text: FORMER APPLICANT(S): ABBVIE DEUTSCHLAND GMBH & CO. KG; ABBOTT LABORATORIES

MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application