WO2007076933A1 - Interleukin-12p40 variants with improved stability - Google Patents

Interleukin-12p40 variants with improved stability Download PDF

Info

Publication number
WO2007076933A1
WO2007076933A1 PCT/EP2006/012301 EP2006012301W WO2007076933A1 WO 2007076933 A1 WO2007076933 A1 WO 2007076933A1 EP 2006012301 W EP2006012301 W EP 2006012301W WO 2007076933 A1 WO2007076933 A1 WO 2007076933A1
Authority
WO
WIPO (PCT)
Prior art keywords
variant
amino acid
protein
substitutions
variants
Prior art date
Application number
PCT/EP2006/012301
Other languages
French (fr)
Inventor
Gordon D. Webster
Suzanne P. Mckenzie
Kin-Ming Lo
Pascal André STEIN
Original Assignee
Merck Patent Gmbh
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 to ES06846989T priority Critical patent/ES2384055T3/en
Priority to DK06846989T priority patent/DK1966238T3/en
Priority to JP2008547880A priority patent/JP5175214B2/en
Priority to CA2635618A priority patent/CA2635618C/en
Priority to AU2006332138A priority patent/AU2006332138B2/en
Priority to AT06846989T priority patent/ATE555125T1/en
Priority to EP20060846989 priority patent/EP1966238B1/en
Priority to EA200870131A priority patent/EA015338B1/en
Application filed by Merck Patent Gmbh filed Critical Merck Patent Gmbh
Priority to PL06846989T priority patent/PL1966238T3/en
Priority to CN2006800498186A priority patent/CN101351475B/en
Priority to SI200631366T priority patent/SI1966238T1/en
Priority to BRPI0620648-4A priority patent/BRPI0620648B1/en
Publication of WO2007076933A1 publication Critical patent/WO2007076933A1/en
Priority to KR1020087018718A priority patent/KR101343682B1/en

Links

Classifications

    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04NPICTORIAL COMMUNICATION, e.g. TELEVISION
    • H04N21/00Selective content distribution, e.g. interactive television or video on demand [VOD]
    • H04N21/40Client devices specifically adapted for the reception of or interaction with content, e.g. set-top-box [STB]; Operations thereof
    • H04N21/43Processing of content or additional data, e.g. demultiplexing additional data from a digital video stream; Elementary client operations, e.g. monitoring of home network or synchronising decoder's clock; Client middleware
    • H04N21/435Processing of additional data, e.g. decrypting of additional data, reconstructing software from modules extracted from the transport stream
    • H04N21/4353Processing of additional data, e.g. decrypting of additional data, reconstructing software from modules extracted from the transport stream involving decryption of additional data
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06QINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES; SYSTEMS OR METHODS SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES, NOT OTHERWISE PROVIDED FOR
    • G06Q20/00Payment architectures, schemes or protocols
    • G06Q20/08Payment architectures
    • G06Q20/10Payment architectures specially adapted for electronic funds transfer [EFT] systems; specially adapted for home banking systems
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06QINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES; SYSTEMS OR METHODS SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES, NOT OTHERWISE PROVIDED FOR
    • G06Q30/00Commerce
    • G06Q30/02Marketing; Price estimation or determination; Fundraising
    • G06Q30/0207Discounts or incentives, e.g. coupons or rebates
    • G06Q30/0215Including financial accounts
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06QINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES; SYSTEMS OR METHODS SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES, NOT OTHERWISE PROVIDED FOR
    • G06Q30/00Commerce
    • G06Q30/02Marketing; Price estimation or determination; Fundraising
    • G06Q30/0207Discounts or incentives, e.g. coupons or rebates
    • G06Q30/0226Incentive systems for frequent usage, e.g. frequent flyer miles programs or point systems
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06QINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES; SYSTEMS OR METHODS SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES, NOT OTHERWISE PROVIDED FOR
    • G06Q30/00Commerce
    • G06Q30/04Billing or invoicing
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06QINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES; SYSTEMS OR METHODS SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES, NOT OTHERWISE PROVIDED FOR
    • G06Q40/00Finance; Insurance; Tax strategies; Processing of corporate or income taxes
    • G06Q40/12Accounting
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04LTRANSMISSION OF DIGITAL INFORMATION, e.g. TELEGRAPHIC COMMUNICATION
    • H04L41/00Arrangements for maintenance, administration or management of data switching networks, e.g. of packet switching networks
    • H04L41/32Specific management aspects for broadband networks
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04LTRANSMISSION OF DIGITAL INFORMATION, e.g. TELEGRAPHIC COMMUNICATION
    • H04L47/00Traffic control in data switching networks
    • H04L47/10Flow control; Congestion control
    • H04L47/24Traffic characterised by specific attributes, e.g. priority or QoS
    • H04L47/2491Mapping quality of service [QoS] requirements between different networks
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04NPICTORIAL COMMUNICATION, e.g. TELEVISION
    • H04N21/00Selective content distribution, e.g. interactive television or video on demand [VOD]
    • H04N21/40Client devices specifically adapted for the reception of or interaction with content, e.g. set-top-box [STB]; Operations thereof
    • H04N21/45Management operations performed by the client for facilitating the reception of or the interaction with the content or administrating data related to the end-user or to the client device itself, e.g. learning user preferences for recommending movies, resolving scheduling conflicts
    • H04N21/462Content or additional data management, e.g. creating a master electronic program guide from data received from the Internet and a Head-end, controlling the complexity of a video stream by scaling the resolution or bit-rate based on the client capabilities
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04NPICTORIAL COMMUNICATION, e.g. TELEVISION
    • H04N21/00Selective content distribution, e.g. interactive television or video on demand [VOD]
    • H04N21/60Network structure or processes for video distribution between server and client or between remote clients; Control signalling between clients, server and network components; Transmission of management data between server and client, e.g. sending from server to client commands for recording incoming content stream; Communication details between server and client 
    • H04N21/61Network physical structure; Signal processing
    • H04N21/6106Network physical structure; Signal processing specially adapted to the downstream path of the transmission network
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04NPICTORIAL COMMUNICATION, e.g. TELEVISION
    • H04N21/00Selective content distribution, e.g. interactive television or video on demand [VOD]
    • H04N21/80Generation or processing of content or additional data by content creator independently of the distribution process; Content per se
    • H04N21/85Assembly of content; Generation of multimedia applications
    • H04N21/854Content authoring
    • H04N21/8549Creating video summaries, e.g. movie trailer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • HELECTRICITY
    • H04ELECTRIC COMMUNICATION TECHNIQUE
    • H04LTRANSMISSION OF DIGITAL INFORMATION, e.g. TELEGRAPHIC COMMUNICATION
    • H04L41/00Arrangements for maintenance, administration or management of data switching networks, e.g. of packet switching networks
    • H04L41/50Network service management, e.g. ensuring proper service fulfilment according to agreements
    • H04L41/5003Managing SLA; Interaction between SLA and QoS
    • H04L41/5019Ensuring fulfilment of SLA

Definitions

  • This invention relates generally to IL-12p40 proteins, including fusion proteins containing IL-12p40, modified to improve their stability.
  • the IL-12p40 proteins of the invention remove a proteolytic site in the region of Lys260 and Arg261 in the p40 subunit.
  • lnterleukin-12 is an inflammatory cytokine that is produced in response to infection by a variety of cells of the immune system, including phagocytic cells, B cells and activated dendritic cells (Colombo and Trinchieri (2002), Cytokine & Growth Factor Reviews, 13: 155-168).
  • IL-12 plays an essential role in mediating the interaction of the innate and adaptive arms of the immune system, acting on T-cells and natural killer (NK) cells, enhancing the proliferation and activity of cytotoxic lymphocytes and the production of other inflammatory cytokines, especially interferon- ⁇ (IFN- ⁇ ).
  • IFN- ⁇ interferon- ⁇
  • IL-12 is a heterodimeric molecule composed of an ⁇ -chain (the p35 subunit, IL-12p35) and a ⁇ -chain (the p40 subunit, IL-12p40) covalently linked by a disulfide bridge to form the biologically active 74 kDa heterodimer.
  • Amino acid sequences of IL-12p35 and IL-12p40 of a mature (wild-type) human IL-12 are depicted in Figures 1 (SEQ ID NO:1) and 2 (SEQ ID NO:2), respectively.
  • lnterleukin-23 (IL-23) is a disulfide-bridged heterodimeric molecule closely related to IL-12, in that it has the same ⁇ chain IL-12p40 as IL-12, but a unique ⁇ chain (the p19 subunit, IL-23p19) (Oppmann et al., (2000), Immunity, 13: 715-725).
  • IL-23 is produced by phagocytic cells and activated dendritic cells, and is believed to be involved in the recruitment and activation of a range of inflammatory cells (Langrish et al., (2004) Immunol. Rev., 202: 96-105).
  • the amino acid sequence of IL-23p19 of a mature human IL-23 is depicted in Figure 3 (SEQ ID NO:3).
  • IL-12 has been demonstrated to have a potent antitumor activity based upon the induction of IFN- ⁇ and the activation of effector cells such as CD8+ T-cells and NK cells (Brunda et al. (1993), J. Exp. Med., 178: 1223-30). As a result of its demonstrated anti-tumor activity, IL-12 has been tested in human clinical trials as an immunotherapeutic agent for the treatment of a wide variety of cancers (Atkins et al. (1997), Clin. Cancer Res., 3: 409-17; Gollob et al. (2000), Clin. Cancer Res., 6: 1678-92; and Hurteau et al. (2001), Gynecol.
  • IL-12 or IL-23 protein can give rise to problems in its production and clinical use as a therapeutic agent. Therefore, there is a need in the art for improved recombinant IL-12 or IL-23 variants that yield a homogeneous protein more resistant to proteolytic cleavage.
  • the invention provides variants of human IL-12 p40 subunits (p40 variants) which have improved stability compared to wild-type IL-12 p40 proteins.
  • the C-terminal region which is normally sensitive to proteolytic cleavage, has been engineered to be more resistant to digestion by proteases.
  • p40 variants of the invention include engineered amino acid alterations in the D3 domain aimed to avoid the creation of potential T-cell epitopes that could make the variant proteins immunogenic and trigger antibody responses in humans.
  • p40 variants of the invention have improved properties as therapeutic agents over wild-type IL-12p40 proteins with regard to their production, formulation, and pharmacokinetics.
  • the invention provides a variant of a human IL-12 p40 D3 domain (D3 variant), wherein the D3 variant has at least 85% identity with a wild-type human IL-12p40 D3 domain and includes an amino acid alteration at one or more positions corresponding to residues 258-266 of mature human IL-12 p40.
  • D3 variant has at least 85% identity with a wild-type human IL-12p40 D3 domain and includes an amino acid alteration at one or more positions corresponding to residues 258-266 of mature human IL-12 p40.
  • amino acid alterations to one or more positions corresponding to residues 258-266 may be deletions, substitutions, or insertions.
  • amino acid substitutions that replace basic amino acids with non-basic amino acids can be used to create variants according to the invention.
  • D3 variants of the invention may include one or more amino acid substitutions at positions selected from the group consisting of Lys258, Ser259, Lys260, Arg261, Lys263, Lys 264, Asp265, and Arg266.
  • amino acid alterations can be used singly or in combination to induce the structural and/or functional changes described above.
  • the D3 variant can incorporate one, two, three, four or more of the following substitutions: Lys258Gln, Ser259Asp, Lys260Ala, Lys260Asn, Lys260Gln, Lys260Gly, Arg261Ala, Arg261Asp, Arg261Thr, Lys263Gly, Lys263Ser, and/or Lys264Gly.
  • the substitution is a position Lys260.
  • the substitution may replace Lys260 with a non-basic amino acid, for example, Ala, Asn, GIn, or GIy. Further substitutions in addition to Lys260 may occur at Ser259 and Arg261.
  • D3 variants of the invention incorporate substitutions Ser259Asp, Lys260Asn, and Arg261Thr.
  • D3 variants of the invention incorporate substitutions Ser259Asp, Lys260Asn, Arg261Thr and Lys264Gly, while optionally deleting Lys263 and Asp265.
  • a D3 variant of the invention incorporates substitutions Ser259Asp, Lys260Asn, Arg261Thr, and Lys264Gly while deleting Lys263, Lys264 and Asp265.
  • a D3 variant including a substitution replacing Lys260 alternatively includes further substitutions at one or more of Lys258, Ser259, Arg261 , Lys263, and Lys264.
  • a D3 variant includes the substitutions Lys258Gln, Ser259Asp, Lys260Gln, Arg261 Asp, and optionally Lys263Ser and Lys264Gly.
  • one or more of residues corresponding to Lys263, Lys264, Asp265, and Arg266 are deleted, while in another embodiment, one or more of Lys263, Lys264, Asp265, and Arg266 are substituted with a non-basic amino acid.
  • the substitution at Lys264 is Lys264Gly and, optionally, Lys263 and Asp265 are deleted.
  • D3 variants of the invention incorporate substitutions Ser259Asp, Lys260Asn, Arg261Thr, and Lys264Gly, and optionally, deletion of residues corresponding to Lys263, Asp265 and Arg 266.
  • p40 variants and active portions thereof that incorporate a D3 variant as described herein are within the scope of the invention.
  • IL-12 proteins and active portions thereof that contain a p40 variant also are within the scope of the invention.
  • the invention further encompasses fusion proteins including IL-12 variants of the invention and a moiety selected from the group consisting of an antibody moiety, a non-IL-12 cytokine, or an active portion thereof.
  • IL-23 proteins and active portions thereof that contain a p40 variant also are within the scope of the invention.
  • the invention further encompasses fusion proteins including IL-23 variants of the invention and a moiety selected from the group consisting of an antibody moiety, a non-IL-23 cytokine, or an active portion thereof.
  • the invention relates to a nucleic acid that encodes any of the D3 variants, p40 variants, IL-12 variants, and IL-23 variants of the invention.
  • the invention further encompasses a cell, e.g., a prokaryotic cell, including such a nucleic acid.
  • the invention also features methods of making such D3 variants, p40 variants, IL-12 variants, IL-23 variants and fusion proteins containing these moieties.
  • the invention provides methods of using the variants of the invention and the nucleic acids encoding same.
  • the invention encompasses a method of treating a patient that includes administering to the patient a therapeutically effective amount of a p40 variant of the invention or an active portion thereof.
  • the invention relates to: • A variant of an IL-12 p40 D3 domain, wherein the variant has at least 85% sequence identity with a wild-type human IL-12 p40 D3 domain and comprises an amino acid alteration at one or more positions corresponding to residues at positions 258-266 of the parent mature human IL-12 p40.
  • a corresponding variant wherein the alteration is a substitution selected from the group consisting of Lys258, Lys260, Lys263, and Lys264. • A corresponding variant, wherein one or more of the following amino acid substitutions are selected:
  • Lys260Ala Lys260Asn, Lys260Gln, Lys260Gly,
  • Lys263Gly Lys263Ser, and Lys264Gly.
  • a corresponding corresponding variant wherein the substitution is at least at position Lys260.
  • a corresponding variant wherein the substitution replaces a basic amino acid with a non-basic amino acid, selected from the group consisting of Ala, Asn, GIn, or GIy.
  • variant further comprises one or more substitutions at positions Ser259, Arg261 or Arg 266.
  • a corresponding variant further comprising one or more substitutions selected from the group consisting of:
  • a corresponding variant comprising the following substitutions: Lys258Gln, Ser259Asp_, Lys260Gln, Arg261Asp_, Lys263Ser, Lvs264Gly. thus forming the sequence track: GIn - Asp - GJn - Asp - GIu - Ser- GIy - Arg at positions 258 - 265 of the variant.
  • a fusion protein comprising a variant, or an IL-12 or IL-23 protein as described , and an antibody or an active portion thereof.
  • a corresponding fusion protein wherein said variant or said protein is fused to the C-terminus of said antibody or antibody portion, preferably to the Fc-portion of said antibody, wherein said antibody targets tumor antigens, such as KS (EpCam) or 14.18, or receptors that are overexpressed on tumor tissue, such as HER2 or EGFR or VEGFR.
  • tumor antigens such as KS (EpCam) or 14.18
  • receptors that are overexpressed on tumor tissue, such as HER2 or EGFR or VEGFR.
  • An expression vector comprising said nucleic acid. • A host cell producing a variant or an IL-12 or IL-23 protein or a fusion protein as described.
  • a pharmaceutical composition suitable for the treatment of IL-12 or IL-23 triggered diseases comprising in an pharmacologically effective amount a variant, an IL-12 or IL-23 protein , or a fusion protein comprising such a variant or IL-12 or IL-23 protein, optionally together with a pharmaceutically acceptable carrier diluent or excipient, or with other pharmacologically effective drugs in a combination therapy regimen.
  • Figure 1 depicts the mature amino acid sequence of the ⁇ chain, i.e., the p35 subunit, of a mature (wild-type) human IL-12 (SEQ ID NO:1 ).
  • Figure 2 depicts the mature amino acid sequence of the ⁇ chain, i.e., the p40 subunit, of a mature (wild-type) human IL-12 (SEQ ID NO:2).
  • Domain D3 corresponding to positions 211-306 (SEQ ID NO:26), is italicized, and the peptide fragment corresponding to positions 258-266 is underlined (SEQ ID NO:5), with Lys260 and Arg261 highlighted in bold.
  • Figure 3 depicts the mature amino acid sequence of the ⁇ chain, i.e., the p19 subunit, of a mature (wild-type) human IL-23 (SEQ ID NO:3).
  • Figure 4A shows the SDS-PAGE gel for several purified batches of human IL-12 produced as antibody fusion proteins (lanes 1 - 8), while Figure 4B shows the SDS-PAGE gel for several purified batches of human IL-23 produced as antibody fusion proteins (lanes 1 - 3).
  • the IL-12p35 and IL-23p19 subunit is covalently attached to the antibody heavy chain.
  • the 6kD band is indicated by an arrow.
  • Molecular weights (kD) are indicated for the markers (lane M).
  • Figure 5 depicts the amino acid sequence of the C-terminal peptide fragment of mature (wild-type) human IL-12p40 subunit. The fragment starts at Arg261 (SEQ ID NO:4).
  • Figure 6 depicts the amino acid sequence of a peptide fragment corresponding to positions 258-266 of a mature (wild-type) human IL-12p40 subunit (SEQ ID NO:5).
  • Figures 7.1 and 7.2 depict an amino acid sequence alignment of IL- 12p40 subunits from various mammals including human, baboon (Papio anubis), rhesus monkey (Macaca mulatta), mangabey (Cercocebus torquatos), dog (Canis familiaris), cat (Felis catus), horse (Equus caballus), pig (Sus scrota), cow (Bos Taurus), goat (Capra hircus), sheep (Ovis aries), deer (Cervus elaphus), water buffalo [Bubalus bubalis), hamster [Mesocricetus auratus), guinea pig (Cavia porcellus), cotton rat (Sigmodon hispidus), rat (Rattus norvegicus), and mouse (Mus musculus).
  • the two amino acids in bold indicate the proteolytic cleavage site.
  • Figure 8 depicts the amino acid sequence of a variant, referred to herein as p40V1 , of a mature human IL-12p40 subunit (SEQ ID NO:6). The engineered sequence is underlined.
  • Figure 9 depicts the amino acid sequence of a variant, referred to herein as p40V2, of a mature human IL-12p40 subunit (SEQ ID N0.7). The engineered sequence is underlined.
  • Figure 10 depicts the amino acid sequence of a variant, referred to herein as p40V3, of a mature human IL-12p40 subunit (SEQ ID NO:8).
  • the engineered sequence is underlined.
  • Figure 11 depicts the amino acid sequence of a variant, referred to herein as p40V4, of a mature human IL-12p40 subunit (SEQ ID NO:9). The engineered sequence is underlined.
  • Figure 12 depicts the amino acid sequence of a variant, referred herein as p40V5, of a mature human IL-12p40 subunit (SEQ ID NO: 10). The alteration is underlined.
  • Figure 13 depicts the amino acid sequence of a variant, referred herein as p40V6, of a mature human IL-12p40 subunit (SEQ ID NO:11). The alterations are underlined.
  • Figure 14 depicts the amino acid sequence of a variant, referred herein as p40V7, of a mature human IL-12p40 subunit (SEQ ID NO: 12). The alteration is underlined.
  • Figure 15 depicts the amino acid sequence of a variant, referred herein as p40V8, of a mature human IL-12p40 subunit (SEQ ID NO:13). The alteration is underlined.
  • Figure 16 depicts the amino acid sequence of a variant, referred herein as p40V9, of a mature human IL-12p40 subunit (SEQ ID NO:14). The alteration is underlined.
  • Figure 17 depicts the amino acid sequence of a variant, referred herein as p40V10, of a mature human IL-12p40 subunit (SEQ ID NO: 15). The alteration is underlined.
  • Figure 18 depicts the amino acid sequence of a variant, referred herein as p40V11 , of a mature human IL-12p40 subunit (SEQ ID NO:16). The alterations are underlined.
  • Figure 19 depicts the nucleic acid sequence encoding the full length (wild-type) human IL-12p40 subunit (SEQ ID NO:21).
  • Figure 20 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred to herein as V1V2, encoding portions of p40 variants p40V1 and p40V2.
  • the V1V2 fragment encompasses a V1 fragment including a region (underlined) encoding SEQ ID NO:17. This is followed by a linker sequence (lower case), and subsequently, a V2 fragment including a region (underlined) encoding SEQ ID NO:18.
  • Figure 21 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred to herein as V3V4, encoding portions of p40 variants p40V3 and p40V4.
  • the V3V4 fragment encompasses a V3 fragment including a region (underlined) encoding SEQ ID NO: 19. This is followed by a linker sequence (lower case), and subsequently, a V4 fragment including a region (underlined) encoding SEQ ID NO:20.
  • FIG. 22 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred herein as V5V6, encoding portions of p40 variants p40V5 and p40V6.
  • the V5V6 fragment encompasses a V5 fragment including a codon substitution (underlined) encoding Arg261Ala. This is followed by a linker sequence (lower case), and subsequently, a V6 fragment including codon substitutions (underlined) encoding Lys260Ala and Arg261Ala.
  • Figure 23 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred herein as V7V8, encoding portions of p40 variants p40V7 and p40V8.
  • the V7V8 fragment encompasses a V7 fragment including a codon substitution (underlined) encoding Lys260Ala. This is followed by a linker sequence (lower case), and subsequently, a V8 fragment including a codon substitution (underlined) encoding Lys260Gly.
  • Figure 24 is a Western blot with a polyclonal anti-hu-p40 antibody.
  • Supematants of cells transfected with wild-type IL-12p40 and IL-12p40 variants were harvested and processed on an SDS-PAGE gel.
  • Two independent clones (a, b) of each IL-12p40 variant p40V1 , p40V2, p40V3, and p40V4 were tested.
  • the arrow points to the band of cleaved IL12p40 lacking C-terminal fragment (lane p40 wt).
  • Figure 25 shows the SDS-PAGE gel for antibody-IL12 fusion proteins containing p40 variants p40V1 , p40V2, p40V3, p40V4, p40V5, p40V6, p40V7, p40V8, and wild type p40 (lanes 1-9).
  • the upper most main band represents the fusion protein between the antibody heavy chain and the p35 subunit, and the lower main band represents the antibody light chain.
  • the 6 kDa band was not detected in any of the lanes of the variant proteins, except lane 5.
  • Molecular weights (kD) are indicated for the markers (lane M).
  • Figure 26 shows pharmacokinetic data of antibody-IL12 fusion proteins containing p40 variants p40V1- p40V8 compared to antibody-(wild-type)-IL12 fusion proteins, administered intravenously.
  • Figure 27A shows pharmacokinetic data of antibody-IL12 fusion proteins containing p40 variants p40V1-p40V4 compared to antibody-(wild-type)- IL12 fusion proteins (panel A) and
  • Figure 27B shows pharmacokinetic data of antibody-IL12 fusion proteins containing p40 variants p40V5 - p40V8 compared to antibody-(wild-type)-IL12 fusion proteins (panel B), administered subcutaneously.
  • Figure 28 is an IL-12p40 variant having mutations outside the D3 region of the protein.
  • the invention describes variants of the cytokine interleukin-12 (IL-12) p40 subunit which have improved stability compared with the wild-type protein.
  • IL-12 cytokine interleukin-12
  • a region of the p40 subunit which is normally unstructured and sensitive to proteolytic cleavage is mutated to be more resistant to proteolytic cleavage.
  • This region, in domain D3, is near the C-terminus of the p40 subunit, encompassing a polypeptide stretch corresponding to amino acids 258-266 in mature human p40 (p40(258-266)).
  • the IL-12p40 subunit is also a component subunit of the cytokine interleukin-23 (IL-23).
  • IL-23 has two subunits, the ⁇ subunit "p19" and the ⁇ subunit "p40.”
  • the p40 subunit of IL-23 is the same as IL-12p40. Therefore, variants of IL-12p40 subunit are likewise variants of the IL-23p40 subunit.
  • one or more mutations are introduced into the region of p40 corresponding to amino acid residues 258-266 to eliminate the cleavage site, which in human p40 corresponds to the site between Lys260 and Arg261.
  • specific mutations are introduced in this region that by modeling generate a more tightly folded structure.
  • the introduced mutations additionally are predicted to avoid making the engineered region of the p40 subunit immunogenic.
  • amino acid substitutions in the engineered region of the p40 subunit are chosen to avoid the creation of peptides that may be recognized as potential T-cell epitopes in humans.
  • Mutations may be introduced into the region of p40 corresponding to amino acid residues 258-266 by a variety of mechanisms.
  • a mutation or mutations are introduced by substitution of one amino acid residue for another.
  • a mutation or mutations are introduced by deletion of one or more residues of the p40 subunit.
  • a mutation or mutations are introduced by insertion of one or more amino acid residues into the p40 subunit.
  • the p40 variants of the invention are contained in protein compositions such as IL-12 proteins, IL-12 fusion proteins, IL-23 proteins, IL-23 fusion proteins or p40 homodimers.
  • an IL-12 protein contains a p35 subunit and a p40 variant according to the invention.
  • the IL-23 protein contains a p19 subunit and p40 variant according to the invention.
  • a fusion protein contains an antibody portion fused to IL-12 containing an IL-12p40 variant.
  • a fusion protein contains an antibody portion fused to IL-23 containing an IL-12p40 variant.
  • the antibody portion of the fusion protein is an intact antibody, an Fc region, an sc-Fv, an antigen binding portion of an antibody, or an active fragment of an antibody.
  • a fusion protein according to the invention includes an IL-12p40 variant fused to an non-IL-12 or non-IL-23 cytokine or active portion thereof.
  • protein compositions that contain one of the p40 variants of the invention have a longer circulating half-life than the corresponding wild-type protein.
  • IL-12 variants or IL-23 variants that include an engineered p40 subunit of the invention have improved properties as therapeutic agents with regard to their production, formulation and pharmacokinetics.
  • mutations to the IL-12p40 amino acid sequence are introduced outside the IL-12p40(258-266) region and optionally outside the IL-12p40 D3 domain of IL-12p40. Such mutations can be introduced elsewhere in the D3 domain of IL-12p40 and can be introduced in the other domains.
  • Figure 28 depicts a sequence of a p40 subunit of IL-12 that has alterations in the amino acid sequence outside of residues 258-266.
  • the three dimensional crystal structure of IL-12p40 is known (Yoon et al., (2000), EMBO J..
  • the invention rests in part on the observation that the IL-12p40 subunit is susceptible to a specific proteolytic cleavage event, and on novel experimental results defining the cleavage site. It was found that a purified recombinant IL-12 protein, produced in NS/0 cells as described in Example 2, consistently exhibited a degree of heterogeneity when the recombinant protein was separated by electrophoresis on an SDS-PAGE gel under reducing conditions, clearly visible as an additional protein band of approximately 6 kD molecular weight. A similar observation was made for recombinant IL-23 protein.
  • Figure 4 shows the SDS-PAGE gel for several purified batches of human IL-12 (Panel A) and human IL-23 protein compositions (Panel B) produced as antibody fusion proteins.
  • the contaminant was purified and its amino acid sequence was determined, as described in Example 3, and was found to correspond to the sequence of the C-terminal 46 amino acid fragment of the IL-12p40 subunit itself, generated by proteolytic cleavage between Lys260 and Arg261 ( Figure 5-SEQ ID NO:4). Cleavage between Lys260 and Arg261 appears to be highly favored despite a prevalence of basic amino acids in the region surrounding the cleavage site (...KSKREKKDR...
  • the mature human p40 subunit is a 306 amino acid protein resembling a soluble class I cytokine D receptor, composed of domains D1 , D2 and D3.
  • the cleavage site (between Lys260 and Arg261 ) is within D3, a fibronectin type III domain of 96 amino acids encompassing the region from 1211 to S306.
  • the sequence for mature human IL-12 p40 subunit is shown in Figure 2.
  • the amino acid sequence for the D3 is shown in Figure 2 in italics. In the published X-ray crystallographic structure of the human IL-12 heterodimer (Yoon et al.
  • FIG. 7.1 and 7.2 A primary structure alignment of the mature p40 subunit from a variety of mammalian species is shown in Figures 7.1 and 7.2, including human; the primates baboon, rhesus monkey and mangabey; dog; cat; horse; pig; the ruminants cow, goat, sheep, deer, water buffalo; and the rodents hamster, guinea pig, cotton rat, rat and mouse.
  • the region around p40(258-266) exhibits sequence variability, particularly with respect to its length. Particularly, in some species, notably ruminants, the sequence is shorter, and in others, such as in rodents, the sequence in some instances may contain an additional insert.
  • the dipeptide motif corresponding to human p40 K260R261 is conserved, either identically or exhibiting two basic amino acids.
  • These species include agriculturally and commercially important species including, but not limited to, the horse, cow, goat, pig, and sheep. Consequently, introducing one or more mutations into the region of non-human IL-12p40 corresponding to residues 258-266 of human IL-12p40 that are analogous to mutations taught herein with respect to human IL-12p40 may prove useful in reducing or eliminating proteolytic cleavage of non-human IL-12p40 at the K260R261 cleavage site.
  • IL-12p40 variant from a species that lacks a positively charged dipeptide motif corresponding to human IL-12p40 K260R261 in a human or other heterologous organism.
  • non-human forms of IL-12p40 will generally lead to anti-p40 antibodies when administered to humans. More broadly, it is not optimal to administer p40 from one species to another.
  • the potential of various non-human p40 subunits to be proteolytically cleaved is generally unknown.
  • p40 subunits from one species may not function in another species, either at the step of assembly with subunits such as p35 or p19, or at the step of interaction with receptor subunits.
  • the invention provides for variant IL-12p40 proteins with mutations in the D3 domain that improve stability.
  • D3 variant refers to a D3 domain of a human p40 subunit of, for example, IL-12 or IL-23, having one or more amino acid alterations as compared to wild-type D3.
  • p40 variant is used herein to refer to a human p40 subunit of, for example, IL-12 or IL-23, with mutations in the D3 domain, i.e., a p40 subunit containing a D3 variant.
  • IL-12 variant is used herein to refer to a human IL-12 protein containing a p40 variant.
  • IL-23 variant is used herein to refer to a human IL-23 protein containing a p40 variant.
  • the D3 domain of a p40 variant has at least 70% or more sequence identity with the D3 domain of wild- type IL-12p40. In a further embodiment, the D3 domain of a p40 variant has at least 75% or more sequence identity with the D3 domain of wild-type IL-12p40.
  • the D3 domain of a p40 variant has at least 80% or more sequence identity with the D3 domain of wild-type IL-12p40, while in further embodiments, the D3 domain of a p40 variant has at least 81 % or more, or at least 82% or more, or at least 83% or more, or at least 84% or more, or at least 85% or more, or at least 86% or more, or at least 87% or more, or at least 88% or more, or at least 89% or more, or at least 90% or more, or at least 91 % or more, or at least 92% or more, or at least 93% or more, or at least 94% or more, or at least 95% or more, or at least 96% or more, or at least 97% or more, or at least 98% or more, or at least 99% or more identity with the D3 domain of wild-type IL- 12p40.
  • the amino acid sequence of a p40 variant has at least 70% or more sequence identity with the amino acid sequence of mature wild-type IL-12 p40. In a further embodiment, the amino acid sequence of a p40 variant has at least 75% or more sequence identity with the amino acid sequence of mature wild-type IL-12p40.
  • the amino acid sequence of a p40 variant has at least 80% or more sequence identity with the amino acid sequence of mature wild-type IL-12p40, while in further embodiments, the amino acid sequence of a p40 variant has at least 81 % or more, or at least 82% or more, or at least 83% or more, or at least 84% or more, or at least 85% or more, or at least 86% or more, or at least 87% or more, or at least 88% or more, or at least 89% or more, or at least 90% or more, or at least 91 % or more, or at least 92% or more, or at least 93% or more, or at least 94% or more, or at least 95% or more, or at least 96% or more, or at least 97% or more, or at least 98% or more, or at least 99% or more identity with the amino acid sequence of mature wild-type IL-12p40.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid sequence for optimal alignment with a second amino acid sequence).
  • the determination of percent identity between two sequences can also be accomplished using a mathematical algorithm.
  • a non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, (1990) Proc. Natl. Acad. Sci. USA. 87:2264-68, modified as in Karlin and Altschul, (1993) Proc. Natl. Acad. Sci. USA. 90:5873-77.
  • Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et a/., (1990) J. MoI. Biol.. 215:403- 10.
  • Gapped BLAST can be utilized as described in Altschul et a/., (1997) Nucleic Acids Research. 25(17):3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • the invention provides D3 variants containing an alteration that removes the proteolytic cleavage site between Lys260 and Arg261.
  • the amino acid at position Lys260 is mutated.
  • Lys260 is replaced with a non-basic amino acid.
  • Non-basic amino acids include, for example, Ala, Asn, Asp, Cys, GIu, GIn, GIy, He, Leu, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or VaI.
  • Lys 260 is replaced with either Ala, Asn, Asp, Cys, GIu, GIn, GIy, lie, Leu, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or VaI.
  • Lys260 is replaced with selenocysteine. Examples of D3 variants where Lys260 has been replaced by another amino acid are shown in Figures 12, 13, 14, 15, 16, and 18.
  • Arg261 is mutated.
  • Arg 261 is replaced with any non-basic amino acid.
  • Arg261 is replaced with Ala, Asn, Asp, Cys, GIu, GIn, GIy, lie, Leu, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or VaI.
  • Arg261 is replaced with selenocysteine. Examples of D3 variants where Arg261 has been replaced by another amino acid are shown in Figures 12, 13, 17, and 18.
  • Lys260 and Arg261 are mutated.
  • Lys260 and Arg261 are each replaced with another amino acid.
  • Lys260 is replaced with either GIn, Ala, Asn, or GIy and Arg261 is replaced with either Ala, Asp, or Thr.
  • Examples of D3 variants where both Lys260 and Arg261 have been replaced by other amino acids are shown in Figures 13 and 18. [0069] In addition, Lys260 and Arg261 are deleted in one embodiment, while in a further embodiment, an amino acid is inserted between Lys260 and Arg261 [0070] In a further embodiment, a D3 variant is created in which one or more of Lys258, Ser259, Lys260, Arg261 , Lys263, Lys264, or Arg266 is each replaced by another amino acid.
  • Lys258, Ser259, Lys260, Arg261 , Lys263, Lys264, or Arg266 is each replaced by a non-basic amino acid.
  • Lys258 is replaced with GIn.
  • Ser259 is replaced with Asp.
  • Lys260 is replaced with Ala, GIy, Asn or GIn.
  • Arg261 is replaced with Ala, Thr or Asp.
  • Lys263 is replaced by Ser.
  • Lys264 is replaced by GIy.
  • Arg266 is replaced with GIn, Asp, Asn, or Thr.
  • Lys263 and Lys264 are replaced each replaced by another amino acid.
  • Lys263 and Lys264 are each replaced by Ser and GIy respectively.
  • Lys258, Ser259, Lys260, Arg261 , Lys263, and Lys264 are replaced by GIn, Asp, GIn, Asp, Ser and GIy respectively.
  • a D3 variant is created in which one or more of Ser259, Lys260, and Arg261 are each replaced by another amino acid.
  • one or more of Ser259, Lys260, and Arg261 are each replaced by a non-basic amino acid.
  • Ser259, Lys260, and Arg261 are replaced by Asp, Asn, and Thr respectively.
  • Lys258, Ser259, Lys260, and Arg261 are each replaced by another amino acid.
  • Ser259, Lys260, and Arg261 are each replaced by a non-basic amino acid.
  • Lys258, Ser259, Lys260, and Arg261 are each replaced by GIn, Asp, GIn, and Asp respectively.
  • Ser259, Lys260, Arg261 and Lys264 are replaced by Asp, Asn, Thr, and GIy respectively.
  • Ser259, Lys260, Arg261 and Lys264 are replaced by Asp, Asn, Thr, and GIy respectively, while Lys263, and Asp265 are deleted.
  • Ser259, Lys260, and Arg261 are each replaced by Asp, Asn, and Thr respectively, while Lys263, Lys264, and Asp265 are deleted.
  • deletions are believed to have the effect of reducing the conformational flexibility of the p40(258-266) region, thus reducing the ability of the Lys260-Arg261 motif to adopt a conformation that allows cleavage by the relevant protease. Therefore, in one embodiment, one or more of Lys258, Ser259, Lys260, Arg261 , Lys263, Lys264, Asp265, or Arg266 is deleted.
  • a D3 variant is created in which one or more of Lys263, Lys264, Asp265, or Arg266 is deleted.
  • Lys263 and Asp265 are deleted, while in another embodiment, Lys263, Lys264, and Asp265 are deleted.
  • Lys263, Lys264, and Asp265 are deleted and replaced by one or more non-basic amino acids.
  • Lys263, Lys264, Asp265, and Arg266 are deleted.
  • one or more of Lys263, Lys264, Asp265 or Arg266 is deleted, while one or more of Ser259, Lys260, or Arg261 is replaced by another amino acid.
  • Ser259, Lys260, and Arg261 are replaced by Asp, Asn, and Thr respectively while Lys263, Lys264 and Asp265 are deleted.
  • Ser259, Lys260, and Arg261 are replaced by Asp, Asn, and Thr respectively while Lys263, Lys264, Asp265, and Arg266 are deleted.
  • the amino acid substitutions are selected such that they avoid creating novel T-cell epitopes.
  • Methods to analyze peptide sequences for their potential to create T-cell epitopes are well known in the art (see, e.g., U.S. Patent Application Publication No. 2003/0153043; International Publication No. WO 00/034317; and Sturniolo et al. (1999), Nature Biotech., 17: 555-61 ).
  • the sequence of human IL-12p40(258-266) is replaced by the sequence KDNTER (SEQ ID NO: 17).
  • sequence of human IL-12p40(258-266) is replaced by the sequence KDNTEGR (SEQ ID NO: 18).
  • Ser259, Lys260, and Arg261 were replaced by Asp, Asn, and Thr respectively while Lys263, Lys264 and Asp265 were deleted and replaced by only a GIy residue such that the resulting sequence from residue 258-264 in the variant is KDNTEGR.
  • the resulting lL-12p40 variant is shown in Figure 9.
  • sequence of human IL-12p40(258-266) is replaced by the sequence QDQDEKKDR (SEQ ID NO:19).
  • Lys258, Ser259, Lys260, and Arg261 were replaced by GIn, Asp, GIn, and Asp respectively, such that the resulting sequence from residue 258-266 in the variant is QDQDEKKDR.
  • the resulting IL-12p40 variant is shown in Figure 10.
  • the sequence of human IL-12p40(258-266) is replaced by the sequence QDQDESGDR (SEQ ID NO:20).
  • Lys258, Ser259, Lys260, Arg261, Lys263, and Lys264 were replaced by GIn, Asp, GIn, Asp, Ser, and GIy respectively such that the resulting sequence from residue 258-266 is QDQDESGDR.
  • the resulting IL-12p40 variant is shown in Figure 11.
  • a D3 variant is contained within an IL-12p40 subunit or active portion thereof.
  • active portions it is meant that an IL-12p40 subunit containing a D3 variant has at least 10% activity in one embodiment, at least 20% in another embodiment, at least 30% in another embodiment, at least 50% activity in another embodiment, at least 70% activity in another embodiment, at least 75% activity in another embodiment, at least 80% activity in another embodiment, at least 90% activity in another embodiment, at least 95% activity in another embodiment, at least 99% activity in a further embodiment, at least 100% activity in another embodiment, at least 150% activity in a further embodiment, at least 200% activity in another embodiment, at least 300% activity in a further embodiment, at least 400% activity in another embodiment, at least 500% activity - -
  • the IL-12p40 variants may be introduced into protein compositions in place of wild-type IL-12p40.
  • biologically active protein compositions that include IL-12p40 are p40 homodimers, IL-12 and IL-12 fusion proteins, and IL-23 and IL-23 fusion proteins.
  • the IL-12 p35/variant p40 heterodimer consists of separate polypeptide chains.
  • the IL-12 p35/variant p40 heterodimer consists of a single polypeptide chain.
  • the IL-23 p19/variant p40 heterodimer consists of separate polypeptide chains.
  • the IL-23 p19/variant p40 heterodimer consists of a single polypeptide chain.
  • the IL-12 fusion partner can be an antibody moiety or part of an antibody moiety.
  • Useful antibody moieties include ones that target the IL-12 fusion protein to the tumor environment, for example to the tumor cells themselves, or to the necrotic core of a tumor or to the supporting stroma.
  • the fusion partner is another cytokine.
  • Useful cytokines include, but are not limited to, IL-2, IL-7, and IL-15.
  • the IL-23 fusion partner can be an antibody moiety or part of an antibody moiety.
  • Useful antibody moieties include ones that target the IL-23 fusion protein to the tumor environment, for example to the tumor cells themselves, or to the necrotic core of a tumor or to the supporting stroma.
  • the fusion partner is another cytokine.
  • Useful cytokines include, but are not limited to, IL-2, IL-7, and IL-15.
  • nucleic acids encoding polypeptides containing p40 variants of the invention are contemplated.
  • Nucleic acids encoding p40 variants of the invention can be constructed, for example, using DNA techniques familiar to those skilled in the art. Exemplary procedures can be found in Example 1.
  • Figure 19 depicts the nucleic acid sequence encoding mature human IL-12p40 subunit.
  • Figures 20-22 depict synthetic nucleotide fragments for encoding exemplary mutations found in p40 variants of the invention.
  • the p40 variants including fusion proteins and IL-12 proteins or IL-23 proteins containing a p40 variant, of the invention are useful as immunotherapeutic agents, such as for the treatment of a wide variety of cancers, based on the demonstrated anti-tumor activity of IL-12 proteins.
  • p40 variants of the invention can be used, preferably as a heterodimer with p35, in the treatment of cancers including but not limited to renal cancer, colon cancer, ovarian cancer, melanoma and T-cell lymphoma, and as an adjuvant for cancer vaccines.
  • p40 variants can also be used as part of a p40/p40 homodimer to reduce a TH 1 response (e.g., a TH 1 response associated with an autoimmune disease).
  • Both IL-12 variants, IL-23 variants, and p40 variants of the invention can be incorporated into a pharmaceutical composition suitable for administration.
  • Such compositions typically comprise IL-12 variant or a fusion protein containing an IL-12 variant and a pharmaceutically-acceptable carrier.
  • pharmaceutically-acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules,
  • Medicaments that contain IL-12 variants, IL-23 variants, or p40 variants of the invention can have a concentration of 0.01 or less to 100% (w/w), though the amount varies according to the dosage form of the medicaments.
  • Administration dose depends on the body weight of the patients, the seriousness of the disease, the particular type of IL-12p40 variant being used, and the doctor's opinion. For example, for an IL-12 variant of the invention, it is generally advisable to administer between about 0.01 to about 10 mg/kg body weight a day, about 0.02 to about 2 mg/kg/day in case of injection, or about 0.5 mg/kg/day.
  • the dose can be administered once or several times daily according to the seriousness of the disease and the doctor's opinion.
  • an antibody-IL-12 fusion protein or antibody-IL23 fusion protein containing a IL-12p40 variant of the invention it is generally advisable to administer between about 0.001 to about 1 mg/kg body weight per day, about 0.002 to about 0.5 mg/kg/day in case of injection, or about 0.1 mg/kg/day.
  • the dose can be administered once or twice per 2, 3 or 4 week period, according to the nature and seriousness of the disease and the doctor's opinion. [0090] Aspects of invention are further illustrated by the following examples.
  • Example 1 Cloning of variants of human IL-12p40 subunits
  • Nucleic acids encoding p40 variants of the invention in particular, p40V1 through p40V8 (SEQ ID NOS:6-13), were constructed using standard DNA techniques familiar to those skilled in the art. In essence, a DNA cassette, encoding a fragment that spans the region encompassing the mutated amino acid residues and that is bracketed by convenient restriction sites, was synthesized de novo (Blue Heron Biotechnology, Bothell, WA), and substituted for the corresponding fragment of wild-type sequence contained in an expression plasmid carrying the p40 sequence (see, e.g., pNC-p40 in U.S. Patent No. 6,838,260). The nucleic acid sequence encoding mature (wild-type) human IL- 12p40 subunit is shown in Figure 19. Expression plasmids encoding the p40 variants were thus obtained.
  • nucleic acids encoding p40V1 and p40V2 were generated as follows.
  • a cloning vector carrying p40V1 and p40V2 DNA cassettes (pBHV1V2), synthesized as a contiguous fragment as shown in Figure 20, was digested with Bpu10 I and either Eco Rl / Sea I or Bbs I, generating an EcoR I / Bpu10 I (for V1 ) and Bbs I / Bpu10 I (for V2) cassette with EcoR I / Bpu10 I compatible ends, respectively.
  • the Sea I digestion was included to eliminate the similarly sized V2 fragment.
  • These purified fragments were cloned into a pNC- p40 expression vector in a triple ligation with the appropriate Bpu10 I / Pvu I and Pvu I / Eco Rl fragments obtained from NC-p40.
  • the plasmid (pBHV3V4) carrying the synthesized sequence shown in Figure 22 was digested with EcoR I / Bbs I / Sea I (the Sea I digestion was included to eliminate the similarly sized V4 fragment), or with Bbs I alone, generating an EcoR I / Bbs I (for V3) and a Bbs I (for V4) cassette, respectively, each with ends compatible with the EcoR I / Bbs I digested expression plasmid pNC-p40.
  • V3 and V4 cassettes were then gel purified and ligated, respectively, into the expression plasmid pNC-p40 in a triple ligation using the appropriate Bbs I / Pvu I and Pvu I / Eco Rl fragments obtained from pNC-p40.
  • Example 2 Expression of p40 variants and of antibodv-IL12; fusion proteins containing p40 variants
  • Standard methods were used to generate cell lines expressing p40 variants of the invention (see U.S. Patent No. 6,838,260).
  • the pNC-p40 expression plasmids encoding p40 variants were electroporated into cells, e.g., NS/0 cells. The cells were plated, and transfected cells were selected on a G418-containing medium. Culture supernatants from drug-resistant clones were assayed for production of p40 by ELISA, and the highest producers were subcloned and tested for stable expression.
  • the sequential transfection approach described in U.S. Patent No. 6,838,360 was followed.
  • the fusion protein DI-NHS- IL12p40V1 was obtained by further transfecting the cell line expressing p40V1 with a second plasmid, pdHL10lambdaDI-NHS-p35, which encodes the NHS76 antibody, wherein the C-terminus of the heavy chain constant region is connected to the N-terminus of the IL-12 p35 subunit.
  • the expression plasmid pdHUOIambda is a derivative of pdHL7, wherein the encoded light chain constant domain is a lambda chain.
  • the cells were selected on a methotrexate- containing medium, and stable transfectants expressing the antibody fusion proteins were cloned by standard methods.
  • Example 3 Purification and characterization of p40 variants [0098] To characterize the integrity of p40 variants p40V1 , p40V2, p40V3, and p40V4 (SEQ ID NO:6-9), spent cell culture media from duplicate transiently transfected NS-O cells expressing these variants were collected, and processed for a Western blot with a polyclonal anti-hu-p40 antibody, shown in Figure 24. The control wild-type p40 subunit was included as a control (lane 1).
  • Example 4 Characterization of a proteolytic cleavage site in the wild-type IL- 12P40 subunit
  • the identity of the contaminating approximately 6 kDa protein fragment was determined by standard methods. Briefly, purified DI-NHS-IL12 protein was denatured and reduced in a buffered 6 M guanidine / 1 mM DTT solution at 55 0 C, and subjected to reverse phase HPLC separation over a Vydac C4 column with a 10% to 90% acetonitrile gradient. The fraction corresponding to the unidentified peptide species was collected, dried and re-suspended to run on an SDS-PAGE gel confirming that it corresponded to the 6 kDa fragment, and to determine the sequence of the peptide by N-terminal sequencing. The sequencing analysis revealed a peptide with the sequence REKKDRVFTD, which corresponds to a sequence in the mature (wild-type) human IL-12p40 subunit beginning at Arg261.
  • Bioactivity of IL-12 proteins containing p40 variants was measured by induction of IFN ⁇ from human PBMC.
  • the antibody fusion proteins Ab-IL-12 containing variants p40V1 to p40V8 were compared to Ab-IL-12 with wild-type p40 and a recombinant human IL-12 protein.
  • the IFN ⁇ induction assay was performed essentially as described in Gately et al. (1995), Current Protocols in Immunology, Section 6.16.4, and Kobayashi et al. (1989), J. Exp Med., 170: 827-845.
  • PBMCs were cultured with PHA-P for 3 days and then 25 IU/ml of hu IL-2 (R&D Systems, Minneapolis MN) was added for an additional 24 hours. The cells were washed, 20 IU/ml of IL-2 was added to all cells, followed by addition of IL-12 fusion proteins, with a series of two-fold dilution starting at 20 ng/ml (in terms of relative mass contribution of IL-12 to the molecule). Twenty-four hours later, the concentration of IFN ⁇ was measured by ELISA using antibody pairs purchased from R&D Systems. [00104] The results of two separate experiments using PBMCs from different donors are summarized Table 1.
  • Table 1 Bioactivity of Ab-ILI 2 variants in a IFN ⁇ induction assay.
  • panel A shows a comparison of wild-type Ab-ILI 2 with Ab-IL12 variants containing p40 V1 , p40 V2, p40 V3, and p40 V4, and panel - -
  • Example 7 Treatment of a human patient with IL-12p40 variants
  • the IL-12p40 variants of the invention are used to prevent and treat human diseases and disorders as follows.
  • the preferred method of administration is by i.v. infusion or i.v. injection, or by subcutaneous injection, inhalation, although oral delivery, and other methods are also possible.
  • a patient with advanced metastatic prostate cancer, with a history of treatment by conventional chemotherapy is treated as follows with an antibody- IL12 fusion protein containing an IL-12p40 variant.
  • the dose of the antibody-IL12 fusion protein per treatment cycle is about 150 micrograms per kg of body weight, and may be delivered on a single day or on two or three adjacent days, with administration by drip infusion.
  • Treatment may be combined with a standard-of- care treatment for prostate cancer as determined by a physician as appropriate for the patient.
  • Non-steroidal anti-inflammatory drugs, for example NaproxenTM are also prescribed. Treatment cycles are repeated about once every three weeks.
  • a patient with hormone-refractory breast cancer is treated by drip infusion with an antibody-IL12 fusion protein containing IL-12p40 variant.
  • Non- steroidal anti-inflammatory drugs for example NaproxenTM are also prescribed.
  • a patient with advanced hormone-refractory prostate cancer or advanced hormone-refractory breast cancer is treated with antibody-IL12 fusion protein containing an IL-12p40 variant about once every three weeks, in combination with an IL-2-containing immunocytokine such as KS-IL2. These two agents may be co-administered by drip infusion. Prior to the treatment, the patient is dosed with an immunostimulatory amount of cyclophosphamide.
  • Non-steroidal antiinflammatory drugs for example NaproxenTM are also prescribed.
  • a patient with rheumatoid arthritis is treated with an Fc-p40 fusion protein, in which the p40 subunit is an IL-12p40 variant, about once every two weeks at a dose of about 8 mg/kg, with administration by drip infusion. Progression of joint destruction is found to be significantly inhibited by monotherapy, even when compared to disease-modifying anti-rheumatic drugs.

Abstract

Modified interleukin-12 (IL-12) p40 polypeptides are disclosed. The modified polypeptides have alterations in the IL-12p40 subunit to eliminate the protease site between positions Lys260 and Arg261. The modified IL-12p40 polypeptides according to the invention have improved stability compared to wild-type mature human IL-12p40 polypeptides.

Description

INTERLEUKIN-12P40 VARIANTS WITH IMPROVED STABILITY
Field of the Invention
[0001] This invention relates generally to IL-12p40 proteins, including fusion proteins containing IL-12p40, modified to improve their stability. In particular, the IL-12p40 proteins of the invention remove a proteolytic site in the region of Lys260 and Arg261 in the p40 subunit.
Background of the Invention
[0002] lnterleukin-12 (IL-12) is an inflammatory cytokine that is produced in response to infection by a variety of cells of the immune system, including phagocytic cells, B cells and activated dendritic cells (Colombo and Trinchieri (2002), Cytokine & Growth Factor Reviews, 13: 155-168). IL-12 plays an essential role in mediating the interaction of the innate and adaptive arms of the immune system, acting on T-cells and natural killer (NK) cells, enhancing the proliferation and activity of cytotoxic lymphocytes and the production of other inflammatory cytokines, especially interferon-γ (IFN-γ). [0003] IL-12 is a heterodimeric molecule composed of an α-chain (the p35 subunit, IL-12p35) and a β-chain (the p40 subunit, IL-12p40) covalently linked by a disulfide bridge to form the biologically active 74 kDa heterodimer. Amino acid sequences of IL-12p35 and IL-12p40 of a mature (wild-type) human IL-12 are depicted in Figures 1 (SEQ ID NO:1) and 2 (SEQ ID NO:2), respectively. [0004] lnterleukin-23 (IL-23) is a disulfide-bridged heterodimeric molecule closely related to IL-12, in that it has the same β chain IL-12p40 as IL-12, but a unique α chain (the p19 subunit, IL-23p19) (Oppmann et al., (2000), Immunity, 13: 715-725). Like IL-12, IL-23 is produced by phagocytic cells and activated dendritic cells, and is believed to be involved in the recruitment and activation of a range of inflammatory cells (Langrish et al., (2004) Immunol. Rev., 202: 96-105). The amino acid sequence of IL-23p19 of a mature human IL-23 is depicted in Figure 3 (SEQ ID NO:3).
[0005] For immune cells to secrete biologically active IL-12 or IL-23 heterodimers, concomitant expression of the α and β subunits in the same cell is required. Secretion by immune cells of the IL-12p35 or IL-23p19 alone has not been observed, whereas cells that produce the biologically active IL-12 or IL-23 heterodimer secrete the p40 subunit in free form in 10 to 100-fold excess over the heterodimer (D1 Andrea et al. (1992), J. Exp. Med., 176: 1387-98, Oppmann etal., (2000), Immunity, 13: 715-725 ). In addition, it has been observed in the mouse that, even in the absence of an α subunit, cells may produce a biologically active IL-12p40 homodimer (Hikawa et a/. (2004), Neurosciencθ, 129: 75-83). [0006] The presence of endogenous IL-12 has been shown to be necessary for immunological resistance to a broad array of pathogens, as well as to transplanted and chemically induced tumors (Gateley et al. (1998), Annu. Rev. Immunol., 16: 495-521 ). IL-12 has been demonstrated to have a potent antitumor activity based upon the induction of IFN-γ and the activation of effector cells such as CD8+ T-cells and NK cells (Brunda et al. (1993), J. Exp. Med., 178: 1223-30). As a result of its demonstrated anti-tumor activity, IL-12 has been tested in human clinical trials as an immunotherapeutic agent for the treatment of a wide variety of cancers (Atkins et al. (1997), Clin. Cancer Res., 3: 409-17; Gollob et al. (2000), Clin. Cancer Res., 6: 1678-92; and Hurteau et al. (2001), Gynecol. Oncol., 82: 7-10), including renal cancer, colon cancer, ovarian cancer, melanoma and T-cell lymphoma, and as an adjuvant for cancer vaccines (Lee et al. (2001 ), J. Clin. Oncol. 19: 3836-47). [0007] For IL-12 or IL-23, production of the recombinant protein in its correctly folded and biologically active, heterodimeric form, requires the concurrent expression of both the α subunit and IL-12p40 in the producing cell line. The purified recombinant protein, however, exhibits a degree of heterogeneity resulting from proteolytic cleavage in the C-terminal region of the IL-12p40. The instability of the IL-12 or IL-23 protein can give rise to problems in its production and clinical use as a therapeutic agent. Therefore, there is a need in the art for improved recombinant IL-12 or IL-23 variants that yield a homogeneous protein more resistant to proteolytic cleavage.
Summary of the Invention
[0008] The invention provides variants of human IL-12 p40 subunits (p40 variants) which have improved stability compared to wild-type IL-12 p40 proteins. In these p40 variants, the C-terminal region, which is normally sensitive to proteolytic cleavage, has been engineered to be more resistant to digestion by proteases. Specifically, p40 variants of the invention include engineered amino acid alterations in the D3 domain aimed to avoid the creation of potential T-cell epitopes that could make the variant proteins immunogenic and trigger antibody responses in humans. As a result, p40 variants of the invention have improved properties as therapeutic agents over wild-type IL-12p40 proteins with regard to their production, formulation, and pharmacokinetics.
[0009] Accordingly, in one aspect, the invention provides a variant of a human IL-12 p40 D3 domain (D3 variant), wherein the D3 variant has at least 85% identity with a wild-type human IL-12p40 D3 domain and includes an amino acid alteration at one or more positions corresponding to residues 258-266 of mature human IL-12 p40. Certain embodiments of the invention are based, in part, on an appreciation that an amino acid alteration or alterations according to the invention have the particular benefit of removing the proteolytic site between Lys260 and Arg261.
[0010] According to the invention, the amino acid alterations to one or more positions corresponding to residues 258-266 may be deletions, substitutions, or insertions. Moreover, amino acid substitutions that replace basic amino acids with non-basic amino acids can be used to create variants according to the invention.
[0011] In particular, D3 variants of the invention may include one or more amino acid substitutions at positions selected from the group consisting of Lys258, Ser259, Lys260, Arg261, Lys263, Lys 264, Asp265, and Arg266. Such amino acid alterations can be used singly or in combination to induce the structural and/or functional changes described above. For example, the D3 variant can incorporate one, two, three, four or more of the following substitutions: Lys258Gln, Ser259Asp, Lys260Ala, Lys260Asn, Lys260Gln, Lys260Gly, Arg261Ala, Arg261Asp, Arg261Thr, Lys263Gly, Lys263Ser, and/or Lys264Gly. [0012] In some embodiments, the substitution is a position Lys260. The substitution may replace Lys260 with a non-basic amino acid, for example, Ala, Asn, GIn, or GIy. Further substitutions in addition to Lys260 may occur at Ser259 and Arg261. Particularly, some D3 variants of the invention incorporate substitutions Ser259Asp, Lys260Asn, and Arg261Thr. In a further embodiment, D3 variants of the invention incorporate substitutions Ser259Asp, Lys260Asn, Arg261Thr and Lys264Gly, while optionally deleting Lys263 and Asp265. Alternately, a D3 variant of the invention incorporates substitutions Ser259Asp, Lys260Asn, Arg261Thr, and Lys264Gly while deleting Lys263, Lys264 and Asp265.
[0013] In other embodiments according to the invention, a D3 variant including a substitution replacing Lys260 alternatively includes further substitutions at one or more of Lys258, Ser259, Arg261 , Lys263, and Lys264. For example, in one embodiment, a D3 variant includes the substitutions Lys258Gln, Ser259Asp, Lys260Gln, Arg261 Asp, and optionally Lys263Ser and Lys264Gly.
[0014] In further embodiments, in addition to substitutions at Ser259, Lys260, and Arg261 , one or more of residues corresponding to Lys263, Lys264, Asp265, and Arg266 are deleted, while in another embodiment, one or more of Lys263, Lys264, Asp265, and Arg266 are substituted with a non-basic amino acid. In a further embodiment, the substitution at Lys264 is Lys264Gly and, optionally, Lys263 and Asp265 are deleted. Other D3 variants of the invention incorporate substitutions Ser259Asp, Lys260Asn, Arg261Thr, and Lys264Gly, and optionally, deletion of residues corresponding to Lys263, Asp265 and Arg 266. [0015] It will be understood by those skilled in the art that p40 variants and active portions thereof that incorporate a D3 variant as described herein are within the scope of the invention. Similarly, IL-12 proteins and active portions thereof that contain a p40 variant (IL-12 variants) also are within the scope of the invention. The invention further encompasses fusion proteins including IL-12 variants of the invention and a moiety selected from the group consisting of an antibody moiety, a non-IL-12 cytokine, or an active portion thereof.
[0016] Similarly, IL-23 proteins and active portions thereof that contain a p40 variant (IL-23 variants) also are within the scope of the invention. The invention further encompasses fusion proteins including IL-23 variants of the invention and a moiety selected from the group consisting of an antibody moiety, a non-IL-23 cytokine, or an active portion thereof.
[0017] In another aspect, the invention relates to a nucleic acid that encodes any of the D3 variants, p40 variants, IL-12 variants, and IL-23 variants of the invention. The invention further encompasses a cell, e.g., a prokaryotic cell, including such a nucleic acid.
[0018] The invention also features methods of making such D3 variants, p40 variants, IL-12 variants, IL-23 variants and fusion proteins containing these moieties.
[0019] In yet another aspect, the invention provides methods of using the variants of the invention and the nucleic acids encoding same. For example, the invention encompasses a method of treating a patient that includes administering to the patient a therapeutically effective amount of a p40 variant of the invention or an active portion thereof.
[0020] The foregoing, and other features and advantages of the invention as well as the invention itself, will be more fully understood from the following figures, description, and claims. [0021] In summary the invention relates to: • A variant of an IL-12 p40 D3 domain, wherein the variant has at least 85% sequence identity with a wild-type human IL-12 p40 D3 domain and comprises an amino acid alteration at one or more positions corresponding to residues at positions 258-266 of the parent mature human IL-12 p40.
• A corresponding variant, wherein the alteration eliminates the protease site between Lys260 and Arg261.
• A corresponding variant, wherein the alteration is a substitution or a deletion.
• A corresponding variant, wherein the alteration is a substitution selected from the group consisting of Lys258, Lys260, Lys263, and Lys264. • A corresponding variant, wherein one or more of the following amino acid substitutions are selected:
Lys258Gln,
Lys260Ala, Lys260Asn, Lys260Gln, Lys260Gly,
Lys263Gly, Lys263Ser, and Lys264Gly.
• A corresponding corresponding variant, wherein the substitution is at least at position Lys260. • A corresponding variant, wherein the substitution replaces a basic amino acid with a non-basic amino acid, selected from the group consisting of Ala, Asn, GIn, or GIy.
• A corresponding variant, wherein the variant further comprises one or more substitutions at positions Ser259, Arg261 or Arg 266.
• A corresponding variant, wherein the substitutions are Ser259 and Arg261.
• A corresponding variant, wherein the substitutions are Ser259Asp, Lys260Asn, and Arg261Thr.
• A corresponding variant, further comprising the substitution Lys264Gly. • A corresponding variant, wherein further one or both of the residues
Lys263 and Asp265 are deleted.
• A corresponding variant, wherein one or more residues corresponding to Lys263, Lys264, Asp 265, and Arg266 are deleted.
• A corresponding variant, further comprising one or more of the substitutions selected from the group consisting of:
Lys263, Lys264, Asp 265, and Arg266, wherein the corresponding original amino acid residue is replaced by a non-basic amino acid.
• A corresponding variant further comprising one or more substitutions selected from the group consisting of:
Lys258, Ser259, Arg261 , Lys263 and Lys264.
• A corresponding variant, wherein the substitutions are Lys258Gln, Ser259Asp, Lys260Gln, and Arg261Asp,
• A corresponding variant, further comprising the substitutions Lys263Ser and Lys264Gly.
• A corresponding variant, specifically comprising
(i) the following substitutions: Ser259Asp, Lvs260Asn, and Arg261Thr. and
(ii) the following deletions: Lys263, Lys264 and Asp265, wherein said positions are related to the unmodified wild-type molecule, thus forming the sequence track Lys - Asg - Asn - Thr - GIu - Arg at positions 258 - 263 of the variant. • A corresponding variant, specifically comprising
(i) the following substitutions: Ser259As2, Lys260Asn, Arg261Thr, and
Lys264Gly, and
(ii) the following deletions: Lys263 and Asp265, wherein said positions are related to the unmodified wild-type molecule, thus forming the sequence track: Lys - Asp - Asn - Thr - GIu - GIy - Arg at positions 258 - 264 of the variant.
• A corresponding variant, comprising the following substitutions: Lys258Gln, Ser259Asρ_, Lys260G|n, Arg261Asp, thus forming the sequence track: GIn - Asp - GIn - Asp - GIu - Lys - Lys
- Arg at positions 258 - 265 of the variant.
• A corresponding variant, comprising the following substitutions: Lys258Gln, Ser259Asp_, Lys260Gln, Arg261Asp_, Lys263Ser, Lvs264Gly. thus forming the sequence track: GIn - Asp - GJn - Asp - GIu - Ser- GIy - Arg at positions 258 - 265 of the variant.
• An IL-12 protein, or a fragment thereof, containing the variant as described above and below.
• An IL-23 protein, or a fragment thereof, containing the variant as described above and below. • A fusion protein comprising a variant, or an IL-12 or IL-23 protein as described , and an antibody or an active portion thereof.
• A corresponding fusion protein, wherein said variant or said protein is fused to the C-terminus of said antibody or antibody portion, preferably to the Fc-portion of said antibody, wherein said antibody targets tumor antigens, such as KS (EpCam) or 14.18, or receptors that are overexpressed on tumor tissue, such as HER2 or EGFR or VEGFR.
• A nucleic acid or DNA molecule encoding a variant, or an IL-12 or IL-23 protein, or a fusion protein as described above or below.
• An expression vector comprising said nucleic acid. • A host cell producing a variant or an IL-12 or IL-23 protein or a fusion protein as described.
• A pharmaceutical composition suitable for the treatment of IL-12 or IL-23 triggered diseases comprising in an pharmacologically effective amount a variant, an IL-12 or IL-23 protein , or a fusion protein comprising such a variant or IL-12 or IL-23 protein, optionally together with a pharmaceutically acceptable carrier diluent or excipient, or with other pharmacologically effective drugs in a combination therapy regimen. • A use of said pharmaceutical composition or its effective components for the manufacture of a medicament for the treatment of cancer or an autoimmune disease.
Brief Description of the Drawings [0022] Figure 1 depicts the mature amino acid sequence of the α chain, i.e., the p35 subunit, of a mature (wild-type) human IL-12 (SEQ ID NO:1 ). [0023] Figure 2 depicts the mature amino acid sequence of the β chain, i.e., the p40 subunit, of a mature (wild-type) human IL-12 (SEQ ID NO:2). Domain D3, corresponding to positions 211-306 (SEQ ID NO:26), is italicized, and the peptide fragment corresponding to positions 258-266 is underlined (SEQ ID NO:5), with Lys260 and Arg261 highlighted in bold.
[0024] Figure 3 depicts the mature amino acid sequence of the α chain, i.e., the p19 subunit, of a mature (wild-type) human IL-23 (SEQ ID NO:3). [0025] Figure 4A shows the SDS-PAGE gel for several purified batches of human IL-12 produced as antibody fusion proteins (lanes 1 - 8), while Figure 4B shows the SDS-PAGE gel for several purified batches of human IL-23 produced as antibody fusion proteins (lanes 1 - 3). The IL-12p35 and IL-23p19 subunit is covalently attached to the antibody heavy chain. The 6kD band is indicated by an arrow. Molecular weights (kD) are indicated for the markers (lane M). [0026] Figure 5 depicts the amino acid sequence of the C-terminal peptide fragment of mature (wild-type) human IL-12p40 subunit. The fragment starts at Arg261 (SEQ ID NO:4).
[0027] Figure 6 depicts the amino acid sequence of a peptide fragment corresponding to positions 258-266 of a mature (wild-type) human IL-12p40 subunit (SEQ ID NO:5).
[0028] Figures 7.1 and 7.2 depict an amino acid sequence alignment of IL- 12p40 subunits from various mammals including human, baboon (Papio anubis), rhesus monkey (Macaca mulatta), mangabey (Cercocebus torquatos), dog (Canis familiaris), cat (Felis catus), horse (Equus caballus), pig (Sus scrota), cow (Bos Taurus), goat (Capra hircus), sheep (Ovis aries), deer (Cervus elaphus), water buffalo [Bubalus bubalis), hamster [Mesocricetus auratus), guinea pig (Cavia porcellus), cotton rat (Sigmodon hispidus), rat (Rattus norvegicus), and mouse (Mus musculus). The two amino acids in bold indicate the proteolytic cleavage site.
[0029] Figure 8 depicts the amino acid sequence of a variant, referred to herein as p40V1 , of a mature human IL-12p40 subunit (SEQ ID NO:6). The engineered sequence is underlined. [0030] Figure 9 depicts the amino acid sequence of a variant, referred to herein as p40V2, of a mature human IL-12p40 subunit (SEQ ID N0.7). The engineered sequence is underlined.
[0031] Figure 10 depicts the amino acid sequence of a variant, referred to herein as p40V3, of a mature human IL-12p40 subunit (SEQ ID NO:8). The engineered sequence is underlined.
[0032] Figure 11 depicts the amino acid sequence of a variant, referred to herein as p40V4, of a mature human IL-12p40 subunit (SEQ ID NO:9). The engineered sequence is underlined. [0033] Figure 12 depicts the amino acid sequence of a variant, referred herein as p40V5, of a mature human IL-12p40 subunit (SEQ ID NO: 10). The alteration is underlined.
[0034] Figure 13 depicts the amino acid sequence of a variant, referred herein as p40V6, of a mature human IL-12p40 subunit (SEQ ID NO:11). The alterations are underlined. [0035] Figure 14 depicts the amino acid sequence of a variant, referred herein as p40V7, of a mature human IL-12p40 subunit (SEQ ID NO: 12). The alteration is underlined.
[0036] Figure 15 depicts the amino acid sequence of a variant, referred herein as p40V8, of a mature human IL-12p40 subunit (SEQ ID NO:13). The alteration is underlined.
[0037] Figure 16 depicts the amino acid sequence of a variant, referred herein as p40V9, of a mature human IL-12p40 subunit (SEQ ID NO:14). The alteration is underlined. [0038] Figure 17 depicts the amino acid sequence of a variant, referred herein as p40V10, of a mature human IL-12p40 subunit (SEQ ID NO: 15). The alteration is underlined. [0039] Figure 18 depicts the amino acid sequence of a variant, referred herein as p40V11 , of a mature human IL-12p40 subunit (SEQ ID NO:16). The alterations are underlined.
[0040] Figure 19 depicts the nucleic acid sequence encoding the full length (wild-type) human IL-12p40 subunit (SEQ ID NO:21). [0041] Figure 20 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred to herein as V1V2, encoding portions of p40 variants p40V1 and p40V2. The V1V2 fragment encompasses a V1 fragment including a region (underlined) encoding SEQ ID NO:17. This is followed by a linker sequence (lower case), and subsequently, a V2 fragment including a region (underlined) encoding SEQ ID NO:18. [0042] Figure 21 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred to herein as V3V4, encoding portions of p40 variants p40V3 and p40V4. The V3V4 fragment encompasses a V3 fragment including a region (underlined) encoding SEQ ID NO: 19. This is followed by a linker sequence (lower case), and subsequently, a V4 fragment including a region (underlined) encoding SEQ ID NO:20.
[0043] Figure 22 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred herein as V5V6, encoding portions of p40 variants p40V5 and p40V6. The V5V6 fragment encompasses a V5 fragment including a codon substitution (underlined) encoding Arg261Ala. This is followed by a linker sequence (lower case), and subsequently, a V6 fragment including codon substitutions (underlined) encoding Lys260Ala and Arg261Ala. [0044] Figure 23 depicts the nucleic acid sequence of a synthetic nucleotide fragment, referred herein as V7V8, encoding portions of p40 variants p40V7 and p40V8. The V7V8 fragment encompasses a V7 fragment including a codon substitution (underlined) encoding Lys260Ala. This is followed by a linker sequence (lower case), and subsequently, a V8 fragment including a codon substitution (underlined) encoding Lys260Gly. [0045] Figure 24 is a Western blot with a polyclonal anti-hu-p40 antibody. Supematants of cells transfected with wild-type IL-12p40 and IL-12p40 variants (V1-V4) were harvested and processed on an SDS-PAGE gel. Two independent clones (a, b) of each IL-12p40 variant p40V1 , p40V2, p40V3, and p40V4 were tested. The arrow points to the band of cleaved IL12p40 lacking C-terminal fragment (lane p40 wt).
[0046] Figure 25 shows the SDS-PAGE gel for antibody-IL12 fusion proteins containing p40 variants p40V1 , p40V2, p40V3, p40V4, p40V5, p40V6, p40V7, p40V8, and wild type p40 (lanes 1-9). The upper most main band represents the fusion protein between the antibody heavy chain and the p35 subunit, and the lower main band represents the antibody light chain. The 6 kDa band was not detected in any of the lanes of the variant proteins, except lane 5. Molecular weights (kD) are indicated for the markers (lane M). [0047] Figure 26 shows pharmacokinetic data of antibody-IL12 fusion proteins containing p40 variants p40V1- p40V8 compared to antibody-(wild-type)-IL12 fusion proteins, administered intravenously.
[0048] Figure 27A shows pharmacokinetic data of antibody-IL12 fusion proteins containing p40 variants p40V1-p40V4 compared to antibody-(wild-type)- IL12 fusion proteins (panel A) and Figure 27B shows pharmacokinetic data of antibody-IL12 fusion proteins containing p40 variants p40V5 - p40V8 compared to antibody-(wild-type)-IL12 fusion proteins (panel B), administered subcutaneously.
[0049] Figure 28 is an IL-12p40 variant having mutations outside the D3 region of the protein.
Detailed Description of the Invention
[0050] The invention describes variants of the cytokine interleukin-12 (IL-12) p40 subunit which have improved stability compared with the wild-type protein. In these variants, a region of the p40 subunit which is normally unstructured and sensitive to proteolytic cleavage is mutated to be more resistant to proteolytic cleavage. This region, in domain D3, is near the C-terminus of the p40 subunit, encompassing a polypeptide stretch corresponding to amino acids 258-266 in mature human p40 (p40(258-266)). [0051] The IL-12p40 subunit is also a component subunit of the cytokine interleukin-23 (IL-23). IL-23 has two subunits, the α subunit "p19" and the β subunit "p40." The p40 subunit of IL-23 is the same as IL-12p40. Therefore, variants of IL-12p40 subunit are likewise variants of the IL-23p40 subunit. [0052] In one general class of embodiments, one or more mutations are introduced into the region of p40 corresponding to amino acid residues 258-266 to eliminate the cleavage site, which in human p40 corresponds to the site between Lys260 and Arg261. In further embodiments, specific mutations are introduced in this region that by modeling generate a more tightly folded structure. In another aspect of the invention, the introduced mutations additionally are predicted to avoid making the engineered region of the p40 subunit immunogenic. For example, amino acid substitutions in the engineered region of the p40 subunit are chosen to avoid the creation of peptides that may be recognized as potential T-cell epitopes in humans. [0053] Mutations may be introduced into the region of p40 corresponding to amino acid residues 258-266 by a variety of mechanisms. For example, in one embodiment, a mutation or mutations are introduced by substitution of one amino acid residue for another. In a further embodiment, a mutation or mutations are introduced by deletion of one or more residues of the p40 subunit. In yet another embodiment, a mutation or mutations are introduced by insertion of one or more amino acid residues into the p40 subunit.
[0054] In one embodiment, the p40 variants of the invention are contained in protein compositions such as IL-12 proteins, IL-12 fusion proteins, IL-23 proteins, IL-23 fusion proteins or p40 homodimers. For example, in one embodiment, an IL-12 protein contains a p35 subunit and a p40 variant according to the invention. In another embodiment, the IL-23 protein contains a p19 subunit and p40 variant according to the invention. In a further embodiment, a fusion protein contains an antibody portion fused to IL-12 containing an IL-12p40 variant. In an even further embodiment, a fusion protein contains an antibody portion fused to IL-23 containing an IL-12p40 variant. In a further embodiment, the antibody portion of the fusion protein is an intact antibody, an Fc region, an sc-Fv, an antigen binding portion of an antibody, or an active fragment of an antibody. In yet another embodiment, a fusion protein according to the invention includes an IL-12p40 variant fused to an non-IL-12 or non-IL-23 cytokine or active portion thereof. [0055] In a further aspect of the invention, protein compositions that contain one of the p40 variants of the invention have a longer circulating half-life than the corresponding wild-type protein. Thus, in comparison with IL-12 or IL-23 proteins that contain the wild-type p40 subunit, IL-12 variants or IL-23 variants that include an engineered p40 subunit of the invention have improved properties as therapeutic agents with regard to their production, formulation and pharmacokinetics. [0056] In a further embodiment, mutations to the IL-12p40 amino acid sequence are introduced outside the IL-12p40(258-266) region and optionally outside the IL-12p40 D3 domain of IL-12p40. Such mutations can be introduced elsewhere in the D3 domain of IL-12p40 and can be introduced in the other domains. For example, Figure 28, depicts a sequence of a p40 subunit of IL-12 that has alterations in the amino acid sequence outside of residues 258-266. Leong et al., (2003), Proc. Natl. Acad. Sci. USA. 100:1163-1168, the contents of which are incorporated by reference herein, teach various residues which may be mutated in the IL-12 p40 amino acid sequence beyond the 258-266 region. Further, because the three dimensional crystal structure of IL-12p40 is known (Yoon et al., (2000), EMBO J.. 19:3530-3541 , the contents of which are incorporated by reference herein), and residues essential to the interaction of the p40 subunit with the p35 subunit of IL-12 are know, selection of other mutations that will not destroy functionality of the p40 subunit are determinable by one of skill in the art.
Determination of the cleavage product
[0057] The invention rests in part on the observation that the IL-12p40 subunit is susceptible to a specific proteolytic cleavage event, and on novel experimental results defining the cleavage site. It was found that a purified recombinant IL-12 protein, produced in NS/0 cells as described in Example 2, consistently exhibited a degree of heterogeneity when the recombinant protein was separated by electrophoresis on an SDS-PAGE gel under reducing conditions, clearly visible as an additional protein band of approximately 6 kD molecular weight. A similar observation was made for recombinant IL-23 protein. This is illustrated in Figure 4 which shows the SDS-PAGE gel for several purified batches of human IL-12 (Panel A) and human IL-23 protein compositions (Panel B) produced as antibody fusion proteins. [0058] The contaminant was purified and its amino acid sequence was determined, as described in Example 3, and was found to correspond to the sequence of the C-terminal 46 amino acid fragment of the IL-12p40 subunit itself, generated by proteolytic cleavage between Lys260 and Arg261 (Figure 5-SEQ ID NO:4). Cleavage between Lys260 and Arg261 appears to be highly favored despite a prevalence of basic amino acids in the region surrounding the cleavage site (...KSKREKKDR... (Figure 6-SEQ ID NO:5) where Lys260 and Arg261 are indicated in bold). However, in spite of being cleaved from the p40 subunit, the C-terminal peptide fragment remains non-covalently associated with the rest of the p40 subunit, making it difficult to remove the resulting heterogeneity by further purification of the recombinant protein.
L-12p40 protein
[0059] The mature human p40 subunit is a 306 amino acid protein resembling a soluble class I cytokine D receptor, composed of domains D1 , D2 and D3. The cleavage site (between Lys260 and Arg261 ) is within D3, a fibronectin type III domain of 96 amino acids encompassing the region from 1211 to S306. The sequence for mature human IL-12 p40 subunit is shown in Figure 2. The amino acid sequence for the D3 is shown in Figure 2 in italics. In the published X-ray crystallographic structure of the human IL-12 heterodimer (Yoon et al. (2000), EMBO J., 19: 3530-41) a portion of a loop within the region of human p40(258- 266) of the D3 domain is not resolved. Without wishing to be bound by theory, unresolved regions in crystal structures are often an indication of flexible, unstructured loops, and may constitute target sites for proteolysis. [0060] A primary structure alignment of the mature p40 subunit from a variety of mammalian species is shown in Figures 7.1 and 7.2, including human; the primates baboon, rhesus monkey and mangabey; dog; cat; horse; pig; the ruminants cow, goat, sheep, deer, water buffalo; and the rodents hamster, guinea pig, cotton rat, rat and mouse. In the alignment the region around p40(258-266) exhibits sequence variability, particularly with respect to its length. Particularly, in some species, notably ruminants, the sequence is shorter, and in others, such as in rodents, the sequence in some instances may contain an additional insert. In many species, the dipeptide motif corresponding to human p40 K260R261 is conserved, either identically or exhibiting two basic amino acids. These species include agriculturally and commercially important species including, but not limited to, the horse, cow, goat, pig, and sheep. Consequently, introducing one or more mutations into the region of non-human IL-12p40 corresponding to residues 258-266 of human IL-12p40 that are analogous to mutations taught herein with respect to human IL-12p40 may prove useful in reducing or eliminating proteolytic cleavage of non-human IL-12p40 at the K260R261 cleavage site.
[0061] In principle, according to the invention, it is possible to use a IL-12p40 variant from a species that lacks a positively charged dipeptide motif corresponding to human IL-12p40 K260R261 in a human or other heterologous organism. However, in practicing the invention, it is important to note that non- human forms of IL-12p40 will generally lead to anti-p40 antibodies when administered to humans. More broadly, it is not optimal to administer p40 from one species to another. In addition, the potential of various non-human p40 subunits to be proteolytically cleaved is generally unknown. In addition, p40 subunits from one species may not function in another species, either at the step of assembly with subunits such as p35 or p19, or at the step of interaction with receptor subunits.
Variant IL-12p40 proteins
[0062] The invention provides for variant IL-12p40 proteins with mutations in the D3 domain that improve stability. As used herein, the term "D3 variant" refers to a D3 domain of a human p40 subunit of, for example, IL-12 or IL-23, having one or more amino acid alterations as compared to wild-type D3. The term "p40 variant" is used herein to refer to a human p40 subunit of, for example, IL-12 or IL-23, with mutations in the D3 domain, i.e., a p40 subunit containing a D3 variant. The term "IL-12 variant" is used herein to refer to a human IL-12 protein containing a p40 variant. The term "IL-23 variant" is used herein to refer to a human IL-23 protein containing a p40 variant.
[0063] According to one embodiment of the invention, the D3 domain of a p40 variant has at least 70% or more sequence identity with the D3 domain of wild- type IL-12p40. In a further embodiment, the D3 domain of a p40 variant has at least 75% or more sequence identity with the D3 domain of wild-type IL-12p40. In yet another embodiment, the D3 domain of a p40 variant has at least 80% or more sequence identity with the D3 domain of wild-type IL-12p40, while in further embodiments, the D3 domain of a p40 variant has at least 81 % or more, or at least 82% or more, or at least 83% or more, or at least 84% or more, or at least 85% or more, or at least 86% or more, or at least 87% or more, or at least 88% or more, or at least 89% or more, or at least 90% or more, or at least 91 % or more, or at least 92% or more, or at least 93% or more, or at least 94% or more, or at least 95% or more, or at least 96% or more, or at least 97% or more, or at least 98% or more, or at least 99% or more identity with the D3 domain of wild-type IL- 12p40.
[0064] According to another embodiment of the invention, the amino acid sequence of a p40 variant has at least 70% or more sequence identity with the amino acid sequence of mature wild-type IL-12 p40. In a further embodiment, the amino acid sequence of a p40 variant has at least 75% or more sequence identity with the amino acid sequence of mature wild-type IL-12p40. In yet another embodiment, the amino acid sequence of a p40 variant has at least 80% or more sequence identity with the amino acid sequence of mature wild-type IL-12p40, while in further embodiments, the amino acid sequence of a p40 variant has at least 81 % or more, or at least 82% or more, or at least 83% or more, or at least 84% or more, or at least 85% or more, or at least 86% or more, or at least 87% or more, or at least 88% or more, or at least 89% or more, or at least 90% or more, or at least 91 % or more, or at least 92% or more, or at least 93% or more, or at least 94% or more, or at least 95% or more, or at least 96% or more, or at least 97% or more, or at least 98% or more, or at least 99% or more identity with the amino acid sequence of mature wild-type IL-12p40.
[0065] To determine the percent identity between two amino acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid sequence for optimal alignment with a second amino acid sequence). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = (# of identical positions/total # of positions)times 100). If the sequences being compared are of unequal length, the shorter of the sequences is used to determine the total number of positions. The determination of percent identity between two sequences can also be accomplished using a mathematical algorithm. [0066] A non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, (1990) Proc. Natl. Acad. Sci. USA. 87:2264-68, modified as in Karlin and Altschul, (1993) Proc. Natl. Acad. Sci. USA. 90:5873-77. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et a/., (1990) J. MoI. Biol.. 215:403- 10. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et a/., (1997) Nucleic Acids Research. 25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used.
[0067] In one embodiment, the invention provides D3 variants containing an alteration that removes the proteolytic cleavage site between Lys260 and Arg261. In one embodiment, the amino acid at position Lys260 is mutated. In a more specific embodiment, Lys260 is replaced with a non-basic amino acid. Non-basic amino acids include, for example, Ala, Asn, Asp, Cys, GIu, GIn, GIy, He, Leu, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or VaI. For example, Lys 260 is replaced with either Ala, Asn, Asp, Cys, GIu, GIn, GIy, lie, Leu, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or VaI. In an alternate embodiment, Lys260 is replaced with selenocysteine. Examples of D3 variants where Lys260 has been replaced by another amino acid are shown in Figures 12, 13, 14, 15, 16, and 18.
[0068] In another embodiment, Arg261 is mutated. For example, in one embodiment, Arg 261 is replaced with any non-basic amino acid. For example, in one embodiment, Arg261 is replaced with Ala, Asn, Asp, Cys, GIu, GIn, GIy, lie, Leu, Met, Phe, Pro, Ser, Thr, Trp, Tyr, or VaI. In an alternate embodiment, Arg261 is replaced with selenocysteine. Examples of D3 variants where Arg261 has been replaced by another amino acid are shown in Figures 12, 13, 17, and 18. In a further embodiment, Lys260 and Arg261 are mutated. For example, in one embodiment, Lys260 and Arg261 are each replaced with another amino acid. For exampled, Lys260 is replaced with either GIn, Ala, Asn, or GIy and Arg261 is replaced with either Ala, Asp, or Thr. Examples of D3 variants where both Lys260 and Arg261 have been replaced by other amino acids are shown in Figures 13 and 18. [0069] In addition, Lys260 and Arg261 are deleted in one embodiment, while in a further embodiment, an amino acid is inserted between Lys260 and Arg261 [0070] In a further embodiment, a D3 variant is created in which one or more of Lys258, Ser259, Lys260, Arg261 , Lys263, Lys264, or Arg266 is each replaced by another amino acid. In one embodiment, one or more of Lys258, Ser259, Lys260, Arg261 , Lys263, Lys264, or Arg266 is each replaced by a non-basic amino acid. For example, in one embodiment, Lys258 is replaced with GIn. In another embodiment, Ser259 is replaced with Asp. In another embodiment, Lys260 is replaced with Ala, GIy, Asn or GIn. In a further embodiment, Arg261 is replaced with Ala, Thr or Asp. In yet another embodiment, Lys263 is replaced by Ser. In a further embodiment, Lys264 is replaced by GIy. In one embodiment, Arg266 is replaced with GIn, Asp, Asn, or Thr. In a further embodiment, Lys263 and Lys264 are replaced each replaced by another amino acid. For example, in one embodiment Lys263 and Lys264 are each replaced by Ser and GIy respectively. In yet another embodiment, Lys258, Ser259, Lys260, Arg261 , Lys263, and Lys264 are replaced by GIn, Asp, GIn, Asp, Ser and GIy respectively.
[0071] In a further embodiment, a D3 variant is created in which one or more of Ser259, Lys260, and Arg261 are each replaced by another amino acid. In a further embodiment, one or more of Ser259, Lys260, and Arg261 are each replaced by a non-basic amino acid. For example, in one embodiment, Ser259, Lys260, and Arg261 are replaced by Asp, Asn, and Thr respectively. [0072] In another embodiment, Lys258, Ser259, Lys260, and Arg261 are each replaced by another amino acid. For example, in one embodiment, Lys258, _
Ser259, Lys260, and Arg261 are each replaced by a non-basic amino acid. In one embodiment, Lys258, Ser259, Lys260, and Arg261 are each replaced by GIn, Asp, GIn, and Asp respectively. In a further embodiment, Ser259, Lys260, Arg261 and Lys264 are replaced by Asp, Asn, Thr, and GIy respectively. In yet a further embodiment, Ser259, Lys260, Arg261 and Lys264 are replaced by Asp, Asn, Thr, and GIy respectively, while Lys263, and Asp265 are deleted. In yet another embodiment, Ser259, Lys260, and Arg261 are each replaced by Asp, Asn, and Thr respectively, while Lys263, Lys264, and Asp265 are deleted. [0073] Without wishing to be bound by theory, deletions are believed to have the effect of reducing the conformational flexibility of the p40(258-266) region, thus reducing the ability of the Lys260-Arg261 motif to adopt a conformation that allows cleavage by the relevant protease. Therefore, in one embodiment, one or more of Lys258, Ser259, Lys260, Arg261 , Lys263, Lys264, Asp265, or Arg266 is deleted. [0074] In a further embodiment, a D3 variant is created in which one or more of Lys263, Lys264, Asp265, or Arg266 is deleted. For example, in one embodiment, Lys263 and Asp265 are deleted, while in another embodiment, Lys263, Lys264, and Asp265 are deleted. In another embodiment, Lys263, Lys264, and Asp265 are deleted and replaced by one or more non-basic amino acids. In a further embodiment, Lys263, Lys264, Asp265, and Arg266 are deleted. In a further embodiment, one or more of Lys263, Lys264, Asp265 or Arg266 is deleted, while one or more of Ser259, Lys260, or Arg261 is replaced by another amino acid. For example, in one embodiment, Ser259, Lys260, and Arg261 are replaced by Asp, Asn, and Thr respectively while Lys263, Lys264 and Asp265 are deleted. In a further embodiment, Ser259, Lys260, and Arg261 are replaced by Asp, Asn, and Thr respectively while Lys263, Lys264, Asp265, and Arg266 are deleted.
[0075] In other embodiments, the amino acid substitutions are selected such that they avoid creating novel T-cell epitopes. Methods to analyze peptide sequences for their potential to create T-cell epitopes are well known in the art (see, e.g., U.S. Patent Application Publication No. 2003/0153043; International Publication No. WO 00/034317; and Sturniolo et al. (1999), Nature Biotech., 17: 555-61 ). In one embodiment, the sequence of human IL-12p40(258-266) is replaced by the sequence KDNTER (SEQ ID NO: 17). In other words, Ser259, Lys260, and Arg261 were replaced by Asp, Asn, and Thr respectively while Lys263, Lys264, and Asp265 were deleted such that the resulting sequence from residue 258-263 in the variant is KDNTER. The resulting IL-12p40 variant is shown in Figure 8.
[0076] In another embodiment, the sequence of human IL-12p40(258-266) is replaced by the sequence KDNTEGR (SEQ ID NO: 18). In other words, Ser259, Lys260, and Arg261 were replaced by Asp, Asn, and Thr respectively while Lys263, Lys264 and Asp265 were deleted and replaced by only a GIy residue such that the resulting sequence from residue 258-264 in the variant is KDNTEGR. The resulting lL-12p40 variant is shown in Figure 9. [0077] In yet another embodiment, the sequence of human IL-12p40(258-266) is replaced by the sequence QDQDEKKDR (SEQ ID NO:19). In other words, Lys258, Ser259, Lys260, and Arg261 were replaced by GIn, Asp, GIn, and Asp respectively, such that the resulting sequence from residue 258-266 in the variant is QDQDEKKDR. The resulting IL-12p40 variant is shown in Figure 10. [0078] In a further embodiment, the sequence of human IL-12p40(258-266) is replaced by the sequence QDQDESGDR (SEQ ID NO:20). In other words, Lys258, Ser259, Lys260, Arg261, Lys263, and Lys264 were replaced by GIn, Asp, GIn, Asp, Ser, and GIy respectively such that the resulting sequence from residue 258-266 is QDQDESGDR. The resulting IL-12p40 variant is shown in Figure 11.
[0079] In a further embodiment, a D3 variant is contained within an IL-12p40 subunit or active portion thereof. By active portions, it is meant that an IL-12p40 subunit containing a D3 variant has at least 10% activity in one embodiment, at least 20% in another embodiment, at least 30% in another embodiment, at least 50% activity in another embodiment, at least 70% activity in another embodiment, at least 75% activity in another embodiment, at least 80% activity in another embodiment, at least 90% activity in another embodiment, at least 95% activity in another embodiment, at least 99% activity in a further embodiment, at least 100% activity in another embodiment, at least 150% activity in a further embodiment, at least 200% activity in another embodiment, at least 300% activity in a further embodiment, at least 400% activity in another embodiment, at least 500% activity - -
in another embodiment, or at least 1000% activity in another embodiment, in comparison to the biological activity of a wild type IL-12p40 moiety.
Proteins containing IL-12p40 variants
[0080] The IL-12p40 variants may be introduced into protein compositions in place of wild-type IL-12p40. Examples of biologically active protein compositions that include IL-12p40 are p40 homodimers, IL-12 and IL-12 fusion proteins, and IL-23 and IL-23 fusion proteins. In one aspect of the invention, the IL-12 p35/variant p40 heterodimer consists of separate polypeptide chains. Alternatively, the IL-12 p35/variant p40 heterodimer consists of a single polypeptide chain. In another aspect of the invention, the IL-23 p19/variant p40 heterodimer consists of separate polypeptide chains. Alternatively, the IL-23 p19/variant p40 heterodimer consists of a single polypeptide chain.
[0081] In another aspect of the invention, as part of an IL-12 fusion protein, the IL-12 fusion partner can be an antibody moiety or part of an antibody moiety. Useful antibody moieties include ones that target the IL-12 fusion protein to the tumor environment, for example to the tumor cells themselves, or to the necrotic core of a tumor or to the supporting stroma. In another embodiment of the invention, the fusion partner is another cytokine. Useful cytokines include, but are not limited to, IL-2, IL-7, and IL-15.
[0082] In another aspect of the invention, as part of an IL-23 fusion protein, the IL-23 fusion partner can be an antibody moiety or part of an antibody moiety. Useful antibody moieties include ones that target the IL-23 fusion protein to the tumor environment, for example to the tumor cells themselves, or to the necrotic core of a tumor or to the supporting stroma. In another embodiment of the invention, the fusion partner is another cytokine. Useful cytokines include, but are not limited to, IL-2, IL-7, and IL-15.
Nucleic acids encoding p40 variants
[0083] In a further aspect of the invention, nucleic acids encoding polypeptides containing p40 variants of the invention are contemplated. Nucleic acids encoding p40 variants of the invention can be constructed, for example, using DNA techniques familiar to those skilled in the art. Exemplary procedures can be found in Example 1.
[0084] Figure 19 depicts the nucleic acid sequence encoding mature human IL-12p40 subunit. Figures 20-22 depict synthetic nucleotide fragments for encoding exemplary mutations found in p40 variants of the invention.
Methods of Treatment Using p40 Variants
[0085] The p40 variants, including fusion proteins and IL-12 proteins or IL-23 proteins containing a p40 variant, of the invention are useful as immunotherapeutic agents, such as for the treatment of a wide variety of cancers, based on the demonstrated anti-tumor activity of IL-12 proteins. For example, p40 variants of the invention can be used, preferably as a heterodimer with p35, in the treatment of cancers including but not limited to renal cancer, colon cancer, ovarian cancer, melanoma and T-cell lymphoma, and as an adjuvant for cancer vaccines. p40 variants can also be used as part of a p40/p40 homodimer to reduce a TH 1 response (e.g., a TH 1 response associated with an autoimmune disease).
Administration
[0086] Both IL-12 variants, IL-23 variants, and p40 variants of the invention can be incorporated into a pharmaceutical composition suitable for administration. Such compositions typically comprise IL-12 variant or a fusion protein containing an IL-12 variant and a pharmaceutically-acceptable carrier. As used herein, the term "pharmaceutically-acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. [0087] A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[0088] Medicaments that contain IL-12 variants, IL-23 variants, or p40 variants of the invention can have a concentration of 0.01 or less to 100% (w/w), though the amount varies according to the dosage form of the medicaments. [0089] Administration dose depends on the body weight of the patients, the seriousness of the disease, the particular type of IL-12p40 variant being used, and the doctor's opinion. For example, for an IL-12 variant of the invention, it is generally advisable to administer between about 0.01 to about 10 mg/kg body weight a day, about 0.02 to about 2 mg/kg/day in case of injection, or about 0.5 mg/kg/day. The dose can be administered once or several times daily according to the seriousness of the disease and the doctor's opinion. For an antibody-IL-12 fusion protein or antibody-IL23 fusion protein containing a IL-12p40 variant of the invention, it is generally advisable to administer between about 0.001 to about 1 mg/kg body weight per day, about 0.002 to about 0.5 mg/kg/day in case of injection, or about 0.1 mg/kg/day. The dose can be administered once or twice per 2, 3 or 4 week period, according to the nature and seriousness of the disease and the doctor's opinion. [0090] Aspects of invention are further illustrated by the following examples.
Examples
Example 1 : Cloning of variants of human IL-12p40 subunits [0091] Nucleic acids encoding p40 variants of the invention, in particular, p40V1 through p40V8 (SEQ ID NOS:6-13), were constructed using standard DNA techniques familiar to those skilled in the art. In essence, a DNA cassette, encoding a fragment that spans the region encompassing the mutated amino acid residues and that is bracketed by convenient restriction sites, was synthesized de novo (Blue Heron Biotechnology, Bothell, WA), and substituted for the corresponding fragment of wild-type sequence contained in an expression plasmid carrying the p40 sequence (see, e.g., pNC-p40 in U.S. Patent No. 6,838,260). The nucleic acid sequence encoding mature (wild-type) human IL- 12p40 subunit is shown in Figure 19. Expression plasmids encoding the p40 variants were thus obtained.
[0092] In particular, the nucleic acids encoding p40V1 and p40V2 were generated as follows. A cloning vector carrying p40V1 and p40V2 DNA cassettes (pBHV1V2), synthesized as a contiguous fragment as shown in Figure 20, was digested with Bpu10 I and either Eco Rl / Sea I or Bbs I, generating an EcoR I / Bpu10 I (for V1 ) and Bbs I / Bpu10 I (for V2) cassette with EcoR I / Bpu10 I compatible ends, respectively. The Sea I digestion was included to eliminate the similarly sized V2 fragment. These purified fragments were cloned into a pNC- p40 expression vector in a triple ligation with the appropriate Bpu10 I / Pvu I and Pvu I / Eco Rl fragments obtained from NC-p40.
[0093] An identical approach was used to generate nucleic acids encoding p40V5 and p40V6, starting with the synthesized sequence shown in Figure 21 , and to generate nucleic acids encoding p40V7 and p40V8, starting with the synthesized sequence shown in Figure 23. [0094] Similarly, to generate nucleic acids encoding p40V3 and p40V4, the plasmid (pBHV3V4) carrying the synthesized sequence shown in Figure 22, was digested with EcoR I / Bbs I / Sea I (the Sea I digestion was included to eliminate the similarly sized V4 fragment), or with Bbs I alone, generating an EcoR I / Bbs I (for V3) and a Bbs I (for V4) cassette, respectively, each with ends compatible with the EcoR I / Bbs I digested expression plasmid pNC-p40. Note that for the Bbs I restriction enzyme the recognition and cleavage sequences are separate, and thus sequences containing multiple Bbs I recognition sites may generate different, sequence-specific overhangs. The V3 and V4 cassettes were then gel purified and ligated, respectively, into the expression plasmid pNC-p40 in a triple ligation using the appropriate Bbs I / Pvu I and Pvu I / Eco Rl fragments obtained from pNC-p40.
[0095] The same general approach may be used to generate further nucleic acid molecules encoding other p40 variants contemplated by the invention.
Example 2: Expression of p40 variants and of antibodv-IL12; fusion proteins containing p40 variants
[0096] Standard methods were used to generate cell lines expressing p40 variants of the invention (see U.S. Patent No. 6,838,260). The pNC-p40 expression plasmids encoding p40 variants were electroporated into cells, e.g., NS/0 cells. The cells were plated, and transfected cells were selected on a G418-containing medium. Culture supernatants from drug-resistant clones were assayed for production of p40 by ELISA, and the highest producers were subcloned and tested for stable expression.
[0097] To generate antibody-IL-12 fusion protein expressing cell lines with p40 variants of the invention, the sequential transfection approach described in U.S. Patent No. 6,838,360 was followed. For example, the fusion protein DI-NHS- IL12p40V1 was obtained by further transfecting the cell line expressing p40V1 with a second plasmid, pdHL10lambdaDI-NHS-p35, which encodes the NHS76 antibody, wherein the C-terminus of the heavy chain constant region is connected to the N-terminus of the IL-12 p35 subunit. The expression plasmid pdHUOIambda is a derivative of pdHL7, wherein the encoded light chain constant domain is a lambda chain. The cells were selected on a methotrexate- containing medium, and stable transfectants expressing the antibody fusion proteins were cloned by standard methods.
Example 3: Purification and characterization of p40 variants [0098] To characterize the integrity of p40 variants p40V1 , p40V2, p40V3, and p40V4 (SEQ ID NO:6-9), spent cell culture media from duplicate transiently transfected NS-O cells expressing these variants were collected, and processed for a Western blot with a polyclonal anti-hu-p40 antibody, shown in Figure 24. The control wild-type p40 subunit was included as a control (lane 1). It was found that the cleaved species lacking the C-terminal 6 kDa fragment, which is normally well-resolved from the intact p40 species by these electrophoretic conditions (see arrow pointing to band in lane 1), was not present in any of the variants tested (lanes 2-9), and only intact p40 could be detected. Thus, the tested p40 variants were resistant to a proteolytic activity present during protein expression. [0099] Antibody fusion proteins containing IL-12p40 variants were purified from cell culture supernatant using standard techniques based on Protein A capture (see U.S. Patent No. 6,838,260). [00100] SDS-PAGE gel of the purified antibody fusion proteins from NS-O stable clones of the p40 variants given in SEQ ID NOS:6-13 (lanes 1 - 8) and of the non-mutated control (lane 9) is shown in Figure 25. The middle of the three major bands represents the non-cleaved p40 subunit, with the faint band slightly above indicating more glycosylated species. The upper most main band represents the fusion protein between the antibody heavy chain and the p35 subunit, and the lower main band represents the antibody light chain. It was found that, with the exception of the sample of the antibody fusion protein containing IL-12p40V5, the 6 kDa band was not present. For IL-12p40V5, a residual 6 kDa band was observed (lane 5).
Example 4: Characterization of a proteolytic cleavage site in the wild-type IL- 12P40 subunit
[00101] The identity of the contaminating approximately 6 kDa protein fragment was determined by standard methods. Briefly, purified DI-NHS-IL12 protein was denatured and reduced in a buffered 6 M guanidine / 1 mM DTT solution at 550C, and subjected to reverse phase HPLC separation over a Vydac C4 column with a 10% to 90% acetonitrile gradient. The fraction corresponding to the unidentified peptide species was collected, dried and re-suspended to run on an SDS-PAGE gel confirming that it corresponded to the 6 kDa fragment, and to determine the sequence of the peptide by N-terminal sequencing. The sequencing analysis revealed a peptide with the sequence REKKDRVFTD, which corresponds to a sequence in the mature (wild-type) human IL-12p40 subunit beginning at Arg261.
Example 5: Bioactivitv of IL-12 proteins containing p40 variants - -
[00102] Bioactivity of IL-12 proteins containing p40 variants was measured by induction of IFNγ from human PBMC. The antibody fusion proteins Ab-IL-12 containing variants p40V1 to p40V8 were compared to Ab-IL-12 with wild-type p40 and a recombinant human IL-12 protein. [00103] The IFNγ induction assay was performed essentially as described in Gately et al. (1995), Current Protocols in Immunology, Section 6.16.4, and Kobayashi et al. (1989), J. Exp Med., 170: 827-845. PBMCs were cultured with PHA-P for 3 days and then 25 IU/ml of hu IL-2 (R&D Systems, Minneapolis MN) was added for an additional 24 hours. The cells were washed, 20 IU/ml of IL-2 was added to all cells, followed by addition of IL-12 fusion proteins, with a series of two-fold dilution starting at 20 ng/ml (in terms of relative mass contribution of IL-12 to the molecule). Twenty-four hours later, the concentration of IFNγ was measured by ELISA using antibody pairs purchased from R&D Systems. [00104] The results of two separate experiments using PBMCs from different donors are summarized Table 1.
Table 1 : Bioactivity of Ab-ILI 2 variants in a IFNγ induction assay.
IFNγ Induction
Protein ED50 (ng/ml) AVG (n=3) SD
Exp I
R&D EL- 12 0.04 0.02
Ab-IL12 0.43 0.21
Ab-IL12 Vl 1.11 0.59
Ab-IL12 V2 1.09 0.32
Ab-H 12 V3 1.09 0.21
Ab-IL 12 V4 1.44 0.48
Exp II
R&D IL-12 0.05 0.03
Ab-IL12 0.53 0.42
Ab-IL12 Vl 1.53 0.58
Ab-IL12 V4 1.79* 0.80
Ab-IL12 V5 0.58 0.39
Ab-H12 V6 1.63 1.46
Figure imgf000029_0001
n=2
[0100] Compared to recombinant hu IL-12, the activity of Ab-ILI 2 with wild type p40 was about 10 fold reduced. It was found that the antibody-IL12 variant proteins tested did not significantly further affect the activity of the protein. Ab- IL12p40V1 to Ab-ILI 2p40V8 had somewhat reduced activity (approximately 1.5 - 3 fold less) compared to the corresponding wild-type antibody-IL-12 fusion protein.
Example 6: Pharmacokinetics of IL-12 proteins containing p40 variants [0101] The pharmacokinetics of the antibody fusion proteins containing the p40 variants was determined. The experiments were performed using standard techniques familiar to those skilled in the art. Briefly, BALB/c mice (n=3 per treatment group) were injected with 25 μg of Ab-ILI 2 or Ab-ILI 2 variants containing the variants p40V1 , p40V2, p40V3, p40V4, p40V5, p40V6, p40V7 and p40V8 in a volume of 0.2 ml, either intravenously in the tail vein or subcutaneously. At various time points up to 24 hours and up to 96 hours, respectively, small volumes of blood was taken by retro-orbital bleeding and collected in heparin-coated tubes to prevent clotting. After centrifugation to remove cells, the plasma was assayed by capture with anti-human IgG H&L antisera and detection with an anti-human IL-12 antibody. Results were normalized to the initial concentration in the plasma of each mouse taken within 30 seconds after injection (t = 0). Figure 26 is a compilation of representative experiments assessing the pharmacokinetics of intravenously administered protein. It was found that compared to the wild-type control protein, antibody-IL- 12 fusion proteins containing variants p40V1 and p40V2, p40V3, p40V4, as well as p40V6, had significantly improved pharmacokinetic values. Particularly, it was found that the half-life of the distribution phase (alpha phase) approximately doubled, and correspondingly the AUC of the variant proteins approximately doubled as well. However, the elimination phase (beta phase) of all Ab-IL-12 fusion proteins remained substantially similar. These results were consistent with the pharmacokinetics of these proteins when administered to the mouse subcutaneously Figure 27, panel A, shows a comparison of wild-type Ab-ILI 2 with Ab-IL12 variants containing p40 V1 , p40 V2, p40 V3, and p40 V4, and panel - -
B shows a comparison of wild-type Ab-ILI 2 with Ab-ILI 2 variants containing p40 V5, p40 V6, p40 V7, and p40 V8.
Example 7. Treatment of a human patient with IL-12p40 variants [0102] The IL-12p40 variants of the invention are used to prevent and treat human diseases and disorders as follows. In general, the preferred method of administration is by i.v. infusion or i.v. injection, or by subcutaneous injection, inhalation, although oral delivery, and other methods are also possible. [0103] A patient with advanced metastatic prostate cancer, with a history of treatment by conventional chemotherapy, is treated as follows with an antibody- IL12 fusion protein containing an IL-12p40 variant. The dose of the antibody-IL12 fusion protein per treatment cycle is about 150 micrograms per kg of body weight, and may be delivered on a single day or on two or three adjacent days, with administration by drip infusion. Treatment may be combined with a standard-of- care treatment for prostate cancer as determined by a physician as appropriate for the patient. Non-steroidal anti-inflammatory drugs, for example Naproxen™, are also prescribed. Treatment cycles are repeated about once every three weeks.
[0104] A patient with hormone-refractory breast cancer is treated by drip infusion with an antibody-IL12 fusion protein containing IL-12p40 variant. Non- steroidal anti-inflammatory drugs, for example Naproxen™ are also prescribed. [0105] In an alternative treatment strategy, a patient with advanced hormone-refractory prostate cancer or advanced hormone-refractory breast cancer is treated with antibody-IL12 fusion protein containing an IL-12p40 variant about once every three weeks, in combination with an IL-2-containing immunocytokine such as KS-IL2. These two agents may be co-administered by drip infusion. Prior to the treatment, the patient is dosed with an immunostimulatory amount of cyclophosphamide. Non-steroidal antiinflammatory drugs, for example Naproxen™ are also prescribed. [0106] A patient with rheumatoid arthritis is treated with an Fc-p40 fusion protein, in which the p40 subunit is an IL-12p40 variant, about once every two weeks at a dose of about 8 mg/kg, with administration by drip infusion. Progression of joint destruction is found to be significantly inhibited by monotherapy, even when compared to disease-modifying anti-rheumatic drugs.

Claims

- -Patent Claims:
1. A variant of an IL-12 p40 D3 domain, wherein the variant has at least 85% sequence identity with a wild-type human IL-12 p40 D3 domain and comprises an amino acid alteration at one or more positions corresponding to residues at positions 258-266 of the parent mature human IL-12 p40.
2. A variant of claim 1 , wherein the alteration eliminates the protease site between Lys260 and Arg261.
3. A variant of claim 1 or 2, wherein the alteration is a substitution or a deletion.
4. A variant of claim 3, wherein the alteration is a substitution selected from the group consisting of Lys258, Lys260, Lys263, and Lys264.
5. A variant of claim 4, wherein one or more of the following amino acid substitutions are selected:
Lys258Gln,
Lys260Ala, Lys260Asn, Lys260Gln, Lys260Gly,
Lys263Gly, Lys263Ser, and Lys264Gly.
6. A variant of claim 4 or 5, wherein the substitution is at least at position Lys260.
7. A variant of any of the claims 4 - 6, wherein the substitution replaces a basic amino acid with a non-basic amino acid, selected from the group consisting of Ala, Asn, GIn, or GIy.
8. A variant of claim 6 or 7, wherein the variant further comprises one or more substitutions at positions Ser259, Arg261 or Arg 266.
9. A variant of claim 8, wherein the substitutions are Ser259 and Arg261.
10. A variant of claim 9, wherein the substitutions are Ser259Asp, Lys260Asn, and Arg261Thr.
11. A variant of claim 10, further comprising the substitution Lys264Gly.
12. A variant of claim 11 , wherein further one or both of the residues Lys263 and Asp265 are deleted.
13. A variant of claim 9 or 10, wherein one or more residues corresponding to Lys263, Lys264, Asp 265, and Arg266 are deleted.
14. A variant of claim 9 or 10, further comprising one or more of the substitutions selected from the group consisting of:
Lys263, Lys264, Asp 265, and Arg266, wherein the corresponding original amino acid residue is replaced by a non-basic amino acid.
15. A variant of claim 6 or 7 further comprising one or more substitutions selected from the group consisting of: Lys258, Ser259, Arg261 , Lys263 and Lys264.
16. A variant of claim 15, wherein the substitutions are Lys258Gln, Ser259Asp, Lys260Gln, and Arg261Asp,
17. A variant of claim 16, further comprising the substitutions Lys263Ser and Lys264Gly.
18. A variant of claim 3, comprising
(i) the following substitutions: Ser259Asp, Lys260Asn. and Arg261Thr. and (ii) the following deletions: Lys263, Lys264 and Asp265, wherein said positions are related to the unmodified wild-type molecule, thus forming the sequence track Lys - Asp - Asn - Thr - GIu - Arg at positions 258 - 263 of the variant.
19. A variant of claim 3, comprising (i) the following substitutions: Ser259Asp_, Lys260Asn, Arg261Thr, and
Lys264Gly, and
(ii) the following deletions: Lys263 and Asp265, wherein said positions are related to the unmodified wild-type molecule, thus forming the sequence track: Lys - Asp - Asn - Thr - GIu - Gjy - Arg at positions 258 - 264 of the variant.
20. A variant of claim 3, comprising the following substitutions: Lys258Gln, Ser259Asp_, Lys260Gln, Arg261Asp. thus forming the sequence track: GIn - Asp - GIn - Asp - GIu - Lys - Lys - Arg at positions 258 - 265 of the variant.
21. A variant of claim 3, comprising the following substitutions: Lys258Gln, Ser259Asp_, Lys260Gln, Arg261Asp_, Lys263Ser, Lvs264Glv. thus forming the sequence track: GIn - Asp - GIn - Asp - GIu - Ser- GIy - Arg at positions 258 - 265 of the variant.
22. A IL-12 protein, or a fragment thereof, containing the variant of any of the claims 1 - 21.
23. A IL-23 protein, or a fragment thereof, containing the variant of any of the claims 1 - 21.
24. A fusion protein comprising a variant of any one of claims 1 - 21 , or a protein of claim 22 or 23, and an antibody or an active portion thereof.
25. A fusion protein, wherein said variant or said protein is fused to the C- terminus of said antibody or antibody portion. - -
26. A nucleic acid encoding a variant according to any one of claims 1 - 21 , or a protein of claim 22 or 23, or a fusion protein of claim 24 or 25.
27. An expression vector comprising the nucleic acid of claim 26.
28. A host cell producing a variant of any of claims 1 - 21 , a protein of claim 22 or 23, or a fusion protein of claim 24 or 25, comprising the expression vector of claim 27.
29. A pharmaceutical composition suitable for the treatment of IL-12 or IL-23 triggered diseases comprising in an pharmacologically effective amount a protein of any of the claims 22 - 25, optionally together with a pharmaceutically acceptable carrier diluent or excipient.
30. Use of a pharmaceutical composition of claim 29 for the manufacture of a medicament for the treatment of cancer or an autoimmune disease.
PCT/EP2006/012301 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability WO2007076933A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
EP20060846989 EP1966238B1 (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability
JP2008547880A JP5175214B2 (en) 2005-12-30 2006-12-20 Interleukin-12P40 variant with improved stability
CA2635618A CA2635618C (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability
AU2006332138A AU2006332138B2 (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability
AT06846989T ATE555125T1 (en) 2005-12-30 2006-12-20 INTERLEUKIN 12P40 VARIANTS WITH IMPROVED STABILITY
ES06846989T ES2384055T3 (en) 2005-12-30 2006-12-20 Variants of interleukin-12p40 with improved stability
EA200870131A EA015338B1 (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability
DK06846989T DK1966238T3 (en) 2005-12-30 2006-12-20 INTERLEUKIN-12P40 variants with improved stability
PL06846989T PL1966238T3 (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability
CN2006800498186A CN101351475B (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability
SI200631366T SI1966238T1 (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability
BRPI0620648-4A BRPI0620648B1 (en) 2005-12-30 2006-12-20 IL-12 P40 VARIANT, IL-12, IL-23 AND FUSION PROTEINS, PHARMACEUTICAL COMPOSITION AND USE THEREOF
KR1020087018718A KR101343682B1 (en) 2005-12-30 2008-07-29 - interleukin-12p40 variants with improved stability

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US75538205P 2005-12-30 2005-12-30
US60/755,382 2005-12-30

Publications (1)

Publication Number Publication Date
WO2007076933A1 true WO2007076933A1 (en) 2007-07-12

Family

ID=38049414

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2006/012301 WO2007076933A1 (en) 2005-12-30 2006-12-20 Interleukin-12p40 variants with improved stability

Country Status (18)

Country Link
US (3) US7872107B2 (en)
EP (1) EP1966238B1 (en)
JP (1) JP5175214B2 (en)
KR (1) KR101343682B1 (en)
CN (1) CN101351475B (en)
AT (1) ATE555125T1 (en)
AU (1) AU2006332138B2 (en)
BR (1) BRPI0620648B1 (en)
CA (1) CA2635618C (en)
CY (1) CY1112884T1 (en)
DK (1) DK1966238T3 (en)
EA (1) EA015338B1 (en)
ES (1) ES2384055T3 (en)
PL (1) PL1966238T3 (en)
PT (1) PT1966238E (en)
SI (1) SI1966238T1 (en)
WO (1) WO2007076933A1 (en)
ZA (1) ZA200806611B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012506394A (en) * 2008-10-21 2012-03-15 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Cancer treatment with radiation and immune cytokines
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
WO2023002952A1 (en) * 2021-07-19 2023-01-26 Chugai Seiyaku Kabushiki Kaisha Protease-mediated target specific cytokine delivery using fusion polypeptide
WO2023242769A1 (en) * 2022-06-17 2023-12-21 Pfizer Inc. Il-12 variants, anti-pd1 antibodies, fusion proteins, and uses thereof
US11851466B2 (en) 2019-10-03 2023-12-26 Xencor, Inc. Targeted IL-12 heterodimeric Fc-fusion proteins

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030105294A1 (en) * 1998-02-25 2003-06-05 Stephen Gillies Enhancing the circulating half life of antibody-based fusion proteins
US20050202538A1 (en) * 1999-11-12 2005-09-15 Merck Patent Gmbh Fc-erythropoietin fusion protein with improved pharmacokinetics
ES2269366T3 (en) 2000-02-11 2007-04-01 Merck Patent Gmbh IMPROVEMENT OF AVERAGE LIFE IN CIRCULATION OF FUSION PROTEINS BASED ON ANTIBODIES.
BR0207854A (en) 2001-03-07 2004-08-24 Merck Patent Gmbh Expression technology for proteins containing a hybrid isotype antibody moiety
WO2002079415A2 (en) 2001-03-30 2002-10-10 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
DK1383785T3 (en) 2001-05-03 2011-05-23 Merck Patent Gmbh Recombinant tumor-specific antibody and its use
DK1454138T3 (en) 2001-12-04 2012-02-13 Merck Patent Gmbh Immunocytokines with modulated selectivity
CN100467488C (en) 2003-12-30 2009-03-11 默克专利有限公司 Il-7 fusion proteins
JP2008504008A (en) * 2003-12-31 2008-02-14 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Fc-erythropoietin fusion protein with improved pharmacokinetics
US7670595B2 (en) * 2004-06-28 2010-03-02 Merck Patent Gmbh Fc-interferon-beta fusion proteins
US20070104689A1 (en) * 2005-09-27 2007-05-10 Merck Patent Gmbh Compositions and methods for treating tumors presenting survivin antigens
CA2635618C (en) 2005-12-30 2015-10-06 Merck Patent Gesellschaft Mit Beschraekter Haftung Interleukin-12p40 variants with improved stability
US20100048482A1 (en) * 2008-08-15 2010-02-25 Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for modulating epsilon protein kinase c-mediated cytoprotection
EA201171259A1 (en) 2009-04-22 2012-05-30 Мерк Патент Гмбх ANTIBODY HYBRID PROTEINS WITH MODIFIED FCRN BINDING SITES
EP2709651B1 (en) 2011-05-19 2017-10-04 Rush University Medical Center Il-12 p40 monomer, monoclonal antibody against p40 homodimer and the combination of the two for autoimmune disease treatment
CA2940018C (en) * 2014-02-19 2018-12-04 Merck Patent Gmbh Cancer-targeted il-12 immunotherapy
LT3458083T (en) 2016-05-18 2023-02-10 Modernatx, Inc. Polynucleotides encoding interleukin-12 (il12) and uses thereof
CN106533815B (en) * 2017-01-13 2019-10-29 邦彦技术股份有限公司 Gateway control method and device based on capability characteristics
CN108574635B (en) * 2017-03-09 2021-06-22 华为技术有限公司 Routing priority configuration method, device and controller
CN108623691B (en) * 2017-03-17 2020-05-15 北京比洋生物技术有限公司 IgG (immunoglobulin G) -like long-acting immune fusion protein and application thereof
JP7285220B2 (en) 2017-05-18 2023-06-01 モデルナティエックス インコーポレイテッド Lipid nanoparticles comprising linked interleukin-12 (IL12) polypeptide-encoding polynucleotides
US20210017247A1 (en) 2017-07-03 2021-01-21 Torque Therapeutics, Inc. Fusion Molecules Targeting Immune Regulatory Cells and Uses Thereof
CN110264184B (en) * 2019-06-28 2021-07-23 Oppo(重庆)智能科技有限公司 Payment control method and related product
WO2022155263A2 (en) * 2021-01-12 2022-07-21 Askgene Pharma Inc. Chimeric molecules comprising il-12 agonist polypeptide
WO2023043978A2 (en) * 2021-09-17 2023-03-23 Bicara Therapeutics Inc. Caix targeting il-12 fusion proteins and methods of use thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0433827B1 (en) * 1989-12-22 1998-03-04 F. Hoffmann-La Roche Ag Cytotoxic lymphocyte maturation factor
WO1999029732A2 (en) * 1997-12-08 1999-06-17 Lexigen Pharmaceuticals Corporation Heterodimeric fusion proteins useful for targeted immune therapy and general immune stimulation
WO2001018051A2 (en) * 1999-09-09 2001-03-15 Schering Corporation Mammalian interleukin-12 p40 and interleukin b30, combinations thereof, antibodies, uses in pharmaceutical compositions
WO2001040257A2 (en) * 1999-12-02 2001-06-07 Maxygen, Inc. Modified il-12 subunits and nucleic acids encoding them
JP2001161378A (en) * 1999-10-01 2001-06-19 Toray Ind Inc Method for producing pure protein, interleukin 12, protein inhibiting activity of interleukin 12, medicine for treating immunological disease of mammal, and method for treating immunological disease

Family Cites Families (102)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US647717A (en) * 1897-12-17 1900-04-17 Guilford S Wood Rotary engine.
US4196265A (en) * 1977-06-15 1980-04-01 The Wistar Institute Method of producing antibodies
US5082658A (en) * 1984-01-16 1992-01-21 Genentech, Inc. Gamma interferon-interleukin-2 synergism
US5679543A (en) * 1985-08-29 1997-10-21 Genencor International, Inc. DNA sequences, vectors and fusion polypeptides to increase secretion of desired polypeptides from filamentous fungi
US4676980A (en) * 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4935233A (en) * 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US5359035A (en) * 1985-12-21 1994-10-25 Hoechst Aktiengesellschaft Bifunctional proteins including interleukin-2 (IL-2) and granuloctyte macrophage colony stimulating factor (GM-CSF)
DE3883899T3 (en) 1987-03-18 1999-04-22 Sb2 Inc CHANGED ANTIBODIES.
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
DE3856559T2 (en) 1987-05-21 2004-04-29 Micromet Ag Multifunctional proteins with predetermined objectives
US5258498A (en) * 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
ATE122238T1 (en) 1987-06-10 1995-05-15 Dana Farber Cancer Inst Inc BIFUNCTIONAL ANTIBODY CONSTRUCTIONS AND METHOD FOR SELECTIVE KILLING OF CELLS.
US5064646A (en) 1988-08-02 1991-11-12 The University Of Maryland Novel infectious bursal disease virus
ES2054753T3 (en) 1987-09-02 1994-08-16 Ciba Geigy Ag CONJUGATES OF CYTOKINES WITH IMMUNOGLOBULINS.
US5677425A (en) * 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
DE3850542T2 (en) 1987-09-23 1994-11-24 Bristol Myers Squibb Co Antibody heteroconjugates for killing HIV-infected cells.
NZ226414A (en) 1987-10-02 1992-07-28 Genentech Inc Cd4 peptide adhesion variants and their preparation and use
JP3040121B2 (en) * 1988-01-12 2000-05-08 ジェネンテク,インコーポレイテッド Methods of treating tumor cells by inhibiting growth factor receptor function
CA1341588C (en) 1988-01-26 2009-01-06 Michel Revel Human ifn-beta2/i1-6, its purification and use
DE3812605A1 (en) 1988-04-15 1990-06-07 Leskovar Peter Dipl Ing Dr Hab IMMUNEGULATIVE SUBSTANCES AND SUBSTANCE MIXTURES FOR ACTIVE INFLUENCING OF THE DISEASE PROCESS
US5601819A (en) * 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
US5457038A (en) * 1988-11-10 1995-10-10 Genetics Institute, Inc. Natural killer stimulatory factor
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5225538A (en) * 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5399346A (en) * 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5073627A (en) * 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
DE69030579T2 (en) 1989-09-20 1997-11-13 Abbott Lab METHOD FOR PRODUCING FUSION PROTEINS
US5196320A (en) * 1989-09-20 1993-03-23 Abbott Biotech, Inc. Method of producing engineered binding proteins
US5314995A (en) * 1990-01-22 1994-05-24 Oncogen Therapeutic interleukin-2-antibody based fusion proteins
CA2077348A1 (en) 1990-03-02 1991-09-03 Stephen D. Gillies Antibody constructs with enhanced binding affinity
CA2082804A1 (en) 1990-07-27 1992-01-28 Theodore Maione Methods and compositions for treatment of angiogenic diseases
WO1992008801A1 (en) 1990-11-09 1992-05-29 Abbott Laboratories Bridging antibody fusion constructs
WO1992008495A1 (en) 1990-11-09 1992-05-29 Abbott Biotech, Inc. Cytokine immunoconjugates
US5650150A (en) * 1990-11-09 1997-07-22 Gillies; Stephen D. Recombinant antibody cytokine fusion proteins
AU9052891A (en) 1990-12-05 1992-07-08 Novo Nordisk A/S Proteins with changed epitopes and methods for the production thereof
GB9105245D0 (en) 1991-03-12 1991-04-24 Lynxvale Ltd Binding molecules
US6072039A (en) 1991-04-19 2000-06-06 Rohm And Haas Company Hybrid polypeptide comparing a biotinylated avidin binding polypeptide fused to a polypeptide of interest
US6797492B2 (en) * 1991-05-17 2004-09-28 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
US5199942A (en) * 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
WO1993003157A1 (en) 1991-07-29 1993-02-18 Dana Farber Cancer Institute Plasmids for the rapid preparation of modified proteins
WO1993005169A1 (en) 1991-08-30 1993-03-18 Fred Hutchinson Cancer Research Center Hybrid cytokines
US20020037558A1 (en) * 1991-10-23 2002-03-28 Kin-Ming Lo E.coli produced immunoglobulin constructs
US6627615B1 (en) * 1991-12-17 2003-09-30 The Regents Of The University Of California Methods and compositions for in vivo gene therapy
GB9206422D0 (en) * 1992-03-24 1992-05-06 Bolt Sarah L Antibody preparation
WO1993024640A2 (en) * 1992-06-04 1993-12-09 The Regents Of The University Of California Methods and compositions for in vivo gene therapy
DE69332485T2 (en) * 1992-08-11 2003-11-13 Harvard College Immunomodulatory peptides
DE4228839A1 (en) 1992-08-29 1994-03-03 Behringwerke Ag Methods for the detection and determination of mediators
US5738849A (en) * 1992-11-24 1998-04-14 G. D. Searle & Co. Interleukin-3 (IL-3) variant fusion proteins, their recombinant production, and therapeutic compositions comprising them
AU6776194A (en) 1993-04-28 1994-11-21 Hybritech Incorporated Method for creating optimized regulatory regions affecting protein expression and protein trafficking
US5554512A (en) * 1993-05-24 1996-09-10 Immunex Corporation Ligands for flt3 receptors
CA2125763C (en) * 1993-07-02 2007-08-28 Maurice Kent Gately P40 homodimer of interleukin-12
GB9316989D0 (en) 1993-08-16 1993-09-29 Lynxvale Ltd Binding molecules
US5585098A (en) * 1993-11-23 1996-12-17 Ovimmune, Inc. Oral administration of chicken yolk immunoglobulins to lower somatic cell count in the milk of lactating ruminants
EP0659439B1 (en) 1993-12-24 2001-10-24 MERCK PATENT GmbH Immunoconjugates
US5541087A (en) * 1994-09-14 1996-07-30 Fuji Immunopharmaceuticals Corporation Expression and export technology of proteins as immunofusins
US6309636B1 (en) * 1995-09-14 2001-10-30 Cancer Research Institute Of Contra Costa Recombinant peptides derived from the Mc3 anti-BA46 antibody, methods of use thereof, and methods of humanizing antibody peptides
US5891680A (en) * 1995-02-08 1999-04-06 Whitehead Institute For Biomedical Research Bioactive fusion proteins comprising the p35 and p40 subunits of IL-12
US6148321A (en) * 1995-05-05 2000-11-14 Intel Corporation Processor event recognition
WO1997000317A1 (en) 1995-06-07 1997-01-03 Osteosa Inc. Osteoclast growth regulatory factor
AU1407997A (en) 1995-12-01 1997-06-19 Beth Israel Hospital Il-12 p40 subunit fusion polypeptides and uses thereof
US5723125A (en) * 1995-12-28 1998-03-03 Tanox Biosystems, Inc. Hybrid with interferon-alpha and an immunoglobulin Fc linked through a non-immunogenic peptide
AU1952497A (en) 1996-02-13 1997-09-02 Regents Of The University Of California, The Novel antibody-cytokine fusion protein, and methods of making and using the same
US5922685A (en) * 1996-06-05 1999-07-13 Powderject Vaccines, Inc. IL-12 gene therapy of tumors
US5994104A (en) * 1996-11-08 1999-11-30 Royal Free Hospital School Of Medicine Interleukin-12 fusion protein
US6100387A (en) * 1997-02-28 2000-08-08 Genetics Institute, Inc. Chimeric polypeptides containing chemokine domains
JPH1187664A (en) * 1997-04-28 1999-03-30 Nippon Steel Corp Semiconductor device and its production
ES2258817T3 (en) 1997-05-21 2006-09-01 Biovation Limited METHOD FOR THE PRODUCTION OF NON-IMMUNOGEN PROTEINS.
CA2296770A1 (en) 1997-07-14 1999-01-28 Bolder Biotechnology, Inc. Derivatives of growth hormone and related proteins
US20030105294A1 (en) * 1998-02-25 2003-06-05 Stephen Gillies Enhancing the circulating half life of antibody-based fusion proteins
EP1071468B1 (en) * 1998-04-15 2006-06-14 Lexigen Pharmaceuticals Corp. Enhancement of antibody-cytokine fusion protein mediated immune responses by co-administration with angiogenesis inhibitor
BR9909677A (en) * 1998-04-17 2000-12-19 Lexigen Pharm Corp Intensification of immune responses, mediated by antibody-cytokine fusion protein, through co-administration with prostaglandin inhibitor
WO1999058662A1 (en) 1998-05-14 1999-11-18 Merck Patent Gmbh Fused protein
BR9913331A (en) * 1998-08-25 2001-05-15 Lexigen Pharm Corp Expression and export of angiogenesis inhibitors as immunofusins
US6646113B1 (en) * 1998-09-17 2003-11-11 The Trustees Of The University Of Pennsylvania Nucleic acid molecule encoding human survival of motor neuron-interacting protein 1 (SIP1) deletion mutants
US6335176B1 (en) * 1998-10-16 2002-01-01 Pharmacopeia, Inc. Incorporation of phosphorylation sites
WO2000034317A2 (en) 1998-12-08 2000-06-15 Biovation Limited Method for reducing immunogenicity of proteins
EP1141013A2 (en) * 1999-01-07 2001-10-10 Lexigen Pharmaceuticals Corp. EXPRESSION AND EXPORT OF ANTI-OBESITY PROTEINS AS Fc FUSION PROTEINS
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
ATE369384T1 (en) * 1999-05-19 2007-08-15 Emd Lexigen Res Ct Corp EXPRESSION AND EXPORT OF INTERFERON-ALPHA PROTEINS AS FC FUSION PROTEINS
SK782002A3 (en) * 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
US7067110B1 (en) * 1999-07-21 2006-06-27 Emd Lexigen Research Center Corp. Fc fusion proteins for enhancing the immunogenicity of protein and peptide antigens
AU778611B2 (en) * 1999-08-09 2004-12-16 Merck Patent Gmbh Multiple cytokine-antibody complexes
US20050202538A1 (en) * 1999-11-12 2005-09-15 Merck Patent Gmbh Fc-erythropoietin fusion protein with improved pharmacokinetics
ES2269366T3 (en) * 2000-02-11 2007-04-01 Merck Patent Gmbh IMPROVEMENT OF AVERAGE LIFE IN CIRCULATION OF FUSION PROTEINS BASED ON ANTIBODIES.
SK982003A3 (en) * 2000-06-29 2004-05-04 Merck Patent Gmbh Enhancement of antibody-cytokine fusion protein mediated immune responses by combined treatment with immunocytokine uptake enhancing agents
US20030133939A1 (en) * 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
MXPA03006358A (en) 2001-01-17 2004-12-02 Trubion Pharmaceuticals Inc Binding domain-immunoglobulin fusion proteins.
EP1392826A2 (en) * 2001-01-18 2004-03-03 MERCK PATENT GmbH Bifunctional fusion proteins with glucocerebrosidase activity
KR100899970B1 (en) 2001-02-19 2009-05-28 메르크 파텐트 게엠베하 Method for identification of t-cell epitopes and use for preparing molecules with reeduced immunogenicity
WO2002066514A2 (en) * 2001-02-19 2002-08-29 Merck Patent Gmbh Artificial fusion proteins with reduced immunogenicity
BR0207854A (en) * 2001-03-07 2004-08-24 Merck Patent Gmbh Expression technology for proteins containing a hybrid isotype antibody moiety
WO2002079415A2 (en) * 2001-03-30 2002-10-10 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
DK1383785T3 (en) * 2001-05-03 2011-05-23 Merck Patent Gmbh Recombinant tumor-specific antibody and its use
DK1454138T3 (en) * 2001-12-04 2012-02-13 Merck Patent Gmbh Immunocytokines with modulated selectivity
US7169904B2 (en) * 2002-12-17 2007-01-30 Emd Lexigen Research Center Corp. Immunocytokine sequences and uses thereof
US20050069521A1 (en) * 2003-08-28 2005-03-31 Emd Lexigen Research Center Corp. Enhancing the circulating half-life of interleukin-2 proteins
CN100467488C (en) * 2003-12-30 2009-03-11 默克专利有限公司 Il-7 fusion proteins
JP2008504008A (en) * 2003-12-31 2008-02-14 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Fc-erythropoietin fusion protein with improved pharmacokinetics
PT1706428E (en) * 2004-01-22 2009-12-29 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US7670595B2 (en) * 2004-06-28 2010-03-02 Merck Patent Gmbh Fc-interferon-beta fusion proteins
CA2591297C (en) * 2004-12-09 2015-01-13 Stephen D. Gillies Il-7 variants with reduced immunogenicity
CA2635618C (en) 2005-12-30 2015-10-06 Merck Patent Gesellschaft Mit Beschraekter Haftung Interleukin-12p40 variants with improved stability

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0433827B1 (en) * 1989-12-22 1998-03-04 F. Hoffmann-La Roche Ag Cytotoxic lymphocyte maturation factor
WO1999029732A2 (en) * 1997-12-08 1999-06-17 Lexigen Pharmaceuticals Corporation Heterodimeric fusion proteins useful for targeted immune therapy and general immune stimulation
WO2001018051A2 (en) * 1999-09-09 2001-03-15 Schering Corporation Mammalian interleukin-12 p40 and interleukin b30, combinations thereof, antibodies, uses in pharmaceutical compositions
JP2001161378A (en) * 1999-10-01 2001-06-19 Toray Ind Inc Method for producing pure protein, interleukin 12, protein inhibiting activity of interleukin 12, medicine for treating immunological disease of mammal, and method for treating immunological disease
WO2001040257A2 (en) * 1999-12-02 2001-06-07 Maxygen, Inc. Modified il-12 subunits and nucleic acids encoding them

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL ET AL., NUCLEIC ACIDS RESEARCH, vol. 25, no. 17, 1997, pages 3389 - 3402
DATABASE JPO Proteins [online] 27 August 2002 (2002-08-27), "Process for producing highly pure protein, interleukin 12, protein inhibiting interleukin-12 activity, remedies for mammalian immunopathy and method of treating immunopathy.", XP002435977, retrieved from EBI accession no. JPOP:BD507325 Database accession no. BD507325 *
DATABASE UniProt [online] 1 June 2003 (2003-06-01), "Interleukin-12 subunit beta precursor (IL-12B) (IL-12 subunit p40) (Cytotoxic lymphocyte maturation factor 40 kDa subunit) (CLMF p40).", XP002435979, retrieved from EBI accession no. UNIPROT:Q865W9 Database accession no. Q865W9 *
DATABASE UniProt [online] 1 November 1999 (1999-11-01), "Interleukin-12 subunit beta precursor (IL-12B) (IL-12 subunit p40) (Cytotoxic lymphocyte maturation factor 40 kDa subunit) (CLMF p40).", XP002435978, retrieved from EBI accession no. UNIPROT:Q9XSQ5 Database accession no. Q9XSQ5 *
GEARING D P ET AL: "Homology of the p40 subunit of natural killer cell stimulatory factor (NKSF) with the extracellular domain of the interleukin-6 receptor [1]", CELL 1991 UNITED STATES, vol. 66, no. 1, 1991, pages 9 - 10, XP002435952, ISSN: 0092-8674 *
KARLIN, ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 2264 - 68
KARLIN, ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 77
ODBILEG R ET AL: "Cloning and sequence analysis of llama cytokines related to cell-mediated immunity", VETERINARY IMMUNOLOGY AND IMMUNOPATHOLOGY, AMSTERDAM, NL, vol. 102, no. 1-2, November 2004 (2004-11-01), pages 93 - 102, XP004586000, ISSN: 0165-2427 *
TRINCHIERI G ET AL: "NATURAL KILLER CELL STIMULATORY FACTOR (NKSF) OR INTERLEUKIN-12 IS A KEY REGULATOR OF IMMUNE RESPONSE AND INFLAMMATION", PROGRESS IN GROWTH FACTOR RESEARCH, PERGAMON PRESS, GB, vol. 4, no. 4, 1992, pages 355 - 368, XP000575698, ISSN: 0955-2235 *
WOLF S F ET AL: "CLONING OF CDNA FOR NATURAL KILLER CELL STIMULATORY FACTOR, A HETERODIMERIC CYTOKINE WITH MULTIPLE BIOLOGIC EFFECTS ON T AND NATURAL KILLER CELLS", JOURNAL OF IMMUNOLOGY, THE WILLIAMS AND WILKINS CO. BALTIMORE, US, vol. 146, no. 9, 1 May 1991 (1991-05-01), pages 3074 - 3081, XP000941940, ISSN: 0022-1767 *
YOON C ET AL: "Charged residues dominate a unique interlocking topography in the heterodimeric cytokine interleukin-12", EMBO JOURNAL 17 JUL 2000 UNITED KINGDOM, vol. 19, no. 14, 17 July 2000 (2000-07-17), pages 3530 - 3541, XP002435951, ISSN: 0261-4189 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012506394A (en) * 2008-10-21 2012-03-15 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Cancer treatment with radiation and immune cytokines
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US11655277B2 (en) 2018-10-03 2023-05-23 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US11851466B2 (en) 2019-10-03 2023-12-26 Xencor, Inc. Targeted IL-12 heterodimeric Fc-fusion proteins
WO2023002952A1 (en) * 2021-07-19 2023-01-26 Chugai Seiyaku Kabushiki Kaisha Protease-mediated target specific cytokine delivery using fusion polypeptide
WO2023242769A1 (en) * 2022-06-17 2023-12-21 Pfizer Inc. Il-12 variants, anti-pd1 antibodies, fusion proteins, and uses thereof

Also Published As

Publication number Publication date
US7872107B2 (en) 2011-01-18
JP2009521911A (en) 2009-06-11
JP5175214B2 (en) 2013-04-03
BRPI0620648A2 (en) 2011-11-22
KR20080090483A (en) 2008-10-08
PL1966238T3 (en) 2012-09-28
AU2006332138B2 (en) 2012-03-22
DK1966238T3 (en) 2012-07-16
SI1966238T1 (en) 2012-08-31
US20070154453A1 (en) 2007-07-05
CY1112884T1 (en) 2016-04-13
PT1966238E (en) 2012-07-31
CA2635618A1 (en) 2007-07-12
BRPI0620648A8 (en) 2019-01-15
BRPI0620648B1 (en) 2022-12-20
CN101351475A (en) 2009-01-21
CA2635618C (en) 2015-10-06
EA015338B1 (en) 2011-06-30
AU2006332138A1 (en) 2007-07-12
EP1966238A1 (en) 2008-09-10
US20130034518A1 (en) 2013-02-07
EP1966238B1 (en) 2012-04-25
US8188248B2 (en) 2012-05-29
US20110097792A1 (en) 2011-04-28
ES2384055T3 (en) 2012-06-28
CN101351475B (en) 2013-05-15
ATE555125T1 (en) 2012-05-15
ZA200806611B (en) 2009-12-30
KR101343682B1 (en) 2013-12-20
EA200870131A1 (en) 2008-12-30
US9029330B2 (en) 2015-05-12

Similar Documents

Publication Publication Date Title
US8188248B2 (en) Nucleic acids encoding interleukin-12P40 variants with improved stability
CN101072793B (en) Il-7 variants with reduced immunogenicity
CN100522242C (en) Artificial proteins with reduced immunogenicity
Han et al. IL-15: IL-15 receptor alpha superagonist complex: high-level co-expression in recombinant mammalian cells, purification and characterization
US11008374B2 (en) Uses of IL-15 antagonists for the treatment of autoimmune and inflammatory diseases
CA2210491C (en) Antagonists of interleukin-15
EP2409991B1 (en) Antibody constant region variant
RU2701301C2 (en) METHOD FOR PURIFYING CONJUGATES BASED ON IL-15/IL-15Rα
EP2409990A1 (en) Antibody constant region variant
CN102648002A (en) IL-17 family cytokine compositions and uses
WO2007027805A2 (en) Method for generating anti-variable region monoclonal antibodies
ES2253887T3 (en) AGONIST ANTIBODIES OF THE G-CSF RECEIVER AND THE SAME METHOD.
CN105229035A (en) Growth hormone compound
JP7175983B2 (en) Novel growth hormone receptor antagonist and its fusion protein
CN111018946B (en) Method for purifying IL-15/IL-15Rα -based conjugates
CN1246529A (en) Coding sequence of human translation initiation factor subunit, its encoded polypeptide and its preparing process
TW201714894A (en) Immunoglobulin fusion proteins

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680049818.6

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006846989

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/008408

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2635618

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2008547880

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 200870131

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2006332138

Country of ref document: AU

Ref document number: 1020087018718

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2006332138

Country of ref document: AU

Date of ref document: 20061220

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006332138

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2006846989

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0620648

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080627