USRE48959E1 - Humanized antibodies to LIV-1 and use of same to treat cancer - Google Patents

Humanized antibodies to LIV-1 and use of same to treat cancer Download PDF

Info

Publication number
USRE48959E1
USRE48959E1 US15/862,389 US201115862389A USRE48959E US RE48959 E1 USRE48959 E1 US RE48959E1 US 201115862389 A US201115862389 A US 201115862389A US RE48959 E USRE48959 E US RE48959E
Authority
US
United States
Prior art keywords
antibody
humanized antibody
liv
seq
linker
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US15/862,389
Other languages
English (en)
Inventor
Maria Leia Smith
Django Sussman
William Arthur
Albina Nesterova
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Seagen Inc
Original Assignee
Seagen Inc
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
Application filed by Seagen Inc filed Critical Seagen Inc
Priority to US15/862,389 priority Critical patent/USRE48959E1/en
Assigned to SEAGEN INC. reassignment SEAGEN INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: SEATTLE GENETICS, INC.
Application granted granted Critical
Publication of USRE48959E1 publication Critical patent/USRE48959E1/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6865Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from skin, nerves or brain cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3015Breast
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3053Skin, nerves, brain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/464Igs containing CDR-residues from one specie grafted between FR-residues from another
    • C07K16/465Igs containing CDR-residues from one specie grafted between FR-residues from another with additional modified FR-residues
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This application includes an electronic sequence listing in a file named 433583SEQLIST.txt, created on Jan. 20, 2015, and having a size of 98 kilobytes. The information in this file is hereby incorporated by reference.
  • LIV-1 is a member of the LZT (LIV-1-ZIP Zinc Transporters) subfamily of zinc transporter proteins. Taylor et al., Biochim. Biophys. Acta 1611:16-30 (2003). Computer analysis of the LIV-1 protein reveals a potential metalloprotease motif, fitting the consensus sequence for the catalytic zinc-binding site motif of the zinc metalloprotease. LIV-1 mRNA is primarily expressed in breast, prostate, pituitary gland and brain tissue.
  • LIV-1 protein has also been implicated in certain cancerous conditions, e.g. breast cancer and prostate cancer.
  • the detection of LIV-1 is associated with estrogen receptor-positive breast cancer, McClelland et al., Br. J. Cancer 77:1653-1656 (1998), and the metastatic spread of these cancers to the regional lymph nodes. Manning et al., Eur. J. Cancer 30A: 675-678 (1994).
  • the invention provides a humanized antibody comprising a mature heavy chain variable region having an amino acid sequence at least 90% identical to SEQ ID NO:53 provided that position H27 is occupied by L, position H29 is occupied by I, H30 by E and H94 by V and a mature light chain variable region at least 90% identical to SEQ ID NO:60 provided position L36 is occupied by Y and position L46 by P.
  • the humanized antibody comprises three CDRs of SEQ ID NO:53 and three CDRs of SEQ ID NO:60. Those CDRs are shown in FIG. 16 .
  • position H76 is occupied by N.
  • the humanized comprises a mature heavy chain variable region having an amino acid sequence at least 95% identical to SEQ ID NO:53 and a mature light chain variable region at least 95% identical to SEQ ID NO:60.
  • the mature heavy chain variable region is fused to a heavy chain constant region and the mature light chain constant region is fused to a light chain constant region.
  • the heavy chain constant region is a mutant form of natural human constant region which has reduced binding to an Fcgamma receptor relative to the natural human constant region.
  • the heavy chain constant region is of IgG1 isotype.
  • the heavy chain constant region has an amino acid sequence comprising SEQ ID NO:44 and the light chain constant region has an amino acid sequence comprising SEQ ID NO:42.
  • the heavy chain constant region has an amino acid sequence comprising SEQ ID NO:46 (S239C) and the light chain constant region has an amino acid sequence comprising SEQ ID NO:42.
  • the mature heavy chain variable region has an amino acid sequence designated SEQ ID NO:52 or 53 and the mature light chain variable region has an amino acid sequence designated SEQ ID NO: 59 or 60.
  • the mature heavy chain variable region has an amino acid sequence designated SEQ ID NO:53 and the mature light chain variable region has an amino acid sequence designated SEQ ID NO:60.
  • Some such humanized antibodies are conjugated to a cytotoxic or cytostatic agent. Some such humanized antibodies have an association constant for human or cynomolgus monkey LIV-1 of 0.5 to 2 ⁇ 10 8 M ⁇ 1 .
  • the invention also provides a humanized antibody comprising a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO:52, wherein position H27 is occupied by L, position H29 is occupied by I, H30 by E, H76 by N, and H94 by V and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO:60 provided position L36 is occupied by Y and position L46 by P.
  • the invention also provides a nucleic acid encoding a mature heavy chain variable region and/or a mature light chain variable region of any of the above defined humanized antibodies.
  • the invention further provides a method of treating a patient having or at risk of cancer, comprising administering to the patient an effective regime of any of the above defined humanized antibodies.
  • the cancer can be for example a breast cancer, cervical cancer, melanoma, or a prostate cancer.
  • the invention further provides a pharmaceutical composition comprising a humanized antibody as defined above.
  • the invention further provides methods of treating a subject afflicted with a melanoma that expresses the LIV-1 protein by administering to the subject a LIV-1 specific antibody or a LIV-1 antibody drug conjugate, in an amount sufficient to inhibit growth of the melanoma cancer cells.
  • the invention further provides methods of treating a subject afflicted with a cervical cancer that expresses the LIV-1 protein by administering to the subject a LIV-1 specific antibody or a LIV-1 antibody drug conjugate, in an amount sufficient to inhibit growth of the cervical cancer cells.
  • the invention further provides a humanized antibody comprising a mature heavy chain variable region having an amino acid sequence at least 90% identical to HB (SEQ ID NO:10) and a mature light chain variable region at least 90% identical to LB (SEQ ID NO:5).
  • the antibody comprises a mature heavy chain variable region having an amino acid sequence at least 95% identical to HB and a mature light chain variable region at least 95% identical to LB.
  • positions H29, H30 and H76 are occupied by I, E and N, and L36 is occupied by Y.
  • any difference in the variable region frameworks of the mature heavy chain variable region and SEQ ID NO:10 is/are selected from the group consisting of H27 occupied by F, H28 occupied by N, H48 occupied by I, H66 occupied by K, H67 occupied by A, H71 occupied by A, H76 occupied by N, H93 occupied by N, H94 occupied by V, L37 occupied by L, L39 occupied by K, L45 occupied by K, and L46 occupied by L.
  • the 3 CDRs of the mature heavy chain variable region are those of SEQ ID NO: 10 and the 3 CDRs of the mature light chain variable region are those of SEQ ID NO:15. The CDRs are shown in FIG. 1 .
  • the mature heavy chain variable region is fused to a heavy chain constant region and the mature light chain constant region is fused to a light chain constant region.
  • the heavy chain constant region is a mutant form of natural human constant region which has reached binding to an Fegamma receptor relative to the natural human constant region.
  • the heavy chain constant region is of IgG1 isotype.
  • the heavy chain constant region has an amino acid sequence comprising SEQ ID NO:6 and the light chain constant region has an amino acid sequence comprising SEQ ID NO:4.
  • the heavy chain constant region has an amino acid sequence comprising SEQ ID NO:8 (S239C) and the light chain constant region has an amino acid sequence comprising SEQ ID NO:4.
  • the mature heavy chain variable region has an amino acid sequence comprising SEQ ID NO:10 and the mature light chain variable region has an amino acid sequence comprising SEQ ID NO:15.
  • the antibody is conjugated to a cytotoxic or cytostatic agent.
  • Preferred humanized antibodies having greater affinity for LIV-1 than the antibody BR2-14a.
  • the humanized antibody has an association constant for human or cynomolgus monkey LIV-1 of 0.5 to 2 ⁇ 10 8 M ⁇ 1 .
  • the invention further provides a humanized antibody comprising a mature heavy chain variable region comprising the 3 CDRs of SEQ ID NO:10 and wherein positions H29, H30 and H76 are occupied by I, E and N respectively, and a mature light chain variable region comprising the 3 CDRs of SEQ ID NO:15, and wherein position L36 is occupied by Y.
  • the invention further provides a nucleic acid encoding a mature heavy chain variable region and/or a mature light chain variable region of any of the humanized antibodies described above.
  • the invention further provides a method of treating a patient having or at risk of cancer, comprising administering to the patient an effective regime of a humanized antibody as described above.
  • the cancer is breast cancer, cervical cancer, melanoma, or a prostate cancer.
  • the invention further provides a pharmaceutical composition comprising a humanized antibody as described above.
  • the invention further provides a method of treating a patient having or at risk of triple negative breast cancer, comprising administering to the patient an effective regime of an antibody that specifically binds to LIV-1.
  • the antibody is conjugated to a cytotoxic or cytostatic agent.
  • FIG. 1 shows an alignment of the amino acid sequences of humanized LIV-1 heavy chain variable regions for hLIV-1 HA (SEQ ID NO:9), hLIV-1 HB (SEQ ID NO:10), hLIV-1 HC (SEQ ID NO:11), hLIV-1 HD (SEQ ID NO:12), and hLIV-1 HE (SEQ ID NO:13) with the heavy chain variable region of the parental murine mAb (referred to as BR2-14a) (SEQ ID NO:86) (upper two panels).
  • BR2-14a the heavy chain variable region of the parental murine mAb
  • hLIV-1 LA (SEQ ID NO:14), hLIV-1 LB (SEQ ID NO:15), hLIV-1 LC (SEQ ID NO:16), hLIV-1 LD (SEQ ID NO:17), hLIV-1 LE (SEQ ID NO:18), and hLIV-1 LF (SEQ ID NO:19) with the light chain variable region of the parental murine mAb (referred to as BR2-14a) (SEQ ID NO:87) (lower two panels).
  • FIG. 2 shows the binding curves for the humanized LIV-1 mAbs and the parental murine antibody (referred to as BR2-14a).
  • FIG. 3 shows the results of competition binding studies of the humanized LIV-1 mAbs and the parental murine antibody (referred to as BR2-14a).
  • the numbers in parentheses after each variant indicate the number of back mutations.
  • FIG. 4 shows the results of saturation binding studies on MCF7 cells.
  • BR2-14a-AF refers to AF-labeled parental murine antibody
  • hLIV-14 refers to AF-labeled HBLB antibody, a humanized antibody that specifically binds to LIV-1.
  • FIG. 5 shows the results of competition binding studies on CHO cells expressing recombinant LIV-1 protein.
  • BR2-14a refers to the parental murine antibody.
  • hLIV-14 HBLB WT refers to the HBLB antibody,
  • hLIV-14 HBLB S239C refers to the HBLB antibody having serine to cysteine substitutions at each position in the heavy chain.
  • FIG. 6 shows an analysis of LIV-1 protein expression by IHC on post-hormone treated breast cancer patient samples.
  • FIG. 7 shows an analysis of LIV-1 protein expression by IHC on hormone-refractory metastatic prostate cancer patient samples.
  • FIG. 8 shows an analysis of LIV-1 protein expression by IHC on triple negative breast cancer patient samples.
  • FIG. 9 shows the results of cytotoxicity assays on hLIV-14 antibody drug conjugates, i.e., the HBLB mAb conjugated to vcMMAE (1006) or mcMMAF (1269), as well as conjugates of control murine (mIgG) and human (hIgG) antibodies.
  • hLIV-14-SEA-1006 refers to a non-fucosylated form of the HBLB mAb conjugated to vcMMAE (1006).
  • FIG. 10 shows the results of an in vitro ADCC assay on MCF7 cells using human NK cells (donor 1; V/V).
  • hLIV-14 WT refers to the HBLB mAb.
  • hLIV-14 SEA refers to the non-fucosylated form of the HBLB mAb.
  • hLIV-14 mcMMAF refers to an antibody drug conjugate of the HBLB mAb conjugated to mcMMAF.
  • hLIV-14 vcMMAE refers to an antibody drug conjugate of the HBLB mAb conjugated to vcMMAE.
  • hLIV-14 SEA vcMMAE refers to a non-fucosylated form of the HBLB mAb-vcMMAE antibody drug conjugate.
  • FIG. 11 shows the results of an in vitro ADCC assay on MCF7 cells using human NK cells (donor 2).
  • hLIV-14 WT refers to the HBLB mAb.
  • hLIV-14 SEA refers to the non-fucosylated form of the HBLB mAb.
  • cLIV-14 SEA refers to the non-fucosylated form of the chimeric parental murine antibody
  • hLIV-14 mcF(4) refers to an antibody drug conjugate of the HBLB mAb with an average of 4 mcMMAF drug linker molecules per antibody.
  • hLIV-14 vcE(4) refers to an antibody drug conjugate of the HBLB mAb with an average of 4 vcMMAE drug linker molecules per antibody.
  • hLIV-14 vcE(4) SEA refers to a non-fucosylated form of the HBLB mAb-vcMMAE antibody drug conjugate having an average of four vcMMAE drug linker molecules per antibody.
  • hIgG refers to control human IgG.
  • H00-mcF(4) refers to a control antibody drug conjugate of a nonbinding antibody with an average of 4 mcMMAF drug linker molecules per antibody.
  • H00-vcE(4) refers to a control antibody drug conjugate of a nonbinding antibody with an average of 4 vcMMAE drug linker molecules per antibody.
  • FIG. 12 shows the results of a xenograft study of the MCF7 breast cancer line in nude mice.
  • cLIV-14-mcMMAF(4) refers to an antibody drug conjugate of the chimeric form of the parental murine antibody having an average of 4 mcMMAF drug linker molecules per antibody.
  • cLIV-14- vcMMAE(4) refers to an antibody drug conjugate of the chimeric form of the present murine antibody having an average of 4 vcMMAE drug linker molecules per antibody.
  • H00-mcMMAF(4) refers to an antibody drug conjugate of a nonbinding control antibody having an average of 4 mcMMAF drug linker molecules per antibody.
  • H00-vcMMAE(4) refers to an antibody drug conjugate of a nonbinding control antibody having an average of 4 vcMMAE drug linker molecules per antibody. The dose and time of administration of indicated on the figures.
  • FIG. 13 shows the results of a xenograft study of the PC3 prostate cancer line in male nude mice.
  • cLIV-14-vcMMAE(4) refers to an antibody drug conjugate of the chimeric form of the parent murine antibody having an average of 4 vcMMAE drug linker molecules per antibody.
  • hBUC12-vcMMAE(4) refers to an antibody drug conjugate of an anti-CD19 antibody having an average of 4 vcMMAE drug linker molecules per antibody. The dose and time of administration of indicated on the figure.
  • FIG. 14 shows the results of a xenograft study of the MCF7 breast cancer line in nude mice
  • hLIV-14-vcMMAE(4) refers to an antibody drug conjugate of the HBLB antibody having an average of 4 vcMMAE drug linker molecules per antibody.
  • hLIV-14d-vcMMAE(2) refers to an antibody drug conjugate of the HBLB antibody having an average of 2 vcMMAE drug linker molecules per antibody, each conjugated at the S239C position of each heavy chain.
  • H00-vcMMAE(4) refers to an antibody drug conjugate of a nonbinding control antibody having an average of 4 vcMMAE drug linker molecules per antibody. The dose and time of administration of indicated on the figure.
  • FIG. 15 shows the results of a xenograft study of the PC3 prostate cancer line in male nude mice.
  • hLIV-14-vcMMAE(4) refers to an antibody drug conjugate of the HBLB antibody having an average of 4 vcMMAE drug linker molecules per antibody.
  • hLIV-14-mcMMAF(4) refers to an antibody drug conjugate of the HBLB antibody having an average of 4 mcMMAF drug linker molecules per antibody.
  • hLIV-14d-mcMMAE(2) refers to an antibody drug conjugate of the HBLB antibody having an average of 2 vcMMAE drug linker molecules per antibody, each conjugated at the S239C position of each heavy chain.
  • hLIV-14d-mcMMAF(2) refers to an antibody drug conjugate of the HBLB antibody having an average of 2 mcMMAF drug linker molecules per antibody, each conjugated at the S239C position of each heavy chain.
  • H00-vcMMAE(4) refers to an antibody drug conjugate of a nonbinding control antibody having an average of 4 vcMMAE drug linker molecules per antibody.
  • H00-mcMMAF(4) refers to an antibody drug conjugate of a nonbinding control antibody having an average of 4 mcMMAF drug linker molecules per antibody. The dose and time of administration of indicated on the figure.
  • FIGS. 16A and 16B show alignments of humanized heavy chain ( FIG. 16A ) and light chain ( FIG. 16B ) mature variable regions with those of the mouse BR2-22a.
  • FIG. 16A shows an alignment of the amino acid sequences of humanized heavy chain variable regions for hLIV-22 HA (SEQ ID NO:47), hLIV-22 HB (SEQ ID NO:48), hLIV-22 HC (SEQ ID NO:49), hLIV-22 HD (SEQ ID NO:50), hLIV-22 HE (SEQ ID NO:51), hLIV-22 HF (SEQ ID NO:52), and hLIV-22 HG (SEQ ID NO:53) with the heavy chain variable region of the parental murine mAb (referred to as BR2-22a) (SEQ ID NO:88).
  • FIG. 16B shows an alignment of the amino acid sequences of humanized light chain variable regions for hLIV-22 LA (SEQ ID NO:54), hLIV-22 LB (SEQ ID NO:55), hLIV-22 LC (SEQ ID NO:56).
  • hLIV-22 LD SEQ ID NO:57
  • hLIV-22 LE SEQ ID NO:58
  • hLIV-22 LF SEQ ID NO:59
  • hLIV-22 LG SEQ ID NO:60
  • FIG. 17 shows competition binding assays of different permutations of humanized heavy chains HA-HF and humanized light chains LA-LF derived from the murine monoclonal anti LIV-1antibody BR2-22a. The total number of murine back mutations in each light or heavy chain is shown in parentheses. Only HELF showed sufficient retention of binding.
  • FIG. 18 shows systemic variation of the HE and LF chains to test contribution of individual backmutations to antigen binding. Sites of potential somatic hypermutations are in parentheses. Mouse residues are underlined. The remaining residues are human germline residues.
  • FIG. 19 shows competition binding of the LF variants on the top of the figure.
  • the tested back mutations are shown in the bottom of the figure.
  • Mouse residues are underlined.
  • the remaining residues are human germline residues.
  • FIG. 20 shows competition binding of the HE variants on the top of the figure. The tested back mutations are shown in the bottom of the figure. Mouse residues are underlined. The remaining residues are human germline residues.
  • FIG. 21 shows competition binding of different permutations of HE, HF, HG and LF and LG.
  • FIG. 22 shows saturation binding of humanized LIV 14 antibody and humanized LIV22 antibody on human and cynomolgus LIV-1 expressed from CHO cells.
  • FIG. 23 shows cytotoxic activity of humanized LIV22-vcMMAE on MCF-7 cells after 144 hr of treatment.
  • h00-1006 is a control drug-conjugated antibody.
  • FIG. 24 shows cytotoxic activity of hLIV22-mcMMAF on MCF-7 cells after 144 hr of treatment.
  • h00-1269 is a control drug-conjugated antibody.
  • FIG. 25 shows the activity of hLIV22- antibody on PC3 (DSMZ) prostate carcinoma model in nude female mice. Dose days are indicated by triangles on the X-axis.
  • FIG. 26 shows that activity of hLIV22 antibody on MCF7 (NCI) breast carcinoma tumors in nude mice.
  • FIG. 27 compares the activity of hLIV22 and hLIV14 in the same mode as FIG. 26 .
  • FIG. 28 shows an analysis of LIV-1 protein expression by IHC on melanoma cancer patient samples.
  • Monoclonal antibodies are typically provided in isolated form. This means that an antibody is typically at least 50% w/w pure of interfering proteins and other contaminants arising from its production or purification but does not exclude the possibility that the monoclonal antibody is combined with an excess of pharmaceutical acceptable carrier(s) or other vehicle intended to facilitate its use. Sometimes monoclonal antibodies are at least 60%, 70%, 80%, 90%, 95 or 99% w/w pure of interfering proteins and contaminants from production or purification.
  • Specific binding of a monoclonal antibody to its target antigen means an affinity of at least 10 6 , 10 7 , 10 8 , 10 9 , or 10 10 M ⁇ 1 . Specific binding is detachably higher in magnitude and distinguishable from non-specific binding occurring to at least one unrelated target. Specific binding can be the result of formation of bonds between particular functional groups or particular spatial fit (e.g., lock and key type) whereas nonspecific binding is usually the result of van der Waals forces. Specific binding does not however necessarily imply that a monoclonal antibody binds one and only one target.
  • the basic antibody structural unit is a tetramer of subunits.
  • Each tetramer includes two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. This variable region is initially expressed linked to a cleavable signal peptide.
  • the variable region without the signal peptide is sometimes referred to as a mature variable region.
  • a light chain mature variable region means a light chain variable region without the light chain signal peptide.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively.
  • the variable and constant regions are joined by a “J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 or more amino acids.
  • variable regions of each light/heavy chain pair form the antibody binding site.
  • an intact antibody has two binding sites.
  • the chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs.
  • the CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope.
  • FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4 From N-terminal to C-terminal, both light and heavy chains comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • Kabat Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1987 and 1991), or Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987); Chothia et al., Nature 342:878-883 (1989).
  • Kabat also provides a widely used numbering convention (Kabat numbering) in which corresponding residues between different heavy chains or between different light chains are assigned the same number.
  • antibody includes intact antibodies and binding fragments thereof. Typically, antibody fragments compete with the intact antibody from which they were derived for specific binding to the target including separate heavy chains, light chains Fab, Fab′, F(ab′) 2 , F(ab)c, diabodies, Dabs, nanobodies, and Fv. Fragments can be produced by recombinant DNA techniques, or by enzymatic or chemical separation of intact immunoglobulins.
  • antibody also includes a diabody (homodimeric Fv fragment) or a minibody (V L -V H -C H 3), a bispecific antibody or the like.
  • a bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites (see, e.g., Songsivilai and Lachmann, Clin. Exp. Immunol., 79:315-321 (1990); Kostelny et al., J. Immunol., 148: 1547-53 (1992)).
  • the term “antibody” includes an antibody by itself (naked antibody) or an antibody conjugated to a cytotoxic or cytostatic drug.
  • epitope refers to a site on an antigen to which an antibody binds.
  • An epitope can be formed from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of one or more proteins. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols, in Methods in Molecular Biology, Vol. 66, Glenn E. Morris, Ed. (1996).
  • Antibodies that recognize the same or overlapping epitopes can be identified in a simple immunoassay showing the ability of one antibody to compete with the binding of another antibody to a target antigen.
  • the epitope of an antibody can also be defined by X-ray crystallography of the antibody bound to its antigen to identify contact residues.
  • two antibodies have the same epitope if all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • Two antibodies have overlapping epitopes if some amino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • Competition between antibodies is determined by an assay in which an antibody under test inhibits specific binding of a reference antibody to a common antigen (see, e.g., Junghans et al., Cancer Res. 50:1495, 1990).
  • a test antibody competes with a reference antibody if an excess of a test antibody (e.g., at least 2 ⁇ , 5 ⁇ , 10 ⁇ , 20 ⁇ or 100 ⁇ ) inhibits binding of the reference antibody by at least 50% but preferably 75%, 90%, or 99% as measured in a competitive binding assay.
  • Antibodies identified by competition assay include antibodies binding to the same epitope as the reference antibody and antibodies binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antibody for steric hindrance to occur.
  • patient includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.
  • amino acids are grouped as follows: Group I (hydrophobic side chains): met, ala, val, len, ile; Group II (neutral hydrophilic side chains): cys, ser, thr; Group III (acidic side chains): asp, glu; Group IV (basic side chains): asn, gln, his, lys, arg: Group V (residues influencing chain orientation): gly, pro; and Group VI (aromatic side chains): trp, tyr, phe. Conservative substitutions involve substitutions between amino acids in the same class. Non-conservative substitutions constitute exchanging a member of one of these classes for a member of another.
  • Percentage sequence identifies are determined with antibody sequences maximally aligned by the Kabat numbering convention. After alignment, if a subject antibody region (e.g., the entire mature variable region of a heavy or light chain) is being compared with the same region of a reference antibody, the percentage sequence identity between the subject and reference antibody regions is the number of positions occupied by the same amino acid in both the subject and reference antibody region divided by the total number of aligned positions of the two regions, with gaps not counted, multiplied by 100 to convert to percentage.
  • a subject antibody region e.g., the entire mature variable region of a heavy or light chain
  • compositions or methods “comprising” one or more recited elements may include other elements not specifically recited.
  • a composition that comprises antibody may contain the antibody alone or in combination with other ingredients.
  • Designation of a range of values includes all integers within or defining the range.
  • An antibody effector function refers to a function contributed by an Fc domain(s) of an Ig.
  • Such functions can be, for example, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis or complement-dependent cytotoxicity.
  • Such function can be effected by, for example, binding of an Fc effector domain(s) to an Fc receptor on an immune cell with phagocytic or lytic activity or by binding of an Fc effector domain(s) to components of the complement system.
  • the effect(s) mediated by the Fc-binding cells or complement components result in inhibition and/or depletion of the LIV-1 targeted cell.
  • Fc regions of antibodies can recruit Fc receptor (FcR)-expressing cells and juxtapose them with antibody-coated target cells.
  • Fc ⁇ RIII CD16
  • Fc ⁇ RII CD32
  • Fc ⁇ RIII CD64
  • effector cells include monocytes, macrophages, natural killer (NK) cells, nentrophils and eosinophils.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • Fc regions of cell-bound antibodies can also activate the complement classical pathway to elicit complement-dependent cytotoxicity (CDC).
  • Clq of the complement system binds to the Fc regions of antibodies when they are complexed with antigens. Binding of Clq to cell-bound antibodies can initiate a cascade of events involving the proteolytic activation of C4 and C2 to generate the C3 convertase. Cleavage of C3 to C3b by C3 convertase enables the activation of terminal complement components including C5b, C6, C7, C8 and C9.
  • ADCC antibody-dependent cellular cytotoxicity
  • immune cells possessing lytic activity also referred to as effector cells.
  • effector cells include natural killer cells, monocytes/macrophages and neutrophils.
  • the effector cells attach to an Fc effector domain(s) of Ig bound to target cells via their antigen-combining sites. Death of the antibody-coated target cell occurs as a result of effector cell activity.
  • ADCP antibody-dependent cellular phagocytosis
  • phagocytic immune cells e.g., macrophages, neutrophils and dendritic cells
  • complement-dependent cytotoxicity refers to a mechanism for inducing cell death in which an Fc effector domain(s) of a target-bound antibody activates a series of enzymatic reactions culminating in the formation of holes in the target cell membrane.
  • antigen-antibody complexes such as those on antibody-coated target cells bind and activate complement component Clq which in turn activates the complement cascade leading to target cell death.
  • Activation of complement may also result in deposition of complement components on the target cell surface that facilitate ADCC by binding complement receptors (e.g., CR3) on lenkocytes.
  • complement receptors e.g., CR3
  • a “cytotoxic effect” refers to the depletion, elimination and/or the killing of a target cell.
  • a “cytotoxic agent” refers to an agent that has a cytotoxic effect on a cell. Cytotoxic agents can be conjugated to an antibody or administration in combination with an antibody.
  • a “cytostatic effect” refers to the inhibition of cell proliferation.
  • a “cytostatic agent” refers to an agent that has a cytostatic effect on a cell, thereby inhibiting the growth and/or expansion of a specific subset of cells. Cytostatic agents can be conjugated to an antibody or administered in combination with an antibody.
  • pharmaceutically acceptable means approved or approvable by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopenia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • pharmaceutically compatible ingredient refers to a pharmaceutically acceptable diluent, adjuvant, excipient, or vehicle with which an anti-LIV-1 antibody.
  • phrases “pharmaceutically acceptable salt,” refers to pharmaceutically acceptable organics or inorganic salts of an anti-LIV-1 antibody or conjugate thereof or agent administered with an anti-LIV-1 antibody.
  • Exemplary salts includes sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, glyconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1,1′ methylene bis-(
  • a pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counterion.
  • the counterion may be any organic or inorganic moiety that stabilizes the charge on the parent compound.
  • a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counterion.
  • the invention provides monoclonal antibodies that specifically bind to LIV-1.
  • the antibodies are useful for treatment and diagnoses of various cancers as well as detecting LIV-1.
  • LIV-1 means a human LIV-1.
  • An exemplary human sequence is assigned Swiss Prot accession number Q13433.
  • Q13433 is included herein as SEQ ID NO:83.
  • Three variant isoforms and one polymorphism are known.
  • a second version of the human LIV-1 protein, accession number AAA96258.2, is included herein as SEQ ID NO:84.
  • Four extracellular domains are bounded by residues 29-325, 377-423, 679-686 and 746-755 of Q13433 respectively.
  • LIV-1 means at least an extracellular domain of the protein and usually the complete protein other than a cleavable signal peptide (amino acids 1-28 of Q13433).
  • the invention provides humanized antibodies derived from two mouse antibodies, BR-2-14and BR2-22a. Unless specifically indicated otherwise, the present disclosures relate to both antibodies.
  • the two mouse antibodies show 94% and 91% sequence identity to one another in the nature heavy and light chain variable regions.
  • the two antibodies bind to the same or overlapping epitopes on human LIV-1.
  • the BR2-22a antibody has about ten-fold higher affinity for human LIV-1 and about 3-fold higher affinity for cynomolgus monkey LIV-1 than BR2-14a as shown in FIG. 22 .
  • the affinity of humanized forms of the mouse BR2-14a antibody (i.e., Ka) is preferably within a factor of five or a factor of two of that of the mouse antibody BR2-14a for human LIV-1.
  • Humanized BR2-14a antibodies specifically bind to human LIV-1 in native form and/or recombinantly expressed from CHO cells as does the mouse antibody from which they were derived.
  • Preferred humanized BR2-14a antibodies have an affinity the same as or greater than (i.e., greater than beyond margin of error in measurement) that of BR2-14a for human LIV-1 (e.g., 1.1-5 fold, 1.1 to 3 fold, 1.5 to 3-fold, 1.7 to 2.3-fold or 1.7-2.1-fold the affinity or about twice the affinity of BR2-14a).
  • Preferred humanized BR2-14a antibodies bind to the same epitope and/or compete with BR2-14a for binding to human LIV-1.
  • Preferred humanized BR2-14a antibodies also bind to the cyno-homolog of LIV-1 thus permitting preclinical in nonhuman primates.
  • the affinity of humanized forms of the mouse BR2-22a antibody (i.e., Ka) for human LIV-1, natively expressed or expressed from CHO cells, is preferably within a factor of five or a factor of two of that of the mouse antibody BR2-22.
  • Some humanized BR2-22a antibodies have an association constant that is essentially the same as that of BR2-22a (i.e., within experimental error).
  • Some humanized BR2-22a antibodies have an association constant within a range of 0.5 to 1 or 0.5-1.5 that of the association constant of the BR2-22a antibody.
  • Preferred humanized BR2-22a antibodies have an association constant greater than 5 ⁇ 10 8 M ⁇ 1 , or in a range of 0.5 to 2 ⁇ 10 9 M ⁇ 1 or about 0.8 ⁇ 10 9 M ⁇ 1 (+/ ⁇ error in measurement) for human LIV-1 expressed from CHO cells.
  • affinities can be measured in accordance with the methods of the Examples.
  • Preferred humanized BR2-22a antibodies bind to the same epitope and/or compete with BR2-22a for binding to human LIV-1.
  • Humanized BR2-22a antibodies bind to the cyno-homolog of LIV-1 as well as human LIV-1.
  • Preferred humanized BR2-22a antibodies bind with essentially the same association constant to human and cynomolgus monkey LIV-1 both expressed from CHO cells (within experimental error) thus permitting and increasing the predictive accuracy of preclinical testing in nonhuman primates.
  • Preferred antibodies inhibit cancer (e.g., growth of cells, metastasis and/or lethality to the organisms) as shown on cancerous cells propagating in culture, in an animal model or clinical trial.
  • Animal models can be formed by implanting LIV-1-expressing human tumor cell lines into appropriate immunodeficient rodent strains, e.g., athymic nude mice or SCID mice. These tumor cell lines can be established in immunodeficient rodent hosts either as solid tumor by subcutaneous injections or as disseminated tumors by intravenous injections. Once established within a host, these tumor models can be applied to evaluate the therapeutic efficacies of the anti-LIV-1 antibodies or conjugated forms thereof as described in the Examples.
  • a humanized antibody is a genetically engineered antibody in which the CDRs from a non-human “donor” antibody are grafted into human “acceptor” antibody sequences (see, e.g., Queen, U.S. Pat. Nos. 5,530,101, and 5,585,089; Winter, U.S. Pat. No. 5,225,539; Carter, U.S. Pat. No. 6,407,213; Adair, U.S. Pat. No. 5,859,205; and Foote, U.S. Pat. No. 6,881,557).
  • the acceptor antibody sequences can be, for example, a mature human antibody sequence, a composite of such sequences, a consensus sequence of human antibody sequences, or a germline region sequence.
  • a preferred acceptor sequence for the heavy chain is the germline V H exon V H 1-2 (also referred to in the literature as HV1-2) (Shin et al., 1991, EMBO J, 10:364-3645) and for the hinge region (J H ), exon J H -6 (Mattila et al., 1995, Eur. J. Immunol. 25:2578-2582).
  • a preferred acceptor sequence is exon VK2-30 (also referred to in the literature as KV2-30) and for the hinge region exon J ⁇ -4 (Hieter et al., 1982, J. Biol. Chem. 257:1516-1522).
  • a humanized antibody is an antibody having some or all CDRs entirely or substantially from a donor antibody and variable region framework sequences and constant regions, if present, entirely or substantially from human antibody sequences.
  • a humanized heavy chain has at least one, two and usually all three CDRs entirely or substantially from a donor antibody heavy chain, and a heavy chain variable region framework sequence and heavy chain constant region, if present, substantially from human heavy chain variable region framework and constant region sequences.
  • a humanized light chain has at least one, two and usually all three CDRs entirely or substantially from a donor antibody light chain, and a light chain variable region framework sequence and light chain constant region, if present, substantially from human light chain variable region framework and constant region sequences.
  • a humanized antibody comprises a humanized heavy chain and a humanized light chain.
  • a CDR in a humanized antibody is substantially from a corresponding CDR in a non-human antibody when at least 60%, 85%, 90%, 95% or 100% of corresponding residues (as defined by Kabat) are identical between the respective CDRs.
  • the variable region framework sequences of an antibody chain or the constant region of an antibody chain are substantially from a human variable region framework sequence or human constant region respectively when at least 85%, 90%, 95% or 100% of corresponding residues defined by Kabat are identical.
  • humanized antibodies often incorporate all six CDRs (preferably as defined by Kabat) from a mouse antibody, they can also be made with less than all CDRs (e.g., at least 3, 4, or 5) CDRs from a mouse antibody (e.g., Pascalis et al., J. Immunol. 169:3076, 2002; Vajdos et al., Journal of Molecular Biology, 320: 415-428, 2002; Iwahashi et al., Mol. Immunol. 36:1079-1091, 1999; Tamura et al, Journal of Immunology, 164:1432-1441, 2000).
  • CDRs e.g., Pascalis et al., J. Immunol. 169:3076, 2002; Vajdos et al., Journal of Molecular Biology, 320: 415-428, 2002; Iwahashi et al., Mol. Immunol. 36:1079-1091, 1999; Tamura et al, Journal of Immunology, 164:1432-1
  • Certain amino acids from the human variable region framework residues can be selected for substitution based on their possible influence on CDR configuration and/or binding to antigen. Investigation of such possible influences is by modeling, examination of the characteristics of the amino acids at particular locations, or empirical observation of the effects of substitution or mutagenesis of particular amino acids.
  • the human framework amino acid when an amino acid differs between a murine variable region framework residue and a selected human variable region framework residue, the human framework amino acid can be substituted by the equivalent framework amino acid from the mouse antibody when it is reasonably expected that the amino acid:
  • the invention provides humanized forms of the mouse BR2-14a antibody including five exemplified humanized heavy chain mature variable regions (HA-HE) and six exemplified humanized light chain mature variable regions (LA-LF).
  • the permutations of these chains having the strongest binding (lowest BC50) and HBLB, HBLF, HCLB, HCLF, HDLB, HDLF, HELE and HELF.
  • HBLB also know as hLIV14
  • it has the strongest binding, about 2 fold stronger than the mouse donor antibody, and has the fewest back mutations (four).
  • the invention provides variants of the HBLB humanized antibody in which the humanized heavy chain mature variable region shows at least 90%, 95% or 99% identity to SEQ ID NO:10 and the humanized light chain mature variable region shows at least 90%, 95% or 99% sequence identity to SEQ ID NO:15.
  • the humanized heavy chain mature variable region shows at least 90%, 95% or 99% identity to SEQ ID NO:10
  • the humanized light chain mature variable region shows at least 90%, 95% or 99% sequence identity to SEQ ID NO:15.
  • some or all of the backmutations in HBLB are retained.
  • at least 1, 2 or preferably all 3 of heavy chain positions H29, H30 and H76 are occupied by I and E and N, respectively.
  • position L36 is preferably occupied by Y.
  • the CDR regions of such humanized antibodies are preferably substantially identical to the CDR regions of HBLB, which are the same as those of the mouse donor antibody.
  • the CDR regions can be defined by any conventional definition (e.g., Chothia) but are preferably as defined by Kabat.
  • the humanized antibody comprises a heavy chain comprising the 3 CDRs of SEQ ID NO:10 and variable region frameworks with a least 95% identity to the variable region frameworks of SEQ ID NO: 10.
  • the humanized antibody comprises a light chain comprising the 3 CDRs of SEQ ID NO:15 and variable region frameworks with at least 95% identity to variable region frameworks of SEQ ID NO:15.
  • the humanized antibody comprises a heavy chain comprising the 3 CDRs of SEQ ID NO:10 and variable region frameworks with at least 95% identity to the variable region frameworks of SEQ ID NO:10, and a light chain comprising the 3 CDRs of SEQ ID NO:15, and variable region frameworks with at least 95% identity to the variable region frameworks of SEQ ID NO:15.
  • variable region frameworks Any or all of the positions backmutated in other exemplified humanized heavy or light chain mature variable regions can also be made (i.e., 1, 2, 3, 4, 5, 6, 7, 8 or all 9 of H27 occupied by P, H28 occupied by N, H48 occupied by I, H66 occupied by K, H67 occupied by A, H71 occupied by A, H76 occupied by N, H93 occupied by N and H94 occupied by V in the heavy chain and 1, 2, 3, 4 or all 5 of L37 occupied by L, L39 occupied by K, L45 occupied by K, and L46 occupied by L in the light chain.
  • additional backmutations are not preferred because they in general do not improve affinity and introducing more mouse residues may give increased risk of immunogenicity.
  • the invention provides humanized forms of the mouse BR2-22a antibody including three exemplified humanized heavy chain mature variable regions (HE, HF and HG) and two exemplified humanized light chain (LF and LG) which can be combined in different permutations with adequate binding (see FIG. 21 ).
  • HGLG also known as hLIV22
  • HGLG is preferred because it has the best combination of binding properties (essentially the same as the mouse BR2-22a antibody within experimental error) and fewest back mutations (seven).
  • the invention provides variants of the HGLG humanized antibody in which the humanized heavy chain mature variable region shows at least 90%, 95%, 98% or 99% identity to SEQ ID NO:53 and the humanized light chain mature variable region shows at least 90%, 95%, 98% or 99% sequence identity to SEQ ID NO:60.
  • the humanized heavy chain mature variable region shows at least 90%, 95%, 98% or 99% identity to SEQ ID NO:53 and the humanized light chain mature variable region shows at least 90%, 95%, 98% or 99% sequence identity to SEQ ID NO:60.
  • some or all of the backmutations in HGLG are retained.
  • at least 1, 2, 3, 4 or preferably all 5 of heavy chain positions H27, H29, H30, H76, and H94 are occupied by L, I, E, N and V (here, as elsewhere in this application Kabat numbering is used to describe positions in the nature variable heavy and light chain variable regions).
  • H94 contributes the most to retention of binding affinity and H76 the least.
  • positions L36 and L46 are preferably occupied by Y and P respectively.
  • the CDR regions of such humanized antibodies are preferably substantially identical to the CDR regions of HGLG, which are the same as those of the mouse donor antibody.
  • the CDR regions can be defined by any conventional definition (e.g., Chothia) but are preferably as defined by Kabat.
  • the humanized antibody comprises a heavy chain comprising the 3 CDRs of SEQ ID NO:53 and variable region frameworks with at least 95% identity to the variable region frameworks of SEQ ID NO:53.
  • the humanized antibody comprises a light chain comprising the 3 CDR's of SEQ ID NO:60 and variable region frameworks with at least 95% identity to the variable region frameworks of SEQ ID NO:60.
  • the humanized antibody comprises a heavy chain comprising the 3 CDRs of SEQ ID NO:53 and variable region frameworks with at least 95% identity to the variable region frameworks of SEQ ID NO:53, and a light chain comprising the 3 CDRs of SEQ ID NO:60, and variable region frameworks with at lest 95% identity to the variable region frameworks of SEQ ID NO:60.
  • variable region frameworks Any or all of the positions backmutated in other exemplified humanized heavy or light chain mature variable regions can also be made (i.e., 1, 2, 3, 4, 5 or all 6, of H28 occupied by N, H48 occupied by I, H66 occupied by K, H67 occupied by A, H71 occupied by A, H93 occupied by T in the heavy chain and 1 or, 2 of L37 occupied by L37 occupied by L, and L45 occupied by K.
  • additional backmutations are not preferred because they in general do not improve affinity and introducing more mouse residues may give increased risk of immunogenicity.
  • CDR residues in the CDRs of the mouse antibody are substitute certain residues in the CDRs of the mouse antibody with corresponding residues from human CDRs sequences, typically from the CDRs of the human acceptor sequences used in designing the exemplified humanized antibodies.
  • SDR residues are needed to retain binding in a humanized antibody.
  • CDR residues not contacting antigen and not in the SDRs can be identified based on previous studies (for example residues H60-H65 in CDR H2 are often not required), from regions of Kabat CDRs lying outside Chothia hypervariable loops (Chothia, J. Mol. Biol.
  • the amino acid occupying the position can be an amino acid occupying the corresponding position (by Kabat numbering) in the acceptor antibody sequence.
  • the number of such substitutions of acceptor for donor amino acids in the CDRs to include reflect a balance of competing considerations.
  • substitutions are potentially advantageous in decreasing the number of mouse amino acids in a humanized antibody and consequently decreasing potential immunogenicity.
  • substitutions can also cause changes of affinity, and significant reductions in affinity are preferably avoided.
  • one or more residues in a CDR of a humanized BR2-22a antibody can be replaced by corresponding residues from a CDR from the mouse BR2-14a antibody (or vice versa). Positions for substitution within CDRs and amino acids to substitute can also be selected empirically.
  • substitutions can be made, for example, in framework residues not in contact with the CDRs, or even some potential CDR-contact residues amino acids within the CDRs.
  • the replacements made in the variant humanized sequences are conservative with respect to the replaced HBLB amino acids (in the case of humanized BR2-14a) or HGLG amino acids (in the case of humanized BR2-22).
  • replacements relative to HBLB or HGLG have no substantial effect on the binding affinity or property of the humanized mAb, that is, its ability to bind human LIV-1 and inhibit growth of cancer cells.
  • Variants typically differ from the heavy and light chain mature variable region sequences of HBLB (hLIV14) or HGLG (hLIV22) by a small number (e.g., typically no more than 1, 2, 3,5 or 10 in either the light chain or heavy chain mature variable region, or both) of replacements, deletions or insertions.
  • a small number e.g., typically no more than 1, 2, 3,5 or 10 in either the light chain or heavy chain mature variable region, or both
  • the heavy and light chain variable regions of humanized antibodies can be linked to at least a portion of a human constant region.
  • the choice of constant region depends, in part, whether antibody-dependence cell-mediated cytotoxicity, antibody dependent cellular phagocytosis and/or complement dependent cytotoxicity are desired.
  • human isotopes IgG1 and IgG3 have strong complement-dependent cytotoxicity
  • human IgG4 lacks complement-dependent cytotoxicity.
  • Human IgG1 and IgG3 also induce stronger cell mediated effector functions than human IgG2 and IgG4.
  • Light chain constant regions can be lambda or kappa.
  • Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab′, F(ab′)2, and Fv, or as single chain antibodies in which heavy and light chain variable domains are linked through a spacer.
  • Isoallotypes differ from allotypes in that sera recognizing an isoallotype binds to a non-polymorphic region of a one or more other isotypes.
  • One or several amino acids at the amino or carboxy terminus of the light and/or heavy chain may be missing or derivatized in a proportion or all of the molecules. Substitutions can be made in the constant regions to reduce or increase effector function such as complement-mediated cytotoxicity or ADCC (see, e.g., Winter et al., U.S. Pat. No. 5,624,821; Tso et al., U.S. Pat. No. 5,834,597; and Lazar et al., Proc. Natl. Acad. Sci. USA 103:4005, 2006), or to prolong half-life in humans (see, e.g., Hinton et al., J. Biol. Chem. 279:6213, 2004).
  • Exemplary substitution include the amino acid substitution of the native amino acid to a cysteine residue is introduced at amino acid positions 234, 235, 237, 239, 267, 298, 299, 326, 330, or 332, preferably an S239C mutation in a human IgG1 isotype (US 200100158909).
  • the presence of an additional cysteine residue allows interchain disulfide bond formation. Such interchain disulfide bond formation can cause steric hindrance, thereby reducing the affinity of the Fc region-Fc ⁇ R binding interaction.
  • the cysteine residue(s) introduced in or in proximity to the Fc region of an IgG constant region can also serve as sites for conjugation to therapeutic agents (i.e., coupling cytotoxic drugs using thiol specific reagents such as maleimide derivatives of drugs.
  • therapeutic agents i.e., coupling cytotoxic drugs using thiol specific reagents such as maleimide derivatives of drugs.
  • the presence of a therapeutic agent causes steric hindrance, thereby further reducing the affinity of the Fc region-Fc ⁇ R binding interaction.
  • Other substitutions at any of positions 234, 235, 236 and/or 237 reduce affinity for Fc ⁇ receptors, particularly Fc ⁇ RI receptor (see, e.g., U.S. Pat. Nos. 6,624,821, 5,624,821.)
  • FcRn is a receptor that is structurally similar to MHC Class 1 antigen that non-covalent associates with ⁇ 2-microglobulin. FcRn regulates the catabolism of IgGs and their transcytosis across tissues (Ghetie and Ward, 2000, Annu. Rev. Immunol. 18:739-766; Ghetie and Ward, 2002, Immunol. Res. 25:97-113).
  • the IgG-FcRn interaction takes place at pH 6.0 (pH of intracellular vesicles) but not at pH 7.4 (pH of blood); this interaction enables IgGs to be recycled back to the circulation (Ghetie and Ward, 2000, Ann. Rev. Immunol. 18:739-766; Ghetie and Ward, 2002, Immunol. Res. 25:97-113).
  • the region on human IgG 1 involved in FcRn binding has been mapped (Shields, et al., 2001, J. Biol. Chem. 276:6591-604).
  • IgG1 Alanine substitutions at positions Pro238, Thr256, Thr307, Gln311, Asp312, Gln380, Gla382, or Asn434 of human IgG1 enhance FcRn binding (Shields et al., 2001, J. Biol. Chem. 276:6591-604). IgG1 molecules harboring these substitutions have longer serum half-lives. Consequently, these modified IgG1 molecules may be able to carry out their effector functions, and hence exert their therapeutic efficacies, over a longer period of time compared to unmodified IgG1.
  • Other exemplary substitutions for increasing binding to FcRn include a Gln at position 250 and/or a Len at position 248. EU numbering is used for all position in the constant region.
  • Oligosaccharides covalently attached to the correspond Asn297 are involved in the ability of the Fc region of an IgG to bind Fc ⁇ R (Lund et al., 1996, J. Immunol. 157:4963-69; Wright and Morrison, 1997, Trends Biotechnol. 15:26-31). Engineering of this glycoform on IgG can significantly improve IgG-mediated ADCC. Addition of bisecting N-acetylglucosamine modifications (Umana et al., 1999, Nat. Biotechnol. 17:176-180, Davies et al., 2001, Biotech. Bioeng.
  • IgG variants with altered Fc ⁇ R binding affinities (Shields et al., 2001, J. Biol. Chem. 276:6591-604).
  • a subset of these variants involving substitutions at Thr256/Ser298, Ser298/Glu333, Ser398/Lys344, or Ser298/Glu333/Lys334 to Ala demonstrate increased in both binding affinity toward Fc ⁇ R and ADCC activity (Shields et al., 2001, J. Biol. Chem. 276:6591-604; Okazaki et al., 2004, J. Mol. Biol. 336:1239-49).
  • Complement fixation activity of antibodies can be improved by substitutions at Lys326 and Glu333 (Idusogie et al., 2001, J. Immunol. 166:2571-2575).
  • the same substitutions on a human IgG2 back bone can convert an antibody isotype that binds poorly to Clq and is severely deficient in complement activation activity to one that can both bond Clq and mediate CDC (Idusogie et al., 2001, J. Immunol. 166:2571-75).
  • Several other methods have also been applied to improve complement fixation activity of antibodies.
  • Complement activity can be reduced by mutating at least one of the amino acid residues 318, 320, and 322 of the heavy chain to a residue having a different side chain, such as Ala.
  • a residue having a different side chain such as Ala.
  • Other alkyl-substituted non-ionic residues such as Gly, Ile, Leu, or Val, or such aromatic non-polar residues as Phe, Tyr, Trp and Pro in place of any one of the three residues also reduce or abolish Clq binding.
  • Ser, Thr, Cys, and Met can be unused at residues 320 and 322, but not 318, to reduce or abolish Clq binding activity.
  • Replacement of the 318 (Glu) residue by a polar residue may modify but not abolish Clq binding activity.
  • Reference to a human constant region includes a constant region with any natural allotype or any permutation of residues occupying polymorphic positions in natural allotypes. Also, up to 1, 2, 5, or 10 mutations may be present relative to a natural human constant region, such as those indicated above to reduce Fcgamma receptor binding or increase binding to FcRN.
  • Humanized antibodies are typically produced by recombinant expression.
  • Recombinant polynucleotide constructs typically include an expression control sequence operably linked to the coding sequences of antibody chains, including naturally-associated of heterologous promoter regions.
  • the expression control sequences are eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and the collection and purification of the crossreacting antibodies.
  • Mammalian cells are a preferred host for expressing nucleotide segments encoding immunoglobulins or fragments thereof. See Winnacker, From Genes to Clones, (VCH Publishers, NY, 1987).
  • a number of suitable host cell lines capable of secreting intact heterologous proteins have been developed in the art, and include CHO cell lines (e.g., DG44), various COS cell lines. HeLa cells, HEK293 cells, L cells, and non-antibody-producing myelomas including Sp2/0 and NS0,
  • the cells are nonhuman.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer (Queen et al., Immunol. Rev.
  • Preferred expression control sequences are promoters derived from endogenous genes, cytomegalovirus, SV40, adenovirus, bovine papillomavirus, and the like. See Co et al., J. Immunol. 148:1149 (1992).
  • antibodies can be purified according to standard procedures of the art, including HPLC purification, column chromatography, gel electrophoresis and the like (see generally, Scopes, Protein Purification (Springer-Verlag, NY, 1982)).
  • the invention further provides nucleic acids encoding any of the humanized heavy and light chains described above.
  • the nucleic acids also encode a signal peptide fused to the mature heavy and light chains.
  • Coding sequences on nucleic acids can be in operable linkage with regulatory sequences to ensure expression of the coding sequences, such as a promoter, enhancer, ribosome binding site, transcription termination signal and the like.
  • the nucleic acids encoding heavy and light chains can occur in isolated form or can be cloned into one or more vectors.
  • the nucleic acids can be synthesized by for example, solid state synthesis or PCR of overlapping oligonucleotides.
  • Nucleic acids encoding heavy and light chains can be joined as one contiguous nucleic acid, e.g., within an expression vector, or can be separate, e.g., each cloned into its own expression vector.
  • Anti-LIV-1 antibodies can be conjugated to cytotoxic or cyostatic moieties (including pharmaceutically compatible salts thereof) to form an antibody drug conjugate (ADC).
  • ADC antibody drug conjugate
  • Particularly suitable moieties for conjugation to antibodies are cytotoxic agents (e.g., chemotherapeutic agents), prodrug converting enzymes, radioactive isotopes or compounds, or toxins (these moieties being collectively referred to as a therapeutic agent).
  • an anti-LIV-1 antibody can be conjugated to a cytotoxic agent such as a chemotherapeutic agent, or a toxin (e.g., a cytostatic or cytocidal agent such as, e.g., abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin).
  • a cytotoxic agent such as a chemotherapeutic agent, or a toxin (e.g., a cytostatic or cytocidal agent such as, e.g., abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin).
  • An anti-LIV-1 antibody can be conjugated to a pro-drug converting enzyme.
  • the pro-drug converting enzyme can be recombinantly fused to the antibody or chemically conjugated thereto using known methods.
  • Exemplary pro-drug converting enzymes are carboxypeptides G2, beta-glucuronidase, penicillin-V-amidase, penicillin-G-amidase, ⁇ -lactamase, ⁇ -glucosidase, nitroreductase and carboxypeptidase A.
  • the therapeutic agent can be conjugated in a manner that reduces its activity unless it is cleaved off the antibody (e.g., by hydrolysis, by antibody degradation or by a cleaving agent).
  • Such therapeutic agent is attached to the antibody with a cleavable linker that is sensitive to cleavage in the intracellular environment of the LIV-1-expressing cancer cell but is not substantially sensitive to the extracellular environment such that the conjugate is cleaved from the antibody when it is internalized by the LIV-1-expressing cancer cell (e.g., in the endosomal or, for example by virtue of pH sensitivity or protease sensitivity, in the lysosomal environment or in the caveolear environment).
  • the ADC comprises a linker region between the therapeutic agent and the anti-LIV-1 antibody.
  • the linker is cleavable under intracellular conditions, such that the cleavage of the linker releases the therapeutic agent from the antibody in the intracellular environment (e.g., within a lysosome or endosome or caveolea).
  • the linker can be, e.g., a peptidyl linker that is cleaved by an intracellular peptidase or protease enzyme, including a lysosomal or endosomal protease.
  • the peptidyl linker is at least two amino acids long or at least three amino acids long.
  • Cleaving agents can include cathepsins B and D and plasmin (see, e.g., Dubowchik and Walker, 1999, Pharm. Therapeutics 83:67-123).
  • Most typical are peptidyl linkers that are cleavable by enzymes that are present in LIV-1-expressing cells.
  • a peptidyl linker that is cleavable by the thiol-dependent protease cathepsin-B, which is highly expressed in cancerous tissue can be used (e.g., a linker comprising a Phe-Leu or a Gly-Phe-Leu-Gly (SEQ ID NO:90) peptide).
  • the peptidyl linker cleavable by an intracellular protease comprises a Val-Cit linker or a Phe-Lys dipeptide (see, e.g., U.S. Pat. No. 6,214,345, which describes the synthesis of doxorubicin with the Val-Cit linker).
  • One advantage of using intracellular proteolytic release of the therapeutic agent is that the agent is typically attenuated when conjugated and the serum stabilities of the conjugates are typically high.
  • the cleavable linker can e pH-sensitive, i.e., sensitive to hydrolysis at certain pH values.
  • the pH-sensitive linker is hydrolyzable under acidic conditions.
  • an acid-labile linker that is hydrolyzable in the lysosome e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide, orthoester, acetal, ketal, or the like
  • an acid-labile linker that is hydrolyzable in the lysosome (e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide, orthoester, acetal, ketal, or the like) can be used.
  • U.S. Pat. Nos. 5,122,368; 5,824,805; 5,622,929, Dubowchik and Walker 1999, Pharm.
  • the hydrolyzable linker is a thioether linker (such as, e.g., a thioether attached to the therapeutic agent via an acylhydrazone bond (see, e.g., U.S. Pat. No. 5,622,929)).
  • Disulfide linkers include those that can be formed using SATA (N-succinimidyl-S-acetylthioacetate), SPDP (N-succinimidyl-3-(2-pyridyldithio)propionate), SPDB (N-succinimidyl-3-(2-pyridyldithio)butyrate) and SMPT (N-succinimidyl-oxycarbonyl-alpha-methyl-alpha-(2-pyridyl-dithio)toluene), SPDB and SMPT.
  • SATA N-succinimidyl-S-acetylthioacetate
  • SPDP N-succinimidyl-3-(2-pyridyldithio)propionate
  • SPDB N-succinimidyl-3-(2-pyridyldithio)butyrate
  • SMPT N-succinimidyl-oxycarbonyl
  • the linker can also be a malonate linker (Johnson et al., 1995, Anticancer Res. 15:1387-93), a maleimidobenzoyl linker (Lau et al., 1995, Bioorg-Med-Chem. 3(10):1299-1304), or a 3′-N-amide analog (Lau et al., 1995, Bioorg-Med-Chem. 3(10):1305-12).
  • the linker also can be a non-cleavable linker, such as an maleimido-alkylene- or maleimide-aryl linker that is directly attached to the therapeutic agent (e.g., a drug).
  • the therapeutic agent e.g., a drug
  • An active drug-linker is released by degradation of the antibody.
  • the linker is not substantially sensitive to the extracellular environment meaning that no more than about 20%, typically no more than about 15%, more typically no more than about 10%, and even more typically no more than about 5%, no more than about 3%, or no more than about 1% of the linkers in a sample of the ADC is cleaved when the ADC present in an extracellular environment (e.g., in plasma).
  • Whether a linker is not substantially sensitive to the extracellular environment can be determined, for example, by including independently with plasma both (a) the ADC (the “ADC sample”) and (B) an equal molar amount of unconjugated antibody or therapeutic agent (the “control sample”) for a predetermined time period (e.g., 2, 4, 8, 16, or 24 hours) and then comparing the amount of unconjugated antibody or therapeutic agent present in the ADC sample with that present in control sample, as measured, for example, by high performance liquid chromatography.
  • a predetermined time period e.g., 2, 4, 8, 16, or 24 hours
  • This linker can also promote cellular internalization.
  • the linker can promote cellular internalization when conjugated to the therapeutic agent (i.e., in the milieu of the linker-therapeutic agent moiety of the ADC or ADC derivative as described herein).
  • the linker can promote cellular internalization when conjugated to both the therapeutic agent and the anti-LIV-1 antibody (i.e., in the milieu of the ADC as described herein).
  • linkers that can be used with the present compositions are described in WO 2004-010957 and have the form -A a -W w —Y y — (1)
  • Representative stretcher units are depicted within the square brackets of Formulas (Ia) and (Ib: see infra), wherein A-, —W—, —Y—, -D, w and y are as defined above and R 1 is selected from —C 1 -C 10 alkylene-, —C 3 -C 8 carbocyclo-, —O—(C 1 -C 8 alkyl)-, -arylene-, —C 1 -C 10 alkylene-arylene-, -arylene-C 1 -C 10 alkylene-, —C 1 -C 10 alkylene-(C 3 -C 8 carbocyclo)-, —(C 3 -C 8 carbocyclo)-C 8 -C 10 alkylene, —C 3 -C 8 heterocyclo-, —C 1 -C 10 alkylene-(C 3 -C 8 heterocyclo)-, —(C 3 -C 8 heterocyclo)-C 1 -
  • the drug loading is represented by p, the number of drug-linker molecules per antibody.
  • p can represent the average number of drug-linker molecules per antibody, also referred to the average drug loading, P ranges from 1 to 20 and is preferably from 1 to 8. In some preferred embodiments, when p represents the average drug loading, p ranges from about 2 to about 5. In some embodiments, p is about 2, about 3, about 4, or about 5.
  • the average number of drugs per antibody in a preparation may be characterized by conventional means such as mass spectroscopy, ELISA assay, and HPLC.
  • —W w — is preferably a dipeptide, tripeptide, tetrapeptide, pentapeptide, hexapeptide, heptapeptide, octapeptide, nonapeptide, decapeptide, undecapeptide or dodecapeptide unit.
  • the Spacer unit when present, links an Amino Acid unit to the Drug unit.
  • Spacer units are of two general types: self-immolative and non self-immolative.
  • a non self-immolative spacer unit is one in which part or all of the Spacer unit remains bound to the Drug unit after enzymatic cleavage of an amino acid unit from the anti-LIV-1 antibody-linker-drug conjugate or the drug-linker compound.
  • Examples of a non self-immolative Spacer unit include a (glycine-glycine) spacer unit and a glycine spacer unit.
  • an anti-LIV-1 antibody-linker-drug conjugate containing a glycine-glycine spacer unit or a glycine spacer unit undergoes enzymatic cleavage via a tumor-cell associated-protease, a cancer-cell-associated protease or a lymphocyte-associated protease, a glycine-glycine-drug moiety or a glycine-drug moiety is cleaved from Ab-A a -W w ,—.
  • an independent hydrolysis reaction should take place within the target cell to cleave the glycine-drug unit bond.
  • an anti-LIV-1 antibody drug conjugate containing a self-immolative spacer unit can release the drug (D) without the need for a separate hydrolysis step.
  • —Y— is a p-aminobenzyl alcohol (PAB) unit that is linked to —W w — via the nitrogen atom of the PAB group, and connected directly to -D via a carbonate, carbamate or ether group.
  • PAB p-aminobenzyl alcohol
  • Other examples of self-immolative spacers include aromatic compounds that are electronically equivalent to the PAB group such as 2-aminoimidazol-5-methanol derivatives (see Hay et al., 1999, Bioorg. Med. Chem. Lett.
  • Spacers can be used that undergo facile cyclization upon amide bond hydrolysis, such as substituted and unsubstituted 4-aminobutyric acid amides (Rodrigues et al., 1995, Chemistry Biology 2:223), appropriately substituted bicyclo[2.2.1] and bicyclo[2.2.2] ring systems (Storm et al., 1972, J. Amer. Chem. Soc. 94:5815) and 2-aminophenylpropionic acid amides (Amsberry et al., 1990, J. Org. Chem. 55:5867).
  • the spacer unit is a branched bis(hydroxymethyl)styrene (BHMS) unit, which can be used to incorporate additional drugs.
  • BHMS branched bis(hydroxymethyl)styrene
  • cytotoxic agents to conjugate to anti-LIV-1 antibodies include, for example, antitubulin agents. DNA minor groove binding agents, DNA replication inhibitors, chemotherapy sensitizers, or the like.
  • Other exemplary classes of cytotoxic agents include anthracyclines, auristatins, camptothecins, duocarmycins, etoposides, maytansinoids and vinca alkaloids.
  • cytotoxic agents include auristatins (e.g., auristatin E, AFP, MMAF, MMAE), DNA minor groove binders (e.g., enediynes and lexitropsins), duocarmycins, taxanes (e.g., paclitaxel and docetaxel), vinca alkaloids, doxorubicin, morpholino-doxorubicin and cyano-morpholino-doxorubicin.
  • auristatins e.g., auristatin E, AFP, MMAF, MMAE
  • DNA minor groove binders e.g., enediynes and lexitropsins
  • duocarmycins e.g., paclitaxel and docetaxel
  • vinca alkaloids doxorubicin, morpholino-doxorubicin and cyano-morpholino-doxorubicin.
  • the cytotoxic agent can be a chemotherapeutic such as, for example, doxorubicin, paclitaxel, melphalan, vinca alkaloids, methotrexate, mitomycin C or etoposide.
  • the agent can also be a CC-1065 analogue, calicheamicin, maytansine, an analogue of dolastatin 10, rhizoxin, or palytoxin.
  • the cytotoxic agent can also be an auristatin.
  • the auristatin can be an auristatin E derivative is, e.g., an ester formed between auristatin E and a keto acid.
  • auristatin E can be reacted with paraacetyl benzoic acid or benzoylvaleric acid to produce AFB and AEVB, respectively.
  • Other typical auristatins include AFP, MMAF, and MMAE.
  • the synthesis and structure of various auristatins are described in, for example, US 2005-0238649 and US2006-0074008.
  • the cytotoxic agent can be a DNA minor groove binding agent.
  • the minor groove binding agent can be a CBI compound or an enediyne (e.g., calicheamicin).
  • the cytotoxic or cytostatic agent can be an anti-rubulin agent.
  • anti-tubulin agents include taxanes (e.g., Taxol® (paclitaxel), Taxotere® (docetaxel)), T67 (Tularik), vinca alkyloids (e.g., vincristine, vinblastine, vindesine, and vinorelbine), and auristatins (e.g., auristatin E, AFP, MMAF, MMAE, AEB, AEVB).
  • Taxol® paclitaxel
  • Taxotere® docetaxel
  • T67 Tularik
  • vinca alkyloids e.g., vincristine, vinblastine, vindesine, and vinorelbine
  • auristatins e.g., auristatin E, AFP, MMAF, MMAE, AEB, AEVB.
  • antitubulin agents include, for example, baccatin derivatives, taxane analogs (e.g., epothilone A and B), nocodazole, colchicine and colcimid, estramustine, cryptophysins, cemadotin, maytansinoids, combretastatins, discodermolide, and elemtherobin.
  • the cytotoxic agent can be a maytanisinoid, another group of anti-tubulin agents.
  • the maytansinoid can be maytansine or a maytansine containing drug linker such as DM-1 or DM-4 (ImmunoGen, Inc.; see also Chari et al., 1992. Cancer Res. 52:127-131).
  • Exemplary antibody drug conjugates include vcMMAE and mcMMAF antibody drug conjugates as follows wherein p and Ab are as previously described herein:
  • LIV-1 binding to an extracellular domain of LIV-1 can be used in some of the methods of the invention, particularly the treatment of triple negative breast cancers.
  • a collection of mouse antibodies to LIV-1 is described in US20080175839.
  • These antibodies include 1.1F10, 1.7A4, BR2-10b, BR2-11a, BR2-13a, BR2-14a, BR2-15a, BR2-16a, BR2-17a, BR2-18a, BR2-19a, BR2-20a, BR2-21a, BR2-22a, BR2-23a, BR2-24a, and BR2-25a, of which BR2-19a produced by the hybridoma ATCC Accession No. PTA-5706 or BR2-23a produced by the hybridoma ATCC Accession No. PTA-5707 in addition to BR2-14a and BR2-22a are preferred.
  • Humanized, chimeric or veneered forms of these antibodies can be made by conventional methods summarized below.
  • LIV-1 antibodies to LIV-1 can be made de novo by immunizing with LIV-1 or one or more extracellular domains thereof.
  • the production of other non-human monoclonal antibodies, e.g., murine, guinea pig, primate, rabbit or rat, against an immunogen can be performed by as described by Harlow & Lane, Antibodies, A Laboratory Manual (CSHP NY, 1988) (incorporated by reference for all purposes).
  • Such an immunogen can be obtained from a natural source, by peptide synthesis or by recombinant expression.
  • a chimeric antibody is an antibody in which the mature variable regions of light and heavy chains of a non-human antibody (e.g., a mouse) are combined with human light and heavy chain constant regions. Such antibodies substantially or entirely retain the binding specificity of the mouse antibody, and are about two-thirds human sequence.
  • a veneered antibody is a type of humanized antibody that retains some and usually all of the CDRs and some of the non-human variable region framework residues of a non-human antibody but replaces other variable region framework residues that may contribute to B- or T-cell epitopes, for example exposed residues (Padlan, Mol. Immunol. 28:489, 1991) with residues from the corresponding positions of a human antibody sequence.
  • the result is an antibody in which the CDRs are entirely or substantially from a non-human antibody and the variable region frameworks of the non-human antibody are made more human-like by the substitutions.
  • Human antibodies against LIV-1 can be provided by a variety of techniques described below. Methods for producing human antibodies include the trioma method of Oestberg et al., Hybridoma 2:361-367 (1983); Oestberg, U.S. Pat. No. 4,634,664; and Engleman et al., U.S. Pat. No. 4,634,666, use of transgenic mice including human immunoglobulin genes (see, e.g., Lonberg et al., WO93/12227 (1993); U.S. Pat. Nos.
  • any of the antibodies can be selected to have the same or overlapping epitope specificity as an exemplary antibody, such as the BR2-14a antibody, by a competitive binding assay or otherwise.
  • the humanized antibodies of the invention can be used to treat cancer.
  • Some such cancers show detectable levels of LIV-1measured at either the protein (e.g., by immunoassay using one of the exemplified antibodies ) or mRNA level.
  • Some such cancers show elevated levels of LIV-1 relative to noncancerous tissue of the same type, preferably from the same patient.
  • An exemplary level of LIV-1 on cancer cells amenable to treatments is 5000-150000 LIV-1 molecules per cell, although higher or lower levels can be treated.
  • a level of LIV-1 in a cancer is measured before performing treatment.
  • cancers associated with LIV-1 expression and amenable to treatment include breast cancer, prostate cancer, ovarian cancer, endometrial cancer, cervical, liver, gastric, kidney, and squamous cell carcinomas (e.g., bladder, head, neck and lung), skin cancers, e.g., melanoma, small lung cell carcinoma or lung carcinoid.
  • the treatment can be applied to patients having primary or metastatic tumors of these kinds.
  • the treatment can also be applied to patients who are refractory to conventional treatments (e.g., hormones, tamoxifen, herceptin), or who have replaced following a response to such treatments.
  • the methods can also be used on triple negative breast cancers.
  • a triple negative breast cancer is a term of art for a cancer lacking detectable estrogen and progesterone receptors and lacking overexpression of HER2/neu when stained with an antibody to any of these receptors, such as described in the examples. Staining can be performed relative to an irrelevant control antibody and lack of expression shown from a background level of straining the same or similar to that of the control within experimental error. Likewise lack of overexpression is shown by staining at the same or similar level within experimental error of noncancerous breast tissue, preferably obtained from the same patient.
  • triple native breast cancers are characterized by lack of responsiveness to hormones interacting with these receptors, aggressive behavior and a distinct pattern of metastasis.
  • hLIV14 antibodies can be used to treat cancers that express LIV-1.
  • an hLIV14 antibody is used treat a subject with a LIV-1-expressing breast cancer.
  • an hLIV14 antibody is used treat a subject with a LIV-1-expressing prostate cancer.
  • an hLIV14 antibody is used treat a subject with a LIV-1-expressing melanoma.
  • an hLIV14 antibody is used treat a subject with a LIV-1-expressing ovarian cancer.
  • an hLIV14 antibody is used treat a subject with a LIV-1-expressing endometrial cancer.
  • an hLIV14 antibody is used treat a subject with a LIV-1-expressing cervical cancer. In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing liver cancer. In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing gastric cancer. In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing kidney cancer. In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing squamous cell carcinomas (e.g., bladder, head, neck and lung cancer). In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing breast cancer.
  • a LIV-1-expressing cervical cancer In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing liver cancer. In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing gastric cancer. In
  • an hLIV14 antibody is used treat a subject with a LIV-1-expressing skin cancer. In another embodiment, an hLIV14 antibody is used treat a subject with a LIV-1-expressing small lung cell carcinoma or lung carcinoid.
  • hLIV22 antibodies can be used to treat cancers that express LIV-1. In one embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing breast cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing prostate cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing melanoma.
  • an hLIV22 antibody is used treat a subject with a LIV-1-expressing ovarian cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing endometrial cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing cervical cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing liver cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing gastric cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing kidney cancer.
  • an hLIV22 antibody is used treat a subject with a LIV-1-expressing squamous cell carcinomas (e.g., bladder, head, neck, and lung cancer). In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing breast cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing skin cancer. In another embodiment, an hLIV22 antibody is used treat a subject with a LIV-1-expressing small lung cell carcinoma or lung carcinoid. This application provides the first disclosure that LIV-1 protein is expressed on the surface of the melanoma cells.
  • antibodies that bind to LIV-1 can be used to treat patients that are afflicted with melanomas that express LIV-1.
  • Such antibodies include antibodies disclosed herein, e.g., hLIV14 and hLIV22, but are not limited to the antibodies disclosed herein.
  • Humanized antibodies are administered in an effective regime meaning a dosage, route of administration and frequency of administration that delays the onset, reduces the severity, inhibits further deterioration, and/or ameliorates at least one sign or symptom of cancer.
  • an effective regime meaning a dosage, route of administration and frequency of administration that delays the onset, reduces the severity, inhibits further deterioration, and/or ameliorates at least one sign or symptom of cancer.
  • the regime can be referred to as a therapeutically effective regime.
  • the patient is at elevated risk of the cancer relative to the general population but is not yet experiencing symptoms, the regime can be referred to as a prophylactically effective regime.
  • therapeutic or prophylactic efficiency can be observed in an individual patient relative to historical controls or past experiences in the same patient.
  • therapeutic or prophylactic efficacy can be demonstrated in a preclinical or clinical trial in a population of treated patients relative to a control population of untreated patients.
  • Exemplary dosages for a monoclonal antibody are 0.1 mg/kg to 50 mg/kg of the patient's body weight, more typically 1 mg/kg to 30 mg/kg, 1 mg/kg to 20 mg/kg, 1 mg/kg to 15 mg/kg, 1 mg/kg to 12 mg/kg, or 1 mg/kg to 10 mg/kgl, or 2 mg/kg to 30 mg/kg, 2 mg/kg to 20 mg/kg, 2 mg/kg to 15 mg/kg, 2 mg/kg to 12 mg/kg, or 2 mg/kg to 10 mg/kg, or 3 mg/kg to 30 mg/kg, 3 mg/kg to 20 mg/kg, 3 mg/kg to 15 mg/kg, 3 mg/kg to 12 mg/kg, or 3 mg/kg to 10 mg/kb.
  • Exemplary dosages for a monoclonal antibody or antibody drug conjugates thereof are 1 mg/kg to 7.5 mg/kg, or 2 mg/kg to 7.5 mg/kg or 3 mg/kg to 7.5 mg/kg of the subject's body weight, or 0.1-20, or 0.5 mg/kg body weight (e.g., 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mg/kg) or 10-1500 or 200-1500 mg as a fixed dosage.
  • the patient is administered a dose of at least 1.5 mg/kg, at least 2 mg/kg or at least 3 mg/kb, administered once every three weeks or greater.
  • the dosage depends on the frequency of administration, condition of the patient and response to prior treatment, if any, whether the treatment is prophylactic or therapeutic and whether the disorder is acute or chronic, among other factors.
  • Administration can be parenteral, intravenous, oral, subcutaneous, intra-arterial, intracranial, intrathecal, intraperitoneal, topical, intranasal or intramuscular. Administration can also be localized directly into a tumor. Administration into the systemic circulation by intravenous or subcutaneous administration is preferred. Intravenous administration can be, for example, by infusion over a period such as 30-90 min or by a single bolus injection.
  • the frequency of administration depends on the half-life of the antibody or conjugate in the circulation, the condition of the patient and the route of administration among other factors.
  • the frequency can be daily, weekly, monthly, quarterly, or at irregular intervals in response to changes in the patient's condition or progression of the cancer being treated.
  • An exemplary frequency for intravenous administration is between twice a week and quarterly over a continuous course of treatment, although more or less frequent dosing is also possible.
  • Other exemplary frequencies for intravenous administration are between weekly or three out of every four weeks over a continuous course of treatment, although more or less frequency dosing is also possible.
  • an exemplary dosing frequency is daily to monthly, although more or less frequent dosing is also possible.
  • the number of dosages administered depends on the nature of the cancer (e.g., whether presenting acute or chronic symptoms) and the response of the disorder to the treatment.
  • acute disorders or acute exacerbations of a chronic disorder between 1 and 10 doses are often sufficient.
  • a single bolus dose, optionally in divided form, is sufficient for an acute disorder or acute exacerbation of a chronic disorder.
  • Treatment can be repeated for recurrence of an acute disorder or acute exacerbation.
  • an antibody can be administered at regular intervals, e.g., weekly, fortnightly, monthly, quarterly, every six months for at least 1, 5 or 10 years, or the life of the patient.
  • compositions for parenteral administration are preferably sterile and substantially isotonic and manufactured under GMP conditions.
  • Pharmaceutical composition can be provided in unit dosage form (i.e., the dosage for a single administration).
  • Pharmaceutical compositions can be formulated using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries. The formulation depends on the route of administration chosen.
  • antibodies can be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution. Ringer's solution, or physiological saline or acetate buffer (to reduce discomfort at the site of injection).
  • the solution can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • antibodies can be in lyophilized form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • concentration of antibody in a liquid formulation can be e.g., 1-100 mg/mL such as 10 mg/ml.
  • Treatment with antibodies of the invention can be combined with chemotherapy, radiation, stem cell treatment, surgery other treatments effective against the disorder being treated.
  • Useful classes of other agents that can be administered with humanized antibodies to LIV-1 include, for example, antibodies to other receptors expressed on cancerous cells, antitubulin agents (e.g., auristatins), DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cis-platin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antibiotics, antifolates, antimetabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, pre-forming compounds, purine antimetabolites, puromycins, radiation sensitizers, steroids, taxanes, topoi
  • Treatment with the humanized anti-LIV-1 antibody can increase the median progression-free survival or overall survival time of patients with tumors (e.g., breast, prostate, melanoma), especially when relapsed or refractory, by at least 30% or 40% but preferably 50%, 60% to 70% or even 100% or longer, compared to the same treatment (e.g., chemotherapy) but without an anti-LIV-1 antibody alone or as a conjugate.
  • tumors e.g., breast, prostate, melanoma
  • Treatment with the humanized anti-LIV-1 antibody can increase the median progression-free survival or overall survival time of patients with tumors (e.g., breast, prostate, melanoma), especially when relapsed or refractory, by at least 30% or 40% but preferably 50%, 60% to 70% or even 100% or longer, compared to the same treatment (e.g., chemotherapy) but without an anti-LIV-1 antibody alone or as a conjugate.
  • treatment e.g., standard chemotherapy
  • treatment including the anti-LIV-1 antibody alone or as a conjugate
  • treatment can increase the complete response rate, partial response rate, or objective response rate (complete+partial) of patients with tumors by at least 30% or 40% but preferably 50%, 60% to 70% or even 100% compared to the same treatment (e.g., chemotherapy) but without the anti-LIV-1 antibody.
  • the complete and partial response rates are determined by objective criteria commonly used in clinical trials for cancer, e.g., as listed or accepted by the National Cancer Institute and/or Food and Drug Administration.
  • the anti-LIV-1 humanized antibodies can be used for detecting LIV-1 in the context of clinical diagnosis or treatment or in research. Expression of LIV-1 on a cancer provides an indication that the cancer is amenable to treatment with the antibodies of the present invention.
  • the antibodies can also be sold as research reagents for laboratory research in detecting cells bearing LIV-1 and their response to various stimuli. In such uses, monoclonal antibodies can be labeled with fluorescent molecules, spin-labeled molecules, enzymes or radioisotypes, and can be provided in the form of kit with all the necessary reagents to perform the assay for LIV-1.
  • the antibodies described herein, BR2-14a, BR2-22a and humanized versions thereof, e.g., hLIV14 and hLIV22, can be used to detect LIV-1 protein expression and determine whether a cancer is amenable to treatment with LIV-1 ADCs.
  • BR2-14a, BR2-22a and humanized versions thereof, e.g., hLIV14 and hLIV 22 can be used to detect LIV-1 expression on breast cancer cells, melanoma cells, cervical cancer cells, or prostate cancer cells.
  • the antibodies can also be used to purify LIV-1, e.g., by affinity chromatography.
  • the invention further provides an amino acid sequence for LIV-1 (CY LIV-1) from cynomolgus monkeys at SEQ ID NO:85 with or without a signal peptide, which occupies approximately residues 1-28 of SEQ ID NO:85, as well as nucleic acids that encode that amino acid sequences. Variants differing by up to 1, 2, 3, 4, or 5 substitutions, deletions or insertions are also included provided CY variants do not include a natural human LIV-1 sequence. Analogous to human LIV-1, reference to CY-LIV-1 means at least one extracellular domain of the protein and usually the complete protein other than a cleavable signal peptide (amino acids 1-28).
  • the invention further provides antibodies that specifically bind to SEQ ID NO:85 with or without specifically binding to human LIV-1 (i.e., binding to human LIV-1 at level of negative control irrelevant antibody).
  • the invention further provides antibodies that preferentially bind CY-LIV-1 over human LIV-1 and vice versa. Preferential binding means an association higher beyond experimental error and preferably at least 2, 3 or 4 fold higher.
  • the invention further provides antibodies that show the same binding profile to human and CY-LIV-1 within experimental error as any of the exemplified antibodies described below.
  • the invention further provides methods of analyzing binding of an antibody to CY LIV-1. Such methods involve contacting an antibody with CY LIV-1, determining whether the antibody specifically binds to CY LIV-1 and optionally determining a measure of binding strength, such as an association constant.
  • 1 ⁇ 10 5 antigen expressing cells (either MCF7 cells (ATCC) expressing human LIV-1, a transfected CHO cell line expressing human LIV-1 or a transferred CHO cell line expressing cyno LIV-1) were aliquoted per well of a 96-well v-bottom plates.
  • AlexaFluor-647 labeled murine LIV-1 mAb e.g., BR2-14a
  • Cells were pelleted and washed 3 ⁇ with PBS/BSA. The cells were the pelleted and resuspended in 125 ⁇ L of PBS/BSA. Fluorescence was analyzed by flow cytometry, using percent of saturated fluorescent signal to determine percent bound and to subsequently calculate apparent Kd.
  • 1 ⁇ 10 5 MCF7 cells expressing LIV-1 in PBS/BSA were aliquoted in each well of a 96-well v-bottom plates on ice. The cells were incubated for 1 hour with 5 nM AlexaFluor-647 labeled murine LIV-1 mAb and increasing concentrations (from 0.38 nM to 600 nM) of unlabeled humanized LIV-1 mAb, combinations of humanized light chains LA-LF and humanized heavy chains HA-HE. Cells were pelleted and washed 3 times with PBS. The cells were pelleted and resuspended in 125 ⁇ L of PBS/BSA.
  • CHO cells expressing recombinant cyno LIV-1 in PBS were aliquoted in each well of a 96-well v-bottom plates on ice. The cells were incubated for 1 hour with 5 nM AlexaFluor-647 labeled murine LIV-1 mAb and increasing concentrations (from 0.038 nM to 600 nM) of unlabeled humanized LIV-1 mAb, combinations of humanized light chains LA-LF and humanized heavy chains HA-HE. Cells were pelleted and washed 3 times with PBS. The cells were pelleted and resuspended in 125 ⁇ L of PBS/BSA.
  • LIV-1 copy number on the cell surfaces was determined using murine LIV-1 mAb as primary antibody and the DAKO QiFiKit flow cytometric indirect assay as described by the manufacturer (DAKO A/S, Glostrup, Denmark) and evaluated with a Becton Dickinson FACS®can flow cytometer.
  • Tumor cells were incubated with LIV-1 antibody drug conjugates for 96-144 hours at 37° C.
  • a non-binding (H00) ADC was used as a negative control.
  • Antibody drug conjugates of the LIV-1 antibodies were prepared as described in US20050238649.
  • the drug linkers vcMMAE (also referred to as 1006) and mcMMAF (referred to as 1269) are both described in US20050238649.
  • Preparation of cysteine mutants of IgG1 antibodies is generally described in US20100158919. US20050238649 and US20100158919 are herein incorporated by reference for all purposes.
  • IGF insulin like growth factor
  • SGD-2084 2-fluorofucose peracetate
  • Conditioned media was collected on day 13 by passing the culture through 0.2 ⁇ m filter.
  • Antibody purification was performed by applying the conditioned media to a protein A column pre-equilibrated with 1 ⁇ phosphate buffered saline (PBS), pH 7.4.
  • PBS phosphate buffered saline
  • ADCC activity was measured using the standard 31 Cr-release assay. Briefly, the MCP-7 target tumor cells were labeled with 100 ⁇ Ci Na 51 CrO 4 , washed, and pre-incubated with test antibodies prior to addition of effector (natural killer, NK) cells.
  • NK (Cd16 + CD56 + ) cells were prepared from non-adherent peripheral blood mononuclear cells (PBMCs) obtained from normal Fc ⁇ RIIIA 158V/V donors (Lifeblood, Memphis, Tenn.) with immunomagnetic beads (EasyStep, StemCell Technologies, Vancouver, BC, Canada). Viable NK cells were added to target cells at an effector to target cell ratio of 10:1.
  • a human IgG1 ⁇ (Ancell, Bayport, Minn.) was used as negative control in this assay. After 4 hours of incubation, supernatants were collected and dried overnight on Luma plates. Gamma radiation emitted from lysed MCF-7 cells was then detected using the TopCount Microplate Scintillation and Luminescence Counter (Perkin Elmer, Waltham, Mass.). ADCC activity is reported as % specific lysis.
  • mice 7-8 animals/group were implanted with tumor cells grown in culture.
  • MCF-7 from NCl (5 ⁇ 10 6 cells in 25% matrigel), PC3 from ATCC (2.5 ⁇ 10 6 cells in 25% matrigel), and PC3 from DSMZ (5 ⁇ 10 5 in 25% matrigel).
  • MCF-7 Female mice also received estrogen supplementation by implanting a slow-release estrogen pellet (90 day release).
  • Tumor volumes were monitored using calipers and animals were euthanized when tumor volume reached ⁇ 800 mm 3 Median tumor volume plots were continued for each group until one or more animals were euthanized. All animal procedures were performed under a protocol approved by the Institutional Animal Care and Use Committee in a facility accredited by the Association for Assessment and Accreditation of Laboratory Animal Care.
  • Tissue microarrays and individual tumor samples were obtained from commercial sources.
  • Tissue microarrays from normal or tumor formalin fixed and paraffin embedded (FFPE) tissues were purchased either from US Biomax Inc. or Cybrdi.
  • FFPE paraffin embedded
  • a frozen array was purchased from BioChain.
  • Single sections were purchased from NDRI, Asterand, Tissue Solution or CHTN.
  • a set of 25 paraffin-embedded samples of metastatic hormone refractory prostate cancer was provided by Dr. R. Vessella, University of Washington, Genitourinary Cancer Department. All samples were processed on Bond-MaxTM auto-stainer (Leica).
  • FPPE slides or TMAs sectioned on glass slides were deparaffinized using BondTM Dewax solution (Leica, cat #AR9222) at 72° C. and rehydrated.
  • Antigen retrieval was performed using EDTA based BondTM Epitope Retrieval Solution 2 (Leica, cat #AR9640) for 20 min at 95-100° C. before incubation with the primary murine LIV-1 mAb (1-2 ⁇ g/ml for 30-45 minutes at 25° C.).
  • Isotype-matched murine IgG1 (Sigma; cat #M5284) was used as negative control for background staining.
  • the K D for the murine LIV-1 monoclonal antibody BR2-14a antibody (US2004141983) was determined for human LIV-1 expressed as an endogenous protein in a human breast cancer cell line or as a recombinant protein in a CHO cell line.
  • the K D for the murine LIV-1 antibody BR2-14a was also determined for cyano LIV-1 expressed as a recombinant protein in a CHO cell line.
  • MCF7 is a human breast cancer cell line.
  • 293F is a human embryonic kidney cell line.
  • Table 1 shows that the antibody had about 5-fold lower dissociation constant for non-recombinant LIV-1 expressed from a human cell line than recombinant LV-1, wherein human (hLIV-1) or from cynomolgus monkeys (cyLIV-1).
  • the starting point or donor antibody for humanization in this Example is the mouse antibody BR2-14a produced by the hybridoma having ATCC Accession No. PTA-5705A and descried in US2004141983.
  • Suitable human acceptor sequences are genomic sequences provided by VH1-02 and JH5 for the heavy chain and by VK2-30 and Jk4 for the light chain.
  • the human acceptor sequences show 68 and 85 percentage identity to the donor sequences in the variable region frameworks.
  • the light chain CDRs of the human acceptor sequences are of the same canonical type as the CDRs of the donor sequences.
  • the heavy chain CDRs of the human acceptor sequences differed in their canonical type (the germline was 1-3 versus 1-2 for the murine donor).
  • Alignment of the donor sequences identified eleven positions in the heavy chain (H27, H28, H29, H30, H48, H66, H67, H71, H76, H93 and H94) and five positions in the light chain (L36, L37, L45, L46 and L39) at which the human acceptor sequence differed from the donor sequence and that may affect antibody binding as a result of contacting antigen directly, affecting conformation of CDRs or affecting packing between heavy and light chains.
  • Five humanized heavy chains and six humanized light chains were made incorporating back mutations at different permutations of these positions ( FIG. 1 (sequence alignment) and Table 2).
  • Humanized antibodies were then expressed representing every permutation of these chains (30 possibilities) of the humanized heavy and light chains.
  • the binding curves for recombinant human LIV-1 expressed from CHO cells are shown in FIG. 2 .
  • the EC50's are summarized in the Table 3 below.
  • the HBLB antibody was selected as the best of the humanized antibodies because it has (together with HELE) the strongest binding but has fewer backmutations versus HELE, there being four back mutations in HBLB and twelve in HELE.
  • the EC50s for the humanized LIV-1 mAb which bound human LIV-1 expressed on CHO cells were determined for human LIV-1 expressed as a native protein in an MCF7 cell line ( FIG. 3 ). Again, LIV-1 mAb HBLB and HELE were determined to be the tightest binders.
  • the Kd for HBLB to human LIV-1 on the MCF7 cell line was determined from the average of several saturation binding curves as 1.5 nM whereas that for the mouse antibody is 2.9 nM.
  • the HBLB antibody has about twice the affinity for native human LIV-1 as the mouse antibody.
  • the saturation binding curve shown in FIG. 4 is a representative example.
  • HBLB S239C The binding curves and EC50's of these antibodies compared with the mouse donor antibody are shown in FIG. 5 .
  • the EC50's of both forms of HBLB were similar to one another (within the error of the study), and both were stronger than the mouse antibody.
  • the EC50s for the humanized LIV-1 mAb HBLB and HBLB S239C were also determined for cyno LIV-1 expressed as a recombinant protein in a CHO cell line. Both antibodies bound with equal affinity (better than murine LIV-1 mAb).
  • Murine LIV-1 mAbs (at least 2 for concordance) were used for immunohistochemical analysis of various tumor types using formalin-fixed paraffin embedded tissues.
  • FIGS. 6 and 7 show a high level of LIV-1 expression in post-hormone (tamoxifen or aromatase inhibitors) treated breast and prostate tumors providing a strong rationale to target these tumors using a LIV-1 ADC.
  • FIG. 8 shows detectable LIV-1 expression in triple negative (ER-, PgR-, Her2-) breast cancer tissues.
  • the LIV-1 level of expression in triple negative breast cancer by immunohistochemistry staining was comparable to the level in the PC3 animal model, where we demonstrated anti-tumor activity of LIV-1 ADC.
  • Triple negative breast cancers were therefore a potential target population, particularly triple negative breast cancers which have been found to express LIV-1.
  • various hLIV-14 ADCs (the HBLB antibody conjugated with vcMMAE (referred to as 1006) or mcMMAF (referred to as 1269) (both small molecules and/or linkers described in US20050238649)) were highly effective in killing MCF-7 cells, as compared with the nonbinding and murine control conjugates (mIgG-1006, mIgG-1269, hIgG-1006, and hIgG-1269).
  • cysteine mutant LIV-14d ADCs having an average of two drug linkers per antibody were also highly effective in killing MCF-7 cells as measured by the cytotoxic assay. Referring to FIGS.
  • an effector function enhanced LIV-1 ADC (indicated as SEA) also had a similar level of cytotoxic activity as wildtype (non-fucosylated) ADCs (compare hLIV-1 SEA 1006 (vcMMAE) with hLIV-1 1006 (vcMMAE)).
  • SEA effector function enhanced LIV-1 ADC
  • LIV-1 ADCs chimeric and humanized (HBLB) mAbs with an average of 4 drugs per antibody
  • FIGS. 12-15 LIV-1 ADCs conjugated to vcMMAE showed significant tumor delay compared to untreated and control ADCs. At least one complete regression (CR) was observed in all the studies using LIV-1-vcMMAE at 3 mg/kg with a number of animals having tumors that were static or grew slowly compared to controls.
  • a chimeric form of the parental murine antibody conjugated to vcMMAE resulted in complete regressions in 3 out of 7 mice.
  • a humanized ADC (HBLB) conjugated to vcMMAE (hLIV-14-mvMMAE)(4)
  • HBLB humanized ADC
  • vcMMAE vcMMAE
  • a cysteine mutant form of the HBLB antibody, a vcMMAE drug linker conjugated to each heavy chain at position 239, producing at conjugate with an average drug load of 2 drug linkers per antibody; designated hLIV-14d-vcMMAE(2)) exhibited similar activity as the 4-loaded form.
  • BR2-22a sometimes also referred to as an mAb2, in a mouse monoclonal antibody of isotype IgG1Kappa.
  • 1 ⁇ 10 5 antigen expressing cells (either MCF7 cells expressing human LIV-1, 293F cells, a transfected CHO cell line expressing human LIV-1 or a transfected CHO cell line expressing cyno LIV-1) were aliquoted per wall of 96-well v-bottom plates. AtexaFlour-647 labeled murine BR2-22a was added in concentrations ranging from 0.66 pM to 690 nM and incubated on ice for 30 minutes. Cells were pelleted and washed 3 ⁇ with PBS/BSA. The cells were then pelleted and resuspended in 125 ⁇ L of PBS/BSA. Fluorescence was analyzed by flow cytometry, using percent of saturated fluorescent signal to determine percent bound and to subsequently calculate apparent Kd.
  • CHO cells expressing recombinant LIV-1 in PBS were aliquoted into each cell well of a 96-well v-bottom plates on ice. The cells were incubated for 1 hour with 5 nM AlexaFluor-647 (AF) labeled parental BR2-22a and increasing concentrations (from 0.038 nM to 600 mM) of unlabeled humanized BR2-22a antibody in all combinations of humanized light Chains LA-LG and humanized heavy chains HA-HG. Cells were pelleted and washed 3 times with PBS. The cells were then pelleted and resuspended in 125 ⁇ L of PBS/BSA.
  • AF AlexaFluor-647
  • mice 7-8 animals/group were implanted with tumor cells grown in culture MCF-7 (NCI) at 5 ⁇ 10 6 in 25% matrigel, PC3 from ATCC (2.5 ⁇ 10 6 cells in 25% matrigel), and PC3 from DSMZ (5 ⁇ 10 5 in 25% matrigel).
  • MCF-7 MCF-7
  • ATCC 2.5 ⁇ 10 6 cells in 25% matrigel
  • DSMZ DSMZ
  • mice Female mice also received estrogen supplementation by implanting a slow-release estrogen pellet (90 day release).
  • Tumor volumes were monitored using calipers and animals were euthanized when tumor volume reached ⁇ 800 mm 3 Median tumor volume plots were continued for each group until one or more animals were euthanized. All animal procedures were performed under a protocol approved by the Institutional Animal Care and Use Committee in a facility accredited by the Association for Assessment and Accreditation of Laboratory Animal Care.
  • BR2-22a shows 94% identity to BR2-14a in the mature heavy chain variable region and 91% identity in the mature light chain variable region.
  • the KD for the murine LivI of BR2-22a (Table 5) was determined for human LIV-1 expressed as an endogenous protein in a human breast cancer cell line, in 293F cells or as a recombinant protein in a CHO cell line.
  • the KD for BR2-22a was also determined for cyno LIV-1 expressed as a recombinant protein in a CHO cell line.
  • the BR2-22a antibody was humanized using a VH1-02 JH5 germline acceptor sequence for the heavy chain and a VK2-30 JK4 acceptor sequence for the light chain. These acceptor sequences were chosen based on their having the highest sequence identity to the mature variable region frameworks of BR2-22A heavy and light chains. Initially five variant heavy chains were constructed. Each included the three Kabat CDRs from the heavy chain of BR2-22a, the chains differing in having from zero (VA) to 11 (VE) backmutations. Initially six variant light chains were constructed. Each included the three Kabat CDRs from the light chain of BR2-22a and from zero (LA) to four backmutations (LF).
  • FIGS. 16A and 16B The full sequence of the mature variable region of each variant is shown in FIGS. 16A and 16B .
  • FIG. 18 shows the variants tested.
  • LF-1 through LF-4 differ from LF in each lacking a different backmutation prevent in LF.
  • HE-I through HE-I1 lack one of the backmutations present in HE.
  • FIG. 19 compares LF-1 through LF-4 (each paired with HE).
  • FIG. 19 shows that LF-2 and LF-3 lose substantial binding affinity relative to LF (indicated as HELF historic control in the graph), whereas LF-1 and LF-4 do not.
  • FIG. 20 shows similar binding curves for the HE variants.
  • FIG. 20 shows that HF-11 lost most of its binding indicating that backmutation at position H94 has the greatest effect on binding affinity of the backmutations tested.
  • Loss of backmutations at positions H27, H29 and H30 also caused significant loss of affinity.
  • the role of H30 can be rationalized by the mouse residue being the result of somatic mutation. Loss of a back mutation at position H76 caused some loss of affinity.
  • the other back mutations at positions H28, H48, H66, H67, H71 and H93 could be dispensed with little or no effect on binding affinity.
  • HF and HG were constructed as was light chain LG.
  • HF included backmutations at H27, H29, H30 and H94 and HG included these mutations and a backmutation at H76.
  • LG contains backmutations at L36 and L46.
  • permutations of HF, HG, LE and LF were tested for competition binding as shown in FIG. 21 and all showed binding within a factor of three of that of mouse BR2-22a.
  • HGLG was selected for further experiments as representing the best combination of binding affinity and fewest backmutations.
  • This antibody is hereafter referred to as hLIV22.
  • the saturation binding affinity of hLIV22 for human and cyano LIV-1 expressed from CHO cells is shown in FIG. 22 compared with that of hLIV14.
  • FIG. 22 shows that hLIV22 has about four fold higher affinity (inverse of dissociation constant) for human LIV-1 that does hLIV14.
  • the affinity of hLIV22 for human LIV-1 is the same within experimental error as its affinity for cynomolgus LIV-1, whereas hLIV14 shows twice the affinity for human LIV-1 as for cynomolgus LIV-1.
  • the affinity of hLIV22 for human LIV-1 is the same within experimental error as that of the parent mouse antibody, BR2-22a.
  • Anti-tumor activity of hLIV22 ADC in vitro was measured using cytotoxicity assays.
  • cytotoxicity assays First, we performed a survey of LIV-1 expression in various cell lines by quantitative FACS analysis. The breast cancer cell line MCF-7 from ATCC had the highest level of LIV-1 binding sites/cell, as compared to the MCF-7 cell line from other sources (data not shown). We used this cell line for in vitro assays.
  • various hLIV22 ADCs configured with vcMMAE (referred to as 1006) or mcMMAF (referred to as 1269) (both small molecules described in US 2005-0238469)) were highly effective in killing MCF-7 cells as measured by the in vitro cytotoxic assay.
  • FIGS. 23 and 24 compare hLIV22-conjugated to 1006 or 1269 with a nonbinding control antibody conjugated to 1006 or 1269.
  • hLIV22 ADCs conjugated to vcMMAE showed significant tumor delay compared to untreated and control ADCs. There were multiple complete regressions was observed in the MCF-7 study using hLIV22-vcMMAE at 3 mg/kg. Additionally, in all studies there were a number of animals that had tumors that were static or grew slowly compared to controls. These studies demonstrate that hLIV 22 ADC can stop or delay growth of LIV-1 expressing cancers, including breast and prostate.
  • hLIV22 and hLIV14 ADCs were also tested in a model of cervical cancer.
  • a HeLA cell xenograft model was used for the assay. After tumors grew to an appropriate six, hLIV22 conjugated to vcMMAE was administered to animals at 3 mg/kg and 1 mg/kg. A control antibody conjugate was administered at 3 mg/kg. Complete and partial regression were observed in animals that received 3 mg/kg hLIV22 vcMMAE conjugate. (Data not shown.)
  • LIV-1 antibodies and antibody drug conjugates can be used to treat LIV-1 expressing cervical cancers.
  • Results are shown in FIG. 28 . Seventy-two percent of the tested melanoma patient samples (21/29) were positive for LIV-1 expression. This indicates that LIV-1 inhibitors, e.g., anti-LIV-1antibodies, can be used to treat melanoma cancers.
  • LIV-1 inhibitors e.g., anti-LIV-1antibodies
  • mice 7-8 animals/group are implanted with 10 ⁇ 10 6 SK-MEL-5 cells (a melanoma tumor-derived cell line) grown in culture. Tumors are allowed to grow in vivo until they are 100 mm 3 , as measured using a caliper.
  • Humanized LIV-1 ADCs e.g., hLIV14 or hLIV22, are administered at 3 mg/kg.
  • Drug conjugates are, e.g., vcMMAE or mcMMAF.
  • Control ADC's are also administered to control animals at 3 mg/kg.
  • ADC's are given as q4d ⁇ 4 intraperitoneal injections.
  • Tumor volumes are monitored using calipers and animals are euthanized when tumor volume reaches ⁇ 800 mm 3 .
  • Administration of hLIV14 ADC or hLIV22 ADC greatly reduced tumor growth in animals as compared to those animals that received control ADC's.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Reproductive Health (AREA)
  • Pregnancy & Childbirth (AREA)
  • Gynecology & Obstetrics (AREA)
  • Neurosurgery (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Endocrinology (AREA)
  • Urology & Nephrology (AREA)
  • Dermatology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US15/862,389 2010-12-06 2011-12-06 Humanized antibodies to LIV-1 and use of same to treat cancer Active 2032-08-20 USRE48959E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/862,389 USRE48959E1 (en) 2010-12-06 2011-12-06 Humanized antibodies to LIV-1 and use of same to treat cancer

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US42029110P 2010-12-06 2010-12-06
US201161446990P 2011-02-25 2011-02-25
US13/990,778 US9228026B2 (en) 2010-12-06 2011-12-06 Humanized antibodies to LIV-1 and use of same to treat cancer
US15/862,389 USRE48959E1 (en) 2010-12-06 2011-12-06 Humanized antibodies to LIV-1 and use of same to treat cancer
PCT/US2011/063612 WO2012078688A2 (en) 2010-12-06 2011-12-06 Humanized antibodies to liv-1 and use of same to treat cancer

Publications (1)

Publication Number Publication Date
USRE48959E1 true USRE48959E1 (en) 2022-03-08

Family

ID=46207694

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/990,778 Ceased US9228026B2 (en) 2010-12-06 2011-12-06 Humanized antibodies to LIV-1 and use of same to treat cancer
US15/862,389 Active 2032-08-20 USRE48959E1 (en) 2010-12-06 2011-12-06 Humanized antibodies to LIV-1 and use of same to treat cancer
US14/948,183 Active 2031-12-08 US9783608B2 (en) 2010-12-06 2015-11-20 Humanized antibodies to LIV-1 and use of same to treat cancer
US15/692,424 Abandoned US20180079810A1 (en) 2010-12-06 2017-08-31 Humanized Antibodies To LIV-1 And Use Of Same To Treat Cancer
US16/784,069 Abandoned US20200165335A1 (en) 2010-12-06 2020-02-06 Humanized antibodies to liv-1 and use of same to treat cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/990,778 Ceased US9228026B2 (en) 2010-12-06 2011-12-06 Humanized antibodies to LIV-1 and use of same to treat cancer

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/948,183 Active 2031-12-08 US9783608B2 (en) 2010-12-06 2015-11-20 Humanized antibodies to LIV-1 and use of same to treat cancer
US15/692,424 Abandoned US20180079810A1 (en) 2010-12-06 2017-08-31 Humanized Antibodies To LIV-1 And Use Of Same To Treat Cancer
US16/784,069 Abandoned US20200165335A1 (en) 2010-12-06 2020-02-06 Humanized antibodies to liv-1 and use of same to treat cancer

Country Status (25)

Country Link
US (5) US9228026B2 (ko)
EP (4) EP2648752B1 (ko)
JP (5) JP6105481B2 (ko)
KR (5) KR20220011811A (ko)
CN (1) CN103533957B (ko)
AU (4) AU2011338480B8 (ko)
BR (2) BR122021020513B1 (ko)
CA (2) CA3211246A1 (ko)
CY (3) CY1118874T1 (ko)
DK (3) DK3156420T3 (ko)
ES (3) ES2620264T3 (ko)
HR (1) HRP20170567T1 (ko)
HU (3) HUE043355T2 (ko)
IL (4) IL299837A (ko)
LT (1) LT2648752T (ko)
MX (2) MX351027B (ko)
NZ (1) NZ611468A (ko)
PL (3) PL2648752T3 (ko)
PT (3) PT2648752T (ko)
RS (1) RS55843B1 (ko)
RU (1) RU2608646C2 (ko)
SG (3) SG190938A1 (ko)
SI (3) SI2648752T1 (ko)
WO (1) WO2012078688A2 (ko)
ZA (1) ZA201303803B (ko)

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RS55843B1 (sr) * 2010-12-06 2017-08-31 Seattle Genetics Inc Humanizovana antitela za liv-1 i upotreba istih u lečenju kancera
JP6538561B2 (ja) 2012-10-25 2019-07-03 バイオベラティブ・ユーエスエイ・インコーポレイテッド 抗補体C1s抗体とそれらの用途
CA2889197A1 (en) 2012-11-02 2014-05-08 True North Therapeutics, Inc. Anti-complement c1s antibodies and uses thereof
WO2014089177A2 (en) 2012-12-04 2014-06-12 Massachusetts Institute Of Technology Compounds, conjugates and compositions of epipolythiodiketopiperazines and polythiodiketopiperazines
MX2015008448A (es) 2012-12-27 2016-04-07 Sanofi Sa Anticuerpos anti-lamp1 y conjugados anticuerpo farmaco, y usos de estos.
SG11201600734YA (en) * 2013-07-31 2016-02-26 Amgen Inc Stabilization of fc-containing polypeptides
MX2021008464A (es) 2013-10-15 2023-03-03 Seattle Genetics Inc Farmaco-enlazadores modificados con polietilenglicol para la farmacocinetica mejorada de conjugados de ligando-farmaco.
KR102647074B1 (ko) 2014-02-17 2024-03-14 씨젠 인크. 친수성 항체-약물 컨쥬게이트
EP3280440B1 (en) 2015-04-06 2022-11-16 Bioverativ USA Inc. Humanized anti-c1s antibodies and methods of use thereof
AU2016363013B2 (en) 2015-12-04 2022-03-10 Seagen Inc. Conjugates of quaternized tubulysin compounds
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
IL311107A (en) 2016-02-17 2024-04-01 Seagen Inc BCMA antibodies and their use for the treatment of cancer and immune disorders
MA45324A (fr) 2016-03-15 2019-01-23 Seattle Genetics Inc Polythérapie utilisant un adc-liv1 et un agent chimiothérapeutique
KR102626498B1 (ko) 2016-03-25 2024-01-19 씨젠 인크. 페길화된 약물-링커 및 그의 중간체의 제조를 위한 공정
WO2017197045A1 (en) 2016-05-11 2017-11-16 Movassaghi Mohammad Convergent and enantioselective total synthesis of communesin analogs
MX2019001302A (es) 2016-08-09 2019-06-12 Seattle Genetics Inc Conjugados de farmaco con enlazadores autoestabilizantes que tienen propiedades fisioquimicas mejoradas.
EA201891851A1 (ru) * 2016-09-16 2019-04-30 Сиэтл Дженетикс, Инк. Антитела против bcma и их применение для лечения злокачественных новообразований и иммунологических нарушений
CN109843334A (zh) 2016-10-18 2019-06-04 西雅图基因公司 烟酰胺腺嘌呤二核苷酸补救途径抑制剂的靶向递送
US10239862B2 (en) 2017-03-15 2019-03-26 Silverback Therapeutics, Inc. Benzazepine compounds, conjugates, and uses thereof
CA3056134A1 (en) 2017-03-24 2018-09-27 Seattle Genetics, Inc. Process for the preparation of glucuronide drug-linkers and intermediates thereof
US11305224B2 (en) 2017-04-18 2022-04-19 3M Innovative Properties Company Air filter media with post-pleat-deposited sorbent particles
TW201904613A (zh) 2017-04-27 2019-02-01 美商西雅圖遺傳學公司 季鹼化菸鹼醯胺腺二核苷酸補救合成抑制劑結合物
US11932650B2 (en) 2017-05-11 2024-03-19 Massachusetts Institute Of Technology Potent agelastatin derivatives as modulators for cancer invasion and metastasis
BR112020004225A2 (pt) * 2017-09-02 2020-09-08 Abbvie Inc. conjugados de fármaco de anticorpo anti-egfr (adc) e usos dos mesmos
WO2019046859A1 (en) * 2017-09-02 2019-03-07 Abbvie Inc. ANTI-EGFR-DRUG ANTIBODY (ADC) CONJUGATES AND USES THEREOF
US10640508B2 (en) 2017-10-13 2020-05-05 Massachusetts Institute Of Technology Diazene directed modular synthesis of compounds with quaternary carbon centers
EP3717518A1 (en) 2017-12-01 2020-10-07 Seattle Genetics, Inc. Humanized anti-liv1 antibodies for the treatment of breast cancer
AU2019225845A1 (en) 2018-02-20 2020-09-24 Seagen Inc. Hydrophobic Auristatin F compounds and conjugates thereof
US20200113912A1 (en) 2018-09-12 2020-04-16 Silverback Therapeutics, Inc. Methods and Compositions for the Treatment of Disease with Immune Stimulatory Conjugates
WO2020056194A1 (en) 2018-09-12 2020-03-19 Silverback Therapeutics, Inc. Benzazepine compounds, conjugates, and uses thereof
CA3112545A1 (en) 2018-09-12 2020-03-19 Silverback Therapeutics, Inc. Substituted benzazepine compounds, conjugates, and uses thereof
CN113164774A (zh) 2018-09-12 2021-07-23 希沃尔拜克治疗公司 Toll样受体激动剂的抗体缀合物
US20210292429A1 (en) * 2018-11-07 2021-09-23 Crispr Therapeutics Ag Anti-liv1 immune cell cancer therapy
TW202112354A (zh) 2019-06-05 2021-04-01 美商西雅圖遺傳學公司 遮蔽抗體調配物
WO2020247574A1 (en) 2019-06-05 2020-12-10 Seattle Genetics, Inc. Methods of purifying masked antibodies
US11535634B2 (en) 2019-06-05 2022-12-27 Massachusetts Institute Of Technology Compounds, conjugates, and compositions of epipolythiodiketopiperazines and polythiodiketopiperazines and uses thereof
WO2021016233A1 (en) * 2019-07-22 2021-01-28 Seagen Inc. Humanized anti-liv1 antibodies for the treatment of cancer
EP4013456A1 (en) 2019-08-15 2022-06-22 Silverback Therapeutics, Inc. Formulations of benzazepine conjugates and uses thereof
AU2020358726A1 (en) 2019-10-01 2022-04-07 Silverback Therapeutics, Inc. Combination therapy with immune stimulatory conjugates
AU2020399976A1 (en) 2019-12-09 2022-06-30 Merck Sharp & Dohme B.V. Combination therapy with LIV1-ADC and PD-1 antagonist
TW202138388A (zh) 2019-12-30 2021-10-16 美商西根公司 以非海藻糖苷化抗-cd70抗體治療癌症之方法
WO2021168274A1 (en) 2020-02-21 2021-08-26 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
TW202216211A (zh) 2020-07-01 2022-05-01 美商希沃爾拜克治療公司 抗asgr1抗體共軛物及其用途
JP2023543026A (ja) 2020-09-28 2023-10-12 シージェン インコーポレイテッド がんの処置のためのヒト化抗liv1抗体
AR124681A1 (es) 2021-01-20 2023-04-26 Abbvie Inc Conjugados anticuerpo-fármaco anti-egfr
EP4370211A1 (en) 2021-07-14 2024-05-22 Seagen Inc. Antibody masking domains
WO2023049825A1 (en) 2021-09-24 2023-03-30 Seagen Inc. Improved antibody masking domains
WO2023111913A1 (en) 2021-12-15 2023-06-22 Crispr Therapeutics Ag Engineered anti-liv1 cell with regnase-1 and/or tgfbrii disruption

Citations (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
US5122368A (en) 1988-02-11 1992-06-16 Bristol-Myers Squibb Company Anthracycline conjugates having a novel linker and methods for their production
US5514554A (en) 1991-08-22 1996-05-07 Becton Dickinson And Company Methods and compositions for cancer therapy and for prognosticating responses to cancer therapy
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US5693465A (en) 1994-09-20 1997-12-02 University Of Wales College Of Medicine Methods for predicting the behaviour of breast tumours
WO1998018945A1 (en) 1996-10-31 1998-05-07 Abbott Laboratories Reagents and methods useful for detecting diseases of the breast
WO1998034118A1 (en) 1997-01-30 1998-08-06 Yale University Diagnostic methods and compositions based on the distribution of rad51
US5824805A (en) 1995-12-22 1998-10-20 King; Dalton Branched hydrazone linkers
WO1999023230A1 (en) 1997-10-31 1999-05-14 Abbott Laboratories Reagents and methods useful for detecting diseases of the breast
WO1999025877A1 (en) 1997-11-18 1999-05-27 Abbott Laboratories Reagents and methods useful for detecting diseases of the breast
WO1999033869A2 (en) 1997-12-24 1999-07-08 Corixa Corporation Compounds for immunotherapy and diagnosis of breast cancer and methods for their use
WO2000008210A1 (en) 1998-08-04 2000-02-17 Diadexus Llc A novel method of diagnosing, monitoring, staging, imaging and treating breast cancer
US6066778A (en) 1996-11-06 2000-05-23 The Regents Of The University Of Michigan Transgenic mice expressing APC resistant factor V
WO2000055174A1 (en) 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Human prostate cancer associated gene sequences and polypeptides
US6130237A (en) 1996-09-12 2000-10-10 Cancer Research Campaign Technology Limited Condensed N-aclyindoles as antitumor agents
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
WO2001055178A2 (en) 2000-01-25 2001-08-02 Genentech, Inc. Liv-1 related protein, polynucleotides encoding the same and use thereof for treatment of cancer
WO2001096372A2 (en) 2000-06-13 2001-12-20 University College Cardiff Consultants Ltd Zinc transporters proteins and their use in medicinal preparations
US6468790B1 (en) 1998-10-15 2002-10-22 Chiron Corporation Metastatic breast and colon cancer regulated genes
WO2003075855A2 (en) 2002-03-08 2003-09-18 Protein Design Labs, Inc. Antibodies against cancer antigen tmeff2 and uses thereof
WO2004010957A2 (en) 2002-07-31 2004-02-05 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US6762020B1 (en) 1999-03-15 2004-07-13 Protein Design Labs, Inc. Methods of diagnosing breast cancer
US20040141983A1 (en) 1999-03-15 2004-07-22 Protein Design Labs, Inc. Compositions against cancer antigen LIV-1 and uses thereof
WO2004067564A2 (en) 2003-01-29 2004-08-12 Protein Design Labs, Inc. Compositions against cancer antigen liv-1 and uses thereof
WO2004066933A2 (en) 2003-01-27 2004-08-12 Biogen Idec Ma Inc. Compositions and methods for treating cancer using igsf9 and liv-1
KR20050010267A (ko) 2003-07-18 2005-01-27 삼성전자주식회사 식각장치
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
WO2005058961A2 (en) 2003-12-12 2005-06-30 Amgen Inc. Antibodies specific for human galanin, and uses thereof
US20050238649A1 (en) 2003-11-06 2005-10-27 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US7022500B1 (en) 1988-12-28 2006-04-04 Protein Design Labs, Inc. Humanized immunoglobulins
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
US20060222653A1 (en) 2004-11-12 2006-10-05 Xencor, Inc. Antibodies operably linked to selected chemoattractants
US7141549B2 (en) 2001-01-11 2006-11-28 Peter Mezes Proteins and nucleic acids encoding same
US20060286112A1 (en) 2005-05-16 2006-12-21 Sirid-Aimee Kellermann Human monoclonal antibodies that bind to very late antigen-1 for the treatment of inflammation and other disorders
WO2007120787A2 (en) 2006-04-13 2007-10-25 Novartis Vaccines & Diagnostics, Inc. Methods of treating, diagnosing or detecting cancers associated with liv-1 overexpression
WO2008131376A2 (en) 2007-04-23 2008-10-30 Wyeth Methods and compositions for treating and monitoring treatment of il-13-associated disorders
US7494775B2 (en) 2001-06-21 2009-02-24 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention and therapy of breast and ovarian cancer
US7501121B2 (en) 2004-06-17 2009-03-10 Wyeth IL-13 binding agents
WO2009068649A2 (en) 2007-11-30 2009-06-04 Glaxo Group Limited Antigen-binding constructs
US7705120B2 (en) 2001-06-21 2010-04-27 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
US20100158909A1 (en) 2006-12-01 2010-06-24 Seattle Genetics, Inc. Variant Target Binding Agents and Uses Thereof
US20100158919A1 (en) 2008-12-19 2010-06-24 Isabelle Bauer Dauphin Pharmaceutical Composition
US20110166838A1 (en) 2008-06-16 2011-07-07 Sividon Diagnostics Algorithms for outcome prediction in patients with node-positive chemotherapy-treated breast cancer
US20110165566A1 (en) 2009-07-09 2011-07-07 Wittliff James L Methods of optimizing treatment of breast cancer
US20110195995A1 (en) 2008-10-14 2011-08-11 Wittliff James L Methods of Optimizing Treatment of Estrogen-Receptor Positive Breast Cancers
US20110280892A1 (en) 2005-09-07 2011-11-17 Medimmune, Llc Toxin conjugated eph receptor antibodies
WO2012078688A2 (en) 2010-12-06 2012-06-14 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
WO2012125712A2 (en) 2011-03-14 2012-09-20 Respira Health, Llc Lung tumor classifier for current and former smokers
US8642270B2 (en) 2009-02-09 2014-02-04 Vm Institute Of Research Prognostic biomarkers to predict overall survival and metastatic disease in patients with triple negative breast cancer
US20140127239A1 (en) 2012-10-12 2014-05-08 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
US8728730B2 (en) 2009-09-03 2014-05-20 Genentech, Inc. Methods for treating, diagnosing, and monitoring rheumatoid arthritis
WO2017161007A1 (en) 2016-03-15 2017-09-21 Seattle Genetics, Inc. Combination therapy using a liv1-adc and a chemotherapeutic
WO2019109007A1 (en) 2017-12-01 2019-06-06 Seattle Genetics, Inc. Humanized anti-liv1 antibodies for the treatment of breast cancer
WO2020095249A1 (en) 2018-11-07 2020-05-14 Crispr Therapeutics Ag Anti-liv1 immune cell cancer therapy
WO2020249483A1 (en) 2019-06-10 2020-12-17 University College Cardiff Consultants Limited Anti-mitotic composition comprising antibodies against zip6 and/or zip10

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH652145A5 (de) 1982-01-22 1985-10-31 Sandoz Ag Verfahren zur in vitro-herstellung von hybridomen welche humane monoklonale antikoerper erzeugen.
US4634666A (en) 1984-01-06 1987-01-06 The Board Of Trustees Of The Leland Stanford Junior University Human-murine hybridoma fusion partner
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
AU600575B2 (en) 1987-03-18 1990-08-16 Sb2, Inc. Altered antibodies
ATE120454T1 (de) 1988-06-14 1995-04-15 Cetus Oncology Corp Kupplungsmittel und sterisch gehinderte, mit disulfid gebundene konjugate daraus.
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
SG48759A1 (en) 1990-01-12 2002-07-23 Abgenix Inc Generation of xenogenic antibodies
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
EP0546073B1 (en) 1990-08-29 1997-09-10 GenPharm International, Inc. production and use of transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5858657A (en) 1992-05-15 1999-01-12 Medical Research Council Methods for producing members of specific binding pairs
US5871907A (en) 1991-05-15 1999-02-16 Medical Research Council Methods for producing members of specific binding pairs
US6407213B1 (en) 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
AU3328493A (en) 1991-12-17 1993-07-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
ATE199392T1 (de) 1992-12-04 2001-03-15 Medical Res Council Multivalente und multispezifische bindungsproteine, deren herstellung und verwendung
US5877218A (en) 1994-01-10 1999-03-02 Teva Pharmaceutical Industries, Ltd. Compositions containing and methods of using 1-aminoindan and derivatives thereof and process for preparing optically active 1-aminoindan derivatives
US5834597A (en) 1996-05-20 1998-11-10 Protein Design Labs, Inc. Mutated nonactivating IgG2 domains and anti CD3 antibodies incorporating the same
ATE385007T1 (de) 1999-02-05 2008-02-15 Samsung Electronics Co Ltd Verfahren zum wiederauffinden von bildtexturen und vorrichtung dafür
WO2004006955A1 (en) 2001-07-12 2004-01-22 Jefferson Foote Super humanized antibodies
EP2298807A3 (en) 2004-07-30 2011-05-18 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US8163551B2 (en) 2008-05-02 2012-04-24 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation

Patent Citations (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
US5122368A (en) 1988-02-11 1992-06-16 Bristol-Myers Squibb Company Anthracycline conjugates having a novel linker and methods for their production
US7022500B1 (en) 1988-12-28 2006-04-04 Protein Design Labs, Inc. Humanized immunoglobulins
US5514554A (en) 1991-08-22 1996-05-07 Becton Dickinson And Company Methods and compositions for cancer therapy and for prognosticating responses to cancer therapy
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US5693465A (en) 1994-09-20 1997-12-02 University Of Wales College Of Medicine Methods for predicting the behaviour of breast tumours
US5824805A (en) 1995-12-22 1998-10-20 King; Dalton Branched hydrazone linkers
US6130237A (en) 1996-09-12 2000-10-10 Cancer Research Campaign Technology Limited Condensed N-aclyindoles as antitumor agents
WO1998018945A1 (en) 1996-10-31 1998-05-07 Abbott Laboratories Reagents and methods useful for detecting diseases of the breast
US6066778A (en) 1996-11-06 2000-05-23 The Regents Of The University Of Michigan Transgenic mice expressing APC resistant factor V
WO1998034118A1 (en) 1997-01-30 1998-08-06 Yale University Diagnostic methods and compositions based on the distribution of rad51
WO1999023230A1 (en) 1997-10-31 1999-05-14 Abbott Laboratories Reagents and methods useful for detecting diseases of the breast
WO1999025877A1 (en) 1997-11-18 1999-05-27 Abbott Laboratories Reagents and methods useful for detecting diseases of the breast
WO1999033869A2 (en) 1997-12-24 1999-07-08 Corixa Corporation Compounds for immunotherapy and diagnosis of breast cancer and methods for their use
WO2000008210A1 (en) 1998-08-04 2000-02-17 Diadexus Llc A novel method of diagnosing, monitoring, staging, imaging and treating breast cancer
US6468790B1 (en) 1998-10-15 2002-10-22 Chiron Corporation Metastatic breast and colon cancer regulated genes
WO2000055174A1 (en) 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Human prostate cancer associated gene sequences and polypeptides
US6762020B1 (en) 1999-03-15 2004-07-13 Protein Design Labs, Inc. Methods of diagnosing breast cancer
US20080175839A1 (en) 1999-03-15 2008-07-24 Protein Design Labs, Inc. Compositions Against Cancer Antigen LIV-1 and Uses Thereof
US8313745B2 (en) 1999-03-15 2012-11-20 Abbott Biotherapeutics Corp. Compositions against cancer antigen LIV-1 and uses thereof
US20040141983A1 (en) 1999-03-15 2004-07-22 Protein Design Labs, Inc. Compositions against cancer antigen LIV-1 and uses thereof
US20080138345A1 (en) 2000-01-25 2008-06-12 Genentech, Inc. Compositions and methods for treatment of cancer
US20070264267A1 (en) 2000-01-25 2007-11-15 Genentech, Inc. Compositions and methods for treatment of cancer
US20030215457A1 (en) 2000-01-25 2003-11-20 Sauvage Frederic De Compositions and methods for treatment of cancer
US7982015B2 (en) 2000-01-25 2011-07-19 Genentech, Inc. Compositions and methods for treatment of cancer
EP1263780A2 (en) 2000-01-25 2002-12-11 Genentech, Inc. Liv-1 related protein, polynulceotides encoding the same and use thereof for treatment of cancer
US7691566B2 (en) 2000-01-25 2010-04-06 Genentech, Inc. Compositions and methods for treatment of cancer
US7285382B2 (en) 2000-01-25 2007-10-23 Genentech, Inc. Compositions and methods for treatment of cancer
WO2001055178A2 (en) 2000-01-25 2001-08-02 Genentech, Inc. Liv-1 related protein, polynucleotides encoding the same and use thereof for treatment of cancer
WO2001096372A2 (en) 2000-06-13 2001-12-20 University College Cardiff Consultants Ltd Zinc transporters proteins and their use in medicinal preparations
US7141549B2 (en) 2001-01-11 2006-11-28 Peter Mezes Proteins and nucleic acids encoding same
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US7098308B2 (en) 2001-04-30 2006-08-29 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US7494775B2 (en) 2001-06-21 2009-02-24 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention and therapy of breast and ovarian cancer
US7705120B2 (en) 2001-06-21 2010-04-27 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
US8323906B2 (en) 2001-06-21 2012-12-04 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention and therapy of breast and ovarian cancer
US20130102482A1 (en) 2001-06-21 2013-04-25 Board Of Regents, The University Of Texas System Compositions, kits, and methods for identification, assessment, prevention and therapy of breast and ovarian cancer
US10533227B2 (en) 2001-06-21 2020-01-14 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention and therapy of breast and ovarian cancer
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
US7288248B2 (en) 2002-03-08 2007-10-30 Pdl Biopharma, Inc. Antibodies against cancer antigen TMEFF2 and uses thereof
US20040096392A1 (en) 2002-03-08 2004-05-20 Eos Biotechnology, Inc. Antibodies against cancer antigen TMEFF2 and uses thereof
WO2003075855A2 (en) 2002-03-08 2003-09-18 Protein Design Labs, Inc. Antibodies against cancer antigen tmeff2 and uses thereof
US7659241B2 (en) 2002-07-31 2010-02-09 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
WO2004010957A2 (en) 2002-07-31 2004-02-05 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US20060074008A1 (en) 2002-07-31 2006-04-06 Senter Peter D Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
CN1849337A (zh) 2003-01-27 2006-10-18 比奥根艾迪克Ma公司 利用igsf9和liv-1的、用于治疗癌症的组合物及方法
WO2004066933A2 (en) 2003-01-27 2004-08-12 Biogen Idec Ma Inc. Compositions and methods for treating cancer using igsf9 and liv-1
US20040258616A1 (en) 2003-01-27 2004-12-23 Idec Pharmaceuticals Corporation Compositions and methods for treating cancer using IGSF9 and LIV-1
US8906342B2 (en) 2003-01-29 2014-12-09 Abbvie Biotherapeutics Inc. Compositions against cancer antigen LIV-1 and uses thereof
WO2004067564A2 (en) 2003-01-29 2004-08-12 Protein Design Labs, Inc. Compositions against cancer antigen liv-1 and uses thereof
US8591863B2 (en) 2003-01-29 2013-11-26 Abbvie Biotherapeutics Inc. Compositions against cancer antigen LIV-1 and uses thereof
US20080171039A1 (en) 2003-01-29 2008-07-17 Protein Design Labs, Inc. Compositions Against Cancer Antigen Liv-1 and Uses Thereof
US20140037540A1 (en) 2003-01-29 2014-02-06 Abbvie Biotherapeutics Inc. Compositions against cancer antigen liv-1 and uses thereof
KR20050010267A (ko) 2003-07-18 2005-01-27 삼성전자주식회사 식각장치
US20050238649A1 (en) 2003-11-06 2005-10-27 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US7498298B2 (en) 2003-11-06 2009-03-03 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2005058961A2 (en) 2003-12-12 2005-06-30 Amgen Inc. Antibodies specific for human galanin, and uses thereof
US7501121B2 (en) 2004-06-17 2009-03-10 Wyeth IL-13 binding agents
US20060222653A1 (en) 2004-11-12 2006-10-05 Xencor, Inc. Antibodies operably linked to selected chemoattractants
US20060286112A1 (en) 2005-05-16 2006-12-21 Sirid-Aimee Kellermann Human monoclonal antibodies that bind to very late antigen-1 for the treatment of inflammation and other disorders
US20110280892A1 (en) 2005-09-07 2011-11-17 Medimmune, Llc Toxin conjugated eph receptor antibodies
US20100196377A1 (en) 2006-04-13 2010-08-05 Jantapour Mary J Methods of treating, diagnosing or detecting cancer
WO2007120787A2 (en) 2006-04-13 2007-10-25 Novartis Vaccines & Diagnostics, Inc. Methods of treating, diagnosing or detecting cancers associated with liv-1 overexpression
US20100158909A1 (en) 2006-12-01 2010-06-24 Seattle Genetics, Inc. Variant Target Binding Agents and Uses Thereof
WO2008131376A2 (en) 2007-04-23 2008-10-30 Wyeth Methods and compositions for treating and monitoring treatment of il-13-associated disorders
WO2009068649A2 (en) 2007-11-30 2009-06-04 Glaxo Group Limited Antigen-binding constructs
US20110166838A1 (en) 2008-06-16 2011-07-07 Sividon Diagnostics Algorithms for outcome prediction in patients with node-positive chemotherapy-treated breast cancer
US20110195995A1 (en) 2008-10-14 2011-08-11 Wittliff James L Methods of Optimizing Treatment of Estrogen-Receptor Positive Breast Cancers
US20100158919A1 (en) 2008-12-19 2010-06-24 Isabelle Bauer Dauphin Pharmaceutical Composition
US8642270B2 (en) 2009-02-09 2014-02-04 Vm Institute Of Research Prognostic biomarkers to predict overall survival and metastatic disease in patients with triple negative breast cancer
US20110165566A1 (en) 2009-07-09 2011-07-07 Wittliff James L Methods of optimizing treatment of breast cancer
US8728730B2 (en) 2009-09-03 2014-05-20 Genentech, Inc. Methods for treating, diagnosing, and monitoring rheumatoid arthritis
US20160185858A1 (en) 2010-12-06 2016-06-30 Seattle Genetics, Inc. Humanized Antibodies To LIV-1 And Use Of Same To Treat Cancer
WO2012078688A2 (en) 2010-12-06 2012-06-14 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
US20130259860A1 (en) 2010-12-06 2013-10-03 Seattle Genetics, Inc. Humanized Antibodies To LIV-1 And Use Of Same To Treat Cancer
US9228026B2 (en) 2010-12-06 2016-01-05 Seattle Genetics, Inc. Humanized antibodies to LIV-1 and use of same to treat cancer
US20200165335A1 (en) 2010-12-06 2020-05-28 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
US20180079810A1 (en) 2010-12-06 2018-03-22 Seattle Genetics, Inc. Humanized Antibodies To LIV-1 And Use Of Same To Treat Cancer
US9783608B2 (en) 2010-12-06 2017-10-10 Seattle Genetics, Inc. Humanized antibodies to LIV-1 and use of same to treat cancer
WO2012125712A2 (en) 2011-03-14 2012-09-20 Respira Health, Llc Lung tumor classifier for current and former smokers
US20140127239A1 (en) 2012-10-12 2014-05-08 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2017161007A1 (en) 2016-03-15 2017-09-21 Seattle Genetics, Inc. Combination therapy using a liv1-adc and a chemotherapeutic
US20190085091A1 (en) 2016-03-15 2019-03-21 Seattle Genetics, Inc. Combination therapy using a liv1-adc and a chemotherapeutic
WO2019109007A1 (en) 2017-12-01 2019-06-06 Seattle Genetics, Inc. Humanized anti-liv1 antibodies for the treatment of breast cancer
WO2020095249A1 (en) 2018-11-07 2020-05-14 Crispr Therapeutics Ag Anti-liv1 immune cell cancer therapy
WO2020249483A1 (en) 2019-06-10 2020-12-17 University College Cardiff Consultants Limited Anti-mitotic composition comprising antibodies against zip6 and/or zip10

Non-Patent Citations (107)

* Cited by examiner, † Cited by third party
Title
Almagro & Fransson, Frontiers in Bioscience 2008; 13:1619-33. *
Almagro, J.C. et al. (Jan. 1, 2008). "Humanization of Antibodies," Frontiers in Bioscience13:1619-1633.
Amsberry, K.L. et al. (Nov. 1990). "The Lactonization of 2′-Hydroxyhydrocinnamic Acid Amides: A Potential Prodrug for Amines," The Journal of Organic Chemistry 55(23):5867-5877.
Anonymous (Feb. 25, 2016). "A Safety Study of SGN-LIV1A in Breast Cancer Patients History of Changes for the Study: NCT01969643," retrived from URL:https://clinicaltrials.gov/ct2/history/NTC01969643?A=32&C=Side-by-Side#StudyPageTop, last visited Oct. 17, 2019, 7 pages.
Arnold et al., "Prostate cancer bone metastasis: Reactive oxygen species, growth factors and heparan sulfate proteoglycans provide a signaling triad that supports progression," AACR, Annual Meeting, Abstract No. 4504, 1 page, (2008).
Balmana et al. Ann Oncol 2009; 20(supp 4):iv19-20. *
Balmaña, J. et al. (May 2009). "BRCA in breast cancer: ESMO Clinical Recommendations," Annals of Oncology 20 (supp 4):iv19-iv20.
Bedognetti, D. et al. (2016, e-pub. Apr. 26, 2016). "Checkpoint Inhibitors and Their Application in Breast Cancer," Breast Care 11(2):108-115.
Bhaskar, V. et al. (Oct. 1, 2003). "E-Selectin Up-Regulation Allows for Targeted Drug Delivery in Prostate Cancer," Cancer Research63:6387-6394.
Börresen-Dale, A-L. (2003). "Genetic Profiling of Breast Cancer: From Molecular Portraits to Clinical Utility," International Journal of Biological Markers 18(1):54-56.
Brown et al., J. Immunol. 1996; 156(9):3285-91. *
Brown, M. et al. (May 1, 1996). "Tolerance of single, but not multiple, amino acid replacements in antibody VH CDR 2: a means of minimizing B cell wastage from somatic hypermutation?," J. Immunol. 156(9):3285-3291.
Cao, A. et al.(Aprii 20, 2016). "Abstract 4914: Auristatin-Based Antibody Drug Conjugates Activate Multiple ER Stress Response Pathways Resulting in Immunogenic Cell Death and Amplified T-Cell Responses," Retrieved from the Internet: URL:https://cancerres.aacrjournals.org/content/76/14_Supplement/4914, last visited Mar. 6, 2020, 4 pages.
Chari, R.V.J. et al. (Jan. 1, 1992). "Immunoconjugates Containing Novel Maytansinoids: Promising Anticancer Drugs," Cancer Research 52:127-131.
Cherkassky, M. (2003). "Doxycycline," in RLE Drug Encyclopedia ed.10, pp. 299-300, 9 pages.
Chou, T.-C. (Jan. 15, 2010, e-pub. Jan. 12, 2010). "Drug Combination Studies and Their Synergy Quantification Using the Chou-Talaly Method," Cancer Res 70(2): 440-446.
Criscitiello, C. et al. (2015). "Immunotherapy of Breast Cancer," Immuno-Oncology Prog. Tumor Res. 42:30-43.
Derisi, J. et al. (Dec. 1996). "Use of a cDNA Microarray to Analyse Gene Expression Patterns in Human Cancer," Nature Genetics 14:457-460.
Doronina S.O. et al. (2003). "Development of potent monoclonal antibody auristatin conjugates for cancer therapy,"Nature Biotechnol. 21(7):778-784.
Dressman et al., "Genes that co-cluster with estrogen receptor alpha in microarray analysis of breast biopsies," The Pharmacogenomics Journal, 1:135-141, (2001).
Dressman, M.A. et al. (2001). "Genes that Co-cluster with Estrogen Receptor Alpha in Microarray Analysis of Breast Biopsies," The Pharmacogenomics Journal 1(2):135-141.
Dubowchik, G.M. et al. (Aug. 1999). "Receptor-Mediated and Enzyme-Dependent Targeting of Cytotoxic Anticancer Drugs," Pharm. Therapeutics 83(2):67-123.
El-Tanani, M.K. et al. (Jul. 23, 1996)."Insulin/IGF-1 Modulation of the Expression of two Estrogen-induced Genes in MCF-7 Cells" Molecular and Cellular Endocrinology 121(1):29-35.
El-Tanani, M.K. et al. (Mar. 1997). "Interaction Between Estradiol and Growth Factors in the Regulation of Specific Gene Expression in MCF-7 Human Breast Cancer Cells," J. Steroid Biochem. Molec. Biol. 60(5-6):269-276.
El-Tanani, M.K. et al. (Nov. 29, 1996). "Interaction Between Estradiol and cAMP in the Regulation of Specific Gene Expression," Molecular and Cellular Endocrinology 124(1-2):71-77.
EMBL Database, (Dec. 23, 1995). Accession No. U41060.
EPO Application No. 11847198.6, European Search Report and Supplementary European Search Opinion dated Apr. 16, 2015.
EPO Application No. 11847198.6, European Search Report and Supplementary European Search Opinion mailed Apr. 16, 2015.
EPO Application No. 18204152.5, European Extended Search Report dated Dec. 4, 2018.
EPO Application No. EP16200557.3, European Search Report and Written Opinion of the International Searching Authority dated Jan. 25, 2017.
Extended Search Report, dated Apr. 6, 2020, for European Patent Application No. 17767448.8, 17 pages.
Forero, A. et al. (Dec. 6-10, 2016). "Phase 1 Study of the Antibody-Drug Conjugate (ADC) SGN-LIV1A in Patients with Heavily Pretreated Metastatic Breast Cancer," Poster-Abstract No. P6-12-04, Presented at Session: 621—ew Drug and Treatment Strategies, San Antonio Breast Cancer Symposium; N, San Antonio, TX, three pages.
Forero, A. et al. (Dec. 8-12, 2015). "Interim Analysis of a Phase 1 Study of the Antibody-Drug Conjugate SGN-LIV1A in Patients with Metastatic Breast Cancer," Poster-Abstract No. 638, Presented at Session: 621—New Drug and Treatment Strategies, San Antonio Breast Cancer Symposium; San Antonio, TX, three pages.
Forero, A. et al. (Dec. 9-13, 2014). "SGN-LIV1A: A Phase 1 Trial Evaluating a Novel Antibody-Drug Conjugate in Patients with LIV-1-Positive Breast Cancer," Poster- Abstract No. 419 Presented at San Antonio Breast Cancer Symposium, San Antonio, TX, three pages.
Francisco, J.A. et al. (2003, e-pub. May 8, 2003). "cAC10-vcMMAE, an Anti-CD30-Monomethyl Auristatin E Conjugate With Potent and Selective Antitumor Activity," Blood 102:1458-1465.
Hay, M.P. et al. (Aug. 2, 1999). "A 2-nitroimidazole Carbamate Prodrug of 5-amino-1-(chloromethyl)-3-[(5,6,7-trimethoxyindol-2-yl)carbonyl]-1,2-dihydro-3H-benz[e]indole (amino-seco-CBI-TMI) for Use With ADEPT and GDEPT," Bioorganic & Medicinal Chemistry Letters 9(15):2237-2242.
Hurvitz, S.A. et al. (Mar. 20, 2013, e-pub. Feb. 4, 2013), "Phase II Randomized Study of Trastuzumab Emtansine Versus Trastuzumab Plus Docetaxel in Patients With Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer," Journal of Clinical Oncology 31(9):1157-1163.
Johnson, D.A. et al. (Jul.-Aug. 1995). "Anti-Tumor Activity of CC49-Doxorubicin Immunoconjugates," Anticancer Res. 15(4):1387-1393.
Khan, J. et al. (1999). "Epression profiling in cancer using cDNA microarrays," Electrophoresis 20:223-229.
King, D.J. et al. (1992). "Expression, purification and characterization of a mouse-human chimeric antibody and chimeric Fab' fragment," Biochem. J. 281:317-323.
Kingsbury, W.D. et al. (Nov. 1984). "A Novel Peptide Delivery System Involving Peptidase Activated Prodrugs as Antimicrobial Agents. Synthesis and Biological Activity of Peptidyl Derivatives of 5-Fluorouracil," Journal of Medicinal Chemistry 27(11):1447-1451.
Kobayashi, M. et al. (Mar. 1997). "Antitumor Activity of TZT-1027, a Novel Dolastatin 10 Derivative," Jpn J Cancer Res. 88:316-327.
Kostelny, S.A. et al. (Mar. 1, 1992). "Formation of a Bispecific Antibody by the Use of Leucine Zippers," J. Immunol. 148(5): 1547-1553.
Kostic, A. et al. (May 30-Jun. 3, 2014). "SGN-LIV1A, an Antibody-drug Conjugate, in Patients with LIV-1-positive Breast Cancer," Poster—Abstract No. TPS1143, American Society of Clinical Oncology, Chicago, IL, one page.
Lau, A. et al. (Oct. 1995). "Conjugation of Doxorubicin to Monoclonal Anti-Carcinoembryonic Antigen Antibody Via Novel Thiol-Directed Cross-Linking Reagents," Bioorganic & Medicinal Chemistry 3(10):1299-1304.
Lau, A. et al. (Oct. 1995). "Novel Doxorubicin-Monoclonal Anti-Carcinoembryonic Antigen Antibody Immunoconjugate Activity in vitro," Bioorganic & Medicinal Chemistry 3(10):1305-1312.
Li, F. et al. (Apr. 16-20, 2016). "Preclinical Combinations of the Antibody-drug Conjugate SGN-LIV1A with Chemotherapies Show Increased Activity," Poster—Abstract No. 2966 Presented at AACR Annual Meeting, New Orleans, LA, two pages.
Liefers, G.-J. et al. (Jul. 23, 1998). "Micrometastases and Survival in Stage II Colorectal Cancer," New England J. Med. 339(4):223-228.
Lue et al., "LIV-1 mediates epithelial to mesenchymal transition and correlates with prostatic cancer progression," AACR, Annual Meeting, Abstract No. 5373, 1 page, (2008).
Lue, H.-W. et al. (Apr. 12-16). "LIV-1 Mediates Epithelial to Mesenchymal Transition and Correlates with Prostatic Cancer Progression," AACR, Annual Meeting, Abstract No. 5373, 1 page.
Manning, D.L. et al. (1994). "Oestrogen-regulated Genes in Breast Cancer: Association of pLIV1 With Lymph Node Involvement," European Journal of Cancer 30A(5):675-678.
McClelland, R.A. et al. (1998). "Oestrogen-Regulated Genes in Breast Cancer: Association of pLIVI with Response to EndocrineTherapy," Br. J. Cancer 77(10):1653-1656.
Modi, S. et al. (Dec. 5-7, 2017). "Phase 1 Study of the Antibody-Drug Conjugate Ladiratuzumab Vedotin (SGN-LIV1A) in Patients with Heavily Pretreated Triple-Negative Metastatic Breast Cancer," Abstract No. PD3-14 Presented at Session 621, New Drugs and Treatment Strategies, San Antonio Breast Cancer Symposium, San Antonio, TX, three pages.
Mohammad, R.M. et al. (1999). "A New Tubulin Polymerization Inhibitor, Auristatin PE, Induces Tumor regression in a Human Waldenstrom's Macroglobulinemia Xenograft Model," Intl J. Oncology 15:367-372.
Müller, P. et al. (Aug. 1, 2014). "Cancer Chemotherapy Agents Target Intratumoral Dendritic Cells to Potentiate Antitumor Immunity," OncoImmunology 3(8):e954460, 3 pages.
Nanda, R. et al. (Jul. 20, 2016, e-pub. May 2, 2016). "Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase lb Keynote-012 Study," J. Clin. Oncology 34(21):2460-2467.
Neville, D.M. et al. (Sep. 5, 1989). "Enhancement of Immunotoxin Efficacy by Acid-cleavable Crosslinking Agents Utilizing Diphtheria Toxin and Toxin Mutants," The Journal of Biological Chemistry 264(25):14653-14661.
Partial Supplementary European Search Report, dated Nov. 27, 2019, for European Patent Application No. 17767448.8, 16 pages.
Payne, J.A. et al. (Jul. 28, 1995). "Primary Structure, Functional Expression, and Chromosomal Localization of the Bumetanide-Sensitive Na-K-Cl Cotransporter in Human Colon," J. Biol. Chem. 270:17977-17985.
Pettit, G.R. et al. (1998). "Antineoplastic Agents 365. Dolastatin 10 SAR probes," Anti-Cancer Design 13:243-277.
Pollack et al., Cancer Chemother Pharmacol 2007; 60: 423-35. *
Pollack, V.A. et al. (Aug. 2007). "Treatment Parameters Modulating Regression of Human Melanoma Xenografts by an Antibody—drug Conjugate (CR011-vcMMAE) Targeting GPNMB," Cancer Chemother. Pharmacol. 60(3):423-435.
Poole, R.M. (2014, e-pub. Oct. 21, 2014). "Pembrolizumab: First Global Approval," Drugs 74:1973-1981.
Reichert & Valge-Archer, Nat. Rev. Drug Disc. 2007; 6:349-356. *
Reichert, J.M. et al. (May 2007). "Development Trends for Monoclonal Antibody Cancer Therapeutics," Nat. Rev. Drug Disc. 6(5):349-356.
Rodrigues, M.L. et al. (Apr. 1995). "Synthesis and β-lactamase-mediated Activation of a Cephalosporin-Taxol Prodrug," Chemistry & Biology 2(4):223-227.
Ross, S. et al. (May 1, 2002). "Prostate Stem Cell Antigen as Therapy Target: Tissue Expression and in vivo Efficacy of an Immunoconjugate," Cancer Research 61:2546-2553.
Rudikoff et al., Proc. Nat'l Acad. Sci. USA 1982; 79:1979-83. *
Rudikoff, S. et al. (Mar. 1, 1982). "Single Amino Acid Substitution Altering Antigen-binding Specificity," Proc. Nat'l Acad. Sci. USA 79(6):1979-1983.
Slamon, D.J. et al. (Mar. 15, 2001). "Use of Chemotherapy Plus a Monoclonal Antibody Against HER2 for Metastatic Breast Cancer That Overexpresses HER2" The New England Journal of Medicine 344(11):783-792.
Smith et al., "LIV-1 Antibody-Drug Conjugate: A Novel Therapeutic Agent for Breast Cancer," CTRC-AACR San Antonio Breast Cancer Symposium, Abstract No. 851651, 1 page, (2010).
Smith, L.M. et al. (Dec. 8-12, 2010). "LIV-1 Antibody-Drug Conjugate: A Novel Therapeutic Agent for Breast Cancer," Presented at CTRC-AACR San Antonio Breast Cancer Symposium, Cancer Res 70(24 Suppl):Abstract nr. P6-15-16, one page.
Songsivilai, S. et al. (1990). "Bispecific Antibody: A Tool for Diagnosis and Treatment of Disease," Clin. Exp. Immunol. 79:315-321.
Storm, D.R. et al. (Aug. 1972). "Effect of Small Changes in Orientation on Reaction Rate," Journal of the American Chemical Society 94(16):5815-5825.
Sussman, D. et al. (2014; e-published on Sep. 24, 2014). "SGN-LIV1A: A Novel Antibody—Drug Conjugate Targeting LIV-1 for the Treatment of Metastatic Breast Cancer," Molecular Cancer Therapeutics 13(12):2991-3000.
Sussman, D. et al. (Apr. 2-6, 2011). "LIV-1 Antibody-Drug Conjugate: A Novel Therapeutic Agent for Breast and Prostate Cancer," Poster—Abstract No. 3620 Presented at AACR, one pages.
Sussman, D. et al. (Apr. 6-10, 2013)."SGN-LIV1A: A Development Stage Antibody-Drug Conjugate Targeting LIV-1 for the Treatment of Metastatic Breast Cancer," Presented at AACR, Annual Meeting, Cancer Res 73(8 Suppl):Abstract No. 3962, 1 page.
Swain, S.M. et al. (May 2013, e-pub. Apr. 18, 2013). "Pertuzumab, Trastuzumab, and Docetaxel for HER2-Positive Metastatic Breast Cancer (Cleopatra Study); Overal Survival Results From a Randomised, Double-Blind, Placebo-Controlled Phase 3 Study," Lancet Oncol. 14:461-471.
Taylor, K.M. (Apr. 2000). "Hypothesis Paper: LIV-1 Breast Cancer Protein Belongs to New Family of Histidine-Rich Membrane Proteins with Potential to Control Intracellular Zn2+ Homeostasis," IUBMB Life 49(4):249-253.
Taylor, K.M. et al. (1999). "The LIV-1 Gene, Implicated in Metastatic Breast Cancer, Codes for a Histidine-Rich Transmembrane Protein," British Journal of Cancer 80(Suppl 2):24.
Taylor, K.M. et al. (Apr. 1, 2003). "The LZT Proteins; the LIV-1 Subfamily of Zinc Transporters," Biochimica et Biophysica Acta 1611(1-2):16-30.
Taylor, K.M. et al. (Nov. 2003). "Structure-function Analysis of LIV-1, the Breast Cancer-associated Protein that Belongs to a New Subfamily of Zinc Transporters," Biochem. J. 375(1):51-59.
Thorpe, P.E. et al. (Nov. 15, 1987). "New Coupling Agents for the Synthesis of Immunotoxins Containing a Hindered Disulfide Bond with Improved Stability in Vivo," Cancer Research 47:5924-5931.
Tse et al., Clin Cancer Res, 2006; 12(4):1373-82. *
TSE, K.F. et al. (Feb. 15, 2006). "CR011, a Fully Human Monoclonal Antibody-Auristatin E Conjugate, for the Treatment of Melanoma," Clin. Cancer Res 12(4):1373-1382.
U.S. Appl. No. 13/990,778, Non-Final Office Action dated Apr. 14, 2015.
U.S. Appl. No. 13/990,778, Notice of Allowance dated Sep. 18, 2015.
U.S. Appl. No. 14/052,905, Non-Final Office Action dated Feb. 25, 2014.
U.S. Appl. No. 14/052,905, Notice of Allowance dated Aug. 8, 2014.
U.S. Appl. No. 14/052,905, Notice of Allowance mailed Aug. 8, 2014.
U.S. Appl. No. 14/052,905, Requirement for Restriction/Election dated Nov. 12, 2013.
U.S. Appl. No. 14/052,905, Response to Non-Final Office Action filed Jun. 19, 2014.
U.S. Appl. No. 14/052,905, Response to Requirement for Restriction/Election filed Dec. 12, 2013.
U.S. Appl. No. 14/948,183, Non-Final Office Action dated Feb. 2, 2017.
U.S. Appl. No. 14/948,183, Notice of Allowance dated May 31, 2017.
Unno et al., "LIV-1 enhances the aggressive phenotype through the induction of epithelial to mesenchymal transition in human pancreatic carcinoma cells," International Journal of Oncology, 35: 813-821, (2009).
Unno, J. et al. (Oct. 2009). "LIV-1 Enhances the Aggressive Phenotype Through the Induction of Epithelial to Mesenchymal Transition in Human Pancreatic Carcinoma Cells," International Journal of Oncology 35(4):813-821.
Vogel, C.-W. (1987). Immunoconjugates: Antibody Conjugates in Radioimaging and Therapy of Cancer, Oxford University Press, New York, Oxford pp. 1-160.
Wang, H. et al. (Mar. 1, 2004). "Pretreatment With Dexamethasone Increase Antitumor Activity of Carboplatin and Gemcitabine in Mice Bearing Human Cancer Xenografts: In Vivo Activity, Pharmacokinetics, and Clinical Implications for Cancer Chemotherapy," Clinical Cancer Research 10(5):1633-1644.
Wawrzynczak et al. (1987). "Methods for Preparing Immunotoxins: Effect of the Linkage on Activity and Stability," In Immunoconjugates: Antibody Conjugates in Radioimagery and Therapy of Cancer, pp. 28-55.
Welford, S.M. et al. (1998). "Detection of Differentially Expressed Genes in Primary Tumor Tissues Using Representational Differences Analysis Coupled to Microarray Hybridization," Nucleic Acids Research 26 (12):3059-3065.
Williams, K. et al. (1998). "Analysis of Differential Expression in Normal and Neoplastic Human Breast Epithelial Cell Lines," Electrophoresis 19:333-343.
WIPO Application No. PCT/US2011/063612, PCT International Preliminary Report on Patentability dated Jun. 20, 2013.
WIPO Application No. PCT/US2011/063612, PCT International Search Report and Written Opinion of the International Searching Authority dated Jun. 27, 2012.
WIPO Application No. PCT/US2017/22541, PCT International Search Report and Written Opinion of the International Searching Authority dated Jul. 31, 2012.
Yamamoto, K. et al. (Mar. 1996). "Clinicial Application of Chimeric Monoclonal Antibody A7-NCS Conjugate," Biotherapy 10:365-367, Abstract.
Yang, G.P. et al. (1999). "Combining SSH and cDNA Microarrays for Rapid Identification of Differentially Expressed Genes," Nucleic Acids Research 27(6):1517-1523.

Also Published As

Publication number Publication date
JP6869218B2 (ja) 2021-05-12
ES2719548T3 (es) 2019-07-11
KR20190076068A (ko) 2019-07-01
EP3156420A1 (en) 2017-04-19
JP2023011709A (ja) 2023-01-24
IL261900A (en) 2018-10-31
AU2019202530A1 (en) 2019-05-02
CY1123883T1 (el) 2022-03-24
MX2013006365A (es) 2013-08-15
US20130259860A1 (en) 2013-10-03
EP2648752B1 (en) 2017-02-15
LT2648752T (lt) 2017-04-10
RU2013130609A (ru) 2015-01-20
BR122021020513B1 (pt) 2022-10-25
EP3461847A1 (en) 2019-04-03
NZ611468A (en) 2015-06-26
MX2020010639A (es) 2020-10-28
AU2011338480A8 (en) 2017-03-23
CN103533957A (zh) 2014-01-22
CA2819038C (en) 2023-10-17
HUE031726T2 (en) 2017-07-28
JP6453924B2 (ja) 2019-01-16
SG10201912646UA (en) 2020-02-27
WO2012078688A2 (en) 2012-06-14
US20200165335A1 (en) 2020-05-28
JP2014506120A (ja) 2014-03-13
IL273607A (en) 2020-05-31
SG190938A1 (en) 2013-07-31
JP2017149717A (ja) 2017-08-31
CY1121546T1 (el) 2020-05-29
CY1118874T1 (el) 2018-01-10
PT2648752T (pt) 2017-03-28
PL2648752T3 (pl) 2017-07-31
EP2648752A2 (en) 2013-10-16
CN103533957B (zh) 2016-06-22
EP3156420B1 (en) 2019-02-20
MX351027B (es) 2017-09-28
SI3156420T1 (sl) 2019-05-31
SI3461847T1 (sl) 2021-03-31
JP6105481B2 (ja) 2017-03-29
CA3211246A1 (en) 2012-06-14
SI2648752T1 (sl) 2017-06-30
EP3786185A1 (en) 2021-03-03
AU2017203851A1 (en) 2017-06-22
IL226754B (en) 2018-10-31
EP3461847A8 (en) 2021-04-14
AU2017203851B2 (en) 2019-01-31
WO2012078688A3 (en) 2012-08-23
DK3156420T3 (en) 2019-04-29
AU2019202530B2 (en) 2021-04-01
HUE043355T2 (hu) 2019-08-28
IL299837A (en) 2023-03-01
CA2819038A1 (en) 2012-06-14
ES2620264T3 (es) 2017-06-28
AU2011338480B8 (en) 2017-03-23
KR20220011811A (ko) 2022-01-28
US9228026B2 (en) 2016-01-05
RU2608646C2 (ru) 2017-01-23
DK2648752T3 (en) 2017-03-27
JP2021106599A (ja) 2021-07-29
AU2021204473A1 (en) 2021-07-29
US20160185858A1 (en) 2016-06-30
DK3461847T3 (da) 2020-12-14
PL3461847T3 (pl) 2021-07-05
HRP20170567T1 (hr) 2017-06-16
PT3156420T (pt) 2019-05-27
US9783608B2 (en) 2017-10-10
KR20200145867A (ko) 2020-12-30
JP7166384B2 (ja) 2022-11-07
KR101993921B1 (ko) 2019-06-28
AU2011338480B2 (en) 2017-03-16
KR102198189B1 (ko) 2021-01-05
JP2019073515A (ja) 2019-05-16
RS55843B1 (sr) 2017-08-31
ES2842895T3 (es) 2021-07-15
KR20230057485A (ko) 2023-04-28
PT3461847T (pt) 2020-12-24
SG10201510041QA (en) 2016-01-28
PL3156420T3 (pl) 2019-08-30
EP3461847B1 (en) 2020-09-23
EP2648752A4 (en) 2015-05-20
KR20130135884A (ko) 2013-12-11
IL273607B2 (en) 2023-06-01
HUE052806T2 (hu) 2021-05-28
ZA201303803B (en) 2014-07-30
US20180079810A1 (en) 2018-03-22
AU2011338480A1 (en) 2013-06-13
BR112013013781A2 (pt) 2016-09-13

Similar Documents

Publication Publication Date Title
US20200165335A1 (en) Humanized antibodies to liv-1 and use of same to treat cancer
TW202221034A (zh) 抗cd228抗體和抗體-藥物共軛體
TWI591176B (zh) 抗liv-1之人化抗體類及彼等於治療癌症上之用途

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: SEAGEN INC., WASHINGTON

Free format text: CHANGE OF NAME;ASSIGNOR:SEATTLE GENETICS, INC.;REEL/FRAME:054102/0821

Effective date: 20201006

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8