US20050276812A1 - Antibody-drug conjugates and methods - Google Patents

Antibody-drug conjugates and methods Download PDF

Info

Publication number
US20050276812A1
US20050276812A1 US11/141,344 US14134405A US2005276812A1 US 20050276812 A1 US20050276812 A1 US 20050276812A1 US 14134405 A US14134405 A US 14134405A US 2005276812 A1 US2005276812 A1 US 2005276812A1
Authority
US
United States
Prior art keywords
antibody
genbank accession
receptor
drug conjugate
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/141,344
Other languages
English (en)
Inventor
Allen Ebens
Frederic Jacobson
Paul Polakis
Ralph Schwall
Mark Sliwkowski
Susan Spencer
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.)
Genentech Inc
Original Assignee
Genentech 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=35262141&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20050276812(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to US11/141,344 priority Critical patent/US20050276812A1/en
Assigned to GENENTECH, INC. reassignment GENENTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JACOBSON, FREDERIC S., SLIWKOWSKI, MARK X., SCHWALL, RALPH H., SPENCER, SUSAN D., POLAKIS, PAUL, EBENS, ALLEN J. JR.
Publication of US20050276812A1 publication Critical patent/US20050276812A1/en
Priority to US11/564,171 priority patent/US7754441B2/en
Priority to US12/052,938 priority patent/US8652479B2/en
Priority to US12/326,721 priority patent/US8142784B2/en
Priority to US14/149,979 priority patent/US20140128580A1/en
Priority to US15/406,583 priority patent/US20170362318A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • A61K47/6875Medicinal 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 being a hybrid immunoglobulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/18Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • C07K16/1045Lentiviridae, e.g. HIV, FIV, SIV
    • C07K16/1063Lentiviridae, e.g. HIV, FIV, SIV env, e.g. gp41, gp110/120, gp160, V3, PND, CD4 binding site
    • 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
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against Fc-receptors, e.g. CD16, CD32, CD64
    • 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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • 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
    • C07K16/3046Stomach, Intestines
    • 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
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • 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

Definitions

  • the invention relates generally to compounds with anti-cancer activity and more specifically to antibodies conjugated with chemotherapeutic maytansinoid drugs or toxins.
  • the invention also relates to methods of using antibody-drug conjugate compounds for in vitro, in situ, and in vivo diagnosis or treatment of mammalian cells, or associated pathological conditions.
  • Antibody therapy has been established for the targeted treatment of patients with cancer, immunological and angiogenic disorders.
  • ADC antibody-drug conjugates
  • cytotoxic or cytostatic agents i.e. drugs to kill or inhibit tumor cells in the treatment of cancer
  • cytotoxic or cytostatic agents i.e. drugs to kill or inhibit tumor cells in the treatment of cancer
  • Efforts to design and refine ADC have focused on the selectivity of monoclonal antibodies (mAbs) as well as drug-linking and drug-releasing properties. Both polyclonal antibodies and monoclonal antibodies have been reported as useful in these strategies (Rowland et al., (1986) Cancer Immunol. Immunother. 21:183-87). Drugs used in these methods include daunomycin, doxorubicin, methotrexate, mitomycin, neocarzinostatin (Takahashi et al (1988) Cancer 61:881-888) and vindesine (Rowland et al., (1986) supra).
  • Toxins used in antibody-toxin conjugates include bacterial toxins such as diphtheria toxin, plant toxins such as ricin (U.S. Pat. No. 4,753,894; U.S. Pat. No. 5,629,197; U.S. Pat. No. 4,958,009; U.S. Pat. No. 4,956,453), small molecule toxins such as geldanamycin (Mandler et al (2000) J. of the Nat. Cancer Inst. 92(19):1573-1581; Mandler et al (2000) Bioorganic & Med. Chem. Letters 10: 1025-1028; Mandler et al (2002) Bioconjugate Chem.
  • bacterial toxins such as diphtheria toxin
  • plant toxins such as ricin
  • ricin U.S. Pat. No. 4,753,894; U.S. Pat. No. 5,629,197; U.S. Pat. No. 4,958,009; U.S.
  • cytotoxic drugs may effect their cytotoxic and cytostatic effects by mechanisms including tubulin binding, DNA binding, or topoisomerase inhibition. Some cytotoxic drugs tend to be inactive or less active when conjugated to large antibodies or protein receptor ligands.
  • ZEVALIN® ibritumomab tiuxetan, Biogen/Idec
  • ZEVALIN® ibritumomab tiuxetan, Biogen/Idec
  • MYLOTARGTM (gemtuzumab ozogamicin, Wyeth Pharmaceuticals), an antibody-drug conjugate composed of a hu CD33 antibody linked to calicheamicin, was approved in 2000 for the treatment of acute myeloid leukemia by injection (Drugs of the Future (2000) 25(7):686; U.S. Pat. No. 4,970,198; U.S. Pat. No. 5,079,233; U.S. Pat. No. 5,585,089; U.S. Pat. No. 5,606,040; U.S. Pat. No. 5,693,762; U.S. Pat. No. 5,739,116; U.S. Pat. No.
  • Cantuzumab mertansine (Immunogen, Inc.), an antibody-drug conjugate composed of the huC242 antibody linked via the disulfide linker SPP to the maytansinoid drug moiety, DM1 (Xie et al (2004) J. of Pharm. and Exp. Ther. 308(3):1073-1082; Tolcher et al (2003) J. Clin. Oncology 21(2):211-222; U.S. Pat. No. 5,208,020), underwent Phase I trials for the treatment of cancers that express CanAg, such as colon, pancreatic, gastric, and others.
  • CanAg such as colon, pancreatic, gastric, and others.
  • MLN-2704 (Millennium Pharm., BZL Biologics, Immunogen Inc.) is an antibody-drug conjugate composed of the anti-prostate specific membrane antigen (PSMA) monoclonal antibody linked to the maytansinoid drug moiety, DM1, under development for the potential treatment of prostate tumors.
  • PSMA anti-prostate specific membrane antigen
  • DM1 maytansinoid drug moiety
  • SMCC non-disulfide linker
  • TA.1 mouse murine monoclonal antibody
  • SMCC linker was considered therein to be “noncleavable” (also, see: U.S. Pat. No. 4,981,979).
  • HERCEPTIN® tacuzumab linked by SMCC to DM1 has been reported (WO 2005/037992).
  • tumor-associated polypeptides that are specifically expressed on the surface of one or more particular type(s) of cancer cell as compared to on one or more normal non-cancerous cell(s). Often, such tumor-associated polypeptides are more abundantly expressed on the surface of the cancer cells as compared to on the surface of the non-cancerous cells.
  • TAA tumor-associated antigens
  • HERCEPTIN® tacuzumab
  • HER2 ErbB2
  • Trastuzumab is an IgG1 kappa antibody that contains human framework regions with the complementarity-determining regions of a murine antibody (4D5) that binds to HER2. Trastuzumab binds to the HER2 antigen and thus inhibits the growth of cancerous cells. Because trastuzumab is a humanized antibody, it minimizes any HAMA response in patients. The humanized antibody against HER2 is produced by a mammalian cell (Chinese Hamster Ovary, CHO) suspension culture.
  • the HER2 (or c-erbB2) proto-oncogene encodes a transmembrane receptor protein of 185 kDa, which is structurally related to the epidermal growth factor receptor.
  • HER2 protein overexpression is observed in 25%-30% of primary breast cancers and can be determined using an immunohistochemistry based assessment of fixed tumor blocks (Press M F, et al (1993) Cancer Res 53:4960-70.
  • Trastuzumab has been shown, in both in vitro assays and in animals, to inhibit the proliferation of human tumor cells that overexpress HER2 (Hudziak R M, et al (1989) Mol Cell Biol 9:1165-72; Lewis G D, et al (1993) Cancer Immunol Immunother; 37:255-63; Baselga J, et al (1998) Cancer Res. 58:2825-2831).
  • Trastuzumab is a mediator of antibody-dependent cellular cytotoxicity, ADCC (Hotaling T E, et al (1996) [abstract]. Proc. Annual Meeting Am Assoc Cancer Res; 37:471; Pegram M D, et al (1997) [abstract].
  • HERCEPTIN® is clinically active in patients with ErbB2-overexpressing metastatic breast cancers that have received extensive prior anti-cancer therapy (Baselga et al, (1996) J. Clin. Oncol. 14:737-744).
  • HERCEPTIN is a breakthrough in treating patients with ErbB2-overexpressing breast cancers that have received extensive prior anti-cancer therapy, the majority of the patients in this population fail to respond or respond only poorly to HERCEPTIN treatment. Therefore, there is a significant clinical need for developing further HER2-directed cancer therapies for those patients with HER2-overexpressing tumors or other diseases associated with HER2 expression that do not respond, or respond poorly, to HERCEPTIN treatment. In addition to HER2, there is an opportunity to exploit other tumor-associated antigens with targeted therapies.
  • the present invention provides novel compounds with biological activity against cancer cells.
  • the compounds may inhibit tumor growth in mammals and may be useful for treating human cancer patients.
  • the present invention relates to the delivery, transport, accumulation or retention of therapeutic antibody-drug conjugate (ADC) compounds inside cells.
  • ADC therapeutic antibody-drug conjugate
  • the invention is more particularly related to attaining high concentrations of active metabolite molecules in cancer cells.
  • Intracellular targeting may be achieved by methods and compounds which allow accumulation or retention of biologically active agents inside cells. Such effective targeting may be applicable to a variety of therapeutic formulations and procedures.
  • antibody-drug conjugates with stable, non-disulfide linkers groups that attach a maytansinoid drug moiety to an antibody result in increased in vitro potency and in vivo efficacy.
  • the antibody-drug conjugates show the unexpected result of better safety in vivo relative to certain disulfide linker conjugates.
  • Antibody-drug conjugate (ADC) compounds comprise an antibody covalently attached by a linker to one or more maytansinoid drug moieties.
  • ADC may be represented by Formula I: Ab-(L-D) p I
  • a substantial amount of the drug moiety is not cleaved from the antibody until the antibody-drug conjugate enters a cell with a cell-surface receptor specific for the antibody of the antibody-drug conjugate, and the drug moiety is cleaved from the antibody when the antibody-drug conjugate does enter the cell.
  • the ADC specifically binds to a receptor encoded by an ErbB gene, such as EGFR, HER2, HER3 and HER4.
  • the ADC may specifically bind to the extracellular domain of the HER2 receptor.
  • the ADC may inhibit growth of tumor cells which overexpress HER2 receptor.
  • the antibody (Ab) of Formula I is a humanized antibody such as huMAb4D5-1, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 or huMAb4D5-8 (trastuzumab).
  • Another aspect of the invention is a pharmaceutical composition including a Formula I compound, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable diluent, carrier, or excipient.
  • Another aspect provides a pharmaceutical combination comprising a Formula I compound and a second compound having anti-cancer properties or other therapeutic effects.
  • Another aspect includes diagnostic and therapeutic uses for the compounds and compositions disclosed herein.
  • Another aspect is a method for killing or inhibiting the proliferation of tumor cells or cancer cells comprising treating the cells with an amount of an antibody-drug conjugate, or a pharmaceutically acceptable salt or solvate thereof, being effective to kill or inhibit the proliferation of the tumor cells or cancer cells.
  • Another aspect are methods of treating cancer comprising administering to a patient a formulation of a Formula I compound.
  • One method is for the treatment of cancer in a mammal, wherein the cancer is characterized by the overexpression of an ErbB receptor.
  • the mammal optionally does not respond, or responds poorly, to treatment with an unconjugated anti-ErbB antibody.
  • the method comprises administering to the mammal a therapeutically effective amount of an antibody-drug conjugate compound.
  • Another aspect is a method of inhibiting the growth of tumor cells that overexpress a growth factor receptor selected from the group consisting of HER2 receptor and EGF receptor comprising administering to a patient an antibody-drug conjugate compound which binds specifically to said growth factor receptor and a chemotherapeutic agent wherein said antibody-drug conjugate and said chemotherapeutic agent are each administered in amounts effective to inhibit growth of tumor cells in the patient.
  • a growth factor receptor selected from the group consisting of HER2 receptor and EGF receptor
  • Another aspect is a method for the treatment of a human patient susceptible to or diagnosed with a disorder characterized by overexpression of ErbB2 receptor, comprising administering a combination of an antibody-drug conjugate compound of Formula I and a chemotherapeutic agent.
  • Another aspect is an assay method for detecting cancer cells comprising: exposing cells to an antibody-drug conjugate compound, and determining the extent of binding of the antibody-drug conjugate compound to the cells.
  • Another aspect concerns methods of screening ADC drug candidates for the treatment of a disease or disorder where the disease or disorder is characterized by the overexpression of HER2.
  • kits comprising an antibody-drug conjugate, a container, and a package insert or label indicating a treatment.
  • Another aspect includes methods of treating a disease or disorder characterized by the overexpression of HER2 in a patient with the antibody-drug conjugate compounds.
  • Another aspect includes methods of making, methods of preparing, methods of synthesis, methods of conjugation, and methods of purification of the antibody-drug conjugate compounds, and the intermediates for the preparation, synthesis, and conjugation of the antibody-drug conjugate compounds.
  • FIG. 1 shows an in vitro, cell proliferation assay with SK-BR-3 cells treated with antibody-drug conjugates: - ⁇ - trastuzumab-SPP-DM1, - ⁇ - trastuzumab-SPDP-DM1, and -o- trastuzumab-SMCC-DM1.
  • FIG. 2 shows an in vitro, cell proliferation assay with BT-474 cells treated with antibody-drug conjugates: - ⁇ - trastuzumab-SPP-DM1, - ⁇ - trastuzumab-SPDP-DM1, and -o- trastuzumab-SMCC-DM1.
  • FIG. 3 shows an in vitro, cell proliferation assay with MCF7 cells treated with antibody-drug conjugates: - ⁇ - trastuzumab-SPP-DM1, - ⁇ - trastuzumab-SPDP-DM1, and -o- trastuzumab-SMCC-DM1.
  • FIG. 4 shows an in vitro, cell proliferation assay with MDA-MB-468 cells treated with antibody-drug conjugates: - ⁇ - trastuzumab-SPP-DM1, - ⁇ - trastuzumab-SPDP-DM1, and -o- trastuzumab-SMCC-DM1.
  • FIG. 5 shows the serum clearance in beige nude mice without tumors of trastuzumab-SMCC-DM1 vs. trastuzumab-SPP-DM1, measuring conjugate and total antibody serum concentration at six time points (5 minutes, 1 hour, 6 hours, 24 hours, 72, 168 hours post-dose) over 7 days.
  • FIG. 6 shows the stability over time in nude mice without tumors of the conjugates: trastuzumab-SPDP-DM1, trastuzumab-SPP-DM1, trastuzumab-SPP-DM3, trastuzumab-SPP-DM4, and trastuzumab-SMCC-DM1, measuring serum concentration at six time points (5 minutes, 1 hour, 6 hours, 24 hours, 72, 168 hours post-dose) over 7 days.
  • FIG. 7 shows the measurement of serum concentrations of total trastuzumab/trastuzumab-SMCC-DM1, and total trastuzumab/trastuzumab-SPP-DM1 in mice, 7 days after treatment, with and without tumor.
  • FIG. 9 shows a plasma concentration clearance study after administration of 10 mg/kg of trastuzumab-SMCC-DM1 to 4 subject rats. Concentrations of total antibody and trastuzumab-SMCC-DM1 were measured.
  • FIG. 10 shows the mean tumor volume change over time in mice dosed with: Vehicle (PBS pH 6.5), trastuzumab-SPP-DM1 (370 ⁇ g DM1/m 2 ), and trastuzumab-SMCC-DM1 (330 ⁇ g DM1/m 2 ) where dose refers to the dose of DM1 administered.
  • Vehicle PBS pH 6.5
  • trastuzumab-SPP-DM1 370 ⁇ g DM1/m 2
  • trastuzumab-SMCC-DM1 330 ⁇ g DM1/m 2
  • FIG. 11 shows the mean tumor volume change over time in athymic nude mice with Fo5 tumor allografts dosed on Day 0 with: Vehicle (PBS pH 6.5), 10 mg/kg trastuzumab-SIAB-DM1 (3.4 DM1/Ab; 168 ⁇ g DM1/kg), and 10 mg/kg trastuzumab-SMCC-DM1 (3.2 DM1/Ab; 158 ⁇ g DM1/kg) where dose refers to the dose amount of the antibody-drug conjugate administered.
  • Vehicle PBS pH 6.5
  • trastuzumab-SIAB-DM1 3.4 DM1/Ab; 168 ⁇ g DM1/kg
  • trastuzumab-SMCC-DM1 3.2 DM1/Ab; 158 ⁇ g DM1/kg
  • FIG. 13 shows the time to double tumor volume and log cell kill analysis for Vehicle (PBS pH 6.5), trastuzumab-SPP-DM1, trastuzumab-SPP-DM4, trastuzumab-SPP-DM3, and trastuzumab-SMCC-DM1 in HER2-Fo5 tumors.
  • FIG. 14 shows the change in body weight over time of rats dosed with: Vehicle (10 mM sodium succinate, 100 mg/mL sucrose, 0.1% Tween 20, pH 5.0), trastuzumab-SPP-DM1 (1860 ⁇ g DM1/m ), trastuzumab-SMCC-DM1 (1860 ⁇ g DM1/m 2), trastuzumab-SMCC-DM1 (3260 ⁇ g DM1/m 2), and free DM1 (650 ⁇ g/m 2 ).
  • Vehicle 10 mM sodium succinate, 100 mg/mL sucrose, 0.1% Tween 20, pH 5.0
  • trastuzumab-SPP-DM1 1860 ⁇ g DM1/m
  • trastuzumab-SMCC-DM1 1860 ⁇ g DM1/m 2
  • trastuzumab-SMCC-DM1 1860 ⁇ g DM1/m 2
  • free DM1 650 ⁇ g/m 2
  • FIG. 15 shows a liver function test measured in AST units per liter over time in the rat model dosed with: Vehicle (10 mM sodium succinate, 100 mg/mL sucrose, 0.1% Tween 20, pH 5.0), trastuzumab-SPP-DM1 (22.3 mg/kg), trastuzumab-SMCC-DM1 (10 mg/kg), trastuzumab-SMCC-DM1 (25 mg/kg), trastuzumab-SMCC-DM1 (50 mg/kg), and free DM1.
  • Vehicle 10 mM sodium succinate, 100 mg/mL sucrose, 0.1% Tween 20, pH 5.0
  • trastuzumab-SPP-DM1 22.3 mg/kg
  • trastuzumab-SMCC-DM1 10 mg/kg
  • trastuzumab-SMCC-DM1 25 mg/kg
  • trastuzumab-SMCC-DM1 50 mg/kg
  • free DM1 free DM1.
  • FIG. 16 shows a safety profile measured in PLT units in cells per liter over time in the rat model dosed with: Vehicle (10 mM sodium succinate, 100 mg/mL sucrose, 0.1% Tween 20, pH 5.0), trastuzumab-SPP-DM1 (22.3 mg/kg), trastuzumab-SMCC-DM1 (10 mg/kg), trastuzumab-SMCC-DM1 (25 mg/kg), trastuzumab-SMCC-DM1 (50 mg/kg), and free DM1.
  • Vehicle 10 mM sodium succinate, 100 mg/mL sucrose, 0.1% Tween 20, pH 5.0
  • trastuzumab-SPP-DM1 22.3 mg/kg
  • trastuzumab-SMCC-DM1 10 mg/kg
  • trastuzumab-SMCC-DM1 25 mg/kg
  • trastuzumab-SMCC-DM1 50 mg/kg
  • free DM1 free DM1.
  • FIG. 17 shows an in vitro, cell proliferation assay with HT1080EphB2 (C8) cells treated with antibody-drug conjugates: - ⁇ - antiEphB2R 2H9-SPP-DM1, and - ⁇ - antiEphB2R 2H9-SMCC-DM1.
  • antibody herein is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments, so long as they exhibit the desired biological activity (Miller et al (2003) Jour. of Immunology 170:4854-4861). Antibodies may be murine, human, humanized, chimeric, or derived from other species. An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen. (Janeway, C., Travers, P., Walport, M., Shlomchik (2001) Immuno Biology, 5 th Ed ., Garland Publishing, New York).
  • a target antigen generally has numerous binding sites, also called epitopes, recognized by CDRs on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody.
  • An antibody includes a full-length immunoglobulin molecule or an immunologically active portion of a full-length immunoglobulin molecule, i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease.
  • the immunoglobulin disclosed herein can be of any type (e.g., IgG, IgE, IgM, IgD, and IgA), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule.
  • the immunoglobulins can be derived from any species. In one aspect, however, the immunoglobulin is of human, murine, or rabbit origin.
  • Antibody fragments comprise a portion of a full length antibody, generally the antigen binding or variable region thereof.
  • Examples of antibody fragments include Fab, Fab′, F(ab′) 2 , and Fv fragments; diabodies; linear antibodies; fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, CDR (complementary determining region), and epitope-binding fragments of any of the above which immunospecifically bind to cancer cell antigens, viral antigens or microbial antigens, single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al (1975) Nature 256:495, or may be made by recombinant DNA methods (see, U.S. Pat. No. 4,816,567).
  • the monoclonal antibodies may also be isolated from phage antibody libraries using the techniques described in Clackson et al (1991) Nature, 352:624-628; Marks et al (1991) J. Mol. Biol., 222:581-597; for example.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al (1984) Proc. Natl. Acad. Sci. USA, 81:6851-6855).
  • Chimeric antibodies of interest herein include “primatized” antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey or Ape) and human constant region sequences.
  • an “intact antibody” herein is one comprising a VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1, CH2 and CH3.
  • the constant domains may be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variant thereof.
  • the intact antibody may have one or more “effector functions” which refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.
  • intact antibodies can be assigned to different “classes.” There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into “subclasses” (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • ErbB receptor is a receptor protein tyrosine kinase which belongs to the ErbB receptor family which are important mediators of cell growth, differentiation and survival.
  • the ErbB receptor family includes four distinct members including epidermal growth factor receptor (EGFR, ErbB1, HER1), HER2 (ErbB2 or p185 neu ), HER3 (ErbB3) and HER4 (ErbB4 or tyro2).
  • EGFR epidermal growth factor receptor
  • HER2 ErbB2 or p185 neu
  • HER3 ErbB3
  • HER4 ErbB4 or tyro2
  • a panel of anti-ErbB2 antibodies has been characterized using the human breast tumor cell line SKBR3 (Hudziak et al (1989) Mol. Cell. Biol. 9(3):1165-1172. Maximum inhibition was obtained with the antibody called 4D5 which inhibited cellular proliferation by 56%.
  • the ErbB receptor will generally comprise an extracellular domain, which may bind an ErbB ligand; a lipophilic transmembrane domain; a conserved intracellular tyrosine kinase domain; and a carboxyl-terminal signaling domain harboring several tyrosine residues which can be phosphorylated.
  • the ErbB receptor may be a “native sequence” ErbB receptor or an “amino acid sequence variant” thereof.
  • the ErbB receptor may be native sequence human ErbB receptor. Accordingly, a “member of the ErbB receptor family” is EGFR (ErbB 1), ErbB2, ErbB3, ErbB4 or any other ErbB receptor currently known or to be identified in the future.
  • ErbB1 refers to EGFR as disclosed, for example, in Carpenter et al (1987) Ann. Rev. Biochem. 56:881-914, including naturally occurring mutant forms thereof (e.g., a deletion mutant EGFR as in Humphrey et al., (1990) PNAS (USA), 87:4207-4211).
  • the term erbB1 refers to the gene encoding the EGFR protein product. Antibodies against HER1 are described, for example, in Murthy et al (1987) Arch. Biochem. Biophys., 252:549-560 and in WO 95/25167.
  • ERRP epidermal growth factor receptor
  • EGF epidermal growth factor receptor
  • ErbB2 and HER2 are used interchangeably herein and refer to human HER2 protein described, for example, in Semba et al (1985) PNAS (USA), 82:6497-6501 and Yamamoto et al (1986) Nature, 319:230-234 (Genbank accession number X03363).
  • the term “erbB2” refers to the gene encoding human ErbB2 and “neu” refers to the gene encoding rat p185neu.
  • ErbB3 and “HER3” refer to the receptor polypeptide as disclosed, for example, in U.S. Pat. No. 5,183,884; U.S. Pat. No. 5,480,968; Kraus et al (1989) PNAS (USA) 86:9193-9197.
  • Antibodies against ErbB3 are known in the art (U.S. Pat. No. 5,183,884; U.S. Pat. No. 5,480,968; WO 97/35885).
  • ErbB4 and HER4 herein refer to the receptor polypeptide as disclosed, for example, in EP Pat Appln No 599,274; Plowman et al., Proc. Natl. Acad. Sci. USA, 90:1746-1750 (1993); and Plowman et al., Nature, 366:473-475 (1993), including isoforms thereof, e.g., as disclosed in WO 99/19488.
  • Antibodies against HER4 are described, for example, in WO 02/18444.
  • Antibodies to ErbB receptors are available commercially from a number of sources, including, for example, Santa Cruz Biotechnology, Inc., California, USA.
  • ErbB ligand is meant a polypeptide which binds to and/or activates an ErbB receptor.
  • the ErbB ligand may be a native sequence human ErbB ligand such as epidermal growth factor (EGF) (Savage et al (1972) J. Biol.
  • TGF- ⁇ transforming growth factor alpha
  • Amphiregulin also known as schwanoma or keratinocyte autocrine growth factor
  • betacellulin Shing et al (1993) Science 259:1604-1607; and Sasada et al (1993) Biochem. Biophys. Res. Commun.
  • HB-EGF heparin-binding epidermal growth factor
  • epiregulin Toyoda et al (1995) J. Biol. Chem. 270:7495-7500; and Komurasaki et al (1997) Oncogene 15:2841-2848); a heregulin (see below); neuregulin-2 (NRG-2) (Carraway et al., Nature, 387:512-516 (1997)); neuregulin-3 (NRG-3) (Zhang et al (1997) Proc. Natl. Acad.
  • ErbB ligands which bind EGFR include EGF, TGF- ⁇ , amphiregulin, betacellulin, HB-EGF and epiregulin.
  • ErbB ligands which bind ErbB3 include heregulins.
  • ErbB ligands capable of binding ErbB4 include betacellulin, epiregulin, HB-EGF, NRG-2, NRG-3, NRG-4 and heregulins.
  • the ErbB ligand may also be a synthetic ErbB ligand.
  • the synthetic ligand may be specific for a particular ErbB receptor, or may recognize particular ErbB receptor complexes.
  • An example of a synthetic ligand is the synthetic heregulin/EGF chimera biregulin (see, for example, Jones et al (1999) FEBS Letters, 447:227-231, which is incorporated by reference).
  • Heregulin refers to a polypeptide encoded by the heregulin gene product as disclosed in U.S. Pat. No. 5,641,869 or Marchionni et al (1993) Nature 362:312-318.
  • Examples of heregulins include heregulin- ⁇ , heregulin- ⁇ 1, heregulin- ⁇ 2 and heregulin- ⁇ 3 (Holmes et al (1992) Science 256:1205-1210; and U.S. Pat. No.
  • the term includes biologically active fragments and/or amino acid sequence variants of a native sequence HRG polypeptide, such as an EGF-like domain fragment thereof (e.g., HRG ⁇ 1177-244).
  • ErbB hetero-oligomer is a noncovalently associated oligomer comprising at least two different ErbB receptors.
  • An “ErbB dimer” is a noncovalently associated oligomer that comprises two different ErbB receptors. Such complexes may form when a cell expressing two or more ErbB receptors is exposed to an ErbB ligand.
  • ErbB oligomers, such as ErbB dimers can be isolated by immunoprecipitation and analyzed by SDS-PAGE as described in Sliwkowski et al (1994) J. Biol. Chem., 269(20):14661-14665, for example.
  • ErbB hetero-oligomers examples include EGFR-ErbB2 (also referred to as HER1/HER2), ErbB2-ErbB3 (HER2/HER3) and ErbB3-ErbB4 (HER3/HER4) complexes.
  • the ErbB hetero-oligomer may comprise two or more ErbB2 receptors combined with a different ErbB receptor, such as ErbB3, ErbB4 or EGFR (ErbB1).
  • Other proteins, such as a cytokine receptor subunit e.g., gp130
  • gp130 cytokine receptor subunit
  • ligand activation of an ErbB receptor is meant signal transduction (e.g., that caused by an intracellular kinase domain of an ErbB receptor phosphorylating tyrosine residues in the ErbB receptor or a substrate polypeptide) mediated by ErbB ligand binding to a ErbB hetero-oligomer comprising the ErbB receptor of interest.
  • this will involve binding of an ErbB ligand to an ErbB hetero-oligomer which activates a kinase domain of one or more of the ErbB receptors in the hetero-oligomer and thereby results in phosphorylation of tyrosine residues in one or more of the ErbB receptors and/or phosphorylation of tyrosine residues in additional substrate polypeptides(s).
  • ErbB receptor activation can be quantified using various tyrosine phosphorylation assays.
  • a “native sequence” polypeptide is one which has the same amino acid sequence as a polypeptide (e.g., ErbB receptor or ErbB ligand) derived from nature.
  • a polypeptide e.g., ErbB receptor or ErbB ligand
  • Such native sequence polypeptides can be isolated from nature or can be produced by recombinant or synthetic means.
  • a native sequence polypeptide can have the amino acid sequence of naturally occurring human polypeptide, murine polypeptide, or polypeptide from any other mammalian species.
  • amino acid sequence variant refers to polypeptides having amino acid sequences that differ to some extent from a native sequence polypeptide. Ordinarily, amino acid sequence variants will possess at least about 70% sequence identity with at least one receptor binding domain of a native ErbB ligand or with at least one ligand binding domain of a native ErbB receptor, or at least about 80%, or at least about 90% homologous with such receptor or ligand binding domains. The amino acid sequence variants possess substitutions, deletions, and/or insertions at certain positions within the amino acid sequence of the native amino acid sequence.
  • Sequence identity is defined as the percentage of residues in the amino acid sequence variant that are identical after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Methods and computer programs for the alignment are well known in the art. One such computer program is “Align 2,” authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, D.C. 20559, on Dec. 10, 1991.
  • Antibody-dependent cell-mediated cytotoxicity and “ADCC” refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g., Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcRs Fc receptors
  • FcR expression on hematopoietic cells in summarized is Table 3 on page 464 of Ravetch and Kinet, (1991) Annu. Rev. Immunol, 9:457-92.
  • ADCC activity of a molecule of interest may be assessed in vitro, e.g., in a animal model such as that disclosed in Clynes et al (1998) PNAS (USA), 95:652-656.
  • Maytansinoid drug moiety means the substructure of an antibody-drug conjugate that has the structure of a maytansine compound. Maytansine was first isolated from the east African shrub Maytenus serrata (U.S. Pat. No. 3,896,111). Subsequently, it was discovered that certain microbes also produce maytansinoids, such as maytansinol and C-3 maytansinol esters (U.S. Pat. No. 4,151,042). Synthetic maytansinol and maytansinol analogues have been reported. See U.S. Pat. Nos.
  • Fc receptor or “FcR” are used to describe a receptor that binds to the Fc region of an antibody, such as a native sequence human FcR.
  • FcR may bind an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIA (an “activating receptor”) and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor Fc ⁇ RIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain.
  • Inhibiting receptor Fc ⁇ RIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain.
  • ITAM immunoreceptor tyrosine-based activation motif
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • FcR FcR
  • FcRn neonatal receptor
  • “Complement dependent cytotoxicity” or “CDC” refers to the ability of a molecule to lyse a target in the presence of complement.
  • the complement activation pathway is initiated by the binding of the first component of the complement system (C1q) to a molecule (e.g., an antibody) complexed with a cognate antigen.
  • a CDC assay e.g., as described in Gazzano-Santoro et al (1996) J. Immunol. Methods, 202:163, may be performed.
  • “Native antibodies” are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end. The constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light-chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains.
  • variable refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FRs).
  • the variable domains of native heavy and light chains each comprise four FRs, largely adopting a ⁇ -sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.).
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody dependent cellular cytotoxicity (ADCC).
  • hypervariable region when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region generally comprises amino acid residues from a “complementarity determining region” or “CDR” (e.g., residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al supra) and/or those residues from a “hypervariable loop” (e.g., residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk (1987) J. Mol. Biol., 196:901-917).
  • “Framework Region” or “FR” residues are those variable domain residue
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual “Fc” fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab′)2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen.
  • “Fv” is the minimum antibody fragment which contains a complete antigen-recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six hypervariable regions confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • the Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain.
  • Fab′ fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region.
  • Fab′-SH is the designation herein for Fab′ in which the cysteine residue(s) of the constant domains bear at least one free thiol group.
  • F(ab′)2 antibody fragments originally were produced as pairs of Fab′ fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • the “light chains” of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequences of their constant domains.
  • Single-chain Fv or “scFv” mean single chain variable region antibody fragments which comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide may further comprise a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding (Plückthun in The Pharmacology of Monoclonal Antibodies , vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994).
  • Anti-ErbB2 antibody scFv fragments are described in WO 93/16185; U.S. Pat. No. 5,571,894; U.S. Pat. No. 5,587,458.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a variable heavy domain (VH) connected to a variable light domain (VL) in the same polypeptide chain (VH-VL).
  • VH variable heavy domain
  • VL variable light domain
  • VH-VL variable light domain
  • linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448.
  • “Humanized” forms of non-human (e.g., rodent) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • Humanized anti-ErbB2 antibodies include huMAb4D5-1, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (HERCEPTIN®, trastuzumab) as described in Table 3 of U.S. Pat. No. 5,821,337 expressly incorporated herein by reference; humanized 520C9 (WO 93/21319) and humanized 2C4 antibodies.
  • an “isolated” antibody is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the antibody may be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, or more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup protein sequencer, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • an antibody “which binds” an antigen of interest is one capable of binding that antigen with sufficient affinity such that the antibody is useful in targeting a cell expressing the antigen.
  • the antibody is one which binds ErbB2
  • it will usually preferentially bind ErbB2 as opposed to other ErbB receptors, and may be one which does not significantly cross-react with other proteins such as EGFR, ErbB3 or ErbB4.
  • the extent of binding of the antibody to these non-ErbB2 proteins e.g., cell surface binding to endogenous receptor
  • FACS fluorescence activated cell sorting
  • RIA radioimmunoprecipitation
  • the anti-ErbB2 antibody will not significantly cross-react with the rat neu protein, e.g., as described in Schecter et al (1984) Nature 312:513, and Drebin et al (1984) Nature, 312:545-548.
  • an antibody which “blocks” ligand activation of an ErbB receptor reduces or prevents such activation, wherein the antibody is able to block ligand activation of the ErbB receptor substantially more effectively than monoclonal antibody 4D5, e.g., about as effectively as monoclonal antibodies 7F3 or 2C4 or Fab fragments thereof.
  • the antibody that blocks ligand activation of an ErbB receptor may be one which is about 50-100% more effective than 4D5 at blocking formation of an ErbB hetero-oligomer.
  • Blocking of ligand activation of an ErbB receptor can occur by any means, e.g., by interfering with: ligand binding to an ErbB receptor, ErbB complex formation, tyrosine kinase activity of an ErbB receptor in an ErbB complex and/or phosphorylation of tyrosine kinase residue(s) in or by an ErbB receptor.
  • An antibody having a “biological characteristic” of a designated antibody such as the monoclonal antibody designated 2C4 (Omnitarg, Genentech, Inc.), is one which possesses one or more of the biological characteristics of that antibody which distinguish it from other antibodies that bind to the same antigen (e.g., ErbB2).
  • an antibody with a biological characteristic of 2C4 may block HRG activation of an ErbB hetero-oligomer comprising ErbB2 and ErbB3, ErbB1 or ErbB4; block EGF, TGF- ⁇ , HB-EGF, epiregulin and/or amphiregulin activation of an ErbB receptor comprising EGFR and ErbB2; block EGF, TGF- ⁇ and/or HRG mediated activation of MAPK; and/or bind the same epitope in the extracellular domain of ErbB2 as that bound by 2C4 (e.g., which blocks binding of monoclonal antibody 2C4 to ErbB2).
  • the expression “monoclonal antibody 2C4” refers to an antibody that has antigen binding residues of, or derived from, the murine 2C4 antibody of the Examples below.
  • the monoclonal antibody 2C4 may be murine monoclonal antibody 2C4 or a variant thereof, such as humanized antibody 2C4, possessing antigen binding amino acid residues of murine monoclonal antibody 2C4 (WO 01/00245).
  • the expression “rhuMAb 2C4” when used herein refers to an antibody comprising the variable light (VL) and variable heavy (VH) sequences of SEQ ID Nos. 3 and 4, respectively, fused to human light and heavy IgG1 (non-A allotype) constant region sequences optionally expressed by a Chinese Hamster Ovary (CHO) cell (WO 01/00245).
  • the term “monoclonal antibody 4D5” refers to an antibody that has antigen binding residues of, or derived from, the murine 4D5 antibody (ATCC CRL 10463).
  • the monoclonal antibody 4D5 may be murine monoclonal antibody 4D5 or a variant thereof, such as a humanized 4D5, possessing antigen binding residues of murine monoclonal antibody 4D5.
  • Exemplary humanized 4D5 antibodies include huMAb4D5-1, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (HERCEPTIN®) as in U.S. Pat. No. 5,821,337.
  • a “growth inhibitory agent” refers to a compound or composition which inhibits growth of a cell, e.g. an ErbB expressing cancer cell either in vitro or in vivo.
  • the growth inhibitory agent may be one which significantly reduces the percentage of ErbB expressing cells in S phase.
  • growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest (The Molecular Basis of Cancer, Mendelsohn and Israel, eds., Chapter 1, entitled “Cell cycle regulation, oncogenes, and antineoplastic drugs” by Murakami et al. (W B Saunders: Philadelphia, 1995), especially p. 13).
  • growth inhibitory antibodies are those which bind to ErbB2 and inhibit the growth of cancer cells overexpressing ErbB2.
  • Growth inhibitory anti-ErbB2 antibodies may inhibit growth of SK-BR-3 breast tumor cells in cell culture by greater than 20%, or greater than 50% (e.g., from about 50% to about 100%) at an antibody concentration of about 0.5 to 30 ⁇ g/ml, where the growth inhibition is determined six days after exposure of the SK-BR-3 cells to the antibody (U.S. Pat. No. 5,677,171).
  • the cell is generally one which expresses the ErbB2 receptor, especially where the cell overexpresses the ErbB2 receptor.
  • the cell may be a cancer cell, e.g., a breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic or bladder cell. In vitro, the cell may be a SK-BR-3, BT474, Calu 3, MDA-MB-453, MDA-MB-361 or SKOV3 cell.
  • Cell death in vitro may be determined in the absence of complement and immune effector cells to distinguish cell death induced by antibody-dependent cell-mediated cytotoxicity (ADCC) or complement dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • the assay for cell death may be performed using heat inactivated serum (i.e., in the absence of complement) and in the absence of immune effector cells.
  • PI propidium iodide
  • trypan blue see Moore et al (1995) Cytotechnology, 17:1-11
  • 7AAD can be assessed relative to untreated cells.
  • Cell death-inducing antibodies are those which induce PI uptake in the PI uptake assay in BT474 cells (see below).
  • An antibody which “induces apoptosis” is one which induces programmed cell death as determined by binding of annexin V, fragmentation of DNA, cell shrinkage, dilation of endoplasmic reticulum, cell fragmentation, and/or formation of membrane vesicles (called apoptotic bodies).
  • the cell is usually one which overexpresses the ErbB2 receptor, including a tumor cell, e.g., a breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic or bladder cell.
  • the cell may be a SK-BR-3, BT474, Calu 3 cell, MDA-MB-453, MDA-MB-361 or SKOV3 cell.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • a “disorder” is any condition that would benefit from treatment of the present invention. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question.
  • disorders to be treated herein include benign and malignant tumors; leukemia and lymphoid malignancies, in particular breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic, prostate or bladder cancer; neuronal, glial, astrocytal, hypothalamic and other glandular, macrophagal, epithelial, stromal and blastocoelic disorders; and inflammatory, angiogenic and immunologic disorders.
  • An exemplary disorder to be treated in accordance with the present invention is a solid, malignant tumor
  • the term “therapeutically effective amount” refers to an amount of a drug effective to treat a disease or disorder in a mammal.
  • the therapeutically effective amount of the drug may: (i) reduce the number of cancer cells; (ii) reduce the tumor size; (iii) inhibit, retard, slow to some extent and preferably stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; and/or (vi) relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • efficacy may be assessed by physical measurements of the tumor during the course following administration of the ADC, and by determining partial and complete remission of tumor.
  • efficacy can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rate (RR).
  • bioavailability refers to the systemic availability (i.e., blood/plasma levels) of a given amount of drug administered to a patient. Bioavailability is an absolute term that indicates measurement of both the time (rate) and total amount (extent) of drug that reaches the general circulation from an administered dosage form.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small-cell lung cancer, non-small cell lung cancer (“NSCLC”), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, gastrointestinal stromal tumor (GIST), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • GIST gastrointestinal stromal tumor
  • An “ErbB-expressing cancer” is one comprising cells which have ErbB protein present at their cell surface.
  • An “ErbB2-expressing cancer” is one which produces sufficient levels of ErbB2 at the surface of cells thereof, such that an anti-ErbB2 antibody can bind thereto and have a therapeutic effect with respect to the cancer.
  • a cancer which “overexpresses” a receptor is one which has significantly higher levels of the receptor, such as ErbB2, at the cell surface thereof, compared to a noncancerous cell of the same tissue type.
  • a receptor e.g. an ErbB receptor
  • Such overexpression may be caused by gene amplification or by increased transcription or translation.
  • ErbB receptor overexpression may be determined in a diagnostic or prognostic assay by evaluating increased levels of the ErbB protein present on the surface of a cell (e.g., via an immunohistochemistry assay; IHC).
  • FISH fluorescent in situ hybridization
  • PCR polymerase chain reaction
  • Overexpression of the ErbB ligand may be determined diagnostically by evaluating levels of the ligand (or nucleic acid encoding it) in the patient, e.g., in a tumor biopsy or by various diagnostic assays such as the IHC, FISH, southern blotting, PCR or in vivo assays described above.
  • shed antigen e.g., ErbB extracellular domain
  • a detectable label e.g., a radioactive isotope
  • Overexpression of HER2 at the 3+ level which leads to ligand-independent activation of the tyrosine kinase (Hudziak et al., (1987) Proc. Natl. Acad. Sci.
  • a cancer which is “not characterized by overexpression of the ErbB2 receptor” is one which, in a diagnostic assay, does not express higher than normal levels of ErbB2 receptor compared to a noncancerous cell of the same tissue type.
  • the murine monoclonal anti-HER2 antibody inhibits the growth of breast cancer cell lines that overexpress HER2 at the 2+ and 3+ (1-2 ⁇ 10 6 HER2 receptors per cell) level, but has no activity on cells that express lower levels of HER2 (Lewis et al (1993) Cancer Immunol. Immunother. 37:255-263). Based on this observation, antibody 4D5 was humanized (huMAb4D5-8, rhuMAb HER2, U.S. Pat. No. 5,821,337; Carter et al (1992) Proc. Natl. Acad. Sci.
  • a “hormone independent” cancer is one in which proliferation thereof is not dependent on the presence of a hormone which binds to a receptor expressed by cells in the cancer. Such cancers do not undergo clinical regression upon administration of pharmacological or surgical strategies that reduce the hormone concentration in or near the tumor.
  • hormone independent cancers include androgen independent prostate cancer, estrogen independent breast cancer, endometrial cancer and ovarian cancer. Such cancers may begin as hormone dependent tumors and progress from a hormone-sensitive stage to a hormone-refractory tumor following anti-hormonal therapy.
  • cytotoxic agent refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells.
  • the term is intended to include radioactive isotopes (e.g., 211 At, 131 I, 125 I, 90 Y, 186 Re, 188 Re, 153 Sm, 212 Bi, 32 P, 60 C, and radioactive isotopes of Lu), chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including synthetic analogs and derivatives thereof.
  • radioactive isotopes e.g., 211 At, 131 I, 125 I, 90 Y, 186 Re, 188 Re, 153 Sm, 212 Bi, 32 P, 60 C, and radioactive isotopes of Lu
  • chemotherapeutic agents e.g., chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial,
  • a “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer.
  • chemotherapeutic agents include Erlotinib (TARCEVA®, Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millenium Pharm.), Fulvestrant (FASLODEX®, Astrazeneca), Sutent (SU11248, Pfizer), Letrozole (FEMARA®, Novartis), Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin®, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), Lapatinib (GSK572016, GlaxoSmithKline), Lonafarnib (SCH 66336), Sorafenib (BAY43-9006, Bayer Labs.), and Gefitini
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esor
  • chemotherapeutic agent include: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX® tamoxifen), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON.
  • SERMs selective estrogen receptor modulators
  • aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® megestrol acetate, AROMASIN® exemestane, formestanie, fadrozole, RIVISOR® vorozole, FEMARA® letrozole, and ARIMIDEX® anastrozole;
  • anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog);
  • aromatase inhibitors such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog
  • Protein kinase inhibitors include tyrosine kinase inhibitors which inhibit to some extent tyrosine kinase activity of a tyrosine kinase such as an ErbB receptor.
  • tyrosine kinase inhibitors include EGFR-targeted drugs such as: (i) antibodies which bind to EGFR, including MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see, U.S. Pat. No.
  • an “anti-angiogenic agent” refers to a compound which blocks, or interferes with to some degree, the development of blood vessels.
  • the anti-angiogenic factor may, for instance, be a small molecule or antibody that binds to a growth factor or growth factor receptor involved in promoting angiogenesis.
  • An exemplary anti-angiogenic agent is an antibody that binds to Vascular Endothelial Growth Factor (VEGF) such as bevacizumab (AVASTIN®, Genentech).
  • VEGF Vascular Endothelial Growth Factor
  • cytokine is a generic term for proteins released by one cell population which act on another cell as intercellular mediators.
  • cytokines are lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor- ⁇ and - ⁇ ; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF- ⁇ ; platelet-growth factor;
  • prodrug refers to a precursor or derivative form of a pharmaceutically active substance that is less cytotoxic to tumor cells compared to the parent drug and is capable of being enzymatically activated or converted into the more active parent form. See, e.g., Wilman, “Prodrugs in Cancer Chemotherapy” Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting Harbor (1986) and Stella et al., “Prodrugs: A Chemical Approach to Targeted Drug Delivery,” Directed Drug Delivery, Borchardt et al., (ed.), pp. 247-267, Humana Press (1985).
  • the prodrugs of this invention include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, P-lactam-containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, 5-fluorocytosine and other 5-fluorouridine prodrugs which can be converted into the more active cytotoxic free drug.
  • cytotoxic drugs that can be derivatized into a prodrug form for use in this invention include, but are not limited to, those chemotherapeutic agents described above.
  • a “liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug (such as the anti-ErbB2 antibodies disclosed herein and, optionally, a chemotherapeutic agent) to a mammal.
  • a drug such as the anti-ErbB2 antibodies disclosed herein and, optionally, a chemotherapeutic agent
  • the components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • a “cardioprotectant” is a compound or composition which prevents or reduces myocardial dysfunction (i.e., cardiomyopathy and/or congestive heart failure) associated with administration of a drug, such as an anthracycline antibiotic and/or an anti-ErbB2 antibody, to a patient.
  • the cardioprotectant may, for example, block or reduce a free-radical-mediated cardiotoxic effect and/or prevent or reduce oxidative-stress injury.
  • cardioprotectants encompassed by the present definition include the iron-chelating agent dexrazoxane (ICRF-187) (Seifert et al., The Annals of Pharmacotherapy, 28:1063-1072 (1994)); a lipid-lowering agent and/or anti-oxidant such as probucol (Singal et al., J. Mol.
  • amifostine (aminothiol 2-[(3-aminopropyl)amino]ethanethiol-dihydrogen phosphate ester, also called WR-2721, and the dephosphorylated cellular uptake form thereof called WR-1065) and S-3-(3-methylaminopropylamino)propylphosphorothioic acid (WR-151327), see Green et al., (1994) Cancer Research, 54:738-741; digoxin (Bristow, M. R. ed. (1980) Drug - Induced Heart Disease .
  • beta-blockers such as metoprolol (Hjalmarson et al (1994) Drugs 47:Suppl 4:31-9; and Shaddy et al (1995) Am. Heart J., 129:197-9); vitamin E; ascorbic acid (vitamin C); free radical scavengers such as oleanolic acid, ursolic acid and N-acetylcysteine (NAC); spin trapping compounds such as alpha-phenyl-tert-butyl nitrone (PBN); (Paracchini et al (1993) Anticancer Res., 13:1607-1612); selenoorganic compounds such as P251 (Elbesen); and the like.
  • an “isolated” nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the antibody nucleic acid.
  • An isolated nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the nucleic acid molecule as it exists in natural cells.
  • an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily express the antibody where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • Alkyl is C 1 -C 18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms.
  • alkyl radicals include C 1 -C 8 hydrocarbon moieties such as, but not limited to: methyl (Me, —CH 3 ), ethyl (Et, —CH 2 CH 3 ), 1-propyl (n-Pr, n-propyl, —CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, —CH(CH 3 ) 2 ), 1-butyl (n-Bu, n-butyl, —CH 2 CH 2 CH 2 CH 3 ), 2-methyl-1-propyl (i-Bu, i-butyl, —CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, —CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl,
  • Alkenyl is C 2 -C 18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp 2 double bond.
  • alkenyl radicals include C 2 -C 8 hydrocarbon moieties such as, but not limited to: ethylene or vinyl (—CH ⁇ CH 2 ), allyl (—CH 2 CH ⁇ CH 2 ), cyclopentenyl (—C 5 H 7 ), and 5-hexenyl (—CH 2 CH 2 CH 2 CH 2 CH ⁇ CH 2 )
  • Alkynyl is C 2 -C 18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond.
  • alkynyl radicals include C 2 -C 8 hydrocarbon moieties such as, but not limited to: acetylenic (—C ⁇ CH) and propargyl (—CH 2 C ⁇ CH),
  • Alkylene refers to a saturated, branched or straight chain or cyclic hydrocarbon radical of 1-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane.
  • Typical alkylene radicals include C 1 -C 8 hydrocarbon moieties such as, but are not limited to: methylene (—CH 2 —) 1,2-ethyl (—CH 2 CH 2 —), 1,3-propyl (—CH 2 CH 2 CH 2 —), 1,4-butyl (—CH 2 CH 2 CH 2 CH 2 —), and the like.
  • Alkenylene refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene.
  • Typical alkenylene radicals include C 2 -C 8 hydrocarbon moieties such as, but are not limited to: 1,2-ethylene (—CH ⁇ CH—).
  • Alkynylene refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne.
  • Typical alkynylene radicals include C 2 -C 8 hydrocarbon moieties such as, but are not limited to: acetylene (—C ⁇ C—), propargyl (—CH 2 C ⁇ C—), and 4-pentynyl (—CH 2 CH 2 CH 2 C ⁇ C—).
  • Aryl alone or in combination, means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system.
  • Aryl radical may contain one, two or three rings wherein such rings may be attached together in a pendent manner, e.g. biphenyl, or may be fused, e.g. napthalene or anthracene.
  • Some aryl groups are represented in the exemplary structures as “Ar”.
  • Typical aryl groups include C 6 -C 12 hydrocarbon moieties such as, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl, and the like.
  • Arylalkyl refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp 3 carbon atom, is replaced with an aryl radical.
  • Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, 2-naphthylethen-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like.
  • the arylalkyl group comprises 6 to 20 carbon atoms, e.g. the alkyl moiety, including alkanyl, alkenyl or alkynyl groups, of the arylalkyl group is 1 to 6 carbon atoms and the aryl moiety is 5 to 14 carbon atoms.
  • Heteroarylalkyl refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp 3 carbon atom, is replaced with a heteroaryl radical.
  • Typical heteroarylalkyl groups include, but are not limited to, 2-benzimidazolylmethyl, 2-furylethyl, and the like.
  • the heteroarylalkyl group comprises 6 to 20 carbon atoms, e.g.
  • the alkyl moiety, including alkanyl, alkenyl or alkynyl groups, of the heteroarylalkyl group is 1 to 6 carbon atoms and the heteroaryl moiety is 5 to 14 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S.
  • the heteroaryl moiety of the heteroarylalkyl group may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system.
  • Alkyl, alkylene, aryl, arylalkyl, and heteroarylalkyl groups may be substituted where one or more hydrogen atoms are each independently replaced with a substituent.
  • Typical substituents include, but are not limited to, —X, —R, —O ⁇ , —OR, —SR, —S ⁇ , —NR 2 , —NR 3 , ⁇ NR, —CX 3 , —CN, —OCN, —SCN, —N ⁇ C ⁇ O, —NCS, —NO, —NO 2 , ⁇ N 2 , —N 3 , —NC( ⁇ O)R, —C( ⁇ O)R, —C( ⁇ O)NR 2 , —SO 3 ⁇ , —SO 3 H, —S( ⁇ O) 2 R, —OS( ⁇ O) 2 OR, —S( ⁇ O) 2 NR, —S( ⁇ O)R, —OP( ⁇
  • Heteroaryl also known as heterocycle or heterocyclyl, refers to a ring system radical in which one or more ring atoms is a heteroatom, e.g. nitrogen, oxygen, and sulfur.
  • the heteroaryl radical comprises 5 to 14 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S.
  • a heteroaryl may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system.
  • Heteroaryl compounds are described in Paquette, Leo A.; “Principles of Modern Heterocyclic Chemistry” (W. A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9; “The Chemistry of Heterocyclic Compounds, A series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566.
  • Linker or “link” means a chemical moiety comprising a covalent bond or a chain of atoms that covalently attaches an antibody to a drug moiety.
  • a linker is specified as L.
  • Linkers include a divalent radical such as an alkylene, an arylene, a heteroarylene, moieties such as: —(CR 2 ) n O(CR 2 ) n —, repeating units of alkyloxy (e.g. polyethylenoxy, PEG, polymethyleneoxy) and alkylamino (e.g. polyethyleneamino, JeffamineTM); and diacid ester and amides including succinate, succinamide, diglycolate, malonate, and caproamide.
  • chiral refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • d and l or (+) and ( ⁇ ) are employed to designate the sign of rotation of plane-polarized light by the compound, with ( ⁇ ) or l meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • these stereoisomers are identical except that they are mirror images of one another.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • the terms “racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • phrases “pharmaceutically acceptable salt,” as used herein, refers to pharmaceutically acceptable organic or inorganic salts of an ADC.
  • Exemplary salts include, but are not limited, to 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, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3-na
  • 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.
  • “Pharmaceutically acceptable solvate” refers to an association of one or more solvent molecules and an ADC.
  • solvents that form pharmaceutically acceptable solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • the compounds of the invention include those with utility for anticancer activity.
  • the compounds include an antibody conjugated, i.e. covalently attached by a linker, to a drug moiety where the drug when not conjugated to an antibody has a cytotoxic or cytostatic effect.
  • the biological activity of the drug moiety is thus modulated by conjugation to an antibody.
  • the antibody-drug conjugates (ADC) of the invention may selectively deliver an effective dose of a cytotoxic agent to tumor tissue whereby greater selectivity, i.e. a lower efficacious dose may be achieved.
  • the bioavailability of the ADC, or an intracellular metabolite of the ADC is improved in a mammal when compared to the corresponding maytansinoid compound alone. Also, the bioavailability of the ADC, or an intracellular metabolite of the ADC is improved in a mammal when compared to the corresponding antibody alone (antibody of the ADC, without the drug moiety or linker).
  • the maytansinoid drug moiety of the ADC is not cleaved from the antibody until the antibody-drug conjugate binds to a cell-surface receptor or enters a cell with a cell-surface receptor specific for the antibody of the antibody-drug conjugate.
  • the drug moiety may be cleaved from the antibody after the antibody-drug conjugate enters the cell.
  • the maytansinoid drug moiety may be intracellularly cleaved in a mammal from the antibody of the compound, or an intracellular metabolite of the compound, by enzymatic action, hydrolysis, oxidation, or other mechanism.
  • the sulfur atom of the maytansinoid drug moiety of the ADC may be oxidized to a sulfone or sulfoxide group.
  • Protons on carbons bound to the sulfone and sulfoxide may be removed under general or enzymatic catalysis inside the cell and result in a beta-elimination fragmentation that cleaves and separates the drug moiety from the antibody of the ADC.
  • other electron withdrawing groups such as amides in the linker, antibody or drug moiety may effect similar fragmentation/cleavage mechanisms inside a cell.
  • Antibody-drug conjugates may be represented by Formula I: Ab-(L-D) p I or a pharmaceutically acceptable salt or solvate thereof, wherein:
  • L is a non-disulfide linker. L includes but is not limited to the structures: where the wavy lines indicate the covalent attachments to Ab and D;
  • the drug to antibody ratio or drug loading is represented by p for Formula I compounds.
  • the drug loading value p is 1 to 8.
  • Formula I compounds include all mixtures of variously loaded and attached antibody-drug conjugates where 1, 2, 3, 4, 5, 6, 7, and 8 drug moieties are covalently attached to the antibody.
  • Ab is an antibody which binds to one or more tumor-associated antigens or cell-surface receptors selected from (1)-(16) and (18)-(36), i.e. not to an ErbB receptor, including HER2.
  • the antibody unit (Ab-) of Formula I includes within its scope any unit of an antibody that binds or reactively associates or complexes with a receptor, antigen or other receptive moiety associated with a given target-cell population.
  • An antibody can be any protein or protein-like molecule that binds to, complexes with, or reacts with a moiety of a cell population sought to be therapeutically or otherwise biologically modified.
  • the antibody unit acts to deliver the maytansinoid drug moiety to the particular target cell population with which the antibody unit reacts.
  • Such antibodies include, but are not limited to, large molecular weight proteins such as, full-length antibodies and antibody fragments.
  • Antibodies comprising the antibody-drug conjugates of the invention preferably retain the antigen binding capability of their native, wild type counterparts.
  • antibodies of the invention are capable of binding, preferably specifically, to antigens.
  • antigens include, for example, tumor-associated antigens (TAA), cell surface receptor proteins and other cell surface molecules, cell survival regulatory factors, cell proliferation regulatory factors, molecules associated with (for e.g., known or suspected to contribute functionally to) tissue development or differentiation, lymphokines, cytokines, molecules involved in cell cycle regulation, molecules involved in vasculogenesis and molecules associated with (for e.g., known or suspected to contribute functionally to) angiogenesis.
  • TAA tumor-associated antigens
  • cell survival regulatory factors include, for example, tumor-associated antigens (TAA), cell surface receptor proteins and other cell surface molecules, cell survival regulatory factors, cell proliferation regulatory factors, molecules associated with (for e.g., known or suspected to contribute functionally to) tissue development or differentiation, lymphokines, cytokines, molecules involved in
  • An antigen to which an antibody of the invention is capable of binding may be a member of a subset of one of the above-mentioned categories, wherein the other subset(s) of said category comprise other molecules/antigens that have a distinct characteristic (with respect to the antigen of interest).
  • the antibody of the antibody-drug conjugates specifically binds to a receptor encoded by an ErbB gene.
  • the antibody may bind specifically to an ErbB receptor selected from EGFR, HER2, HER3 and HER4.
  • the ADC may specifically bind to the extracellular domain (ECD) of the HER2 receptor and inhibit the growth of tumor cells which overexpress HER2 receptor.
  • the antibody of the ADC may be a monoclonal antibody, e.g. a murine monoclonal antibody, a chimeric antibody, or a humanized antibody.
  • a humanized antibody may be huMAb4D5-1, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 or huMAb4D5-8 (trastuzumab).
  • the antibody may be an antibody fragment, e.g. a Fab fragment.
  • Antibodies in Formula I antibody-drug conjugates and which may be useful in the treatment of cancer include, but are not limited to, antibodies against cell surface receptors and tumor-associated antigens (TAA).
  • TAA tumor-associated antigens
  • TAA tumor-associated antigens
  • TAA tumor-associated antigens
  • tumor-associated polypeptides are more abundantly expressed on the surface of the cancer cells as compared to on the surface of the non-cancerous cells.
  • the identification of such tumor-associated cell surface antigen polypeptides has given rise to the ability to specifically target cancer cells for destruction via antibody-based therapies.
  • TAA Tumor-Associated Antigens
  • TAA Tumor-Associated Antigens
  • NCBI National Center for Biotechnology Information
  • Tumor-associated antigens targeted by antibodies include all amino acid sequence variants and isoforms possessing at least about 70%, 80%, 85%, 90%, or 95% sequence identity relative to the sequences identified in the cited references, or which exhibit substantially the same biological properties or characteristics as a TAA having a sequence found in the cited references.
  • a TAA having a variant sequence generally is able to bind specifically to an antibody that binds specifically to the TAA with the corresponding sequence listed.
  • MAbs monoclonal antibodies
  • Hybridoma technology which refers to a cloned cell line that produces a single type of antibody, uses the cells of various species, including mice (murine), hamsters, rats, and humans.
  • Polyclonal antibodies may be raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant.
  • Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • Binding specificity of monoclonal antibodies produced by hybridoma cells may be determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson et al (1980) Anal. Biochem. 107:220.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells serve as a source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells (U.S. 2005/0048572; U.S. 2004/0229310).
  • monoclonal antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al (1990) Nature 348:552-554; Clackson et al (1991) Nature 352:624-628; and Marks et al (1991) J. Mol. Biol., 222:581-597 describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy chain and light chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567); and Morrison et al (1984) Proc. Natl. Acad. Sci. USA 81:6851), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • the production of antibodies will be illustrated with reference to anti-ErbB2 antibodies but it will be apparent for those skilled in the art that antibodies to other members of the ErbB receptor family, as well as any other receptor or tumor-associated antigen or target, can be produced and modified in a similar manner.
  • the ErbB2 antigen to be used for production of antibodies may be, e.g., a soluble form of the extracellular domain of ErbB2 or a portion thereof, containing the desired epitope.
  • cells expressing ErbB2 at their cell surface e.g. NIH-3T3 cells transformed to overexpress ErbB2; or a carcinoma cell line such as SK-BR-3 cells (Stancovski et al (1991) PNAS (USA) 88:8691-8695), can be used to generate antibodies.
  • Other forms of ErbB2 useful for generating antibodies will be apparent to those skilled in the art.
  • Example 1 describes production of an exemplary humanized anti-ErbB2 antibody.
  • the humanized antibody may, for example, comprise nonhuman hypervariable region residues incorporated into a human variable heavy domain and may further comprise a framework region (FR) substitution at a position selected from the group consisting of 69H, 71H and 73H utilizing the variable domain numbering system set forth in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991).
  • the humanized antibody comprises FR substitutions at two or all of positions 69H, 71H and 73H.
  • human antibodies can be generated.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • U.S. Pat. No. 5,591,669; U.S. Pat. No. 5,589,369; U.S. Pat. No. 5,545,807 are examples of transgenic animals that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production.
  • phage display technology can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors (Johnson, Kevin S. and Chiswell, David J. (1993) Current Opinion in Structural Biology 3:564-571).
  • V immunoglobulin variable
  • a repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially (Marks et al (1991) J. Mol. Biol. 222:581-597; Griffith et al (1993) EMBO J. 12:725-734; U.S. Pat. No.
  • Human antibodies may also be generated by in vitro activated B cells (U.S. Pat. No. 5,567,610; U.S. Pat. No. 5,229,275). Human anti-ErbB2 antibodies are described (U.S. Pat. No. 5,772,997 and WO 97/00271.
  • antibody fragments can also be produced directly by recombinant host cells and the antibody phage libraries discussed above.
  • Fab′-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab′) 2 fragments (Carter et al (1992) Bio/Technology 10:163-167). According to another approach, F(ab′) 2 fragments can be isolated directly from recombinant host cell culture.
  • the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; U.S. Pat. No. 5,571,894; and U.S. Pat. No. 5,587,458.
  • the antibody fragment may also be a “linear antibody”, e.g., as described in U.S. Pat. No. 5,641,870 for example. Such linear antibody fragments may be monospecific or bispecific.
  • Bispecific antibodies with binding specificities for at least two different epitopes may bind to two different epitopes of the ErbB2 protein.
  • Other such antibodies may combine an ErbB2 binding site with binding site(s) for EGFR, ErbB3 and/or ErbB4.
  • an anti-ErbB2 arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g.
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express ErbB2 (WO 96/16673; U.S. Pat. No. 5,837,234; WO98/02463; U.S. Pat. No. 5,821,337). Purification methods for bispecific antibodies have been disclosed (WO 93/08829; Traunecker et al (1991) EMBO J.
  • Bispecific antibodies can be produced using leucine zippers (Kostelny et al (1992) J. Immunol. 148(5):1547-1553), and single-chain Fv (sFv) dimers (Gruber et al (1994) J. Immunol. 152:5368).
  • bispecific antibodies from antibody fragments
  • Techniques for generating bispecific antibodies from antibody fragments have also been described, such as using chemical linkage wherein intact antibodies are proteolytically cleaved to generate F(ab′) 2 fragments (Brennan et al (1985) Science 229:81).
  • Fab′-SH fragments can be recovered from E. coli and chemically coupled to form bispecific antibodies (Shalaby et al (1992) J. Exp. Med. 175:217-225.
  • the “diabody” technology provides an alternative method for making bispecific antibody fragments (Hollinger et al (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448).
  • Antibodies with more than two valencies are contemplated.
  • Multivalent, “Octopus” antibodies with three or more antigen binding sites and two or more variable domains can be readily produced by recombinant expression of nucleic acid encoding the polypeptide chains of the antibody (U.S. 2002/0004586; WO 01/77342).
  • trispecific antibodies can be prepared (Tutt et al (1991) J. Immunol. 147:60.
  • Amino acid sequence modification(s) of antibodies are contemplated. For example, mutants and various isoforms of antibodies which bind to tumor-associated antigens are contemplated to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody are prepared by introducing appropriate nucleotide changes into the nucleic acid encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of, residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • the amino acid changes also may alter post-translational processes of the antibody, such as changing the number or position of glycosylation sites.
  • a useful method for identification of certain residues or regions of the antibody that are preferred locations for mutagenesis is “alanine scanning mutagenesis” (Cunningham and Wells (1989) Science 244:1081-1085) where an amino acid residue, or group of target residues, are identified (e.g., charged residues such as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino acid, such as alanine or polyalanine, to optimize the interaction of the amino acids with antigen.
  • alanine scanning mutagenesis (Cunningham and Wells (1989) Science 244:1081-1085) where an amino acid residue, or group of target residues, are identified (e.g., charged residues such as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino acid, such as alanine or polyalanine, to optimize the interaction of the amino acids with antigen.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an anti-ErbB2 antibody with an N-terminal methionyl residue or the antibody fused to a cytotoxic polypeptide.
  • Other insertional variants of the anti-ErbB2 antibody molecule include the fusion to the N- or C-terminus of the anti-ErbB2 antibody to an enzyme (e.g. for ADEPT: Tietze et al (2003) Current Pharm. Design 9:2155-2175) or a polypeptide which increases the serum half-life of the antibody, such as an albumin-binding peptide.
  • Plasma-protein binding can be an effective means of improving the pharmacokinetic properties of short lived molecules.
  • Albumin is the most abundant protein in plasma.
  • Serum albumin binding peptides (ABP) can alter the pharmacodynamics of fused active domain proteins, including alteration of tissue uptake, penetration, and diffusion. These pharmacodynamic parameters can be modulated by specific selection of the appropriate serum albumin binding peptide sequence (U.S. 20040001827).
  • a series of albumin binding peptides were identified by phage display screening (Dennis et al (2002) “Albumin Binding As A General Strategy For Improving The Pharmacokinetics Of Proteins” J Biol. Chem. 277:35035-35043; WO 01/45746).
  • Compounds of the invention include ABP sequences taught by: (i) Dennis et al (2002) J Biol. Chem. 277:35035-35043 at Tables III and IV, page 35038; (ii) U.S. 20040001827 at [0076] SEQ ID NOS: 9-22; and (iii) WO 01/45746 at pages 12-13, SEQ ID NOS: z1-z14, and all of which are incorporated herein by reference.
  • the amino acid sequence is usually altered by altering the underlying nucleic acid sequence.
  • Nucleic acid molecules encoding amino acid sequence variants of the antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the antibody.
  • the sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but FR alterations are also contemplated.
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking.
  • cysteine bond(s) may be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
  • the term “salvage receptor binding epitope” refers to an epitope of the Fc region of an IgG molecule (e.g., IgG 1 , IgG 2 , IgG 3 , or IgG 4 ) that is responsible for increasing the in vivo serum half-life of the IgG molecule (U.S. 2003/0190311, U.S. Pat. No. 6,821,505; U.S. Pat. No. 6,165,745; U.S. Pat. No. 5,624,821; U.S. Pat. No. 5,648,260; U.S. Pat. No. 6,165,745; U.S. Pat. No. 5,834,597).
  • Glycosylation variants of antibodies are variants in which the glycosylation pattern of an antibody is altered.
  • altering is meant deleting one or more carbohydrate moieties found in the antibody, adding one or more carbohydrate moieties to the antibody, changing the composition of glycosylation (glycosylation pattern), or the extent of glycosylation.
  • Antibodies may be glycosylated at conserved positions (N-linked or O-linked) in their constant regions (Hse et al (1997) J. Biol. Chem. 272:9062-9070; Jefferis and Lund, (1997) Chem. Immunol. 65:111-128; Wright and Morrison, (1997) TibTECH 15:26-32).
  • the oligosaccharide side chains of the immunoglobulins affect the protein's function (Boyd et al (1996) Mol. Immunol. 32:1311-1318; Wittwe and Howard, (1990) Biochem.
  • Oligosaccharides may also serve to target a given glycoprotein to certain molecules based upon specific recognition structures (Malhotra et al (1995) Nature Med. 1:237-243; Umana et al (1999) Nature Biotech. 17:176-180). Removal of the oligosaccharides may optimize antigen binding and other properties of the antibody (Boyd et al (1996) Mol. Immunol. 32:1311-1318).
  • Factors which affect glycosylation during recombinant production of antibodies include growth mode, media formulation, culture density, oxygenation, pH, purification schemes and the like (U.S. Pat. No. 5,047,335; U.S. Pat. No. 5,510,261; U.S. Pat. No. 5,278,299).
  • Glycosylation, or certain types of glycosylation can be enzymatically removed from the glycoprotein, for example using endoglycosidase H (Endo H).
  • the recombinant host cell can be genetically engineered, e.g. make defective in processing certain types of polysaccharides.
  • glycosylation structure of antibodies can be readily analyzed by conventional techniques of carbohydrate analysis, including lectin chromatography, NMR, Mass spectrometry, HPLC, GPC, monosaccharide compositional analysis, sequential enzymatic digestion, and HPAEC-PAD, which uses high pH anion exchange chromatography to separate oligosaccharides based on charge.
  • Methods for releasing oligosaccharides for analytical purposes include, without limitation, enzymatic treatment (commonly performed using peptide-N-glycosidase F/endo- ⁇ -galactosidase), elimination using harsh alkaline environment to release mainly O-linked structures, and chemical methods using anhydrous hydrazine to release both N- and O-linked oligosaccharides.
  • Maytansine compounds inhibit cell proliferation by inhibiting the formation of microtubules during mitosis through inhibition of polymerization of the microtubulin protein, tubulin (Remillard et al (1975) Science 189:1002-1005; U.S. Pat. No. 5,208,020).
  • Maytansine and maytansinoids are highly cytotoxic but their clinical use in cancer therapy has been greatly limited by their severe systemic side-effects primarily attributed to their poor selectivity for tumors.
  • Clinical trials with maytansine had been discontinued due to serious adverse effects on the central nervous system and gastrointestinal system (Issel et al (1978) Can. Treatment. Rev. 5:199-207.
  • Maytansinoid drug moieties are attractive drug moieties in antibody-drug conjugates because they are: (i) relatively accessible to prepare by fermentation or chemical modification, derivatization of fermentation products, (ii) amenable to derivatization with functional groups suitable for conjugation through the non-disulfide linkers to antibodies, (iii) stable in plasma, and (iv) effective against a variety of tumor cell lines.
  • Maytansine compounds suitable for use as maytansinoid drug moieties are well known in the art, and can be isolated from natural sources according to known methods, produced using genetic engineering techniques (see Yu et al (2002) PNAS 99:7968-7973), or maytansinol and maytansinol analogues prepared synthetically according to known methods.
  • Exemplary maytansinoid drug moieties include those having a modified aromatic ring, such as: C-19-dechloro (U.S. Pat. No. 4,256,746) (prepared by lithium aluminum hydride reduction of ansamitocin P2); C-20-hydroxy (or C-20-demethyl)+/ ⁇ C-19-dechloro (U.S. Pat. Nos. 4,361,650 and 4,307,016) (prepared by demethylation using Streptomyces or Actinomyces or dechlorination using LAH); and C-20-demethoxy, C-20-acyloxy (—OCOR), +/ ⁇ dechloro (U.S. Pat. No. 4,294,757) (prepared by acylation using acyl chlorides). and those having modifications at other positions
  • Exemplary maytansinoid drug moieties also include those having modifications such as: C-9-SH, prepared by the reaction of maytansinol with H 2 S or P 2 S 5 (U.S. Pat. No. 4,424,219); C-14-alkoxymethyl(demethoxy/CH 2 OR) (U.S. Pat. No. 4,331,598); C-14-hydroxymethyl or acyloxymethyl (CH 2 OH or CH 2 OAc) prepared from Nocardia (U.S. Pat. No. 4,450,254); C-15-hydroxy/acyloxy, prepared by the conversion of maytansinol by Streptomyces (U.S. Pat. No.
  • the linkage position is known to be useful as the linkage position, depending upon the type of link.
  • the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the C-15 position modified with a hydroxyl group and the C-20 position having a hydroxyl group are all suitable.
  • Maytansinoid drug moieties include those having the structure: where the wavy line indicates the covalent attachment of the sulfur atom of D to a linker (L) of an antibody-drug conjugate (ADC).
  • R may independently be H or a C 1 -C 6 alkyl.
  • the alkylene chain attaching the amide group to the sulfur atom may be methanyl, ethanyl, or propyl, i.e. m is 1, 2, or 3 (U.S. Pat. Nos. 633,410, 5,208,020, Chari et al (1992) Cancer Res. 52:127-131; Liu et al (1996) Proc. Natl. Acad. Sci 93:8618-8623).
  • the maytansinoid drug moiety (D) will have the following stereochemistry:
  • alkyl groups such as the methyl groups on the carbon adjacent to the sulfur atom of DM3 and DM4 may affect the rate of intracellular cleavage of the ADC (U.S. 2004/0235840 A1).
  • the variable alkyl unit (CR 2 ) m may therefore affect potency, efficacy, and safety/toxicity in vitro and in vivo.
  • the linker, L attaches the antibody to a drug moiety through covalent bond(s), not comprising a disulfide group.
  • the linker is a bifunctional or multifunctional moiety which can be used to link one or more Drug moieties (D) and an antibody unit (Ab) to form antibody-drug conjugates (ADC) of Formula I.
  • Antibody-drug conjugates (ADC) can be conveniently prepared using a linker having reactive functionality for binding to the Drug and to the Antibody.
  • a cysteine thiol, or an amine, e.g. N-terminus or amino acid side chain such as lysine, of the antibody (Ab) can form a bond with a functional group of a linker reagent, drug moiety or drug-linker reagent.
  • the linkers are preferably stable extracellularly.
  • the antibody-drug conjugate (ADC) is preferably stable and remains intact, i.e. the antibody remains linked to the drug moiety.
  • the linkers are stable outside the target cell and may be cleaved at some efficacious rate inside the cell.
  • An effective linker will: (i) maintain the specific binding properties of the antibody; (ii) allow intracellular delivery of the conjugate or drug moiety; (iii) remain stable and intact, i.e. not cleaved, until the conjugate has been delivered or transported to its targetted site; and (iv) maintain a cytotoxic, cell-killing effect or a cytostatic effect of the maytansinoid drug moiety. Stability of the ADC may be measured by standard analytical techniques such as mass spectroscopy, HPLC, and the separation/analysis technique LC/MS.
  • bivalent linker reagents which are useful to attach two or more functional or biologically active moieties, such as peptides, nucleic acids, drugs, toxins, antibodies, haptens, and reporter groups are known, and methods have been described their resulting conjugates (Hermanson, G. T. (1996) Bioconjugate Techniques ; Academic Press: New York, p234-242).
  • Linkers may have structures selected from: where the wavy lines indicate the covalent attachments to Ab and D in either orientation.
  • X may have the structures, in either orientation: where R is independently H or C 1 -C 6 alkyl; and n is 1 to 12.
  • Y may have the structures, in either orientation: where R is independently H or C 1 -C 6 alkyl; and n is 1 to 12.
  • the linker may have the structure, designated as SMCC:
  • linker (L) has the structure: where the wavy lines indicate the covalent attachments to Ab and D in either orientation.
  • the linker may have the structure, designated as SIAB:
  • linker (L) has the structure:
  • the linker may be substituted with groups which modulated solubility or reactivity.
  • a sulfonate substituent may increase water solubility of the reagent and facilitate the coupling reaction of the linker reagent with the antibody or the drug moiety, or facilitate the coupling reaction of Ab-L with D, or D-L with Ab, depending on the synthetic route employed to prepare the ADC.
  • a Linker has a reactive functional group which has a nucleophilic group that is reactive to an electrophilic group present on an antibody.
  • Useful electrophilic groups on an antibody include, but are not limited to, aldehyde and ketone carbonyl groups.
  • the heteroatom of a nucleophilic group of a Linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit.
  • Useful nucleophilic groups on a Linker include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide.
  • the electrophilic group on an antibody provides a convenient site for attachment to a Linker.
  • Linkers can be peptidic, comprising one or more amino acid units.
  • Peptide linker reagents may be prepared by solid phase or liquid phase synthesis methods (E. Schröder and K. Lübke, The Peptides , volume 1, pp 76-136 (1965) Academic Press) that are well known in the field of peptide chemistry, including t-BOC chemistry (Geiser et al “Automation of solid-phase peptide synthesis” in Macromolecular Sequencing and Synthesis , Alan R. Liss, Inc., 1988, pp. 199-218) and Fmoc/HBTU chemistry (Fields, G. and Noble, R.
  • the compounds expressly contemplate, but are not limited to, ADC prepared with cross-linker reagents: BMPEO, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate), and including bis-maleimide reagents: DTME, BMB, BMDB, BMH, BMOE, BM(PEO) 3 , and BM(PEO) 4 , which are commercially available from Pierce Biotechnology, Inc., Customer Service Department, P.O.
  • Bis-maleimide reagents allow the attachment of a free thiol group of a cysteine residue of an antibody to a thiol-containing drug moiety, label, or linker intermediate, in a sequential or concurrent fashion.
  • Suitable functional groups besides maleimide, which are reactive with a thiol group of an antibody, maytansinoid drug moiety, or linker intermediate include iodoacetamide, bromoacetamide, vinyl pyridine, disulfide, pyridyl disulfide, isocyanate, and isothiocyanate.
  • Exemplary antibody-drug conjugates where DM1 is linked through a BMPEO linker to a thiol group of trastuzumab have the structure:
  • Useful linker reagents can also be obtained via other commercial sources, such as Molecular Biosciences Inc. (Boulder, Colo.), or synthesized in accordance with procedures described in Toki et al (2002) J. Org. Chem. 67:1866-1872; U.S. Pat. No. 6,214,345 to Firestone et al; WO 02/088172; U.S. 2003130189; U.S. 2003096743; WO 03/026577; WO 03/043583; and WO 04/032828.
  • the Linker may be a dendritic type linker for covalent attachment of more than one drug moiety through a branching, multifunctional linker moiety to an antibody (Sun et al (2002) Bioorganic & Medicinal Chemistry Letters 12:2213-2215; Sun et al (2003) Bioorganic & Medicinal Chemistry 11:1761-1768; King et al (2002) Tetrahedron Letters 43:1987-1990).
  • Dendritic linkers can increase the molar ratio of drug to antibody, i.e. loading, which is related to the potency of the ADC.
  • an antibody bears only one reactive cysteine thiol group, a multitude of drug moieties may be attached through a dendritic linker.
  • dendritic linker reagents allow up to nine nucleophilic drug moiety reagents to be conjugated by reaction with the chloroethyl nitrogen mustard functional groups: Drug Loading
  • the drug loading is represented by p in a molecule of Formula I, the average number of maytansinoid drugs per antibody.
  • Drug loading may range from 1 to 8 drugs (D) per antibody (Ab), i.e. where 1, 2, 3, 4, 5, 6, 7, and 8 drug moieties are covalently attached to the antibody.
  • Compositions of ADC of Formula I include collections of antibodies conjugated with a range of drugs, from 1 to 8.
  • the average number of drugs per antibody in preparations of ADC from conjugation reactions may be characterized by conventional means such as mass spectroscopy, ELISA assay, electrophoresis, and HPLC.
  • the quantitative distribution of ADC in terms of p may also be determined.
  • ELISA the averaged value of p in a particular preparation of ADC may be determined (Hamblett et al (2004) Clinical Cancer Res. 10:7063-7070; Sanderson et al (2005) Clinical Cancer Res. 11:843-852).
  • the distribution of p (drug) values is not discernible by the antibody-antigen binding and detection limitation of ELISA.
  • ELISA assay for detection of antibody-drug conjugates does not determine where the drug moieties are attached to the antibody, such as the heavy chain or light chain fragments, or the particular amino acid residues.
  • separation, purification, and characterization of homogeneous ADC where p is a certain value from ADC with other drug loadings may be achieved by means such as reverse phase HPLC or electrophoresis.
  • p may be limited by the number of attachment sites on the antibody.
  • an antibody may have only one or several cysteine thiol groups, or may have only one or several sufficiently reactive thiol groups through which a linker may be attached.
  • Higher drug loading e.g. p>5, may cause aggregation, insolubility, toxicity, or loss of cellular permeability of certain antibody-drug conjugates.
  • an antibody may contain, for example, many lysine residues that do not react with the drug-linker intermediate (D-L) or linker reagent. Only the most reactive lysine groups may react with an amine-reactive linker reagent. Also, only the most reactive cysteine thiol groups may react with a thiol-reactive linker reagent. Generally, antibodies do not contain many, if any, free and reactive cysteine thiol groups which may be linked to a drug moiety.
  • cysteine thiol residues in the antibodies of the compounds exist as disulfide bridges and must be reduced with a reducing agent such as dithiothreitol (DTT) or TCEP, under partial or total reducing conditions. Additionally, the antibody must be subjected to denaturing conditions to reveal reactive nucleophilic groups such as lysine or cysteine.
  • the loading (drug/antibody ratio) of an ADC may be controlled in several different manners, including: (i) limiting the molar excess of drug-linker intermediate (D-L) or linker reagent relative to antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) partial or limiting reductive conditions for cysteine thiol modification.
  • the resulting product is a mixture of ADC compounds with a distribution of drug moieties attached to an antibody, e.g. 1, 2, 3, etc.
  • Liquid chromatography methods such as polymeric reverse phase (PLRP) and hydrophobic interaction (HIC) may separate compounds in the mixture by drug loading value.
  • Preparations of ADC with a single drug loading value (p) may be isolated (“Effect of drug loading on the pharmacology, pharmacokinetics, and toxicity of an anti-CD30 antibody-drug conjugate”, Hamblett, K. J., et al, Abstract No.
  • ADCs may still be heterogeneous mixtures because the drug moieties may be attached, via the linker, at different sites on the antibody.
  • the ADC of Formula I may be prepared by several routes, employing organic chemistry reactions, conditions, and reagents known to those skilled in the art, including: (1) reaction of a nucleophilic group or an electrophilic group of an antibody with a bivalent linker reagent, to form antibody-linker intermediate Ab-L, via a covalent bond, followed by reaction with an activated drug moiety D; and (2) reaction of a nucleophilic group or an electrophilic group of a drug moiety with a linker reagent, to form drug-linker intermediate D-L, via a covalent bond, followed by reaction with the nucleophilic group or an electrophilic group of an antibody. Conjugation methods (1) and (2) may be employed with a variety of antibodies, drug moieties, and linkers to prepare the antibody-drug conjugates of Formula I.
  • Nucleophilic groups on antibodies include, but are not limited to: (i) N-terminal amine groups, (ii) side chain amine groups, e.g. lysine, (iii) side chain thiol groups, e.g. cysteine, and (iv) sugar hydroxyl or amino groups where the antibody is glycosylated.
  • Amine, thiol, and hydroxyl groups are nucleophilic and capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups. Certain antibodies have reducible interchain disulfides, i.e. cysteine bridges.
  • Antibodies may be made reactive for conjugation with linker reagents by treatment with a reducing agent such as DTT (Cleland's reagent, dithiothreitol) or TCEP (tris(2-carboxyethyl)phosphine hydrochloride; Getz et al (1999) Anal. Biochem. Vol 273:73-80; Soltec Ventures, Beverly, Mass.).
  • a reducing agent such as DTT (Cleland's reagent, dithiothreitol) or TCEP (tris(2-carboxyethyl)phosphine hydrochloride; Getz et al (1999) Anal. Biochem. Vol 273:73-80; Soltec Ventures, Beverly, Mass.).
  • a reducing agent such as DTT (Cleland's reagent, dithiothreitol) or TCEP (tris(2-carboxyethyl)phosphine hydrochloride
  • Antibody-drug conjugates may also be produced by modification of the antibody to introduce electrophilic moieties, which can react with nucleophilic substituents on the linker reagent or drug.
  • the sugars of glycosylated antibodies may be oxidized, e.g. with periodate oxidizing reagents, to form aldehyde or ketone groups which may react with the amine group of linker reagents or drug moieties.
  • the resulting imine Schiff base groups may form a stable linkage, or may be reduced, e.g. by borohydride reagents to form stable amine linkages.
  • reaction of the carbohydrate portion of a glycosylated antibody with either galactose oxidase or sodium meta-periodate may yield carbonyl (aldehyde and ketone) groups in the protein that can react with appropriate groups on the drug (Hermanson, G. T. (1996) Bioconjugate Techniques; Academic Press: New York, p234-242).
  • proteins containing N-terminal serine or threonine residues can react with sodium meta-periodate, resulting in production of an aldehyde in place of the first amino acid (Geoghegan & Stroh, (1992) Bioconjugate Chem. 3:138-146; U.S. Pat. No. 5,362,852).
  • Such aldehyde can be reacted with a drug moiety or linker nucleophile.
  • nucleophilic groups on a drug moiety include, but are not limited to: amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide groups capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups.
  • Maytansine may, for example, be converted to May-SSCH 3 , which can be reduced to the free thiol, May-SH, and reacted with a modified antibody (Chari et al (1992) Cancer Research 52:127-131) to generate a maytansinoid-antibody immunoconjugate with a disulfide linker.
  • Antibody-maytansinoid conjugates with disulfide linkers have been reported (WO 04/016801; U.S. Pat. No. 6,884,874; U.S. 2004/039176 A1; WO 03/068144; U.S. 2004/001838 A1; U.S. Pat. No. 6,441,163; U.S. Pat. No.
  • the disulfide linker SPP is constructed with linker reagent N-succinimidyl 4-(2-pyridylthio) pentanoate.
  • Antibody-SPP-DM1 conjugates are represented by the structure:
  • Disulfide (S—S) linker antibody-drug conjugates were tested for purposes of comparison to the non-disulfide linker ADC of the invention.
  • Trastuzumab-SPP-DM1 was prepared according to Example 3 (Ranson, M. and Sliwkowski M. (2002) Oncology 63(suppl 1):17-24).
  • Disulfide antibody-drug conjugates: trastuzumab-SPDP-DM1, trastuzumab-SPP-DM3, and trastuzumab-SPP-DM4 were also tested, and have the structures below:
  • ADC of the invention include SMCC linkers and the DM1 maytansinoid drug moiety, represented as Ab-SMCC-DM1:
  • Transgenic animals and cell lines are particularly useful in screening antibody-drug conjugates (ADC) that have potential as prophylactic or therapeutic treatments of diseases or disorders involving overexpression of tumor-associated antigens and cell surface receptors, e.g. HER2 (U.S. Pat. No. 6,632,979).
  • Screening for a useful ADC may involve administering candidate ADC over a range of doses to the transgenic animal, and assaying at various time points for the effect(s) of the ADC on the disease or disorder being evaluated. Alternatively, or additionally, the drug can be administered prior to or simultaneously with exposure to an inducer of the disease, if applicable.
  • Candidate ADC may be screened serially and individually, or in parallel under medium or high-throughput screening format. The rate at which ADC may be screened for utility for prophylactic or therapeutic treatments of diseases or disorders is limited only by the rate of synthesis or screening methodology, including detecting/measuring/analysis of data.
  • One embodiment is a screening method comprising (a) transplanting cells from a stable breast cancer cell line into a non-human animal, (b) administering an ADC drug candidate to the non-human animal and (c) determining the ability of the candidate to inhibit the formation of tumors from the transplanted cell line.
  • the invention also concerns a method of screening ADC candidates for the treatment of a disease or disorder characterized by the overexpression of a receptor protein comprising (a) contacting cells from a stable breast cancer cell line with a drug candidate and (b) evaluating the ability of the ADC candidate to inhibit the growth of the stable cell line.
  • One embodiment is a screening method comprising (a) contacting cells from a stable breast cancer cell line with an ADC drug candidate and (b) evaluating the ability of the ADC candidate to block ligand activation of HER2. Another embodiment the ability of the ADC candidate to block heregulin binding is evaluated. In another embodiment the ability of the ADC candidate to block ligand-stimulated tyrosine phosphorylation is evaluated.
  • Another embodiment is a screening method comprising (a) contacting cells from a stable breast cancer cell line with an ADC drug candidate and (b) evaluating the ability of the ADC candidate to induce cell death. In one embodiment the ability of the ADC candidate to induce apoptosis is evaluated.
  • Another embodiment is a screening method comprising (a) administering an ADC drug candidate to a transgenic non-human mammal that overexpresses, e.g. in its mammary gland cells, a native human protein, e.g. HER2 or a fragment thereof, wherein such transgenic mammal has stably integrated into its genome a nucleic acid sequence encoding the native human protein or a fragment thereof having the biological activity of the native human protein, operably linked to transcriptional regulatory sequences directing its expression, and develops a tumor, e.g. a mammary tumor, not responding or poorly responding to antibody treatment, e.g.
  • the disease or disorder may be a HER2-overexpressing cancer, such as breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, thyroid, pancreatic and bladder cancer.
  • the cancer may be breast cancer which expressed HER2 in at least about 500,000 copies per cell, or at least about 2,000,000 copies per cell.
  • ADC drug candidates may, for example, be evaluated for their ability to induce cell death and/or apoptosis, using assay methods well known in the art and described hereinafter.
  • candidate ADC are screened by being administered to the transgenic animal over a range of doses, and evaluating the animal's physiological response to the compounds over time.
  • Administration may be oral, or by suitable injection, depending on the chemical nature of the compound being evaluated. In some cases, it may be appropriate to administer the compound in conjunction with co-factors that would enhance the efficacy of the compound.
  • cell lines derived from the subject transgenic animals are used to screen for compounds useful in treating various disorders associated with overexpression of certain tumor-associated antigen proteins or cell surface receptors, e.g. HER2-overexpression, the test compounds are added to the cell culture medium at an appropriate time, and the cellular response to the compound is evaluated over time using the appropriate biochemical and/or histological assays. In some cases, it may be appropriate to apply the compound of interest to the culture medium in conjunction with co-factors that would enhance the efficacy of the compound.
  • the present invention provides assays for identifying ADC which specifically target and bind the overexpressed HER2 protein, the presence of which is correlated with abnormal cellular function, and in the pathogenesis of cellular proliferation and/or differentiation of mammary gland that is causally related to the development of breast tumors.
  • the ability of the compound to block ErbB ligand binding to cells expressing the ErbB (ErbB2) receptor may be determined.
  • cells isolated from the transgenic animal overexpressing HER2 and transfected to express another ErbB receptor (with which HER2 forms hetero-oligomer) may be incubated, i.e. culturing, with the ADC and then exposed to labeled ErbB ligand.
  • the ability of the compound to block ligand binding to the ErbB receptor in the ErbB hetero-oligomer may then be evaluated.
  • inhibition of heregulin (HRG) binding to breast tumor cell lines, overexpressing HER2 and established from the transgenic non-human mammals (e.g. mice) herein, by the candidate ADC may be performed using monolayer cultures on ice in a 24-well-plate format.
  • Anti-ErbB2 monoclonal antibodies may be added to each well and incubated for 30 minutes.
  • 125 I-labeled rHRG ⁇ 1 177-224 (25,000 cpm) may then be added, and the incubation may be continued for 4 to 16 hours.
  • Dose response curves may be prepared and an IC 50 value may be calculated for the compound of interest.
  • an ADC to block ErbB ligand-stimulated tyrosine phosphorylation of an ErbB receptor present in an ErbB hetero-oligomer may be assessed.
  • cell lines established from the transgenic animals herein may be incubated with a test ADC and then assayed for ErbB ligand-dependent tyrosine phosphorylation activity using an anti-phosphotyrosine monoclonal antibody (which is optionally conjugated with a detectable label).
  • the kinase receptor activation assay described in U.S. Pat. No. 5,766,863 is also available for determining ErbB receptor activation and blocking of that activity by the compound.
  • one may screen for ADC which inhibit HRG stimulation of p180 tyrosine phosphorylation in MCF7 cells essentially as described below.
  • a cell line established from a HER2-transgenic animal may be plated in 24-well plates and the compound may be added to each well and incubated for 30 minutes at room temperature; then rHRG ⁇ 1177-244 may be added to each well to a final concentration of 0.2 nM, and the incubation may be continued for about 8 minutes.
  • Media may be aspirated from each well, and reactions may be stopped by the addition of 100 ⁇ l of SDS sample buffer (5% SDS, 25 mM DTT, and 25 mM Tris-HCl, pH 6.8).
  • Each sample (25 ⁇ l) may be electrophoresed on a 4-12% gradient gel (Novex) and then electrophoretically transferred to polyvinylidene difluoride membrane.
  • Antiphosphotyrosine (at 1 ⁇ g/ml) immunoblots may be developed, and the intensity of the predominant reactive band at M r ⁇ 180,000 may be quantified by reflectance densitometry.
  • An alternate method to evaluate inhibition of receptor phosphorylation is the KIRA (kinase receptor activation) assay (Sadick et al (1998) Jour. of Pharm. and Biomed. Anal. 1-9).
  • Some of the well-established monoclonal antibodies against HER2 that are known to inhibit HRG stimulation of p180 tyrosine phosphorylation can be used as positive control in this assay.
  • a dose-response curve for inhibition of HRG stimulation of p180 tyrosine phosphorylation as determined by reflectance densitometry may be prepared and an IC 50 for the compound of interest may be calculated.
  • the cells may be treated with a test compound at various concentrations for 4 days and stained with crystal violet or the redox dye Alamar Blue. Incubation with the compound may show a growth inhibitory effect on this cell line similar to that displayed by monoclonal antibody 2C4 on MDA-MB-175 cells (Schaefer et al., supra).
  • exogenous HRG will not significantly reverse this inhibition.
  • ADC growth inhibitory ADC compounds that specifically target HER2
  • HER2 overexpressing cells are grown in a 1:1 mixture of F12 and DMEM medium supplemented with 10% fetal bovine serum, glutamine and penicillin streptomycin.
  • the cells are plated at 20,000 cells in a 35 mm cell culture dish (2 mls/35 mm dish) and the test compound is added at various concentrations. After six days, the number of cells, compared to untreated cells is counted using an electronic COULTERTM cell counter.
  • Those ADC which inhibit cell growth by about 20-100% or about 50-100% may be selected as growth inhibitory compounds.
  • the PI uptake assay uses cells isolated from the breast tumor tissue of a transgenic animal. According to this assay, the cells are cultured in Dulbecco's Modified Eagle Medium (D-MEM): Ham's F-12 (50:50) supplemented with 10% heat-inactivated FBS (Hyclone) and 2 mM L-glutamine. Thus, the assay is performed in the absence of complement and immune effector cells. The cells are seeded at a density of 3 ⁇ 106 per dish in 100 ⁇ 20 mm dishes and allowed to attach overnight.
  • D-MEM Dulbecco's Modified Eagle Medium
  • the medium is then removed and replaced with fresh medium alone or medium containing various concentrations of the compound.
  • the cells are incubated for a 3-day time period. Following each treatment, monolayers are washed with PBS and detached by trypsinization. Cells are then centrifuged at 1200 rpm for 5 minutes at 4° C., the pellet resuspended in 3 ml cold Ca 2+ binding buffer (10 mM Hepes, pH 7.4, 140 mM NaCl, 2.5 mM CaCl 2 ) and aliquoted into 35 mm strainer-capped 12 ⁇ 75 mm tubes (1 ml per tube, 3 tubes per treatment group) for removal of cell clumps. Tubes then receive PI (10 ⁇ g/ml).
  • Samples may be analyzed using a FACSCANTM flow cytometer and FACSCONVERTTM CellQuest software (Becton Dickinson). Those compounds which induce statistically significant levels of cell death as determined by PI uptake may be selected as cell death-inducing compounds.
  • an annexin binding assay using cells established from the breast tumor tissue of the transgenic animal is performed.
  • the cells are cultured and seeded in dishes as discussed in the preceding paragraph.
  • the medium is then removed and replaced with fresh medium alone or medium containing 10 ⁇ g/ml of the antibody-drug conjugate (ADC).
  • ADC antibody-drug conjugate
  • monolayers are washed with PBS and detached by trypsinization.
  • Cells are then centrifuged, resuspended in Ca 2+ binding buffer and aliquoted into tubes as discussed above for the cell death assay. Tubes then receive labeled annexin (e.g.
  • annexin V-FITC (1 ⁇ g/ml). Samples may be analyzed using a FACSCANTM flow cytometer and FACSCONVERTTM CellQuest software (Becton Dickinson). Those compounds which induce statistically significant levels of annexin binding relative to control are selected as apoptosis-inducing compounds.
  • the cytotoxic or cytostatic activity of an antibody-drug conjugate is measured by: exposing mammalian cells having tumor-associated antigens or receptor proteins to the antibody of the ADC in a cell culture medium; culturing the cells for a period from about 6 hours to about 5 days; and measuring cell viability.
  • Cell-based in vitro assays were used to measure viability, i.e. proliferation (IC 50 ), cytotoxicity (EC 50 ), and induction of apoptosis (caspase activation) of the ADC.
  • the in vitro potency of antibody-drug conjugates was measured by a cell proliferation assay ( FIGS. 1-4 ).
  • the CellTiter-Glo® Luminescent Cell Viability Assay is a commercially available (Promega Corp., Madison, Wis.), homogeneous assay method based on the recombinant expression of Coleoptera luciferase (U.S. Pat. No. 5,583,024; U.S. Pat. No. 5,674,713; U.S. Pat. No. 5,700,670).
  • This cell proliferation assay determines the number of viable cells in culture based on quantitation of the ATP present, an indicator of metabolically active cells (Crouch et al (1993) J. Immunol. Meth.
  • the CellTiter-Glo® Assay was conducted in 96 well format, making it amenable to automated high-throughput screening (HTS) (Cree et al (1995) AntiCancer Drugs 6:398-404).
  • HTS high-throughput screening
  • the homogeneous assay procedure involves adding the single reagent (CellTiter-Glo® Reagent) directly to cells cultured in serum-supplemented medium. Cell washing, removal of medium and multiple pipetting steps are not required.
  • the system detects as few as 15 cells/well in a 384-well format in 10 minutes after adding reagent and mixing.
  • the homogeneous “add-mix-measure” format results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present.
  • the amount of ATP is directly proportional to the number of cells present in culture.
  • the CellTiter-Glo® Assay generates a “glow-type” luminescent signal, produced by the luciferase reaction, which has a half-life generally greater than five hours, depending on cell type and medium used. Viable cells are reflected in relative luminescence units (RLU).
  • the substrate, Beetle Luciferin is oxidatively decarboxylated by recombinant firefly luciferase with concomitant conversion of ATP to AMP and generation of photons.
  • This cell proliferation assay an be used with various multiwell formats, e.g. 96 or 384 well format. Data can be recorded by luminometer or CCD camera imaging device. The luminescence output is presented as relative light units (RLU), measured over time.
  • RLU relative light units
  • FIG. 1 shows the potency measurements at increasing concentrations of trastuzumab-SPP-DM1, trastuzumab-SPDP-DM1 and trastuzumab-SMCC-DM1 after a 3 day treatment on SK-BR-3 (HER2 3+) breast tumor cells.
  • FIG. 2 shows the potency measurements of at increasing concentrations of trastuzumab-SPP-DM1, trastuzumab-SPDP-DM1 and trastuzumab-SMCC-DM1 after a 3 day treatment on BT-474 (HER2 3+) breast tumor cells.
  • FIG. 1 shows the potency measurements at increasing concentrations of trastuzumab-SPP-DM1, trastuzumab-SPDP-DM1 and trastuzumab-SMCC-DM1 after a 3 day treatment on BT-474 (HER2 3+) breast tumor cells.
  • FIG. 3 shows the potency measurements of at increasing concentrations of trastuzumab-SPP-DM1, trastuzumab-SPDP-DM1 and trastuzumab-SMCC-DM1 after a 3 day treatment on MCF7 (HER2 low) breast tumor cells.
  • FIG. 4 shows the potency measurements at increasing concentrations of trastuzumab-SPP-DM1, trastuzumab-SPDP-DM1 and trastuzumab-SMCC-DM1 after a 3 day treatment on MDA-MB-468 (HER2 negative) breast tumor cells.
  • IC 50 values were established for SK-BR-3 and BT-474 which are known to overexpress HER2 receptor protein.
  • a lot of conjugate trastuzumab-SPP-DM1 with 2.8 DM1 per trastuzumab (drug/Ab) gave a mean IC 50 of 14.4 ⁇ g/ml with a range 9.1 to 22.3 ⁇ g/ml for 6 experiments against SK-BR-3 cells and a mean IC 50 of 51.7 ⁇ g/ml with a range 28.7 to 63.1 ⁇ g/ml for 4 experiments against BT-474 cells.
  • trastuzumab-SMCC-DM1 with 2.7 DM1 per trastuzumab (drug/Ab) gave a mean IC 50 of 15.2 ⁇ g/ml with a range 12.6 to 18.8 ⁇ g/ml for 4 experiments against SK-BR-3 cells and a mean IC 50 of 94.9 ⁇ g/ml with a range 75.2 to 114.6 ⁇ g/ml for 2 experiments against BT-474 cells.
  • the conjugates were inactive against cells MCF7 and MDA-MB-468 which do not overexpress HER2.
  • FIG. 17 shows an in vitro, cell proliferation assay (Example 5) with HT1080EphB2 (C8) cells treated with antiEphB2R 2H9 antibody-drug conjugates: 2H9-SPP-DM1 (IC 50 80 ng/ml), and 2H9-SMCC-DM1 (IC 50 50 ng/ml).
  • FIG. 5 shows the serum clearance in beige nude mice without tumors of trastuzumab-SMCC-DM1 vs. trastuzumab-SPP-DM1, measuring conjugate and total antibody serum concentration at six time points over 7 days.
  • a difference in the amount of total antibody and ADC indicates cleavage of the linker and separation of the antibody from its DM1 moiety.
  • SMCC-linked ADC remained intact in vivo longer than did SPP-linked conjugates.
  • FIG. 6 shows the stability over time in nude mice without tumors of the conjugates: trastuzumab-SPDP-DM1, trastuzumab-SPP-DM1, trastuzumab-SPP-DM3, trastuzumab-SPP-DM4, and trastuzumab-SMCC-DM1, measuring serum concentration at six time points over 7 days.
  • ADC with the SMCC linker were more stable in vivo than were conjugates linked by SPP or SPDP, although trastuzumab-SMCC-DM1 had approximately the same stability as the most hindered disulfide conjugate, trastuzumab-SPP-DM4.
  • FIGS. 8 and 9 show the comparative stability and clearance profiles of a disulfide linker ADC (trastuzumab-SPP-DM1 and a non-disulfide linker ADC (trastuzumab-SMCC-DM1) in rats.
  • the parameters of the study include: trastuzumab- trastuzumab- Parameter SPP-DM1 SMCC-DM1 Vd (ml/kg 41 41 Clearance (ml/day/kg) 52 15 T 1 ⁇ 2 alpha 0.09 0.15 T 1 ⁇ 2 beta 0.7 0.85 T 1 ⁇ 2 gamma (days) 2.6 5.5
  • the efficacy of the antibody-drug conjugates of the invention may be measured in vivo by implanting allografts or xenografts of cancer cells in rodents and treating the tumors with ADC. Variable results are to be expected depending on the cell line, the specificity of antibody binding of the ADC to receptors present on the cancer cells, dosing regimen, and other factors.
  • the in vivo efficacy of anti-HER2 ADC was measured by a high expressing HER2 transgenic explant mouse model. An allograft was propagated from the Fo5 mmtv transgenic mouse which does not respond to, or responds poorly to, HERCEPTIN therapy. Subjects were treated once with ADC and monitored over 3-6 weeks to measure the time to tumor doubling, log cell kill, and tumor shrinkage. Follow up dose-response and multi-dose experiments were conducted.
  • transgenic mice were produced using a HER2 cDNA plasmid in which an upstream ATG was deleted in order to prevent initiation of translation at such upstream ATG codons, which would otherwise reduce the frequency of translation initiation from the downstream authentic initiation codon of HER2 (for example, see Child et al (1999) J. Biol. Chem. 274: 24335-24341). Additionally, a chimeric intron was added to the 5′ end, which should also enhance the level of expression as reported earlier (Neuberger and Williams (1988) Nucleic Acids Res. 16: 6713; Buchman and Berg (1988) Mol. Cell. Biol. 8: 4395; Brinster et al (1988) Proc. Natl.
  • the chimeric intron was derived from a Promega vector, pCI-neo mammalian expression vector (bp 890-1022).
  • the cDNA 3′-end is flanked by human growth hormone exons 4 and 5, and polyadenylation sequences.
  • FVB mice were used because this strain is more susceptible to tumor development.
  • the promoter from MMTV-LTR was used to ensure tissue-specific HER2 expression in the mammary gland. Animals were fed the AIN 76A diet in order to increase susceptibility to tumor formation (Rao et al (1997) Breast Cancer Res. and Treatment 45:149-158).
  • FIGS. 10-13 show that the ADC have strong anti-tumor activity in the allograft of a HER2 positive tumor (Fo5) that originally arose in an MMTV-HER2 transgenic mouse.
  • the antibody alone e.g. trastuzumab
  • trastuzumab does not have significant anti-tumor activity in this model (Erickson et al U.S. Pat. No. 6,632,979).
  • FIGS. 10 and 11 the growth of the tumors was retarded by treatment with ADC as compared to control (Vehicle) level of growth. Tumor growth was slowed most by treatment with trastuzumab-SMCC-DM1 and trastuzumab-SIAB-DM1 conjugates.
  • FIGS. 10-13 show that the ADC have strong anti-tumor activity in the allograft of a HER2 positive tumor (Fo5) that originally arose in an MMTV-HER2 transgenic mouse.
  • the antibody alone e.g
  • the trastuzumab-SMCC-DM1 conjugate slowed tumor growth more than conjugates with SPP linkers, i.e. more potency, whether measured as doubling time of tumors in nude mice or as Log Cell Kill corresponding to the doubling time measurements shown ( FIG. 13 ).
  • the in vivo efficacy of anti-CD22 ADC was measured with a mouse tumor xenograft model.
  • Groups of eight SCID mice with 20 million Bjab-luc (luciferase expressing Bjab cells) xenograft tumor cells per mouse were dosed once at day 1 (except where noted) with an anti-CD22 antibody-drug conjugate, or a naked antibody (Example 8).
  • Tr-SMCC-DM1 200 4.2 3.2 3 7A2-SMCC-DM1 200 3.8 3.6 6 5E8-SMCC-DM1 200 3.8 3.6 10
  • RFB4-SMCC-DM1 200 3.2 4.25 18
  • RFB4-SMCC-DM1 405 10 2.75 55 (dosed 3 ⁇ at 1, 7, 14 days) 7A2 — 4 — 3 5E8 — 4 — 3 RFB4 — 4 — 3
  • the time for tumor size to double was measured (MTD, mean tumor doubling time).
  • the three naked anti-CD22 antibodies showed essentially no efficacy relative to a non-specific binding ADC (trastuzumab-SMCC-DM1).
  • the corresponding conjugates all showed the effect of significantly retarding tumor growth.
  • the effect of multiple dosing was established with RFB4-SMCC-DM1 where the MTD for singly dosed mice was 18 days whereas the MTD for mice dosed three times, at days 1, 7, and 14, was 55 days. Complete remission of tumor occurred in all 8 mice in the triple-dosed group.
  • antiCD22-SMCC-DM1 conjugates with antibodies 12F7, 9A8, 8C9, 8G10, 3F11, 10D2, 6C9, 14D1, and 11H10, showed shrinkage of initial tumor volume or retardation of tumor growth relative to control (trastuzumab-SMCC-DM1), after 7 days following a single dose (400 ⁇ g DM1/m2) in SCID mice with 20 million Bjab-luc xenograft tumor cells per mouse.
  • AntiCD22 conjugates RFB4-SMCC-DM1, 5E8-SMCC-DM1, and 7A2-SMCC-DM1 were also effective in retardation of tumor growth relative to control (trastuzumab-SMCC-DM1), after 11 days following a single dose (200 ⁇ g DM1/m2) in SCID mice with 5 million Ramos RA1 xenograft tumor cells per mouse.
  • the conjugate RFB4-SMCC-DM1 was studied at three different drug loadings on groups of ten SCID mice with Bjab-luc xenografts (Example 8).
  • the low (1.95) and medium (3.7) drug loaded conjugates each showed the effect of significantly retarding tumor growth, with an MTD of about 15 days.
  • the high loaded (6.75) conjugate did not show an effect significantly different than control conjugate GP120-SMCC-DM1, or naked antibody RFB4.
  • AntiCD19-SMCC-DM1 and antiCD22-SMCC-DM1 conjugates did not show in vivo activity in a Raji cell mouse tumor xenograft model.
  • Other antiCD19 and antiCD22 conjugates may have in vivo activity against other cancer cell tumor models.
  • conjugate SN8 antiCD79b-SMCC-DM1 gave partial remission (PR) in 4 animals and complete remission (CR) in 2 animals.
  • Conjugate 17A7 antiCD79b-SMCC-DM1 gave CR in 1 animal.
  • Conjugate 8H9 antiCD79a-SMCC-DM1 gave PR in 2 animals and CR in 1 animal.
  • antiCD79b-SMCC-DM1 conjugates with antibodies 2F2, 5C3, 7H7, 8D11, 15E4, and 16C11, showed retardation of tumor growth or shrinkage of initial tumor volume, relative to control (trastuzumab-SMCC-DM1), after 8 days following a single dose (192 ⁇ g DM1/m2) in CB17 ICR SCID mice with 20 million Bjab-luc xenograft tumor cells per mouse.
  • mice administered with antiCD79b-SMCC-DM1 were dosed at day 1 with samples in the table below (Example 8).
  • the antiCD79b-SMCC-DM1 was dosed at levels of 0.5, 2.0, and 3.64 mg Ab/kg mouse.
  • Vehicle control (PBS) 4 antiCD79b-SMCC-DM1 32 0.5 10 antiCD79b-SMCC-DM1 130 2.0 35 antiCD79b-SMCC-DM1 236 3.64 >70
  • TENB2 is a tumor antigen that is shown to be almost exclusively expressed in the human prostate and overexpressed in human prostate tumors (Glynne-Jones et al (2001) Int J Cancer. October 15;94(2):178-84).
  • PC3-TVA-919cv1:5 is a human prostate cancer cell line expressing high level of TENB2.
  • mice were subcutaneously injected with 5 million PC3-TVA-919 high expressor or medium expressor cells in a volume of 0.2 ml per mouse. Cells were suspended in HBSS. When mean tumor size reached 100-200 mm 3 , the mice were randomly grouped into 8 groups of 8-10 mice each and give a single IV treatment of the samples below (Example 8).
  • Murine anti-TENB2-DM1 conjugates showed anti-tumor efficacy against PC3-TENB2 tumors, relative to negative control and vehicle control.
  • Murine 10H1 antiNaPi3b-SMCC-DM1 conjugate showed no anti-tumor efficacy against PC3-NaPi3b tumors, relative to negative control and vehicle control.
  • Other antibody variants of antiNaPi3b conjugates may have in vivo activity against PC3-NaPi3b tumors, or other cancer cell lines.
  • a 5-day acute toxicity study in adolescent female rats was conducted by a single injection of trastuzumab-SMCC-DM1 (two doses: 1860 and 3260 ⁇ g DM1/m 2 ), a comparison disulfide ADC, trastuzumab-SPP-DM1 (two doses: 1860 and 3260 ⁇ g DM1/m 2 ), free DM1 maytansine (thiol) and a control Vehicle (day 0).
  • Body weight was measured daily.
  • Clinical chemistry, serum enzymes and hematology analysis was conducted on days 3 and 5; concluding with complete necropsy with histopathological assessment. Toxicity signals included the clinical observation of weight loss.
  • FIG. 14 shows the changes in body weight (grams) over 5 days. Rats receiving the disulfide ADC, trastuzumab-SPP-DM1 showed a marked, dose-dependent toxicity, indicated by lethality at the higher dose and a decline in body weight at the lower dose. In contrast, rats receiving trastuzumab-SMCC-DM1 gained weight, with the lower dosed rats showing no decline in the rate of weight gain, relative to the placebo Vehicle-dosed rats. Rats dosed at the higher level of trastuzumab-SMCC-DM1 also gained weight, comparable to the free DM1 cytotoxin.
  • Hepatotoxicity was measured by elevated liver enzymes, increased numbers of mitotic and apoptotic figures and hepatocyte necrosis. Hematolymphoid toxicity was observed by depletion of leukocytes, primarily granuloctyes (neutrophils), and/or platelets, and lymphoid organ involvement, i.e. atrophy or apoptotic activity. Toxicity was also noted by gastrointestinal tract lesions such as increased numbers of mitotic and apoptotic figures and degenerative entercolitis.
  • Enzymes indicative of liver injury that were studied include:
  • FIGS. 15 and 16 show that rats exposed to 22.3 mg/kg trastuzumab-SPP-DM1, (Group 2) demonstrated the most severe clinical toxicity in this five day acute toxicity study. These animals showed the most profound loss of body weight, elevations of liver function tests, leuko- and thrombocytopenia and morphologic evidence of toxicity against hematolymphoid tissues. The extent of toxicity was similar compared to that encountered in previous studies using a dose of 25 mg/kg. In contrast, animals in Groups 3 and 4 given trastuzumab-SMCC-DM1 at 10 and 25 mg/kg, respectively, were indistinguishable from vehicle-treated animals based on clinical pathology and body weight data. Morphologically, these animals showed a mildly increased number of mitotic figures in the liver, however, peripheral lymphoid and hematopoietic tissues were within normal limits.
  • trastuzumab-SMCC-DM1 Group 5 animals, 50 mg/kg trastuzumab-SMCC-DM1, showed evidence of toxicity. However, with the exception of one liver function test (ALT), the severity of toxicity was less than in animals receiving 50% of the same drug dose as trastuzumab-SPP-DM1 (Group 2). At about the same dose, trastuzumab-SMCC-DM1 (Group 4, 22.3 mg/kg) showed about 25% of the AST level as trastuzumab-SPP-DM1 (Group 2, 25 mg/kg). By day 5 of this study, Group 5 animals showed increases in body weights (following a transitory loss during days 3 and 4), a decreasing serum bilirubin and a rising platelet count ( FIG. 15 ).
  • Toxicity and safety of ADC administered to Cynomolgus monkeys may be assessed.
  • a toxicity/safety study of the antibody-drug conjugate, trastuzumab-SMCC-DM1 was conducted in Cynomolgus monkeys. Three groups of monkeys were studied to assess the toxicity of trastuzumab-SMCC-DM1 administered via intravenous injection at escalating doses, relative to control (Vehicle).
  • Group 1 (4 subjects) received only Vehicle (PBS, pH 6.5, i.e. formulation minus ADC) at day 1 and day 22, followed by necropsy at day 36.
  • Hepatotoxicity was inferred by measurement of the elevated liver enzymes from the Rodent Toxicity study.
  • Cynomolgus monkeys were dosed with Vehicle (Group 1) and trastuzumab-SMCC-DM1 (Group 2: 4900 ⁇ g/m 2 ; Group 3: 7200 ⁇ g/m 2 .
  • Liver enzyme AST, platelet counts, white blood cells, absolute neutrophil, red blood cells, reticulocytes, and a comparison of 2 IV dose regimens were measured for trastuzumab-SMCC-DM1 in Cynomolgus Monkeys (Example 10).
  • Therapeutic antibody-drug conjugates may be administered by any route appropriate to the condition to be treated.
  • the ADC will typically be administered parenterally, i.e. infusion, subcutaneous, intramuscular, intravenous, intradermal, intrathecal, bolus, intratumor injection or epidural (Shire et al (2004) J. Pharm. Sciences 93(6):1390-1402).
  • Pharmaceutical formulations of therapeutic antibody-drug conjugates (ADC) are typically prepared for parenteral administration with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form.
  • An antibody-drug conjugate (ADC) having the desired degree of purity is optionally mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers, in the form of a lyophilized formulation or an aqueous solution (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.).
  • Acceptable parenteral vehicles, diluents, carriers, excipients, and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glut
  • Zn-protein complexes Zn-protein complexes
  • non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol (PEG).
  • TWEENTM TWEENTM
  • PLURONICSTM polyethylene glycol
  • PEG polyethylene glycol
  • An exemplary formulation of an ADC such as trastuzumab-SMCC-DM1 contains about 100 mg/ml of trehalose (2-(hydroxymethyl)-6-[3,4,5-trihydroxy-6-(hydroxymethyl)tetrahydropyran-2-yl]oxy-tetrahydropyran-3,4,5-triol; C 12 H 22 O 11 ; CAS Number 99-20-7) and about 0.1% TWEENTM 20 (polysorbate 20; dodecanoic acid 2-[2-[3,4-bis(2-hydroxyethoxy)tetrahydrofuran-2-yl]-2-(2-hydroxyethoxy)ethoxy]ethyl ester; C 26 H 50 O 10 ; CAS Number 9005-64-5) at approximately pH 6.
  • compositions of a therapeutic antibody-drug conjugate may contain certain amounts of unreacted drug moiety (D), antibody-linker intermediate (Ab-L), and/or drug-linker intermediate (D-L), as a consequence of incomplete purification and separation of excess reagents, impurities, and by-products, in the process of making the ADC; or time/temperature hydrolysis or degradation upon storage of the bulk ADC or formulated ADC composition.
  • a formulation of the ADC trastuzumab-SMCC-DM1 may contain a detectable amount of free drug DM1.
  • it may contain a detectable amount of drug-linker intermediate DM1-SMCC.
  • trastuzumab may contain a detectable amount of the antibody, trastuzumab.
  • An exemplary formulation of trastuzumab-SMCC-DM1 may contain up to 10% molar equivalent of DM1-SMCC.
  • DM1-SMCC IC 50 0.05 ⁇ M
  • DM1-SMCC IC 50 0.05 ⁇ M
  • free drug DM1 IC 50 0.0045 ⁇ M
  • the active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semi permeable matrices of solid hydrophobic polymers containing the ADC, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and gamma-ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( ⁇ )-3-hydroxybutyric acid.
  • the formulations to be used for in vivo administration must be sterile, which is readily accomplished by filtration through sterile filtration membranes.
  • the formulations include those suitable for the foregoing administration routes.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, Pa.). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Aqueous suspensions contain the active materials (ADC) in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan
  • the aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.
  • compositions of ADC may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • Subcutaneous (bolus) administration may be effected with about 1.5 ml or less of total volume and a concentration of about 100 mg ADC per ml.
  • the subcutaneous route may be employed, such as by pre-filled syringe or autoinjector device technology.
  • the initial pharmaceutically effective amount of ADC administered per dose will be in the range of about 0.01-100 mg/kg, namely about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day.
  • human patients may be initially dosed at about 1.5 mg ADC per kg patient body weight.
  • the dose may be escalated to the maximally tolerated dose (MTD).
  • MTD maximally tolerated dose
  • the dosing schedule may be about every 3 weeks, but according to diagnosed condition or response, the schedule may be more or less frequent.
  • the dose may be further adjusted during the course of treatment to be at or below MTD which can be safely administered for multiple cycles, such as about 4 or more.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • formulations of ADC suitable for oral administration may be prepared as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the ADC.
  • the formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use.
  • sterile liquid carrier for example water
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • Exemplary unit dosage formulations contain a daily dose or unit daily sub-dose, or an appropriate fraction thereof, of the active ingredient.
  • the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
  • the antibody-drug conjugates (ADC) of the present invention may be used to treat various diseases or disorders, such as cancer and autoimmune conditions.
  • exemplary conditions or disorders include benign or malignant tumors; leukemia and lymphoid malignancies; other disorders such as neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic and immunologic disorders.
  • Cancer susceptible to ADC treatment include those which are characterized by the overexpression of certain tumor associated antigens or cell surface receptors, e.g. HER2
  • the ADC compounds which are identified in the animal models and cell-based assays can be further tested in tumor-bearing higher primates and human clinical trials.
  • Human clinical trials can be designed similar to the clinical trials testing the efficacy of the anti-HER2 monoclonal antibody HERCEPTIN in patients with HER2 overexpressing metastatic breast cancers that had received extensive prior anti-cancer therapy as reported by Baselga et al. (1996) J. Clin. Oncol. 14:737-744.
  • the clinical trial may be designed to evaluate the efficacy of an ADC in combination with known therapeutic regimens, such as radiation and/or chemotherapy involving known chemotherapeutic and/or cytotoxic agents (Pegram et al (1999) Oncogene 18:2241-2251).
  • cancer the disease or disorder to be treated is cancer.
  • cancer to be treated herein include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, gastrointestinal stromal tumor (GIST), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • GIST gastrointestinal stromal tumor
  • the cancer to be treated herein may be one characterized by excessive activation of an ErbB receptor, e.g. HER2. Such excessive activation may be attributable to overexpression or increased production of the ErbB receptor or an ErbB ligand.
  • a diagnostic or prognostic assay will be performed to determine whether the patient's cancer is characterized by excessive activation of an ErbB receptor. For example, ErbB gene amplification and/or overexpression of an ErbB receptor in the cancer may be determined.
  • Assays for determining such amplification/overexpression are available in the art and include the IHC, FISH and shed antigen assays described above.
  • levels of an ErbB ligand, such as TGF-alpha., in or associated with the tumor may be determined according to known procedures. Such assays may detect protein and/or nucleic acid encoding it in the sample to be tested.
  • ErbB ligand levels in the tumor may be determined using immunohistochemistry (IHC); see, for example, Scher et al. (1995) Clin. Cancer Research 1:545-550.
  • IHC immunohistochemistry
  • one may evaluate levels of ErbB ligand-encoding nucleic acid in the sample to be tested; e.g. via FISH, southern blotting, or PCR techniques.
  • ErbB2 overexpression may be analyzed by IHC, e.g.
  • Parrafin embedded tissue sections from a tumor biopsy may be subjected to the IHC assay and accorded a ErbB2 protein staining intensity criteria as follows: Score 0, no staining is observed or membrane staining is observed in less than 10% of tumor cells; Score 1+, a faint/barely perceptible membrane staining is detected in more than 10% of the tumor cells, the cells are only stained in part of their membrane; Score 2+, a weak to moderate complete membrane staining is observed in more than 10% of the tumor cells; Score 3+, a moderate to strong complete membrane staining is observed in more than 10% of the tumor cells. Those tumors with 0 or 1+ scores for ErbB2 overexpression assessment may be characterized as not overexpressing ErbB2, whereas those tumors with 2+ or 3+ scores may be characterized as overexpressing ErbB2.
  • FISH assays such as the INFORMTM (Ventana Co., Ariz.) or PATHVISIONTM (Vysis, Ill.) may be carried out on formalin-fixed, paraffin-embedded tumor tissue to determine the extent (if any) of ErbB2 overexpression in the tumor.
  • ErbB receptor or ErbB ligand overexpression or amplification may be evaluated using an in vivo diagnostic assay, e.g. by administering a molecule (such as an antibody) which binds the molecule to be detected and is tagged with a detectable label (e.g. a radioactive isotope) and externally scanning the patient for localization of the label.
  • a detectable label e.g. a radioactive isotope
  • an ADC for the prevention or treatment of disease, the appropriate dosage of an ADC will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the molecule is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the molecule is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. 0.1-20 mg/kg) of molecule is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • a typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • An exemplary dosage of ADC to be administered to a patient is in the range of about 0.1 to about 10 mg/kg of patient weight.
  • An exemplary dosing regimen comprises administering an initial loading dose of about 4 mg/kg, followed by a weekly maintenance dose of about 2 mg/kg of the anti-ErbB2 antibody. Other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • An antibody-drug conjugate may be combined in a pharmaceutical combination formulation, or dosing regimen as combination therapy, with a second compound having anti-cancer properties.
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the ADC of the combination such that they do not adversely affect each other.
  • the second compound may be a chemotherapeutic agent, cytotoxic agent, cytokine, growth inhibitory agent, anti-hormonal agent, aromatase inhibitor, protein kinase inhibitor, lipid kinase inhibitor, anti-androgen, antisense oligonucleotide, ribozyme, gene therapy vaccine, anti-angiogenic agent and/or cardioprotectant.
  • a chemotherapeutic agent such as a tubulin-forming inhibitor, a topoisomerase inhibitor, or a DNA binder.
  • the second compound may be an antibody which binds ErbB2 and blocks ligand activation of an ErbB receptor.
  • the second antibody may be monoclonal antibody 2C4 or humanized 2C4 “Omnitarg” (WO 01/00245).
  • the second antibody may be conjugated with a cytotoxic or chemotherapeutic agent, e.g., a maytansinoid, an auristatin, a calicheamicin, or a 1,8 bis-naphthalimide moiety.
  • a cytotoxic or chemotherapeutic agent e.g., a maytansinoid, an auristatin, a calicheamicin, or a 1,8 bis-naphthalimide moiety.
  • VEGF vascular endot
  • the combination therapy may be administered as a simultaneous or sequential regimen.
  • the combination may be administered in two or more administrations.
  • the combined administration includes coadministration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • treatment with an ADC of the present invention involves the combined administration of an anticancer agent identified herein, and one or more chemotherapeutic agents or growth inhibitory agents, including coadministration of cocktails of different chemotherapeutic agents, optionally along with treatment with an anti-ErbB2 antibody, such as trastuzumab.
  • Chemotherapeutic agents include Erlotinib HCl (CP-358774, TARCEVATM; Genentech/OSI), taxanes (such as paclitaxel and doxetaxel) and/or anthracycline antibiotics. Preparation and dosing schedules for such chemotherapeutic agents may be used according to manufacturers's instructions or as determined empirically by the skilled practitioner. Preparation and dosing schedules for such chemotherapy are also described in Chemotherapy Service Ed., M. C. Perry, Williams & Wilkins, Baltimore, Md. (1992).
  • the anticancer agent may be combined with an anti-hormonal compound; e.g., an anti-estrogen compound such as tamoxifen; an anti-progesterone such as onapristone (EP 616812); or an anti-androgen such as flutamide, in dosages known for such molecules.
  • an anti-hormonal compound e.g., an anti-estrogen compound such as tamoxifen; an anti-progesterone such as onapristone (EP 616812); or an anti-androgen such as flutamide
  • an anti-hormonal compound such as tamoxifen
  • an anti-progesterone such as onapristone (EP 616812)
  • an anti-androgen such as flutamide
  • Suitable dosages for any of the above coadministered agents are those presently used and may be lowered due to the combined action (synergy) of the newly identified agent and other chemotherapeutic agents or treatments.
  • the combination therapy may provide “synergy” and prove “synergistic”, i.e. the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect may be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered by alternation or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect may be attained when the compounds are administered or delivered sequentially, e.g. by different injections in separate syringes.
  • an effective dosage of each active ingredient is administered sequentially, i.e. serially, whereas in combination therapy, effective dosages of two or more active ingredients are administered together.
  • the invention includes novel and unobvious compounds produced by a process comprising contacting a compound of this invention with a mammal for a period of time sufficient to yield a metabolic product thereof.
  • Metabolite products may be identified by preparing a radiolabelled (e.g. 14 C or 3 H) ADC, administering it parenterally in a detectable dose (e.g. greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples.
  • a detectable dose e.g. greater than about 0.5 mg/kg
  • an animal such as rat, mouse, guinea pig, monkey, or to man
  • a detectable dose e.g. greater than about 0.5 mg/kg
  • an animal such as rat, mouse, guinea pig, monkey, or to man
  • sufficient time for metabolism to occur typically about 30 seconds to 30 hours
  • isolating its conversion products from the urine, blood or other biological samples typically about 30 seconds to 30 hours
  • the metabolite structures are determined in conventional fashion,
  • Metabolites include the products of in vivo cleavage of the ADC where cleavage of any bond occurs that links the drug moiety to the antibody. Metabolic cleavage may thus result in the naked antibody, or an antibody fragment. The antibody metabolite may be linked to a part, or all, of the linker. Metabolic cleavage may also result in the production a drug moiety or part thereof. The drug moiety metabolite may be linked to a part, or all, of the linker.
  • an article of manufacture containing ADC and materials useful for the treatment of the disorders described above.
  • the article of manufacture comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, or blister pack.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds an antibody-drug conjugate (ADC) composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • ADC antibody-drug conjugate
  • At least one active agent in the composition is an ADC.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as cancer.
  • the label or package inserts indicates that the composition comprising the antibody which binds ErbB2 can be used to treat cancer which expresses an ErbB receptor selected from the group consisting of epidermal growth factor receptor (EGFR), ErbB2, ErbB3 and ErbB4.
  • the label or package insert may indicate that the patient to be treated is one having cancer characterized by excessive activation of an ErbB receptor selected from EGFR, ErbB2, ErbB3 or ErbB4.
  • the cancer may be one which overexpresses one of these receptors and/or which overexpresses an ErbB ligand (such as TGF- ⁇ ).
  • the label or package insert may also indicate that the composition can be used to treat cancer, wherein the cancer is not characterized by overexpression of the ErbB2 receptor.
  • the package insert may indicate that the ADC composition can be used also to treat hormone independent cancer, prostate cancer, colon cancer or colorectal cancer.
  • the article of manufacture may comprise (a) a first container with a compound contained therein, wherein the compound comprises an ADC of the present invention in which the antibody of the ADC is a first antibody which binds ErbB2 and inhibits growth of cancer cells which overexpress ErbB2; and (b) a second container with a compound contained therein, wherein the compound comprises a second antibody which binds ErbB2 and blocks ligand activation of an ErbB receptor, or a conjugate of this second antibody with a maytansinoid.
  • the article of manufacture in this embodiment may further comprise a package insert indicating that the first and second compounds can be used to treat cancer.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • Ringer's solution such as phosphate
  • the murine monoclonal antibody 4D5 which specifically binds the extracellular domain of ErbB2 was produced as described in Fendly et al (1990) Cancer Research 50:1550-1558. Briefly, NIH 3T3/HER2-3 400 cells (expressing approximately 1 ⁇ 10 5 ErbB2 molecules/cell) produced as described in Hudziak et al (1987) Proc. Natl. Acad. Sci. (USA) 84:7158-7163 were harvested with phosphate buffered saline (PBS) containing 25 mM EDTA and used to immunize BALB/c mice. The mice were given injections i.p. of 10 7 cells in 0.5 ml PBS on weeks 0, 2, 5 and 7.
  • PBS phosphate buffered saline
  • mice with antisera that immunoprecipitated 32 P-labeled ErbB2 were given i.p. injections of a wheat germ agglutinin-Sepharose (WGA) purified ErbB2 membrane extract on weeks 9 and 13. This was followed by an i.v. injection of 0.1 ml of the ErbB2 preparation and the splenocytes were fused with mouse myeloma line X63-Ag8.653. Hybridoma supernatants were screened for ErbB2-binding by ELISA and radioimmunoprecipitation.
  • WGA wheat germ agglutinin-Sepharose
  • the murine monoclonal antibody 4D5 was humanized, using a “gene conversion mutagenesis” strategy, as described in U.S. Pat. No. 5,821,337, the entire disclosure of which is hereby expressly incorporated by reference.
  • the humanized monoclonal antibody 4D5 used in the following experiments is designated huMAb4D5-8. This antibody is of IgG1 isotype.
  • HERCEPTIN® (huMAb4D5-8, rhuMAb HER2, U.S. Pat. No. 5,821,337) antibody
  • 50 mL MES buffer 25 mM MES, 50 mM NaCl, pH 5.6
  • a cation exchange column Sepharose S, 15 cm ⁇ 1.7 cm
  • Trastuzumab was eluted by raising the NaCl concentration of the buffer to 200 mM.
  • Fractions containing the antibody were pooled, diluted to 10 mg/mL, and dialyzed into a buffer containing 50 mm potassium phosphate, 50 mM NaCl, 2 mM EDTA, pH 6.5.
  • trastuzumab was derivatized with N-succinimidyl-4-(2-pyridylthio)pentanoate to introduce dithiopyridyl groups.
  • Trastuzumab (376.0 mg, 8 mg/mL) in 44.7 mL of 50 mM potassium phosphate buffer (pH 6.5) containing NaCl (50 mM) and EDTA (1 mM) was treated with SPP (5.3 molar equivalents in 2.3 mL ethanol).
  • the reaction mixture was gel filtered through a Sephadex G25 column equilibrated with 35 mM sodium citrate, 154 mM NaCl, 2 mM EDTA.
  • Antibody containing fractions were pooled and assayed. The degree of modification of the antibody was determined as described above. Recovery of the modified antibody (trastuzumab-SPP-Py) was 337 mg (89.7%) with 4.5 releasable 2-thiopyridine groups linked per antibody (p′).
  • Trastuzumab-SPP-Py (337.0 mg, 9.5 ⁇ mols of releasable 2-thiopyridine groups) was diluted with the above 35 mM sodium citrate buffer, pH 6.5, to a final concentration of 2.5 mg/mL.
  • DM1 N 2′ -deacetyl-N 2′ -(3-mercapto-1-oxopropyl)-maytansine
  • FIG. 1 (1.7 equivalents, 16.1 ⁇ mols) in 3.0 mM dimethylacetamide (DMA, 3% v/v in the final reaction mixture) was then added to the antibody solution.
  • DMA dimethylacetamide
  • the reaction was loaded on a Sephacryl S300 gel filtration column (5.0 cm ⁇ 90.0 cm, 1.77 L) equilibrated with 35 mM sodium citrate, 154 mM NaCl, pH 6.5.
  • the flow rate was 5.0 L/min and 65 fractions (20.0 mL each) were collected.
  • a major peak centered around fraction No. 47 ( FIG. 3 ).
  • the major peak comprises monomeric trastuzumab-SPP-DM1.
  • Fractions 44-51 were pooled and assayed.
  • the number of DM1 drug molecules linked per antibody molecule (p′) was determined by measuring the absorbance at 252 nm and 280 nm, and found to be 3.7 drug molecules per antibody molecule.
  • trastuzumab was derivatized with (Succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate, SMCC, Pierce Biotechnology, Inc) to introduce the SMCC linker.
  • Trastuzumab was purified from HERCEPTIN® as in Example 2 and buffer-exchange treated at 20 mg/mL in 50 mM potassium phosphate/50 mM sodium chloride/2 mM EDTA, pH 6.5 with 7.5 to 10 molar equivalents of SMCC (20 mM in DMSO or DMA (dimethylacetamide), 6.7 mg/mL).
  • the reaction mixture was filtered through a Sephadex G25 column equilibrated with 50 mM potassium phosphate/50 mM sodium chloride/2 mM EDTA, pH 6.5. Alternatively, the reaction mixture was gel filtered with 30 mM citrate and 150 mM sodium chloride at pH 6. Antibody containing fractions were pooled and assayed. Recovery of trastuzumab-SMCC was 88%.
  • the reaction was stirred at ambient temperature under argon for 4 to about 16 hours.
  • reaction mixture was gel filtered with 10 mM succinate and 150 mM sodium chloride at pH 5.
  • the DM1/trastuzumab ratio (p) was 3.1, as measured by the absorbance at 252 nm and at 280 nm.
  • the drug to antibody ratio (p) may also be measured by mass spectrometry.
  • Conjugation may also be monitored by SDS polyacrylamide gel electrophoresis. Aggregation may be assessed by laser light scattering analysis.
  • ADC Antibody-drug average drug conjugate
  • Ab antibody
  • DM1/Ab 10H-SMCC-DM1 10H1 (murine) anti-NaPi3b NaPi3b 5.0 2H9-SMCC-DM1 2H9 Mab anti-EphB2R 4.0 ragweed-SMCC-DM1 ragweed 2.7 antiTENB2-SMCC-DM1 anti-TENB2-3416#2 Mouse MAb TENB2 2.5 anti-TENB2 20D1 Fc Chimeric Human Mouse anti-TENB2 TENB2 3.32 Chimera-SMCC-DM1 antiCD19-SMCC-DM1 antiCD19 (mIgG1 isotype) CD19 5.05 2H7-SMCC-DM1 antiCD20 (mIgG2a isotype) CD20 2.44, 3.84 (two lots) CD 20LC-SMCC-DM1 antiCD20
  • the antibody may be reduced with a reducing agent such as dithiothreitol (DTT) or TCEP, in the presence of EDTA. After incubating for about one hour at 37° C., the pH is adjusted to about 7 with 100 mM potassium phosphate.
  • the reduced antibody is modified by the bis-maleimido reagent BM(PEO)4 (Pierce Chemical), leaving an unreacted maleimido group on the surface of the antibody.
  • BM(PEO)4 This may be accomplished by dissolving BM(PEO)4 in a 50% ethanol/water mixture to a concentration of 10 mM and adding a tenfold molar excess to a solution containing antibody in phosphate buffered saline at a concentration of approximately 1.6 mg/ml (10 ⁇ M) and allowing it to react for 1 hour to form Ab-BMPEO.
  • Excess BM(PEO)4 is removed by gel filtration (HiTrap column, Pharmacia) in 30 mM citrate, pH 6 with 150 mM NaCl buffer.
  • An approximate 10 fold molar excess DM1 is dissolved in dimethyl acetamide (DMA) and added to the Ab-BMPEO intermediate.
  • DMA dimethyl acetamide
  • DMF Dimethyl formamide
  • antibody-drug conjugates with the BMPEO linker and DM1 drug moiety were prepared, including: Antibody-drug average drug conjugate (ADC) antibody (Ab) antigen loading (DM1/Ab) trastuzumab- trastuzumab HER2 2.94 BM(PEO)-DM1 (Herceptin ®) CD120-BMPEO4-DM1 Mouse anti- GP120 2 GP120 MAb RFB4-BMPEO4-DM1 antiCD22 CD22 3.7 and 4.25 (two lots)
  • mice in six Groups of four animals were studied. At Day 0, each mouse received a single 2 mg/kg dose of ADC in 200 ⁇ l aqueous carrier, except the Vehicle Group which received only carrier. Blood was collected by cardiac puncture under anesthesia at each time point (5 minutes, 1 hour, 6 hours, 24 hours, 72 hours, and 168 hours) post-dose. The serum was isolated and antibody and ADC were measured.
  • mice suitable for transgenic experiments can be obtained from standard commercial sources such as Taconic (Germantown, N.Y.). Many strains are suitable, but FVB female mice are preferred because of their higher susceptibility to tumor formation. FVB males were used for mating and vasectomized CD.1 studs were used to stimulate pseudopregnancy. Vasectomized mice can be obtained from any commercial supplier. Founders were bred with either FVB mice or with 129/BL6 x FVB p53 heterozygous mice. The mice with heterozygosity at p53 allele were used to potentially increase tumor formation. However, this has proven unnecessary. Therefore, some F1 tumors are of mixed strain. Founder tumors are FVB only. Six founders were obtained with some developing tumors without having litters.
  • Animals having tumors (allograft propagated from Fo5 mmtv transgenic mice) were treated with a single injection of a trastuzumab-DM1 maytansinoid conjugate (10 mg/kg dose) and tumor volume was assessed for over 20 days after injection.
  • mice Groups of eight to ten SCID mice with 20 million Bjab-luc xenograft tumor cells per mouse were dosed at day 1 with an antibody-drug conjugate, or a naked antibody. A group of eight mice were tested each with anti-CD22 ADC, naked anti-CD22 antibodies, and control.
  • trastuzumab-SPP-DM1 disulfide linker
  • free DM1 and trastuzumab-SMCC-DM1 non-disulfide linker
  • trastuzumab-SMCC-DM1 for Group 2 and Group 3 studies contained PBS, 5.4 mM sodium phosphate, 4.2 mM potassium phosphate, 140 mM sodium chloride, pH 6.5.
  • Blood was collected for hematology analysis prior to first dosing (acclimation period) and at days 3, 7, 11, and 14 after the first dose (Groups 1 and 2) and at days 3, 7, 11, 14, and 21 after the second dose (Groups 1, 2 and 3).
  • Erythrocyte (RBC) and platelet (PLT) counts were measured by the light scattering method.
  • Leukocyte (WBC) count was measured by the peroxidase/basophil method.
  • Reticulocyte count was measured by the light scattering method with cationic dye.
  • Cell counts were measured on an Advia 120 apparatus.
  • ALT aminotransferase
  • AST aminotransferase
US11/141,344 2002-05-08 2005-05-31 Antibody-drug conjugates and methods Abandoned US20050276812A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US11/141,344 US20050276812A1 (en) 2004-06-01 2005-05-31 Antibody-drug conjugates and methods
US11/564,171 US7754441B2 (en) 2003-11-17 2006-11-28 Compositions and methods for the treatment of tumor of hematopoietic origin
US12/052,938 US8652479B2 (en) 2004-06-01 2008-03-21 Antibody-drug conjugates and methods
US12/326,721 US8142784B2 (en) 2004-06-01 2008-12-02 Antibody-drug conjugates and methods
US14/149,979 US20140128580A1 (en) 2004-06-01 2014-01-08 Antibody-drug conjugates and methods
US15/406,583 US20170362318A1 (en) 2002-05-08 2017-01-13 Compositions and methods for the treatment of tumor hematopoietic origin

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US57651704P 2004-06-01 2004-06-01
US61609804P 2004-10-05 2004-10-05
US11/141,344 US20050276812A1 (en) 2004-06-01 2005-05-31 Antibody-drug conjugates and methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/018829 Continuation-In-Part WO2005117986A2 (en) 2002-05-08 2005-05-31 Antibody drug conjugates and methods

Related Child Applications (4)

Application Number Title Priority Date Filing Date
PCT/US2005/018829 Continuation-In-Part WO2005117986A2 (en) 2002-05-08 2005-05-31 Antibody drug conjugates and methods
US11/564,171 Continuation-In-Part US7754441B2 (en) 2003-11-17 2006-11-28 Compositions and methods for the treatment of tumor of hematopoietic origin
US12/052,938 Division US8652479B2 (en) 2004-06-01 2008-03-21 Antibody-drug conjugates and methods
US12/326,721 Continuation US8142784B2 (en) 2004-06-01 2008-12-02 Antibody-drug conjugates and methods

Publications (1)

Publication Number Publication Date
US20050276812A1 true US20050276812A1 (en) 2005-12-15

Family

ID=35262141

Family Applications (4)

Application Number Title Priority Date Filing Date
US11/141,344 Abandoned US20050276812A1 (en) 2002-05-08 2005-05-31 Antibody-drug conjugates and methods
US12/052,938 Active 2027-09-13 US8652479B2 (en) 2004-06-01 2008-03-21 Antibody-drug conjugates and methods
US12/326,721 Active 2026-09-18 US8142784B2 (en) 2004-06-01 2008-12-02 Antibody-drug conjugates and methods
US14/149,979 Abandoned US20140128580A1 (en) 2004-06-01 2014-01-08 Antibody-drug conjugates and methods

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/052,938 Active 2027-09-13 US8652479B2 (en) 2004-06-01 2008-03-21 Antibody-drug conjugates and methods
US12/326,721 Active 2026-09-18 US8142784B2 (en) 2004-06-01 2008-12-02 Antibody-drug conjugates and methods
US14/149,979 Abandoned US20140128580A1 (en) 2004-06-01 2014-01-08 Antibody-drug conjugates and methods

Country Status (14)

Country Link
US (4) US20050276812A1 (zh)
EP (2) EP2286844A3 (zh)
JP (2) JP5234734B2 (zh)
KR (2) KR101200133B1 (zh)
CN (2) CN114053429A (zh)
AU (1) AU2005249490B2 (zh)
BR (1) BRPI0510883B8 (zh)
CA (1) CA2567520A1 (zh)
IL (1) IL179140A (zh)
MX (1) MXPA06014065A (zh)
NO (1) NO20066075L (zh)
NZ (2) NZ551180A (zh)
RU (1) RU2404810C9 (zh)
WO (1) WO2005117986A2 (zh)

Cited By (152)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050136063A1 (en) * 2003-11-21 2005-06-23 Schering Corporation Anti-IGFR antibody therapeutic combinations
US20060013819A1 (en) * 2004-06-16 2006-01-19 Genentech, Inc. Therapy of platinum-resistant cancer
WO2007090631A1 (en) * 2006-02-07 2007-08-16 Affectis Pharmaceuticals Ag Methods for the diagnosis and treatment of affective disorders and cushing's syndromes
US20080050310A1 (en) * 2006-05-30 2008-02-28 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
US20080096879A1 (en) * 2006-09-08 2008-04-24 University Of Pittsburgh-- Of The Commonwealth System Of Higher Education Synthesis of fr901464 and analogs with antitumor activity
US20080226659A1 (en) * 2000-03-16 2008-09-18 Sharon Erickson Methods of treatment using anti-erbb antibody-maytansinoid conjugates
WO2009012256A1 (en) 2007-07-16 2009-01-22 Genentech, Inc. Humanized anti-cd79b antibodies and immunoconjugates and methods of use
US20090028856A1 (en) * 2007-07-16 2009-01-29 Genentech, Inc. Anti-CD79B Antibodies and Immunoconjugates and Methods of Use
US20090041791A1 (en) * 2004-02-23 2009-02-12 Bainian Feng Heterocyclic self-immolative Linkers and Conjugates
WO2009032661A1 (en) * 2007-08-29 2009-03-12 Sanofi-Aventis Humanized anti-cxcr5 antibodies, derivatives thereof and their uses
US20090081223A1 (en) * 2005-01-21 2009-03-26 Genentech, Inc. Fixed dosing of her antibodies
WO2009054001A1 (en) * 2007-10-22 2009-04-30 Biocon Limited A pharmaceutical composition and a process thereof
US20090155259A1 (en) * 2005-02-23 2009-06-18 Genentech, Inc. Extending time to disease progression or survival in cancer patients
WO2009134870A1 (en) * 2008-04-30 2009-11-05 Immunogen, Inc. Potent cell-binding agent drug conjugates
WO2010009124A2 (en) 2008-07-15 2010-01-21 Genentech, Inc. Anthracycline derivative conjugates, process for their preparation and their use as antitumor compounds
US20100092495A1 (en) * 2008-04-30 2010-04-15 Immunogen Inc. Potent cell-binding agent drug conjugates
WO2009117277A3 (en) * 2008-03-18 2010-04-29 Genentech, Inc. Combinations of an anti-her2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
US7723485B2 (en) 2007-05-08 2010-05-25 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
US20100129314A1 (en) * 2008-04-30 2010-05-27 Immunogen Inc. Potent conjugates and hydrophilic linkers
US20100215669A1 (en) * 2008-01-31 2010-08-26 Genentech, Inc. Anti-cd79b antibodies and immunoconjugates and methods of use
WO2010120561A1 (en) 2009-04-01 2010-10-21 Genentech, Inc. Anti-fcrh5 antibodies and immunoconjugates and methods of use
WO2010124797A1 (en) * 2009-04-29 2010-11-04 Bayer Schering Pharma Aktiengesellschaft Anti-mesothelin immunoconjugates and uses therefor
US20110045005A1 (en) * 2001-10-19 2011-02-24 Craig Crowley Compositions and methods for the treatment of tumor of hematopoietic origin
US20110059852A1 (en) * 2008-03-26 2011-03-10 Cellerant Therapeutics, Inc. Compositions and methods for treating haematological proliferative disorders of meyloid origin
US20110070243A1 (en) * 2002-05-08 2011-03-24 Craig Crowley Compositions and methods for the treatment of tumor of hematopoietic
US20110117089A1 (en) * 2008-04-02 2011-05-19 Macrogenics, Inc. BCR-Complex-Specific Antibodies And Methods Of Using Same
WO2011069074A2 (en) 2009-12-04 2011-06-09 Genentech, Inc. Methods of treating metastatic breast cancer with trastuzumab-mcc-dm1
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011109308A1 (en) * 2010-03-02 2011-09-09 Seattle Genetics, Inc. Methods for screening antibodies
US8163287B2 (en) 2005-07-22 2012-04-24 Genentech, Inc. Combination therapy of her expressing tumors
US8183347B2 (en) 2006-03-17 2012-05-22 Genentech, Inc. Anti-TAT226 antibodies and immunoconjugates
WO2012074757A1 (en) 2010-11-17 2012-06-07 Genentech, Inc. Alaninyl maytansinol antibody conjugates
WO2012078771A1 (en) 2010-12-09 2012-06-14 Genentech, Inc. Treatment of her2-positive cancer with paclitaxel and trastuzumab-mcc-dm1
EP2468776A2 (en) 2007-02-09 2012-06-27 Genentech, Inc. Anti-Robo4 antibodies and uses therefor
US8268970B2 (en) 2007-10-01 2012-09-18 Bristol-Myers Squibb Company Human antibodies that bind mesothelin, and uses thereof
WO2012145112A2 (en) * 2011-04-18 2012-10-26 Immunogen, Inc. Novel maytansinoid derivatives with sulfoxide linker
WO2012154809A1 (en) 2011-05-09 2012-11-15 University Of Virginia Patent Foundation Compositions and methods for treating cancer
USRE43899E1 (en) 1999-10-01 2013-01-01 Immunogen Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
WO2013033380A1 (en) 2011-08-31 2013-03-07 Genentech, Inc. Diagnostic markers
WO2013043933A2 (en) 2011-09-22 2013-03-28 Amgen Inc. Cd27l antigen binding proteins
WO2013055874A2 (en) 2011-10-14 2013-04-18 Genentech, Inc. Uses for and article of manufacture including her2 dimerization inhibitor pertuzumab
WO2013063001A1 (en) 2011-10-28 2013-05-02 Genentech, Inc. Therapeutic combinations and methods of treating melanoma
WO2013134743A1 (en) 2012-03-08 2013-09-12 Halozyme, Inc. Conditionally active anti-epidermal growth factor receptor antibodies and methods of use thereof
WO2013149159A1 (en) 2012-03-30 2013-10-03 Genentech, Inc. Anti-lgr5 antibodies and immunoconjugates
WO2013165940A1 (en) 2012-05-01 2013-11-07 Genentech, Inc. Anti-pmel17 antibodies and immunoconjugates
WO2013173761A2 (en) 2012-05-18 2013-11-21 Amgen Inc. St2 antigen binding proteins
US8597654B2 (en) 2005-05-13 2013-12-03 Genentech, Inc. Adjuvant therapy with an anti-ERBB2 antibody conjugated to a maytansiniod
WO2013182668A1 (en) 2012-06-08 2013-12-12 F. Hoffmann-La Roche Ag Mutant selectivity and combinations of a phosphoinositide 3 kinase inhibitor compound and chemotherapeutic agents for the treatment of cancer
US8652474B2 (en) 2008-01-30 2014-02-18 Genentech, Inc. Composition comprising antibody that binds to domain II of HER2 and acidic variants thereof
US20140110297A1 (en) * 2007-06-25 2014-04-24 The Regents Of The University Of Colorado "Medical Articles Coated With Organopolysiloxane Containing a Protein Solution"
WO2014089335A2 (en) 2012-12-07 2014-06-12 Amgen Inc. Bcma antigen binding proteins
US8771966B2 (en) 2010-06-03 2014-07-08 Genentech, Inc. Immuno-PET imaging of antibodies and immunoconjugates and uses therefor
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
US8853154B2 (en) 2012-09-13 2014-10-07 Bristol-Myers Squibb Company Fibronectin based scaffold domain proteins that bind to myostatin
WO2014174316A1 (en) * 2013-04-26 2014-10-30 Adc Biotechnology Ltd Method of synthesising adcs using affinity resins
US8911732B2 (en) 2010-12-20 2014-12-16 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US8940302B2 (en) 2007-03-02 2015-01-27 Genentech, Inc. Predicting response to a HER inhibitor
WO2015038984A2 (en) 2013-09-12 2015-03-19 Halozyme, Inc. Modified anti-epidermal growth factor receptor antibodies and methods of use thereof
WO2015042108A1 (en) 2013-09-17 2015-03-26 Genentech, Inc. Methods of using anti-lgr5 antibodies
US9017671B2 (en) 2004-10-20 2015-04-28 Genentech, Inc. Method of treating cancer with a pharmaceutical formulation comprising a HER2 antibody
US9023356B2 (en) 2007-03-15 2015-05-05 Ludwig Institute For Cancer Research Ltd Treatment method using EGFR antibodies and SRC inhibitors and related formulations
WO2015089344A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US9072798B2 (en) 2009-02-18 2015-07-07 Ludwig Institute For Cancer Research Ltd. Specific binding proteins and uses thereof
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US9090693B2 (en) 2007-01-25 2015-07-28 Dana-Farber Cancer Institute Use of anti-EGFR antibodies in treatment of EGFR mutant mediated disease
WO2015112909A1 (en) 2014-01-24 2015-07-30 Genentech, Inc. Methods of using anti-steap1 antibodies and immunoconjugates
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
WO2015164665A1 (en) 2014-04-25 2015-10-29 Genentech, Inc. Methods of treating early breast cancer with trastuzumab-mcc-dm1 and pertuzumab
WO2015179658A2 (en) 2014-05-22 2015-11-26 Genentech, Inc. Anti-gpc3 antibodies and immunoconjugates
WO2016014360A1 (en) * 2014-07-24 2016-01-28 Genentech, Inc. Methods of conjugating an agent to a thiol moiety in a protein that contains at least one trisulfide bond
US9283276B2 (en) 2007-08-14 2016-03-15 Ludwig Institute For Cancer Research Ltd. Monoclonal antibody 175 targeting the EGF receptor and derivatives and uses thereof
WO2016040868A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
WO2016044396A1 (en) 2014-09-17 2016-03-24 Genentech, Inc. Immunoconjugates comprising anti-her2 antibodies and pyrrolobenzodiazepines
AU2013203265B2 (en) * 2007-08-29 2016-06-23 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
WO2016115345A1 (en) * 2015-01-14 2016-07-21 The Brigham And Women's Hospital, Treatment of cancer with anti-lap monoclonal antibodies
EP3056576A3 (en) * 2007-10-23 2016-09-28 Clinical Genomics Pty Ltd A method of diagnosing neoplasms
AU2014240234B2 (en) * 2008-03-18 2016-10-06 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
WO2016196373A2 (en) 2015-05-30 2016-12-08 Genentech, Inc. Methods of treating her2-positive metastatic breast cancer
US9562102B2 (en) 2001-05-11 2017-02-07 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
WO2017087280A1 (en) 2015-11-16 2017-05-26 Genentech, Inc. Methods of treating her2-positive cancer
US20170151345A1 (en) * 2012-08-30 2017-06-01 The General Hospital Corporation Compositions and methods for treating cancer
AU2014299561B2 (en) * 2013-06-24 2017-06-08 Ablbio Antibody-drug conjugate having improved stability and use thereof
US9676864B2 (en) 2009-10-02 2017-06-13 Sanofi Antibodies that specifically bind to the EphA2 receptor
US20170183394A1 (en) * 2005-05-25 2017-06-29 Hoffmann-La Roche Inc. Method for the purification of antibodies
WO2017137457A1 (en) * 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
WO2017161206A1 (en) 2016-03-16 2017-09-21 Halozyme, Inc. Conjugates containing conditionally active antibodies or antigen-binding fragments thereof, and methods of use
US9771377B2 (en) 2012-12-21 2017-09-26 University of Pittsburgh—of the Commonwealth System of Higher Education Synthesis of FR901464 and analogs with antitumor activity
US9815904B2 (en) 2013-04-16 2017-11-14 Genetech, Inc. Pertuzumab variants and evaluation thereof
US20170326251A1 (en) * 2014-10-28 2017-11-16 Adc Biotechnology Ltd. Method of synthesizing antibody drug conjugates using affinity resins
WO2017197234A1 (en) 2016-05-13 2017-11-16 Bioatla, Llc Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
WO2018009811A1 (en) 2016-07-08 2018-01-11 Genentech, Inc. Use of human epididymis protein 4 (he4) for assessing responsiveness of muc 16-positive cancer treatment
WO2018085513A1 (en) 2016-11-04 2018-05-11 Genentech, Inc. Treatment of her2-positive breast cancer
WO2018091724A1 (en) 2016-11-21 2018-05-24 Cureab Gmbh Anti-gp73 antibodies and immunoconjugates
WO2018125589A1 (en) 2016-12-28 2018-07-05 Genentech, Inc. Treatment of advanced her2 expressing cancer
WO2018136412A2 (en) 2017-01-17 2018-07-26 Genentech, Inc. Subcutaneous her2 antibody formulations
US10059768B2 (en) 2014-09-12 2018-08-28 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2018160654A2 (en) 2017-03-02 2018-09-07 Genentech, Inc. Adjuvant treatment of her2-positive breast cancer
WO2018200505A1 (en) 2017-04-24 2018-11-01 Genentech, Inc. Erbb2/her2 mutations in the transmbrane or juxtamembrane domain
USRE47123E1 (en) * 2006-07-18 2018-11-13 Sanofi EPHA2 receptor antagonist antibodies
USRE47223E1 (en) 2005-06-20 2019-02-05 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP3308801A4 (en) * 2015-06-09 2019-02-27 XDCExplorer (Shanghai) Co., Ltd. ANTIBODY-MEDICINAL CONJUGATE, INTERMEDIATE, PREPARATION METHOD, PHARMACEUTICAL COMPOSITIONS AND USES THEREOF
EP3456743A1 (en) 2013-05-30 2019-03-20 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
US10316084B2 (en) 2014-06-11 2019-06-11 Genentech, Inc. Anti-LGR5 antibodies and uses thereof
WO2019118937A1 (en) 2017-12-15 2019-06-20 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
US10383878B2 (en) 2014-04-10 2019-08-20 Daiichi Sankyo Company, Limited Anti-HER3 antibody-drug conjugate
WO2019175125A1 (en) 2018-03-13 2019-09-19 F. Hoffmann-La Roche Ag Combination therapy with targeted 4-1bb (cd137) agonists
CN110730787A (zh) * 2017-05-09 2020-01-24 赛安诺生物技术有限责任公司 修饰的微囊藻毒素和节球藻毒素
WO2019118411A3 (en) * 2017-12-11 2020-03-26 Triphase Accelerator U.S. Corporation Anti-cd22 antibody-maytansine conjugates, combinations, and methods of use thereof
WO2020081119A1 (en) 2018-10-15 2020-04-23 Genentech, Inc. Methods of treating residual breast cancer with trastuzumab emtansine
US10633636B2 (en) 2012-07-24 2020-04-28 The General Hospital Corporation Oncolytic virus therapy for resistant tumors
US10676515B2 (en) 2011-01-31 2020-06-09 The General Hospital Corporation Multimodal trail molecules expressed in T cells
WO2020117257A1 (en) 2018-12-06 2020-06-11 Genentech, Inc. Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
US10689457B2 (en) 2008-06-16 2020-06-23 Genentech, Inc. Treatment of metastatic breast cancer
US10729782B2 (en) 2012-10-19 2020-08-04 Daiichi Sankyo Company, Limited Antibody-drug conjugate produced by binding through linker having hydrophilic structure
EP3689910A2 (en) 2014-09-23 2020-08-05 F. Hoffmann-La Roche AG Method of using anti-cd79b immunoconjugates
US10736976B2 (en) 2016-12-01 2020-08-11 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-PD-L1 antibodies for immuno-PET imaging
WO2020232169A1 (en) 2019-05-14 2020-11-19 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat follicular lymphoma
US10844135B2 (en) 2003-10-10 2020-11-24 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates and methods of making said
US10874746B2 (en) 2016-02-08 2020-12-29 Synaffix B.V. Sulfamide linkers for use in bioconjugates
US10898570B2 (en) 2015-07-07 2021-01-26 Genentech, Inc. Combination therapy with an anti-HER2 antibody-drug conjugate and a Bcl-2 inhibitor
US10914720B2 (en) 2016-02-10 2021-02-09 Becton Dickinson France Method to evaluate the stability of a protein-based formulation
EP3782654A1 (en) 2014-09-12 2021-02-24 Genentech, Inc. Anti-her2 antibodies and immunoconjugates
US10973924B2 (en) 2012-10-11 2021-04-13 Daiichi Sankyo Company, Limited Antibody-drug conjugate
WO2021076196A1 (en) 2019-10-18 2021-04-22 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
US11008398B2 (en) 2013-12-25 2021-05-18 Daiichi Sankyo Company, Limited Anti-TROP2 antibody-drug conjugate
US11077202B2 (en) 2017-05-15 2021-08-03 Daiichi Sankyo Company, Limited Anti-CDH6 antibody and anti-CDH6 antibody-drug conjugate
US11130802B2 (en) 2018-10-10 2021-09-28 Tilos Therapeutics, Inc. Anti-lap antibody variants
US11149088B2 (en) 2016-04-15 2021-10-19 Bioatla, Inc. Anti-Axl antibodies, antibody fragments and their immunoconjugates and uses thereof
WO2021217051A1 (en) 2020-04-24 2021-10-28 Genentech, Inc. Methods of using anti-cd79b immunoconjugates
US11173213B2 (en) 2015-06-29 2021-11-16 Daiichi Sankyo Company, Limited Method for selectively manufacturing antibody-drug conjugate
US11185594B2 (en) 2014-04-10 2021-11-30 Daiichi Sankyo Company, Limited (Anti-HER2 antibody)-drug conjugate
US11230601B2 (en) 2017-10-10 2022-01-25 Tilos Therapeutics, Inc. Methods of using anti-lap antibodies
WO2022047243A1 (en) 2020-08-27 2022-03-03 Enosi Life Sciences Corp. Methods and compositions to treat autoimmune diseases and cancer
US11273155B2 (en) 2016-12-12 2022-03-15 Daiichi Sankyo Company, Limited Combination of antibody-drug conjugate and immune checkpoint inhibitor
US11318212B2 (en) 2017-08-31 2022-05-03 Daiichi Sankyo Company, Limited Method for producing antibody-drug conjugate
US11338043B2 (en) 2016-02-08 2022-05-24 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting HER2 tumours and method for improving therapeutic index of antibody-conjugates
RU2781195C2 (ru) * 2008-03-18 2022-10-07 Дженентек, Инк. Комбинации конъюгата анти-her2-антитело-лекарственное средство и химиотерапевтических средств и способы применения
WO2022241446A1 (en) 2021-05-12 2022-11-17 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023019092A1 (en) 2021-08-07 2023-02-16 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
US11584927B2 (en) 2014-08-28 2023-02-21 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified T-cells
US11584800B2 (en) 2014-01-31 2023-02-21 Daiichi Sankyo Company, Limited Method of treating cancer comprising administration of anti-HER2 antibody-drug conjugate
US11872289B2 (en) 2018-05-18 2024-01-16 Daiichi Sankyo Co., Ltd. Anti-MUC1 antibody-drug conjugate
US11938193B2 (en) * 2016-01-08 2024-03-26 Washington University Compositions comprising chemerin and methods of use thereof
US11945882B2 (en) 2017-08-31 2024-04-02 Daiichi Sankyo Company, Limited Method for producing antibody-drug conjugate
US11951175B2 (en) 2016-02-08 2024-04-09 Synaffix B.V. Bioconjugates containing sulfamide linkers for use in treatment

Families Citing this family (193)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6136311A (en) 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
EP1992643A3 (en) 2001-06-20 2008-12-10 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
US20050272120A1 (en) 2001-06-20 2005-12-08 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20070134243A1 (en) * 2004-12-01 2007-06-14 Gazzard Lewis J Antibody drug conjugates and methods
US7947839B2 (en) 2004-12-01 2011-05-24 Genentech, Inc. Heterocyclic-substituted bis-1,8 naphthalimide compounds, antibody drug conjugates, and methods of use
AU2006213662B2 (en) * 2005-02-11 2010-08-05 Immunogen, Inc. Process for preparing stable drug conjugates
US8735394B2 (en) 2005-02-18 2014-05-27 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
EP2301531B1 (en) 2005-02-18 2018-06-06 Abraxis BioScience, LLC Combinations and modes of administration of therapeutic agents and combination therapy
US20070166388A1 (en) 2005-02-18 2007-07-19 Desai Neil P Combinations and modes of administration of therapeutic agents and combination therapy
AU2006283726C1 (en) * 2005-08-24 2015-05-07 Immunogen, Inc. Process for preparing maytansinoid antibody conjugates
EP1806365A1 (en) 2006-01-05 2007-07-11 Boehringer Ingelheim International GmbH Antibody molecules specific for fibroblast activation protein and immunoconjugates containing them
WO2007100634A2 (en) * 2006-02-22 2007-09-07 3M Innovative Properties Company Immune response modifier conjugates
EP1996716B1 (en) 2006-03-20 2011-05-11 The Regents of the University of California Engineered anti-prostate stem cell antigen (psca) antibodies for cancer targeting
EP1864682A1 (en) * 2006-06-09 2007-12-12 Sanofi-Aventis Leptomycin derivatives
LT2117520T (lt) 2006-12-14 2018-12-10 Abraxis Bioscience, Llc Krūties vėžio terapija hormonų receptoriaus statuso pagrindu su nanodalelėmis, apimančiomis taksaną
JP6126773B2 (ja) 2007-09-04 2017-05-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 癌のターゲッティングおよび検出のための高親和性抗前立腺幹細胞抗原(psca)抗体
AU2008312457B2 (en) * 2007-10-19 2014-04-17 Genentech, Inc. Cysteine engineered anti-TENB2 antibodies and antibody drug conjugates
PT2242772E (pt) * 2007-12-26 2015-02-09 Biotest Ag Imunoconjugados dirigidos contra cd138 e as suas utilizações
US9011864B2 (en) * 2007-12-26 2015-04-21 Biotest Ag Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
EP2801584B1 (en) * 2007-12-26 2019-07-10 Biotest AG Agents targeting CD138 and uses thereof
JP2011508738A (ja) * 2007-12-26 2011-03-17 バイオテスト・アクチエンゲゼルシヤフト Cd138発現腫瘍細胞のターゲティングを向上させる方法及び剤
BRPI0907648B1 (pt) 2008-01-29 2022-01-11 Institute Of Molecular Biology And Genetics National Academy Of Science Of Ukraine Anticorpo isolado humanizado, usos de um anticorpo, métodos para preparar um anticorpo, para gerar anticorpos imunorreativos com slc34a2 e para triar ou identificar agentes ou compostos que modulam slc34a2, composições, ácido nucleico isolado, e, microorganismo hospedeiro
AU2009223124B2 (en) 2008-03-14 2015-03-26 Genentech, Inc. Genetic variations associated with drug resistance
US8236319B2 (en) 2008-04-30 2012-08-07 Immunogen, Inc. Cross-linkers and their uses
HUE025618T2 (en) * 2008-05-13 2016-04-28 Univ Yale Kimer small molecules to select antibodies against cancer cells
US8852630B2 (en) 2008-05-13 2014-10-07 Yale University Chimeric small molecules for the recruitment of antibodies to cancer cells
JP2011530535A (ja) 2008-08-07 2011-12-22 セントローズ, エルエルシー グリコシド化合物およびその医薬組成物
WO2010055950A1 (ja) 2008-11-17 2010-05-20 財団法人ヒューマンサイエンス振興財団 癌間質の構成因子に対して特異的結合能を有する物質と抗腫瘍性化合物との複合体による新規の癌ターゲティング治療
RU2636046C2 (ru) 2009-01-12 2017-11-17 Сайтомкс Терапьютикс, Инк Композиции модифицированных антител, способы их получения и применения
EP3360879A1 (en) 2009-02-05 2018-08-15 ImmunoGen, Inc. Benzodiazepine derivatives as cytotoxic agents
WO2010096486A1 (en) 2009-02-17 2010-08-26 Cornell Research Foundation, Inc. Methods and kits for diagnosis of cancer and prediction of therapeutic value
US9238878B2 (en) 2009-02-17 2016-01-19 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
SG174930A1 (en) 2009-04-01 2011-11-28 Genentech Inc Anti-fcrh5 antibodies and immunoconjugates and methods of use
ES2726945T3 (es) 2009-06-03 2019-10-10 Immunogen Inc Métodos de conjugación
US9345661B2 (en) 2009-07-31 2016-05-24 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
JP2013504585A (ja) 2009-09-09 2013-02-07 セントローズ, エルエルシー 細胞外標的化薬物複合体
AR078161A1 (es) 2009-09-11 2011-10-19 Hoffmann La Roche Formulaciones farmaceuticas muy concentradas de un anticuerpo anti cd20. uso de la formulacion. metodo de tratamiento.
EP2486023A4 (en) * 2009-10-06 2014-05-07 Immunogen Inc EFFICIENT CONJUGATES AND HYDROPHILIC BINDER
CA2782333C (en) 2009-12-02 2019-06-04 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
EP2510012B1 (en) * 2009-12-09 2017-04-19 Bayer Pharma Aktiengesellschaft Anti-c4.4a antibodies and uses thereof
MX2012009215A (es) 2010-02-23 2012-11-23 Genentech Inc Composiciones y metodos para el diagnostico y tratamiento de tumores.
KR20130028727A (ko) 2010-03-29 2013-03-19 아브락시스 바이오사이언스, 엘엘씨 치료제의 약물 전달 및 유효성 향상 방법
WO2011123395A1 (en) 2010-03-29 2011-10-06 Abraxis Bioscience, Llc Methods of treating cancer
EP2569334A4 (en) * 2010-05-13 2014-01-22 Fox Chase Cancer Ct RECOMBINANT-PRODUCED ANTIBODIES FOR THE TARGETING OF ERBB SIGNALING MOLECULES AND METHOD FOR THEIR USE FOR DISEASE DIAGNOSIS AND TREATMENT
NZ604031A (en) 2010-06-04 2015-05-29 Abraxis Bioscience Llc Methods of treatment of pancreatic cancer
CA3220104A1 (en) 2010-06-08 2011-12-15 Genentech, Inc. Cysteine engineered antibodies and conjugates
CN103298489A (zh) 2010-10-29 2013-09-11 伊缪诺金公司 新型egfr结合分子及其免疫偶联物
EP2632947A4 (en) 2010-10-29 2015-03-18 Immunogen Inc NON-ANTAGONIST MOLECULES BINDING TO THE EGF RECEPTOR AND IMMUNOCONJUGATES THEREOF
AU2011323316B2 (en) * 2010-11-03 2016-02-25 Immunogen, Inc. Cytotoxic agents comprising new ansamitocin derivatives
JP6294076B2 (ja) 2011-01-06 2018-03-14 ビオノール イミュノ エーエスBionor Immuno As 多量体ペプチド
RU2606016C2 (ru) 2011-01-14 2017-01-10 Редвуд Байосайнс, Инк. Меченые альдегидом полипептиды иммуноглобулина и способы их применения
MX346635B (es) 2011-02-15 2017-03-27 Immunogen Inc Derivados citotoxicos de la benzodiazepina.
WO2012135517A2 (en) 2011-03-29 2012-10-04 Immunogen, Inc. Preparation of maytansinoid antibody conjugates by a one-step process
ME03353B (me) 2011-03-29 2019-10-20 Immunogen Inc Priprema konjugata antitela i majtanzinoida jednostepenim postupkom
WO2012177837A2 (en) 2011-06-21 2012-12-27 Immunogen, Inc. Novel maytansinoid derivatives with peptide linker and conjugates thereof
EP2589609A1 (en) * 2011-11-03 2013-05-08 Pierre Fabre Medicament Antigen binding protein and its use as addressing product for the treatment of cancer
KR20140105765A (ko) 2011-11-21 2014-09-02 이뮤노젠 아이엔씨 EGfr 항체 세포독성제 접합체에 의한 EGfr 치료에 내성을 나타내는 종양의 치료 방법
BR112014013694A2 (pt) 2011-12-08 2017-06-13 Biotest Ag método para tratar uma doença, e, kit
US9814782B2 (en) * 2012-02-24 2017-11-14 Alteogen Inc. Modified antibody in which motif comprising cysteine residue is bound, modified antibody-drug conjugate comprising the modified antibody, and production method for same
KR102433686B1 (ko) 2012-05-15 2022-08-19 씨젠 인크. 자가-안정화 링커 접합체
US9504756B2 (en) 2012-05-15 2016-11-29 Seattle Genetics, Inc. Self-stabilizing linker conjugates
CA2872327A1 (en) 2012-05-21 2013-11-28 Genentech, Inc. Anti-ly6e antibodies and immunoconjugates and methods of use
CA2874923C (en) 2012-06-06 2021-08-31 Bionor Immuno As Peptides derived from viral proteins for use as immunogens and dosage reactants
US9334332B2 (en) 2012-07-25 2016-05-10 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies
EP2879711A4 (en) 2012-08-02 2016-03-16 Genentech Inc ANTI-ETBR ANTIBODIES AND IMMUNOCONJUGATES
WO2014022679A2 (en) 2012-08-02 2014-02-06 Genentech, Inc. Anti-etbr antibodies and immunoconjugates
EP2887965A1 (en) 2012-08-22 2015-07-01 ImmunoGen, Inc. Cytotoxic benzodiazepine derivatives
NZ707091A (en) 2012-10-04 2018-12-21 Immunogen Inc Use of a pvdf membrane to purify cell-binding agent cytotoxic agent conjugates
CN103288957B (zh) * 2012-12-21 2015-01-28 百奥泰生物科技(广州)有限公司 一种抑制肿瘤生长的抗体药物衍生物及其制备方法和用途
CN103333245B (zh) * 2012-12-21 2015-03-18 百奥泰生物科技(广州)有限公司 一种针对细胞受体并抑制癌细胞生长的药物分子及其制备方法和用途
CN103933575B (zh) * 2013-01-23 2017-09-29 上海新理念生物医药科技有限公司 一种三齿型连接子及其应用
JP6423804B2 (ja) 2013-02-28 2018-11-14 イミュノジェン・インコーポレーテッド 細胞結合剤及び細胞毒性剤を含む複合体
JP6494533B2 (ja) 2013-02-28 2019-04-03 イミュノジェン・インコーポレーテッド 細胞結合剤及び細胞毒性剤としてのマイタンシノイドを含む複合体
MY183572A (en) 2013-03-15 2021-02-26 Regeneron Pharma Biologically active molecules, conjugates thereof, and therapeutic uses
EP2994164B1 (en) 2013-05-08 2020-08-05 Zymeworks Inc. Bispecific her2 and her3 antigen binding constructs
WO2014194030A2 (en) 2013-05-31 2014-12-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
AR096687A1 (es) 2013-06-24 2016-01-27 Genentech Inc Anticuerpos anti-fcrh5
CA2921412A1 (en) 2013-08-26 2015-03-05 Regeneron Pharmaceuticals, Inc. Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
RU2699289C2 (ru) 2013-08-26 2019-09-04 Байонтек Рисерч Энд Дивелопмент, Инк. НУКЛЕИНОВЫЕ КИСЛОТЫ, КОДИРУЮЩИЕ АНТИТЕЛА ПРОТИВ СИАЛИРОВАННОГО АНТИГЕНА ЛЬЮИСАа ЧЕЛОВЕКА
GB201315486D0 (en) * 2013-08-30 2013-10-16 Ucb Pharma Sa Antibodies
EP3043810B1 (en) 2013-09-09 2019-10-23 CanImGuide Therapeutics AB Immune system modulators
CN103483357B (zh) * 2013-10-12 2015-11-18 齐鲁制药有限公司 一种抗体-美登素偶联物的中间体新晶型及其制备方法
WO2015061209A1 (en) 2013-10-21 2015-04-30 Genentech, Inc. ANTI-Ly6E ANTIBODIES AND METHODS OF USE
WO2015073721A1 (en) 2013-11-13 2015-05-21 Zymeworks Inc. Monovalent antigen binding constructs targeting egfr and/or her2 and uses thereof
KR101854443B1 (ko) * 2013-11-19 2018-06-15 레메젠 리미티드 항-her2 항체 및 이의 접합체
MX2016006572A (es) 2013-11-27 2016-12-09 Zymeworks Inc Construcciones de union a antigenos biespecificas dirigidas a her2.
DK3083689T3 (da) 2013-12-17 2020-08-03 Genentech Inc Anti-CD3-antistoffer og fremgangsmåder til anvendelse
JP2015124162A (ja) * 2013-12-26 2015-07-06 日本化薬株式会社 フェノール性水酸基を有するhsp90阻害剤のアクティブターゲティング型高分子誘導体及びその用途
CA2935077C (en) * 2013-12-27 2022-03-15 Geoffrey C. Winters Sulfonamide-containing linkage systems for drug conjugates
US10562977B2 (en) 2014-01-29 2020-02-18 Shanghai Hengrui Pharmaceutical Co., Ltd. Ligand-cytotoxic drug conjugate, preparation method thereof, and uses thereof
TW202214691A (zh) 2014-03-21 2022-04-16 美商艾伯維有限公司 抗-egfr抗體及抗體藥物結合物
SI3134124T1 (sl) * 2014-04-25 2019-06-28 Pierre Fabre Medicament Konjugat protitelesa zdravila IGF-1R in uporaba le-tega za zdravljenje raka
EP3138568B1 (en) 2014-04-29 2019-04-17 Genequantum Healthcare (Suzhou) Co., Ltd. New stable antibody-drug conjugate, preparation method therefor, and use thereof
LT3137114T (lt) * 2014-04-30 2021-03-25 Pfizer Inc. Anti-ptk7 antikūnų ir vaistų konjugatai
CA2950155C (en) 2014-06-02 2021-11-23 Regeneron Pharmaceuticals, Inc. Biologically active molecule conjugates, reagents and methods of manufacture, and therapeutic uses
PT3154583T (pt) 2014-06-04 2021-03-24 Biontech Res And Development Inc Anticorpos monoclonais humanos para o gangliósido gd2
EP3151830A4 (en) * 2014-06-06 2018-02-07 Redwood Bioscience, Inc. Anti-her2 antibody-maytansine conjugates and methods of use thereof
EP3164420A4 (en) * 2014-06-30 2018-05-23 Tarveda Therapeutics, Inc. Targeted conjugates and particles and formulations thereof
WO2016008112A1 (en) 2014-07-16 2016-01-21 Medshine Discovery Inc. Linkers and application towards adc thereof
WO2016020791A1 (en) 2014-08-05 2016-02-11 Novartis Ag Ckit antibody drug conjugates
KR102626976B1 (ko) 2014-09-02 2024-01-18 이뮤노젠 아이엔씨 항체 약물 컨쥬게이트 조성물의 제형화 방법
US10988531B2 (en) 2014-09-03 2021-04-27 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
ES2815353T3 (es) 2014-09-03 2021-03-29 Immunogen Inc Derivados de benzodiazepina citotóxicos
JP2017527562A (ja) 2014-09-03 2017-09-21 イミュノジェン・インコーポレーテッド 細胞毒性ベンゾジアゼピン誘導体
CA2957354A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
MA40934A (fr) 2014-11-19 2017-09-27 Immunogen Inc Procédé de préparation de conjugués agent de liaison cellulaire-agent cytotoxique
US9975949B2 (en) 2014-12-05 2018-05-22 Genentech, Inc. Anti-CD79b antibodies and methods of use
US10898582B2 (en) 2014-12-11 2021-01-26 University Of Utah Research Foundation Bi-functional allosteric protein-drug molecules for targeted therapy
US20170369525A1 (en) * 2014-12-31 2017-12-28 Development Center For Biotechnology Site-specific conjugation through glycoproteins linkage and method thereof
WO2016144650A1 (en) 2015-03-06 2016-09-15 Canimguide Therapeutics Ab Immune system modulators and compositions
RU2739617C2 (ru) * 2015-03-09 2020-12-28 Эдженсис, Инк. Конъюгаты антитело-лекарственное средство (adc), которые связываются с белками flt3
EA034950B1 (ru) 2015-03-27 2020-04-09 Регенерон Фармасьютикалз, Инк. Производные майтанзиноида, их конъюгаты и способы использования
GB201506870D0 (en) 2015-04-22 2015-06-03 Ucb Biopharma Sprl Method
TWI731861B (zh) 2015-06-16 2021-07-01 美商建南德克公司 FcRH5之人源化及親和力成熟抗體及使用方法
LT3313845T (lt) 2015-06-29 2020-12-10 Immunogen, Inc. Cisteino inžinerijos antikūnų konjugatai
EP3328393B1 (en) * 2015-07-31 2023-12-20 Centrose, Llc Extracellular drug conjugates targeting cd20
NZ739721A (en) 2015-08-07 2019-09-27 Imaginab Inc Antigen binding constructs to target molecules
MA43345A (fr) 2015-10-02 2018-08-08 Hoffmann La Roche Conjugués anticorps-médicaments de pyrrolobenzodiazépine et méthodes d'utilisation
CA3001712A1 (en) 2015-10-28 2017-05-04 Tarveda Therapeutics, Inc. Sstr-targeted conjugates and particles and formulations thereof
US10618935B2 (en) 2015-11-03 2020-04-14 Industrial Technology Research Institute Antibody-drug conjugate (ADC) and method for forming the same
CN106729743B (zh) * 2015-11-23 2021-09-21 四川科伦博泰生物医药股份有限公司 抗ErbB2抗体-药物偶联物及其组合物、制备方法和应用
TWI727380B (zh) 2015-11-30 2021-05-11 美商輝瑞股份有限公司 位點專一性her2抗體藥物共軛體
CN114478801A (zh) 2016-01-25 2022-05-13 里珍纳龙药品有限公司 美登素类化合物衍生物、其偶联物和使用方法
CN108601848A (zh) 2016-02-05 2018-09-28 伊缪诺金公司 用于制备细胞结合剂-细胞毒性剂缀合物的有效方法
WO2017147240A1 (en) 2016-02-23 2017-08-31 Tarveda Therapeutics, Inc. Hsp90 targeted conjugates and particles and formulations thereof
US11786603B2 (en) 2016-02-26 2023-10-17 Regeneron Pharmaceuticals, Inc. Optimized transglutaminase site-specific antibody conjugation
ES2929010T3 (es) * 2016-04-06 2022-11-24 Alteogen Inc Conjugado de anticuerpo-fármaco que comprende anticuerpo modificado
CA3019398A1 (en) 2016-04-26 2017-11-02 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
WO2017214456A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-cd98 antibodies and antibody drug conjugates
EP3468599A2 (en) 2016-06-08 2019-04-17 AbbVie Inc. Anti-cd98 antibodies and antibody drug conjugates
CN109563167A (zh) 2016-06-08 2019-04-02 艾伯维公司 抗b7-h3抗体和抗体药物偶联物
WO2017214335A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
CA3027103A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
TW201811376A (zh) 2016-07-01 2018-04-01 英商葛蘭素史密斯克藍智慧財產權有限公司 抗體-藥物結合物及使用其之治療方法
MA45945A (fr) 2016-08-09 2019-06-19 Seattle Genetics Inc Conjugués de médicaments avec des lieurs auto-stabilisants, aux propriétés physiochimiques améliorées
WO2018089373A2 (en) 2016-11-08 2018-05-17 Regeneron Pharmaceuticals, Inc. Steroids and protein-conjugates thereof
TWI755450B (zh) 2016-11-23 2022-02-21 美商伊繆諾金公司 苯二氮平衍生物之選擇性磺化
US10864279B2 (en) 2016-12-16 2020-12-15 Industrial Technology Research Institute Linker-drug and antibody-drug conjugate (ADC) employing the same
WO2018147960A1 (en) 2017-02-08 2018-08-16 Imaginab, Inc. Extension sequences for diabodies
JP7106560B2 (ja) 2017-02-28 2022-07-26 イミュノジェン・インコーポレーテッド 自己犠牲型ペプチドリンカーを有するメイタンシノイド誘導体及びそのコンジュゲート
JP6931072B2 (ja) * 2017-03-24 2021-09-01 モガム・インスティテュート・フォー・バイオメディカル・リサーチMogam Institute For Biomedical Research 抗ceacam1抗体およびその使用
WO2018181059A1 (ja) 2017-03-30 2018-10-04 日油株式会社 ヘテロ二官能性単分散ポリエチレングリコール及びそれを用いた複合体
EP3604384B1 (en) 2017-03-30 2021-09-08 NOF Corporation Hydrophilic polymer derivative having self-immolative acetal linker and composite using same
US11932694B2 (en) 2017-04-19 2024-03-19 Bluefin Biomedicine, Inc. Anti-VTCN1 antibodies and antibody drug conjugates
WO2018195243A1 (en) 2017-04-20 2018-10-25 Immunogen, Inc. Cytotoxic benzodiazepine derivatives and conjugates thereof
MX2019013690A (es) 2017-05-18 2020-01-27 Regeneron Pharma Conjugados de farmaco-proteina con ciclodextrina.
JP7279026B2 (ja) * 2017-09-20 2023-05-22 メルサナ セラピューティクス インコーポレイテッド Napi2b標的化療法に対する応答を予測するための組成物および方法
US11707522B2 (en) 2017-10-13 2023-07-25 Boehringer Ingelheim International Gmbh Human antibodies to Tn antigen
KR20200085807A (ko) 2017-11-07 2020-07-15 리제너론 파마슈티칼스 인코포레이티드 항체 약물 접합제용 친수성 링커
WO2019133652A1 (en) 2017-12-28 2019-07-04 Immunogen, Inc. Benzodiazepine derivatives
CA3093645A1 (en) 2018-03-13 2019-09-19 Nof Corporation Heterobifunctional compound having monodispersed polyethylene glycol in main chain and side chain
EP3560945A1 (en) 2018-04-27 2019-10-30 F. Hoffmann-La Roche AG Methods for purification of polypeptides using polysorbates
CA3098453A1 (en) 2018-05-09 2019-11-14 Regeneron Pharmaceuticals, Inc. Anti-msr1 antibodies and methods of use thereof
CN110507824A (zh) * 2018-05-21 2019-11-29 荣昌生物制药(烟台)有限公司 一种抗间皮素抗体及其抗体药物缀合物
EP3802825A1 (en) 2018-06-08 2021-04-14 Intellia Therapeutics, Inc. Compositions and methods for immunooncology
EP3808376A4 (en) 2018-06-17 2021-09-01 L&L Biopharma Co., Ltd. CLDN18.2 ANTIBODY, BISPECIFIC ANTIBODY, ADC AND CAR, AND APPLICATIONS THEREOF
AR116109A1 (es) 2018-07-10 2021-03-31 Novartis Ag Derivados de 3-(5-amino-1-oxoisoindolin-2-il)piperidina-2,6-diona y usos de los mismos
WO2020075817A1 (ja) * 2018-10-10 2020-04-16 武田薬品工業株式会社 抗体薬物複合体の製造方法
AU2019402189B2 (en) 2018-12-20 2023-04-13 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
US20210260208A1 (en) 2018-12-21 2021-08-26 Regeneron Pharmaceuticals, Inc. Tubulysins and protein-tubulysin conjugates
WO2020146541A2 (en) 2019-01-08 2020-07-16 Regeneron Pharmaceuticals, Inc. Traceless linkers and protein-conjugates thereof
AU2020222346B2 (en) 2019-02-15 2021-12-09 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
EA202192019A1 (ru) 2019-02-15 2021-11-02 Новартис Аг Производные 3-(1-оксо-5-(пиперидин-4-ил)изоиндолин-2-ил)пиперидин-2,6-диона и пути их применения
CA3133155A1 (en) 2019-03-19 2020-09-24 Fundacio Privada Institut D'investigacio Oncologica De Vall Hebron Combination therapy for the treatment for cancer
JPWO2021060439A1 (zh) 2019-09-26 2021-04-01
US20220387618A1 (en) 2019-11-15 2022-12-08 Seagen Inc. Methods of treating her2 positive breast cancer with tucatinib in combination with an anti-her2 antibody-drug conjugate
WO2021127621A2 (en) * 2019-12-19 2021-06-24 Overa, Inc. Compositions and methods for oocyte-specific contraception
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
CN115551552A (zh) 2020-02-25 2022-12-30 祐方有限公司 喜树碱衍生物及其缀合物
CA3179154A1 (en) 2020-04-16 2021-10-21 Regeneron Pharmaceuticals, Inc. Diels-alder conjugation methods
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
AU2021296449A1 (en) 2020-06-24 2023-01-19 Regeneron Pharmaceuticals, Inc. Tubulysins and protein-tubulysin conjugates
JP2023534437A (ja) 2020-07-13 2023-08-09 リジェネロン ファーマシューティカルズ,インク. タンパク質中のグルタミン残基にコンジュゲートされたカンプトテシンアナログおよびその使用
CN116134027A (zh) 2020-08-03 2023-05-16 诺华股份有限公司 杂芳基取代的3-(1-氧代异吲哚啉-2-基)哌啶-2,6-二酮衍生物及其用途
KR102320536B1 (ko) * 2020-09-04 2021-11-03 주식회사 베르티스 피분석물을 검출 또는 측정하기 위한 조성물
JP2023548975A (ja) 2020-11-10 2023-11-21 レジェネロン ファーマシューティカルズ, インコーポレイテッド セレン抗体複合体
BR112023014128A2 (pt) 2021-01-15 2023-10-31 Seagen Inc Conjugados de anticorpo-fármaco imunomodulatórios
UY39610A (es) 2021-01-20 2022-08-31 Abbvie Inc Conjugados anticuerpo-fármaco anti-egfr
WO2022170002A1 (en) 2021-02-03 2022-08-11 Seagen Inc. Immunostimulatory compounds and conjugates
KR20230158004A (ko) 2021-03-08 2023-11-17 진콴텀 헬스케어 (쑤저우) 씨오., 엘티디. 항체 면역 작용제 접합체 및 이의 적용
TW202304979A (zh) 2021-04-07 2023-02-01 瑞士商諾華公司 抗TGFβ抗體及其他治療劑用於治療增殖性疾病之用途
CN117177775A (zh) 2021-04-14 2023-12-05 启德医药科技(苏州)有限公司 连接子、缀合物及其应用
TW202309294A (zh) 2021-04-27 2023-03-01 瑞士商諾華公司 病毒載體生產系統
CA3223227A1 (en) * 2021-05-21 2022-11-24 NorthStar Medical Technologies, LLC Urokinase plasminogen activator receptor-targeted radiopharmaceutical
CA3221398A1 (en) 2021-05-28 2022-12-01 Seagen Inc. Anthracycline antibody conjugates
KR102604934B1 (ko) * 2021-10-27 2023-11-23 주식회사 베르티스 피분석물을 검출 또는 측정하기 위한 조성물
WO2023129518A1 (en) 2021-12-29 2023-07-06 Regeneron Pharmaceuticals, Inc. Tubulysins and protein-tubulysin conjugates
WO2023137026A1 (en) 2022-01-12 2023-07-20 Regeneron Pharmaceuticals, Inc. Camptothecin analogs conjugated to a glutamine residue in a protein, and their use
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023215740A1 (en) 2022-05-06 2023-11-09 Seagen Inc. Immunomodulatory antibody-drug conjugates
US20240108744A1 (en) 2022-07-27 2024-04-04 Mediboston Limited Auristatin derivatives and conjugates thereof
WO2024030577A1 (en) 2022-08-03 2024-02-08 Seagen Inc. Immunostimulatory anti-pd-l1-drug conjugates

Citations (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4137230A (en) * 1977-11-14 1979-01-30 Takeda Chemical Industries, Ltd. Method for the production of maytansinoids
US4248870A (en) * 1978-10-27 1981-02-03 Takeda Chemical Industries, Ltd. Maytansinoids and use
US4256746A (en) * 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
US4260608A (en) * 1978-11-14 1981-04-07 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and methods of use thereof
US4265814A (en) * 1978-03-24 1981-05-05 Takeda Chemical Industries Matansinol 3-n-hexadecanoate
US4294757A (en) * 1979-01-31 1981-10-13 Takeda Chemical Industries, Ltd 20-O-Acylmaytansinoids
US4307016A (en) * 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4308269A (en) * 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4308268A (en) * 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4309428A (en) * 1979-07-30 1982-01-05 Takeda Chemical Industries, Ltd. Maytansinoids
US4313946A (en) * 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4315929A (en) * 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
US4317821A (en) * 1979-06-08 1982-03-02 Takeda Chemical Industries, Ltd. Maytansinoids, their use and pharmaceutical compositions thereof
US4322348A (en) * 1979-06-05 1982-03-30 Takeda Chemical Industries, Ltd. Maytansinoids
US4331598A (en) * 1979-09-19 1982-05-25 Takeda Chemical Industries, Ltd. Maytansinoids
US4362663A (en) * 1979-09-21 1982-12-07 Takeda Chemical Industries, Ltd. Maytansinoid compound
US4364866A (en) * 1979-09-21 1982-12-21 Takeda Chemical Industries, Ltd. Maytansinoids
US4371533A (en) * 1980-10-08 1983-02-01 Takeda Chemical Industries, Ltd. 4,5-Deoxymaytansinoids, their use and pharmaceutical compositions thereof
US4424219A (en) * 1981-05-20 1984-01-03 Takeda Chemical Industries, Ltd. 9-Thiomaytansinoids and their pharmaceutical compositions and use
US4450254A (en) * 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4753894A (en) * 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US4956453A (en) * 1985-12-06 1990-09-11 Cetus Corporation Anti-human ovarian cancer immunotoxins and methods of use thereof
US4958009A (en) * 1985-12-06 1990-09-18 Cetus Corporation Anti-human ovarian cancer immunotoxins and methods of use thereof
US4981979A (en) * 1987-09-10 1991-01-01 Neorx Corporation Immunoconjugates joined by thioether bonds having reduced toxicity and improved selectivity
US5206018A (en) * 1978-11-03 1993-04-27 Ayerst, Mckenna & Harrison, Inc. Use of rapamycin in treatment of tumors
US5208020A (en) * 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5629197A (en) * 1984-02-08 1997-05-13 Cetus Oncology Corporation Monoclonal anti-human breast cancer antibodies
US5725856A (en) * 1988-01-12 1998-03-10 Genentech, Inc. Monoclonal antibodies directed to the HER2 receptor
US5821337A (en) * 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US6333410B1 (en) * 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
US20020001587A1 (en) * 2000-03-16 2002-01-03 Sharon Erickson Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
US6441163B1 (en) * 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
US20030103985A1 (en) * 2001-05-18 2003-06-05 Boehringer Ingelheim International Gmbh Cytotoxic CD44 antibody immunoconjugates
US6632979B2 (en) * 2000-03-16 2003-10-14 Genentech, Inc. Rodent HER2 tumor model
US20030235582A1 (en) * 2002-06-14 2003-12-25 Immunogen, Inc. Anti-IGF-I receptor antibody
US20040057952A1 (en) * 2002-07-03 2004-03-25 Immunogen Inc. Antibodies to non-shed Muc1 and Muc16, and uses thereof
US6716821B2 (en) * 2001-12-21 2004-04-06 Immunogen Inc. Cytotoxic agents bearing a reactive polyethylene glycol moiety, cytotoxic conjugates comprising polyethylene glycol linking groups, and methods of making and using the same
US20040120949A1 (en) * 2002-11-08 2004-06-24 Boehringer Ingelheim International Gmbh Compositions and methods for treating cancer using cytotoxic CD44 antibody immunoconjugates and radiotherapy
US20040126379A1 (en) * 2002-08-21 2004-07-01 Boehringer Ingelheim International Gmbh Compositions and methods for treating cancer using cytotoxic CD44 antibody immunoconjugates and chemotherapeutic agents
US20040235068A1 (en) * 2001-09-05 2004-11-25 Levinson Arthur D. Methods for the identification of polypeptide antigens associated with disorders involving aberrant cell proliferation and compositions useful for the treatment of such disorders
US20040235840A1 (en) * 2003-05-20 2004-11-25 Immunogen, Inc. Cytotoxic agents comprising new maytansinoids
US20040241174A1 (en) * 2003-05-14 2004-12-02 Immunogen, Inc. Drug conjugate composition
US20050016993A1 (en) * 2003-04-17 2005-01-27 Koskey James Donald Heated pet mat
US6913748B2 (en) * 2002-08-16 2005-07-05 Immunogen, Inc. Cross-linkers with high reactivity and solubility and their use in the preparation of conjugates for targeted delivery of small molecule drugs
US20050169933A1 (en) * 2003-10-10 2005-08-04 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates

Family Cites Families (343)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
CU22545A1 (es) 1994-11-18 1999-03-31 Centro Inmunologia Molecular Obtención de un anticuerpo quimérico y humanizado contra el receptor del factor de crecimiento epidérmico para uso diagnóstico y terapéutico
US3896111A (en) 1973-02-20 1975-07-22 Research Corp Ansa macrolides
US4151042A (en) 1977-03-31 1979-04-24 Takeda Chemical Industries, Ltd. Method for producing maytansinol and its derivatives
IL55477A (en) * 1978-09-01 1982-04-30 Yeda Res & Dev System for utilizing solar energy
US4361658A (en) * 1980-04-03 1982-11-30 Exxon Research And Engineering Co. Process for polymeric gelation
US4364533A (en) * 1980-11-13 1982-12-21 The Boeing Company Sidewall panel window assembly and method of installing
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JPS6098584A (ja) 1983-11-02 1985-06-01 Canon Inc カメラ―体形vtr
AU3934085A (en) 1984-01-30 1985-08-09 Icrf Patents Ltd. Improvements relating to growth factors
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US4970198A (en) 1985-10-17 1990-11-13 American Cyanamid Company Antitumor antibiotics (LL-E33288 complex)
US5583024A (en) 1985-12-02 1996-12-10 The Regents Of The University Of California Recombinant expression of Coleoptera luciferase
US5401638A (en) 1986-06-04 1995-03-28 Oncogene Science, Inc. Detection and quantification of neu related proteins in the biological fluids of humans
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US5079233A (en) 1987-01-30 1992-01-07 American Cyanamid Company N-acyl derivatives of the LL-E33288 antitumor antibiotics, composition and methods for using the same
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
AU6355090A (en) 1989-08-23 1991-04-03 Scripps Clinic And Research Foundation Compositions and methods for detection and treatment of epstein-barr virus infection and immune disorders
CA2066428C (en) 1989-09-08 2000-11-28 Bert Vogelstein Structural alterations of the egf receptor gene in human gliomas
DE68926248T2 (de) 1989-09-29 1996-12-19 Oncogene Science Inc p100 "neu" menschlisches Protein and Verwendung dieses Proteins zum Nachweis von preneoplasmatischen- oder neoplasmatischen beim Menschen
CA2026147C (en) * 1989-10-25 2006-02-07 Ravi J. Chari Cytotoxic agents comprising maytansinoids and their therapeutic use
US5183884A (en) 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
DE4014540A1 (de) 1990-05-07 1991-11-14 Klaus Dr Tschaikowsky Immunkonjugate zur prophylaxe und therapie von organschaeden bei entzuendlichen prozessen
US5256643A (en) 1990-05-29 1993-10-26 The Government Of The United States Human cripto protein
AU9016591A (en) 1990-10-25 1992-05-26 Tanox Biosystems, Inc. Glycoproteins associated with membrane-bound immunoglobulins as antibody targets on B cells
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5278299A (en) 1991-03-18 1994-01-11 Scripps Clinic And Research Foundation Method and composition for synthesizing sialylated glycosyl compounds
CA2407304A1 (en) 1991-03-29 1992-10-15 Genentech, Inc. Human pf4a receptors and their use
US5440021A (en) 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
US5543503A (en) 1991-03-29 1996-08-06 Genentech Inc. Antibodies to human IL-8 type A receptor
IL101943A0 (en) 1991-05-24 1992-12-30 Genentech Inc Structure,production and use of heregulin
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
JP3050424B2 (ja) 1991-07-12 2000-06-12 塩野義製薬株式会社 ヒトエンドセリンリセプター
US5264557A (en) 1991-08-23 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Polypeptide of a human cripto-related gene, CR-3
ES2136092T3 (es) 1991-09-23 1999-11-16 Medical Res Council Procedimientos para la produccion de anticuerpos humanizados.
US5362852A (en) 1991-09-27 1994-11-08 Pfizer Inc. Modified peptide derivatives conjugated at 2-hydroxyethylamine moieties
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
US6153408A (en) 1991-11-15 2000-11-28 Institut Pasteur And Institut National De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
US6011146A (en) 1991-11-15 2000-01-04 Institut Pasteur Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
WO1993010260A1 (en) 1991-11-21 1993-05-27 The Board Of Trustees Of The Leland Stanford Junior University Controlling degradation of glycoprotein oligosaccharides by extracellular glycosisases
JPH07501451A (ja) 1991-11-25 1995-02-16 エンゾン・インコーポレイテッド 多価抗原結合タンパク質
AU675929B2 (en) 1992-02-06 1997-02-27 Curis, Inc. Biosynthetic binding protein for cancer marker
ATE244763T1 (de) 1992-02-11 2003-07-15 Cell Genesys Inc Erzielen von homozygotem durch zielgerichtete genetische ereignisse
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
AU4025193A (en) 1992-04-08 1993-11-18 Cetus Oncology Corporation Humanized C-erbB-2 specific antibodies
WO1993022332A2 (en) 1992-04-24 1993-11-11 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
AU687346B2 (en) 1992-06-30 1998-02-26 Oncologix, Inc. A combination of anti-erbB-2 monoclonal antibodies and method of using
ATE149570T1 (de) 1992-08-17 1997-03-15 Genentech Inc Bispezifische immunoadhesine
IL107366A (en) 1992-10-23 2003-03-12 Chugai Pharmaceutical Co Ltd Genes coding for megakaryocyte potentiator
CA2103323A1 (en) 1992-11-24 1994-05-25 Gregory D. Plowman Her4 human receptor tyrosine kinase
US5644033A (en) 1992-12-22 1997-07-01 Health Research, Inc. Monoclonal antibodies that define a unique antigen of human B cell antigen receptor complex and methods of using same for diagnosis and treatment
US5801005A (en) 1993-03-17 1998-09-01 University Of Washington Immune reactivity to HER-2/neu protein for diagnosis of malignancies in which the HER-2/neu oncogene is associated
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
ATE186219T1 (de) 1993-03-24 1999-11-15 Berlex Biosciences Kombination von antihormonale und bindende moleküle zur krebsbehandlung
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US5773223A (en) 1993-09-02 1998-06-30 Chiron Corporation Endothelin B1, (ETB1) receptor polypeptide and its encoding nucleic acid methods, and uses thereof
AU697142B2 (en) 1993-11-23 1998-10-01 Genentech Inc. Protein tyrosine kinases named Rse
ATE207366T1 (de) 1993-12-24 2001-11-15 Merck Patent Gmbh Immunokonjugate
PT699237E (pt) 1994-03-17 2003-07-31 Merck Patent Gmbh Fvs de cadeia anti-egfr e anticorpos anti-egfr
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
BR9508409A (pt) 1994-07-21 1997-12-23 Akzo Nobel Nv Composição de peróxido transportável estável em armazenagem e uso de uma formulação de peróxido orgânico
WO2001016318A2 (en) 1999-09-01 2001-03-08 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US5750370A (en) 1995-06-06 1998-05-12 Human Genome Sciences, Inc. Nucleic acid encoding human endothlein-bombesin receptor and method of producing the receptor
AU4289496A (en) 1994-12-02 1996-06-19 Chiron Corporation Method of promoting an immune response with a bispecific antibody
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
EP2295415A1 (en) 1995-03-30 2011-03-16 OSI Pharmaceuticals, Inc. Quinazoline derivatives
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
JPH08336393A (ja) 1995-04-13 1996-12-24 Mitsubishi Chem Corp 光学活性なγ−置換−β−ヒドロキシ酪酸エステルの製造法
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
GB9508538D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quinazoline derivatives
GB9508565D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quiazoline derivative
US6410506B1 (en) 1995-05-19 2002-06-25 Human Genome Sciences, Inc. Transforming growth factor α HII
US5837234A (en) 1995-06-07 1998-11-17 Cytotherapeutics, Inc. Bioartificial organ containing cells encapsulated in a permselective polyether suflfone membrane
CA2222231A1 (en) 1995-06-07 1996-12-19 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth of tumors
US5977322A (en) 1995-06-14 1999-11-02 The Regents Of The University Of California High affinity human antibodies to tumor antigens
US5707829A (en) 1995-08-11 1998-01-13 Genetics Institute, Inc. DNA sequences and secreted proteins encoded thereby
JPH11510170A (ja) 1995-07-27 1999-09-07 ジェネンテック インコーポレーテッド タンパク質の処方
US20020193567A1 (en) 1995-08-11 2002-12-19 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
US5783186A (en) 1995-12-05 1998-07-21 Amgen Inc. Antibody-induced apoptosis
JP3646191B2 (ja) 1996-03-19 2005-05-11 大塚製薬株式会社 ヒト遺伝子
PT896586E (pt) 1996-03-27 2007-01-31 Genentech Inc Anticorpos de erbb3
PL190489B1 (pl) 1996-04-12 2005-12-30 Warner Lambert Co Nieodwracalne inhibitory kinaz tyrozyny, kompozycja farmaceutyczna je zawierająca i ich zastosowanie
EP0910636A1 (en) 1996-05-17 1999-04-28 Schering Corporation Human b-cell antigens, related reagents
US5834597A (en) 1996-05-20 1998-11-10 Protein Design Labs, Inc. Mutated nonactivating IgG2 domains and anti CD3 antibodies incorporating the same
US5922845A (en) 1996-07-11 1999-07-13 Medarex, Inc. Therapeutic multispecific compounds comprised of anti-Fcα receptor antibodies
US5945511A (en) 1997-02-20 1999-08-31 Zymogenetics, Inc. Class II cytokine receptor
US20030185830A1 (en) 1997-02-25 2003-10-02 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
US7033827B2 (en) 1997-02-25 2006-04-25 Corixa Corporation Prostate-specific polynucleotide compositions
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US6261791B1 (en) 1997-03-10 2001-07-17 The Regents Of The University Of California Method for diagnosing cancer using specific PSCA antibodies
US6541212B2 (en) 1997-03-10 2003-04-01 The Regents Of The University Of California Methods for detecting prostate stem cell antigen protein
NZ337413A (en) 1997-03-10 2003-02-28 Univ California Antibodies that bind to Prostate Stem Cell Antigen (PSCA) to treat prostate cancer.
UA73073C2 (uk) 1997-04-03 2005-06-15 Уайт Холдінгз Корпорейшн Заміщені 3-ціанохіноліни, спосіб їх одержання та фармацевтична композиція
US5994071A (en) 1997-04-04 1999-11-30 Albany Medical College Assessment of prostate cancer
US6555339B1 (en) 1997-04-14 2003-04-29 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human protein-coupled receptors
US6319688B1 (en) 1997-04-28 2001-11-20 Smithkline Beecham Corporation Polynucleotide encoding human sodium dependent phosphate transporter (IPT-1)
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US6890749B2 (en) 1997-05-15 2005-05-10 Abbott Laboratories Reagents and methods useful for detecting diseases of the prostate
WO1998051824A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting disease of the urinary tract
ZA986729B (en) 1997-07-29 1999-02-02 Warner Lambert Co Irreversible inhibitors of tyrosine kinases
ZA986732B (en) 1997-07-29 1999-02-02 Warner Lambert Co Irreversible inhibitiors of tyrosine kinases
TW436485B (en) 1997-08-01 2001-05-28 American Cyanamid Co Substituted quinazoline derivatives
US6602677B1 (en) 1997-09-19 2003-08-05 Promega Corporation Thermostable luciferases and methods of production
AU9805398A (en) 1997-10-15 1999-05-03 Children's Medical Center Corporation Novel human egf receptors and use thereof
US20030060612A1 (en) 1997-10-28 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20020034749A1 (en) 1997-11-18 2002-03-21 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
US6110695A (en) 1997-12-02 2000-08-29 The Regents Of The University Of California Modulating the interaction of the chemokine, B Lymphocyte Hemoattractant, and its Receptor, BLR1
WO1999046284A2 (en) 1998-03-13 1999-09-16 The Burnham Institute Molecules that home to various selected organs or tissues
AU4078599A (en) 1998-05-13 1999-11-29 Epimmune, Inc. Expression vectors for stimulating an immune response and methods of using the same
US20020187472A1 (en) 2001-03-09 2002-12-12 Preeti Lal Steap-related protein
US20030064397A1 (en) 1998-05-22 2003-04-03 Incyte Genomics, Inc. Transmembrane protein differentially expressed in prostate and lung tumors
WO2002016429A2 (en) 2000-08-24 2002-02-28 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2000012130A1 (en) 1998-08-27 2000-03-09 Smithkline Beecham Corporation Rp105 agonists and antagonists
JP4689781B2 (ja) 1998-09-03 2011-05-25 独立行政法人科学技術振興機構 アミノ酸輸送蛋白及びその遺伝子
WO2000020579A1 (en) 1998-10-02 2000-04-13 Mcmaster University Spliced form of erbb-2/neu oncogene
WO2001057188A2 (en) 2000-02-03 2001-08-09 Hyseq, Inc. Novel nucleic acids and polypeptides
US6468546B1 (en) 1998-12-17 2002-10-22 Corixa Corporation Compositions and methods for therapy and diagnosis of ovarian cancer
US6962980B2 (en) 1999-09-24 2005-11-08 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US6858710B2 (en) 1998-12-17 2005-02-22 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030091580A1 (en) 2001-06-18 2003-05-15 Mitcham Jennifer L. Compositions and methods for the therapy and diagnosis of ovarian cancer
US20020119158A1 (en) 1998-12-17 2002-08-29 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
JP2002536966A (ja) 1998-12-30 2002-11-05 ベス・イスラエル・ディーコニス・メディカル・センター・インコーポレーテッド カルシウムチャネルファミリーの特徴付け
US20030190669A1 (en) 1998-12-30 2003-10-09 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
CA2361009C (en) 1999-01-29 2012-10-23 Corixa Corporation Her-2/neu fusion proteins
GB9905124D0 (en) 1999-03-05 1999-04-28 Smithkline Beecham Biolog Novel compounds
AU3395900A (en) 1999-03-12 2000-10-04 Human Genome Sciences, Inc. Human lung cancer associated gene sequences and polypeptides
US7074403B1 (en) * 1999-06-09 2006-07-11 Immunomedics, Inc. Immunotherapy of autoimmune disorders using antibodies which target B-cells
US7304126B2 (en) 1999-05-11 2007-12-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU5003200A (en) 1999-05-14 2000-12-05 United States Of America As Represented By The Department Of Veterans Affairs, The Isolation and characterization of epidermal growth factor related protein
US7049410B2 (en) 1999-05-14 2006-05-23 Majumdar Adhip P N Antibodies to a novel EGF-receptor related protein (ERRP)
AU4952600A (en) 1999-06-03 2000-12-28 Takeda Chemical Industries Ltd. Screening method with the use of cd100
DK2283866T3 (en) * 1999-06-25 2015-05-18 Genentech Inc METHODS OF TREATMENT USING ANTI-ERBB ANTIBODY-MAYTANSINOID CONJUGATES
EP1189641B1 (en) 1999-06-25 2009-07-29 Genentech, Inc. HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
US20030119113A1 (en) 1999-07-20 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7297770B2 (en) 1999-08-10 2007-11-20 Genentech, Inc. PRO6496 polypeptides
US7294696B2 (en) 1999-08-17 2007-11-13 Genentech Inc. PRO7168 polypeptides
US20030232056A1 (en) 1999-09-10 2003-12-18 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030206918A1 (en) 1999-09-10 2003-11-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030129192A1 (en) 1999-09-10 2003-07-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
EP1216258A1 (en) 1999-09-28 2002-06-26 Universität Zürich Factors having prion-binding activity in serum and plasma and agents to detect transmissible spongiform encephalopathitis
CA2385528C (en) * 1999-10-01 2013-12-10 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
IL149116A0 (en) 1999-10-29 2002-11-10 Genentech Inc Anti-prostate stem cell antigen (psca) antibody compositions and methods of use
EP2418212B1 (en) 1999-11-29 2016-06-15 The Trustees of Columbia University in the City of New York Isolation of five novel genes coding for new fc receptors-type melanoma involved in the pathogenesis of lymphoma/melanoma
CA2392510A1 (en) 1999-11-30 2001-06-07 Corixa Corporation Compositions and methods for therapy and diagnosis of breast cancer
EP1568373A3 (en) 1999-12-10 2005-12-21 Epimmune Inc. Inducing cellular immune responses to HER2/neu using peptide and nucleic acid compositions
US6610286B2 (en) 1999-12-23 2003-08-26 Zymogenetics, Inc. Method for treating inflammation using soluble receptors to interleukin-20
JP2003535037A (ja) 1999-12-23 2003-11-25 ザイモジェネティクス,インコーポレイティド 炎症を治療する方法
NZ502058A (en) 1999-12-23 2003-11-28 Ovita Ltd Isolated mutated nucleic acid molecule for regulation of ovulation rate
RU2279441C2 (ru) 1999-12-23 2006-07-10 Займодженетикс, Инк. Выделенный растворимый il-20 рецептор (варианты)
US20040001827A1 (en) 2002-06-28 2004-01-01 Dennis Mark S. Serum albumin binding peptides for tumor targeting
ATE337403T1 (de) 1999-12-24 2006-09-15 Genentech Inc Verfahren und verbindungen zur verlängerung der halbwertzeiten bei der ausscheidung von biowirksamen verbindungen
US7294695B2 (en) 2000-01-20 2007-11-13 Genentech, Inc. PRO10268 polypeptides
EP1252294A2 (en) 2000-01-21 2002-10-30 Corixa Corporation Compounds and methods for prevention and treatment of her-2/neu associated malignancies
US20030224379A1 (en) 2000-01-21 2003-12-04 Tang Y. Tom Novel nucleic acids and polypeptides
US20030186372A1 (en) 2000-02-11 2003-10-02 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU2001238596A1 (en) 2000-02-22 2001-09-03 Millennium Pharmaceuticals, Inc. 18607, a novel human calcium channel
US20030219806A1 (en) 2000-02-22 2003-11-27 Millennium Pharmaceuticals, Inc. Novel 18607, 15603, 69318, 12303, 48000, 52920, 5433, 38554, 57301, 58324, 55063, 52991, 59914, 59921 and 33751 molecules and uses therefor
US20040002068A1 (en) 2000-03-01 2004-01-01 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US20040005561A1 (en) 2000-03-01 2004-01-08 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
WO2001066689A2 (en) 2000-03-07 2001-09-13 Hyseq, Inc. Novel nucleic acids and polypeptides
AU4941101A (en) 2000-03-24 2001-10-08 Fahri Saatcioglu Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods
EP1268554A2 (de) 2000-03-31 2003-01-02 IPF Pharmaceuticals GmbH Diagnostik- und arzneimittel zur untersuchung des zelloberflächenproteoms von tumor- und entzündungszellen sowie zur behandlung von tumorerkrankungen und entzündlichen erkrankungen vorzugsweise mit hilfe einer spezifischen chemokinrezeptor-analyse und der chemokinrezeptor-ligand-interaktion
AU2001253140A1 (en) 2000-04-03 2001-10-15 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Tumor markers in ovarian cancer
MXPA02009902A (es) 2000-04-07 2003-06-17 Arena Pharm Inc Receptores acoplados a proteina g, conocidos, activados constitutivamente, no endogenos.
ES2528794T3 (es) 2000-04-11 2015-02-12 Genentech, Inc. Anticuerpos multivalentes y usos de los mismos
WO2001088133A2 (en) 2000-05-18 2001-11-22 Lexicon Genetics Incorporated Human semaphorin homologs and polynucleotides encoding the same
AU2001274888A1 (en) 2000-05-19 2001-12-03 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
WO2001094641A2 (en) 2000-06-09 2001-12-13 Idec Pharmaceuticals Corporation Gene targets and ligands that bind thereto for treatment and diagnosis of ovarian carcinomas
AU2001268471A1 (en) 2000-06-16 2002-01-02 Incyte Genomics, Inc. G-protein coupled receptors
US20020165347A1 (en) 2000-06-30 2002-11-07 Amgen, Inc. B7-like molecules and uses thereof
WO2002002587A1 (en) 2000-06-30 2002-01-10 Human Genome Sciences, Inc. B7-like polynucleotides, polypeptides, and antibodies
CA2413186A1 (en) 2000-06-30 2002-01-10 Incyte Genomics, Inc. Extracellular matrix and cell adhesion molecules
WO2002006339A2 (en) 2000-07-03 2002-01-24 Curagen Corporation Proteins and nucleic acids encoding same
US20040044179A1 (en) 2000-07-25 2004-03-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2002010187A1 (en) 2000-07-27 2002-02-07 Mayo Foundation For Medical Education And Research B7-h3 and b7-h4, novel immunoregulatory molecules
US7205108B2 (en) 2000-07-28 2007-04-17 Ulrich Wissenbach Trp8, Trp9 and Trp10, novel markers for cancer
US7229623B1 (en) 2000-08-03 2007-06-12 Corixa Corporation Her-2/neu fusion proteins
WO2002013847A2 (en) 2000-08-14 2002-02-21 Corixa Corporation Methods for diagnosis and therapy of hematological and virus-associated malignancies
WO2002014503A2 (en) 2000-08-14 2002-02-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of her-2/neu-associated malignancies
GB0020953D0 (en) 2000-08-24 2000-10-11 Smithkline Beecham Biolog Vaccine
US20020119148A1 (en) 2000-09-01 2002-08-29 Gerritsen Mary E. ErbB4 antagonists
AU2001290548A1 (en) 2000-09-11 2002-03-26 Hyseq, Inc. Novel nucleic acids and polypeptides
AR030612A1 (es) 2000-09-12 2003-08-27 Smithkline Beecham Corp Procedimiento e intermedios
US6613567B1 (en) 2000-09-15 2003-09-02 Isis Pharmaceuticals, Inc. Antisense inhibition of Her-2 expression
US7582293B2 (en) 2000-09-15 2009-09-01 Genentech, Inc. Anti-PRO6308 antibodies
AU2001290837A1 (en) 2000-09-15 2002-03-26 Zymogenetics Inc. Use of a polypeptide comprising the extracellular domains of il-20rb for the treatment of inflammation
AU2001292724B2 (en) 2000-09-18 2005-05-26 Biogen Idec Ma Inc. CRIPTO mutant and uses thereof
UA83458C2 (uk) 2000-09-18 2008-07-25 Байоджен Айдек Ма Інк. Виділений поліпептид baff-r (рецептор фактора активації в-клітин сімейства tnf)
MXPA03003151A (es) 2000-10-13 2003-08-19 Eos Biotechnology Inc Metodos de diagnostico de cancer de prostata, composiciones y metodos para seleccionar moduladores de cancer de prostata.
US20030092164A1 (en) 2000-11-07 2003-05-15 Gross Jane A. Human tumor necrosis factor receptor
US20020150573A1 (en) 2000-11-10 2002-10-17 The Rockefeller University Anti-Igalpha-Igbeta antibody for lymphoma therapy
EP2341060B1 (en) 2000-12-12 2019-02-20 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
WO2002061087A2 (en) 2000-12-19 2002-08-08 Lifespan Biosciences, Inc. Antigenic peptides, such as for g protein-coupled receptors (gpcrs), antibodies thereto, and systems for identifying such antigenic peptides
AU2002243495A1 (en) 2001-01-12 2002-07-24 University Of Medicine And Dentistry Of New Jersey Bone morphogenetic protein-2 in the treatment and diagnosis of cancer
US20030119119A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119126A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
MXPA03006617A (es) 2001-01-24 2004-12-02 Protein Design Labs Inc Metodos de diagnostico de cancer de pecho, composiciones y metodos de rastreo de moduladores de cancer de pecho.
US20030073144A1 (en) 2001-01-30 2003-04-17 Corixa Corporation Compositions and methods for the therapy and diagnosis of pancreatic cancer
WO2002064798A1 (en) 2001-02-12 2002-08-22 Bionomics Limited Dna sequences differentially expressed in tumour cell lines
US20030087250A1 (en) 2001-03-14 2003-05-08 Millennium Pharmaceuticals, Inc. Nucleic acid molecules and proteins for the identification, assessment, prevention, and therapy of ovarian cancer
AU2002311787A1 (en) 2001-03-28 2002-10-15 Zycos Inc. Translational profiling
WO2003008537A2 (en) 2001-04-06 2003-01-30 Mannkind Corporation Epitope sequences
US6820011B2 (en) 2001-04-11 2004-11-16 The Regents Of The University Of Colorado Three-dimensional structure of complement receptor type 2 and uses thereof
AU2002254615A1 (en) 2001-04-17 2002-10-28 The Board Of Trustees Of The University Of Arkansas Repeat sequences of the ca125 gene and their use for diagnostic and therapeutic interventions
AU2002309583A1 (en) 2001-04-18 2002-11-05 Protein Desing Labs, Inc. Methods of diagnosis of lung cancer, compositions and methods of screening for modulators of lung cancer
WO2003083041A2 (en) 2002-03-22 2003-10-09 Biogen, Inc. Cripto-specific antibodies
PT1390389E (pt) 2001-04-26 2009-04-03 Biogen Idec Inc Anticorpos que bloqueiam o cripto e as utilizações dos mesmos
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
WO2002089747A2 (en) 2001-05-09 2002-11-14 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
WO2002092836A2 (en) 2001-05-11 2002-11-21 Sloan-Kettering Institute For Cancer Research Nucleic acid sequence encoding ovarian antigen, ca125, and uses thereof
SI1436003T1 (sl) 2001-05-24 2010-02-26 Zymogenetics Inc TACI-imunoglobulinski fuzijski proteini
WO2003000842A2 (en) 2001-06-04 2003-01-03 Curagen Corporation Novel proteins and nucleic acids encoding same
AU2002314901A1 (en) 2001-06-04 2002-12-16 Eos Biotechnology, Inc. Methods of diagnosis and treatment of androgen-dependent prostate cancer, prostate cancer undergoing androgen-withdrawal, and androgen-independent prostate cancer
WO2002098356A2 (en) 2001-06-05 2002-12-12 Exelixis Inc. Ppp2cs as modifiers of the p53 pathway and methods of use
ATE483976T1 (de) 2001-06-05 2010-10-15 Exelixis Inc Gfats als modifikatoren des p53-wegs und verwendungsverfahren
US7235358B2 (en) 2001-06-08 2007-06-26 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
WO2002101075A2 (en) 2001-06-13 2002-12-19 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
US7189507B2 (en) 2001-06-18 2007-03-13 Pdl Biopharma, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
CA2451465A1 (en) 2001-06-18 2002-12-27 Eos Biotechnology Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
US20050107595A1 (en) 2001-06-20 2005-05-19 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1992643A3 (en) * 2001-06-20 2008-12-10 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
AU2002322280A1 (en) 2001-06-21 2003-01-21 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
WO2003002717A2 (en) 2001-06-28 2003-01-09 Schering Corporation Biological activity of ak155
US20030120040A1 (en) 2001-06-29 2003-06-26 Genentech, Inc. Secreted and Transmembrane polypeptides and nucleic acids encoding the same
AU2002314433A1 (en) 2001-07-02 2003-01-21 Licentia Ltd. Ephrin-tie receptor materials and methods
US20040076955A1 (en) 2001-07-03 2004-04-22 Eos Biotechnology, Inc. Methods of diagnosis of bladder cancer, compositions and methods of screening for modulators of bladder cancer
WO2003003984A2 (en) 2001-07-05 2003-01-16 Curagen Corporation Novel proteins and nucleic acids encoding same
US7446185B2 (en) 2001-07-18 2008-11-04 The Regents Of The University Of California Her2/neu target antigen and use of same to stimulate an immune response
US20030108963A1 (en) 2001-07-25 2003-06-12 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kit, and methods for identification, assessment, prevention and therapy of prostate cancer
WO2003014294A2 (en) 2001-08-03 2003-02-20 Genentech, Inc. Tacis and br3 polypeptides and uses thereof
AU2002324700A1 (en) 2001-08-14 2003-03-03 Bayer Ag Nucleic acid and amino acid sequences involved in pain
US20030092013A1 (en) 2001-08-16 2003-05-15 Vitivity, Inc. Diagnosis and treatment of vascular disease
WO2003018621A2 (en) 2001-08-23 2003-03-06 Oxford Biomedica (Uk) Limited Genes
AU2002357643A1 (en) 2001-08-29 2003-04-14 Vanderbilt University The human mob-5 (il-24) receptors and uses thereof
US20030124579A1 (en) 2001-09-05 2003-07-03 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
AU2002336446B2 (en) 2001-09-06 2008-03-06 Agensys, Inc. Nucleic acid and corresponding protein entitled STEAP-1 useful in treatment and detection of cancer
AU2002330039A1 (en) 2001-09-17 2003-04-01 Eos Biotechnology, Inc. Methods of diagnosis of cancer compositions and methods of screening for modulators of cancer
WO2003024392A2 (en) * 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
IL160933A0 (en) 2001-09-18 2004-08-31 Proteologics Inc Methods and compositions for treating ?cap associated diseases
WO2003025148A2 (en) 2001-09-19 2003-03-27 Nuvelo, Inc. Novel nucleic acids and polypeptides
WO2003026577A2 (en) 2001-09-24 2003-04-03 Seattle Genetics, Inc. P-amidobenzylethers in drug delivery agents
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
US7355012B2 (en) * 2001-09-26 2008-04-08 United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Mutated anti-CD22 antibodies with increased affinity to CD22-expressing leukemia cells
AU2002327792A1 (en) 2001-09-28 2003-04-07 Bing Yang Diagnosis and treatment of diseases caused by mutations in cd72
WO2003029277A2 (en) 2001-10-03 2003-04-10 Rigel Pharmaceuticals, Inc. Modulators of lymphocyte activation and migration
US20040249144A1 (en) 2001-10-03 2004-12-09 Zairen Sun Regulated breast cancer genes
WO2003088808A2 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
JP2005522986A (ja) 2001-10-19 2005-08-04 ジェネンテック・インコーポレーテッド 炎症性腸疾患の診断と治療のための組成物と方法
US20050123925A1 (en) 2002-11-15 2005-06-09 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US6942972B2 (en) * 2001-10-24 2005-09-13 Beckman Coulter, Inc. Efficient synthesis of protein-oligonucleotide conjugates
AU2002356858A1 (en) 2001-10-24 2003-05-06 National Jewish Medical And Research Center Structure of tall-1 and its cognate receptor
KR20100013352A (ko) 2001-10-31 2010-02-09 알콘, 인코퍼레이티드 뼈 형태발생 단백질(bmp), bmp 수용체 및 bmp 결합 단백질 및 녹내장 진단 및 치료에서 그의 용도
WO2003042661A2 (en) 2001-11-13 2003-05-22 Protein Design Labs, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
US20030232350A1 (en) 2001-11-13 2003-12-18 Eos Biotechnology, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
WO2003043583A2 (en) 2001-11-20 2003-05-30 Seattle Genetics, Inc. Treatment of immunological disorders using anti-cd30 antibodies
AU2002339691A1 (en) 2001-11-29 2003-06-10 Genset Agonists and antagonists of prolixin for the treatment of metabolic disorders
WO2003048202A2 (en) 2001-12-03 2003-06-12 Asahi Kasei Pharma Corporation Nf-kappab activating genes
AU2002366951A1 (en) 2001-12-10 2003-07-09 Nuvelo,Inc. Novel nucleic acids and polypeptides
US20030134790A1 (en) 2002-01-11 2003-07-17 University Of Medicine And Dentistry Of New Jersey Bone Morphogenetic Protein-2 And Bone Morphogenetic Protein-4 In The Treatment And Diagnosis Of Cancer
US7452675B2 (en) 2002-01-25 2008-11-18 The Queen's Medical Center Methods of screening for TRPM4b modulators
JP2005526501A (ja) * 2002-02-21 2005-09-08 デューク・ユニヴァーシティ 自己免疫疾患用の試薬および治療方法
CA2476518A1 (en) 2002-02-22 2003-09-04 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US20030165966A1 (en) 2002-03-01 2003-09-04 Marcia Belvin MSRAs as modifiers of the p53 pathway and methods of use
WO2003104399A2 (en) 2002-06-07 2003-12-18 Avalon Pharmaceuticals, Inc Cancer-linked gene as target for chemotherapy
EP1572087B1 (en) * 2002-03-08 2008-05-14 PDL BioPharma, Inc. Antibodies against cancer antigen tmeff2 and uses thereof
EP2258712A3 (en) 2002-03-15 2011-05-04 Multicell Immunotherapeutics, Inc. Compositions and Methods to Initiate or Enhance Antibody and Major-histocompatibility Class I or Class II-restricted T Cell Responses by Using Immunomodulatory, Non-coding RNA Motifs
CA2486490A1 (en) 2002-03-19 2003-12-31 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003081210A2 (en) 2002-03-21 2003-10-02 Sunesis Pharmaceuticals, Inc. Identification of kinase inhibitors
US7193069B2 (en) 2002-03-22 2007-03-20 Research Association For Biotechnology Full-length cDNA
EP1490085A2 (en) 2002-03-25 2004-12-29 Uab Research Foundation Fc receptor homolog, reagents, and uses thereof
WO2003083074A2 (en) 2002-03-28 2003-10-09 Idec Pharmaceuticals Corporation Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
US20030194704A1 (en) 2002-04-03 2003-10-16 Penn Sharron Gaynor Human genome-derived single exon nucleic acid probes useful for gene expression analysis two
WO2003087306A2 (en) 2002-04-05 2003-10-23 Agensys, Inc. Nucleic acid and corresponding protein entitled 98p4b6 useful in treatment and detection of cancer
AU2003223520A1 (en) 2002-04-12 2003-10-27 Mitokor Targets for therapeutic intervention identified in the mitochondrial proteome
US20030224467A1 (en) 2002-04-17 2003-12-04 Osborne C. Kent AIB1 as a prognostic marker and predictor of resistance to endocrine therapy
AU2003228869A1 (en) 2002-05-03 2003-11-17 Incyte Corporation Transporters and ion channels
AU2003229294A1 (en) 2002-05-15 2003-12-02 Avalon Pharmaceuticals Cancer-linked gene as target for chemotherapy
WO2003101388A2 (en) 2002-05-30 2003-12-11 Bristol-Myers Squibb Company Human solute carrier family 7 member 11 (hslc7a11)
AU2003240495A1 (en) 2002-06-04 2003-12-19 Incyte Corporation Diagnostics markers for lung cancer
EP1575492A4 (en) 2002-06-04 2007-05-09 Avalon Pharmaceuticals GENERAL ASSOCIATED WITH CANCER AS TARGET FOR CHEMOTHERAPY
AU2003242633A1 (en) 2002-06-06 2003-12-22 Molecular Engines Laboratories Dudulin genes, non-human animal model: uses in human hematological disease
BRPI0311822B8 (pt) 2002-06-06 2021-05-25 Oncotherapy Science Inc composição farmacêutica para o tratamento de câncer
AU2003245441A1 (en) 2002-06-12 2003-12-31 Avalon Pharmaceuticals, Inc. Cancer-linked gene as target for chemotherapy
US20040249130A1 (en) 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
US20040022727A1 (en) 2002-06-18 2004-02-05 Martin Stanton Aptamer-toxin molecules and methods for using same
AU2003245615A1 (en) 2002-06-20 2004-01-06 The Regents Of The University Of California Compositions and methods for modulating lymphocyte activity
JP2005530856A (ja) 2002-06-21 2005-10-13 ジョンズ ホプキンス ユニバーシティー スクール オブ メディシン 膜関連腫瘍内皮マーカー
WO2004009622A2 (en) 2002-07-19 2004-01-29 Cellzome Ag Protein complexes of cellular networks underlying the development of cancer and other diseases
KR100471357B1 (ko) 2002-07-24 2005-03-10 미래산업 주식회사 반도체 소자 테스트 핸들러용 캐리어 모듈
CA2492447A1 (en) 2002-07-25 2004-02-05 Genentech, Inc. Taci antibodies and uses thereof
US20050180972A1 (en) 2002-07-31 2005-08-18 Wahl Alan F. Anti-CD20 antibody-drug conjugates for the treatment of cancer and immune disorders
AU2003251471A1 (en) 2002-08-06 2004-02-25 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with human cxc chemokine receptor 5(cxcr5)
JP2004121218A (ja) 2002-08-06 2004-04-22 Jenokkusu Soyaku Kenkyusho:Kk 気管支喘息または慢性閉塞性肺疾患の検査方法
AU2003259913A1 (en) 2002-08-19 2004-03-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1391213A1 (en) 2002-08-21 2004-02-25 Boehringer Ingelheim International GmbH Compositions and methods for treating cancer using maytansinoid CD44 antibody immunoconjugates and chemotherapeutic agents
JP2006515742A (ja) 2002-08-27 2006-06-08 ブリストル−マイヤーズ スクイブ カンパニー 乳癌細胞でのプロテインチロシンキナーゼおよび/またはプロテインチロシンキナーゼ経路と相互作用および/またはモデュレートする化合物の活性を予測するためのポリヌクレオチドの同定
WO2004020595A2 (en) 2002-08-29 2004-03-11 Five Prime Therapeutics, Inc. Novel human polypeptides encoded by polynucleotides
AU2002951346A0 (en) 2002-09-05 2002-09-26 Garvan Institute Of Medical Research Diagnosis of ovarian cancer
CN1691964A (zh) 2002-09-06 2005-11-02 曼康公司 表位序列
JP2006513702A (ja) 2002-09-09 2006-04-27 ヌラ インコーポレーティッド Gタンパク質共役受容体およびその使用
JP2004113151A (ja) 2002-09-27 2004-04-15 Sankyo Co Ltd 癌遺伝子及びその用途
AU2003278002A1 (en) 2002-10-03 2004-04-23 Mcgill Univeristy Antibodies and cyclic peptides which bind cea (carcinoembryonic antigen) and their use as cancer therapeutics
CA2501131A1 (en) 2002-10-04 2004-04-22 Van Andel Research Institute Molecular sub-classification of kidney tumors and the discovery of new diagnostic markers
US7608429B2 (en) 2002-10-31 2009-10-27 Genentech, Inc. Methods and compositions for increasing antibody production
CA2503748A1 (en) 2002-11-08 2004-05-27 Genentech, Inc. Compositions and methods for the treatment of natural killer cell related diseases
WO2004044178A2 (en) 2002-11-13 2004-05-27 Genentech, Inc. Methods and compositions for diagnosing dysplasia
AU2003294355A1 (en) 2002-11-15 2004-06-15 The Board Of Trustees Of The University Of Arkansas Ca125 gene and its use for diagnostic and therapeutic interventions
EP2292259A3 (en) 2002-11-15 2011-03-23 MUSC Foundation For Research Development Complement receptor 2 targeted complement modulators
US20080213166A1 (en) 2002-11-20 2008-09-04 Biogen Idec Inc. Novel Gene Targets and Ligands that Bind Thereto for Treatment and Diagnosis of Colon Carcinomas
AU2003294462C1 (en) 2002-11-21 2011-06-30 University Of Utah Research Foundation Purinergic modulation of smell
AU2003298786A1 (en) 2002-11-26 2004-06-18 Protein Design Labs, Inc. Methods of detecting soft tissue sarcoma, compositions and methods of screening for soft tissue sarcoma modulators
EP1581641A4 (en) 2002-12-06 2006-11-15 Diadexus Inc COMPOSITIONS, SPREADING VARIATIONS AND METHODS RELATED TO OVARAL SPECIFIC GENES AND PROTEINS
US20040157278A1 (en) 2002-12-13 2004-08-12 Bayer Corporation Detection methods using TIMP 1
WO2004058171A2 (en) 2002-12-20 2004-07-15 Protein Design Labs, Inc. Antibodies against gpr64 and uses thereof
WO2004058309A1 (en) 2002-12-23 2004-07-15 Human Genome Sciences, Inc. Neutrokine-alpha conjugate, neutrokine-alpha complex, and uses thereof
WO2004063709A2 (en) 2003-01-08 2004-07-29 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
US20050227301A1 (en) 2003-01-10 2005-10-13 Polgen Cell cycle progression proteins
US20050181375A1 (en) 2003-01-10 2005-08-18 Natasha Aziz Novel methods of diagnosis of metastatic cancer, compositions and methods of screening for modulators of metastatic cancer
US20040171823A1 (en) 2003-01-14 2004-09-02 Nadler Steven G. Polynucleotides and polypeptides associated with the NF-kappaB pathway
JP2007520996A (ja) 2003-01-15 2007-08-02 ミレニアム・ファーマシューティカルズ・インコーポレイテッド 44390、54181、211、5687、884、1405、636、4421、5410、30905、2045、16405、18560、2047、33751、52872、14063、20739、32544、43239、44373、51164、53010、16852、1587、2207、22245、2387、52908、69112、14990、18547、115、579、15985、15625、760、18603、2395、2554、8675、32720、4809、14303、16816、17827、32620、577、619、1423、2158、8263、15402、16209、16386、21165、30911、41897、1643、2543、9626、13231、32409、84260、2882、8203、32678または55053を用いて泌尿器科障害を処置するための方法および組成物
CA2513113A1 (en) 2003-01-23 2004-08-05 Genentech, Inc. Methods for producing humanized antibodies and improving yield of antibodies or antigen binding fragments in cell culture
EP1594893A2 (en) 2003-02-14 2005-11-16 Sagres Discovery, Inc. Therapeutic targets in cancer
US20030224411A1 (en) 2003-03-13 2003-12-04 Stanton Lawrence W. Genes that are up- or down-regulated during differentiation of human embryonic stem cells
JP4491576B2 (ja) * 2003-04-22 2010-06-30 株式会社 神崎高級工機製作所 油圧供給装置
ZA200601182B (en) * 2003-10-10 2007-04-25 Immunogen Inc Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
EP1689432B1 (en) * 2003-11-17 2009-12-30 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
EP1718667B1 (en) * 2004-02-23 2013-01-09 Genentech, Inc. Heterocyclic self-immolative linkers and conjugates
CA2559870A1 (en) * 2004-04-07 2005-10-27 Genetech,Inc. Mass spectrometry of antibody conjugates
DK2644194T3 (en) * 2008-03-18 2017-07-03 Genentech Inc Combinations of an anti-HER2 antibody-drug conjugate and docetaxel

Patent Citations (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4137230A (en) * 1977-11-14 1979-01-30 Takeda Chemical Industries, Ltd. Method for the production of maytansinoids
US4265814A (en) * 1978-03-24 1981-05-05 Takeda Chemical Industries Matansinol 3-n-hexadecanoate
US4307016A (en) * 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4361650A (en) * 1978-03-24 1982-11-30 Takeda Chemical Industries, Ltd. Fermentation process of preparing demethyl maytansinoids
US4248870A (en) * 1978-10-27 1981-02-03 Takeda Chemical Industries, Ltd. Maytansinoids and use
US5206018A (en) * 1978-11-03 1993-04-27 Ayerst, Mckenna & Harrison, Inc. Use of rapamycin in treatment of tumors
US4256746A (en) * 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
US4260608A (en) * 1978-11-14 1981-04-07 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and methods of use thereof
US4294757A (en) * 1979-01-31 1981-10-13 Takeda Chemical Industries, Ltd 20-O-Acylmaytansinoids
US4322348A (en) * 1979-06-05 1982-03-30 Takeda Chemical Industries, Ltd. Maytansinoids
US4317821A (en) * 1979-06-08 1982-03-02 Takeda Chemical Industries, Ltd. Maytansinoids, their use and pharmaceutical compositions thereof
US4308268A (en) * 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4308269A (en) * 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4309428A (en) * 1979-07-30 1982-01-05 Takeda Chemical Industries, Ltd. Maytansinoids
US4331598A (en) * 1979-09-19 1982-05-25 Takeda Chemical Industries, Ltd. Maytansinoids
US4364866A (en) * 1979-09-21 1982-12-21 Takeda Chemical Industries, Ltd. Maytansinoids
US4362663A (en) * 1979-09-21 1982-12-07 Takeda Chemical Industries, Ltd. Maytansinoid compound
US4371533A (en) * 1980-10-08 1983-02-01 Takeda Chemical Industries, Ltd. 4,5-Deoxymaytansinoids, their use and pharmaceutical compositions thereof
US4450254A (en) * 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4315929A (en) * 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
US4313946A (en) * 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4424219A (en) * 1981-05-20 1984-01-03 Takeda Chemical Industries, Ltd. 9-Thiomaytansinoids and their pharmaceutical compositions and use
US4753894A (en) * 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US5629197A (en) * 1984-02-08 1997-05-13 Cetus Oncology Corporation Monoclonal anti-human breast cancer antibodies
US4956453A (en) * 1985-12-06 1990-09-11 Cetus Corporation Anti-human ovarian cancer immunotoxins and methods of use thereof
US4958009A (en) * 1985-12-06 1990-09-18 Cetus Corporation Anti-human ovarian cancer immunotoxins and methods of use thereof
US4981979A (en) * 1987-09-10 1991-01-01 Neorx Corporation Immunoconjugates joined by thioether bonds having reduced toxicity and improved selectivity
US5725856A (en) * 1988-01-12 1998-03-10 Genentech, Inc. Monoclonal antibodies directed to the HER2 receptor
US5208020A (en) * 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5416064A (en) * 1989-10-25 1995-05-16 Immunogen, Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5821337A (en) * 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US6632979B2 (en) * 2000-03-16 2003-10-14 Genentech, Inc. Rodent HER2 tumor model
US20020001587A1 (en) * 2000-03-16 2002-01-03 Sharon Erickson Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
US6333410B1 (en) * 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
US20030103985A1 (en) * 2001-05-18 2003-06-05 Boehringer Ingelheim International Gmbh Cytotoxic CD44 antibody immunoconjugates
US6441163B1 (en) * 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
US20040235068A1 (en) * 2001-09-05 2004-11-25 Levinson Arthur D. Methods for the identification of polypeptide antigens associated with disorders involving aberrant cell proliferation and compositions useful for the treatment of such disorders
US6716821B2 (en) * 2001-12-21 2004-04-06 Immunogen Inc. Cytotoxic agents bearing a reactive polyethylene glycol moiety, cytotoxic conjugates comprising polyethylene glycol linking groups, and methods of making and using the same
US20030235582A1 (en) * 2002-06-14 2003-12-25 Immunogen, Inc. Anti-IGF-I receptor antibody
US20040057952A1 (en) * 2002-07-03 2004-03-25 Immunogen Inc. Antibodies to non-shed Muc1 and Muc16, and uses thereof
US6913748B2 (en) * 2002-08-16 2005-07-05 Immunogen, Inc. Cross-linkers with high reactivity and solubility and their use in the preparation of conjugates for targeted delivery of small molecule drugs
US20040126379A1 (en) * 2002-08-21 2004-07-01 Boehringer Ingelheim International Gmbh Compositions and methods for treating cancer using cytotoxic CD44 antibody immunoconjugates and chemotherapeutic agents
US20040120949A1 (en) * 2002-11-08 2004-06-24 Boehringer Ingelheim International Gmbh Compositions and methods for treating cancer using cytotoxic CD44 antibody immunoconjugates and radiotherapy
US20050016993A1 (en) * 2003-04-17 2005-01-27 Koskey James Donald Heated pet mat
US20040241174A1 (en) * 2003-05-14 2004-12-02 Immunogen, Inc. Drug conjugate composition
US20040235840A1 (en) * 2003-05-20 2004-11-25 Immunogen, Inc. Cytotoxic agents comprising new maytansinoids
US20050169933A1 (en) * 2003-10-10 2005-08-04 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates

Cited By (337)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE43899E1 (en) 1999-10-01 2013-01-01 Immunogen Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
USRE44704E1 (en) 1999-10-01 2014-01-14 Immunogen Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
US8337856B2 (en) 2000-03-16 2012-12-25 Immunogen, Inc. Methods of treatment using anti-ERBB antibody-maytansinoid conjugates
US20080226659A1 (en) * 2000-03-16 2008-09-18 Sharon Erickson Methods of treatment using anti-erbb antibody-maytansinoid conjugates
US9562102B2 (en) 2001-05-11 2017-02-07 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US20110045005A1 (en) * 2001-10-19 2011-02-24 Craig Crowley Compositions and methods for the treatment of tumor of hematopoietic origin
US20110206658A1 (en) * 2002-05-08 2011-08-25 Craig Crowley Compositions and methods for the treatment of tumor of hematopoietic origin
US20110070243A1 (en) * 2002-05-08 2011-03-24 Craig Crowley Compositions and methods for the treatment of tumor of hematopoietic
US9085630B2 (en) 2002-11-15 2015-07-21 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
US10844135B2 (en) 2003-10-10 2020-11-24 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates and methods of making said
US8017735B2 (en) * 2003-11-21 2011-09-13 Schering Corporation Anti-IGFR1 antibody therapeutic combinations
US20050136063A1 (en) * 2003-11-21 2005-06-23 Schering Corporation Anti-IGFR antibody therapeutic combinations
US7989434B2 (en) 2004-02-23 2011-08-02 Seattle Genetics, Inc. Heterocyclic self-immolative linkers and conjugates
US20090041791A1 (en) * 2004-02-23 2009-02-12 Bainian Feng Heterocyclic self-immolative Linkers and Conjugates
US20100273843A1 (en) * 2004-02-23 2010-10-28 Bainian Feng Heterocyclic self-immolative linkers and conjugates
US7754681B2 (en) 2004-02-23 2010-07-13 Seattle Genetics, Inc. Heterocyclic self-immolative linkers and conjugates
US20060013819A1 (en) * 2004-06-16 2006-01-19 Genentech, Inc. Therapy of platinum-resistant cancer
US9017671B2 (en) 2004-10-20 2015-04-28 Genentech, Inc. Method of treating cancer with a pharmaceutical formulation comprising a HER2 antibody
US20090081223A1 (en) * 2005-01-21 2009-03-26 Genentech, Inc. Fixed dosing of her antibodies
US8404234B2 (en) 2005-01-21 2013-03-26 Genentech, Inc. Fixed dosing of HER antibodies
US8691232B2 (en) 2005-02-23 2014-04-08 Genentech, Inc. Extending time to disease progression or survival in cancer patients
US20090155259A1 (en) * 2005-02-23 2009-06-18 Genentech, Inc. Extending time to disease progression or survival in cancer patients
US8597654B2 (en) 2005-05-13 2013-12-03 Genentech, Inc. Adjuvant therapy with an anti-ERBB2 antibody conjugated to a maytansiniod
US11034754B2 (en) * 2005-05-25 2021-06-15 Hoffmann-La Roche Inc. Method for the purification of antibodies
US20170183394A1 (en) * 2005-05-25 2017-06-29 Hoffmann-La Roche Inc. Method for the purification of antibodies
USRE47223E1 (en) 2005-06-20 2019-02-05 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8163287B2 (en) 2005-07-22 2012-04-24 Genentech, Inc. Combination therapy of her expressing tumors
US8153127B2 (en) 2006-02-07 2012-04-10 Phenoquest Ag Method of treating depression
WO2007090631A1 (en) * 2006-02-07 2007-08-16 Affectis Pharmaceuticals Ag Methods for the diagnosis and treatment of affective disorders and cushing's syndromes
US20100069463A1 (en) * 2006-02-07 2010-03-18 Affectis Pharmaceuticals Ag Methods for the diagnosis and treatment of affective disorders and cushing's syndromes
US9109035B2 (en) 2006-03-17 2015-08-18 Genentech, Inc. Anti-TAT226 antibodies and immunoconjugates
US9675707B2 (en) 2006-03-17 2017-06-13 Genentech, Inc. Anti-TAT226 antibodies and immunoconjugates
US8183347B2 (en) 2006-03-17 2012-05-22 Genentech, Inc. Anti-TAT226 antibodies and immunoconjugates
US8524865B2 (en) 2006-05-30 2013-09-03 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
US8968741B2 (en) 2006-05-30 2015-03-03 Genentech, Inc. Anti-CD22 antibodies and immunoconjugates and methods of use
US8226945B2 (en) 2006-05-30 2012-07-24 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
US20080050310A1 (en) * 2006-05-30 2008-02-28 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
US8394607B2 (en) 2006-05-30 2013-03-12 Genentech, Inc. Anti-CD22 antibodies and immunoconjugates and methods of use
US20110142859A1 (en) * 2006-05-30 2011-06-16 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
USRE47123E1 (en) * 2006-07-18 2018-11-13 Sanofi EPHA2 receptor antagonist antibodies
US7825267B2 (en) 2006-09-08 2010-11-02 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Synthesis of FR901464 and analogs with antitumor activity
US20080096879A1 (en) * 2006-09-08 2008-04-24 University Of Pittsburgh-- Of The Commonwealth System Of Higher Education Synthesis of fr901464 and analogs with antitumor activity
US20110086909A1 (en) * 2006-09-08 2011-04-14 University Of Pittsburgh-- Of The Commonwealth System Of Higher Education Synthesis of fr901464 and analogs with antitumor activity
US8309599B2 (en) 2006-09-08 2012-11-13 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Synthesis of FR901464 and analogs with antitumor activity
US9090693B2 (en) 2007-01-25 2015-07-28 Dana-Farber Cancer Institute Use of anti-EGFR antibodies in treatment of EGFR mutant mediated disease
EP2468776A2 (en) 2007-02-09 2012-06-27 Genentech, Inc. Anti-Robo4 antibodies and uses therefor
US8940302B2 (en) 2007-03-02 2015-01-27 Genentech, Inc. Predicting response to a HER inhibitor
US9023356B2 (en) 2007-03-15 2015-05-05 Ludwig Institute For Cancer Research Ltd Treatment method using EGFR antibodies and SRC inhibitors and related formulations
US7723485B2 (en) 2007-05-08 2010-05-25 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
USRE47194E1 (en) 2007-05-08 2019-01-08 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
US20140110297A1 (en) * 2007-06-25 2014-04-24 The Regents Of The University Of Colorado "Medical Articles Coated With Organopolysiloxane Containing a Protein Solution"
US20090068202A1 (en) * 2007-07-16 2009-03-12 Genentech, Inc. Humanized Anti-CD79B Antibodies and Immunoconjugates and Methods of Use
US10981987B2 (en) 2007-07-16 2021-04-20 Genentech, Inc. Humanized anti-CD79b antibodies and immunoconjugates and methods of use
WO2009012256A1 (en) 2007-07-16 2009-01-22 Genentech, Inc. Humanized anti-cd79b antibodies and immunoconjugates and methods of use
EP2474557A2 (en) 2007-07-16 2012-07-11 Genentech, Inc. Anti-CD79b antibodies and immunoconjugates and methods of use
US11866496B2 (en) 2007-07-16 2024-01-09 Genentech, Inc. Humanized anti-CD79B antibodies and immunoconjugates and methods of use
USRE48558E1 (en) 2007-07-16 2021-05-18 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US20110135667A1 (en) * 2007-07-16 2011-06-09 Genentech, Inc. Anti-cd79b antibodies and immunoconjugates and methods of use
US8691531B2 (en) 2007-07-16 2014-04-08 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
EP2502937A2 (en) 2007-07-16 2012-09-26 Genentech, Inc. Anti-CD 79b Antibodies And Immunoconjugates And Methods Of Use
EP2641618A2 (en) 2007-07-16 2013-09-25 Genentech, Inc. Humanized anti-CD79B antibodies and immunoconjugates and methods of use
US20090028856A1 (en) * 2007-07-16 2009-01-29 Genentech, Inc. Anti-CD79B Antibodies and Immunoconjugates and Methods of Use
US9845355B2 (en) 2007-07-16 2017-12-19 Genentech, Inc. Humanized anti-CD79b antibodies and immunoconjugates and methods of use
RU2791984C2 (ru) * 2007-07-16 2023-03-15 Дженентек, Инк. АНТИ-CD79b АНТИТЕЛА И ИММУНОКОНЪЮГАТЫ И СПОСОБЫ ИХ ПРИМЕНЕНИЯ
US10494432B2 (en) 2007-07-16 2019-12-03 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US8545850B2 (en) 2007-07-16 2013-10-01 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US8088378B2 (en) 2007-07-16 2012-01-03 Genetech Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US9283276B2 (en) 2007-08-14 2016-03-15 Ludwig Institute For Cancer Research Ltd. Monoclonal antibody 175 targeting the EGF receptor and derivatives and uses thereof
KR20100061815A (ko) * 2007-08-29 2010-06-09 사노피-아벤티스 인간화된 항-cxcr5 항체, 이의 유도체 및 이들의 용도
US8980262B2 (en) * 2007-08-29 2015-03-17 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
CN111909273A (zh) * 2007-08-29 2020-11-10 塞诺菲-安万特股份有限公司 人源化的抗-cxcr5抗体、其衍生物及它们的应用
CN104017078B (zh) * 2007-08-29 2020-09-08 塞诺菲-安万特股份有限公司 人源化的抗-cxcr5抗体、其衍生物及它们的应用
CN101842115B (zh) * 2007-08-29 2014-07-30 塞诺菲-安万特股份有限公司 人源化的抗-cxcr5抗体、其衍生物及它们的应用
US20170044263A1 (en) * 2007-08-29 2017-02-16 Sanofi Humanized Anti-CXCR5 Antibodies, Derivatives Thereof and Their Uses
KR20150082692A (ko) * 2007-08-29 2015-07-15 사노피 인간화된 항-cxcr5 항체, 이의 유도체 및 이들의 용도
RU2571515C2 (ru) * 2007-08-29 2015-12-20 Санофи-Авентис Гуманизированные антитела к cxcr5, их производные и их применение
US20130236476A1 (en) * 2007-08-29 2013-09-12 Sanofi Humanized Anti-CXCR5 Antibodies, Derivatives Thereof And Their Use
US9228019B2 (en) * 2007-08-29 2016-01-05 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9243067B2 (en) * 2007-08-29 2016-01-26 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US20130243783A1 (en) * 2007-08-29 2013-09-19 Sanofi Humanized Anti-CXCR5 Antibodies, Derivatives Thereof And Their Use
US20130246035A1 (en) * 2007-08-29 2013-09-19 Sanofi Humanized Anti-CXCR5 Antibodies, Derivatives Thereof And Their Use
AU2008296538B2 (en) * 2007-08-29 2014-08-14 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
US20130251714A1 (en) * 2007-08-29 2013-09-26 Sanofi Humanized Anti-CXCR5 Antibodies, Derivatives Thereof And Their Use
AU2013203265B2 (en) * 2007-08-29 2016-06-23 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
AU2013203262B2 (en) * 2007-08-29 2016-07-07 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
US9175087B2 (en) * 2007-08-29 2015-11-03 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9815902B2 (en) * 2007-08-29 2017-11-14 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
US20110027266A1 (en) * 2007-08-29 2011-02-03 Sanofi-Aventis Humanized anti-CXCR5 antibodies, derivatives thereof and their use
AU2013203264B2 (en) * 2007-08-29 2016-07-07 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
CN104017078A (zh) * 2007-08-29 2014-09-03 塞诺菲-安万特股份有限公司 人源化的抗-cxcr5抗体、其衍生物及它们的应用
EP3354662A1 (en) * 2007-08-29 2018-08-01 Sanofi Humanized anti-cxcr5 antibodies, derivatives thereof and their uses
TWI608017B (zh) * 2007-08-29 2017-12-11 賽諾菲 安萬特公司 人化抗cxcr5抗體類、彼之衍生物類及彼等之用途
EP3792284A1 (en) * 2007-08-29 2021-03-17 Sanofi Humanized anti-cxcr5 antibodies, derivatives thereof and their uses
WO2009032661A1 (en) * 2007-08-29 2009-03-12 Sanofi-Aventis Humanized anti-cxcr5 antibodies, derivatives thereof and their uses
US8647622B2 (en) * 2007-08-29 2014-02-11 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
KR101701140B1 (ko) * 2007-08-29 2017-02-01 사노피 인간화된 항-cxcr5 항체, 이의 유도체 및 이들의 용도
KR101599704B1 (ko) * 2007-08-29 2016-03-07 사노피 인간화된 항-cxcr5 항체, 이의 유도체 및 이들의 용도
US8268970B2 (en) 2007-10-01 2012-09-18 Bristol-Myers Squibb Company Human antibodies that bind mesothelin, and uses thereof
US8383779B2 (en) 2007-10-01 2013-02-26 Bristol-Myers Squibb Company Human antibodies that bind mesothelin, and uses thereof
US8399623B2 (en) 2007-10-01 2013-03-19 Bristol-Myers Squibb Company Human antibodies that bind mesothelin, and uses thereof
US8425904B2 (en) 2007-10-01 2013-04-23 Bristol-Myers Squibb Company Human antibodies that bind mesothelin, and uses thereof
WO2009054001A1 (en) * 2007-10-22 2009-04-30 Biocon Limited A pharmaceutical composition and a process thereof
EP3056576A3 (en) * 2007-10-23 2016-09-28 Clinical Genomics Pty Ltd A method of diagnosing neoplasms
US8652474B2 (en) 2008-01-30 2014-02-18 Genentech, Inc. Composition comprising antibody that binds to domain II of HER2 and acidic variants thereof
US9181346B2 (en) 2008-01-30 2015-11-10 Genentech, Inc. Composition comprising antibody that binds to domain II of HER2 and acidic variants thereof
US11414498B2 (en) 2008-01-30 2022-08-16 Genentech, Inc. Composition comprising antibody that binds to domain II of HER2 and acidic variants thereof
US11597776B2 (en) 2008-01-30 2023-03-07 Genentech, Inc. Composition comprising antibody that binds to domain II of HER2 and acidic variants thereof
US9896506B2 (en) 2008-01-31 2018-02-20 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
EP2657253A2 (en) 2008-01-31 2013-10-30 Genentech, Inc. Anti-CD79b antibodies and immunoconjugates and methods of use
US8722857B2 (en) 2008-01-31 2014-05-13 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US20100215669A1 (en) * 2008-01-31 2010-08-26 Genentech, Inc. Anti-cd79b antibodies and immunoconjugates and methods of use
US10544218B2 (en) 2008-01-31 2020-01-28 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
KR101751065B1 (ko) * 2008-03-18 2017-06-26 제넨테크, 인크. 항-her2 항체-약물 접합체와 화학요법제의 병용물, 및 사용 방법
EP2638907A2 (en) 2008-03-18 2013-09-18 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and lapatinib
TWI449536B (zh) * 2008-03-18 2014-08-21 Genentech Inc 抗her2抗體-藥物結合物及化療劑之組合及使用方法
IL274393B (en) * 2008-03-18 2022-09-01 Genentech Inc Aggregates of frequent anti-her-2 antibody conjugates and co-therapeutic agents and methods of using them
RU2671489C2 (ru) * 2008-03-18 2018-11-01 Дженентек, Инк. Комбинации конъюгата анти-her2-антитело-лекарственное средство и химиотерапевтических средств и способы применения
WO2009117277A3 (en) * 2008-03-18 2010-04-29 Genentech, Inc. Combinations of an anti-her2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
US8663643B2 (en) 2008-03-18 2014-03-04 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
AU2009225877B2 (en) * 2008-03-18 2014-11-20 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
EP3269366A2 (en) 2008-03-18 2018-01-17 Genentech, Inc. Combinations of an anti-her2 antibody-drug conjugate and lapatinib, and methods of use
EP2644204A3 (en) * 2008-03-18 2014-02-12 Genentech, Inc. Combinations of an Anti-HER2 antibody-drug conjugate and pertuzumab
EP2644194A3 (en) * 2008-03-18 2014-01-01 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and docetaxel
TWI675668B (zh) * 2008-03-18 2019-11-01 美商建南德克公司 抗her2抗體-藥物結合物及化療劑之組合及使用方法
CN102036660A (zh) * 2008-03-18 2011-04-27 健泰科生物技术公司 抗her2抗体-药物偶联物和化疗剂的组合,及使用方法
EP3692988A2 (en) 2008-03-18 2020-08-12 Genentech, Inc. Combinations of an anti-her2 antibody-drug conjugate and 5-fu, anti-vegf antibody, carboplatin or abt-869 and methods of use
EP2644204A2 (en) 2008-03-18 2013-10-02 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and pertuzumab
AU2017200047B2 (en) * 2008-03-18 2018-11-15 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
EP2254571A2 (en) * 2008-03-18 2010-12-01 Genentech, Inc. Combinations of an anti-her2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
TWI574698B (zh) * 2008-03-18 2017-03-21 建南德克公司 抗her2抗體-藥物結合物及化療劑之組合及使用方法
EP2644194A2 (en) 2008-03-18 2013-10-02 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and docetaxel
RU2781195C2 (ru) * 2008-03-18 2022-10-07 Дженентек, Инк. Комбинации конъюгата анти-her2-антитело-лекарственное средство и химиотерапевтических средств и способы применения
EP2638907A3 (en) * 2008-03-18 2014-05-14 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and lapatinib
AU2014240234B2 (en) * 2008-03-18 2016-10-06 Genentech, Inc. Combinations of an anti-HER2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
RU2510272C2 (ru) * 2008-03-18 2014-03-27 Дженентек, Инк. Комбинации конъюгата анти-her2-антитело-лекарственное средство и химиотерапевтических средств и способы применения
US20110059852A1 (en) * 2008-03-26 2011-03-10 Cellerant Therapeutics, Inc. Compositions and methods for treating haematological proliferative disorders of meyloid origin
US9371390B2 (en) 2008-03-26 2016-06-21 Cellerant Therapeutics, Inc. Cytokine receptors associated with myelogenous haematological proliferative disorders and uses thereof
US8715619B2 (en) * 2008-03-26 2014-05-06 Cellerant Therapeutics, Inc. Compositions and methods for treating haematological proliferative disorders of myeloid origin
US8709715B2 (en) 2008-03-26 2014-04-29 Cellerant Therapeutics, Inc. Cytokine receptors associated with myelogenous haematological proliferative disorders and uses thereof
US20130216558A1 (en) * 2008-03-26 2013-08-22 Cellerant Therapeutics, Inc. Compositions and methods for treating haematological proliferative disorders of myeloid origin
US10479831B2 (en) 2008-04-02 2019-11-19 Macrogenics, Inc BCR-complex-specific antibodies and methods of using same
US8993730B2 (en) 2008-04-02 2015-03-31 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US20110117089A1 (en) * 2008-04-02 2011-05-19 Macrogenics, Inc. BCR-Complex-Specific Antibodies And Methods Of Using Same
US8669349B2 (en) 2008-04-02 2014-03-11 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US9695236B2 (en) 2008-04-02 2017-07-04 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US20120226026A1 (en) * 2008-04-30 2012-09-06 Immunogen, Inc. Potent conjugates and hydrophilic linkers
EP2276506A1 (en) * 2008-04-30 2011-01-26 Immunogen, Inc. Potent conjugates and hydrophilic linkers
WO2009134870A1 (en) * 2008-04-30 2009-11-05 Immunogen, Inc. Potent cell-binding agent drug conjugates
US20100092495A1 (en) * 2008-04-30 2010-04-15 Immunogen Inc. Potent cell-binding agent drug conjugates
US20100129314A1 (en) * 2008-04-30 2010-05-27 Immunogen Inc. Potent conjugates and hydrophilic linkers
EP2276506A4 (en) * 2008-04-30 2014-05-07 Immunogen Inc EFFICIENT CONJUGATES AND HYDROPHILIC BINDER
US9150649B2 (en) 2008-04-30 2015-10-06 Immunogen, Inc. Potent conjugates and hydrophilic linkers
US10689457B2 (en) 2008-06-16 2020-06-23 Genentech, Inc. Treatment of metastatic breast cancer
US11655305B2 (en) 2008-06-16 2023-05-23 Genentech, Inc. Treatment of metastatic breast cancer
WO2010009124A2 (en) 2008-07-15 2010-01-21 Genentech, Inc. Anthracycline derivative conjugates, process for their preparation and their use as antitumor compounds
US9072798B2 (en) 2009-02-18 2015-07-07 Ludwig Institute For Cancer Research Ltd. Specific binding proteins and uses thereof
WO2010120561A1 (en) 2009-04-01 2010-10-21 Genentech, Inc. Anti-fcrh5 antibodies and immunoconjugates and methods of use
KR101681795B1 (ko) 2009-04-29 2016-12-01 바이엘 인텔렉쳐 프로퍼티 게엠베하 항메소텔린 면역접합체 및 그의 용도
TWI472343B (zh) * 2009-04-29 2015-02-11 Bayer Ip Gmbh 抗-間皮素的免疫接合物及其用途
WO2010124797A1 (en) * 2009-04-29 2010-11-04 Bayer Schering Pharma Aktiengesellschaft Anti-mesothelin immunoconjugates and uses therefor
US10781263B2 (en) 2009-04-29 2020-09-22 Bayer Intellectual Property Gmbh Anti-mesothelin immunoconjugates and uses therefor
US20120189644A1 (en) * 2009-04-29 2012-07-26 Bayer Pharma Aktiengesellschaft Anti-mesothelin immunoconjugates and uses therefor
US20150322160A1 (en) * 2009-04-29 2015-11-12 Bayer Intellectual Property Gmbh Anti-mesothelin immunoconjugates and uses therefor
AU2010243942B2 (en) * 2009-04-29 2015-01-29 Bayer Intellectual Property Gmbh Anti-mesothelin immunoconjugates and uses therefor
US9084829B2 (en) * 2009-04-29 2015-07-21 Bayer Intellectual Property Gmbh Anti-mesothelin immunoconjugates and uses therefor
JP2017039735A (ja) * 2009-04-29 2017-02-23 バイエル・インテレクチュアル・プロパティ・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツングBayer Intellectual Property GmbH 抗メソテリン免疫複合体およびその使用
US10647779B2 (en) 2009-04-29 2020-05-12 Bayer Intellectual Property Gmbh Anti-mesothelin immunoconjugates and uses therefor
KR20120031265A (ko) * 2009-04-29 2012-04-02 바이엘 파마 악티엔게젤샤프트 항메소텔린 면역접합체 및 그의 용도
US9676864B2 (en) 2009-10-02 2017-06-13 Sanofi Antibodies that specifically bind to the EphA2 receptor
WO2011069074A2 (en) 2009-12-04 2011-06-09 Genentech, Inc. Methods of treating metastatic breast cancer with trastuzumab-mcc-dm1
US20110165155A1 (en) * 2009-12-04 2011-07-07 Genentech, Inc. Methods of treating metastatic breast cancer with trastuzumab-mcc-dm1
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011109308A1 (en) * 2010-03-02 2011-09-09 Seattle Genetics, Inc. Methods for screening antibodies
US9725500B2 (en) 2010-03-02 2017-08-08 Seattle Genetics, Inc. Methods for screening antibodies
US8771966B2 (en) 2010-06-03 2014-07-08 Genentech, Inc. Immuno-PET imaging of antibodies and immunoconjugates and uses therefor
US9492565B2 (en) 2010-06-15 2016-11-15 Genmab A/S Human antibody drug conjugates against tissue factor
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
WO2012074757A1 (en) 2010-11-17 2012-06-07 Genentech, Inc. Alaninyl maytansinol antibody conjugates
WO2012078771A1 (en) 2010-12-09 2012-06-14 Genentech, Inc. Treatment of her2-positive cancer with paclitaxel and trastuzumab-mcc-dm1
US10022452B2 (en) 2010-12-20 2018-07-17 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US9719996B2 (en) 2010-12-20 2017-08-01 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US8911732B2 (en) 2010-12-20 2014-12-16 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US10676515B2 (en) 2011-01-31 2020-06-09 The General Hospital Corporation Multimodal trail molecules expressed in T cells
US9156854B2 (en) 2011-04-18 2015-10-13 Immunogen, Inc. Maytansinoid derivatives with sulfoxide linker
WO2012145112A2 (en) * 2011-04-18 2012-10-26 Immunogen, Inc. Novel maytansinoid derivatives with sulfoxide linker
WO2012145112A3 (en) * 2011-04-18 2014-05-01 Immunogen, Inc. Novel maytansinoid derivatives with sulfoxide linker
US9624240B2 (en) 2011-04-18 2017-04-18 Immunogen, Inc. Maytansinoid derivatives with sulfoxide linker
WO2012154809A1 (en) 2011-05-09 2012-11-15 University Of Virginia Patent Foundation Compositions and methods for treating cancer
WO2013033380A1 (en) 2011-08-31 2013-03-07 Genentech, Inc. Diagnostic markers
US9574008B2 (en) * 2011-09-22 2017-02-21 Amgen Inc. CD27L antigen binding proteins
WO2013043933A2 (en) 2011-09-22 2013-03-28 Amgen Inc. Cd27l antigen binding proteins
US20150218284A1 (en) * 2011-09-22 2015-08-06 Amgen Inc. Cd27l antigen binding proteins
US10266604B2 (en) 2011-09-22 2019-04-23 Amgen Inc. CD27L antigen binding proteins
EP4241849A2 (en) 2011-10-14 2023-09-13 F. Hoffmann-La Roche AG Uses for and article of manufacture including her2 dimerization inhibitor pertuzumab
EP4234034A2 (en) 2011-10-14 2023-08-30 F. Hoffmann-La Roche AG Uses for and article of manufacture including her2 dimerization inhibitor pertuzumab
EP3598981A2 (en) 2011-10-14 2020-01-29 F. Hoffmann-La Roche AG Uses for and article of manufacture including her2 dimerization inhibitor pertuzumab
EP4234033A2 (en) 2011-10-14 2023-08-30 F. Hoffmann-La Roche AG Uses for and article of manufacture including her2 dimerization inhibitor pertuzumab
WO2013055874A2 (en) 2011-10-14 2013-04-18 Genentech, Inc. Uses for and article of manufacture including her2 dimerization inhibitor pertuzumab
WO2013063001A1 (en) 2011-10-28 2013-05-02 Genentech, Inc. Therapeutic combinations and methods of treating melanoma
EP3296320A1 (en) 2012-03-08 2018-03-21 Halozyme, Inc. Conditionally active anti-epidermal growth factor receptor antibodies and methods of use thereof
WO2013134743A1 (en) 2012-03-08 2013-09-12 Halozyme, Inc. Conditionally active anti-epidermal growth factor receptor antibodies and methods of use thereof
US10525130B2 (en) 2012-03-26 2020-01-07 Sanofi Stable IGG4 based binding agent formulations
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
WO2013149159A1 (en) 2012-03-30 2013-10-03 Genentech, Inc. Anti-lgr5 antibodies and immunoconjugates
US9175089B2 (en) 2012-03-30 2015-11-03 Genentech, Inc. Anti-LGR5 antibodies and immunoconjugates
US9597411B2 (en) 2012-05-01 2017-03-21 Genentech, Inc. Anti-PMEL17 antibodies and immunoconjugates
US9056910B2 (en) 2012-05-01 2015-06-16 Genentech, Inc. Anti-PMEL17 antibodies and immunoconjugates
WO2013165940A1 (en) 2012-05-01 2013-11-07 Genentech, Inc. Anti-pmel17 antibodies and immunoconjugates
US10196454B2 (en) 2012-05-01 2019-02-05 Genentech, Inc. Anti-PMEL17 antibodies and immunoconjugates
EP3693395A1 (en) 2012-05-18 2020-08-12 Amgen Inc. St2 antigen binding proteins
WO2013173761A2 (en) 2012-05-18 2013-11-21 Amgen Inc. St2 antigen binding proteins
WO2013182668A1 (en) 2012-06-08 2013-12-12 F. Hoffmann-La Roche Ag Mutant selectivity and combinations of a phosphoinositide 3 kinase inhibitor compound and chemotherapeutic agents for the treatment of cancer
EP3545968A1 (en) 2012-06-08 2019-10-02 F. Hoffmann-La Roche AG Mutant selectivity and combinations of a phosphoinositide 3 kinase inhibitor compound and chemotherapeutic agents for the treatment of cancer
US10633636B2 (en) 2012-07-24 2020-04-28 The General Hospital Corporation Oncolytic virus therapy for resistant tumors
US10130680B2 (en) * 2012-08-30 2018-11-20 The General Hospital Corporation Methods for treating cancer with EGFR nanobodies linked to DR5 binding moieties
US20170151345A1 (en) * 2012-08-30 2017-06-01 The General Hospital Corporation Compositions and methods for treating cancer
US8853154B2 (en) 2012-09-13 2014-10-07 Bristol-Myers Squibb Company Fibronectin based scaffold domain proteins that bind to myostatin
US10973924B2 (en) 2012-10-11 2021-04-13 Daiichi Sankyo Company, Limited Antibody-drug conjugate
US11633493B2 (en) 2012-10-11 2023-04-25 Daiichi Sankyo Company, Limited Antibody-drug conjugate
US10729782B2 (en) 2012-10-19 2020-08-04 Daiichi Sankyo Company, Limited Antibody-drug conjugate produced by binding through linker having hydrophilic structure
WO2014089335A2 (en) 2012-12-07 2014-06-12 Amgen Inc. Bcma antigen binding proteins
US9771377B2 (en) 2012-12-21 2017-09-26 University of Pittsburgh—of the Commonwealth System of Higher Education Synthesis of FR901464 and analogs with antitumor activity
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
US11053311B2 (en) 2013-01-25 2021-07-06 Amgen Inc. Antibodies targeting CDH19 for melanoma
US9920121B2 (en) * 2013-01-25 2018-03-20 Amgen Inc. Antibodies targeting CDH19 for melanoma
US20150322151A1 (en) * 2013-01-25 2015-11-12 Amgen Inc. Antibodies targeting cdh19 for melanoma
US11230600B2 (en) 2013-03-14 2022-01-25 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US10150813B2 (en) 2013-03-14 2018-12-11 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
EP3299391A1 (en) 2013-03-14 2018-03-28 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US9969811B2 (en) 2013-04-16 2018-05-15 Genentech, Inc. Pertuzumab variants and evaluation thereof
US9815904B2 (en) 2013-04-16 2017-11-14 Genetech, Inc. Pertuzumab variants and evaluation thereof
US10201544B2 (en) 2013-04-26 2019-02-12 Adc Biotechnology Ltd. Method of synthesising ADCs using affinity resins
WO2014174316A1 (en) * 2013-04-26 2014-10-30 Adc Biotechnology Ltd Method of synthesising adcs using affinity resins
EP3456743A1 (en) 2013-05-30 2019-03-20 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
EP3971212A1 (en) 2013-05-30 2022-03-23 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
EP4349864A2 (en) 2013-05-30 2024-04-10 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
US10071170B2 (en) 2013-06-24 2018-09-11 Ablbio Antibody-drug conjugate having improved stability and use thereof
AU2014299561B2 (en) * 2013-06-24 2017-06-08 Ablbio Antibody-drug conjugate having improved stability and use thereof
WO2015038984A2 (en) 2013-09-12 2015-03-19 Halozyme, Inc. Modified anti-epidermal growth factor receptor antibodies and methods of use thereof
US10246515B2 (en) 2013-09-17 2019-04-02 Genentech, Inc. Methods of treating hedgehog-related diseases with an anti-LGR5 antibody
WO2015042108A1 (en) 2013-09-17 2015-03-26 Genentech, Inc. Methods of using anti-lgr5 antibodies
WO2015089344A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
EP3461845A1 (en) 2013-12-13 2019-04-03 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US11008398B2 (en) 2013-12-25 2021-05-18 Daiichi Sankyo Company, Limited Anti-TROP2 antibody-drug conjugate
WO2015112909A1 (en) 2014-01-24 2015-07-30 Genentech, Inc. Methods of using anti-steap1 antibodies and immunoconjugates
US11584800B2 (en) 2014-01-31 2023-02-21 Daiichi Sankyo Company, Limited Method of treating cancer comprising administration of anti-HER2 antibody-drug conjugate
US11795236B2 (en) 2014-01-31 2023-10-24 Daiichi Sankyo Company, Limited Method for treating cancer comprising administration of anti-HER2 antibody-drug conjugate
US11298359B2 (en) 2014-04-10 2022-04-12 Daiichi Sankyo Company, Limited Anti-HER3 antibody-drug conjugate
US11185594B2 (en) 2014-04-10 2021-11-30 Daiichi Sankyo Company, Limited (Anti-HER2 antibody)-drug conjugate
US10383878B2 (en) 2014-04-10 2019-08-20 Daiichi Sankyo Company, Limited Anti-HER3 antibody-drug conjugate
WO2015164665A1 (en) 2014-04-25 2015-10-29 Genentech, Inc. Methods of treating early breast cancer with trastuzumab-mcc-dm1 and pertuzumab
WO2015179658A2 (en) 2014-05-22 2015-11-26 Genentech, Inc. Anti-gpc3 antibodies and immunoconjugates
US10316084B2 (en) 2014-06-11 2019-06-11 Genentech, Inc. Anti-LGR5 antibodies and uses thereof
WO2016014360A1 (en) * 2014-07-24 2016-01-28 Genentech, Inc. Methods of conjugating an agent to a thiol moiety in a protein that contains at least one trisulfide bond
US11370838B2 (en) 2014-07-24 2022-06-28 Genentech, Inc. Methods of conjugating an agent to a thiol moiety in a protein that contains at least one sulfide bond
US11584927B2 (en) 2014-08-28 2023-02-21 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified T-cells
US11286302B2 (en) 2014-09-12 2022-03-29 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2016040868A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
EP3693391A1 (en) 2014-09-12 2020-08-12 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
EP3782654A1 (en) 2014-09-12 2021-02-24 Genentech, Inc. Anti-her2 antibodies and immunoconjugates
US10059768B2 (en) 2014-09-12 2018-08-28 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2016044396A1 (en) 2014-09-17 2016-03-24 Genentech, Inc. Immunoconjugates comprising anti-her2 antibodies and pyrrolobenzodiazepines
US11000510B2 (en) 2014-09-23 2021-05-11 Genentech, Inc. Methods of using anti-CD79b immunoconjugates
EP3689910A2 (en) 2014-09-23 2020-08-05 F. Hoffmann-La Roche AG Method of using anti-cd79b immunoconjugates
US20170326251A1 (en) * 2014-10-28 2017-11-16 Adc Biotechnology Ltd. Method of synthesizing antibody drug conjugates using affinity resins
GB2557389A (en) * 2015-01-14 2018-06-20 Brigham & Womens Hospital Inc Treatment of cancer with anti-lap monoclonal antibodies
GB2557389B (en) * 2015-01-14 2020-12-23 Brigham & Womens Hospital Inc Treatment of cancer with anti-lap monoclonal antibodies
US11407823B2 (en) 2015-01-14 2022-08-09 The Brigham And Women's Hospital, Inc. Treatment of cancer with anti-LAP monoclonal antibodies
US10287347B2 (en) 2015-01-14 2019-05-14 The Brigham And Women's Hospital, Inc. Treatment of cancer with anti-lap monoclonal antibodies
WO2016115345A1 (en) * 2015-01-14 2016-07-21 The Brigham And Women's Hospital, Treatment of cancer with anti-lap monoclonal antibodies
US10017567B2 (en) 2015-01-14 2018-07-10 The Brigham And Women's Hospital, Inc. Treatment of cancer with anti-LAP monoclonal antibodies
WO2016196373A2 (en) 2015-05-30 2016-12-08 Genentech, Inc. Methods of treating her2-positive metastatic breast cancer
US11406715B2 (en) 2015-05-30 2022-08-09 Genentech, Inc. Methods of treating HER2-positive metastatic breast cancer
US10449258B2 (en) 2015-06-09 2019-10-22 Xdcexplorer (Shanghai) Co., Ltd. Antibody drug conjugate, intermediate, preparation method, pharmaceutical composition and uses thereof
EP3308801A4 (en) * 2015-06-09 2019-02-27 XDCExplorer (Shanghai) Co., Ltd. ANTIBODY-MEDICINAL CONJUGATE, INTERMEDIATE, PREPARATION METHOD, PHARMACEUTICAL COMPOSITIONS AND USES THEREOF
US11173213B2 (en) 2015-06-29 2021-11-16 Daiichi Sankyo Company, Limited Method for selectively manufacturing antibody-drug conjugate
US10898570B2 (en) 2015-07-07 2021-01-26 Genentech, Inc. Combination therapy with an anti-HER2 antibody-drug conjugate and a Bcl-2 inhibitor
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
WO2017087280A1 (en) 2015-11-16 2017-05-26 Genentech, Inc. Methods of treating her2-positive cancer
US11938193B2 (en) * 2016-01-08 2024-03-26 Washington University Compositions comprising chemerin and methods of use thereof
US10874746B2 (en) 2016-02-08 2020-12-29 Synaffix B.V. Sulfamide linkers for use in bioconjugates
US11590239B2 (en) 2016-02-08 2023-02-28 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting CD30 tumours and method for improving therapeutic index of antibody-conjugates
US11957763B2 (en) 2016-02-08 2024-04-16 Synaffix B.V. Sulfamide linkers for use in bioconjugates
WO2017137457A1 (en) * 2016-02-08 2017-08-17 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting cd30 tumours and method for improving therapeutic index of antibody-conjugates
US11951175B2 (en) 2016-02-08 2024-04-09 Synaffix B.V. Bioconjugates containing sulfamide linkers for use in treatment
US11338043B2 (en) 2016-02-08 2022-05-24 Synaffix B.V. Antibody-conjugates with improved therapeutic index for targeting HER2 tumours and method for improving therapeutic index of antibody-conjugates
US10914720B2 (en) 2016-02-10 2021-02-09 Becton Dickinson France Method to evaluate the stability of a protein-based formulation
WO2017161206A1 (en) 2016-03-16 2017-09-21 Halozyme, Inc. Conjugates containing conditionally active antibodies or antigen-binding fragments thereof, and methods of use
US11897959B2 (en) 2016-04-15 2024-02-13 Bioatla, Inc. Anti-AXL antibodies, antibody fragments and their immunoconjugates and uses thereof
US11149088B2 (en) 2016-04-15 2021-10-19 Bioatla, Inc. Anti-Axl antibodies, antibody fragments and their immunoconjugates and uses thereof
US11879011B2 (en) 2016-05-13 2024-01-23 Bioatla, Inc. Anti-ROR2 antibodies, antibody fragments, their immunoconjucates and uses thereof
WO2017197234A1 (en) 2016-05-13 2017-11-16 Bioatla, Llc Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
US11254742B2 (en) 2016-05-13 2022-02-22 Bioatla, Inc. Anti-Ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
EP4122958A1 (en) 2016-05-13 2023-01-25 BioAtla, Inc. Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
US11440969B2 (en) 2016-07-08 2022-09-13 Genentech, Inc. Use of human epididymis protein 4 (HE4) for assessing responsiveness of MUC 16-positive cancer treatment
WO2018009811A1 (en) 2016-07-08 2018-01-11 Genentech, Inc. Use of human epididymis protein 4 (he4) for assessing responsiveness of muc 16-positive cancer treatment
WO2018085513A1 (en) 2016-11-04 2018-05-11 Genentech, Inc. Treatment of her2-positive breast cancer
WO2018091724A1 (en) 2016-11-21 2018-05-24 Cureab Gmbh Anti-gp73 antibodies and immunoconjugates
EP4015532A1 (en) 2016-11-21 2022-06-22 cureab GmbH Anti-gp73 antibodies and immunoconjugates
US10736976B2 (en) 2016-12-01 2020-08-11 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-PD-L1 antibodies for immuno-PET imaging
US11273155B2 (en) 2016-12-12 2022-03-15 Daiichi Sankyo Company, Limited Combination of antibody-drug conjugate and immune checkpoint inhibitor
WO2018125589A1 (en) 2016-12-28 2018-07-05 Genentech, Inc. Treatment of advanced her2 expressing cancer
US11654105B2 (en) 2017-01-17 2023-05-23 Genentech, Inc. Subcutaneous HER2 antibody formulations
WO2018136412A2 (en) 2017-01-17 2018-07-26 Genentech, Inc. Subcutaneous her2 antibody formulations
EP3868404A1 (en) 2017-01-17 2021-08-25 F. Hoffmann-La Roche AG Subcutaneous her2 antibody formulations
US10849849B2 (en) 2017-01-17 2020-12-01 Genentech Inc. Subcutaneous HER2 antibody formulations
WO2018160654A2 (en) 2017-03-02 2018-09-07 Genentech, Inc. Adjuvant treatment of her2-positive breast cancer
US11077189B2 (en) 2017-03-02 2021-08-03 Genentech Inc. Adjuvant treatment of HER2-positive breast cancer
US11638756B2 (en) 2017-03-02 2023-05-02 Genentech, Inc. Adjuvant treatment of HER2-positive breast cancer
WO2018200505A1 (en) 2017-04-24 2018-11-01 Genentech, Inc. Erbb2/her2 mutations in the transmbrane or juxtamembrane domain
US11512114B2 (en) 2017-05-09 2022-11-29 Cyano Biotech Gmbh Modified microcystins and nodularins
CN110730787A (zh) * 2017-05-09 2020-01-24 赛安诺生物技术有限责任公司 修饰的微囊藻毒素和节球藻毒素
US11077202B2 (en) 2017-05-15 2021-08-03 Daiichi Sankyo Company, Limited Anti-CDH6 antibody and anti-CDH6 antibody-drug conjugate
US11446386B2 (en) 2017-05-15 2022-09-20 Daiichi Sankyo Company, Limited Anti-CDH6 antibody and method of producing an anti-CDH6 antibody-drug conjugate
US11318212B2 (en) 2017-08-31 2022-05-03 Daiichi Sankyo Company, Limited Method for producing antibody-drug conjugate
US11945882B2 (en) 2017-08-31 2024-04-02 Daiichi Sankyo Company, Limited Method for producing antibody-drug conjugate
US11230601B2 (en) 2017-10-10 2022-01-25 Tilos Therapeutics, Inc. Methods of using anti-lap antibodies
WO2019118411A3 (en) * 2017-12-11 2020-03-26 Triphase Accelerator U.S. Corporation Anti-cd22 antibody-maytansine conjugates, combinations, and methods of use thereof
WO2019118937A1 (en) 2017-12-15 2019-06-20 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
WO2019175125A1 (en) 2018-03-13 2019-09-19 F. Hoffmann-La Roche Ag Combination therapy with targeted 4-1bb (cd137) agonists
US11872289B2 (en) 2018-05-18 2024-01-16 Daiichi Sankyo Co., Ltd. Anti-MUC1 antibody-drug conjugate
US11130802B2 (en) 2018-10-10 2021-09-28 Tilos Therapeutics, Inc. Anti-lap antibody variants
WO2020081119A1 (en) 2018-10-15 2020-04-23 Genentech, Inc. Methods of treating residual breast cancer with trastuzumab emtansine
EP4227320A2 (en) 2018-10-15 2023-08-16 F. Hoffmann-La Roche AG Methods of treating residual breast cancer with trastuzumab emtansine
WO2020117257A1 (en) 2018-12-06 2020-06-11 Genentech, Inc. Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
WO2020232169A1 (en) 2019-05-14 2020-11-19 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat follicular lymphoma
WO2021076196A1 (en) 2019-10-18 2021-04-22 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2021217051A1 (en) 2020-04-24 2021-10-28 Genentech, Inc. Methods of using anti-cd79b immunoconjugates
WO2022047243A1 (en) 2020-08-27 2022-03-03 Enosi Life Sciences Corp. Methods and compositions to treat autoimmune diseases and cancer
WO2022241446A1 (en) 2021-05-12 2022-11-17 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023019092A1 (en) 2021-08-07 2023-02-16 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma

Also Published As

Publication number Publication date
CA2567520A1 (en) 2005-12-15
BRPI0510883A (pt) 2007-12-26
KR20120064120A (ko) 2012-06-18
NZ579482A (en) 2011-02-25
US8652479B2 (en) 2014-02-18
RU2006147264A (ru) 2008-07-20
US20080171040A1 (en) 2008-07-17
NO20066075L (no) 2007-02-28
CN114053429A (zh) 2022-02-18
KR20070037575A (ko) 2007-04-05
US8142784B2 (en) 2012-03-27
KR101200133B1 (ko) 2012-11-13
RU2404810C9 (ru) 2015-06-20
JP2012036202A (ja) 2012-02-23
EP2286844A2 (en) 2011-02-23
AU2005249490A1 (en) 2005-12-15
CN102973947A (zh) 2013-03-20
EP2286844A3 (en) 2012-08-22
MXPA06014065A (es) 2007-01-31
BRPI0510883B1 (pt) 2021-04-13
JP5234734B2 (ja) 2013-07-10
EP1753463A2 (en) 2007-02-21
NZ551180A (en) 2009-10-30
IL179140A (en) 2016-12-29
US20140128580A1 (en) 2014-05-08
RU2404810C2 (ru) 2010-11-27
JP2008501029A (ja) 2008-01-17
AU2005249490B2 (en) 2010-07-29
WO2005117986A3 (en) 2006-06-15
IL179140A0 (en) 2007-03-08
US20090202536A1 (en) 2009-08-13
BRPI0510883B8 (pt) 2021-05-25
WO2005117986A2 (en) 2005-12-15

Similar Documents

Publication Publication Date Title
US8142784B2 (en) Antibody-drug conjugates and methods
JP2008501029A5 (zh)
US20090226465A1 (en) Macrocyclic depsipeptide antibody-drug conjugates and methods
ZA200610159B (en) Antibody-drug conjugates and methods
US20180000962A1 (en) Cysteine engineered antibodies and conjugates

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENENTECH, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:EBENS, ALLEN J. JR.;JACOBSON, FREDERIC S.;POLAKIS, PAUL;AND OTHERS;REEL/FRAME:016661/0572;SIGNING DATES FROM 20050718 TO 20050812

STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION