US20040013607A1 - Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells - Google Patents

Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells Download PDF

Info

Publication number
US20040013607A1
US20040013607A1 US10/446,689 US44668903A US2004013607A1 US 20040013607 A1 US20040013607 A1 US 20040013607A1 US 44668903 A US44668903 A US 44668903A US 2004013607 A1 US2004013607 A1 US 2004013607A1
Authority
US
United States
Prior art keywords
antibody
antibodies
chimeric
humanized
cells
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
US10/446,689
Inventor
Shui-on Leung
Hans Hansen
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.)
Immunomedics Inc
Original Assignee
Immunomedics 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=23112129&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040013607(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Immunomedics Inc filed Critical Immunomedics Inc
Priority to US10/446,689 priority Critical patent/US20040013607A1/en
Publication of US20040013607A1 publication Critical patent/US20040013607A1/en
Priority to US10/974,678 priority patent/US20050106108A1/en
Priority to US11/676,466 priority patent/US20070172920A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • 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
    • C07K16/3061Blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6867Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of a blood cancer
    • 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
    • A61K47/6877Medicinal 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 the antibody being an immunoglobulin containing regions, domains or residues from different species
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention relates generally to immunoconjugates for diagnostic and therapeutic uses in cancer.
  • the invention relates to recombinantly produced chimeric and humanized monoclonal antibodies directed against B-cell lymphoma and leukemia cells, which antibodies can be covalently conjugated to a diagnostic or therapeutic reagent without loss of antibody binding and internalization function and with reduced production of human anti-mouse antibodies.
  • Non-Hodgkins lymphoma (NHL) and chronic lymphocytic leukemia are B-cell malignancies that remain important contributors to cancer mortality.
  • the response of these malignancies to various forms of treatment is mixed. They respond reasonably well to chemotherapy, and, in cases where adequate clinical staging of NHL is possible, as for patients with localized disease, satisfactory treatment may be provided using field radiation therapy (Hall et al., Radiology for the Radiologist, Lippincott, Philadelphia, 1989, pp 365-376).
  • field radiation therapy Hall et al., Radiology for the Radiologist, Lippincott, Philadelphia, 1989, pp 365-376.
  • the toxic side effects associated with chemotherapy and the toxicity to the hematopoietic system from local, as well as whole body, radiotherapy limits the use of these therapeutic methods. About one-half of the patients die from the disease (Posner et al., Blood, 61: 705 (1983)).
  • LL-2 (EPB2) is a highly specific anti-B-cell lymphoma and anti-lymphocytic leukemia cell murine monoclonal antibody (mAb) that is rapidly internalized by such cells and that can overcome some of the aforementioned difficulties (Shih et al., Int. J. Cancer, 56: 538 (1994)).
  • LL2 which is of the IgG2a antibody type, was developed using the Raji B-lymphoma cell line as the source of antigen (Pawlak-Byczkowska et al., Cancer Res., 49: 4568 (1989)).
  • Murine LL2 (mLL2) is known to react with an epitope of CD22 (Belisle et al., Proc Amer. Assn. Clin. Res., 34: A2873 (1993)). CD22 molecules are expressed in the cytoplasm of progenitor and early pre-B cells, and appear in the cell surface of mature B-cells.
  • mLL2 was shown to react with 50 of 51 B-cell lymphomas tested.
  • mLL2 is a highly sensitive means of detecting B-cell lymphoma cell in vivo, as determined by a radioimmunodetection method (Murthy et al., Eur. J. Nucl. Med., 19: 394 (1992)).
  • the Fab′ fragment of mLL2 labeled with 99m Tc localized to 63 of 65 known lesions in Phase II trial patients with B-cell lymphoma (Mills et al., Proc. Amer. Assn. Cancer Res., 14: A2857 (1993)).
  • mLL2 Fab′ conjugated to the exotoxin PE38KDEL induced complete remissions of measurable human lymphoma xenografts (CA-46) growing in nude mice (Kreitman et al., Cancer Res., 53: 819 (1993)).
  • fragments of mLL2 such as F(ab′)2 and Fab′, partially alleviate/circumvent these problems of immunogenicity, there are circumstances in which whole IgG is more desirable, such as when induction of cellular immunity is intended for therapy, or where an antibody with enhanced survival time is required.
  • mLL2 IgG antibody In order to maximize the value of the mLL2 IgG antibody as a therapeutic or diagnostic modality and increase its utility in multiple and continuous administration modalities, it is an object of this invention to produce a mouse/human chimeric mAb (cLL2) and a humanized mAb (hLL2) related to mLL2 that retain the antigen-binding specificity of mLL2, but that elicit reduced HAMA in a subject receiving same.
  • cLL2 mouse/human chimeric mAb
  • hLL2 humanized mAb
  • CDR complementarity determining regions
  • It is also an object of this invention provide conjugates of the hLL2 and cLL2 mAbs containing therapeutic or diagnostic modalities.
  • a cLL2 mAb related to mLL2 mAb in which the murine light (VK) and heavy (VH) chain variable regions are joined to the human constant light (kappa) and heavy (IgG 1 ) chains.
  • This chimeric mAb retains the B-lymphoma and leukemia cell targeting and internalization properties of the parental mLL2.
  • a hLL2 mAb related to mLL2 mAb in which the complementarity-determining regions (CDRs) of the light and heavy chains of the mLL2 mAb are joined to the framework (FR) sequence of human VK and VH regions, respectively, and subsequently to the human kappa and IgG 1 constant region domains, respectively.
  • This humanized antibody retains the B-lymphoma and leukemia cell targeting and internalization characteristics of the parental mLL2 mAb, and can exhibit a lowered HAMA reaction.
  • isolated polynucleotides comprising DNA sequences encoding the amino acid sequences of the variable light and heavy chains, respectively, of the hLL2 and cLL2 mAbs.
  • amino acid sequences of the CDRs of the VK and VH chains are provided.
  • conjugates in which the hLL2 or cLL2 mAb is covalently bonded to a diagnostic or therapeutic reagent.
  • FIG. 1 is a comparison of the murine with the humanized LL2 VK (FIG. 1A) and VH (FIG. 1B) domains. Only hFR sequences (designated as REIHuVK and EUHuVH) different than mFR sequences (designated as murine) are shown, and designated by asterisks. More residues in these positions were retained in the humanized structure. CDRs are boxed. FR residues showing CDR contacts by computer modeling are underlined.
  • FIG. 2 shows vicinal relationships of the LL2 CDRs to their framework regions (FRs).
  • FRs framework regions
  • FIG. 3 shows the light chain (FIG. 3A) staging (VKpBR) and mammalian expression (pKH) vectors, and the heavy chain (FIG. 3B) staging (VHpBS) and mammalian expression (pG1g) vectors.
  • FIG. 4 shows the double-stranded DNA and amino acid sequences of the LL2 VK domain (FIG. 4A) and the LL2 VH domain (FIG. 4B). Amino acid sequences encoded by the corresponding DNA sequences are given as one letter codes. CDR amino acid sequences are boxed. The Asn-glycosylation site located in FR1 of LL2VK (FIG. 4A) is shown as the underlined NVT sequence.
  • FIG. 5A shows the double stranded DNA and corresponding amino acid residues of the hLL2 VK domain. CDR amino acid sequences are boxed. The corresponding data for the VH domain is shown in FIG. 5B.
  • FIG. 6 is a schematic diagram representation of the PCR/gene synthesis of the humanized VH region and the subcloning into the staging vector, VHpBS.
  • FIG. 7 shows SDS-PAGE analysis of mLL2 and cLL2 antibodies under non-reducing (lanes 6-8) and reducing (lanes 3-5, light and heavy chains) conditions. Lanes 3 and 6 include a control antibody.
  • FIG. 8 shows SDS-PAGE analysis of different versions of cLL2 and hLL2 antibodies under reducing (lanes 3-5) and non-reducing (lanes 6-8) conditions.
  • FIG. 9 shows SDS-PAGE analysis on mix-and-match cLL2 and hLL2 antibodies under reducing (lanes 3-6) and non-reducing (lanes 7-10) conditions.
  • cLL2 serves as the control.
  • FIG. 10 shows the results of a comparative Raji cell competitive antibody binding assay involving mLL2 and cLL2 antibodies competing for binding to cells against tracer radiolabeled mLL2.
  • FIG. 11 shows the results of a comparative Raji cell competitive antibody binding assay in which mixed humanized/chimeric LL2s were compared to cLL2 (FIG. 11A), and two versions of hLL2 compared to cLL2 (FIG. 11B).
  • FIG. 12 shows a comparison of antibody internalization:surface binding ratios as a function of time for cLL2, cLL2 (Q to V mutagenesis), hLL2 and mLL2 antibodies.
  • FIG. 13 shows an SDS-PAGE analysis of mLL2 and cLL2 after deglycosylation by endoglycosidase F.
  • FIG. 14 shows the effect of deglycosylation of mLL2 on its binding affinity to Raji cells.
  • cDNAs encoding the VL and VH regions of the mLL2 mAb have been isolated and separately recombinantly subcloned into mammalian expression vectors containing the genes encoding kappa and IgG 1 constant regions, respectively, of human antibodies. Cotransfection of mammalian cells with these two recombinant DNAs expressed a cLL2 mAb that, like the parent mLL2 mAb, bound avidly to, and was rapidly internalized by, B-lymphoma cells.
  • the CDRs of the VK and VH DNAs have been similarly recombinantly linked to the framework (FR) sequences of the human VK and VH regions, respectively, which are subsequently linked, respectively, to the human kappa and IgG 1 constant regions, so as to express in mammalian cells as described above hLL2.
  • cLL2 or “cLL2 mAb” are intended to refer to the chimeric monoclonal antibody constructed by joining or subcloning the murine VK and VH regions to the human constant light and heavy chains, respectively.
  • hLL2 or “hLL2 mAb” are intended to refer to the humanization of the chimeric monoclonal antibody by replacing the murine FR sequences in cLL2 with that of human framework regions.
  • Covalent conjugates between cLL2 and hLL2 mAbs and a diagnostic or chemotherapeutic reagent, formulated in pharmaceutically acceptable vehicles can be prepared that have the advantages, compared to prior art antibody conjugates, of B-cell lymphoma-specific and leukemia cell-specific targeting, rapid internalization into target cells, rapid liberation of the diagnostic or chemotherapeutic reagent intracellularly (thereby increasing effectiveness of the reagent), and a potential reduction of the HAMA response in the human patient.
  • VK-appended carbohydrate moiety of the cLL2 mAb is shown herein not to be involved in binding to B-lymphoma cells, it is preferred to use conjugates in which the reagent is bound to the antibody through such carbohydrate moieties, such as through oxidized carbohydrate derivatives.
  • Methods for the production of such conjugates and their use in diagnostics and therapeutics are provided, for example. in Shih et al., U.S. Pat. No. 5,057,313, Shih et al., Int. J. Cancer 41: 832 (1988), and copending, commonly owned Hansen el al., U.S. Ser. No. 08/162,912, the contents of which are incorporated herein by reference.
  • Direct linkage of the reagent to oxidized carbohydrate without the use of a polymeric carrier is described in McKearn et al., U.S. Pat. No. 5,156,840, which is also incorporated by reference.
  • a wide variety of diagnostic and therapeutic reagents can be advantageously conjugated to the antibodies of the invention. These include: chemotherapeutic drugs such as doxorubicin, methotrexate, taxol, and the like; chelators, such as DTPA, to which detectable labels such as fluorescent molecules or cytotoxic agents such as heavy metals or radionuclides can be complexed; and toxins such as Pseudomonas exotoxin, and the like.
  • chemotherapeutic drugs such as doxorubicin, methotrexate, taxol, and the like
  • chelators such as DTPA, to which detectable labels such as fluorescent molecules or cytotoxic agents such as heavy metals or radionuclides can be complexed
  • toxins such as Pseudomonas exotoxin, and the like.
  • Cell lines and culture media used in the present invention include LL2 (EPB-2) hybridoma cells (Pawlak-Byczkowska et al. 1989 above), Sp2/0-Ag14 myeloma cells (ATCC, Rockville, Md.) and Raji cells. These cells are preferably cultured in Dulbecco's modified Eagle's Medium (DMEM) supplemented with 10% FCS (Gibco/BRL, Gaithersburg, Mass.), 2 mM L-glutamine and 75 ⁇ g/ml gentamicin, (complete DMEM).
  • DMEM Dulbecco's modified Eagle's Medium
  • Transfectomas are grown in Hybridoma Serum Free Medium, HSFM, (Gibco/BRL, Gaithersburg, Mass.) containing 10% of FCS and 75 ⁇ g/ml gentamicin (complete HSFM) or, where indicated, in HSFM containing only antibiotics. Selection of the transfectomas may be carried out in complete HSFM containing 500 ⁇ g/ml of hygromycin (Calbiochem, San Diego, Calif.). All cell lines are preferably maintained at 37° C. in 5% CO 2 .
  • antibody variable domains can be modeled by computer modeling (see, for example, Dion, in Goldenberg et al. eds., Cancer Therapy With Radiolabeled Antibodies, CRC Press, Boca Raton, Fla., 1994), which is incorporated by reference.
  • 3-D structure for both the mLL22 and hLL2 mAbs are best modeled by homology.
  • the high frequency of residue identities (75.0 to 92.3%) between the deduced primary sequences of mLL2 light chain FR regions and human REI (VK) facilitates this approach because of the availability of crystallographic data from the Protein Data Bank (PDR Code 1REI, Bernstein et al., J. Mol. Biol.
  • antibody EU (VH) sequences can be selected as the computer counterparts for FR1 to FR3 of the mLL2 heavy chain; FR4 was based on NEWM.
  • NEWM structural data PDR Code 3FAB
  • amino acid side groups can be replaced to correspond to mLL2 or EU (hLL2) as needed.
  • the CDR of the light chain can be modeled from the corresponding sequence of 1MCP (L1 and L2) and 1REI (L3).
  • H1 and H2 can be based on 2HFL and 1MCP, respectively, while H3 can be modeled de novo. Wherever possible, side group replacements should be performed so as to maintain the torsion angle between C ⁇ and C ⁇ . Energy minimization may be accomplished by the AMBER forcefield (Weiner et al, J. Amer. Chem. Soc. 106: 765 (1984) using the convergent method. Potentially critical FR-CDR interactions can be determined by initially modeling the light and heavy variable chains of mLL2. All FR residues within a 4.5 ⁇ radius of all atoms within each CDR can thereby be identified and retained in the final design model of hLL2.
  • CDR engrafting can be accomplished by gene synthesis using long synthetic DNA oligonucleotides as templates and short oligonucleotides as primers in a PCR reaction.
  • the DNA encoding the VK or VH domain will be approximately 350 bp long.
  • a unique AvrII site can be introduced while maintaining the originally designed amino acid sequence (FIG. 4B).
  • Two long non-overlapping single-stranded DNA oligonucleotides ⁇ 150 bp upstream and downstream of the AvrII site (see, for example, oligo A and oligo B, Example 3 below) can be generated by automated DNA oligonucleotide synthesizer (Cyclone Plus DNA Synthesizer, Milligen-Biosearch).
  • oligonucleotides such as oligos A and B may be expected to be low, they can be amplified by two pairs of flanking oligonucleotides (oligo Seq. ID Nos. 7 and 8 for oligo A; oligo Seq. ID Nos. 9 and 10 for oligo B, Example 3) in a PCR reaction.
  • the primers can be designed with the necessary restriction sites to facilitate subsequent subcloning.
  • Primers for oligo A and for oligo B should contain overlapping sequence at the AvrII site so that the resultant PCR product for oligo A and B, respectively, can be joined in-frame at the AvrII site to form a full length DNA sequence (ca 350 bp) encoding the hLL2 VH domain.
  • the ligation of the PCR products for oligo A (restriction-digested with PstI and AvrII) and B (restriction-digested with AvrII and BstEII) at the AvrII site and their subcloning into the PstII/BstEII sites of the staging vector, VHpBS, can be completed in a single three-fragment-ligation step (See, for example, Example 3).
  • the subcloning of the correct sequence into VHpBS can be first analyzed by restriction digestion analysis and subsequently confirmed by sequencing reaction according to Sanger et al., Proc. Natl. Acad. Sci. USA 74: 5463 (1977).
  • the HindIII/BamHI fragment containing the Ig promoter, leader sequence and the hLL2 VH sequence can be excised from the staging vector and subcloned to the corresponding sites in a pSVgpt-based vector, pG1g, which contains the genomic sequence of the human IgG constant region, an Ig enhancer and a gpt selection marker, forming the final expression vector, hLL2pG1g. Similar strategies can be employed for the construction of the hLL2 VK sequence.
  • the restriction site chosen for the ligation of the PCR products for the long oligonucloetides can be NruI in this case.
  • the DNA sequence containing the Ig promoter, leader sequence and the hLL2 VK sequence can be excised from the staging vector VKpBR by treatment with BamHI/HindIII, and can be subcloned into the corresponding sites of a pSVhyg-based vector, pKh, which contains the genomic sequence of human kappa chain constant regions, a hygromycin selection marker, an Ig and a kappa enhancer, forming the final expression vector, hLL2pKh.
  • VH and VK chains of mLL2 can be obtained by PCR cloning using DNA products and primers. Orlandi et al., infra, and Leung et al., infra.
  • the VK PCR primers may be subcloned into a pBR327 based staging vector (VKpBR) as described above.
  • VKpBR pBR327 based staging vector
  • the VH PCR products may be subcloned into a similar pBluescript-based staging vector (VHpBS) as described above.
  • the fragments containing the VK and VH sequences, along with the promoter and signal peptide sequences, can be excised from the staging vectors using HindIII and BamHI restriction endonucleases.
  • the VK fragments (about 600 bp) can be subcloned into a mammalian expression vector (for example, pKh) conventionally.
  • pKh is a pSVhyg-based expression vector containing the genomic sequence of the human kappa constant region. an Ig enhancer, a kappa enhancer and the hygromucin-resistant gene.
  • the about 800 bp VH fragments can be subcloned into pG1g, a pSVgpt-based expression vector carrying the genomic sequence of the human IgG1 constant region, an Ig enhancer and the xanthine-guanine phosphoribosyl transferase (gpt) gene.
  • the two plasmids may be transfected into mammalian expression cells, such as Sp2/0-Ag14 cells, by electroporation and selected for hygromycin resistance. Colonies surviving selection are expanded, and supernatant fluids monitored for production of cLL2 mAb by an ELISA method.
  • a transfection efficiency of about 1-10 ⁇ 10 6 cells is desirable.
  • An antibody expression level of between 0.10 and 2.5 ⁇ g/ml can be expected with this system.
  • RNA isolation, cDNA synthesis, and amplification can be carried out as follows.
  • Total cell RNA can be prepared from a LL2 hybridoma cell line, using a total of about 10 7 cells, according to Sambrook et al., ( Molecular Cloning: A Laboratory Manual, Second ed., Cold Spring Harbor Press, 1989), which is incorporated by reference.
  • First strand cDNA can be reverse transcribed from total RNA conventionally, such as by using the SuperScript preamplification system (Gibco/BRL., Gaithersburg, Md.).
  • RNAs in a reaction volume of 20 ⁇ l, 50 ng of random primers can be annealed to 5 ⁇ g of RNAs in the presence of 2 ⁇ l of 10 ⁇ synthesis buffer [200 mM Tris-HCl (pH 8.4), 500 mM KCl, 25 mM MgCl 2 , 1 mg/ml BSA], 1 ⁇ l of 10 mM dNTP mix, 2 ⁇ l of 0.1 M DTT, and 200 units of SuperScript reverse transcriptase.
  • the elongation step is initially allowed to proceed at room temperature for 10 min followed by incubation at 42° C. for 50 min.
  • the reaction can be terminated by heating the reaction mixture at 90° C. for 5 min.
  • VK and VH sequences for cLL2 or hLL2 can amplified by PCR as described by Orlandi et al., (Proc. Natl. Acad. Sci., USA, 86: 3833 (1989)) which is incorporated by reference.
  • VK sequences may be amplified using the primers CK3BH and VK5-3 (Leung et al., BioTechniques, 15: 286 (1993), which is incorporated by reference), while VH sequences can be amplified using the primer CH1B which anneals to the CHI region of murine IgG, and VHIBACK (Orlandi et al., 1989 above).
  • the PCR reaction mixtures containing 10 ⁇ l of the first strand cDNA product, 9 ⁇ l of 10 ⁇ PCR buffer [500 mM KCl, 100 mM Tris-HCl (pH 8.3), 15 mM MgCl2, and 0.01% (w/v) gelatin] can be subjected to 30 cycles of PCR.
  • Each PCR cycle preferably consists of denaturation at 94° C. for 1 min, annealing at 50° C. for 1.5 min, and polymerization at 72° C. for 1.5 min.
  • Amplified VK and VH fragments can be purified on 2% agarose (BioRad, Richmond, Calif.).
  • PCR products for VK can be subcloned into a staging vector, such as a pBR327-based staging vector VKpBR that contains an Ig promoter, a signal peptide sequence and convenient restriction sites to facilitate in-frame ligation of the VK PCR products.
  • PCR products for VH can be subcloned into a similar staging vector, such as the pBluescript-based VHpBS. Individual clones containing the respective PCR products may be sequenced by, for example, the method of Sanger et al., Proc. Natl. Acad. Sci., USA, 74: 5463 (1977) which is incorporated by reference.
  • DNA sequences described herein are to be taken as including all alleles, mutants and variants thereof, whether occurring naturally or induced.
  • the two plasmids can be co-transfected into an appropriate cell, e.g., myeloma Sp2/0-Ag14, colonies selected for hygromycin resistance, and supernatant fluids monitored for production of cLL2 or hLL2 antibodies by, for example, an ELISA assay, as described below.
  • an appropriate cell e.g., myeloma Sp2/0-Ag14, colonies selected for hygromycin resistance, and supernatant fluids monitored for production of cLL2 or hLL2 antibodies by, for example, an ELISA assay, as described below.
  • Transfection, and assay for antibody secreting clones by ELISA can be carried out as follows. About 10 ⁇ g of hLL2pKh (light chain expression vector) and 20 ⁇ g of hLL2pG1g (heavy chain expression vector) can be used for the transfection of 5 ⁇ 10 6 SP2/0 myeloma cells by electroporation (BioRad, Richmond, Calif.) according to Co et al., J. Immunol., 148: 1149 (1992) which is incorporated by reference. Following transfection, cells may be grown in 96-well microtiter plates in complete HSFM medium (GIBCO, Gaithersburg, Md.) at 37° C., 5% CO 2 .
  • the selection process can be initiated after two days by the addition of hygromycin selection medium (Calbiochem, San Diego, Calif.) at a final concentration of 500 ⁇ g/ml of hygromycin. Colonies typically emerge 2-3 weeks post-electroporation. The cultures can then be expanded for further analysis.
  • hygromycin selection medium Calbiochem, San Diego, Calif.
  • Transfectoma clones that are positive for the secretion of chimeric or humanized heavy chain can be identified by ELISA assay. Briefly, supernatant samples (100 ⁇ l) from transfectoma cultures are added in triplicate to ELISA microtiter plates precoated with goat anti-human (GAH)-IgG, F(ab′) 2 fragment-specific antibody (Jackson ImmunoResearch, West Grove, Pa.). Plates are incubated for 1 h at room temperature. Unbound proteins are removed by washing three times with wash buffer (PBS containing 0.05% polysorbate 20).
  • wash buffer PBS containing 0.05% polysorbate 20
  • HRP horseradish peroxidase conjugated GAH-IgG, Fc fragment-specific antibodies
  • HRA horseradish peroxidase conjugated GAH-IgG, Fc fragment-specific antibodies
  • OPD orthophenylene-diamine
  • reaction is stopped by the addition of 50 ⁇ l of 4 N HCl solution into each well before measuring absorbance at 490 nm in an automated ELISA reader (Bio-Tek instruments, Winooski, Vt.). Bound chimeric antibodies are than determined relative to an irrelevant chimeric antibody standard (obtainable from Scotgen, Ltd., Edinburg, Scotland).
  • Antibodies can be isolated from cell culture media as follows. Transfectoma cultures are adapted to serum-free medium. For production of chimeric antibody, cells are grown as a 500 ml culture in roller bottles using HSFM. Cultures are centrifuged and the supernatant filtered through a 0.2 micron membrane. The filtered medium is passed through a protein A column (1 ⁇ 3 cm) at a flow rate of 1 ml/min. The resin is then washed with about 10 column volumes of PBS and protein A-bound antibody is eluted from the column with 0.1 M glycine buffer (pH 3.5) containing 10 mM EDTA.
  • 0.1 M glycine buffer pH 3.5
  • Fractions of 1.0 ml are collected in tubes containing 10 ⁇ l of 3 M Tris (pH 8.6), and protein concentrations determined from the absorbancies at 280/260 nm. Peak fractions are pooled, dialyzed against PBS, and the antibody concentrated, for example, with the Centricon 30 (Amicon, Beverly, Mass.). The antibody concentration is determined by ELISA, as before, and its concentration adjusted to about 1 mg/ml using PBS. Sodium azide, 0.01% (w/v), is conveniently added to the sample as preservative.
  • Comparative binding affinities of the mLL2, cLL2 and hcLL2 antibodies thus isolated may be determined by direct radioimmunoassay.
  • mLL2 can be labeled with 131 I or 125 I using the chloramine T method (see, for example, Greenwood et al., Biochem. J., 89: 123 (1963) which is incorporated by reference).
  • the specific activity of the iodinated antibody is typically adjusted to about 10 ⁇ Ci/ ⁇ g.
  • Unlabeled and labeled antibodies are diluted to the appropriate concentrations using reaction medium (HSFM supplemented with 1% horse serum and 100 ⁇ g/ml gentamicin).
  • the appropriate concentrations of both labeled and unlabeled antibodies are added together to the reaction tubes in a total volume of 100 ⁇ l.
  • a culture of Raji cells is sampled and the cell concentration determined. The culture is centrifuged and the collected cells washed once in reaction medium followed by resuspension in reaction medium to a final concentration of about 10 7 cells/ml. All procedures are carried out in the cold at 4° C.
  • the cell suspension, 100 ⁇ l, is added to the reaction tubes.
  • the reaction is carried out at 4° C. for 2 h with periodic gentle shaking of the reaction tubes to resuspend the cells. Following the reaction period, 5 ml of wash buffer (PBS containing 1% BSA) is added to each tube.
  • PBS containing 1% BSA wash buffer
  • the suspension is centrifuged and the cell pellet washed a second time with another 5 ml of wash buffer. Following centrifugation, the amount of remaining radioactivity remaining in the cell pellet is determined in a gamma counter (Minaxi, Packard Instruments, Sterling, Va.).
  • the Raji cell surface antigen binding affinities of mix-and-match and fully humanized antibodies can be compared to that of cLL2 using various concentrations of mLL2 F(ab′) 2 fragments devoid of the Fc portion as competitors, as evaluated by flow cytometry assay. Residual surface-bound LL2 antibodies carrying the human Fe portions (cLL2 and mix-and-match LL2) can be detected by a FITC-labeled anti-human Fc specific antibody in a flow cytometry assay. Where mix-and-match LL2 antibodies exhibit antigen-binding affinities similar to that of cLL2, it can be concluded that the original designs for the humanization of both the light and heavy chains retain the mLL2 immunoreactivity.
  • the rate of antibody internalization can be determined according to Opresko et al., ( J. Biol. Chem., 262: 4116 (1987)), using radioiodinated antibody as tracer. Briefly, radiolabeled antibodies (1 ⁇ 10 4 cpm) are incubated with the Raji cells (1 ⁇ 10 6 cells/ml) at 4° C. for 2 h in 0.5 ml of DMEM medium containing 1% human serum. Following the reaction interval, non-specifically bound antibodies are removed by washing three times with 0.5 ml of DMEM medium. To each of the reaction tubes 0.5 ml of DMEM medium is added and the suspension incubated at 37° C. for the determination of internalization.
  • triplicates of cells are removed and chilled immediately in an ice bath to stop further internalization.
  • Cells are centrifuged at 1000 ⁇ g for 5 min at 4° C. The supernatant is removed and counted for radioactivity.
  • the surface-bound radioactivity is removed by treatment with 1 ml 0.1 M acetate/0.1 M glycine buffer at pH 3.0 for 8 min. in the cold. Radioactivity removed by the acid treatment, and that remaining associated with the cells, are determined.
  • the ratio of the CPM internalization /CPM surface is plotted versus time to determine the rate of internalization from the slope.
  • Asn-linked glycosylation sites may be introduced into antibodies using conventional site-directed oligonucleotide mutagenesis reactions. For example, to introduce an Asn in position 18 of a kappa protein, one may alter codon 18 from AGG to AAC. To accomplish this, a single stranded DNA template containing the antibody light chain sequence is prepared from a suitable strain of E. coli (e.g., dut ⁇ ung ⁇ ) in order to obtain a DNA molecule containing a small number of uracils in place of thymidine. Such a DNA template can be obtained by M13 cloning or by in vitro transcription using a SP6 promoter.
  • E. coli e.g., dut ⁇ ung ⁇
  • oligonucleotide containing the mutated sequence is synthesized conventionally, annealed to the single-stranded template and the product treated with T4 DNA polymerase and T4 DNA ligase to produce a double-stranded DNA molecule. Transformation of wild type E. coli (dut + ung + ) cells with the double-stranded DNA provides an efficient recovery of mutated DNA.
  • an Asn-linked glycosylation site can be introduced into an antibody light chain using an oligonucleotide containing the desired mutation as the primer and DNA clones of the variable regions for the, VL chain, or by using RNA from cells that produce the antibody of interest as a template.
  • RNA from cells that produce the antibody of interest as a template.
  • Site-directed mutagenesis can be performed, for example, using the TRANSFORMERTM kit (Clontech, Palo Alto, Calif.) according to the manufacturer's instructions.
  • a glycosylation site can be introduced by synthesizing an antibody chain with mutually priming oligonucleotides, one such containing the desired mutation. See, for example, Uhlmann, Gene 71: 29 (1988); Wosnick et al., Gene 60: 115 (1988); Ausubel et al., above, which are incorporated by reference.
  • V murine variable
  • FR region framework
  • the REI and EU FRs were selected as the human frameworks onto which the CDRs for LL2 VK and VH were grafted, respectively.
  • the FR4 sequence of NEWM was used to replace the EU FR4 sequence for the humanization of LL2 heavy chain.
  • murine FR residues having potential CDR contacts were retained in the design of the humanized FR sequences (FIG. 1).
  • variable regions for both heavy (VH) and light (VK) chains of mLL2 (IgG2a) were obtained by PCR cloning using DNA primers as described in general above and in greater detail in Example 3, below. As PCR is prone to mutations, the variable region sequence of multiple individual clones for either the heavy or light chains was determined for six clones and confirmed to be identical prior to use for the construction of the chimeric antibody.
  • VKpBR pBR327-based staging vector
  • VHpBS VHpBS
  • oligo A 149-mer
  • oligo B 140-mer
  • Oligo A (Sequence ID No. 7 below) represents the minus strand of the hLL2 VH domain complementary to nt 24 to 172.
  • Oligo B (Sequence ID No. 8 below) represents the minus strand of the hLL2 VH domain complementary to nt 180 to 320.
  • Sequence ID No. 8 5′-CCC CAG TAG AAC GTA ATA TCC CTT GCA CAA AAA TAA AAT GCC GTG TCC TCA GAC CTC AGG CTG CTC AGC TCC ATG TAG GCT GTA TTG GTG GAT TCG TCT GCA GTT ATT GTG GCC TTG TCC TTG AAG TTC TGA TT-3′
  • Oligos A and B were cleaved from the support and deprotected by treatment with concentrated ammonium hydroxide. After the samples were vacuum-dried (SpeedVac, Savant, Farmingdale, N.Y.) and resuspended in 100 ⁇ l of water, incomplete oligomers (less than 100-mer) were removed by centrifugation through a CHROMOSPIN-100TM column (Clonetech, Palo Alto, Calif.) before the DNA oligomers were amplified by PCR. All flanking primers for the separate amplifications and PCR cloning of oligos A and B were purified by SDS-PAGE essentially according to the methods of Sambrook et al., 1989, above. From the CHROMASPIN-purified oligo A, 1 ⁇ l of sample stock was PCR-amplified in a reaction volume of 100 ⁇ l by adding 5 ⁇ l of 5 ⁇ M of oligo Sequence ID No. 9:
  • Oligo B was PCR-amplified by the primer pairs Sequence ID No. 11:
  • Double-stranded PCR-amplified products for oligos A and B were gel-purified, restriction-digested with PstI/AvrII (PCR product of oligo A) and BstEII/AvrII (PCR product of oligo B), and sucloned into the complementary PstI/BstEII sites of the heavy chain staging vector, VHpBS.
  • the humanized VH sequence was subcloned into the pG1g vector, resulting in the final human IgG1 heavy chain expression vector, hLL2pG1g.
  • oligo E 150-mer
  • oligo F 121-mer
  • Oligo E Sequence ID No. 13 5′-CCT AGT GGA TGC CCA GTA GAT CAG CAG TTT AGG TGC TTT CCC TGG TTT CTG GTG GTA CCA GGC CAA GTA GTT CTT GTG ATT TGC ACT GTA TAA AAC ACT TTG ACT GGA CTT ACA GCT CAT AGT GAC CCT ATC TCC AAC AGA TGC GCT CAG-3′
  • [0093] represents the minus strand of the humanized VK domain complementary to nt 31 to 180, and this sequence was PCR-amplified by oligo Sequence ID No. 14:
  • Oligo F Sequence ID No. 16 5′-GCA CCT TGG TCC CTC CAC CGA ACG TCC ACG AGG AGA GGT ATT GGT GAC AAT AAT ATG TTG CAA TGT CTT CTG GTT GAA GAG AGC TGA TGG TGA AAG TAA AAT CTG TCC CAG ATC CGC TGC C-3′
  • [0098] represents the minus strand of the humanized LL2 VK domain complementary to nt 208 to 327, and was PCR amplified by oligo Sequence ID No. 17:
  • FIG. 1 Sequence ID Nos. 1 and 2
  • FIG. 1A amino acid sequence between murine and humanized LL2 VK domains
  • FIG. 1B human REI framework sequences were used for all FRs.
  • human EU framework sequences were used for FR 1-3
  • NEWM sequences were used for FR-4. Only human FR sequences that are different from that of the mouse are shown. Asterisks indicate murine FR sequences that are different from that of the human FR at corresponding positions. Murine residues at these positions were retained in the humanized structure. CDRs are boxed.
  • FIG. 4A (Sequence ID No. 3) there are shown the double stranded DNA and corresponding amino acid sequences (shown by single letter code) of the humanized LL2 VK domain. CDR 1-3 amino acid sequences are boxed. The corresponding display for VH is shown in FIG. 4B (Sequence ID No. 4).
  • FIG. 5A Sequence ID No. 5
  • FIG. 5B Sequence ID No. 6
  • Amino acid sequences are shown by the single-letter code, and CDR amino acid sequences are boxed.
  • pKh is a pSVhyg-based expression vector containing the genomic sequence of the human kappa constant region, an Ig enhancer, a kappa enhancer and the hygromycin-resistant gene. Similarly, the ca.
  • the two plasmids were co-transfected into Sp2/0-Ag14 cells by electroporation and selected for hygromycin resistance. Supernatants from colonies surviving selection were monitored for chimeric antibody secretion by ELISA assay (see above). The transfection efficiency was approximately 1-10 ⁇ 10 6 cells. The antibody expression level, in a terminal culture, was found to vary in the range between ⁇ 0.10 and 2.5 ⁇ g/ml.
  • FIG. 7 shows the results of analyzing protein A-purified mLL2 (lanes 4 and 7) and cLL2 (lanes 5 and 8) by SDS-PAGE under reducing and nonreducing conditions, respectively.
  • HMW stands for high molecular weight protein markers
  • LMW light molecular weight markers.
  • the light chains of both mLL2 and cLL2 (lanes 4 and 5) migrated primarily as a doublet band, with a higher than expected apparent molecular weight.
  • the human kappa constant region of cLL2 is known to contain no potential glycosylation site, it can be inferred that the potential glycosylation site identified in the FR1 region of LL2 VK domain was utilized.
  • FIG. 8 shows the results of analyzing different versions of hLL2 and cLL2 antibodies by SDS-PAGE under reducing and non-reducing conditions.
  • LMW and HMW are molecular weight markers.
  • Lanes 3 and 6 are cLL2 antibodies.
  • Lanes 4 and 7 are hLL2 with seven murine FR residues in the VH domain (hLL2-1).
  • Lanes 5 and 8 are hLL2 with 6 murine FR residues in the VH domain (hLL2-2).
  • the humanized light chains migrated more rapidly and as more discrete bands compared to chimeric light chains.
  • FIG. 9 shows the results of SDS-PAGE analysis on mix-and-match and cLL2 and hLL2 antibodies under both reducing and non-reducing conditions.
  • Lanes 1 and 2 are molecular weight markers.
  • Lanes 3 and 7 are cLL2.
  • Lanes 4 and 8 are mix-and-match with a humanized light and chimeric heavy chain [(hL/cH)LL2].
  • Lanes 5 and 9 are chimeric light and humanized heavy (Version 1) chains [(cL/hH)LL2-1].
  • Lanes 6 and 10 are chimeric light and a humanized heavy (version 2) chains [(cL/hH)LL2-2].
  • the humanized LL2 version 1 contains 7 murine FR residues in the VH domain, while version 2 contains 6 murine FR residues in the VH domain. It is noteworthy that the position of the light chain of (hL/cH)LL2 (lane 4) is different from that of the others, suggesting that there is no carbohydrate attachment to the humanized LL2 light chain.
  • a competition cell binding assay was carried out to assess the immunoreactivity of cLL2 relative to the parent mLL2. Using 131 I-labeled mLL2 (0.025 ⁇ g/ml) as a probe, Raji cells were incubated with the antibodies and the relative binding to the cells determined from the amount of cell-bound labeled mLL2 (see above). As shown by the competition assays described in FIG. 10, both mLL2 and cLL2 antibodies exhibited similar binding activities.
  • cLL2, mix-and-match LL2, hLL2-1 or hLL2-2 antibodies were incubated with 10 8 Raji cells in the presence of varying concentrations of mLL2 F(ab′) 2 fragments (as competitor) in a final volume of 100 ⁇ l of PBS buffer supplemented with 1% FCS and 0.01% sodium azide. The mixture was incubated for 30 minutes at 4° C., and washed three times with PBS to remove unbound antibodies.
  • the binding levels of the antibodies were assessed by adding a 20 ⁇ diluted FITC-labeled goat anti-human IgG1, Fc fragment-specific antibodies (Jackson ImmunoResearch, West Grove, Pa.). The cells were washed three times with PBS, and fluorescence intensities measured by a FACSCAN fluorescence activated cell sorter (Becton-Dickinson, Bedford, Mass.). The results are shown in FIG. 11A.
  • FIGS. 11A and B demonstrate that the immunoreactivity of cLL2 is similar or identical to that of humanized or mix-and-match antibodies. Taken together with the comparison of cLL2 with mLL2 (FIG. 10), the authenticity of the sequences for chimeric and humanized VK and VH obtained is established, and the functionality of cLL2 and hLL2 confirmed.
  • LL2 antibody One of the unique characteristics of the LL2 antibody is its rapid internalization upon binding to Raji cells (Shih et al., 1994 above). Murine LL2 after internalization is likely to be rapidly transferred to the Golgi apparatus and from there to the lysosomes, the organelle responsible for the degradation of a wide variety of biochemicals (Keisari et al., Immunochem., 10: 565 (1973)).
  • Rates of antibody internalization were determined according to Opresko et al., 1987 above. The ratio of CPM intracellular /CPM surface was determined as a function of time.
  • Rates of LL2 antibody internalization were determined by incubating radiolabeled LL2 antibody (1 ⁇ 10 6 cpm) with 0.5 ⁇ 10 6 Raji cells in 0.5 ml of DMEM buffer containing 1% human serum for 2 hrs. at 4° C. Excess human serum was included to saturate Raji cell surface Fc receptors in order to exclude or minimize non-antigen-specific internalization mediated through the Fc receptors. Unbound radiolabeled LL2 antibodies were removed from the cells by washing three times with 0.5 ml portions of DMEM at 4° C. Cells were then incubated at 37° C., and, at timed intervals, aliquots of the cell suspension were transferred to ice in order to stop internalization.
  • the cells in these aliquots were isolated by centrifugation at 1,000 ⁇ g for 5 mins. at 4° C., and surface bound radiolabeled LL2 stripped off cells with 1 ml of 0.1 M glycine acetate buffer, pH 3, for 8 mins. at 4° C. Radioactivity thus obtained (CPM surface) and radioactivity remaining in the cells (CPM intracellular) were determined. Rates of internalization were calculated from the slope of the plot of intracellular:surface radioactivity ratios as a function of time.
  • the pattern of internalization for mLL2, cLL2 and hLL2 was also monitored by fluorescence microscopy on a time-course basis using a FITC-labeled second antibody probe as described in the specification. Internalization of both antibodies was observed in at the earliest time point measurable. At 5 minutes, antibodies were seen both on the cell surface and internalized in areas immediately adjacent to the membrane as cytoplasmic micro-vesicles. At 15 min. post-incubation, the fine dots dispersed around the intramembrane began to merge into a group of granules, at locations believed to be the Golgi apparatus. As more antibodies were being internalized after 30 min.
  • A103 antibody an IgG2a antibody that binds to the surface of all human epithelial cells but does not internalize efficiently (Mattes et al., Hybridoma, 2: 253 (1983)) showed strong membrane staining at up to 2 h, while the anti-transferrin receptor antibody (5F9) internalized rapidly, just as did LL2.
  • Murine and chimeric LL2 light chains were treated with (+) or without ( ⁇ ) endoglycosidase F conventionally, and the antibody products examined by SDS-PAGE under reducing and non-reducing conditions (FIG. 13). There was no distinction between the antibody types as to electrophoretic behavior. In both cases, deglycosylation reduced the rate of migration of the light chain.
  • a mutation was introduced at position 18 of the light chain so that the Asn was replaced with Gin to produce LL2Q VK FR1.
  • SDS-PAGE analyses demonstrated that the NVT to QVT mutation abolished glycosylation of the antibody.
  • Comparison of the Raji cell binding affinity for cLL2 with and without light chain VK glycosylation demonstrated that the carbohydrate moiety was without influence on binding of the antibody to these cells.
  • glycosylation site can be used for conjugating therapeutic or diagnostic agents to LL2 without compromising the ability of the antibody to bind and internalize in B-lymphoma or leukemia cells.
  • variable region carbohydrate moiety in the functional activities of mLL2, cLL2 and hLL2 mAbs indicates that this moiety could profitably be used as the site of attachment of cytotoxic or detection agents such as radionuclides or toxins, and thereby avoid potential interference with the binding of the conjugate to a cell surface.
  • dextran (18-40 kDa) was converted to an amino dextran by oxidation of the dextran by NaIO 4 , Schiff base formation with NH 2 —CH 2 —CHOH—CH 2 —NH 2 , and reduction with NaBH 4 .
  • the amino dextran was then condensed with doxorubicin (DOX) in the presence of succinic anhydride and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide to produce DOX-aminodextran.
  • DOX doxorubicin
  • the latter was then condensed with an aldehydic group on LL2 VK FR-1 produced by oxidizing the carbohydrate moiety of the antibody fragment with NaIO 4 .
  • This conjugation system is not limited to the mLL2 antibody. In a comparative study, 15-19 moles of DOX/mole of cLL2 were bound.
  • conjugation possibilities are not limited to the use of a carrier dextran as in the example above.
  • the carbohydrate moiety of the LL2 VK FR1 region can be oxidized to produce aldehydic groups. These in turn can be reacted with an amino group on any drug to produce a Schiff base which, upon reduction, produces multiple copies of the drug stably linked to the antibody via alkyamine groups.
  • a LL2 covalently bound to a chelator there is produced a LL2 covalently bound to a chelator.
  • the chelator can be used to deliver to target tissues, for example, a radionuclide or paramagnetic metal ion, with a potential for diagnostic and therapeutic uses.
  • DTPA-LL2 conjugates were produced containing 5.5 moles of the chelator/mole of antibody which, in turn, chelated 47.3% of Y-90 and 97.4% In-111

Abstract

A chimeric LL2 monoclonal antibody is described in which the complementarity determining regions (CDRs) of the light and heavy chains of the murine LL2 anti-B-lymphoma, anti-leukemia cell monoclonal antibody has been recombinantly joined to the human kappa and IgG1 constant region domains, respectively, which retains the immunospecificity and B-cell lymphoma and leukemia cell internalization capacity of the parental murine LL2 monoclonal antibody, and which has the potential of exhibiting reduced human anti-mouse antibody production activity. A humanized LL2 monoclonal antibody is described in which the CDRs of the light and heavy chains have been recombinantly joined to a framework sequence of human light and heavy chains variable regions, respectively, and subsequently linked to human kappa and IgG1 constant region domains, respectively, which retains the immunospecificity and B-lymphoma and leukemia cell internalization capacities of the parental murine and chimeric LL2 monoclonal antibodies, and which has the potential for exhibiting reduced human anti-mouse antibody production activity. Vectors for producing recombinant chimeric and humanized chimeric monoclonal antibodies are provided. Isolated DNAs encoding the amino acid sequences of the LL2 variable light and heavy chain and CDR framework regions are described. Conjugates of chimeric and humanized chimeric LL2 antibodies with cytotoxic agents or labels find use in therapy and diagnosis of B-cell lymphomas and leukemias.

Description

    CROSS-REFERENCE TO RELATED PATENT APPLICATIONS
  • This application is a continuation of U.S. Ser. No. 09/741,843 filed on Dec. 22, 2000, which is a continuation of U.S. Ser. No. 09/127,902 filed on Aug. 3, 1998, now U.S. Pat. No. 6,187,287, which is a continuation of U.S. Ser. No. 08/690,102 filed on Jul. 31, 1996, now U.S. Pat. No. 5,789,554, which is a continuation of U.S. Ser. No. 08/289,576 filed on Aug. 12, 1994, now abandoned. This application claims only subject matter disclosed in the parent applications and therefore presents no new matter.[0001]
  • BACKGROUND OF THE INVENTION
  • The invention relates generally to immunoconjugates for diagnostic and therapeutic uses in cancer. In particular, the invention relates to recombinantly produced chimeric and humanized monoclonal antibodies directed against B-cell lymphoma and leukemia cells, which antibodies can be covalently conjugated to a diagnostic or therapeutic reagent without loss of antibody binding and internalization function and with reduced production of human anti-mouse antibodies. [0002]
  • Non-Hodgkins lymphoma (NHL) and chronic lymphocytic leukemia are B-cell malignancies that remain important contributors to cancer mortality. The response of these malignancies to various forms of treatment is mixed. They respond reasonably well to chemotherapy, and, in cases where adequate clinical staging of NHL is possible, as for patients with localized disease, satisfactory treatment may be provided using field radiation therapy (Hall et al., [0003] Radiology for the Radiologist, Lippincott, Philadelphia, 1989, pp 365-376). However, the toxic side effects associated with chemotherapy and the toxicity to the hematopoietic system from local, as well as whole body, radiotherapy, limits the use of these therapeutic methods. About one-half of the patients die from the disease (Posner et al., Blood, 61: 705 (1983)).
  • The use of targeting monoclonal antibodies conjugated to radionuclides or other cytotoxic agents offers the possibility of delivering such agents directly to the tumor site, thereby limiting the exposure of normal tissues to toxic agents (Goldenberg, [0004] Semin. Nucl. Med., 19: 332 (1989)). In recent years, the potential of antibody-based therapy and its accuracy in the localization of tumor-associated antigens have been demonstrated both in the laboratory and clinical studies (see., e.g., Thorpe, TIBTECH, 11: 42 (1993); Goldenberg, Scientific American, Science & Medicine, 1: 64 (1994); Baldwin et al., U.S. Pat. Nos. 4,925,922 and 4,916,213; Young, U.S. Pat. No. 4,918,163; U.S. Pat. No. 5,204,095; Irie et al., U.S. Pat. No. 5,196,337; Hellstrom et al., U.S. Pat. Nos. 5,134,075 and 5,171,665). In general, the use of radiolabeled antibodies or antibody fragments against tumor-associated markers for localization of tumors has been more successful than for therapy, in part because antibody uptake by the tumor is generally low, ranging from only 0.01% to 0.001% of the total dose injected (Vaughan et al., Brit. J. Radiol., 60: 567 (1987)). Increasing the concentration of the radiolabel to increase the dosage to the tumor is counterproductive generally as this also increases exposure of healthy tissue to radioactivity.
  • LL-2 (EPB2) is a highly specific anti-B-cell lymphoma and anti-lymphocytic leukemia cell murine monoclonal antibody (mAb) that is rapidly internalized by such cells and that can overcome some of the aforementioned difficulties (Shih et al., [0005] Int. J. Cancer, 56: 538 (1994)). LL2, which is of the IgG2a antibody type, was developed using the Raji B-lymphoma cell line as the source of antigen (Pawlak-Byczkowska et al., Cancer Res., 49: 4568 (1989)). Murine LL2 (mLL2) is known to react with an epitope of CD22 (Belisle et al., Proc Amer. Assn. Clin. Res., 34: A2873 (1993)). CD22 molecules are expressed in the cytoplasm of progenitor and early pre-B cells, and appear in the cell surface of mature B-cells.
  • By immunostaining of tissue sections, mLL2 was shown to react with 50 of 51 B-cell lymphomas tested. mLL2 is a highly sensitive means of detecting B-cell lymphoma cell in vivo, as determined by a radioimmunodetection method (Murthy et al., [0006] Eur. J. Nucl. Med., 19: 394 (1992)). The Fab′ fragment of mLL2 labeled with 99mTc localized to 63 of 65 known lesions in Phase II trial patients with B-cell lymphoma (Mills et al., Proc. Amer. Assn. Cancer Res., 14: A2857 (1993)). In addition, 131I-labeled mLL2 was therapeutically effective in B-cell lymphoma patients (Goldenberg et al., J. Clin. Oncol., 9: 548 (1991)). mLL2 Fab′ conjugated to the exotoxin PE38KDEL induced complete remissions of measurable human lymphoma xenografts (CA-46) growing in nude mice (Kreitman et al., Cancer Res., 53: 819 (1993)).
  • The clinical use of mLL2, just as with most other promising murine antibodies, has been limited by the development in humans of a HAMA response. While a HAMA response is not invariably observed following injection of mLL2, in a significant number of cases patients developed HAMA following a single treatment with mLL2. This can limit the diagnostic and therapeutic usefulness of such antibody conjugates, not only because of the potential anaphylactic problem, but also as a major portion of the circulating conjugate may be complexed to and sequestered by the circulating anti-mouse antibodies. This is exemplified by one study in which about 30% of the patients developed low level HAMA response following a single injection of about 6 mg of mLL2 [0007] 131I-IgG and nearly all developed a strong HAMA response with additional injections. On the other hand, with mLL2 Fab′ labeled with 99mTc, no HAMA response was observed. Such HAMA responses in general pose a potential obstacle to realizing the full diagnostic and therapeutic potential of the mLL2 antibody.
  • Although, as noted above, the use of fragments of mLL2, such as F(ab′)2 and Fab′, partially alleviate/circumvent these problems of immunogenicity, there are circumstances in which whole IgG is more desirable, such as when induction of cellular immunity is intended for therapy, or where an antibody with enhanced survival time is required. [0008]
  • In order to maximize the value of the mLL2 IgG antibody as a therapeutic or diagnostic modality and increase its utility in multiple and continuous administration modalities, it is an object of this invention to produce a mouse/human chimeric mAb (cLL2) and a humanized mAb (hLL2) related to mLL2 that retain the antigen-binding specificity of mLL2, but that elicit reduced HAMA in a subject receiving same. [0009]
  • It is another object of this invention to provide DNA sequences encoding the amino acid sequences of the variable regions of the light and heavy chains of the cLL2 and hLL2 mAbs, including the complementarity determining regions (CDR). [0010]
  • It is also an object of this invention provide conjugates of the hLL2 and cLL2 mAbs containing therapeutic or diagnostic modalities. [0011]
  • It is a further object of this invention to provide methods of therapy and diagnosis that utilize the humanized and chimeric mAbs of the invention. [0012]
  • These objects have been achieved by the invention described below in the specification and appended claims. [0013]
  • SUMMARY OF THE INVENTION
  • In one aspect of the invention, there is provided a cLL2 mAb related to mLL2 mAb, in which the murine light (VK) and heavy (VH) chain variable regions are joined to the human constant light (kappa) and heavy (IgG[0014] 1) chains.
  • This chimeric mAb retains the B-lymphoma and leukemia cell targeting and internalization properties of the parental mLL2. [0015]
  • In another aspect of the invention, there is provided a hLL2 mAb related to mLL2 mAb, in which the complementarity-determining regions (CDRs) of the light and heavy chains of the mLL2 mAb are joined to the framework (FR) sequence of human VK and VH regions, respectively, and subsequently to the human kappa and IgG[0016] 1 constant region domains, respectively. This humanized antibody retains the B-lymphoma and leukemia cell targeting and internalization characteristics of the parental mLL2 mAb, and can exhibit a lowered HAMA reaction.
  • In still another aspect, there is provided isolated polynucleotides comprising DNA sequences encoding the amino acid sequences of the variable light and heavy chains, respectively, of the hLL2 and cLL2 mAbs. [0017]
  • In an additional aspect, there is provided the amino acid sequences of the CDRs of the VK and VH chains. [0018]
  • In yet another aspect, there are provided conjugates in which the hLL2 or cLL2 mAb is covalently bonded to a diagnostic or therapeutic reagent. [0019]
  • In still another aspect, there are provided methods whereby the aforementioned mAb conjugates can be used to diagnose or treat B-cell lymphomas and lymphocytic leukemias. [0020]
  • These and other aspects and embodiments of the invention will become apparent by reference to the following specification and appended claims.[0021]
  • DESCRIPTION OF THE FIGURES
  • FIG. 1 is a comparison of the murine with the humanized LL2 VK (FIG. 1A) and VH (FIG. 1B) domains. Only hFR sequences (designated as REIHuVK and EUHuVH) different than mFR sequences (designated as murine) are shown, and designated by asterisks. More residues in these positions were retained in the humanized structure. CDRs are boxed. FR residues showing CDR contacts by computer modeling are underlined. [0022]
  • FIG. 2 shows vicinal relationships of the LL2 CDRs to their framework regions (FRs). Separate energy-minimized models for the VL and VH domains of mLL2 were constructed, and all FR residues within a radius of 4.5 Å or any CDR atom were identified as potential CDR-FR contacts. CDRs of the light (L1, L2, and L3, FIG. 2A) and heavy (H1, H2, and H3, FIG. 2B)) chains are shown as “ball and stick” representations superimposed on their respective, space-filling FRs. [0023]
  • FIG. 3 shows the light chain (FIG. 3A) staging (VKpBR) and mammalian expression (pKH) vectors, and the heavy chain (FIG. 3B) staging (VHpBS) and mammalian expression (pG1g) vectors. [0024]
  • FIG. 4 shows the double-stranded DNA and amino acid sequences of the LL2 VK domain (FIG. 4A) and the LL2 VH domain (FIG. 4B). Amino acid sequences encoded by the corresponding DNA sequences are given as one letter codes. CDR amino acid sequences are boxed. The Asn-glycosylation site located in FR1 of LL2VK (FIG. 4A) is shown as the underlined NVT sequence. [0025]
  • FIG. 5A shows the double stranded DNA and corresponding amino acid residues of the hLL2 VK domain. CDR amino acid sequences are boxed. The corresponding data for the VH domain is shown in FIG. 5B. [0026]
  • FIG. 6 is a schematic diagram representation of the PCR/gene synthesis of the humanized VH region and the subcloning into the staging vector, VHpBS. [0027]
  • FIG. 7 shows SDS-PAGE analysis of mLL2 and cLL2 antibodies under non-reducing (lanes 6-8) and reducing (lanes 3-5, light and heavy chains) conditions. [0028] Lanes 3 and 6 include a control antibody.
  • FIG. 8 shows SDS-PAGE analysis of different versions of cLL2 and hLL2 antibodies under reducing (lanes 3-5) and non-reducing (lanes 6-8) conditions. [0029]
  • FIG. 9 shows SDS-PAGE analysis on mix-and-match cLL2 and hLL2 antibodies under reducing (lanes 3-6) and non-reducing (lanes 7-10) conditions. cLL2 serves as the control. [0030]
  • FIG. 10 shows the results of a comparative Raji cell competitive antibody binding assay involving mLL2 and cLL2 antibodies competing for binding to cells against tracer radiolabeled mLL2. [0031]
  • FIG. 11 shows the results of a comparative Raji cell competitive antibody binding assay in which mixed humanized/chimeric LL2s were compared to cLL2 (FIG. 11A), and two versions of hLL2 compared to cLL2 (FIG. 11B). [0032]
  • FIG. 12 shows a comparison of antibody internalization:surface binding ratios as a function of time for cLL2, cLL2 (Q to V mutagenesis), hLL2 and mLL2 antibodies. [0033]
  • FIG. 13 shows an SDS-PAGE analysis of mLL2 and cLL2 after deglycosylation by endoglycosidase F. [0034]
  • FIG. 14 shows the effect of deglycosylation of mLL2 on its binding affinity to Raji cells.[0035]
  • DETAILED DESCRIPTION OF THE INVENTION
  • cDNAs encoding the VL and VH regions of the mLL2 mAb have been isolated and separately recombinantly subcloned into mammalian expression vectors containing the genes encoding kappa and IgG[0036] 1 constant regions, respectively, of human antibodies. Cotransfection of mammalian cells with these two recombinant DNAs expressed a cLL2 mAb that, like the parent mLL2 mAb, bound avidly to, and was rapidly internalized by, B-lymphoma cells.
  • The CDRs of the VK and VH DNAs have been similarly recombinantly linked to the framework (FR) sequences of the human VK and VH regions, respectively, which are subsequently linked, respectively, to the human kappa and IgG[0037] 1 constant regions, so as to express in mammalian cells as described above hLL2.
  • In this specification, the expressions “cLL2” or “cLL2 mAb” are intended to refer to the chimeric monoclonal antibody constructed by joining or subcloning the murine VK and VH regions to the human constant light and heavy chains, respectively. The expressions “hLL2” or “hLL2 mAb” are intended to refer to the humanization of the chimeric monoclonal antibody by replacing the murine FR sequences in cLL2 with that of human framework regions. [0038]
  • Covalent conjugates between cLL2 and hLL2 mAbs and a diagnostic or chemotherapeutic reagent, formulated in pharmaceutically acceptable vehicles (see, e.g., [0039] Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Co., Easton, Pa., 1990) can be prepared that have the advantages, compared to prior art antibody conjugates, of B-cell lymphoma-specific and leukemia cell-specific targeting, rapid internalization into target cells, rapid liberation of the diagnostic or chemotherapeutic reagent intracellularly (thereby increasing effectiveness of the reagent), and a potential reduction of the HAMA response in the human patient.
  • As the VK-appended carbohydrate moiety of the cLL2 mAb is shown herein not to be involved in binding to B-lymphoma cells, it is preferred to use conjugates in which the reagent is bound to the antibody through such carbohydrate moieties, such as through oxidized carbohydrate derivatives. Methods for the production of such conjugates and their use in diagnostics and therapeutics are provided, for example. in Shih et al., U.S. Pat. No. 5,057,313, Shih et al., [0040] Int. J. Cancer 41: 832 (1988), and copending, commonly owned Hansen el al., U.S. Ser. No. 08/162,912, the contents of which are incorporated herein by reference. Direct linkage of the reagent to oxidized carbohydrate without the use of a polymeric carrier is described in McKearn et al., U.S. Pat. No. 5,156,840, which is also incorporated by reference.
  • A wide variety of diagnostic and therapeutic reagents can be advantageously conjugated to the antibodies of the invention. These include: chemotherapeutic drugs such as doxorubicin, methotrexate, taxol, and the like; chelators, such as DTPA, to which detectable labels such as fluorescent molecules or cytotoxic agents such as heavy metals or radionuclides can be complexed; and toxins such as Pseudomonas exotoxin, and the like. Several embodiments of these conjugates are described in the examples below. [0041]
  • Cell lines and culture media used in the present invention include LL2 (EPB-2) hybridoma cells (Pawlak-Byczkowska et al. 1989 above), Sp2/0-Ag14 myeloma cells (ATCC, Rockville, Md.) and Raji cells. These cells are preferably cultured in Dulbecco's modified Eagle's Medium (DMEM) supplemented with 10% FCS (Gibco/BRL, Gaithersburg, Mass.), 2 mM L-glutamine and 75 μg/ml gentamicin, (complete DMEM). Transfectomas are grown in Hybridoma Serum Free Medium, HSFM, (Gibco/BRL, Gaithersburg, Mass.) containing 10% of FCS and 75 μg/ml gentamicin (complete HSFM) or, where indicated, in HSFM containing only antibiotics. Selection of the transfectomas may be carried out in complete HSFM containing 500 μg/ml of hygromycin (Calbiochem, San Diego, Calif.). All cell lines are preferably maintained at 37° C. in 5% CO[0042] 2.
  • An important aspect of this invention is that antibody variable domains can be modeled by computer modeling (see, for example, Dion, in Goldenberg et al. eds., [0043] Cancer Therapy With Radiolabeled Antibodies, CRC Press, Boca Raton, Fla., 1994), which is incorporated by reference. In general, the 3-D structure for both the mLL22 and hLL2 mAbs are best modeled by homology. The high frequency of residue identities (75.0 to 92.3%) between the deduced primary sequences of mLL2 light chain FR regions and human REI (VK) facilitates this approach because of the availability of crystallographic data from the Protein Data Bank (PDR Code 1REI, Bernstein et al., J. Mol. Biol. 112: 535 (1977)), which is incorporated by reference. Similarly, antibody EU (VH) sequences can be selected as the computer counterparts for FR1 to FR3 of the mLL2 heavy chain; FR4 was based on NEWM. As X-ray coordinate data is currently lacking for the EU sequence, NEWM structural data (PDR Code 3FAB) for FRs 1 to 4 can be used, and amino acid side groups can be replaced to correspond to mLL2 or EU (hLL2) as needed. The CDR of the light chain can be modeled from the corresponding sequence of 1MCP (L1 and L2) and 1REI (L3). For heavy chain CDRs, H1 and H2 can be based on 2HFL and 1MCP, respectively, while H3 can be modeled de novo. Wherever possible, side group replacements should be performed so as to maintain the torsion angle between Cα and Cβ. Energy minimization may be accomplished by the AMBER forcefield (Weiner et al, J. Amer. Chem. Soc. 106: 765 (1984) using the convergent method. Potentially critical FR-CDR interactions can be determined by initially modeling the light and heavy variable chains of mLL2. All FR residues within a 4.5 Å radius of all atoms within each CDR can thereby be identified and retained in the final design model of hLL2.
  • Once the sequences for the hLL2 VK and VH domains are designed, CDR engrafting can be accomplished by gene synthesis using long synthetic DNA oligonucleotides as templates and short oligonucleotides as primers in a PCR reaction. In most cases, the DNA encoding the VK or VH domain will be approximately 350 bp long. By taking advantage of codon degeneracy, a unique restriction site may easily be introduced, without changing the encoded amino acids, at regions close to the middle of the V gene DNA sequence. For example, at DNA nucleotide positions 157-162 (amino acid positions 53 and 54) for the hLL2 VH domain, a unique AvrII site can be introduced while maintaining the originally designed amino acid sequence (FIG. 4B). Two long non-overlapping single-stranded DNA oligonucleotides (˜150 bp) upstream and downstream of the AvrII site (see, for example, oligo A and oligo B, Example 3 below) can be generated by automated DNA oligonucleotide synthesizer (Cyclone Plus DNA Synthesizer, Milligen-Biosearch). As the yields of full length DNA oligonucleotides such as oligos A and B may be expected to be low, they can be amplified by two pairs of flanking oligonucleotides (oligo Seq. ID Nos. 7 and 8 for oligo A; oligo Seq. ID Nos. 9 and 10 for oligo B, Example 3) in a PCR reaction. The primers can be designed with the necessary restriction sites to facilitate subsequent subcloning. Primers for oligo A and for oligo B should contain overlapping sequence at the AvrII site so that the resultant PCR product for oligo A and B, respectively, can be joined in-frame at the AvrII site to form a full length DNA sequence ([0044] ca 350 bp) encoding the hLL2 VH domain. The ligation of the PCR products for oligo A (restriction-digested with PstI and AvrII) and B (restriction-digested with AvrII and BstEII) at the AvrII site and their subcloning into the PstII/BstEII sites of the staging vector, VHpBS, can be completed in a single three-fragment-ligation step (See, for example, Example 3). The subcloning of the correct sequence into VHpBS can be first analyzed by restriction digestion analysis and subsequently confirmed by sequencing reaction according to Sanger et al., Proc. Natl. Acad. Sci. USA 74: 5463 (1977).
  • The HindIII/BamHI fragment containing the Ig promoter, leader sequence and the hLL2 VH sequence can be excised from the staging vector and subcloned to the corresponding sites in a pSVgpt-based vector, pG1g, which contains the genomic sequence of the human IgG constant region, an Ig enhancer and a gpt selection marker, forming the final expression vector, hLL2pG1g. Similar strategies can be employed for the construction of the hLL2 VK sequence. The restriction site chosen for the ligation of the PCR products for the long oligonucloetides (oligos C and D, see examples below) can be NruI in this case. [0045]
  • The DNA sequence containing the Ig promoter, leader sequence and the hLL2 VK sequence can be excised from the staging vector VKpBR by treatment with BamHI/HindIII, and can be subcloned into the corresponding sites of a pSVhyg-based vector, pKh, which contains the genomic sequence of human kappa chain constant regions, a hygromycin selection marker, an Ig and a kappa enhancer, forming the final expression vector, hLL2pKh. [0046]
  • As humanization sometimes results in a reduction or even loss of antibody affinity, additional modification might be required in order to restore the original affinity (See, for example, Tempest et al., [0047] Bio/Technology 9: 266 (1991); Verhoeyen et al., Science 239: 1534 (1988)), which are incorporated by reference. Knowing that cLL2 exhibits a binding affinity comparable to that of its murine counterpart (see Example 5 below), defective designs, if any, in the original version of hLL2 can be identified by mixing and matching the light and heavy chains of cLL2 to those of the humanized version. SDS-PAGE analysis of the different mix-and-match humanized chimeric LL2 under non-reducing (the disulfide L-H chain connections remain intact) and reducing conditions (the chains separate, permitting analyses of the relationships of the different types of light and heavy chains on the properties of the molecule). For example, migration as multiple bands as a higher apparent molecular size can be due to the presence of a glycan group at the N-linked glycosylation site found at the FRI region of the murine VK domain of LL2. For another example, a discrete band migrating at about 25 kDa is the expected molecular size for a non-glycosylated light chain.
  • In general, to prepare cLL2 mAb, VH and VK chains of mLL2 can be obtained by PCR cloning using DNA products and primers. Orlandi et al., infra, and Leung et al., infra. The VK PCR primers may be subcloned into a pBR327 based staging vector (VKpBR) as described above. The VH PCR products may be subcloned into a similar pBluescript-based staging vector (VHpBS) as described above. The fragments containing the VK and VH sequences, along with the promoter and signal peptide sequences, can be excised from the staging vectors using HindIII and BamHI restriction endonucleases. The VK fragments (about 600 bp) can be subcloned into a mammalian expression vector (for example, pKh) conventionally. pKh is a pSVhyg-based expression vector containing the genomic sequence of the human kappa constant region. an Ig enhancer, a kappa enhancer and the hygromucin-resistant gene. Similarly, the about 800 bp VH fragments can be subcloned into pG1g, a pSVgpt-based expression vector carrying the genomic sequence of the human IgG1 constant region, an Ig enhancer and the xanthine-guanine phosphoribosyl transferase (gpt) gene. The two plasmids may be transfected into mammalian expression cells, such as Sp2/0-Ag14 cells, by electroporation and selected for hygromycin resistance. Colonies surviving selection are expanded, and supernatant fluids monitored for production of cLL2 mAb by an ELISA method. A transfection efficiency of about 1-10×10[0048] 6 cells is desirable. An antibody expression level of between 0.10 and 2.5 μg/ml can be expected with this system.
  • RNA isolation, cDNA synthesis, and amplification can be carried out as follows. Total cell RNA can be prepared from a LL2 hybridoma cell line, using a total of about 10[0049] 7 cells, according to Sambrook et al., (Molecular Cloning: A Laboratory Manual, Second ed., Cold Spring Harbor Press, 1989), which is incorporated by reference. First strand cDNA can be reverse transcribed from total RNA conventionally, such as by using the SuperScript preamplification system (Gibco/BRL., Gaithersburg, Md.). Briefly, in a reaction volume of 20 μl, 50 ng of random primers can be annealed to 5 μg of RNAs in the presence of 2 μl of 10× synthesis buffer [200 mM Tris-HCl (pH 8.4), 500 mM KCl, 25 mM MgCl2, 1 mg/ml BSA], 1 μl of 10 mM dNTP mix, 2 μl of 0.1 M DTT, and 200 units of SuperScript reverse transcriptase. The elongation step is initially allowed to proceed at room temperature for 10 min followed by incubation at 42° C. for 50 min. The reaction can be terminated by heating the reaction mixture at 90° C. for 5 min.
  • The VK and VH sequences for cLL2 or hLL2 can amplified by PCR as described by Orlandi et al., (Proc. Natl. Acad. Sci., USA, 86: 3833 (1989)) which is incorporated by reference. VK sequences may be amplified using the primers CK3BH and VK5-3 (Leung et al., [0050] BioTechniques, 15: 286 (1993), which is incorporated by reference), while VH sequences can be amplified using the primer CH1B which anneals to the CHI region of murine IgG, and VHIBACK (Orlandi et al., 1989 above). The PCR reaction mixtures containing 10 μl of the first strand cDNA product, 9 μl of 10× PCR buffer [500 mM KCl, 100 mM Tris-HCl (pH 8.3), 15 mM MgCl2, and 0.01% (w/v) gelatin] (Perkin Elmer Cetus, Norwalk, Conn.), can be subjected to 30 cycles of PCR. Each PCR cycle preferably consists of denaturation at 94° C. for 1 min, annealing at 50° C. for 1.5 min, and polymerization at 72° C. for 1.5 min. Amplified VK and VH fragments can be purified on 2% agarose (BioRad, Richmond, Calif.). See Example 3 for a method for the synthesis of an oligo A (149-mer) and an oligo B (140-mer) on an automated Cyclone Plus DNA synthesizer (Milligan-Biosearch) for use in constructing humanized V genes.
  • PCR products for VK can be subcloned into a staging vector, such as a pBR327-based staging vector VKpBR that contains an Ig promoter, a signal peptide sequence and convenient restriction sites to facilitate in-frame ligation of the VK PCR products. PCR products for VH can be subcloned into a similar staging vector, such as the pBluescript-based VHpBS. Individual clones containing the respective PCR products may be sequenced by, for example, the method of Sanger et al., [0051] Proc. Natl. Acad. Sci., USA, 74: 5463 (1977) which is incorporated by reference.
  • The DNA sequences described herein are to be taken as including all alleles, mutants and variants thereof, whether occurring naturally or induced. [0052]
  • The two plasmids can be co-transfected into an appropriate cell, e.g., myeloma Sp2/0-Ag14, colonies selected for hygromycin resistance, and supernatant fluids monitored for production of cLL2 or hLL2 antibodies by, for example, an ELISA assay, as described below. [0053]
  • Transfection, and assay for antibody secreting clones by ELISA, can be carried out as follows. About 10 μg of hLL2pKh (light chain expression vector) and 20 μg of hLL2pG1g (heavy chain expression vector) can be used for the transfection of 5×10[0054] 6 SP2/0 myeloma cells by electroporation (BioRad, Richmond, Calif.) according to Co et al., J. Immunol., 148: 1149 (1992) which is incorporated by reference. Following transfection, cells may be grown in 96-well microtiter plates in complete HSFM medium (GIBCO, Gaithersburg, Md.) at 37° C., 5% CO2. The selection process can be initiated after two days by the addition of hygromycin selection medium (Calbiochem, San Diego, Calif.) at a final concentration of 500 μg/ml of hygromycin. Colonies typically emerge 2-3 weeks post-electroporation. The cultures can then be expanded for further analysis.
  • Transfectoma clones that are positive for the secretion of chimeric or humanized heavy chain can be identified by ELISA assay. Briefly, supernatant samples (100 μl) from transfectoma cultures are added in triplicate to ELISA microtiter plates precoated with goat anti-human (GAH)-IgG, F(ab′)[0055] 2 fragment-specific antibody (Jackson ImmunoResearch, West Grove, Pa.). Plates are incubated for 1 h at room temperature. Unbound proteins are removed by washing three times with wash buffer (PBS containing 0.05% polysorbate 20). Horseradish peroxidase (HRP) conjugated GAH-IgG, Fc fragment-specific antibodies (Jackson ImmunoResearch, West Grove, Pa.) are added to the wells, (100 μl of antibody stock diluted×104, supplemented with the unconjugated antibody to a final concentration of 1.0 μg/ml). Following an incubation of 1 h, the plates are washed, typically three times. A reaction solution, [100 μl, containing 167 μg of orthophenylene-diamine (OPD) (Sigma, St. Louis, Mo.), 0.025% hydrogen peroxide in PBS], is added to the wells. Color is allowed to develop in the dark for 30 minutes. The reaction is stopped by the addition of 50 μl of 4 N HCl solution into each well before measuring absorbance at 490 nm in an automated ELISA reader (Bio-Tek instruments, Winooski, Vt.). Bound chimeric antibodies are than determined relative to an irrelevant chimeric antibody standard (obtainable from Scotgen, Ltd., Edinburg, Scotland).
  • Antibodies can be isolated from cell culture media as follows. Transfectoma cultures are adapted to serum-free medium. For production of chimeric antibody, cells are grown as a 500 ml culture in roller bottles using HSFM. Cultures are centrifuged and the supernatant filtered through a 0.2 micron membrane. The filtered medium is passed through a protein A column (1×3 cm) at a flow rate of 1 ml/min. The resin is then washed with about 10 column volumes of PBS and protein A-bound antibody is eluted from the column with 0.1 M glycine buffer (pH 3.5) containing 10 mM EDTA. Fractions of 1.0 ml are collected in tubes containing 10 μl of 3 M Tris (pH 8.6), and protein concentrations determined from the absorbancies at 280/260 nm. Peak fractions are pooled, dialyzed against PBS, and the antibody concentrated, for example, with the Centricon 30 (Amicon, Beverly, Mass.). The antibody concentration is determined by ELISA, as before, and its concentration adjusted to about 1 mg/ml using PBS. Sodium azide, 0.01% (w/v), is conveniently added to the sample as preservative. [0056]
  • Comparative binding affinities of the mLL2, cLL2 and hcLL2 antibodies thus isolated may be determined by direct radioimmunoassay. mLL2 can be labeled with [0057] 131I or 125I using the chloramine T method (see, for example, Greenwood et al., Biochem. J., 89: 123 (1963) which is incorporated by reference). The specific activity of the iodinated antibody is typically adjusted to about 10 μCi/μg. Unlabeled and labeled antibodies are diluted to the appropriate concentrations using reaction medium (HSFM supplemented with 1% horse serum and 100 μg/ml gentamicin). The appropriate concentrations of both labeled and unlabeled antibodies are added together to the reaction tubes in a total volume of 100 μl. A culture of Raji cells is sampled and the cell concentration determined. The culture is centrifuged and the collected cells washed once in reaction medium followed by resuspension in reaction medium to a final concentration of about 107 cells/ml. All procedures are carried out in the cold at 4° C. The cell suspension, 100 μl, is added to the reaction tubes. The reaction is carried out at 4° C. for 2 h with periodic gentle shaking of the reaction tubes to resuspend the cells. Following the reaction period, 5 ml of wash buffer (PBS containing 1% BSA) is added to each tube. The suspension is centrifuged and the cell pellet washed a second time with another 5 ml of wash buffer. Following centrifugation, the amount of remaining radioactivity remaining in the cell pellet is determined in a gamma counter (Minaxi, Packard Instruments, Sterling, Va.).
  • The Raji cell surface antigen binding affinities of mix-and-match and fully humanized antibodies can be compared to that of cLL2 using various concentrations of mLL2 F(ab′)[0058] 2 fragments devoid of the Fc portion as competitors, as evaluated by flow cytometry assay. Residual surface-bound LL2 antibodies carrying the human Fe portions (cLL2 and mix-and-match LL2) can be detected by a FITC-labeled anti-human Fc specific antibody in a flow cytometry assay. Where mix-and-match LL2 antibodies exhibit antigen-binding affinities similar to that of cLL2, it can be concluded that the original designs for the humanization of both the light and heavy chains retain the mLL2 immunoreactivity.
  • The internalization of mLL2, cLL2 and hLL2 antibodies into target cells can be followed by fluorescence labeling, essentially according to the procedure of Pirker et al., [0059] J. Clin. Invest., 76: 1261 (1985), which is incorporated by reference. Cultured Raji cells are centrifuged and the cells resuspended in fresh medium to a concentration of about 5×106 cells/ml. To each well of a 96-well microtiter plate, 100 μl of the cell suspension is added. The antibodies, 40 μg/ml, in a volume of 100 μl are added to the reaction wells at timed intervals so as to terminate all reactions simultaneously. The plate is incubated at 37° C. in a CO2 cell culture incubator. Unbound antibodies are removed by washing the cells three times with cold 1% FCS/PBS at the end of the incubation. The cells are then treated with 1 ml of Formaid-Fresh [10% formalin solution (Fisher, Fair Lawn, N.J.)] for 15 min at 4° C. After washing, antibodies present either on the cell surface or inside the cells are detected by treatment with FITC-labeled goat anti-mouse antibody (Tago, Burlingame, Calif.), or FITC-labeled goat anti-human antibody (Jackson ImmunoResearch, West Grove, Pa.), depending on whether the antibody being assayed for is murine, chimeric, or humanized, respectively. Fluorescence distributions are evaluated using a BH-2 fluorescence microscope (Olympus, Lake Success, N.Y.).
  • The rate of antibody internalization can be determined according to Opresko et al., ([0060] J. Biol. Chem., 262: 4116 (1987)), using radioiodinated antibody as tracer. Briefly, radiolabeled antibodies (1×104 cpm) are incubated with the Raji cells (1×106 cells/ml) at 4° C. for 2 h in 0.5 ml of DMEM medium containing 1% human serum. Following the reaction interval, non-specifically bound antibodies are removed by washing three times with 0.5 ml of DMEM medium. To each of the reaction tubes 0.5 ml of DMEM medium is added and the suspension incubated at 37° C. for the determination of internalization. At timed intervals, triplicates of cells are removed and chilled immediately in an ice bath to stop further internalization. Cells are centrifuged at 1000×g for 5 min at 4° C. The supernatant is removed and counted for radioactivity. The surface-bound radioactivity is removed by treatment with 1 ml 0.1 M acetate/0.1 M glycine buffer at pH 3.0 for 8 min. in the cold. Radioactivity removed by the acid treatment, and that remaining associated with the cells, are determined. The ratio of the CPMinternalization/CPMsurface is plotted versus time to determine the rate of internalization from the slope.
  • Detailed protocols for oligonucleotide-directed mutagenesis and related techniques for mutagenesis of cloned DNA are well-known. For example, see Sambrook et al. and Ausubel et al. above. [0061]
  • Asn-linked glycosylation sites may be introduced into antibodies using conventional site-directed oligonucleotide mutagenesis reactions. For example, to introduce an Asn in position 18 of a kappa protein, one may alter codon 18 from AGG to AAC. To accomplish this, a single stranded DNA template containing the antibody light chain sequence is prepared from a suitable strain of [0062] E. coli (e.g., dutung−) in order to obtain a DNA molecule containing a small number of uracils in place of thymidine. Such a DNA template can be obtained by M13 cloning or by in vitro transcription using a SP6 promoter. See, for example, Ausubel et al., eds., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, 1987. An oligonucleotide containing the mutated sequence is synthesized conventionally, annealed to the single-stranded template and the product treated with T4 DNA polymerase and T4 DNA ligase to produce a double-stranded DNA molecule. Transformation of wild type E. coli (dut+ ung+) cells with the double-stranded DNA provides an efficient recovery of mutated DNA.
  • Alternatively, an Asn-linked glycosylation site can be introduced into an antibody light chain using an oligonucleotide containing the desired mutation as the primer and DNA clones of the variable regions for the, VL chain, or by using RNA from cells that produce the antibody of interest as a template. Also see, Huse, in ANTIBODY ENGINEERING: A PRACTICAL GUIDE, Boerrebaeck, ed., W. H. Freeman & Co., pp 103-120, 1992. Site-directed mutagenesis can be performed, for example, using the TRANSFORMER™ kit (Clontech, Palo Alto, Calif.) according to the manufacturer's instructions. [0063]
  • Alternatively, a glycosylation site can be introduced by synthesizing an antibody chain with mutually priming oligonucleotides, one such containing the desired mutation. See, for example, Uhlmann, [0064] Gene 71: 29 (1988); Wosnick et al., Gene 60: 115 (1988); Ausubel et al., above, which are incorporated by reference.
  • Although the general description above referred to the introduction of an Asn glycosylation site in position 18 of the light chain of an antibody, it will occur to the skilled artisan that it is possible to introduce Asn-linked glycosylation sites elsewhere in the light chain, or even in the heavy chain variable region. [0065]
  • The representative embodiments described below are simply used to illustrate the invention. Those skilled in these arts will recognize that variations of the present materials fall within the broad generic scope of the claimed invention. The contents of all references mentioned herein are incorporated by reference. [0066]
  • EXAMPLE 1 Choice of Human Frameworks and Sequence Design for the Humanization of LL2 Monoclonal Antibody
  • By comparing the murine variable (V) region framework (FR) sequences of LL2 to that of human antibodies in the Kabat data base (Kabat et al., [0067] Sequences of Proteins of Immunological Interest, 5th ed., U.S. Department of Health and Human Services, U.S. Government Printing Office, Washington, D.C.), which is incorporated by reference, the human REI (FIG. 1A, Sequence ID No. 1) and EU (FIG. 1B, Sequence ID No. 2) sequences were found to exhibit the highest degree of sequence homology to the FRs of VK and VH domains of LL2, respectively. Therefore, the REI and EU FRs were selected as the human frameworks onto which the CDRs for LL2 VK and VH were grafted, respectively. The FR4 sequence of NEWM, however, rather than that of EU, was used to replace the EU FR4 sequence for the humanization of LL2 heavy chain. Based on the results of computer modeling studies (FIGS. 2A and 2B), murine FR residues having potential CDR contacts, which might affect the affinity and specificity of the resultant antibody, were retained in the design of the humanized FR sequences (FIG. 1).
  • Two versions of humanized heavy chain were constructed. In the first version (hLL2-1), the glutamine (Q) at amino acid position 5 (Kabat numbering) was introduced to include a PstI restriction site to facilitate its subcloning into the staging vector (FIG. 3). This murine residue was converted, by oligo-directed mutagenesis, to the human EU residue valine (V) in hLL2-2. It should be noted that in the original murine kappa chain variable sequence, a potential N-linked glycosylation site was identified at positions 18-20 (FIG. 1) and was used for carbohydrate addition. This glycosylation site was not included in the REI FR sequence used for LL2 light chain humanization. [0068]
  • See Example 3 for more oligonucleotide detail. [0069]
  • EXAMPLE 2 PCR Cloning and Sequence Elucidation for LL2 Heavy and Light Chain Variable Regions
  • The variable regions for both heavy (VH) and light (VK) chains of mLL2 (IgG2a) were obtained by PCR cloning using DNA primers as described in general above and in greater detail in Example 3, below. As PCR is prone to mutations, the variable region sequence of multiple individual clones for either the heavy or light chains was determined for six clones and confirmed to be identical prior to use for the construction of the chimeric antibody. [0070]
  • The PCR products for VK were subcloned into a pBR327-based staging vector, VKpBR, which contained an Ig promoter, a signal peptide sequence and convenient restriction sites to facilitate in-frame ligation of the VK PCR products (FIG. 3A). The PCR products for VH were subcloned into a similar pBluescript-based staging vector, VHpBS (FIG. 3B). [0071]
  • As noted above, at least six individual clones containing the respective PCR products were sequenced according to the method of Sanger et al., 1977, above. All were shown to bear identical sequences and their respective sequences were elucidated, as shown in FIG. 4A for LL2 VK (Sequence ID NO. 3) and in FIG. 4B for LL2 VH (Sequence ID NO. 4). No defective mutations were identified within the sequences encoding the VK and VH regions. Comparison of the PCR-amplified variable region sequences of LL2 with the Kabat database (Kabat et al., above) suggested that the VK and VH sequences of LL2 belong to [0072] subgroup 5 and 2B, respectively. Important residues such as Cys for intra-domain disulfide linkage were retained at appropriate positions.
  • In the FRI framework region of VK, an N-linked carbohydrate attachment site, Asn-Val-Ser, was identified at position 18-20 (FIG. 4A), suggesting that the VK of LL2 might be glycosylated. As will be detailed below, SDS-PAGE analysis under reducing conditions demonstrated that this Asn glycosylation site is indeed utilized for carbohydrate addition. The presence of the glycosylation site in the variable region does not, however, appear to affect the immunoreactivity of the antibody. A comparison of the immunoreactivity of mLL2 with that of cLL2 in a competitive RIA showed that the two antibodies have nearly identical activities. [0073]
  • EXAMPLE 3 PCR/Gene Synthesis of the Humanized V Genes
  • The designed sequence for the hLL2 VH domain, the construction of the hLL2 VH domain by long oligonucleotides and PCR, and the staging vector VHpBS containing the hLL2 VH domain are summarized in the sketch shown in FIG. 6. [0074]
  • For the construction of the hLL2 VH domain, oligo A (149-mer) and oligo B (140-mer) were synthesized on an automated CYCLONE PLUS™ DNA synthesizer (Milligen Bioresearch). [0075]
  • Oligo A (Sequence ID No. 7 below) represents the minus strand of the hLL2 VH domain complementary to nt 24 to 172. [0076]
  • Sequence ID No. 7 [0077]
    5′-TAT AAT CAT TCC TAG GAT TAA TGT ATC CAA TCC ATT
    CCA GAC CCT GTC CAG GTG CCT GCC TGA CCC AGT GCA
    GCC AGT AGC TAG TAA AGG TGT AGC CAG AAG CCT TGC
    AGG AGA CCT TCA CTG ATG ACC CAG GTT TCT TGA CTT
    CAG CC-3′
  • Oligo B (Sequence ID No. 8 below) represents the minus strand of the hLL2 VH domain complementary to nt 180 to 320. [0078]
  • Sequence ID No. 8 [0079]
    5′-CCC CAG TAG AAC GTA ATA TCC CTT GCA CAA AAA TAA
    AAT GCC GTG TCC TCA GAC CTC AGG CTG CTC AGC TCC
    ATG TAG GCT GTA TTG GTG GAT TCG TCT GCA GTT ATT
    GTG GCC TTG TCC TTG AAG TTC TGA TT-3′
  • Oligos A and B were cleaved from the support and deprotected by treatment with concentrated ammonium hydroxide. After the samples were vacuum-dried (SpeedVac, Savant, Farmingdale, N.Y.) and resuspended in 100 μl of water, incomplete oligomers (less than 100-mer) were removed by centrifugation through a CHROMOSPIN-100™ column (Clonetech, Palo Alto, Calif.) before the DNA oligomers were amplified by PCR. All flanking primers for the separate amplifications and PCR cloning of oligos A and B were purified by SDS-PAGE essentially according to the methods of Sambrook et al., 1989, above. From the CHROMASPIN-purified oligo A, 1 μl of sample stock was PCR-amplified in a reaction volume of 100 μl by adding 5 μl of 5 μM of oligo Sequence ID No. 9: [0080]
  • 5′-CCA GCT GCA GCA ATC AGG GGC TGA AGT CAA GAA ACC TG-3′[0081]
  • and oligo Sequence ID No. 10: [0082]
  • 5′-AAG TGG ATC CTA TAA TCA TTC CTA GGA TTA ATG-3′[0083]
  • in the presence of 10 μl of 10×PCR Buffer (500 mM KCl, 100 mM Tris.HCL buffer, pH 8.3, 15 mM MgCl[0084] 2) and 5 units of AMPLITAQ™ DNA polymerase (Perkin Elmer Cetus, Norwalk, Conn.). This reaction mixture was subjected to 30 cycles of PCR reaction consisting of denaturation at 94° C. for 1 minute, annealing at 50° C. for 1.5 minutes, and polymerization at 72° C. for 1.5 minutes.
  • Oligo B was PCR-amplified by the primer pairs Sequence ID No. 11: [0085]
  • 5′-TAA TCC TAG GAA TGA TTA TAC TGA GTA CAA TCA GAA CTT CAA GGA CCA G-3′[0086]
  • and Sequence ID No. 12: [0087]
  • 5′-GGA GAC GGT GAC CGT GGT GCC TTG GCC CCA GTA GAA CGT AGT AA-3′[0088]
  • under similar conditions. [0089]
  • Double-stranded PCR-amplified products for oligos A and B were gel-purified, restriction-digested with PstI/AvrII (PCR product of oligo A) and BstEII/AvrII (PCR product of oligo B), and sucloned into the complementary PstI/BstEII sites of the heavy chain staging vector, VHpBS. The humanized VH sequence was subcloned into the pG1g vector, resulting in the final human IgG1 heavy chain expression vector, hLL2pG1g. [0090]
  • For constructing the full length DNA of the humanized VK sequence, oligo E (150-mer) and oligo F (121-mer) were synthesized as described above. [0091]
  • Oligo E Sequence ID No. 13: [0092]
    5′-CCT AGT GGA TGC CCA GTA GAT CAG CAG TTT AGG TGC
    TTT CCC TGG TTT CTG GTG GTA CCA GGC CAA GTA GTT
    CTT GTG ATT TGC ACT GTA TAA AAC ACT TTG ACT GGA
    CTT ACA GCT CAT AGT GAC CCT ATC TCC AAC AGA TGC
    GCT CAG-3′
  • represents the minus strand of the humanized VK domain complementary to nt 31 to 180, and this sequence was PCR-amplified by oligo Sequence ID No. 14: [0093]
  • 5′-GAC AAG CTT CAG CTG ACC CAG TCT CCA TCA TCT CTG AGC GCA TCT GTT GGA G-3′[0094]
  • and oligo Sequence ID No. 15 [0095]
  • 5′-AGA GAA TCG CGA AGG GAC ACC AGA TTC CCT AGT GGA TGC CCA GTA-3′. [0096]
  • Oligo F Sequence ID No. 16: [0097]
    5′-GCA CCT TGG TCC CTC CAC CGA ACG TCC ACG AGG AGA
    GGT ATT GGT GAC AAT AAT ATG TTG CAA TGT CTT CTG
    GTT GAA GAG AGC TGA TGG TGA AAG TAA AAT CTG TCC
    CAG ATC CGC TGC C-3′
  • represents the minus strand of the humanized LL2 VK domain complementary to nt 208 to 327, and was PCR amplified by oligo Sequence ID No. 17: [0098]
  • 5′-GAC AAG CTT TCG CGA TTC TCT GGC AGC GGA TCT GGG ACA G-3′[0099]
  • and oligo Sequence ID No. 18: [0100]
  • 5′-GAC CGG CAG ATC TGC ACC TTG GTC CCT CCA CCG-3′. [0101]
  • Gel-purified PCR products for oligos E and F were restriction-digested with PvuII/NruI and NruI/BglII, respectively. The two PCR fragments E and F were then joined at the NruI site and ligated to the complementary PvuI/BcII sites of the light chain staging vector, VKpBR. The humanized VK sequence was subcloned into vector pKh to form the final human kappa chain expression vector, hLL2pKh. [0102]
  • To express the humanized antibodies, about 10 μg of linearized hLL2pKh and 20 μg of linearized hLL2pG1g were used to transfect 5×10[0103] 6 SP2/0 cells by electroporation. The transfectomas were selected with hygromycin at 500 μg/ml and secreted antibody was purified on a 1×3 cm column of protein A. After concentrating the purified antibody by Centricon 30 centifugation, antibody concentration was determined by ELISA. The final concentration of the antibody was adjusted to 1 mg/ml in PBS buffer containing 0.01% (w/v) sodium azide as a preservative.
  • In FIG. 1 (Sequence ID Nos. 1 and 2), there is compared the amino acid sequence between murine and humanized LL2 VK domains (FIG. 1A) and between murine and humanized LL2 VH domains (FIG. 1B). In the VK chain, human REI framework sequences were used for all FRs. In the VH chain, human EU framework sequences were used for FR 1-3, and NEWM sequences were used for FR-4. Only human FR sequences that are different from that of the mouse are shown. Asterisks indicate murine FR sequences that are different from that of the human FR at corresponding positions. Murine residues at these positions were retained in the humanized structure. CDRs are boxed. [0104]
  • In FIG. 4A (Sequence ID No. 3) there are shown the double stranded DNA and corresponding amino acid sequences (shown by single letter code) of the humanized LL2 VK domain. CDR 1-3 amino acid sequences are boxed. The corresponding display for VH is shown in FIG. 4B (Sequence ID No. 4). [0105]
  • In FIG. 5A (Sequence ID No. 5) and FIG. 5B (Sequence ID No. 6) there are shown double-stranded DNA sequences and amino acid sequences of humanized LL2 VK and LL2 VH, respectively. Amino acid sequences are shown by the single-letter code, and CDR amino acid sequences are boxed. [0106]
  • EXAMPLE 4 Construction, Expression and Purification of Chimeric LL2 Antibodies
  • The fragments containing the VK and VH sequences of LL2, together with the promoter and signal peptide sequences, were excised from LL2VKpBR and LL2VHpBS, respectively, by double restriction digestion with HindIII and BamHI. The about 600 bp VK fragments were then subcloned into the HindIII/BamHI site of a mammalian expression vector, pKh (FIG. 3A). pKh is a pSVhyg-based expression vector containing the genomic sequence of the human kappa constant region, an Ig enhancer, a kappa enhancer and the hygromycin-resistant gene. Similarly, the ca. 800 bp VH fragments were subcloned into the corresponding HindIII/BamHI site of pG1g (FIG. 3B), a pSVgpt-based expression vector carrying the genomic sequence of the human IgG1 constant region, an Ig enhancer and the xanthine-guanine phosphoribosyltransferase (gpt) gene. The final expression vectors are designated as LL2pKh and LL2pG1g, respectively. [0107]
  • The two plasmids were co-transfected into Sp2/0-Ag14 cells by electroporation and selected for hygromycin resistance. Supernatants from colonies surviving selection were monitored for chimeric antibody secretion by ELISA assay (see above). The transfection efficiency was approximately 1-10×10[0108] 6 cells. The antibody expression level, in a terminal culture, was found to vary in the range between <0.10 and 2.5 μg/ml.
  • FIG. 7 shows the results of analyzing protein A-purified mLL2 ([0109] lanes 4 and 7) and cLL2 (lanes 5 and 8) by SDS-PAGE under reducing and nonreducing conditions, respectively. HMW stands for high molecular weight protein markers, and LMW for light molecular weight markers. The light chains of both mLL2 and cLL2 (lanes 4 and 5) migrated primarily as a doublet band, with a higher than expected apparent molecular weight. As the human kappa constant region of cLL2 is known to contain no potential glycosylation site, it can be inferred that the potential glycosylation site identified in the FR1 region of LL2 VK domain was utilized.
  • FIG. 8 shows the results of analyzing different versions of hLL2 and cLL2 antibodies by SDS-PAGE under reducing and non-reducing conditions. As before, LMW and HMW are molecular weight markers. [0110] Lanes 3 and 6 are cLL2 antibodies. Lanes 4 and 7 are hLL2 with seven murine FR residues in the VH domain (hLL2-1). Lanes 5 and 8 are hLL2 with 6 murine FR residues in the VH domain (hLL2-2). The humanized light chains migrated more rapidly and as more discrete bands compared to chimeric light chains.
  • FIG. 9 shows the results of SDS-PAGE analysis on mix-and-match and cLL2 and hLL2 antibodies under both reducing and non-reducing conditions. [0111] Lanes 1 and 2 are molecular weight markers. Lanes 3 and 7 are cLL2. Lanes 4 and 8 are mix-and-match with a humanized light and chimeric heavy chain [(hL/cH)LL2]. Lanes 5 and 9 are chimeric light and humanized heavy (Version 1) chains [(cL/hH)LL2-1]. Lanes 6 and 10 are chimeric light and a humanized heavy (version 2) chains [(cL/hH)LL2-2]. The humanized LL2 version 1 contains 7 murine FR residues in the VH domain, while version 2 contains 6 murine FR residues in the VH domain. It is noteworthy that the position of the light chain of (hL/cH)LL2 (lane 4) is different from that of the others, suggesting that there is no carbohydrate attachment to the humanized LL2 light chain.
  • EXAMPLE 5 Binding of cLL2 Antibody to Raji Cell Surface Antigens
  • A competition cell binding assay was carried out to assess the immunoreactivity of cLL2 relative to the parent mLL2. Using [0112] 131I-labeled mLL2 (0.025 μg/ml) as a probe, Raji cells were incubated with the antibodies and the relative binding to the cells determined from the amount of cell-bound labeled mLL2 (see above). As shown by the competition assays described in FIG. 10, both mLL2 and cLL2 antibodies exhibited similar binding activities.
  • The results were confirmed by a second competition assay based on flow cytometry. Briefly, using Raji cells as before and varying the concentration of one antibody relative to other, as before, the amount of bound mLL2 or cLL2 was determined with FITC-labeled anti-mouse Fc or anti-human Fc antibodies followed by analysis using flow cytometry. [0113]
  • EXAMPLE 6 Binding of hLL2 Antibodies to Raji Cells
  • In experiments similar to those of Example 5, the antigen binding affinities of the three different combinations of mix-and-match or humanized LL2 were compared with that of cLL2 in the flow cytometry assay. [0114]
  • Briefly, 1 μg of cLL2, mix-and-match LL2, hLL2-1 or hLL2-2 antibodies were incubated with 10[0115] 8 Raji cells in the presence of varying concentrations of mLL2 F(ab′)2 fragments (as competitor) in a final volume of 100 μl of PBS buffer supplemented with 1% FCS and 0.01% sodium azide. The mixture was incubated for 30 minutes at 4° C., and washed three times with PBS to remove unbound antibodies. By taking advantage of the presence of human Fc portions in the antibodies, the binding levels of the antibodies were assessed by adding a 20× diluted FITC-labeled goat anti-human IgG1, Fc fragment-specific antibodies (Jackson ImmunoResearch, West Grove, Pa.). The cells were washed three times with PBS, and fluorescence intensities measured by a FACSCAN fluorescence activated cell sorter (Becton-Dickinson, Bedford, Mass.). The results are shown in FIG. 11A.
  • Using the same methods, cLL2 was compared to two versions of hLL2 (FIG. 11B). [0116]
  • The results shown in FIGS. 11A and B demonstrate that the immunoreactivity of cLL2 is similar or identical to that of humanized or mix-and-match antibodies. Taken together with the comparison of cLL2 with mLL2 (FIG. 10), the authenticity of the sequences for chimeric and humanized VK and VH obtained is established, and the functionality of cLL2 and hLL2 confirmed. [0117]
  • EXAMPLE 7 Internalization of mLL2 and cLL2 by Raji Cells
  • One of the unique characteristics of the LL2 antibody is its rapid internalization upon binding to Raji cells (Shih et al., 1994 above). Murine LL2 after internalization is likely to be rapidly transferred to the Golgi apparatus and from there to the lysosomes, the organelle responsible for the degradation of a wide variety of biochemicals (Keisari et al., [0118] Immunochem., 10: 565 (1973)).
  • Rates of antibody internalization were determined according to Opresko et al., 1987 above. The ratio of CPM[0119] intracellular/CPMsurface was determined as a function of time.
  • Rates of LL2 antibody internalization were determined by incubating radiolabeled LL2 antibody (1×10[0120] 6 cpm) with 0.5×106 Raji cells in 0.5 ml of DMEM buffer containing 1% human serum for 2 hrs. at 4° C. Excess human serum was included to saturate Raji cell surface Fc receptors in order to exclude or minimize non-antigen-specific internalization mediated through the Fc receptors. Unbound radiolabeled LL2 antibodies were removed from the cells by washing three times with 0.5 ml portions of DMEM at 4° C. Cells were then incubated at 37° C., and, at timed intervals, aliquots of the cell suspension were transferred to ice in order to stop internalization. The cells in these aliquots were isolated by centrifugation at 1,000×g for 5 mins. at 4° C., and surface bound radiolabeled LL2 stripped off cells with 1 ml of 0.1 M glycine acetate buffer, pH 3, for 8 mins. at 4° C. Radioactivity thus obtained (CPM surface) and radioactivity remaining in the cells (CPM intracellular) were determined. Rates of internalization were calculated from the slope of the plot of intracellular:surface radioactivity ratios as a function of time.
  • As shown in FIG. 12, mLL2, cLL2, cLL2Q and hLL2 antibodies were internalized at a similar rate (Ke=0.107 (mLL2) to 0.1221 (cLL2Q, NVT to QVT mutation). Those numbers suggested that approximately 50% of the surface-bound antibody could be internalized in 10 min. The results show that neither chimerization nor humanization nor deglycosylation by mutagenesis of mLL2 antibodies impair rates of internalization. [0121]
  • The pattern of internalization for mLL2, cLL2 and hLL2 was also monitored by fluorescence microscopy on a time-course basis using a FITC-labeled second antibody probe as described in the specification. Internalization of both antibodies was observed in at the earliest time point measurable. At 5 minutes, antibodies were seen both on the cell surface and internalized in areas immediately adjacent to the membrane as cytoplasmic micro-vesicles. At 15 min. post-incubation, the fine dots dispersed around the intramembrane began to merge into a group of granules, at locations believed to be the Golgi apparatus. As more antibodies were being internalized after 30 min. of incubation, redistribution of the grouped antibodies to scattered locations, probably the lysosomes in which the antibodies were degraded, was observed. At 2 hrs post-incubation, most of the antibodies were found inside the cell. Only strong surface staining was observed when LL2 was incubated for 20 min on ice. Both mLL2 and cLL2 were internalized with a similar pattern. The internalization of LL2 was associated specifically with antigen-antibody binding, as the irrelevant control humanized antibody demonstrated only dull surface staining. [0122]
  • A103 antibody (an IgG2a antibody that binds to the surface of all human epithelial cells but does not internalize efficiently (Mattes et al., [0123] Hybridoma, 2: 253 (1983)) showed strong membrane staining at up to 2 h, while the anti-transferrin receptor antibody (5F9) internalized rapidly, just as did LL2.
  • EXAMPLE 8 Role of Glycosylation Site in FRI Region of LL2 VK Sequence
  • Of particular inventive interest is the identification of an Asn-glycosylation site at position 18-20 within the FRI region of the LL2 NVT light chain sequence (FIG. 4A). As shown above, SDS-PAGE analysis under reducing condition suggests that the Asn glycosylation site is utilized for carbohydrate addition. [0124]
  • In this example, the influence of the carbohydrate moiety at position 18-20 on the functional activities of the light chains was examined. [0125]
  • Murine and chimeric LL2 light chains were treated with (+) or without (−) endoglycosidase F conventionally, and the antibody products examined by SDS-PAGE under reducing and non-reducing conditions (FIG. 13). There was no distinction between the antibody types as to electrophoretic behavior. In both cases, deglycosylation reduced the rate of migration of the light chain. [0126]
  • The effect of deglycosylation on the binding affinity to Raji cells of the mLL2 antibody is shown in FIG. 14. Removing carbohydrate by endoglycosidase F was without influence on the binding activity. [0127]
  • A mutation was introduced at position 18 of the light chain so that the Asn was replaced with Gin to produce LL2Q VK FR1. SDS-PAGE analyses demonstrated that the NVT to QVT mutation abolished glycosylation of the antibody. Comparison of the Raji cell binding affinity for cLL2 with and without light chain VK glycosylation demonstrated that the carbohydrate moiety was without influence on binding of the antibody to these cells. [0128]
  • It can be concluded that the presence of the carbohydrate site in the variable region does not affect the immunoreactivity of the antibody. Computer modeling studies suggested that the VK carbohydrate moiety in LL2 is remotely positioned from the CDRs and forms a “cap” over the bottom loops of the FR-associated B-barrels supporting the CDRs. [0129]
  • Humanization without inclusion of the original glycosylation site resulted in a CDR-grafted LL2 antibody with immunoreactivity comparable to that of its murine counterpart. [0130]
  • These characteristics indicate that the glycosylation site can be used for conjugating therapeutic or diagnostic agents to LL2 without compromising the ability of the antibody to bind and internalize in B-lymphoma or leukemia cells. [0131]
  • EXAMPLE 9 Conjugation of LL2 at its Carbohydrate-Bearing Site
  • The apparent lack of involvement of the variable region carbohydrate moiety in the functional activities of mLL2, cLL2 and hLL2 mAbs indicates that this moiety could profitably be used as the site of attachment of cytotoxic or detection agents such as radionuclides or toxins, and thereby avoid potential interference with the binding of the conjugate to a cell surface. [0132]
  • Using procedures described in Shih et al., U.S. Pat. No. 5,057,313 (which is incorporated by reference) for preparing antibody conjugates through an oxidized carbohydrate moiety of the antibody and a primary alkylamino group of a polymeric carrier to which are covalently one or more of a variety of drugs, toxins, chelators and detectable labels, a doxorubicin-dextran-LL2 antibody fragment devoid of appended glycans was produced containing multiple copies of the drug. The carbohydrate moieties of the cLL2 VK FR1 region involved were those covalently bound to the Asn glycosylation site. [0133]
  • In one synthesis, dextran (18-40 kDa) was converted to an amino dextran by oxidation of the dextran by NaIO[0134] 4, Schiff base formation with NH2—CH2—CHOH—CH2—NH2, and reduction with NaBH4. The amino dextran was then condensed with doxorubicin (DOX) in the presence of succinic anhydride and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide to produce DOX-aminodextran. The latter was then condensed with an aldehydic group on LL2 VK FR-1 produced by oxidizing the carbohydrate moiety of the antibody fragment with NaIO4.
  • In one preparation of DOX-LL2, the number of moles of DOX attached to dextran was 14 moles per mole dextran, and the number of moles of doxorubicin per mole F(ab′)2 was 8.9. The immunoreactivity in the Raji cell binding assay above was about 80% of control values. [0135]
  • This conjugation system is not limited to the mLL2 antibody. In a comparative study, 15-19 moles of DOX/mole of cLL2 were bound. [0136]
  • The conjugation possibilities are not limited to the use of a carrier dextran as in the example above. For example, the carbohydrate moiety of the LL2 VK FR1 region can be oxidized to produce aldehydic groups. These in turn can be reacted with an amino group on any drug to produce a Schiff base which, upon reduction, produces multiple copies of the drug stably linked to the antibody via alkyamine groups. [0137]
  • For example, where the drug is aminohexyl DTPA (a chelating agent), there is produced a LL2 covalently bound to a chelator. The chelator can be used to deliver to target tissues, for example, a radionuclide or paramagnetic metal ion, with a potential for diagnostic and therapeutic uses. DTPA-LL2 conjugates were produced containing 5.5 moles of the chelator/mole of antibody which, in turn, chelated 47.3% of Y-90 and 97.4% In-111 [0138]
  • It should be emphasized that the above-described examples merely describe several specific embodiments of the invention, and applicants do not intend to be limited as to scope of claims by these specific examples. [0139]
  • Applicants also incorporate by reference all publications and patents cited in the specification. [0140]

Claims (10)

We claim:
1. An immunoconjugate comprising an antibody or a fragment thereof specific for B-cells which is conjugated to Y-90 through a DTPA chelating agent.
2. The immunoconjugate of claim 1, wherein the DTPA chelating agent is aminohexyl DTPA.
3. The immunoconjugate of claim 1, wherein at least 47.3% of Y-90 is chelated.
4. The immunoconjugate of claim 1, wherein the immunoconjugate is internalizing.
5. The immunoconjugate of claim 4, wherein conjugation through the DPTA chelating agent to Y-90 does not reduce binding to B-cells or internalization function relative to the unconjugated antibody.
6. A method of treating B-cell lymphoma and lymphocytic leukemia comprising administering an immunoconjugate as claimed in claim 1 to a subject suffering from B-cell lymphoma or lymphocytic leukemia.
7. A method of treating B-cell lymphoma and lymphocytic leukemia comprising administering an immunoconjugate as claimed in claim 2 to a subject suffering from B-cell lymphoma or lymphocytic leukemia.
8. A method of treating B-cell lymphoma and lymphocytic leukemia comprising administering an immunoconjugate as claimed in claim 3 to a subject suffering from B-cell lymphoma or lymphocytic leukemia.
9. A method of treating B-cell lymphoma and lymphocytic leukemia comprising administering an immunoconjugate as claimed in claim 4 to a subject suffering from B-cell lymphoma or lymphocytic leukemia.
10. A method of treating B-cell lymphoma and lymphocytic leukemia comprising administering an immunoconjugate as claimed in claim 5 to a subject suffering from B-cell lymphoma or lymphocytic leukemia.
US10/446,689 1994-08-12 2003-05-29 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells Abandoned US20040013607A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/446,689 US20040013607A1 (en) 1994-08-12 2003-05-29 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US10/974,678 US20050106108A1 (en) 1994-08-12 2004-10-28 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US11/676,466 US20070172920A1 (en) 1994-08-12 2007-02-19 Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US28957694A 1994-08-12 1994-08-12
US08/690,102 US5789554A (en) 1994-08-12 1996-07-31 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US09/127,902 US6187287B1 (en) 1994-08-12 1998-08-03 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US09/741,843 US20020102254A1 (en) 1994-08-12 2000-12-22 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US10/446,689 US20040013607A1 (en) 1994-08-12 2003-05-29 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/741,843 Continuation US20020102254A1 (en) 1994-08-12 2000-12-22 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/974,678 Division US20050106108A1 (en) 1994-08-12 2004-10-28 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells

Publications (1)

Publication Number Publication Date
US20040013607A1 true US20040013607A1 (en) 2004-01-22

Family

ID=23112129

Family Applications (6)

Application Number Title Priority Date Filing Date
US08/690,102 Expired - Lifetime US5789554A (en) 1994-08-12 1996-07-31 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US09/127,902 Expired - Lifetime US6187287B1 (en) 1994-08-12 1998-08-03 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US09/741,843 Abandoned US20020102254A1 (en) 1994-08-12 2000-12-22 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US10/446,689 Abandoned US20040013607A1 (en) 1994-08-12 2003-05-29 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US10/974,678 Abandoned US20050106108A1 (en) 1994-08-12 2004-10-28 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US11/676,466 Abandoned US20070172920A1 (en) 1994-08-12 2007-02-19 Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US08/690,102 Expired - Lifetime US5789554A (en) 1994-08-12 1996-07-31 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US09/127,902 Expired - Lifetime US6187287B1 (en) 1994-08-12 1998-08-03 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US09/741,843 Abandoned US20020102254A1 (en) 1994-08-12 2000-12-22 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/974,678 Abandoned US20050106108A1 (en) 1994-08-12 2004-10-28 Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US11/676,466 Abandoned US20070172920A1 (en) 1994-08-12 2007-02-19 Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells

Country Status (10)

Country Link
US (6) US5789554A (en)
EP (1) EP0771208B1 (en)
JP (1) JP3053873B2 (en)
AT (1) ATE306930T1 (en)
AU (1) AU3272695A (en)
CA (1) CA2195557C (en)
DE (1) DE69534530T2 (en)
ES (1) ES2251723T3 (en)
IL (1) IL114909A (en)
WO (1) WO1996004925A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010094142A (en) * 2008-10-14 2010-04-30 Nikkiso Co Ltd Heat exchange device

Families Citing this family (392)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8771694B2 (en) * 1994-08-12 2014-07-08 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
EP0771208B1 (en) * 1994-08-12 2005-10-19 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells
US5874540A (en) * 1994-10-05 1999-02-23 Immunomedics, Inc. CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
WO1997034632A1 (en) 1996-03-20 1997-09-25 Immunomedics, Inc. Glycosylated humanized b-cell specific antibodies
DE59711533D1 (en) * 1996-07-26 2004-05-27 Aventis Pharma Gmbh Insulin derivatives with increased zinc binding
UA76934C2 (en) * 1996-10-04 2006-10-16 Chugai Pharmaceutical Co Ltd Reconstructed human anti-hm 1.24 antibody, coding dna, vector, host cell, method for production of reconstructed human antibody, pharmaceutical composition and drug for treating myeloma containing reconstructed human anti-hm 1.24 antibody
EP1007569B1 (en) * 1997-02-11 2009-04-08 Immunomedics, Inc. Stimulation of an immune response with antibodies labeled with the alpha-galactosyl epitope
US6183744B1 (en) * 1997-03-24 2001-02-06 Immunomedics, Inc. Immunotherapy of B-cell malignancies using anti-CD22 antibodies
US6306393B1 (en) 1997-03-24 2001-10-23 Immunomedics, Inc. Immunotherapy of B-cell malignancies using anti-CD22 antibodies
DE69831224T2 (en) * 1997-05-02 2006-03-23 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services IMMUNTOXINES CONTAINING AN ONC PROTEIN AGAINST POORED CELLS
CN1326565C (en) * 1997-10-14 2007-07-18 中外制药株式会社 Potentiator for antibody against lymphoid tumor
US20010033839A1 (en) * 1999-10-04 2001-10-25 Emilio Barbera-Guillem Vaccine and immunotherapy for solid nonlymphoid tumor and related immune dysregulation
US6224866B1 (en) 1998-10-07 2001-05-01 Biocrystal Ltd. Immunotherapy of B cell involvement in progression of solid, nonlymphoid tumors
US7696325B2 (en) 1999-03-10 2010-04-13 Chugai Seiyaku Kabushiki Kaisha Polypeptide inducing apoptosis
US20030099629A1 (en) * 1999-03-11 2003-05-29 Immunomedics, Inc. Recombinant onconase and chemical conjugates and fusion proteins of recombinant onconase
EP2039368A3 (en) 1999-04-30 2009-04-01 La Jolla Institute For Allergy And Immunology Methods for preventing reactivation of latent virus and controlling virus replication
US7829064B2 (en) 1999-05-10 2010-11-09 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods
US8383081B2 (en) 1999-05-10 2013-02-26 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods of use
US8119101B2 (en) 1999-05-10 2012-02-21 The Ohio State University Anti-CD74 immunoconjugates and methods of use
US6221597B1 (en) 1999-05-21 2001-04-24 Rosetta Inpharmatics, Inc. Essential genes of yeast as targets for antifungal agents, herbicides, insecticides and anti-proliferative drugs
US6200803B1 (en) 1999-05-21 2001-03-13 Rosetta Inpharmatics, Inc. Essential genes of yeast as targets for antifungal agents, herbicides, insecticides and anti-proliferative drugs
US6197517B1 (en) 1999-05-21 2001-03-06 Rosetta Inpharmatics, Inc. Essential genes of yeast as targets for antifungal agents, herbicides, insecticides and anti-proliferative drugs
US6451284B1 (en) * 1999-08-11 2002-09-17 Idec Pharmaceuticals Corporation Clinical parameters for determining hematologic toxicity prior to radioimmunotheraphy
TWI248365B (en) 1999-08-23 2006-02-01 Chugai Pharmaceutical Co Ltd HM1.24 antigen expression potentiators
WO2001022922A2 (en) * 1999-09-27 2001-04-05 The Regents Of The University Of California Engineering antibodies that bind irreversibly
US7118745B1 (en) 1999-09-27 2006-10-10 The Regents Of The University Of California Engineering antibodies that bind irreversibly
US20020028178A1 (en) * 2000-07-12 2002-03-07 Nabil Hanna Treatment of B cell malignancies using combination of B cell depleting antibody and immune modulating antibody related applications
JP2004500412A (en) * 2000-03-31 2004-01-08 アイデック ファーマスーティカルズ コーポレイション Combination of anti-cytokine antibody or antagonist and anti-CD20 for treatment of B-cell lymphoma
AU6836301A (en) 2000-06-20 2002-01-02 Idec Pharma Corp Treatment of b-cell associated diseases such as malignancies and autoimmune diseases using a cold anti-cd20 antibody/radiolabeled anti-cd22 antibody combination
AU2001270252B2 (en) * 2000-07-03 2007-02-08 Catalent Pharma Solutions, Llc Expression vectors
US20030224415A1 (en) * 2001-06-29 2003-12-04 Gala Design, Inc. Selection free growth of host cells containing multiple integrating vectors
US20040235173A1 (en) * 2000-07-03 2004-11-25 Gala Design, Inc. Production of host cells containing multiple integrating vectors by serial transduction
AU2001271614B2 (en) 2000-07-03 2007-05-31 Catalent Pharma Solutions, Llc Host cells containing multiple integrating vectors
RU2408606C2 (en) 2000-10-20 2011-01-10 Тугаи Сейяку Кабусики Кайся Compound-agonist of thrombopoietin
US7090843B1 (en) * 2000-11-28 2006-08-15 Seattle Genetics, Inc. Recombinant anti-CD30 antibodies and uses thereof
US20030211107A1 (en) * 2002-01-31 2003-11-13 Kandasamy Hariharan Use of CD23 antagonists for the treatment of neoplastic disorders
CA2436180C (en) 2001-01-31 2011-11-08 Idec Pharmaceutical Corporation Immunoregulatory antibodies and uses thereof
US20070065436A1 (en) * 2001-01-31 2007-03-22 Biogen Idec Inc. Anti-cd80 antibody having adcc activity for adcc mediated killing of b cell lymphoma cells alone or in combination with other therapies
US20030103971A1 (en) * 2001-11-09 2003-06-05 Kandasamy Hariharan Immunoregulatory antibodies and uses thereof
US20020159996A1 (en) * 2001-01-31 2002-10-31 Kandasamy Hariharan Use of CD23 antagonists for the treatment of neoplastic disorders
WO2002064159A1 (en) 2001-02-07 2002-08-22 Chugai Seiyaku Kabushiki Kaisha Remedies for tumor in hematopoietic organs
US20030118585A1 (en) * 2001-10-17 2003-06-26 Agy Therapeutics Use of protein biomolecular targets in the treatment and visualization of brain tumors
US7321026B2 (en) * 2001-06-27 2008-01-22 Skytech Technology Limited Framework-patched immunoglobulins
RU2004127458A (en) 2002-02-14 2005-10-27 Иммуномедикс, Инк. (Us) ANTI-CD20 ANTIBODIES, THEIR HYBRID PROTEINS AND WAYS OF THEIR USE
US8287864B2 (en) * 2002-02-14 2012-10-16 Immunomedics, Inc. Structural variants of antibodies for improved therapeutic characteristics
US20040001828A1 (en) * 2002-02-21 2004-01-01 Joseph Tuscano Treatment methods using anti-CD22 antibodies
ES2350477T5 (en) 2002-03-01 2020-05-06 Immunomedics Inc Internalization of anti-CD74 antibodies and methods of use
EP1487879B1 (en) 2002-03-01 2012-12-26 Immunomedics, Inc. Bispecific antibody point mutations for enhancing rate of clearance
EP2287201B1 (en) 2002-03-01 2018-12-12 Immunomedics, Inc. RS7 antibodies
US9770517B2 (en) 2002-03-01 2017-09-26 Immunomedics, Inc. Anti-Trop-2 antibody-drug conjugates and uses thereof
US20160279239A1 (en) 2011-05-02 2016-09-29 Immunomedics, Inc. Subcutaneous administration of anti-cd74 antibody for systemic lupus erythematosus and autoimmune disease
US20060099142A1 (en) * 2002-03-14 2006-05-11 Delaney Allen D Cancer associated araf1 protein kinase and its uses
US20030180292A1 (en) * 2002-03-14 2003-09-25 Idec Pharmaceuticals Treatment of B cell malignancies using anti-CD40L antibodies in combination with anti-CD20 antibodies and/or chemotherapeutics and radiotherapy
US7384738B2 (en) * 2002-03-28 2008-06-10 Bremel Robert D Retrovirus-based genomic screening
US20040038304A1 (en) * 2002-03-28 2004-02-26 Gala Design, Inc. Antibody libraries
GB0210121D0 (en) 2002-05-02 2002-06-12 Celltech R&D Ltd Biological products
PL224150B1 (en) 2002-05-02 2016-11-30 Wyeth Corp Composition containing drug conjugate including the calicheamicin derivatives and the antibody, and the pharmaceutical composition containing it
WO2003106497A1 (en) * 2002-06-14 2003-12-24 Immunomedics, Inc. Monoclonal antibody pam4 and its use for diagnosis and therapy of pancreatic cancer
CN1675245B (en) * 2002-06-14 2011-01-12 免疫医疗公司 Monoclonal antibody hPAM4
US20060024289A1 (en) * 2002-10-02 2006-02-02 Ruggles Sandra W Cleavage of VEGF and VEGF receptor by wild-type and mutant proteases
US7939304B2 (en) * 2002-10-02 2011-05-10 Catalyst Biosciences, Inc. Mutant MT-SP1 proteases with altered substrate specificity or activity
DE60324700D1 (en) 2002-10-11 2008-12-24 Chugai Pharmaceutical Co Ltd CELL TOD INDUCTIVE ACTIVE SUBSTANCE
US7429382B2 (en) 2002-10-16 2008-09-30 Corixa Corporation Antibodies that bind cell-associated CA 125/O772P and methods of use thereof
US8034831B2 (en) * 2002-11-06 2011-10-11 Celgene Corporation Methods for the treatment and management of myeloproliferative diseases using 4-(amino)-2-(2,6-Dioxo(3-piperidyl)-isoindoline-1,3-dione in combination with other therapies
US7563810B2 (en) * 2002-11-06 2009-07-21 Celgene Corporation Methods of using 3-(4-amino-1-oxo-1,3-dihydroisoindol-2-yl)-piperidine-2,6-dione for the treatment and management of myeloproliferative diseases
US8420086B2 (en) 2002-12-13 2013-04-16 Immunomedics, Inc. Camptothecin conjugates of anti-CD22 antibodies for treatment of B cell diseases
US7534427B2 (en) 2002-12-31 2009-05-19 Immunomedics, Inc. Immunotherapy of B cell malignancies and autoimmune diseases using unconjugated antibodies and conjugated antibodies and antibody combinations and fusion proteins
US7820787B2 (en) * 2003-01-23 2010-10-26 The Regents Of The University Of California Multi-functional antibodies
ATE517638T1 (en) * 2003-01-31 2011-08-15 Immunomedics Inc METHOD AND PREPARATIONS FOR ADMINISTERING THERAPEUTIC AND DIAGNOSTIC AGENTS
JP2004279086A (en) 2003-03-13 2004-10-07 Konica Minolta Holdings Inc Radiation image conversion panel and method for manufacturing it
WO2004087763A1 (en) * 2003-03-31 2004-10-14 Chugai Seiyaku Kabushiki Kaisha Modified antibody against cd22 and utilization thereof
WO2005004809A2 (en) 2003-07-01 2005-01-20 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
US7902338B2 (en) 2003-07-31 2011-03-08 Immunomedics, Inc. Anti-CD19 antibodies
WO2005012493A2 (en) * 2003-07-31 2005-02-10 Immunomedics, Inc. Anti-cd19 antibodies
NZ547279A (en) 2003-10-22 2008-04-30 Keck Graduate Inst Methods of synthesizing heteromultimeric polypeptides in yeast using a haploid mating strategy
MXPA06005104A (en) * 2003-11-05 2007-01-25 Palingen Inc Enhanced b cell cytotoxicity of cdim binding antibody.
EP2962699A3 (en) 2003-12-01 2016-04-06 Immunomedics Inc. Improved method for preparing conjugates of proteins and chelating agents
TW200530266A (en) * 2003-12-12 2005-09-16 Chugai Pharmaceutical Co Ltd Method of reinforcing antibody activity
AU2004297109A1 (en) * 2003-12-12 2005-06-23 Chugai Seiyaku Kabushiki Kaisha Cell death inducing agent
TW200530269A (en) * 2003-12-12 2005-09-16 Chugai Pharmaceutical Co Ltd Anti-Mpl antibodies
WO2005056602A1 (en) * 2003-12-12 2005-06-23 Chugai Seiyaku Kabushiki Kaisha Method of screening modified antibody having agonistic activity
US20050221429A1 (en) * 2004-01-16 2005-10-06 Cardinal Health Pts, Llc Host cells containing multiple integrating vectors comprising an amplifiable marker
US8883160B2 (en) 2004-02-13 2014-11-11 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for therapeutic and diagnostic use
US8652484B2 (en) 2004-02-13 2014-02-18 Immunomedics, Inc. Delivery system for cytotoxic drugs by bispecific antibody pretargeting
US9481878B2 (en) 2004-02-13 2016-11-01 Immunomedics, Inc. Compositions and methods of use of immunotoxins comprising ranpirnase (Rap) show potent cytotoxic activity
US20110064754A1 (en) * 2005-03-03 2011-03-17 Center For Molecular Medicine And Immunology Immunoconjugates Comprising Poxvirus-Derived Peptides and Antibodies Against Antigen-Presenting Cells for Subunit-Based Poxvirus Vaccines
US8491914B2 (en) * 2004-02-13 2013-07-23 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for delivery of interference RNA
US8551480B2 (en) 2004-02-13 2013-10-08 Immunomedics, Inc. Compositions and methods of use of immunotoxins comprising ranpirnase (Rap) show potent cytotoxic activity
US9550838B2 (en) 2004-02-13 2017-01-24 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for therapeutic and diagnostic use
US8435539B2 (en) * 2004-02-13 2013-05-07 Immunomedics, Inc. Delivery system for cytotoxic drugs by bispecific antibody pretargeting
US20110020273A1 (en) * 2005-04-06 2011-01-27 Ibc Pharmaceuticals, Inc. Bispecific Immunocytokine Dock-and-Lock (DNL) Complexes and Therapeutic Use Thereof
ES2741574T3 (en) 2004-03-31 2020-02-11 Massachusetts Gen Hospital Method to determine the response of cancer to treatments directed at the epidermal growth factor receptor
JP4799405B2 (en) * 2004-04-09 2011-10-26 中外製薬株式会社 Cell death inducer
JP5629423B2 (en) * 2004-04-12 2014-11-19 カタリスト バイオサイエンシーズ, インコーポレイテッド Cleavage of VEGF and VEGF receptors by wild type and mutant MT-SP1
MXPA06014069A (en) * 2004-06-04 2007-04-25 Genentech Inc Method for treating multiple sclerosis.
TW200608994A (en) * 2004-06-04 2006-03-16 Genentech Inc Method for treating lupus
CN105820249A (en) * 2004-07-09 2016-08-03 中外制药株式会社 Anti-glypican 3 antibody
WO2006012508A2 (en) * 2004-07-22 2006-02-02 Genentech, Inc. Method of treating sjögren's syndrome
PL2213683T3 (en) 2004-08-04 2013-10-31 Mentrik Biotech Llc Variant Fc regions
CA2580271A1 (en) * 2004-10-05 2006-04-20 Genentech, Inc. Method for treating vasculitis
WO2006084092A2 (en) * 2005-02-03 2006-08-10 Raven Biotechnologies, Inc. Antibodies to oncostatin m receptor
US10058621B2 (en) 2015-06-25 2018-08-28 Immunomedics, Inc. Combination therapy with anti-HLA-DR antibodies and kinase inhibitors in hematopoietic cancers
US9707302B2 (en) 2013-07-23 2017-07-18 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
AU2006218454B2 (en) 2005-03-03 2011-11-17 Immunomedics, Inc. Humanized L243 antibodies
US20160355591A1 (en) 2011-05-02 2016-12-08 Immunomedics, Inc. Subcutaneous anti-hla-dr monoclonal antibody for treatment of hematologic malignancies
TWI671403B (en) 2005-03-31 2019-09-11 中外製藥股份有限公司 Method for controlling controlled assembly of polypeptide
EP1870458B1 (en) 2005-03-31 2018-05-09 Chugai Seiyaku Kabushiki Kaisha sc(Fv)2 STRUCTURAL ISOMERS
US8475794B2 (en) 2005-04-06 2013-07-02 Ibc Pharmaceuticals, Inc. Combination therapy with anti-CD74 antibodies provides enhanced toxicity to malignancies, Autoimmune disease and other diseases
US8349332B2 (en) 2005-04-06 2013-01-08 Ibc Pharmaceuticals, Inc. Multiple signaling pathways induced by hexavalent, monospecific and bispecific antibodies for enhanced toxicity to B-cell lymphomas and other diseases
US20090022687A1 (en) * 2005-05-18 2009-01-22 Chugai Seiyaku Kabushiki Kaisha Novel Pharmaceuticals That Use Anti-HLA Antibodies
US7601335B2 (en) * 2005-05-20 2009-10-13 Genentech, Inc. Pretreatment of a biological sample from an autoimmune disease subject
CN101237890A (en) 2005-06-10 2008-08-06 中外制药株式会社 Stabilizer for protein preparation comprising meglumine and use thereof
US9241994B2 (en) 2005-06-10 2016-01-26 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical compositions containing sc(Fv)2
CA2610839C (en) 2005-06-14 2019-06-25 Amgen Inc. Self-buffering protein formulations
EP1893647A2 (en) 2005-06-23 2008-03-05 MedImmune, Inc. Antibody formulations having optimized aggregation and fragmentation profiles
AU2006275475A1 (en) * 2005-07-29 2007-02-08 Amgen Inc. Formulations that inhibit protein aggregation
US20070087005A1 (en) 2005-10-14 2007-04-19 Lazar Gregory A Anti-glypican-3 antibody
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
EP2674440B1 (en) 2005-12-16 2019-07-03 IBC Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US8389688B2 (en) 2006-03-06 2013-03-05 Aeres Biomedical, Ltd. Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
EP2639242A3 (en) * 2006-03-06 2013-10-16 MedImmune, Inc. Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
KR100701923B1 (en) 2006-03-15 2007-03-30 주식회사 녹십자 - monoclonal antibody against b-lymphoma and hybridoma cell line producing the same
JP5144499B2 (en) 2006-03-31 2013-02-13 中外製薬株式会社 Antibody modification method for purifying bispecific antibodies
KR101454508B1 (en) * 2006-05-30 2014-11-04 제넨테크, 인크. Antibodies and immunoconjugates and uses therefor
KR20170014023A (en) 2006-07-05 2017-02-07 카탈리스트 바이오사이언시즈, 인코포레이티드 Protease screening methods and proteases identified thereby
AU2007273507A1 (en) * 2006-07-13 2008-01-17 Chugai Seiyaku Kabushiki Kaisha Cell death inducer
BRPI0717902A2 (en) 2006-12-01 2013-10-29 Medarex Inc "HUMAN MONOCLONAL ANTIBODY ISOLATED, COMPOSITION, ASSOCIATED ANTIBODY-MOLLECLE PARTNERSHIP, IMMUNOCOUGHTED, ISOLATED NUCLEIC ACID MOLECULES, EXPRESSION VECTOR, HOSPEDIC CELL FOR PREPARING A CDT FOR THE PREPARATION OF A CD22 AND METHOD FOR TREATING INFLAMMATORY DISEASE OR SELF-IMMUNEING AN INDIVIDUAL "
EP2377527B1 (en) 2007-01-22 2013-07-17 Genentech, Inc. Polyelectrolyte precipitation and purification of antibodies
CL2008000719A1 (en) * 2007-03-12 2008-09-05 Univ Tokushima Chugai Seiyaku THERAPEUTIC AGENT FOR CANCER RESISTANT TO CHEMOTHERAPEUTIC AGENTS THAT UNDERSTAND AN ANTIBODY THAT RECOGNIZES IT CLASS I AS ACTIVE INGREDIENT; PHARMACEUTICAL COMPOSITION THAT INCLUDES SUCH ANTIBODY; AND METHOD TO TREAT CANCER RESISTANT TO
US7960139B2 (en) 2007-03-23 2011-06-14 Academia Sinica Alkynyl sugar analogs for the labeling and visualization of glycoconjugates in cells
TWI561530B (en) 2007-05-21 2016-12-11 Alderbio Holdings Llc Antibodies to il-6 and use thereof
US8062864B2 (en) 2007-05-21 2011-11-22 Alderbio Holdings Llc Nucleic acids encoding antibodies to IL-6, and recombinant production of anti-IL-6 antibodies
JP5570989B2 (en) * 2007-08-31 2014-08-13 アムジエン・インコーポレーテツド Solid protein formulation
AU2008304111B2 (en) 2007-09-27 2014-04-24 Amgen Inc. Pharmaceutical formulations
EP3381445B1 (en) 2007-11-15 2023-10-25 Amgen Inc. Aqueous formulation of antibody stablised by antioxidants for parenteral administration
JP2011507897A (en) * 2007-12-21 2011-03-10 ジェネンテック, インコーポレイテッド Treatment of patients with rituximab-resistant rheumatoid arthritis
MX2010011955A (en) 2008-04-29 2011-01-21 Abbott Lab Dual variable domain immunoglobulins and uses thereof.
US20090269339A1 (en) * 2008-04-29 2009-10-29 Genentech, Inc. Responses to immunizations in rheumatoid arthritis patients treated with a cd20 antibody
NZ589436A (en) 2008-06-03 2012-12-21 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
NZ590074A (en) 2008-07-08 2012-12-21 Abbott Lab Prostaglandin e2 dual variable domain immunoglobulins and uses thereof
DK2318832T3 (en) 2008-07-15 2014-01-20 Academia Sinica Glycan arrays on PTFE-like aluminum coated slides and related methods
UA108735C2 (en) * 2008-07-21 2015-06-10 STRUCTURAL OPTIONS OF ANTIBODIES TO IMPROVE THERAPEUTIC CHARACTERISTICS
TW201014605A (en) * 2008-09-16 2010-04-16 Genentech Inc Methods for treating progressive multiple sclerosis
AU2009323007B2 (en) 2008-11-25 2016-01-28 H. Lundbeck A/S. Antagonists of IL-6 to prevent or treat thrombosis
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
DK3903829T3 (en) 2009-02-13 2023-06-26 Immunomedics Inc IMMUNE CONJUGATES WITH AN INTRACELLULAR CLEAVABLE BOND
WO2010096394A2 (en) 2009-02-17 2010-08-26 Redwood Biosciences, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
SG173705A1 (en) 2009-03-05 2011-09-29 Abbott Lab Il-17 binding proteins
CA2772628A1 (en) * 2009-09-01 2011-03-10 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
EP4134095A1 (en) 2009-09-15 2023-02-15 The Board of Trustees of the Leland Stanford Junior University Synergistic anti-cd47 therapy for hematologic cancers
IN2012DN01663A (en) 2009-09-16 2015-06-05 Immunomedics Inc
US20110070227A1 (en) * 2009-09-18 2011-03-24 Anna-Marie Novotney-Barry Treatment of Autoimmune and Inflammatory Diseases
AR078651A1 (en) * 2009-10-15 2011-11-23 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
CA2778105C (en) 2009-10-23 2019-04-02 Amgen Inc. Vial adapter and system
UY32979A (en) * 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
CA2782194C (en) 2009-12-02 2018-01-16 Immunomedics, Inc. Combination of radiolabelled antibodies (rait) and antibody-drug conjugates (adc) for treatment of pancreatic cancer
US11377485B2 (en) 2009-12-02 2022-07-05 Academia Sinica Methods for modifying human antibodies by glycan engineering
EP2523680A4 (en) * 2010-01-11 2013-06-19 Ct Molecular Med & Immunology Enhanced cytotoxicity of anti-cd74 and anti-hla-dr antibodies with interferon-gamma
KR20130009760A (en) 2010-02-10 2013-01-23 이뮤노젠 아이엔씨 Cd20 antibodies and uses thereof
AU2011230537C1 (en) 2010-03-26 2018-08-02 Trustees Of Dartmouth College Vista regulatory T cell mediator protein, vista binding agents and use thereof
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
WO2011130332A1 (en) 2010-04-12 2011-10-20 Academia Sinica Glycan arrays for high throughput screening of viruses
EP2568976B1 (en) 2010-05-10 2015-09-30 Academia Sinica Zanamivir phosphonate congeners with anti-influenza activity and determining oseltamivir susceptibility of influenza viruses
AU2011252883B2 (en) 2010-05-14 2015-09-10 Abbvie Inc. IL-1 binding proteins
KR102513760B1 (en) 2010-06-07 2023-03-27 암젠 인크 Drug delivery device
US20130189268A1 (en) 2010-06-22 2013-07-25 Precision Biologics, Inc. Colon and pancreas cancer specific antigens and antibodies
UY33492A (en) 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
AU2011285852B2 (en) 2010-08-03 2014-12-11 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2012019168A2 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
KR20130139884A (en) 2010-08-26 2013-12-23 애브비 인코포레이티드 Dual variable domain immunoglobulins and uses thereof
US9068014B2 (en) 2010-09-23 2015-06-30 Precision Biologics, Inc. Colon and pancreas cancer peptidomimetics
PL3590949T3 (en) 2010-10-01 2022-08-29 Modernatx, Inc. Ribonucleic acids containing n1-methyl-pseudouracils and uses thereof
JP2013541594A (en) 2010-11-08 2013-11-14 ジェネンテック, インコーポレイテッド Anti-IL-6 receptor antibody administered subcutaneously
CA2818813C (en) 2010-11-23 2020-10-06 Alder Biopharmaceuticals, Inc. Anti-il-6 antibodies for the treatment of oral mucositis
US9783602B2 (en) 2010-12-01 2017-10-10 Alderbio Holdings Llc Anti-NGF compositions and use thereof
RU2627171C2 (en) 2010-12-21 2017-08-03 Эббви Инк. Il-1 alpha and beta bispecific immunoglobulins with double variable domains and their application
CN103415621A (en) 2011-01-14 2013-11-27 雷德伍德生物科技股份有限公司 Aldehyde-tagged immunoglobulin polypeptides and method of use thereof
AU2012236573B2 (en) 2011-03-31 2016-06-02 Amgen Inc. Vial adapter and system
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
CA3021845C (en) 2011-04-20 2022-03-29 Amgen Inc. Autoinjector apparatus
CA2831572C (en) 2011-05-02 2019-11-26 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
LT2710114T (en) 2011-05-20 2021-12-10 H. Lundbeck A/S High-purity production of multi-subunit proteins such as antibodies in transformed microbes such as pichia pastoris
EP2709662B1 (en) 2011-05-20 2019-07-31 AlderBio Holdings LLC Use of anti-cgrp or anti-cgrp-r antibodies or antibody fragments to treat or prevent chronic and acute forms of diarrhea
NZ732970A (en) 2011-05-20 2022-08-26 H Lundbeck As Use of anti-cgrp antibodies and antibody fragments to prevent or inhibit photophobia or light aversion in subjects in need thereof, especially migraine sufferers
TR201904088T4 (en) 2011-05-20 2019-05-21 Alderbio Holdings Llc Anti-CGRP compounds and their uses.
EP2714738B1 (en) 2011-05-24 2018-10-10 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
MX347514B (en) 2011-05-25 2017-04-28 Innate Pharma Sa Anti-kir antibodies for the treatment of inflammatory disorders.
CA2845579C (en) 2011-08-19 2020-03-24 Alderbio Holdings Llc Multi-copy strategy for high-titer and high-purity production of multi-subunit proteins such as antibodies in transformed microbes such as pichia pastoris
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RS62993B1 (en) 2011-10-03 2022-03-31 Modernatx Inc Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
ES2675035T3 (en) 2011-10-14 2018-07-05 Amgen, Inc Injector and assembly method
US8999331B2 (en) 2011-10-24 2015-04-07 Abbvie Inc. Immunobinders directed against sclerostin
US9757458B2 (en) 2011-12-05 2017-09-12 Immunomedics, Inc. Crosslinking of CD22 by epratuzumab triggers BCR signaling and caspase-dependent apoptosis in hematopoietic cancer cells
WO2013085893A1 (en) 2011-12-05 2013-06-13 Immunomedics, Inc. Therapeutic use of anti-cd22 antibodies for inducing trogocytosis
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
WO2013096455A1 (en) 2011-12-20 2013-06-27 Dana-Farber Cancer Institute, Inc. Methods for diagnosing and treating oncogenic kras-associated cancer
TW201333035A (en) 2011-12-30 2013-08-16 Abbvie Inc Dual specific binding proteins directed against IL-13 and/or IL-17
JP2015513914A (en) 2012-04-02 2015-05-18 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. Modified polynucleotides for the production of secreted proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
JP2015516984A (en) * 2012-04-26 2015-06-18 バイオアトラ、エルエルシー Anti-CD22 antibody
US10138479B2 (en) 2012-05-24 2018-11-27 Dana-Farber Cancer Institute, Inc. Targeting the glutamine to pyruvate pathway for treatment of oncogenic Kras-associated cancer
EP2859017B1 (en) 2012-06-08 2019-02-20 Sutro Biopharma, Inc. Antibodies comprising site-specific non-natural amino acid residues, methods of their preparation and methods of their use
US9890215B2 (en) 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
EP3421486B1 (en) 2012-06-22 2023-09-27 The Trustees Of Dartmouth College Novel vista-ig constructs and the use of vista-ig for treatment of autoimmune, allergic and inflammatory disorders
AR091755A1 (en) 2012-07-12 2015-02-25 Abbvie Inc PROTEINS OF UNION TO IL-1
WO2014014518A1 (en) 2012-07-18 2014-01-23 Dana-Farber Cancer Institute, Inc. Methods for treating, preventing and predicting risk of developing breast cancer
EP3539563A1 (en) 2012-07-19 2019-09-18 Redwood Bioscience, Inc. Antibody specific for cd22 and methods of use thereof
CA2874864C (en) 2012-08-14 2023-02-21 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
CA2880701A1 (en) 2012-08-18 2014-02-27 Academia Sinica Cell-permeable probes for identification and imaging of sialidases
WO2014031762A1 (en) 2012-08-21 2014-02-27 Academia Sinica Benzocyclooctyne compounds and uses thereof
EP3584255B1 (en) 2012-08-31 2022-02-16 Sutro Biopharma, Inc. Modified amino acids comprising an azido group
CN109793893B (en) 2012-09-07 2023-05-26 达特茅斯大学理事会 VISTA modulators for diagnosis and treatment of cancer
EA201791717A1 (en) 2012-09-07 2018-03-30 Кохерус Байосайенсис, Инк. STABLE WATER COMPOSITIONS ADALIMUMAB
LT2906251T (en) 2012-10-12 2017-12-11 Adc Therapeutics Sa Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
KR20180008921A (en) 2012-11-01 2018-01-24 애브비 인코포레이티드 Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
JP2015535464A (en) 2012-11-21 2015-12-14 アムジエン・インコーポレーテツド Drug delivery device
SI2922554T1 (en) 2012-11-26 2022-06-30 Modernatx, Inc. Terminally modified rna
WO2015012904A2 (en) 2012-12-13 2015-01-29 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
US9492566B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
US10137196B2 (en) 2012-12-13 2018-11-27 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10413539B2 (en) 2012-12-13 2019-09-17 Immunomedics, Inc. Therapy for metastatic urothelial cancer with the antibody-drug conjugate, sacituzumab govitecan (IMMU-132)
HUE057977T2 (en) 2012-12-13 2022-06-28 Immunomedics Inc Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
US9931417B2 (en) 2012-12-13 2018-04-03 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10744129B2 (en) 2012-12-13 2020-08-18 Immunomedics, Inc. Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting Trop-2
US10206918B2 (en) 2012-12-13 2019-02-19 Immunomedics, Inc. Efficacy of anti-HLA-DR antiboddy drug conjugate IMMU-140 (hL243-CL2A-SN-38) in HLA-DR positive cancers
WO2017004144A1 (en) 2015-07-01 2017-01-05 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
KR20150119848A (en) 2012-12-21 2015-10-26 바이오얼라이언스 씨.브이. Hydrophilic self-immolative linkers and conjugates thereof
TWI693073B (en) 2012-12-21 2020-05-11 日商中外製藥股份有限公司 Therapeutic agent for GPC3 target is the effective therapeutic agent for GPC3 target administered to patients
US9938344B2 (en) 2012-12-28 2018-04-10 Precision Biologics, Inc. Humanized monoclonal antibodies and methods of use for the diagnosis and treatment of colon and pancreas cancer
US10092703B2 (en) 2013-03-15 2018-10-09 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
EP3597664A3 (en) 2013-03-15 2020-03-11 Alder Biopharmaceuticals, Inc. Temperature shift for high yield expression of polypeptides in yeast and other transformed cells
CA2904448A1 (en) 2013-03-15 2014-09-18 Tariq Ghayur Dual specific binding proteins directed against il-1.beta. and/or il-17
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EA038918B1 (en) 2013-03-15 2021-11-09 Зинджения, Инк. Peptide binding an epidermal growth factor receptor, multispecific complexes comprising peptide and antibodies and use thereof
CA2904661C (en) 2013-03-15 2022-03-15 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
AR095615A1 (en) 2013-03-15 2015-10-28 Alder Biopharmaceuticals Inc PURIFICATION OF ANTIBODIES AND PURITY MONITORING
JP6670740B2 (en) 2013-03-22 2020-03-25 アムジエン・インコーポレーテツド Injector and assembly method
EP3013365B1 (en) 2013-06-26 2019-06-05 Academia Sinica Rm2 antigens and use thereof
WO2014210564A1 (en) 2013-06-27 2014-12-31 Academia Sinica Glycan conjugates and use thereof
EP3016684B1 (en) 2013-07-03 2022-02-23 H. Lundbeck A/S Regulation of glucose metabolism using anti-cgrp antibodies
ES2865473T3 (en) 2013-07-10 2021-10-15 Sutro Biopharma Inc Antibodies Comprising Multiple Site-Specific Unnatural Amino Acid Residues, Methods for Their Preparation, and Methods of Use
US11253606B2 (en) 2013-07-23 2022-02-22 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, Bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US9782476B2 (en) 2013-09-06 2017-10-10 Academia Sinica Human iNKT cell activation using glycolipids with altered glycosyl groups
EP3050896B1 (en) 2013-09-27 2021-07-07 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
WO2015050844A1 (en) 2013-10-01 2015-04-09 Dana-Farber Cancer Institute, Inc. Methods of treating cancer with atovaquone-related compounds
EP3052521A1 (en) 2013-10-03 2016-08-10 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
US9840493B2 (en) 2013-10-11 2017-12-12 Sutro Biopharma, Inc. Modified amino acids comprising tetrazine functional groups, methods of preparation, and methods of their use
KR102458637B1 (en) 2013-10-24 2022-10-24 암겐 인코포레이티드 Injector and method of assembly
AU2014340174B2 (en) 2013-10-24 2019-09-12 Amgen Inc. Drug delivery system with temperature-sensitive control
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
EP4043493A1 (en) 2013-12-24 2022-08-17 Janssen Pharmaceutica NV Anti-vista antibodies and fragments
US11046763B2 (en) 2014-01-08 2021-06-29 The Board Of Trustees Of The Leland Stanford Junior University Targeted therapy for small cell lung cancer
US10150818B2 (en) 2014-01-16 2018-12-11 Academia Sinica Compositions and methods for treatment and detection of cancers
WO2015109180A2 (en) 2014-01-16 2015-07-23 Academia Sinica Compositions and methods for treatment and detection of cancers
WO2015119906A1 (en) 2014-02-05 2015-08-13 Amgen Inc. Drug delivery system with electromagnetic field generator
MX2016010683A (en) 2014-02-21 2017-05-11 Ibc Pharmaceuticals Inc Disease therapy by inducing immune response to trop-2 expressing cells.
US9139649B2 (en) 2014-02-25 2015-09-22 Immunomedics, Inc. Humanized anti-CD22 antibody
US11059898B2 (en) 2014-03-24 2021-07-13 Cancer Research Technology Limited Modified antibodies containing modified IGG2 domains which elicit agonist or antagonistic properties and use thereof
CN106415244B (en) 2014-03-27 2020-04-24 中央研究院 Reactive marker compounds and uses thereof
WO2015149006A2 (en) 2014-03-27 2015-10-01 Dana-Farber Cancer Institute, Inc. Compositions and methods for modulating ncoa4-mediated autophagic targeting of ferritin
JP2017511148A (en) 2014-04-17 2017-04-20 ベーリンガー インゲルハイム エルツェーファウ ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディトゲゼルシャフト Recombinant host cells modified to overexpress helper proteins
JP2017511147A (en) 2014-04-17 2017-04-20 ベーリンガー インゲルハイム エルツェーファウ ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディトゲゼルシャフト Recombinant host cell for expressing the target protein
JP6640113B2 (en) 2014-05-07 2020-02-05 アムジエン・インコーポレーテツド Auto-injector with impact reducing element
JP6827319B2 (en) 2014-05-08 2021-02-10 中外製薬株式会社 GPC3 targeted therapies GPC3 targeted therapies administered to patients for whom therapy is effective
US10118969B2 (en) 2014-05-27 2018-11-06 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
CA2950415A1 (en) 2014-05-27 2015-12-03 Academia Sinica Anti-cd20 glycoantibodies and uses thereof
CA2950440A1 (en) 2014-05-27 2015-12-03 Academia Sinica Anti-her2 glycoantibodies and uses thereof
JP6894239B2 (en) 2014-05-27 2021-06-30 アカデミア シニカAcademia Sinica Compositions and methods for universal glycoforms for enhanced antibody efficacy
TWI732738B (en) 2014-05-28 2021-07-11 中央研究院 Anti-tnf-alpha glycoantibodies and uses thereof
JP6822843B2 (en) 2014-06-03 2021-01-27 アムジエン・インコーポレーテツド Systems and methods for remotely processing data collected by drug delivery devices
EP2952584A1 (en) 2014-06-04 2015-12-09 Boehringer Ingelheim RCV GmbH & Co KG Improved protein production
CA2951885C (en) 2014-06-11 2023-07-04 Kathy A. Green Use of vista agonists and antagonists to suppress or enhance humoral immunity
WO2015196089A1 (en) * 2014-06-20 2015-12-23 Bioalliance C.V. Anti-cd22 antibody-drug conjugates and methods of using thereof
JP2017528418A (en) 2014-06-20 2017-09-28 バイオアライアンス コマンディテール フェンノートシャップ Antifolate receptor alpha (FRA) antibody-drug conjugates and methods of use thereof
WO2015200260A1 (en) 2014-06-24 2015-12-30 Immunomedics, Inc. Anti-histone therapy for vascular necrosis in severe glomerulonephritis
WO2016040369A2 (en) 2014-09-08 2016-03-17 Academia Sinica HUMAN iNKT CELL ACTIVATION USING GLYCOLIPIDS
MA40764A (en) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd THERAPEUTIC AGENT INDUCING CYTOTOXICITY
PL3204018T3 (en) 2014-10-07 2022-01-03 Immunomedics, Inc. Neoadjuvant use of antibody-drug conjugates
CA2957960C (en) 2014-10-14 2023-08-22 Amgen, Inc. Drug injection device with visual and audible indicators
CN107405398A (en) 2014-12-05 2017-11-28 伊穆奈克斯特股份有限公司 VSIG8 is identified as presumption VISTA acceptors and its to produce the purposes of VISTA/VSIG8 activators and antagonist
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
CA2973819A1 (en) 2014-12-19 2016-06-23 Alder Biopharmaceuticals, Inc. Humanized anti-acth antibodies and use thereof
WO2016100055A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with live button or user interface field
JP6716566B2 (en) 2014-12-19 2020-07-01 アムジエン・インコーポレーテツド Drug delivery device with proximity sensor
US10495645B2 (en) 2015-01-16 2019-12-03 Academia Sinica Cancer markers and methods of use thereof
US9975965B2 (en) 2015-01-16 2018-05-22 Academia Sinica Compositions and methods for treatment and detection of cancers
SI3656443T1 (en) 2015-01-21 2021-11-30 The Board Of Trustees Of The Leland Stanford Junior University Use of trl agonist and anti-cd47 agent to enhance phagocytosis of cancer cells
WO2016118191A1 (en) 2015-01-24 2016-07-28 Academia Sinica Novel glycan conjugates and methods of use thereof
EP3258988B1 (en) 2015-02-17 2019-08-28 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
ES2905870T3 (en) 2015-02-27 2022-04-12 Amgen Inc Drug delivery device having a needle guard mechanism with an adjustable threshold resistance to movement of the needle guard
CA2981543A1 (en) 2015-04-22 2016-10-27 Immunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating trop-2-positive cancer cells
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
JP7026509B2 (en) 2015-06-24 2022-02-28 ヤンセン ファーマシューティカ エヌブイ Anti-VISTA antibody and fragment
US10195175B2 (en) 2015-06-25 2019-02-05 Immunomedics, Inc. Synergistic effect of anti-Trop-2 antibody-drug conjugate in combination therapy for triple-negative breast cancer when used with microtubule inhibitors or PARP inhibitors
ES2953441T3 (en) 2015-06-25 2023-11-13 Immunomedics Inc Combination of anti-hla-dr or anti-Trop-2 antibodies with microtubule inhibitors, parp inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US11376326B2 (en) 2015-07-01 2022-07-05 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting therapeutic agent which is administered to patient for whom the GPC3-targeting therapeutic agent is effective
HRP20231544T1 (en) 2015-08-13 2024-03-15 Amgen Inc. Charged depth filtration of antigen-binding proteins
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
WO2017100501A1 (en) 2015-12-09 2017-06-15 Amgen Inc. Auto-injector with signaling cap
AU2016381992B2 (en) 2015-12-28 2024-01-04 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11154661B2 (en) 2016-01-06 2021-10-26 Amgen Inc. Auto-injector with signaling electronics
JP7078536B2 (en) 2016-01-08 2022-05-31 アルトゥルバイオ, インコーポレイテッド Tetravalent anti-PSGL-1 antibody and its use
US20170224837A1 (en) 2016-02-10 2017-08-10 Immunomedics, Inc. Combination of abcg2 inhibitors with sacituzumab govitecan (immu-132) overcomes resistance to sn-38 in trop-2 expressing cancers
EP3413910A1 (en) 2016-02-12 2018-12-19 Janssen Pharmaceutica NV Anti-vista (b7h5) antibodies
JP2019515876A (en) 2016-03-08 2019-06-13 アカデミア シニカAcademia Sinica Methods for module synthesis of N-glycans and their arrays
WO2017160799A1 (en) 2016-03-15 2017-09-21 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
JP7277047B2 (en) 2016-04-15 2023-05-18 イミュネクスト インコーポレイテッド ANTI-HUMAN VISTA ANTIBODY AND USES THEREOF
SG10202109689QA (en) 2016-04-15 2021-10-28 Alder Biopharmaceuticals Inc Humanized anti-pacap antibodies and uses thereof
WO2017184880A1 (en) 2016-04-20 2017-10-26 Coherus Biosciences, Inc. A method of filling a container with no headspace
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
RU2725292C2 (en) 2016-04-27 2020-06-30 Иммьюномедикс, Инк. Efficacy of conjugates of anti-trop-2 antibody with drug sn-38 for therapy of recurrent/refractory to inhibitors of tumor control point
US11541168B2 (en) 2016-04-29 2023-01-03 Amgen Inc. Drug delivery device with messaging label
WO2017192287A1 (en) 2016-05-02 2017-11-09 Amgen Inc. Syringe adapter and guide for filling an on-body injector
AU2017263558B2 (en) 2016-05-13 2022-12-22 Amgen Inc. Vial sleeve assembly
WO2017200989A1 (en) 2016-05-16 2017-11-23 Amgen Inc. Data encryption in medical devices with limited computational capability
US11541176B2 (en) 2016-06-03 2023-01-03 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
WO2017218698A1 (en) 2016-06-15 2017-12-21 Sutro Biopharma, Inc. Antibodies with engineered ch2 domains, compositions thereof and methods of using the same
EP3478342A1 (en) 2016-07-01 2019-05-08 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
WO2018034784A1 (en) 2016-08-17 2018-02-22 Amgen Inc. Drug delivery device with placement detection
AU2017316663B2 (en) 2016-08-22 2024-02-22 CHO Pharma Inc. Antibodies, binding fragments, and methods of use
AU2017345490B2 (en) 2016-10-21 2022-07-07 Amgen Inc. Pharmaceutical formulations and methods of making the same
EP3532127A1 (en) 2016-10-25 2019-09-04 Amgen Inc. On-body injector
CN110248668B (en) 2016-12-15 2023-05-30 杜克大学 Antibodies and methods for depleting regulatory B10 cells and combination with immune checkpoint inhibitors
MX2019008432A (en) 2017-01-17 2019-11-18 Amgen Inc Injection devices and related methods of use and assembly.
KR102607101B1 (en) 2017-01-26 2023-11-29 제트엘아이피 홀딩 리미티드 CD47 antigen binding unit and its uses
CA3048520A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Drug delivery device with sterile fluid flowpath and related method of assembly
AU2018221351B2 (en) 2017-02-17 2023-02-23 Amgen Inc. Insertion mechanism for drug delivery device
EP3592403A1 (en) 2017-03-06 2020-01-15 Amgen Inc. Drug delivery device with activation prevention feature
JP2020509839A (en) 2017-03-07 2020-04-02 アムジエン・インコーポレーテツド Needle insertion due to overpressure
KR20240005194A (en) 2017-03-09 2024-01-11 암겐 인코포레이티드 Insertion mechanism for drug delivery device
JP2020512314A (en) 2017-03-27 2020-04-23 イミューノメディクス、インコーポレイテッドImmunomedics, Inc. Treatment of Trop-2-expressing triple-negative breast cancer with sacituzumab gobitecan and RAD51 inhibitor
KR102517342B1 (en) 2017-03-28 2023-03-31 암겐 인코포레이티드 Plunger rod and syringe assembly systems and methods
EP3606964A4 (en) 2017-04-03 2020-12-09 Immunomedics, Inc. Subcutaneous administration of antibody-drug conjugates for cancer therapy
EP3634539A1 (en) 2017-06-08 2020-04-15 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
AU2018282077B2 (en) 2017-06-08 2023-11-23 Amgen Inc. Torque driven drug delivery device
MX2019015472A (en) 2017-06-22 2020-02-19 Amgen Inc Device activation impact/shock reduction.
MA49461A (en) 2017-06-23 2020-04-29 Amgen Inc ELECTRONIC DRUG DELIVERY DEVICE INCLUDING A CAP ACTIVATED BY A SWITCH ASSEMBLY
IL270784B2 (en) 2017-07-14 2023-11-01 Amgen Inc Needle insertion-retraction system having dual torsion spring system
IL271173B1 (en) 2017-07-21 2023-12-01 Amgen Inc Gas permeable sealing member for drug container and methods of assembly
WO2019022950A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with container access system and related method of assembly
EP3658203B1 (en) 2017-07-25 2022-08-31 Amgen Inc. Drug delivery device with gear module and related method of assembly
EP3664863A2 (en) 2017-08-09 2020-06-17 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
US11077246B2 (en) 2017-08-18 2021-08-03 Amgen Inc. Wearable injector with sterile adhesive patch
US11103636B2 (en) 2017-08-22 2021-08-31 Amgen Inc. Needle insertion mechanism for drug delivery device
EP3691717B1 (en) 2017-10-04 2023-02-08 Amgen Inc. Flow adapter for drug delivery device
WO2019070552A1 (en) 2017-10-06 2019-04-11 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
EP3694578A1 (en) 2017-10-09 2020-08-19 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
MA50528A (en) 2017-11-03 2020-09-09 Amgen Inc SYSTEMS AND APPROACHES TO STERILIZE A DRUG DELIVERY DEVICE
EP3707075A1 (en) 2017-11-06 2020-09-16 Amgen Inc. Fill-finish assemblies and related methods
JP2021501616A (en) 2017-11-06 2021-01-21 アムジエン・インコーポレーテツド Drug delivery device with placement and flow detection
EP3706826A1 (en) 2017-11-10 2020-09-16 Amgen Inc. Plungers for drug delivery devices
WO2019099322A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Autoinjector with stall and end point detection
MX2020004996A (en) 2017-11-16 2020-08-27 Amgen Inc Door latch mechanism for drug delivery device.
US10835685B2 (en) 2018-05-30 2020-11-17 Amgen Inc. Thermal spring release mechanism for a drug delivery device
US11083840B2 (en) 2018-06-01 2021-08-10 Amgen Inc. Modular fluid path assemblies for drug delivery devices
WO2020023444A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
CA3103682A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
US20210260279A1 (en) 2018-07-24 2021-08-26 Amgen Inc. Hybrid drug delivery devices with optional grip portion and related method of preparation
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
MA53320A (en) 2018-07-31 2021-11-03 Amgen Inc FLUID PATH ASSEMBLY FOR DRUG DELIVERY DEVICE
JP2022500095A (en) 2018-09-24 2022-01-04 アムジエン・インコーポレーテツド Intervention dosing system and method
AU2019348440A1 (en) 2018-09-26 2021-03-25 Ascendis Pharma A/S Degradable hyaluronic acid hydrogels
AU2019350660A1 (en) 2018-09-28 2021-03-18 Amgen Inc. Muscle wire escapement activation assembly for a drug delivery device
JP2022503983A (en) 2018-10-02 2022-01-12 アムジエン・インコーポレーテツド Injection system for drug delivery with internal force transmission
CA3112214A1 (en) 2018-10-05 2020-04-09 Amgen Inc. Drug delivery device having dose indicator
EA202191038A1 (en) 2018-10-15 2021-07-06 Эмджен Инк. METHOD OF PLATFORM ASSEMBLY FOR MEDICINE DELIVERY DEVICE
WO2020081479A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Drug delivery device having damping mechanism
US11213620B2 (en) 2018-11-01 2022-01-04 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
US20220031953A1 (en) 2018-11-01 2022-02-03 Amgen Inc. Drug delivery devices with partial needle retraction
EP3873563A1 (en) 2018-11-01 2021-09-08 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
CN116063520A (en) 2019-01-30 2023-05-05 真和制药有限公司 anti-GAL 3 antibodies and uses thereof
MX2021012557A (en) 2019-04-24 2021-11-12 Amgen Inc Syringe sterilization verification assemblies and methods.
US20220306734A1 (en) 2019-07-24 2022-09-29 H. Lundbeck A/S Anti-mglur5 antibodies and uses thereof
US20220273887A1 (en) 2019-08-23 2022-09-01 Amgen Inc. Drug delivery device with configurable needle shield engagement components and related methods
US20230095053A1 (en) 2020-03-03 2023-03-30 Sutro Biopharma, Inc. Antibodies comprising site-specific glutamine tags, methods of their preparation and methods of their use
SG10202003296VA (en) 2020-04-06 2021-11-29 H Lundbeck As Treatment of most bothersome symptom (mbs) associated with migraine using anti-cgrp antibodies
WO2022171827A1 (en) 2021-02-12 2022-08-18 Boehringer Ingelheim Rcv Gmbh & Co Kg Signal peptides for increased protein secretion
IL308296A (en) 2021-05-07 2024-01-01 Viela Bio Inc Use of an anti-cd19 antibody to treat myasthenia gravis
JPWO2022239720A1 (en) 2021-05-10 2022-11-17
CA3217207A1 (en) 2021-05-21 2022-11-24 Amgen Inc. Method of optimizing a filling recipe for a drug container
TW202340245A (en) 2021-07-27 2023-10-16 瑞典商阿斯特捷利康公司 Treatment of lupus
WO2023016488A1 (en) 2021-08-13 2023-02-16 昆山新蕴达生物科技有限公司 Microtubule inhibitor-based antibody-drug conjugate
WO2023024949A1 (en) 2021-08-24 2023-03-02 昆山新蕴达生物科技有限公司 Antibody-drug conjugate conjugated via breakable linker
CA3229059A1 (en) 2021-08-27 2023-03-02 H. Lundbeck A/S Treatment of cluster headache using anti-cgrp antibodies

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5057313A (en) * 1986-02-25 1991-10-15 The Center For Molecular Medicine And Immunology Diagnostic and therapeutic antibody conjugates
US5258498A (en) * 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5789554A (en) * 1994-08-12 1998-08-04 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US6083477A (en) * 1996-10-17 2000-07-04 Immunomedics, Inc. Non-antigenic toxin-conjugate and fusion protein of internalizing receptor system

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807715A (en) * 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
GB8607679D0 (en) * 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US7321026B2 (en) * 2001-06-27 2008-01-22 Skytech Technology Limited Framework-patched immunoglobulins

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5057313A (en) * 1986-02-25 1991-10-15 The Center For Molecular Medicine And Immunology Diagnostic and therapeutic antibody conjugates
US5258498A (en) * 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5789554A (en) * 1994-08-12 1998-08-04 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US6083477A (en) * 1996-10-17 2000-07-04 Immunomedics, Inc. Non-antigenic toxin-conjugate and fusion protein of internalizing receptor system

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010094142A (en) * 2008-10-14 2010-04-30 Nikkiso Co Ltd Heat exchange device

Also Published As

Publication number Publication date
JP3053873B2 (en) 2000-06-19
US6187287B1 (en) 2001-02-13
IL114909A (en) 1999-10-28
WO1996004925A1 (en) 1996-02-22
US20050106108A1 (en) 2005-05-19
ES2251723T3 (en) 2006-05-01
AU3272695A (en) 1996-03-07
JPH10505231A (en) 1998-05-26
CA2195557A1 (en) 1996-02-22
US20020102254A1 (en) 2002-08-01
US5789554A (en) 1998-08-04
DE69534530D1 (en) 2006-03-02
EP0771208A4 (en) 2000-01-19
ATE306930T1 (en) 2005-11-15
DE69534530T2 (en) 2006-07-06
IL114909A0 (en) 1995-12-08
EP0771208A1 (en) 1997-05-07
EP0771208B1 (en) 2005-10-19
US20070172920A1 (en) 2007-07-26
CA2195557C (en) 2006-10-17

Similar Documents

Publication Publication Date Title
US8771694B2 (en) Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US6187287B1 (en) Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US6254868B1 (en) Glycosylated humanized B-cell specific antibodies
WO1997034632A9 (en) Glycosylated humanized b-cell specific antibodies
US6417337B1 (en) High affinity humanized anti-CEA monoclonal antibodies
King et al. Preparation and preclinical evaluation of humanised A33 immunoconjugates for radioimmunotherapy
US6676924B2 (en) CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
AU6338898A (en) High affinity humanized anti-cea monoclonal antibodies
AU689331C (en) CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION