US20060147444A1 - Biosynthetic binding proteins for immuno-targeting - Google Patents

Biosynthetic binding proteins for immuno-targeting Download PDF

Info

Publication number
US20060147444A1
US20060147444A1 US10/684,237 US68423703A US2006147444A1 US 20060147444 A1 US20060147444 A1 US 20060147444A1 US 68423703 A US68423703 A US 68423703A US 2006147444 A1 US2006147444 A1 US 2006147444A1
Authority
US
United States
Prior art keywords
sfv
formulation
amino acid
polypeptide
antigen
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
US10/684,237
Inventor
James Huston
Lou Houston
David Ring
Hermann Oppermann
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/684,237 priority Critical patent/US20060147444A1/en
Publication of US20060147444A1 publication Critical patent/US20060147444A1/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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3015Breast
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/55Fusion polypeptide containing a fusion with a toxin, e.g. diphteria toxin

Definitions

  • This invention relates in general to novel biosynthetic compositions of matter having particular utility as in vivo targeting agents and more specifically, to biosynthetic dimeric constructs of single-chain binding proteins (sFv), conjugates thereof, and to methods for their production.
  • sFv single-chain binding proteins
  • Fv fragments which consist of one V H and one V L domain held together by noncovalent interactions, form the minimal region of an antibody that contains a complete antigen combining site, dissociation of the V H and V L domains in vivo can preclude their use as therapeutic or imaging agents.
  • sFv polypeptide chains of about 27 kD have been developed containing covalently linked V H -V L polypeptides.
  • the V H - and V L -domains are connected by a polypeptide linker.
  • the resulting sFv polypeptide chains are also referred to in the art as biosynthetic antibody binding sites or BABS and preferably are encoded by a single DNA sequence.
  • biosynthetic polypeptide chains see for example, Huston et al., 1988, Proc. Nat. Aca. Sci. USA 85: 5879-5883 or U.S. Pat. Nos. 5,091,513 and 5,132,405, all of which are hereby incorporated by reference.
  • sFv polypeptide chains provide attractive alternatives to intact immunoglobulins and Fab fragments due to their small size and their stability at concentrations that typically promote dissociation of natural Fv fragments.
  • U.S. Pat. Nos. 5,091,513 and 5,132,405; Huston et al., ((1991) Methods in Enzymology 203: 46-88; Huston et al (1993) Int. Rev. Immunol. 10: 195-217) disclose the utility of sFv polypeptides, as well as single chain constructs synthesized from single DNA sequences, which may further comprise ancillary effector proteins, such as a second sFv or a cytotoxic agent.
  • mini-antibodies are sFv polypeptide chains which also include an 11 oligomerization domain” at their C-termini, separated from the sFv by a hinge region.
  • the oligomerization domains comprise self-associating ⁇ -helices, for example, leucine zippers, that can be further stabilized by additional disulfide bonds.
  • the domains a;re designed to be compatible with vectorial folding across a membrane, a process thought to facilitate in vivo folding of the polypeptide into a functional binding protein.
  • PCT application PCT/US92/09965, published Jun. 10, 1993 also discloses the construction of bivalent sFv constructs, including crosslinked dimers.
  • PCT application PCT/US92/07986, published Apr. 1, 1993 discloses bifunctional (Fab′) 2 molecules composed of two Fab′ monomers linked through cysteine amino acids located at the C-terminus of the first constant domain of each heavy chain.
  • PCT application PCT/US92/10140, published Jun. 10, 1993 also discloses bifunctional (Fab′) 2 dimers which, in addition to the cysteine residues located in the hinge region, also contain C-terminal leucine zipper domains that further stabilize the (Fab′) 2 dimers.
  • the resulting (Fab′) 2 dimers ( ⁇ 100 kD in size), although smaller than intact immunoglobulins, are significantly larger than sFv polypeptides and are anticipated to have slower tissue biodistribution and clearance rates following in vivo administration.
  • Cumber et al. disclose the generation of (Fv-Cys) 2 heterodimers by chemically crosslinking two V H -cys domains together (Cumber et al., 1992, J. Immunology 149B: 120-126). Although the crosslinked V H chains appear to be stable, dissociation of the V L polypeptides from each Fv reduces the pharmacological value of these constructs in vivo.
  • biocompatible constructs having accelerated in vivo biodistribution and body clearance rates than that of antibodies or antibody fragments.
  • biosynthetic constructs having enhanced avidity in vivo, including enhanced target tissue specificity and target tissue retention.
  • dimeric biosynthetic constructs having improved tissue imaging and drug targeting properties in vivo.
  • diagnostic and therapeutic formulations comprising these constructs, having particular utility in the diagnosis and treatment of malignancies.
  • the invention features a formulation for targeting an epitope on an antigen expressed in a mammal, where the formulation contains a pharmaceutically acceptable carrier in combination with a biosynthetic construct for binding at least one preselected antigen.
  • the dimeric construct has particular utility in diagnostic and therapeutic applications in vivo.
  • the invention features the synthesis and use of monomers and dimers of polypeptide constructs belonging to the class of proteins known as single-chain Fv (sFv) polypeptides.
  • sFv single-chain Fv
  • the sFv proteins described herein have superior in vivo pharmacokinetic properties, including accelerated tissue biodistribution and clearance rates relative to either intact IgG, (Fab) 2 dimers or Fab.
  • the dimeric biosynthetic construct of the invention contains two sFv polypeptide chains defined herein as follows.
  • Each sFv polypeptide chain comprises an amino acid sequence defining at least two polypeptide domains. These domains are connected by a polypeptide linker spanning the distance between the C-terminus of one domain and the N-terminus of the other.
  • each domain includes complementarity determining regions (CDRS) interposed between framework regions (FRs) where the CDRs and FRs of each polypeptide chain together define a binding site immunologically reactive with a preselected antigen.
  • CDRS complementarity determining regions
  • FRs framework regions
  • each biosynthetic binding site polypeptide chain can have an amino acid sequence peptide bonded and thus contiguous with the C-terminus of each polypeptide chain, referred to herein as a “C-terminal tail” sequence.
  • C-terminal tail The term “sFv′” refers hereinafter, to an sFv molecule containing such a C-terminal tail sequence.
  • This tail sequence preferably does not contain an ⁇ -helical motif that self-associates with another polypeptide chain of similar sequence but still contains a means for covalently crosslinking two such polypeptide chains together.
  • the resulting dimeric construct has a conformation that permits the independent binding of a preselected antigen or antigens to the binding site of each polypeptide chain in vitro and in vivo.
  • the resulting dimeric constructs have superior in vivo pharmacokinetic properties that include significantly enhanced avidity, including enhanced target tissue retention and/or antigen localization properties, as compared with intact IgG, Fab, (Fab) 2 dimers or monomeric sFv.
  • C-terminal tail sequence referred to herein generally as a “C-terminal tail” sequence, peptide bonded to the C-terminus of an sFv and comprising means for crosslinking two sFv polypeptide chains, alternatively may occur at the N-terminus of an sFv (“N-terminal tail”) or may comprise part of the polypeptide linker spanning the domains of an individual sFv.
  • the dimeric species created by the crosslinking of sFvs having these alternative “tail” sequences also are contemplated to have a conformation permitting the in vivo binding of a preselected antigen by the binding sites of each of the sFv polypeptide chains.
  • both polypeptide chains bind the same epitope on a preselected antigen, and the resulting dimeric construct is termed a “homodimer.”
  • the polypeptide chains bind different epitopes on a preselected antigen and the resulting dimeric construct is termed a “heterodimer.”
  • the two polypeptide chains bind different epitopes on two different, preselected antigens.
  • epitope refers to a portion of an antigen that makes contact with a particular antibody or antibody analogue. In a typical protein, it is likely that any residue accessible from the surface can form part of one or more antigenic determinants.
  • antigen refers to a molecule that can elicit an immune response and that can react specifically with corresponding antibodies or antibody analogues.
  • domain refers to an amino acid sequence that folds into a single globular region in its native conformation, and which may exhibit discrete binding or functional properties.
  • polypeptide linker refers to an amino acid sequence that links the C-terminus of one domain to the N-terminus of the other domain, while still permitting the two domains to maintain their proper physiologically active binding conformations.
  • the currently preferred polypeptide linkers that connect the C-terminus of one domain to the N-terminus of the other domain include part or all of amino acid sequence ((Gly) 4 Ser) 3 set forth in the SEQ. ID. NO.: 7, or ((Ser) 4 Gly) 3 as set forth in SEQ. ID. NO.: 8.
  • the amino acid sequence of each of the polypeptide domains includes complementarity determining regions interposed between framework regions.
  • complementarity determining regions or “CDRs”, as used herein, refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native immunoglobulin binding site, or a synthetic polypeptide which mimics this function. CDRs are not necessarily wholly homologous to hypervariable regions of natural Fv molecules, and also may include specific amino acids or amino acid sequences which flank the hypervariable region and have heretofore been considered framework not directly determinative of complementarity.
  • framework regions or “FRs”, as used herein, refers to amino acid sequences which are found naturally occurring between CDRs in immunoglobulins.
  • FR sequences may be derived in whole or part from the same immunoglobulin as the CDRs, or in whole or part from a different immunoglobulin.
  • the FR sequences can be derived from a human immunoglobulin and so the resulting humanized sFv will be less immunogenic than a murine monoclonal antibody.
  • each variable domain includes three CDRs interspersed between four FRs.
  • the two polypeptide domains that define an sFv molecule contain CDRs interspersed between FRs which together form a binding site immunologically reactive with a preselected antigen.
  • immunologically reactive refers to the noncovalent interactions of the type that occur between an immunoglobulin molecule and an antigen for which the immunoglobulin is specific.
  • vidity describes the stability of a complex formed by a multivalent antibody or antibody analogue, with its binding conjugate.
  • the term “apparent avidity” describes the stability of a complex formed by an antibody or an antibody analogue with its binding conjugate as determined by in vivo immunolocalization studies.
  • the CDRs of the polypeptide chain can have an amino acid sequence substantially homologous with at least a portion of the amino acid sequence of CDRs from a variable region of an immunoglobulin molecule from a first species, together with FRs that are substantially homologous with at least a portion of the amino acid sequence of FRs from a variable region of an immunoglobulin molecule from a second species.
  • the first species is mouse and the second species is human.
  • the CDR sequences in the sFv′ polypeptides are preferably substantially homologous to an immunoglobulin CDR retaining at least 70%, or more preferably 80% or 90%, of the amino acid sequence of the immunoglobulin CDR, and also retains the immunological binding properties of the immunoglobulin.
  • Each sFv′ molecule has a C-terminal polypeptide tail that has a non-self-associating structure and contains at least one crosslinking means.
  • Useful crosslinking means include derivatizable amino acid side chains, particularly those selected from the group consisting of cysteine, lysine, arginine, histidine, glutamate, aspartate, and derivatives and modified forms thereof.
  • cysteine amino acids are incorporated into the C-terminal tail sequences as the crosslinking means.
  • the crosslinking means includes one or more amino acids that can be posttranslationally modified.
  • the crosslinking means can include one or more glycosylation sites, wherein the incorporated carbohydrate moieties can be crosslinked in vitro.
  • Preferred glycosylation sequences include Asn-Xaa-Thr and Asn-Xaa-Ser, where Xaa can be any amino acid, wherein the carbohydrate is typically N-linked to asparagine or O-linked to serine or threonine.
  • the tail also may comprise an amino acid sequence that defines a metal ion chelation motif, and which facilitates purification of the sFv′ monomers by metal ion affinity chromatography, such as the IMAC 2+ chromatography system.
  • chelation motifs can be used for binding detectable moieties, such as Technetium ⁇ 99m ( 99m Tc) for in vivo imaging.
  • Preferred examples of useful C-terminal tail amino acid sequences wherein the crosslinking means is provided by the sulfhydryl group of a cysteine include: Ser-Cys; (Gly) 4 -Cys; and (His) 6 -(Gly) 4 -Cys; set forth in the Sequence Listing as SEQ. ID. NOS.: 9, 10 and 11, respectively.
  • the (Gly) 4 -Cys sequence facilitates the coordination of 99m Tc by this tail.
  • monomeric sFv′ molecules can be coupled together through the crosslinking means in the C-terminal tails to form either homo- or heterodimeric (sFv′) 2 species.
  • sFv coupler refers to the chemical bridge that links two sFv′ polypeptide chains together to form a dimeric species.
  • the crosslinking means is a cysteine residue
  • the linkage is by a disulfide bond.
  • sulfhydryl-specific homobifunctional crosslinking reagents such as bismaleimidohexane, or heterobifunctional crosslinking reagents, can be used to join the two sFv′ molecules together.
  • sFv couplers of preselected length also can be designed to limit interaction between the two sFv′ polypeptide chains or to optimize binding of two preselected antigens, including, for example, multiple copies of a receptor expressed on a cell surface in a mammal.
  • An example of such a variable length coupler includes the bismaleimidocaproyl amino acid (MCA) synthetic peptide bridge.
  • MCA bismaleimidocaproyl amino acid
  • GlySer 3 Gly 2 Ser 3 Lys peptide spacer is used, in theory, any amino acid sequence can be introduced into this type of chemical bridge with a variety of reactive moieties at either end. Consequently, it is possible to design specific linkage groups that can have a predetermined length and flexibility.
  • a polylysine or polyproline peptide may be used.
  • Another benefit of the MCA linkers over many other commercially available linkers is that they are soluble in water.
  • the chemical bridge also may be created to enhance the imaging or therapeutic properties of the construct in vivo (vide infra).
  • the separation distance between, and interaction of, the sFv′ monomers in a dimeric construct of the invention also can be modulated by the judicious choice of amino acids in the tail sequences themselves.
  • the dimeric constructs of this invention preferably target a pharmacologically active drug (or other ancillary protein) to a site of interest utilizing the bivalent capability of the dimer.
  • pharmacologically active drugs include molecules that inhibit cell proliferation and cytotoxic agents that kill cells.
  • cytotoxic agent refers to any molecule that kills cells, and includes anti-cancer therapeutic agents such as doxorubicin.
  • toxins for instance, the toxic portion of the Pseudomonas exotoxin, phytolaccin, ricin, ricin A chain, or diptheria toxin, or other related proteins known as ricin A chain-like ribosomal inhibiting proteins, i.e., proteins capable of inhibiting protein synthesis at the level of the ribosome, such as pokeweed antiviral protein, gelonin, and barley ribosomal protein inhibitor.
  • one sFv′ can be immunologically reactive with a binding site on an antigen at the site of interest
  • the second sFv′ in the dimer can be immunologically reactive with a binding site on the drug to be targeted.
  • the construct may bind one or more antigens at the the site of interest and the drug to be targeted is otherwise associated with the dimer, for example, crosslinked to the chemical bridge itself.
  • the biosynthetic dimeric constructs of this invention also may be used as part of human therapies to target cytotoxic cells such as cytotoxic T-lymphocytes, or pharmacologically active drugs to a preselected site.
  • ADCC antibody dependent cellular cytotoxicity
  • Still another bispecific dimeric construct having cytotoxic properties is a bispecific construct with one sFv′ capable of targeting a tumor cell and the second sFv′ having catalytic properties that binds an inactive drug, subsequently converting it into an active compound (see for example, U.S. Pat. No. 5,219,732).
  • Such a construct would be capable of inducing the formation of a toxic substance in situ.
  • a catalytic sFv′ molecule having ⁇ -lactamase-like activity can be designed to bind and catalyze the conversion of an inactive lactam derivative of doxorubicin into its active form.
  • the bispecific dimer having binding affinities for both the preselected antigen and the inactive-lactam derivative, is administered to an individual and allowed to accumulate at the desired location.
  • the inactive and nontoxic cytotoxin-lactam derivative then is administered to the individual.
  • Interaction of the derivative with the bispecific (sFv′) 2 heterodimer at the site of interest releases the active form of the drug in situ, enhancing both the cytotoxicity and specificity of the drug.
  • the homo- and heterodimeric biosynthetic constructs also may include a detectable moiety bound either to the polypeptide chain, e.g., to the tail sequence, or to the chemical coupler.
  • detectable moiety refers to the moiety bound to or otherwise complexed with the construct and which can be detected external to, and at a distance from, the site of the complex formation, to permit the imaging of cells or cell debris expressing a preselected antigen.
  • Preferable detectable moieties for imaging include radioactive atoms such as Technetium ⁇ 99m ( 99m Tc), a gamma emitter with a half-life of about 6 hours.
  • Non-radioactive moieties useful for in vivo magnetic resonance imaging applications include nitroxide spin labels as well as lanthanide and transition metal ions which induce proton relaxation in situ.
  • the complexed radioactive moieties also may be used in standard radioimmunotherapy protocols to destroy the targeted cell.
  • Preferable nucleotides for high dose radioimmunotherapy include radioactive atoms such as, 90 Yttrium ( 90 Yt), 131 Iodine ( 131 I) or 111 Indium ( 111 In).
  • the sFv, sFv′ and (sFv′ ) 2 constructs disclosed herein have particular utility as in vivo targeting agents of tumor antigens, including antigens characteristic of breast and ovarian malignancies, such as the c-erbB-2 or c-erbB-2 related antigens. Accordingly, these constructs have particular utility in diagnostic applications as imaging agents of malignant cells, and in therapeutic applications as targeting agents for cytotoxins and other cancer therapeutic agents.
  • the CDRs of the sFv or sFv′ polypeptide chain have an amino acid sequence substantially homologous with the CDRs of the variable region of any one of the following monoclonal antibodies: 741F8, 520C9, and 454C11, all of which bind to c-erbB-2 or c-erbB-2-related antigens.
  • Exemplary sFv′ and sFv sequences having CDRs corresponding to the monoclonal antibodies 741F8 and 520C9 are set forth in the Sequence Listing SEQ. ID. NOS.: 1 and 5, respectively.
  • c-erbB-2 refers to a protein antigen that is an approximately 200 kD acidic glycoprotein having an isoelectric point of about 5.3 and having an extracellular domain overexpressed on the surface of tumor cells, such as breast and ovarian tumor cells in about 25% of cases of breast and ovarian cancer.
  • a “c-erbB-2-related tumor antigen” is a protein located on the surface of tumor cells, such as breast and ovarian tumor cells and which is antigenically related to the c-etbB-2 antigen. That is, the related antigen can be bound by an immunoglobulin that is capable of binding the c-erbB-2 antigen (e.g. 741F8, 520C9, and 454C11 antibodies.
  • antigens also include antigens comprising an amino acid sequence that is at least 80% homologous, preferably 90% homologous, with the amino acid sequence of c-erbB-2 or an amino acid sequence encoded by a DNA that hybridizes under stringent conditions with a nucleic acid sequence encoding c-erbB-2.
  • stringent hybridization conditions are those set forth in Sambrook, et al., 1989, Molecular Cloning; A Laboratory Manual 2nd ed.
  • hybridization conditions for example, include 50% formamide, 5 ⁇ Denhardt's Solution, 5 ⁇ SSC, 0.1% SDS and 100 ⁇ g/ml denatured salmon sperm DNA and the washing conditions include 2 ⁇ SSC, 0.1% SDS at 37° C. followed by 1 ⁇ SSC, 0.1% SDS at 68° C.
  • An example of a c-erbB-2-related antigen is the receptor for the epidermal growth factor.
  • the biosynthetic antibody binding site is a humanized hybrid molecule which includes CDRs from the mouse 741F8 antibody interposed between FRs derived from one or more human immunoglobulin molecule.
  • the CDRs that bind to the c-erbB-2 epitope can be found in the amino acid residue numbers 31-37, 52-68, 101-110, 159-169, 185-191 and 224-233 in SEQ ID NOS.: 1 and 2.
  • the hybrid molecule thus contains binding sites which are highly specific for the c-erbB-2 antigen or c-erbB-2 related antigens held in proper immunochemical binding conformation by human FR amino acid sequences, which are less likely to be recognized as foreign by the human body.
  • the dimeric (sFv′) 2 construct can either be homodimeric, wherein the CDR sequences on both monomers define the same binding site, or heterodimeric, wherein the CDR sequences of each sFv′ monomer define a different binding site.
  • An example of an (sFv′) 2 heterodimer described herein having specificity for both c-erbB-2 and digoxin epitopes can be generated by combining the anti-c-erbB-2 sFv′, shown in SEQ. ID. NOS.: 1 and 2 with the anti-digoxin sFv′, shown in SEQ. ID. NOS.: 3 and 4.
  • the CDRs that bind to the digoxin epitope can be derived from the anti-digoxin murine monoclonal antibody 26-10 (Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85: 5879-5883) and can be found in the amino acid residue numbers 32 through 36, 48 through 65, 101 through 107, 157 through 170, 188 through 194 and 229 through 234 in the Sequence Listing as SEQ. ID. NOS.: 3 and 4.
  • Radioimaging or radioimmunotherapy of tumor tissues and malignant cells are preferred aspects of this invention.
  • Overexpression of tumor antigens such as c-erbB-2 and related cell surface antigens in malignant cells allows imaging of the malignant cell or tissue, whether it is well localized, has undergone metastasis or is exposed following cell lysis.
  • the imaging method includes the steps of administering to a mammal a formulation comprising an sfv′ or (sFv′) 2 dimeric construct having specificity for the antigen tumor and containing a detectable moiety at a concentration sufficient to permit extracorporeal detection of the construct bound to the tumor antigen; and then detecting the biosynthetic construct bound to the tumor antigen.
  • the formulation can be used to particular advantage in gamma scintigraphy or magnetic resonance imaging.
  • Overexpression of c-erbB-2 or related receptors on malignant cells thus allows targeting of sFv′ species to the tumor cells, whether the tumor is well-localized or metastatic.
  • internalization of an sFv-toxin fusion protein permits specific destruction of tumor cells bearing the overexpressed c-erbB-2 or related antigen.
  • the present invention discloses monomeric and dimeric biosynthetic constructs having enhanced properties as in vivo targeting agents when compared with intact monoclonal antibodies or their Fab fragments.
  • the dimeric biosynthetic constructs of the invention also permit the in vivo targeting of an epitope on an antigen with greater apparent avidity, including greater tumor specificity, tumor localization and tumor retention properties than that of the Fab fragment having the same CDRs as the construct.
  • the dimeric constructs also permit the in vivo targeting of an epitope on an antigen with a greater apparent avidity, including greater tumor localization and tumor retention properties, than either of the monomeric polypeptides individually.
  • the invention also includes methods for producing the homo- and heterodimeric biosynthetic constructs, which include the steps of designing, constructing, expressing, purifying, and refolding the monomeric sFv′ polypeptide chains in vitro, followed by joining two polypeptide chains together through the crosslinking means in the C-terminal tail sequence, without relying on the tail structure to otherwise assist in dimer formation or enhance transport across a membrane.
  • the invention also includes methods for imaging a preselected antigen in a mammal expressing the preselected antigen. The antigen may be expressed on a cell surface or may be released as part of the cell debris from a dying cell.
  • FIG. 1A is a schematic representation of a DNA construct encoding the sFv′ biosynthetic binding protein of the invention
  • FIG. 1B is a schematic representation of the polypeptide chain encoded by the DNA construct in FIG. 1A ;
  • FIG. 2A is a schematic representation of a refolded sFv′ protein in its native conformation
  • FIG. 2B is a schematic representation showing two folded sFv′ polypeptides covalently linked by a disulfide bond
  • FIG. 3 is a graphic representation of an in vitro competition assay comparing the c-erbB-2 binding activity of an Fab fragment of the 520C9 monoclonal antibody (filled dots), with that of biosynthetic 520C9 sFv at two different stages of purification: mixture of folded and unfolded sFv (+) or affinity-purified sFv (squares), and with a material that did not bind to the affinity column (*);
  • FIG. 4 lists in tabular form the tumor:organ ratios calculated for various sF and sFv′ species injected into tumor-containing mice;
  • FIG. 5 lists in tabular form the percentage of injected dose localized to tumor tissue for various sFv and sFv's species.
  • FIG. 6 is a graphic representation summarizing the comparative tumor retention properties of monomeric and dimeric forms of different sFv′ constructs and Fabs represented by bars 1-6.
  • the sFv′ species represented by bars 1-5 are based on the V regions of the 741F8 monoclonal antibody.
  • Bar 1 refers to intravenously (i.v.) administered glutathionyl-(sFv′-SerCys) monomer, bar 2 to disulfide linked (sFv′-Gly 4 -Cys) 2 , bar 3 to MCA combined (sFv′-Ser-Cys) 2 , bar 4 to BMH cross-linked (sFv′-Ser-Cys) 2 , bar 5 to 741F8 Fab and bar 6 to the 26-10 disulfide linked (sFv′-Ser-Cys) 2 .
  • intravenously administered single-chain Fv (sFv) proteins exhibit superior in vivo pharmacokinetic properties relative to intact monoclonal antibodies (IgG), (Fab) 2 dimers or Fab fragments. These pharmacokinetic properties include accelerated rates of tissue biodistribution, enhanced target tissue specificity, and exceptionally fast clearance rates.
  • the sFv constructs can be designed to bind to preselected antigens and to have particular utility for in vivo immunoimaging and immunotherapy applications.
  • dimeric forms of the constructs which do not rely on self-associating tail sequences for dimerization or transport across a membrane, can be easily prepared and have improved target tissue localization properties, target tissue retention properties and/or avidity for their targets in vivo, relative to monomeric sFv′, Fab fragments or intact IgG.
  • the invention features a formulation for targeting an epitope on an antigen expressed in a mammal.
  • the formulation contains a pharmaceutically acceptable carrier in combination with a dimeric biosynthetic construct for binding at least one preselected antigen.
  • the preselected antigen either may be an antigen expressed on the surface of a cell or an intracellular component exposed upon lysis of the cell.
  • the sFv, sFv′ and (sFv′) 2 constructs disclosed herein have particular utility as in vivo targeting agents for detecting malignant cells in a mammal.
  • the constructs disclosed can be used to target the c-erbB-2 or c-erbB-2-related antigens which are overexpressed in certain breast and ovarian cancers.
  • radioimmunotargeting using radiolabeled (sFv′) 2 constructs will be useful for therapeutic as well as diagnostic applications.
  • biosynthetic sFv, sFv′ and (sFv′) 2 dimers useful in the compositions and methods of the invention, together with methods for their construction and administration.
  • numerous, non-limiting examples which demonstrate the suitability of these constructs as in vivo targeting reagents for diagnostic and therapeutic applications. More specifically, the examples demonstrate: the construction and expression of sFv polypeptides (Example 1); the renaturation, dimerization and purification of sFv′ proteins (Example 2); and the immunoreactivity of the monomeric and dimeric sFv proteins (Example 3).
  • Each of the sFv and sFv′ proteins have amino acid sequences that define at least two polypeptide domains.
  • the polypeptide domains are connected by a polypeptide linker spanning the distance between the C-terminus of one domain and the N-terminus of the other.
  • the amino acid sequence of each domain includes complementarity determining regions (CDRs) interposed between framework regions (FRs), where the CDRs and FRs of each polypeptide chain together define a binding site immunologically reactive with a preselected antigen.
  • each polypeptide chain has an additional C-terminal tail amino acid sequence having a substantially non-self-associating structure. More specifically, this is a sequence that does not interact appreciably with a similar sequence under physiological conditions, as is the case for example with the ⁇ -helical leucine zipper motifs found in DNA binding proteins.
  • Each tail sequence also contains a means for crosslinking two such sFv′ polypeptide chains together to form an (sFv′) 2 dimer.
  • the resulting (sFv′) 2 dimers have conformations which permit the in vivo binding of the preselected antigen by the binding sites of each of the polypeptide chains.
  • FIG. 1A is a schematic representation of the DNA construct
  • FIG. 1B is a schematic representation of the resulting encoded polypeptide chain
  • FIG. 2 is a schematic representation of the folded sFv′ monomer ( FIG. 2A ) and the dimeric (sFv′) 2 construct ( FIG. 2B ).
  • a single-chain Fv (sFv′) polypeptide, shown in FIGS. 1 and 2 A, comprises: a heavy chain variable region (V H ) 10 , and a light chain variable region, (V L ) 14 , wherein the V H and V L domains are attached by polypeptide linker 12 .
  • the binding domains defined by V L and V H include the CDRs 2 , 4 , 6 and 2 ′, 4 ′, 6 ′, respectively, and FRs 32 , 34 , 36 , 38 and 32 ′, 34 ′, 36 ′ 38 ′, respectively which, as shown in FIG. 2 , together define an immunologically reactive binding site or antigenic determinant, 8 .
  • the CDRs and FRs may be derived from different immunoglobulins (see infra).
  • the sFv′ molecules also contain a C-terminal tail amino acid sequence, 16 , comprising an amino acid sequence that will not self-associate with a polypeptide chain having a similar amino acid sequence under physiological conditions, and which contains a means, 18 , for the site-directed crosslinking of two such tail sequences.
  • the crosslinking means is the sulfhydryl group of a cysteine amino acid.
  • the cross-linking means, 18 may be blocked by a blocking group, 20 .
  • the blocking group may be a glutathionyl moiety when the crosslinking means, 18 , is a cysteine amino acid.
  • C-terminal tail sequence referred to herein generally as a “C-terminal tail” sequence, peptide bonded to the C-terminus of an sFv and comprising means for crosslinking two sFv polypeptide chains, alternatively may occur at the N-terminus of an sFv (“N-terminal tail”) or may comprise part of the polypeptide linker spanning the domains of an individual sFv′.
  • the dimeric species created by the crosslinking of sFvs having these alternative “tail” sequences also are contemplated to have a conformation permitting the in vivo binding of a preselected antigen by the binding sites of each of the sFv polypeptide chains.
  • the CDR and FR polypeptide segments are designed empirically based on sequence analysis of Fv regions of preexisting antibodies, such as those described in U.S. Pat. No. 4,753,894, hereby incorporated by reference.
  • the sFv and sFv′ polypeptide chains of the invention are biosynthetic in the sense that they are synthesized, transfected into a cellular host, and protein expressed from a nucleic acid containing genetic sequences based in part on synthetic DNA.
  • Synthetic DNA is understood to include recombinant DNA made by ligation of fragments of DNA derived from the genome of a hybridoma, mature B cell clones, a cDNA library derived from natural sources, or by ligation of plural, chemically synthesized oligonucleotides.
  • the proteins of the invention are properly characterized as “antibody binding sites”, in that these synthetic single polypeptide chains are able to refold into a 3-dimensional conformation with specificity and affinity for a preselected epitope on an antigen.
  • polypeptide chains of the invention are antibody-like in that their structure is patterned after, regions of native antibodies known to be responsible for antigen recognition.
  • the single-chain polypeptide chains of the invention are first derived at the DNA-level.
  • the sFv DNAs are preferably expressed in E. coli, the resulting polypeptide chains being solubilized from inclusion bodies, refolded in vitro, labeled with a detectable moiety, such as 99m Tc, and dimerized to form a biosynthetic (sFv′) 2 construct.
  • a detectable moiety such as 99m Tc
  • dimerized to form a biosynthetic (sFv′) 2 construct e.g., the constructs disclosed herein may also be engineered for secretion from the host cell, for example, secretion into the periplasmic space of an E.
  • the ability to design the single polypeptide chains of the invention depends on the ability to identify Fv binding domains of interest, and to obtain the DNA encoding these variable regions.
  • Hybridoma technology enables the production of cell lines that secrete antibodies to essentially any desired substance that elicits an immune response.
  • U.S. Pat. No. 4,753,894 describes some monoclonal antibodies of interest which recognize c-erbB-2 related antigens on breast cancer cells, and explains how such antibodies were obtained.
  • One monoclonal antibody that is particularly useful in targeting the c-erbB-2 antigen is 741F8 (Bjorn et al., 1985, Cancer Res. 45: 1214-1221; U.S. Pat. No. 4,753,894).
  • This antibody specifically recognizes the c-erbB-2 antigen expressed on the surface of various tumor cell lines, and exhibits very little binding to normal tissues.
  • Other monoclonal antibodies that bind c-erbB-2 or related antigens include 520C9 and 454C11 (Frankel et al., 1985, J. Biol. Resp. Modif. 4: 273-286; Ring et al., 1989, Cancer Res. 49: -3070-3080, Ring et al., 1991, Molec. Immunol. 28: 915-917; U.S. Pat. Nos. 4,753,894 and 5,169,774).
  • sFv′ sequences with the desired specificity can also be derived from phage antibody cloning of combinatorial V gene libraries. Such sequences could be based on cDNA derived from mice preimmunized with tumor cell membranes bearing c-erbB-2 or related antigenic fragments, (See, for example, Clackson et al, (1991) Nature 352: 624-628).
  • the process of designing DNA encoding the single polypeptide chain of interest can be accomplished as follows. Either synthetic DNA duplexes can be ligated together to form a synthetic gene or relevant DNA fragments can be cloned from libraries. In the latter procedure, mRNA encoding the light and heavy chains of the desired immunoglobulin may be isolated from hybridomas producing the immunoglobulin and reverse transcribed into cDNA.
  • V H and V L genes subsequently can be isolated by standard procedures, for instance, by colony hybridization of cDNA libraries (see for example, Sambrook et al., eds., 1989, Molecular Cloning, Cold Spring Harbor Laboratories Press, NY) or by polymerase chain reaction (PCR) (see for example, Innis et al., eds., 1990, PCR Protocols, A guide to methods and applications, Academic Press). Both procedures are well known in the art.
  • Still another approach involves the design and construction of synthetic variable domain genes encoding a predetermined, specific Fv binding site.
  • a computer program such as Compugene
  • the resulting V H and V L gene sequences can then be genetically linked together by means of a linker connecting the C-terminus of one chain with the N-terminus of the other.
  • the FRs may include amino acid sequences which are similar or identical to at least a portion of the FR amino acid sequences of antibodies native to that species.
  • the amino acid sequences constituting the CDRs may be analogous to the sequences from a second, different preexisting antibody having specificity for the antigen of interest (e.g. a murine or other human IgG).
  • the CDRs and FRs may be copied in their entirety from a single pre-existing monoclonal antibody cell line or a desirable sFv species may be cloned from a repertoire library derived from preimmunized or naive animals.
  • V L and V H domains in the DNA sequence of FIG. 1 is not critical. That is, although the sequence represented in FIG. 1A encodes a heavy chain variable region followed by the light chain variable region, as will be appreciated by those skilled in the art, the sFv may be constructed so that the light and heavy chain domains are in reverse order.
  • the V H and V L domains of the sFv are linked in the gene construct by means of a linker 12 ( FIG. 1A ).
  • the linker should be at least long enough (e.g., about 10 to 15 amino acids or at least 40 Angstroms in length) to permit domains 10 and 14 to assume their proper conformations and interdomain relationships.
  • the linkers preferably comprise hydrophilic amino acids that assume an unstructured configuration under physiological conditions, and are free of residues having large side groups that could interfere with proper folding of the V H , V L , or pendant chains. Examples of currently preferred linkers include either part or all of the amino acid sequences ((Gly) 4 Ser) 3 and ((Ser) 4 Gly) 3 , set forth in the Sequence Listing as SEQ. ID. NOS.: 7 and 8, respectively.
  • the linker may also include an amino acid sequence homologous to a sequence identified as “self” by the species into which it will be introduced, particularly if a therapeutic application is intended.
  • the sFv′ polypeptide chains further comprise a C-terminal tail containing at least one amino acid that can be derivatized or post-translationally modified to enable crosslinking of two such sFv′ monomers.
  • the tail sequences include one or more of the sequences Ser-Cys, (Gly) 4 -Cys and (His) 6 -(Gly) 4 -Cys, set forth in the Sequence Listing as SEQ. ID. NOS.: 9, 10, and 11, respectively.
  • the C-terminal tails preferably do not form ⁇ -helical structures which self-associate under physiological conditions, such as the ⁇ -helical leucine zipper motifs found in DNA binding proteins (O'Shea et al., 1989, Science 243: 538-542, O'Shea et al., 1991, Science 254:
  • Suitable derivatizable amino acid side chains may be selected from the group consisting of cysteine, lysine, arginine, histidine, glutamate, aspartate and derivatives or modified forms thereof.
  • cysteine amino acids are incorporated into the C-terminal tail sequences as the crosslinking means.
  • glycosyl moieties present on glycosylated amino acids can be covalently attached in vitro using bifunctional linkers on standard sugar chemistry (see for example, E. A. Davidson (1967) Carbohydrate Chemistry, Holt, Kinehart and Winston, N.Y.; W. J. Lennarz (1980) The Biochemsitry of Glycoproteins and Proteoglycans, Plenum Press, N.Y.).
  • Particularly useful glycosylation sites include the sequences Asn-Xaa-Thr and Asn-Xaa-Ser, wherein Xaa is any amino acid.
  • the glycosylation sequences need not include a cysteine.
  • the tail also may comprise an amino acid sequence defining an ion chelation motif which can be used as part of a purification protocol for isolating of the sFv′ monomers by metal ion affinity chromatography (e.g., by means of a (His) 6 tail on an IMAC chromatography column), as well as for chelating ions of detectable moieties such as Technetium ⁇ 99m or 111 Indium for in vivo imaging applications.
  • metal ion affinity chromatography e.g., by means of a (His) 6 tail on an IMAC chromatography column
  • detectable moieties such as Technetium ⁇ 99m or 111 Indium for in vivo imaging applications.
  • sFv coupler refers to chemical bridges that join the crosslinking residues in each of the sFv′ molecules.
  • the chemical bridge can be a disulfide bond.
  • sulfhydryl-specific crosslinking reagents can be used to join two sFv′ molecules together.
  • An example of such a cysteine-specific chemical bridge includes the bifunctional crosslinking reagent bismaleimidohexane (BMH), a water insoluble linker that can be obtained from Pierce, Rockford, Ill.
  • bifunctional crosslinking agents include heterobifunctional crosslinkers which can be used to join two sFv′ species together where the crosslinking residues in each of the sFv′ C-terminal tail sequences are different, such as, a C-terminal cysteine on one sFv′ and a C-terminal lysine on the other.
  • Useful heterobifunctional crosslinking agents include 4-succinimidyloxycarbonyl-methyl-(2-pyridyldithio)-toluene (SMPT) or N-succinimidyl-3-(2-pyridyldithio)propionate (SPDP), both of which can be obtained from Pierce, Rockland, Ill.
  • sFv couplers of variable length also can be prepared to limit steric interaction of two coupled sFv′ proteins.
  • An example of such an sFv coupler includes a peptide bridge, such as the water soluble bismaleimidocaproyl amino acid (MCA) linker.
  • MCA water soluble bismaleimidocaproyl amino acid
  • an MCA-, GlySer 3 Gly 2 Ser 3 Lys-MCA linker is used, in theory, any amino acid sequence can be introduced into this type of chemical bridge-spacer group.
  • Suitable MCA-peptide chemical bridges can be synthesized on polystyrene resins functionalized with hydroxymethylphenoxyacetic acid (HMP) to allow formation of free acids at the C-terminus following deblocking.
  • HMP hydroxymethylphenoxyacetic acid
  • Gly-Ser 3 -Gly 2 -Ser 3 -Lys the C-terminal lysine is esterified to the resin and other amino acids are added as N- ⁇ -Fmoc protected derivatives.
  • DIC/hydroxybenzotriazol activated amino acids are coupled for 90 minutes after which the N- ⁇ -Fmoc protected groups are deprotected with 20% piperidine in dimethylformamide (DMF).
  • the peptide Upon completion of the synthesis, the peptide is cleaved from the resin and deprotected with 95% trifluoroacetic acid (TFA) in water. The crude peptide then is dissolved in 0.1M phosphate buffer pH 7 and reacted overnight at room temperature with maleimidocaproic acid N-hydroxy-succinimide ester. The resulting homobifunctional peptide crosslinker can be purified by reverse phase HPLC, for example, on a Vydac 1 ⁇ 25 cm column using acetonitrile/water/TFA buffers.
  • TFA trifluoroacetic acid
  • linkers having specific lengths and flexibilities. Since polypeptides having particular secondary structures and flexibilities are well documented in the art, it is possible to judiciously design the peptide couplers with optimal length and flexibility to optimize binding to two preselected antigens on a cell surface. As will be appreciated by those skilled in the art, the separation distance between, and interaction of, the sFv′ monomers in a dimeric construct of the invention also can be modulated by the judicious choice of amino acids in the tail sequences themselves.
  • (sFv′) 2 dimers readily can be prepared wherein the resulting dimers either can be homodimeric, where the CDR sequences define the same epitope binding site, or heterodimeric, where the CDR sequences of each sFv′ monomer define different epitope binding sites.
  • the dimeric constructs of this invention preferably target a pharmacologically active drug (or other ancillary protein) to a site of interest utilizing the bivalent capability of the dimer.
  • pharmacolcogically active drugs include molecules that inhibit cell proliferation and cytotoxic agents that kill cells.
  • toxins for instance, the toxic portion of the Pseudomonas exotoxin, phytolaccin, ricin, ricin A chain, or diptheria toxin, or other related proteins known as ricin A chain-like ribosomal inhibiting proteins, i.e., proteins capable of inhibiting protein synthesis at the level of the ribosome, such as pokeweed antiviral protein, gelonin, and barley ribosomal protein inhibitor.
  • one sFv′ can be immunologically reactive with a binding site on an antigen at the site of interest
  • the second sFv′ in the dimer can be immunologically reactive with a binding site on the drug to be targeted.
  • the (sFv′) 2 dimers may have specificity for both c-erbB-2 and a pharmacologically active drug or cytotoxic agent. The resulting dimer can thus target the agent or drug to tissues expressing the c-erbB-2 antigen in vivo.
  • the construct may bind one or more antigens at the the site of interest and the drug to be targeted is otherwise associated with the dimer, for example, by crosslinking to the chemical bridge itself.
  • sFv′ 2 constructs having particular utility in targeting malignant cells, include constructs wherein one has specificity for a c-erbB-2 or related tumor antigen, and the second determinant has specificity for a different cell surface protein, such as the CD3 antigen found on cytotoxic T-lymphocytes.
  • the heterodimeric (sFv′) 2 construct then could mediate antibody dependent cellular cytotoxicity (ADCC) or cytotoxic T-lymphocyte-induced lysis of the tumor cells expressing the c-erbB-2 antigen.
  • Still another bispecific dimeric construct having cytotoxic properties is a bispecific construct with one sFv′ capable of targeting a tumor cell and the second being a catalytic sFv′ that binds an inactive drug, and subsequently converts it into an active compound (see for example, U.S. Pat. No. 5,219,732).
  • a construct would be capable of inducing the formation of a toxic substance in situ.
  • a catalytic sFv′ molecule having ⁇ -lactamase-like activity can be designed to bind and catalyze the conversion of an inactive lactam derivative of doxorubicin into the active, cytotoxic form.
  • the bispecific dimer having binding affinities for both the preselected antigen and the cytotoxic-lactam derivative, is administered to an individual and allowed to accumulate at the desired location.
  • the inactive, nontoxic cytotoxin-lactam derivative then is administered to the individual.
  • the derivative is complexed with the bispecific (sFv′) 2 heterodimer in situ the active form of the drug is released, enhancing both the cytotoxicity and specificity of the drug.
  • a humanized single-chain Fv is envisioned whereby the recombinant sFv′ contains CDRS of the murine 741F8 antibody interposed between human FR sequences to generate a humanized c-erbB-2 binding protein.
  • the humanized Fv would be capable of binding c-erbB-2 while eliciting little or no immune response when administered to a patient.
  • a nucleic acid sequence encoding a humanized sFv may be designed and constructed as follows.
  • FR regions identified by homology searches of the GenBank database can be introduced into an sFv of interest by site-directed mutagenesis to reproduce the corresponding human sequence.
  • homologous human V H and V L sequences can be derived from a collection of PCR-cloned human V regions, after which the human FR sequences can be ligated with murine CDR regions to create humanized V L and V H genes.
  • a humanized sFv hybrid thus can be created, for instance, where the human FR regions of the human myeloma antibody are introduced between the murine CDR sequences of the murine monoclonal antibody 741F8.
  • the resulting sFv, containing the sequences FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4 contains a murine binding site in a human framework.
  • the resulting sFv DNA constructs then are integrated into expression vectors and transfected into appropriate host cells for protein expression. After being translated, the protein may be purified from the cells themselves or recovered from the culture medium.
  • the expression vectors also may include various sequences to promote correct expression of the recombinant protein. Typical sequences include transcription promoters and termination sequences, enhancer sequences, preferred ribosome binding site sequences, preferred mRNA leader sequences, preferred protein processing sequences, preferred signal sequences for protein secretion, and the like.
  • the DNA sequence encoding the gene of interest also may be manipulated to remove potentially inhibiting sequences or to minimize unwanted secondary structure formation.
  • the resulting synthetic genes can be expressed in appropriate prokaryotic hosts such as various strains of E. coli, or in eucaryotic hosts such as Chinese hamster ovary cells (CHO), mouse myeloma, hybridoma, transfectoma, and human myeloma cells.
  • the currently preferred expression system for the present invention is E. coli, as disclosed herein.
  • the gene When the gene is to be expressed in E. coli, it is cloned into an expression vector downstream of a strong promoter sequence, such as Trp or Tac, and optionally also may include a gene coding for a leader polypeptide, such as the fragment B (FB) of staphylococcal protein A.
  • a strong promoter sequence such as Trp or Tac
  • FB fragment B
  • the resulting fusion protein when expressed, accumulates in retractile bodies (also known as inclusion bodies) in the cytoplasm, and may be harvested after disruption of the cells by French press or sonication. The proteins then are solubilized, and refolded in vitro, as described herein.
  • the construct is engineered as a fusion protein, the protein is solubilized and the leader sequence preferably cleaved before renaturation.
  • the cleavage site for the leader sequence preferably is immediately adjacent to the sFv polypeptide chain and includes one amino acid or a sequence of amino acids exclusive of any one amino
  • the cleavage site preferably is designed for specific cleavage by a selected agent. Endopeptidases are preferred, although non-enzymatic (e.g., chemical) cleavage agents may be used. Many useful cleavage agents, for instance, cyanogen bromide (CNBr), dilute acid, trypsin, Staphylococcus aureus V-8 protease, post-proline cleaving enzyme, blood coagulation Factor Xa, enterokinase, and renin, recognize and preferentially or exclusively cleave at particular cleavage sites.
  • One currently preferred peptide sequence cleavage agent is V-8 protease.
  • the currently preferred cleavage site is at a Glu residue.
  • cleavage site e.g., factor Xa (Ile-Glu-Gly-Arg) or enterokinase (Asp-Asp-Asp-Asp-Lys).
  • factor Xa Ile-Glu-Gly-Arg
  • enterokinase Asp-Asp-Asp-Asp-Lys.
  • Dilute acid preferentially cleaves the peptide bond between Asp-Pro residues, and CNBr in acid cleaves after Met, unless it is followed by Tyr.
  • the engineered gene may be incorporated into a vector without a sequence encoding a leader polypeptide, and the engineered gene expressed to produce a polypeptide chain that is secreted into the E. coli periplasmic space.
  • the secreted protein then can be isolated and, optionally, purified further using standard methodologies. (See, for example, Pack et al. (1992) Biochem 31:1579-1584.)
  • the engineered gene is to be expressed in eucaryotic hybridoma cells
  • the conventional expression host for immunoglobulins the gene preferably is inserted into an expression vector containing, for example, the immunoglobulin promoter, a secretion signal, and immunoglobulin enhancers.
  • This plasmid also may contain sequences encoding other polypeptide chains, including part or all of a toxin, enzyme, cytokine, or hormone.
  • the gene then is transfected into myeloma cells via established electroporation or protoplast fusion methods.
  • the transfected cells then may express V H -linker-V L -tail or V L -linker-V H -tail single-chain Fv′ polypeptide chains.
  • the sFv polypeptide chains can be expressed as either inactive or active polypeptide chains.
  • Spontaneously refolded sFv polypeptide chains can be obtained from either prokaryotic or eukaryotic expression systems when the polypeptide chains are secreted for instance, either into the E. coli periplasmic space or the mammalian cell culture medium. These spontaneously refolded polypeptide chains readily can be purified by affinity chromatography. Where the sFv polypeptide chains are obtained in an unfolded, inactive sFv form, for instance, when overexpression of the sFv polypeptide chain in E. coli results in the formation of inclusion bodies, the proteins can be refolded in vitro.
  • inclusion bodies are harvested by centrifugation, the sFv, solubilized with denaturants such as guanidine hydrochloride (GuHCl) or urea, and then refolded by dilution of the denaturant under appropriate redox (reduction/oxidation) conditions (see below).
  • denaturants such as guanidine hydrochloride (GuHCl) or urea
  • refolded sFv polypeptide chains then can be purified by affinity chromatography. Details for the isolation of inclusion bodies, solubilization and renaturation of the sFv polypeptide chains are well known in the art (see for example, U.S. Pat. No. 5,091,513 and Huston et al., 1988, supra).
  • the sFv′ monomers of the present invention can be dimerized in vivo or in vitro.
  • two sFv′ genes can be cotransfected into the host cell wherein the coexpressed sFv′ polypeptide chains spontaneously dimerize.
  • the refolded, secreted sFv′ polypeptide chain monomers can be isolated from two expression hosts and subsequently dimerized in vitro.
  • the sFv′ polypeptide chains comprising a single cysteine C-terminal tail residue are expressed in E. coli and form inclusion bodies.
  • the resulting sFv′ polypeptide chains are solubilized with denaturants and renatured in vitro, either in the presence or absence of exogenously added glutathione.
  • the additional C-terminal cysteine residues apparently do not interfere with the refolding process.
  • sFv and sFv′ constructs may refold poorly in vitro. These constructs can be “preoxidized prior” to refolding as taught in Huston et al., (1991) Meth. Enzymol.
  • polypeptide chains can be secreted across a membrane bilayer.
  • the latter process spontaneously separates the extra C-terminal cysteine residue from the cysteine residues normally found in the Fv domain, minimizing inappropriate disulfide bond formation. Secretion is the preferred method if the sFv′ constructs refold poorly in vitro.
  • (sFv′) 2 dimers readily can be prepared in vitro by air oxidation if cysteine amino acids are present in the C-terminal tail sequences.
  • sulfhydryl specific crosslinking reagents for instance, the BMH crosslinker or the MCA-peptide-MCA bridge may be used to covalently couple two sFv′ chains.
  • the resultant homo or heterodimers then can be purified by standard size exclusion chromatography.
  • a preferred purification protocol uses a sequential two step affinity chromatography procedure.
  • the heterodimer is exposed to a first chromatographic system having an epitope that interacts specifically with one sFv of the heterodimer.
  • the eluant containing the heterodimer is then exposed to a second system having an epitope that interacts specifically with the other sFv′.
  • a first chromatographic system having an epitope that interacts specifically with one sFv of the heterodimer.
  • the eluant containing the heterodimer is then exposed to a second system having an epitope that interacts specifically with the other sFv′.
  • the dimeric constructs may be administered either by intravenous or intramuscular injection. Effective dosages for the single-chain Fv constructs in antitumor therapies or in effective tumor imaging can be determined by routine experimentation, keeping in mind the objective of the treatment.
  • the pharmaceutical forms suitable for injection include sterile aqueous solutions or dispersions.
  • the form must be sterile and must be fluid so as to be easily administered by syringe. It must be stable under the conditions of manufacture and storage, and must be preserved against the contaminating action of,microorganisms. This may, for example, be achieved by filtration through a sterile 0.22 micron filter and/or lyophilization followed by sterilization with a gamma ray source.
  • Sterile injectable solutions are prepared by incorporating the desirable amount of the constructs, disclosed herein, into an appropriate solvent, such as sodium phosphate buffered saline (PBS), followed by filter sterilization.
  • a physiologically acceptable carriers includes any and all solvents, dispersion media, antibacterial and antifungal agents that are non-toxic to humans, and the like. The use of such media and agents as pharmaceutically active substances are well known in the art. The media or agent must be compatible with maintenance of proper conformation of the single-chain Fv polypeptide chains, and its use in the therapeutic compositions. Supplementary active ingredients can also be incorporated into the compositions.
  • a preferred remotely detectable moiety for in vivo imaging includes the radioactive atom Technetium ⁇ 99m ( 99m Tc), a gamma emitter with a half-life of about 6 hours.
  • Non-radioactive moieties also useful in imaging include nitroxide spin labels as well as lanthanide and transition metal ions all of which induce proton relaxation in situ.
  • the complexed radioactive moieties may be used in standard radioimmunotherapy protocols to destroy the targeted cell.
  • Preferred nucleotides for high dose radioimmunotherapy include the radioactive atoms 90 Yttrium ( 90 Yt), 131 Iodine ( 131 I) and 111 Indium ( 111 In).
  • Either the single polypeptide chain sFv′ itself, or the spacer groups for linking the sFv′ constructs can be labeled with radioisotopes such as 131 I, 111 In and 99m Tc. 99m Tc and 111 In are preferred because they can be detected with gamma cameras and have favorable half-lives for in vivo imaging applications.
  • the single polypeptide chains can be labeled, for example, with radioactive atoms such as 90 Ty, 99M Tc or 111 I via a conjugated metal chelator (see, e.g., Khaw et al., 1980, Science 209: 295; U.S. Pat. No. 4,472,509; U.S. Patent No. 4,479,930), or by other standard means of linking isotopes to proteins, known to those with skill in the art (see for example, Thankur et al., 1991, J. Immuniol. Methods 237: 217-224).
  • V H and V L genes of the 741F8 anti-c-erbB-2 monoclonal antibody were isolated from the cDNA of the parental 741F8 hybridoma line by PCR using primers homologous to the N-terminal coding regions of V H , V L , C H 1, and C L .
  • the PCR-amplified V H and V L genes were isolated by polyacrylamide gel electrophoresis and cloned into a pUC cloning vector.
  • the first FR region of the 741F8 V H gene however contained spurious mutations due to the PCR procedure. Errors were rectified by the replacement of the first 70 nucleotides of 741F8 V H with a similar sequence from 520C9 V H , another c-erbB-2 specific monoclonal antibody.
  • Restriction sites then were introduced into the ends of the heavy and light chain variable gene segments by site-directed mutagenesis (Kunkel et al., 1985, Proc. Natl. Acad. Sci. USA 82: 488-492).
  • a Nco I site encoding methionine was positioned at the N-terminus of V H for expression in E. coli .
  • a Sac I site was created at the 3′ end of V H gene.
  • a Xho I site, together with an adjacent Eco RV site, were created at the N-terminus of V L .
  • a stop codon and a Pst I site were placed at the. C-terminal end of V L .
  • the single-chain Fv gene was constructed by connecting the V H and V L genes together with a DNA sequence encoding the 14 residue polypeptide linker, (Ser 4 Gly) 2 Ser 4 ′ as set forth as amino acids 122 through 135 in the Sequence Listing as SEQ. ID. NOS.: 1 and 2.
  • a synthetic DNA duplex encoding the C-terminal amino acid sequence, (Gly) 4 -Cys was inserted into a Hpa I site located near the stop codon at the 3′ end of the 741F8 sFv gene.
  • the resulting 741F8 anti-c-erbB-2 sFv′ gene was excised from the pUC cloning vector, with the restriction enzymes Nco I and Bam HI (a Bam HI site is located 3′ to the C-terminal Pst I site), and inserted into the same sites of a commercial T7 expression vector pET-3d (In-vitrogen, Inc.).
  • the resulting gene set forth in the Sequence Listing as SEQ. ID. NOS.: 1 and 2, was transformed into E. coli BL21-DE (In-vitrogen, Inc.). Protein expression was induced by the addition of IPTG to the culture medium.
  • the synthetic gene was constructed by ligating multiple synthetic DNA duplexes together.
  • the C-terminal DNA duplex coding for the amino acid sequence (Gly) 4 -Cys subsequently was ligated into a Hpa I restriction site close to the 3′ end of the 26-10 sFv gene.
  • the resulting sFv′ gene set forth in the Sequence Listing as SEQ. ID. NOS.: 3 and 4, was then inserted into the E. coli expression vector pET-3d.
  • This plasmid was subsequently transformed into E. coli BL21-DE (In-vitrogen, Inc.) and protein expression induced by the addition of IPTG to the culture medium.
  • the 520C9 sFv was generated by linking together the V H and V L genes, cloned from a 520C9 hybridoma cDNA library, with a serine rich linker. Briefly, the V H and V L genes were cloned from the 520C9 hybridoma cDNA library using probes directed toward the antibody constant (C) and joining (J) regions. Appropriate restriction sites were introduced at the ends of each gene by site-directed mutagenesis (Kunkel et al., 1985, Proc. Natl. Acad. Sci. USA 82: 488-492). The V H and V L genes were then ligated together with a serine rich linker.
  • the resulting 520C9 sFv gene set forth in the Sequence Listing as SEQ. ID. NOS.: 5 and 6, was transformed into the E. coli expression vector and expressed as described above and in co-pending U.S. Ser. No. 831,967, incorporated therein by reference.
  • Protocols for renaturing sFv monomers derived from E. coli inclusion bodies are described below.
  • the 7418, 26-10 and 520C9 sFv polypeptides were expressed in E. coli .
  • the unfolded sFv proteins were solubilized from inclusion bodies and refolded under appropriate redox conditions.
  • the refolded sFv polypeptide chains were purified by affinity chromatography or by a combination of ion-exchange and size exclusion chromatography when affinity chromatography was not feasible or expedient.
  • Inclusion bodies containing the 741F8 sFv′ proteins were washed in a buffer containing 25 mM Tris, 10 mM EDTA, 1.5 M GuHCl, pH 8.0 and solubilized in 25 mM Tris, 10 mM EDTA, 7 M GuHCl, pH 9.0 to an OD 280 nm of about 25-50.
  • the sample was reduced overnight at room temperature by the addition of dithiothreitol (DTT) to a final concentration of 10 mM.
  • DTT dithiothreitol
  • the thiol groups were converted into mixed disulfides with qlutathione by the addition of solid oxidized glutathione to a final concentration of 100 mM.
  • the solution was adjusted to pH 9.0 and incubated for 4 hr at room temperature.
  • the 741F8 sFv′ polypeptide chains then were refolded in vitro to generate stable monomers with their C-terminal cysteines remaining blocked with glutathione.
  • the 741F8 sFv′ mixed disulfide preparation was diluted to an OD 280 of about 0.15 by the addition of 10 mM Tris, 4 mM EDTA, 6 M urea, pH 8.5 at 4° C. After two hours an equal volume of 10 mM Tris, 4 mM EDTA, 1 mM reduced glutathione, pH 8.5, precooled to 4° C., was added with rapid mixing to reduce the urea concentration to 3 M. After dilution, the samples were allowed to renature for 72 hr at 4° C.
  • Inclusion bodies containing the 26-10 sFv′ proteins were washed with 25 mM Tris, 10 mM EDTA and solubilized in 6 M GuHCl, 25 mM Tris, 10 mM EDTA, pH 8.7 to an OD 280 nm of about 10 to 20.
  • the dissolved proteins were reduced by overnight incubation at room temperature after the addition of DTT to 10 mM.
  • the reduced protein could also be blocked with oxidized glutathione as noted above for the 741F8 sFv′ polypeptide.
  • the reduced, denatured 26-10 sFv′ polypeptides were refolded in a manner similar to that for the 741F8 sFv′ by diluting the preparation into a buffer containing 3 M urea, 0.1 mM oxidized and 0.01 mM reduced glutathione to give a final protein concentration of about 0.15 mg/ml. After overnight incubation at 4° C, the mixture was dialyzed against PBS containing 0.05 M KH 2 PO 4 , 0.15 M NaCl, pH 7 for two days at 4° C.
  • the inclusion bodies containing the 520C9 sFv were washed with 25 mM Tris, 10 mM EDTA, pH 8.0, 1 M GuHCl and solubilized in 25 mM Tris, 10 mM EDTA, 6 M GuHCl, 10 mM dithiothreitol (DTT), pH 9.0.
  • the material was ethanol precipitated and resuspended in 25 mM Tris, 10 mM EDTA, 6M urea, 10 mM DTT, pH 8.0 and fractionated by ion exchange chromatography to remove contaminating nucleic acids and E. coli proteins before renaturation of the sFv′.
  • the material that did not bind to a DEAE Sepharose Fast Flow (FF) column was precipitated by lowering the pH to 5.5 with 1 M acetic acid.
  • the pellet was resolubilized in 25 mM Tris, 10 mM EDTA, 6 M GuHCl, 10 mM DTT, pH 9.0 and oxidized by overnight incubation at room temperature following dilution into a buffer containing 25 mM Tris, 10 mM EDTA 6 M GuHCl, 1 mM oxidized glutathione, 0.1 mM reduced glutathione, pH 9.0.
  • the sample was dialyzed against 10 mM NaH 2 PO 4 , 1 mM EDTA, 150 mM NaCl, 500 mM urea, pH 8.0 and the sample clarified by filtration through a membrane with a 100 kD mol. wt. cut-off prior to purification on a c-erbB-2 affinity column.
  • the refolded 26-10 sFv′ polypeptide chains were purified by ouabain-Sepharose affinity chromatography, as described for the 26-10 sFv constructs (Huston, et. al., 1988, Proc. Natl Acad. Sci. USA 85; 5879-5883 and Tai, et al., 1990, Biochem. 29, 8024-3080, both of which are hereby incorporated by reference).
  • the refolded 520C9 sFv polypeptide chain was similarly purified using a c-erbB-2-agarose affinity column.
  • the refolded samples were loaded onto a c-erbB-2 affinity column, the column washed with PBS, and the 520C9 sFv polypeptides eluted with PBS pH 641 containing 3 M LiCl. The buffer was then exchanged by dialysis.
  • the c-erbB-2 affinity column preferably was prepared by linking the extracellular domain of c-erbB-2 onto agarose beads.
  • the c-erbB-2 sequence coding for its extracellular domain was derived from the baculovirus expression vector described previously (Ring et al., 1992, Mol. Immunol. 28; 915-917).
  • a DNA duplex encoding the His 6 peptide was ligated to the 3′ end of the ECD gene, and the construct expressed in CHO cells.
  • the ECD polypeptide was purified from the CHO cell culture medium on an IMAC metal affinity column (Pharmacia, Piscataway, N.J.), as described in Skerra, et al., 1991, Bio/Technology 9: 273-278, and the eluted ECD proteins attached onto agarose beads to generate the c-erbB-2-agarose affinity resin.
  • the renatured 741F8 sFv′ polypeptides were purified by a combination of ion exchange and size exclusion chromatography. Briefly, the renatured 741F8 sFv′ preparation was passed through a DEAE-cellulose column and the 741F8 sFv′ in the unbound fraction adjusted to pH.5.0 before loading on an S-Sepharose FF column. The 741F8 sFv′ polypeptide chains were eluted with PBS containing 2 mM EDTA and 3 M urea, and dialyzed against 10 mM Tris, 2 mM EDTA, 20 mM NaCl, pH 7.5 at 20° C.
  • the precipitate was harvested by centrifugation, dissolved in a suitable buffer, and passed through a Q-Sepharose FF column. The unbound material was adjusted to pH 5.5 and reloaded onto a S-Sepharose FF column.
  • the 741F8 sFv′ polypeptides were eluted with a PBS, 2 mM EDTA, 100 mM NaCl, 3 M urea buffer and dialyzed against PBS, 2 mM EDTA.
  • the precipitate was harvested again by centrifugation, dissolved in a suitable buffer, sucrose added to 5% (w/v), and the 741F8 sFv′ concentrated to 5 mg/ml in a YM10 membrane concentrator (Amicon).
  • the 741F8 sFv′ polypeptide chains were fractionated by gel filtration chromatography using a S-200 HR column (Pharmacia LKB Biotechnology) and a PBS, 2 mM EDTA buffer.
  • Dimerization of sFv′ monomers can be induced using standard crosslinking conditions. Where disulfide bond formation is desired, the monovalent sFv′ polypeptide chains initially are deblocked by mild reduction and (sFv′) 2 dimers formed by crosslinking the sFv′ polypeptides either by disulfide linkages or by thioether linkages with the BMH or MCA-peptide-MCA crosslinking reagents.
  • the purified 741F8 and 26-10 sFv′ preparations were dialyzed against 50 mM Tris, 150 mM NaCl, pH 8.5.
  • the C-terminal glutathionyl blocking groups were removed by the addition DTT to a concentration of 2 mM followed by overnight incubation at room temperature.
  • Excess reducing agent was removed by extensive dialysis against 50 mM Tris, 150 mM NaCl, pH 8.5, during which the majority of the sFv′ polypeptides oxidized into the homodimeric form.
  • sFv′ polypeptide chains in PBS first were reduced for two hours at room temperature by the addition of DTT to a final concentration of 1 mM.
  • the samples were desalted by gel filtration chromatography using a PBS, 1 mM EDTA buffer.
  • a 4-5 fold molar excess of either the BMH or MCA-peptide-MCA linkers, both dissolved in dimethylsulfoxide, were added to the reduced protein and incubated for at least 12 hours at room temperature.
  • the resulting dimers were then purified by HPLC gel filtration chromatography.
  • a modification of the procedure of Brennan, et al. (1985, Science 229: 81-83) may be used to generate disulfide linked sFv′ heterodimers.
  • a thionitrobenzoate (TNB) derivative of the 26-10 sFv′ (26-10 sFv′-TNB) was mixed with mildly reduced 741F8 sFv′.
  • the 26-10 sFv′-TNB was prepared by reducing the 26-10 sFv′ in PBS with 15 mM 2-mercaptoethylamine for 30 minutes at room temperature.
  • the reducing agent was removed by gel filtration and the reduced 26-10 sFv′ reacted with 2.2 mM dithionitrobenzoate (DTNB) for 3 hours.
  • the active 26-10 sFv′-TNB was adsorbed onto onto ouabain-Sepharose.
  • the glutathionyl blocked 741F8 sFv′ monomer in 25 mM Tris, 150 mM NaCl, pH 8.2 was reduced for 2 hours at room temperature by the addition of DTT to a final concentration of 1 mM.
  • the excess DTT was removed by gel filtration and the reduced 741F8 sFv′ reacted overnight at room temperature with the 26-10 sFv′-TNB complexed to ouabain-Sepharose.
  • the progress of the reaction was monitored spectroscopically at 412 nm, the absorbance maximum of the TNB anion.
  • the (sFv′) 2 homodimers may be separated from the sFv′ monomers by gel filtration chromatography. Following dimerization, the sFv′ preparations are dialyzed against PBS containing 1 mM EDTA, 3M urea, 0.03% azide, to disrupt any non-covalent homodimers and fractionated by HPLC on a TSK-G20000SW column using the same buffer. The procedure requires two passes for purification of the (sFv′) 2 homodimers to homogeneity. The purified hoinodimers may be dialyzed either against PBS or any other suitable buffer prior to use.
  • the (sFv′) 2 heterodimers can be separated by a two step affinity chromatography procedure taking advantage of the bivalent nature of the dimer. For instance, during the the purification of the 741F8/26-10 heterodimer the mixture initially was loaded onto an ouabain-Sepharose column, washed with a PBS, 1 M NaCl buffer, to remove any non-specifically adsorbed material, and rewashed with PBS to reduce the salt concentration. The reactive 26-10 sFv′ species bound to the resin were eluted with 20 mM ouabain in PBS and the eluate dialyzed against PBS to remove the cardiac glycoside.
  • the 741F8/26-10 heterodimers were then repurified on a c-erbB-2-agarose affinity column taking advantage of the ECD binding site in the heterodimer. After the preparation was loaded onto the c-erbB-2 affinity column, it is washed with PBS and the (sFv′) 2 heterodimer eluted with 25 mM Tris, 10 mM EDTA, 5M LiCl, pH 6.8. Prior to use, the buffer was exchanged with PBS by dialysis.
  • the sFv′ polypeptides may be labeled by the chloramine-T method as described (DeNardo, et al., 1986, Nucl. Med. Biol. 13: 303-310). Briefly, 1.0-2.0 mg of sFv′ was combined with 125 I [14-17 mCi/ ⁇ g] (Amersham, Arlington Heights, Ill.) at an iodine to protein ratio of 1:10 in a 12 ⁇ 75 mm plastic test tube. 10 ⁇ l (1 mg/ml] of chloramine-T (Sigma, St. Louis, Mo.) per 100 ⁇ g of protein was added and the mixture incubated for three minutes at room temperature.
  • SK-Br-3 breast cancer cells (Ring et al., 1989, Cancer Res. 49: 3070-3080), were harvested and resuspended in 10 mM NaCl, 0.5% Nonidet-P40, pH 8. Insoluble debris was removed by centrifugation and the extract filtered through 0.45 Millex HA and 0.2 Millex GV filters. 40 ⁇ l of the extract was added to each well of a 96 well plate and incubated overnight at 37° C. The plates then were washed with PBS and non-specific binding sites blocked following the addition of PBS containing 1% skim milk by incubation for one hour at room temperature. The sFv and 520C9 Fab samples, diluted in PBS, were added to the wells and incubated for 30 mins at room temperature. A control containing only dilution buffer was also included.
  • FIG. 3 compares the binding ability of the parental 520C9 Fab fragment, together with the 520C9 sFv single-chain binding protein.
  • the 520C9 sFv samples included the material obtained following renaturation of the polypeptide in vitro, a sample purified on a c-erbB-2 agarose affinity column, and the material that did not bind to the column.
  • the fully purified 520C9 sFv polypeptide exhibits an affinity for c-erbB-2 indistinguishable from the parent 520C9 Fab fragment.
  • SK-OV-3 cells a human ovarian cancer cell line that expresses c-erbB-2 on the cell surface
  • SK-OV-3 cells a human ovarian cancer cell line that expresses c-erbB-2 on the cell surface
  • the radiolabeled sFv′ and Fab preparations were diluted in PBS for these studies.
  • the biodistribution of the glutathionyl-blocked 741F8 sFv′ monomers, and the 741F8 and 26-10 (sFv′) 2 constructs were compared after identical doses of the radiolabeled protein was administered by injection in each case.
  • the total injected doses were determined by counting each animal on a Series 30 multichannel analyzer/probe system (probe model #2007, Canaberra, Meridian, Conn.). Groups of 3-6 mice were sacrificed twenty four hours after injection, the tumors and organs were removed, weighed and counted in a gamma counter to determine the amount of radiolabel incorporated into the tissues.
  • the disulfide linked 741F8 (sFv′) 2 homodimers exhibit identical tumor specificities when compared to the monomeric 741F8 sFv′ polypeptide chains.
  • the T:O ratios of the 741F8 sFv′ constructs consistently exceed those for the 26-10 sFv′ constructs, demonstrating the binding specificity of the 741F8 constructs for the tumors ( FIG. 4 ).
  • the 741F8 (sFv′ 2 dimers generally exhibit higher T:O ratios relative to that of the monomeric species, particularly for the disulfide bonded sFv′ 741F8 (sFv′-(Gly) 4 Cys) 2 and the MCA linked 741F8 (sFv′ ) 2 homodimers.
  • the 741F8 (sFv′ ) 2 homodimers localize in greater amounts in the tumors relative to the monomeric sFv′ species ( FIG. 5 ).
  • the tumor localization properties of all of the dimeric 741F8 (sFv′ ) 2 constructs are significantly greater than those observed with the 741F8 Fab, the 741F8 sFv′ monomer and the 26-10 (sFv′) 2 dimer ( FIG. 6 ).
  • the results demonstrate that the increased apparent avidity and enhanced in vivo imaging of the (sFv′) 2 dimer is due, at least in part, to its improved retention in tumor tissue.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Oncology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Reproductive Health (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed is a formulation for targeting an epitope on an antigen expressed in a mammal. The formulation comprises a pharmaceutically acceptable carrier together with a dimeric biosynthetic construct for binding at least one preselected antigen. The biosynthetic construct contains two polypeptide chains, each of which define single-chain Fv (sFv) binding proteins and have C-terminal tails that facilitate the crosslinking of two sFv polypeptides. The resulting dimeric constructs have a conformation permitting binding of a said preselected antigen by the binding site of each said polypeptide chain when administered to said mammal. The formulation has particular utility in in vivo imaging and drug targeting experiments.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of co-pending application Ser. No. 831,967 filed Feb. 6, 1992, incorporated herein by reference.
  • The U.S. Government may have certain rights in the invention described herein, by virtue of National Institutes of Health Grant No. UO1 CA51880.
  • FIELD OF THE INVENTION
  • This invention relates in general to novel biosynthetic compositions of matter having particular utility as in vivo targeting agents and more specifically, to biosynthetic dimeric constructs of single-chain binding proteins (sFv), conjugates thereof, and to methods for their production.
  • BACKGROUND OF THE INVENTION
  • The development of murine monoclonal antibodies and their proteolytic Fab fragments has raised interest in their utility as diagnostic and therapeutic reagents for in vivo imaging and drug targeting. However, successful in vivo targeting of radionuclides, drugs or toxins using 150 kD intact antibodies or their 50 kD Fab fragments (an antibody fragment consisting of one light chain and approximately half of the heavy chain held together by a single disulfide bond) have been restricted by the limited penetration of these molecules from the vasculature into the tissues of interest, and by their slow clearance rates in vivo, which for IgG leads to behavior that requires several days to clear the background enough for imaging to be possible. Other disadvantages of the intact antibodies or their Fab fragments include: their immunogenicity when prepared from different species, their non-specific binding to many normal tissues and organs, and the fact that they contain multiple proteolytic cleavage sites which result in their degradation during their circulation in vivo.
  • Although Fv fragments, which consist of one VH and one VL domain held together by noncovalent interactions, form the minimal region of an antibody that contains a complete antigen combining site, dissociation of the VH and VL domains in vivo can preclude their use as therapeutic or imaging agents.
  • Although Moore et al., (U.S. Pat. No. 4,642,334) and Glockshuber et al., (1990, Biochem. 29, 1362-1367) disclose attempts to stabilize these Fv fragments with engineered intermolecular disulfide bonds, monovalent 50 kD Fab and Fab, fragments have, until recently, been the smallest proteins available for effective immunotargeting.
  • Recently, single-chain Fv (sFv) polypeptide chains of about 27 kD have been developed containing covalently linked VH-VL polypeptides. The VH- and VL-domains are connected by a polypeptide linker. The resulting sFv polypeptide chains are also referred to in the art as biosynthetic antibody binding sites or BABS and preferably are encoded by a single DNA sequence. For a detailed description of these biosynthetic polypeptide chains see for example, Huston et al., 1988, Proc. Nat. Aca. Sci. USA 85: 5879-5883 or U.S. Pat. Nos. 5,091,513 and 5,132,405, all of which are hereby incorporated by reference. The sFv polypeptide chains provide attractive alternatives to intact immunoglobulins and Fab fragments due to their small size and their stability at concentrations that typically promote dissociation of natural Fv fragments. U.S. Pat. Nos. 5,091,513 and 5,132,405; Huston et al., ((1991) Methods in Enzymology 203: 46-88; Huston et al (1993) Int. Rev. Immunol. 10: 195-217) disclose the utility of sFv polypeptides, as well as single chain constructs synthesized from single DNA sequences, which may further comprise ancillary effector proteins, such as a second sFv or a cytotoxic agent.
  • Pack et al. ((1992) Biochem 31: 1579-1584) disclose the construction of “mini-antibodies”. The mini-antibodies are sFv polypeptide chains which also include an 11oligomerization domain” at their C-termini, separated from the sFv by a hinge region. The oligomerization domains comprise self-associating α-helices, for example, leucine zippers, that can be further stabilized by additional disulfide bonds. The domains a;re designed to be compatible with vectorial folding across a membrane, a process thought to facilitate in vivo folding of the polypeptide into a functional binding protein.
  • PCT application PCT/US92/09965, published Jun. 10, 1993 also discloses the construction of bivalent sFv constructs, including crosslinked dimers.
  • However, the pharmacokinetic properties of these constructs or those disclosed by Pack et al. are not measured in vivo.
  • PCT application PCT/US92/07986, published Apr. 1, 1993 discloses bifunctional (Fab′)2 molecules composed of two Fab′ monomers linked through cysteine amino acids located at the C-terminus of the first constant domain of each heavy chain. PCT application PCT/US92/10140, published Jun. 10, 1993 also discloses bifunctional (Fab′)2 dimers which, in addition to the cysteine residues located in the hinge region, also contain C-terminal leucine zipper domains that further stabilize the (Fab′)2 dimers. In both cases, the resulting (Fab′)2 dimers (≧100 kD in size), although smaller than intact immunoglobulins, are significantly larger than sFv polypeptides and are anticipated to have slower tissue biodistribution and clearance rates following in vivo administration.
  • Cumber et al. disclose the generation of (Fv-Cys)2 heterodimers by chemically crosslinking two VH-cys domains together (Cumber et al., 1992, J. Immunology 149B: 120-126). Although the crosslinked VH chains appear to be stable, dissociation of the VL polypeptides from each Fv reduces the pharmacological value of these constructs in vivo.
  • It is an object of the instant invention to provide biosynthetic constructs having enhanced pharmacokinetic properties as in vivo targeting agents.
  • In particular, it is an object of this invention to provide biocompatible constructs having accelerated in vivo biodistribution and body clearance rates than that of antibodies or antibody fragments. It is another object of the invention to provide biosynthetic constructs having enhanced avidity in vivo, including enhanced target tissue specificity and target tissue retention. Yet another object is to provide dimeric biosynthetic constructs having improved tissue imaging and drug targeting properties in vivo. Still another object is to provide diagnostic and therapeutic formulations comprising these constructs, having particular utility in the diagnosis and treatment of malignancies. Still another object is to provide constructs having enhanced pharmacokinetic properties as in vivo targeting agents, particularly as in vivo imaging agents, for ovarian and breast tumor tissue.
  • These and other objects and features of the invention will be apparent from the description, figures and claims which follow.
  • SUMMARY OF THE INVENTION
  • In its broadest aspect, the invention features a formulation for targeting an epitope on an antigen expressed in a mammal, where the formulation contains a pharmaceutically acceptable carrier in combination with a biosynthetic construct for binding at least one preselected antigen. The dimeric construct has particular utility in diagnostic and therapeutic applications in vivo.
  • The invention features the synthesis and use of monomers and dimers of polypeptide constructs belonging to the class of proteins known as single-chain Fv (sFv) polypeptides. The sFv proteins described herein have superior in vivo pharmacokinetic properties, including accelerated tissue biodistribution and clearance rates relative to either intact IgG, (Fab)2 dimers or Fab.
  • The dimeric biosynthetic construct of the invention contains two sFv polypeptide chains defined herein as follows. Each sFv polypeptide chain comprises an amino acid sequence defining at least two polypeptide domains. These domains are connected by a polypeptide linker spanning the distance between the C-terminus of one domain and the N-terminus of the other.
  • The amino acid sequence of each domain includes complementarity determining regions (CDRS) interposed between framework regions (FRs) where the CDRs and FRs of each polypeptide chain together define a binding site immunologically reactive with a preselected antigen. Additionally, each biosynthetic binding site polypeptide chain can have an amino acid sequence peptide bonded and thus contiguous with the C-terminus of each polypeptide chain, referred to herein as a “C-terminal tail” sequence. The term “sFv′” refers hereinafter, to an sFv molecule containing such a C-terminal tail sequence. This tail sequence preferably does not contain an α-helical motif that self-associates with another polypeptide chain of similar sequence but still contains a means for covalently crosslinking two such polypeptide chains together. When the two sFv′ polypeptide chains are crosslinked together, the resulting dimeric construct has a conformation that permits the independent binding of a preselected antigen or antigens to the binding site of each polypeptide chain in vitro and in vivo. The resulting dimeric constructs have superior in vivo pharmacokinetic properties that include significantly enhanced avidity, including enhanced target tissue retention and/or antigen localization properties, as compared with intact IgG, Fab, (Fab)2 dimers or monomeric sFv.
  • As will be appreciated by those having ordinary skill in the art, the sequence referred to herein generally as a “C-terminal tail” sequence, peptide bonded to the C-terminus of an sFv and comprising means for crosslinking two sFv polypeptide chains, alternatively may occur at the N-terminus of an sFv (“N-terminal tail”) or may comprise part of the polypeptide linker spanning the domains of an individual sFv. The dimeric species created by the crosslinking of sFvs having these alternative “tail” sequences also are contemplated to have a conformation permitting the in vivo binding of a preselected antigen by the binding sites of each of the sFv polypeptide chains. Accordingly, descriptions of how to make and use sFv′ monomers and dimers comprising a C-terminal tail sequence are extended hereby to include sFv monomers and dimers wherein the tail sequence having crosslinking means occurs at the N-terminus of an sFv or comprises part of the polypeptide linker sequence.
  • In one embodiment, both polypeptide chains bind the same epitope on a preselected antigen, and the resulting dimeric construct is termed a “homodimer.” In another embodiment, the polypeptide chains bind different epitopes on a preselected antigen and the resulting dimeric construct is termed a “heterodimer.” In still another embodiment, the two polypeptide chains bind different epitopes on two different, preselected antigens.
  • The term “epitope”, as used herein, refers to a portion of an antigen that makes contact with a particular antibody or antibody analogue. In a typical protein, it is likely that any residue accessible from the surface can form part of one or more antigenic determinants. The term “antigen”, as used herein, refers to a molecule that can elicit an immune response and that can react specifically with corresponding antibodies or antibody analogues.
  • The term “domain”, as used herein, refers to an amino acid sequence that folds into a single globular region in its native conformation, and which may exhibit discrete binding or functional properties. The term “polypeptide linker”, as used herein, refers to an amino acid sequence that links the C-terminus of one domain to the N-terminus of the other domain, while still permitting the two domains to maintain their proper physiologically active binding conformations.
  • In a particular aspect of the invention, the currently preferred polypeptide linkers that connect the C-terminus of one domain to the N-terminus of the other domain include part or all of amino acid sequence ((Gly)4 Ser)3 set forth in the SEQ. ID. NO.: 7, or ((Ser)4 Gly)3 as set forth in SEQ. ID. NO.: 8.
  • The amino acid sequence of each of the polypeptide domains includes complementarity determining regions interposed between framework regions. The term complementarity determining regions” or “CDRs”, as used herein, refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native immunoglobulin binding site, or a synthetic polypeptide which mimics this function. CDRs are not necessarily wholly homologous to hypervariable regions of natural Fv molecules, and also may include specific amino acids or amino acid sequences which flank the hypervariable region and have heretofore been considered framework not directly determinative of complementarity. The term “framework regions” or “FRs”, as used herein, refers to amino acid sequences which are found naturally occurring between CDRs in immunoglobulins. These FR sequences may be derived in whole or part from the same immunoglobulin as the CDRs, or in whole or part from a different immunoglobulin. For example, in order to enhance biocompatibility of an sFv to be administered to a human, the FR sequences can be derived from a human immunoglobulin and so the resulting humanized sFv will be less immunogenic than a murine monoclonal antibody.
  • The amino acid sequence of each variable domain includes three CDRs interspersed between four FRs. The two polypeptide domains that define an sFv molecule contain CDRs interspersed between FRs which together form a binding site immunologically reactive with a preselected antigen. The term “immunologically reactive”, as used herein, refers to the noncovalent interactions of the type that occur between an immunoglobulin molecule and an antigen for which the immunoglobulin is specific. As used herein, the term “avidity” describes the stability of a complex formed by a multivalent antibody or antibody analogue, with its binding conjugate. Also as used herein, the term “apparent avidity” describes the stability of a complex formed by an antibody or an antibody analogue with its binding conjugate as determined by in vivo immunolocalization studies.
  • In a preferred aspect of the invention, the CDRs of the polypeptide chain can have an amino acid sequence substantially homologous with at least a portion of the amino acid sequence of CDRs from a variable region of an immunoglobulin molecule from a first species, together with FRs that are substantially homologous with at least a portion of the amino acid sequence of FRs from a variable region of an immunoglobulin molecule from a second species. Preferably, the first species is mouse and the second species is human. The CDR sequences in the sFv′ polypeptides are preferably substantially homologous to an immunoglobulin CDR retaining at least 70%, or more preferably 80% or 90%, of the amino acid sequence of the immunoglobulin CDR, and also retains the immunological binding properties of the immunoglobulin.
  • Each sFv′ molecule has a C-terminal polypeptide tail that has a non-self-associating structure and contains at least one crosslinking means. Useful crosslinking means include derivatizable amino acid side chains, particularly those selected from the group consisting of cysteine, lysine, arginine, histidine, glutamate, aspartate, and derivatives and modified forms thereof. In a preferred aspect of the invention, cysteine amino acids are incorporated into the C-terminal tail sequences as the crosslinking means. In another aspect of the invention, the crosslinking means includes one or more amino acids that can be posttranslationally modified. For example, the crosslinking means can include one or more glycosylation sites, wherein the incorporated carbohydrate moieties can be crosslinked in vitro. Preferred glycosylation sequences include Asn-Xaa-Thr and Asn-Xaa-Ser, where Xaa can be any amino acid, wherein the carbohydrate is typically N-linked to asparagine or O-linked to serine or threonine.
  • Additionally, the tail also may comprise an amino acid sequence that defines a metal ion chelation motif, and which facilitates purification of the sFv′ monomers by metal ion affinity chromatography, such as the IMAC2+ chromatography system. Furthermore, chelation motifs can be used for binding detectable moieties, such as Technetium−99m (99mTc) for in vivo imaging. Preferred examples of useful C-terminal tail amino acid sequences wherein the crosslinking means is provided by the sulfhydryl group of a cysteine, include: Ser-Cys; (Gly)4-Cys; and (His)6-(Gly)4-Cys; set forth in the Sequence Listing as SEQ. ID. NOS.: 9, 10 and 11, respectively. The (Gly)4-Cys sequence facilitates the coordination of 99mTc by this tail.
  • In the present invention, monomeric sFv′ molecules can be coupled together through the crosslinking means in the C-terminal tails to form either homo- or heterodimeric (sFv′)2 species. The term “sFv coupler”, as used herein, refers to the chemical bridge that links two sFv′ polypeptide chains together to form a dimeric species. In a preferred aspect of the invention, where the crosslinking means is a cysteine residue, the linkage is by a disulfide bond. Alternatively, sulfhydryl-specific homobifunctional crosslinking reagents, such as bismaleimidohexane, or heterobifunctional crosslinking reagents, can be used to join the two sFv′ molecules together. sFv couplers of preselected length also can be designed to limit interaction between the two sFv′ polypeptide chains or to optimize binding of two preselected antigens, including, for example, multiple copies of a receptor expressed on a cell surface in a mammal. An example of such a variable length coupler includes the bismaleimidocaproyl amino acid (MCA) synthetic peptide bridge. Although, in a preferred aspect of the invention a GlySer3Gly2 Ser3Lys peptide spacer is used, in theory, any amino acid sequence can be introduced into this type of chemical bridge with a variety of reactive moieties at either end. Consequently, it is possible to design specific linkage groups that can have a predetermined length and flexibility. If a substantially inflexible coupler is desired, then for instance, a polylysine or polyproline peptide may be used. Another benefit of the MCA linkers over many other commercially available linkers is that they are soluble in water. Moreover, the chemical bridge also may be created to enhance the imaging or therapeutic properties of the construct in vivo (vide infra). As will be appreciated by those having ordinary skill in the art, the separation distance between, and interaction of, the sFv′ monomers in a dimeric construct of the invention also can be modulated by the judicious choice of amino acids in the tail sequences themselves.
  • The dimeric constructs of this invention preferably target a pharmacologically active drug (or other ancillary protein) to a site of interest utilizing the bivalent capability of the dimer. Examples of pharmacologically active drugs include molecules that inhibit cell proliferation and cytotoxic agents that kill cells. The term “cytotoxic agent”, as used herein, refers to any molecule that kills cells, and includes anti-cancer therapeutic agents such as doxorubicin. Other, useful molecules include toxins, for instance, the toxic portion of the Pseudomonas exotoxin, phytolaccin, ricin, ricin A chain, or diptheria toxin, or other related proteins known as ricin A chain-like ribosomal inhibiting proteins, i.e., proteins capable of inhibiting protein synthesis at the level of the ribosome, such as pokeweed antiviral protein, gelonin, and barley ribosomal protein inhibitor.
  • In such cases, one sFv′ can be immunologically reactive with a binding site on an antigen at the site of interest, and the second sFv′ in the dimer can be immunologically reactive with a binding site on the drug to be targeted. Alternatively, the construct may bind one or more antigens at the the site of interest and the drug to be targeted is otherwise associated with the dimer, for example, crosslinked to the chemical bridge itself. The biosynthetic dimeric constructs of this invention also may be used as part of human therapies to target cytotoxic cells such as cytotoxic T-lymphocytes, or pharmacologically active drugs to a preselected site. A bispecific (sFv′)2 heterodimer having specificity for both a tumor antigen and a CD3 antigen, the latter of which is present on cytotoxic T-lymphocytes, thus could mediate antibody dependent cellular cytotoxicity (ADCC) or cytotoxic T-lymphocyte-induced lysis of the tumor cells.
  • Still another bispecific dimeric construct having cytotoxic properties is a bispecific construct with one sFv′ capable of targeting a tumor cell and the second sFv′ having catalytic properties that binds an inactive drug, subsequently converting it into an active compound (see for example, U.S. Pat. No. 5,219,732). Such a construct would be capable of inducing the formation of a toxic substance in situ. For example, a catalytic sFv′ molecule having β-lactamase-like activity can be designed to bind and catalyze the conversion of an inactive lactam derivative of doxorubicin into its active form. Here the bispecific dimer, having binding affinities for both the preselected antigen and the inactive-lactam derivative, is administered to an individual and allowed to accumulate at the desired location. The inactive and nontoxic cytotoxin-lactam derivative then is administered to the individual. Interaction of the derivative with the bispecific (sFv′)2 heterodimer at the site of interest releases the active form of the drug in situ, enhancing both the cytotoxicity and specificity of the drug.
  • The homo- and heterodimeric biosynthetic constructs also may include a detectable moiety bound either to the polypeptide chain, e.g., to the tail sequence, or to the chemical coupler. The term “detectable moiety”, as used herein, refers to the moiety bound to or otherwise complexed with the construct and which can be detected external to, and at a distance from, the site of the complex formation, to permit the imaging of cells or cell debris expressing a preselected antigen. Preferable detectable moieties for imaging include radioactive atoms such as Technetium−99m (99mTc), a gamma emitter with a half-life of about 6 hours. Non-radioactive moieties useful for in vivo magnetic resonance imaging applications include nitroxide spin labels as well as lanthanide and transition metal ions which induce proton relaxation in situ. In addition to immunoimaging, the complexed radioactive moieties also may be used in standard radioimmunotherapy protocols to destroy the targeted cell. Preferable nucleotides for high dose radioimmunotherapy include radioactive atoms such as, 90Yttrium (90Yt), 131Iodine (131I) or 111Indium (111In).
  • The sFv, sFv′ and (sFv′ )2 constructs disclosed herein have particular utility as in vivo targeting agents of tumor antigens, including antigens characteristic of breast and ovarian malignancies, such as the c-erbB-2 or c-erbB-2 related antigens. Accordingly, these constructs have particular utility in diagnostic applications as imaging agents of malignant cells, and in therapeutic applications as targeting agents for cytotoxins and other cancer therapeutic agents. In one preferred aspect of the invention, the CDRs of the sFv or sFv′ polypeptide chain have an amino acid sequence substantially homologous with the CDRs of the variable region of any one of the following monoclonal antibodies: 741F8, 520C9, and 454C11, all of which bind to c-erbB-2 or c-erbB-2-related antigens. Exemplary sFv′ and sFv sequences having CDRs corresponding to the monoclonal antibodies 741F8 and 520C9 are set forth in the Sequence Listing SEQ. ID. NOS.: 1 and 5, respectively.
  • The term “c-erbB-2” refers to a protein antigen that is an approximately 200 kD acidic glycoprotein having an isoelectric point of about 5.3 and having an extracellular domain overexpressed on the surface of tumor cells, such as breast and ovarian tumor cells in about 25% of cases of breast and ovarian cancer. A “c-erbB-2-related tumor antigen” is a protein located on the surface of tumor cells, such as breast and ovarian tumor cells and which is antigenically related to the c-etbB-2 antigen. That is, the related antigen can be bound by an immunoglobulin that is capable of binding the c-erbB-2 antigen (e.g. 741F8, 520C9, and 454C11 antibodies. Related antigens also include antigens comprising an amino acid sequence that is at least 80% homologous, preferably 90% homologous, with the amino acid sequence of c-erbB-2 or an amino acid sequence encoded by a DNA that hybridizes under stringent conditions with a nucleic acid sequence encoding c-erbB-2. As used herein, stringent hybridization conditions are those set forth in Sambrook, et al., 1989, Molecular Cloning; A Laboratory Manual 2nd ed. Cold Spring Harbor Press wherein the hybridization conditions, for example, include 50% formamide, 5×Denhardt's Solution, 5×SSC, 0.1% SDS and 100 μg/ml denatured salmon sperm DNA and the washing conditions include 2×SSC, 0.1% SDS at 37° C. followed by 1×SSC, 0.1% SDS at 68° C. An example of a c-erbB-2-related antigen is the receptor for the epidermal growth factor.
  • In one embodiment, the biosynthetic antibody binding site is a humanized hybrid molecule which includes CDRs from the mouse 741F8 antibody interposed between FRs derived from one or more human immunoglobulin molecule. The CDRs that bind to the c-erbB-2 epitope can be found in the amino acid residue numbers 31-37, 52-68, 101-110, 159-169, 185-191 and 224-233 in SEQ ID NOS.: 1 and 2. The hybrid molecule thus contains binding sites which are highly specific for the c-erbB-2 antigen or c-erbB-2 related antigens held in proper immunochemical binding conformation by human FR amino acid sequences, which are less likely to be recognized as foreign by the human body.
  • The dimeric (sFv′)2 construct can either be homodimeric, wherein the CDR sequences on both monomers define the same binding site, or heterodimeric, wherein the CDR sequences of each sFv′ monomer define a different binding site. An example of an (sFv′)2 heterodimer described herein having specificity for both c-erbB-2 and digoxin epitopes can be generated by combining the anti-c-erbB-2 sFv′, shown in SEQ. ID. NOS.: 1 and 2 with the anti-digoxin sFv′, shown in SEQ. ID. NOS.: 3 and 4. The CDRs that bind to the digoxin epitope can be derived from the anti-digoxin murine monoclonal antibody 26-10 (Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85: 5879-5883) and can be found in the amino acid residue numbers 32 through 36, 48 through 65, 101 through 107, 157 through 170, 188 through 194 and 229 through 234 in the Sequence Listing as SEQ. ID. NOS.: 3 and 4.
  • Radioimaging or radioimmunotherapy of tumor tissues and malignant cells are preferred aspects of this invention. Overexpression of tumor antigens such as c-erbB-2 and related cell surface antigens in malignant cells allows imaging of the malignant cell or tissue, whether it is well localized, has undergone metastasis or is exposed following cell lysis. The imaging method includes the steps of administering to a mammal a formulation comprising an sfv′ or (sFv′)2 dimeric construct having specificity for the antigen tumor and containing a detectable moiety at a concentration sufficient to permit extracorporeal detection of the construct bound to the tumor antigen; and then detecting the biosynthetic construct bound to the tumor antigen. The formulation can be used to particular advantage in gamma scintigraphy or magnetic resonance imaging. Overexpression of c-erbB-2 or related receptors on malignant cells thus allows targeting of sFv′ species to the tumor cells, whether the tumor is well-localized or metastatic. In addition, internalization of an sFv-toxin fusion protein permits specific destruction of tumor cells bearing the overexpressed c-erbB-2 or related antigen.
  • The present invention discloses monomeric and dimeric biosynthetic constructs having enhanced properties as in vivo targeting agents when compared with intact monoclonal antibodies or their Fab fragments. The dimeric biosynthetic constructs of the invention also permit the in vivo targeting of an epitope on an antigen with greater apparent avidity, including greater tumor specificity, tumor localization and tumor retention properties than that of the Fab fragment having the same CDRs as the construct. Furthermore, the dimeric constructs also permit the in vivo targeting of an epitope on an antigen with a greater apparent avidity, including greater tumor localization and tumor retention properties, than either of the monomeric polypeptides individually.
  • The invention also includes methods for producing the homo- and heterodimeric biosynthetic constructs, which include the steps of designing, constructing, expressing, purifying, and refolding the monomeric sFv′ polypeptide chains in vitro, followed by joining two polypeptide chains together through the crosslinking means in the C-terminal tail sequence, without relying on the tail structure to otherwise assist in dimer formation or enhance transport across a membrane. The invention also includes methods for imaging a preselected antigen in a mammal expressing the preselected antigen. The antigen may be expressed on a cell surface or may be released as part of the cell debris from a dying cell.
  • The foregoing and other objects, features and advantages of the present invention will be made more apparent from the following detailed description of the invention.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The foregoing and other objects of this invention, the various features thereof, as well as the invention itself, will be more fully understood from the following description, when read together with the accompanying drawings:
  • FIG. 1A is a schematic representation of a DNA construct encoding the sFv′ biosynthetic binding protein of the invention;
  • FIG. 1B is a schematic representation of the polypeptide chain encoded by the DNA construct in FIG. 1A;
  • FIG. 2A is a schematic representation of a refolded sFv′ protein in its native conformation;
  • FIG. 2B is a schematic representation showing two folded sFv′ polypeptides covalently linked by a disulfide bond;
  • FIG. 3 is a graphic representation of an in vitro competition assay comparing the c-erbB-2 binding activity of an Fab fragment of the 520C9 monoclonal antibody (filled dots), with that of biosynthetic 520C9 sFv at two different stages of purification: mixture of folded and unfolded sFv (+) or affinity-purified sFv (squares), and with a material that did not bind to the affinity column (*);
  • FIG. 4 lists in tabular form the tumor:organ ratios calculated for various sF and sFv′ species injected into tumor-containing mice;
  • FIG. 5 lists in tabular form the percentage of injected dose localized to tumor tissue for various sFv and sFv's species; and
  • FIG. 6 is a graphic representation summarizing the comparative tumor retention properties of monomeric and dimeric forms of different sFv′ constructs and Fabs represented by bars 1-6. The sFv′ species represented by bars 1-5 are based on the V regions of the 741F8 monoclonal antibody. Bar 1 refers to intravenously (i.v.) administered glutathionyl-(sFv′-SerCys) monomer, bar 2 to disulfide linked (sFv′-Gly4-Cys)2, bar 3 to MCA combined (sFv′-Ser-Cys)2, bar 4 to BMH cross-linked (sFv′-Ser-Cys)2, bar 5 to 741F8 Fab and bar 6 to the 26-10 disulfide linked (sFv′-Ser-Cys)2.
  • DETAILED DESCRIPTION OF THE INVENTION
  • It has been discovered that intravenously administered single-chain Fv (sFv) proteins exhibit superior in vivo pharmacokinetic properties relative to intact monoclonal antibodies (IgG), (Fab)2 dimers or Fab fragments. These pharmacokinetic properties include accelerated rates of tissue biodistribution, enhanced target tissue specificity, and exceptionally fast clearance rates. The sFv constructs can be designed to bind to preselected antigens and to have particular utility for in vivo immunoimaging and immunotherapy applications. In addition, it also has been discovered that dimeric forms of the constructs, which do not rely on self-associating tail sequences for dimerization or transport across a membrane, can be easily prepared and have improved target tissue localization properties, target tissue retention properties and/or avidity for their targets in vivo, relative to monomeric sFv′, Fab fragments or intact IgG.
  • In its broadest aspect, the invention features a formulation for targeting an epitope on an antigen expressed in a mammal. The formulation contains a pharmaceutically acceptable carrier in combination with a dimeric biosynthetic construct for binding at least one preselected antigen. The preselected antigen either may be an antigen expressed on the surface of a cell or an intracellular component exposed upon lysis of the cell. The sFv, sFv′ and (sFv′)2 constructs disclosed herein have particular utility as in vivo targeting agents for detecting malignant cells in a mammal. In a particularly useful embodiment, the constructs disclosed can be used to target the c-erbB-2 or c-erbB-2-related antigens which are overexpressed in certain breast and ovarian cancers. In another embodiment, radioimmunotargeting using radiolabeled (sFv′)2 constructs will be useful for therapeutic as well as diagnostic applications.
  • Provided below are detailed descriptions of biosynthetic sFv, sFv′ and (sFv′)2 dimers, useful in the compositions and methods of the invention, together with methods for their construction and administration. Also provided are numerous, non-limiting examples which demonstrate the suitability of these constructs as in vivo targeting reagents for diagnostic and therapeutic applications. More specifically, the examples demonstrate: the construction and expression of sFv polypeptides (Example 1); the renaturation, dimerization and purification of sFv′ proteins (Example 2); and the immunoreactivity of the monomeric and dimeric sFv proteins (Example 3).
  • Construction of Biosynthetic Single-chain Fv Proteins.
  • Each of the sFv and sFv′ proteins have amino acid sequences that define at least two polypeptide domains. The polypeptide domains are connected by a polypeptide linker spanning the distance between the C-terminus of one domain and the N-terminus of the other. The amino acid sequence of each domain includes complementarity determining regions (CDRs) interposed between framework regions (FRs), where the CDRs and FRs of each polypeptide chain together define a binding site immunologically reactive with a preselected antigen.
  • In the case of the sFv′ proteins, each polypeptide chain has an additional C-terminal tail amino acid sequence having a substantially non-self-associating structure. More specifically, this is a sequence that does not interact appreciably with a similar sequence under physiological conditions, as is the case for example with the α-helical leucine zipper motifs found in DNA binding proteins. Each tail sequence also contains a means for crosslinking two such sFv′ polypeptide chains together to form an (sFv′)2 dimer. The resulting (sFv′)2 dimers have conformations which permit the in vivo binding of the preselected antigen by the binding sites of each of the polypeptide chains.
  • The sFv′ constructs of this invention can be further understood by referring to the accompanying FIGS. 1 and 2. FIG. 1A is a schematic representation of the DNA construct, and FIG. 1B is a schematic representation of the resulting encoded polypeptide chain. FIG. 2 is a schematic representation of the folded sFv′ monomer (FIG. 2A) and the dimeric (sFv′)2 construct (FIG. 2B). A single-chain Fv (sFv′) polypeptide, shown in FIGS. 1 and 2A, comprises: a heavy chain variable region (VH) 10, and a light chain variable region, (VL) 14, wherein the VH and VL domains are attached by polypeptide linker 12. The binding domains defined by VL and VH include the CDRs 2, 4, 6 and 2′, 4′, 6′, respectively, and FRs 32, 34, 36, 38 and 32′, 34′, 3638′, respectively which, as shown in FIG. 2, together define an immunologically reactive binding site or antigenic determinant, 8. Furthermore, the CDRs and FRs may be derived from different immunoglobulins (see infra). The sFv′ molecules also contain a C-terminal tail amino acid sequence, 16, comprising an amino acid sequence that will not self-associate with a polypeptide chain having a similar amino acid sequence under physiological conditions, and which contains a means, 18, for the site-directed crosslinking of two such tail sequences. In a currently preferred embodiment, represented in FIGS. 1 and 2, the crosslinking means is the sulfhydryl group of a cysteine amino acid. In the monomeric form of the sFv′ the cross-linking means, 18, may be blocked by a blocking group, 20. For instance, the blocking group may be a glutathionyl moiety when the crosslinking means, 18, is a cysteine amino acid.
  • As will be appreciated by those having ordinary skill in the art, the sequence referred to herein generally as a “C-terminal tail” sequence, peptide bonded to the C-terminus of an sFv and comprising means for crosslinking two sFv polypeptide chains, alternatively may occur at the N-terminus of an sFv (“N-terminal tail”) or may comprise part of the polypeptide linker spanning the domains of an individual sFv′. The dimeric species created by the crosslinking of sFvs having these alternative “tail” sequences also are contemplated to have a conformation permitting the in vivo binding of a preselected antigen by the binding sites of each of the sFv polypeptide chains. Accordingly, descriptions of how to make and use sFv′ monomers and dimers comprising a C-terminal tail sequence are extended hereby to include sFv monomers and dimers wherein the tail sequence having crosslinking means occurs at the N-terminus of an sFv or comprises part of the polypeptide linker sequence.
  • The CDR and FR polypeptide segments are designed empirically based on sequence analysis of Fv regions of preexisting antibodies, such as those described in U.S. Pat. No. 4,753,894, hereby incorporated by reference.
  • Numerous examples of sFv polypeptide chains now exist in the art and are summarized in Huston et al., 1993, Intern. Rev. Immunol. 10: 195-217, hereby incorporated by reference.
  • The sFv and sFv′ polypeptide chains of the invention are biosynthetic in the sense that they are synthesized, transfected into a cellular host, and protein expressed from a nucleic acid containing genetic sequences based in part on synthetic DNA. Synthetic DNA is understood to include recombinant DNA made by ligation of fragments of DNA derived from the genome of a hybridoma, mature B cell clones, a cDNA library derived from natural sources, or by ligation of plural, chemically synthesized oligonucleotides. The proteins of the invention are properly characterized as “antibody binding sites”, in that these synthetic single polypeptide chains are able to refold into a 3-dimensional conformation with specificity and affinity for a preselected epitope on an antigen.
  • A detailed description for engineering and producing sFv proteins by recombinant means appears in U.S. Pat. No. 5,091,513 claiming priority from U.S. Ser. No. 052,800, filed May 21, 1987, assigned to Creative BioMolecules, Inc., hereby incorporated by reference. The polypeptide chains of the invention are antibody-like in that their structure is patterned after, regions of native antibodies known to be responsible for antigen recognition.
  • The single-chain polypeptide chains of the invention are first derived at the DNA-level. The sFv DNAs are preferably expressed in E. coli, the resulting polypeptide chains being solubilized from inclusion bodies, refolded in vitro, labeled with a detectable moiety, such as 99mTc, and dimerized to form a biosynthetic (sFv′)2 construct. Of course, the constructs disclosed herein may also be engineered for secretion from the host cell, for example, secretion into the periplasmic space of an E. coli cell, as described by Pack and Pluckthun, (Biochem., 1992, 31: 1579-1584), or into the culture supernatant of a mammalian cell (for example, as described by Traunecker, et al., 1991, EMBO J. 10: 3655-3659).
  • The ability to design the single polypeptide chains of the invention depends on the ability to identify Fv binding domains of interest, and to obtain the DNA encoding these variable regions. Hybridoma technology enables the production of cell lines that secrete antibodies to essentially any desired substance that elicits an immune response. For example, U.S. Pat. No. 4,753,894 describes some monoclonal antibodies of interest which recognize c-erbB-2 related antigens on breast cancer cells, and explains how such antibodies were obtained. One monoclonal antibody that is particularly useful in targeting the c-erbB-2 antigen is 741F8 (Bjorn et al., 1985, Cancer Res. 45: 1214-1221; U.S. Pat. No. 4,753,894). This antibody specifically recognizes the c-erbB-2 antigen expressed on the surface of various tumor cell lines, and exhibits very little binding to normal tissues. Other monoclonal antibodies that bind c-erbB-2 or related antigens include 520C9 and 454C11 (Frankel et al., 1985, J. Biol. Resp. Modif. 4: 273-286; Ring et al., 1989, Cancer Res. 49: -3070-3080, Ring et al., 1991, Molec. Immunol. 28: 915-917; U.S. Pat. Nos. 4,753,894 and 5,169,774). sFv′ sequences with the desired specificity can also be derived from phage antibody cloning of combinatorial V gene libraries. Such sequences could be based on cDNA derived from mice preimmunized with tumor cell membranes bearing c-erbB-2 or related antigenic fragments, (See, for example, Clackson et al, (1991) Nature 352: 624-628).
  • The process of designing DNA encoding the single polypeptide chain of interest can be accomplished as follows. Either synthetic DNA duplexes can be ligated together to form a synthetic gene or relevant DNA fragments can be cloned from libraries. In the latter procedure, mRNA encoding the light and heavy chains of the desired immunoglobulin may be isolated from hybridomas producing the immunoglobulin and reverse transcribed into cDNA. The VH and VL genes subsequently can be isolated by standard procedures, for instance, by colony hybridization of cDNA libraries (see for example, Sambrook et al., eds., 1989, Molecular Cloning, Cold Spring Harbor Laboratories Press, NY) or by polymerase chain reaction (PCR) (see for example, Innis et al., eds., 1990, PCR Protocols, A guide to methods and applications, Academic Press). Both procedures are well known in the art.
  • Still another approach involves the design and construction of synthetic variable domain genes encoding a predetermined, specific Fv binding site. For example, with the help of a computer program, such as Compugene, one may design and directly synthesize native or near native FR sequences from a first, antibody molecule, and CDR sequences from a second antibody molecule. The resulting VH and VL gene sequences can then be genetically linked together by means of a linker connecting the C-terminus of one chain with the N-terminus of the other.
  • Practice of the invention enables the design and synthesis of various single-chain binding proteins, all of which are characterized by a region having affinity for a preselected epitope on an antigen. Other regions of the biosynthetic protein are designed with the particular planned utility of the protein in mind. Thus, if the reagent is designed for intravascular use in mammals, the FRs may include amino acid sequences which are similar or identical to at least a portion of the FR amino acid sequences of antibodies native to that species. The amino acid sequences constituting the CDRs may be analogous to the sequences from a second, different preexisting antibody having specificity for the antigen of interest (e.g. a murine or other human IgG). Alternatively, the CDRs and FRs may be copied in their entirety from a single pre-existing monoclonal antibody cell line or a desirable sFv species may be cloned from a repertoire library derived from preimmunized or naive animals.
  • It is noted however, that the linear arrangement of the VL and VH domains in the DNA sequence of FIG. 1 is not critical. That is, although the sequence represented in FIG. 1A encodes a heavy chain variable region followed by the light chain variable region, as will be appreciated by those skilled in the art, the sFv may be constructed so that the light and heavy chain domains are in reverse order.
  • As mentioned above, the VH and VL domains of the sFv are linked in the gene construct by means of a linker 12 (FIG. 1A). The linker should be at least long enough (e.g., about 10 to 15 amino acids or at least 40 Angstroms in length) to permit domains 10 and 14 to assume their proper conformations and interdomain relationships. The linkers preferably comprise hydrophilic amino acids that assume an unstructured configuration under physiological conditions, and are free of residues having large side groups that could interfere with proper folding of the VH, VL, or pendant chains. Examples of currently preferred linkers include either part or all of the amino acid sequences ((Gly)4Ser)3 and ((Ser)4Gly)3, set forth in the Sequence Listing as SEQ. ID. NOS.: 7 and 8, respectively. The linker may also include an amino acid sequence homologous to a sequence identified as “self” by the species into which it will be introduced, particularly if a therapeutic application is intended.
  • Considerations for Suitable C-terminal Tail Sequences.
  • As mentioned above, the sFv′ polypeptide chains further comprise a C-terminal tail containing at least one amino acid that can be derivatized or post-translationally modified to enable crosslinking of two such sFv′ monomers. In preferred aspects of the invention, the tail sequences include one or more of the sequences Ser-Cys, (Gly)4-Cys and (His)6-(Gly)4-Cys, set forth in the Sequence Listing as SEQ. ID. NOS.: 9, 10, and 11, respectively. The C-terminal tails preferably do not form α-helical structures which self-associate under physiological conditions, such as the α-helical leucine zipper motifs found in DNA binding proteins (O'Shea et al., 1989, Science 243: 538-542, O'Shea et al., 1991, Science 254:
  • 539-544) or the four-helix bundle motifs found in recombinant ion channels (Hill et al., 1990, Science 294: 543-546).
  • Suitable derivatizable amino acid side chains may be selected from the group consisting of cysteine, lysine, arginine, histidine, glutamate, aspartate and derivatives or modified forms thereof. In a preferred aspect of the invention, cysteine amino acids are incorporated into the C-terminal tail sequences as the crosslinking means.
  • Also envisioned to be useful are posttranslationally modified amino acids that can be crosslinked in vitro. More specifically, the glycosyl moieties present on glycosylated amino acids, following secretion out of the cell, can be covalently attached in vitro using bifunctional linkers on standard sugar chemistry (see for example, E. A. Davidson (1967) Carbohydrate Chemistry, Holt, Kinehart and Winston, N.Y.; W. J. Lennarz (1980) The Biochemsitry of Glycoproteins and Proteoglycans, Plenum Press, N.Y.). Particularly useful glycosylation sites include the sequences Asn-Xaa-Thr and Asn-Xaa-Ser, wherein Xaa is any amino acid. Where crosslinking of glycosyl moieties is contemplated, the glycosylation sequences need not include a cysteine.
  • The tail also may comprise an amino acid sequence defining an ion chelation motif which can be used as part of a purification protocol for isolating of the sFv′ monomers by metal ion affinity chromatography (e.g., by means of a (His)6 tail on an IMAC chromatography column), as well as for chelating ions of detectable moieties such as Technetium−99m or 111Indium for in vivo imaging applications.
  • sFv′ Coupler Considerations.
  • In the present invention, two monomeric sFv′ proteins are crosslinked together through their C-terminal tails to form an (sFv′)2 dimer. The term “sFv coupler”, as used herein, refers to chemical bridges that join the crosslinking residues in each of the sFv′ molecules.
  • In one preferred aspect of the invention, where the crosslinking residue is a cysteine residue, the chemical bridge can be a disulfide bond. Alternatively, sulfhydryl-specific crosslinking reagents can be used to join two sFv′ molecules together. An example of such a cysteine-specific chemical bridge includes the bifunctional crosslinking reagent bismaleimidohexane (BMH), a water insoluble linker that can be obtained from Pierce, Rockford, Ill. Other bifunctional crosslinking agents include heterobifunctional crosslinkers which can be used to join two sFv′ species together where the crosslinking residues in each of the sFv′ C-terminal tail sequences are different, such as, a C-terminal cysteine on one sFv′ and a C-terminal lysine on the other. Useful heterobifunctional crosslinking agents include 4-succinimidyloxycarbonyl-methyl-(2-pyridyldithio)-toluene (SMPT) or N-succinimidyl-3-(2-pyridyldithio)propionate (SPDP), both of which can be obtained from Pierce, Rockland, Ill. sFv couplers of variable length also can be prepared to limit steric interaction of two coupled sFv′ proteins. An example of such an sFv coupler includes a peptide bridge, such as the water soluble bismaleimidocaproyl amino acid (MCA) linker. Although in a preferred aspect of the invention, an MCA-, GlySer3Gly2Ser3Lys-MCA linker is used, in theory, any amino acid sequence can be introduced into this type of chemical bridge-spacer group.
  • Suitable MCA-peptide chemical bridges can be synthesized on polystyrene resins functionalized with hydroxymethylphenoxyacetic acid (HMP) to allow formation of free acids at the C-terminus following deblocking. During the synthesis of the preferred peptide sequence Gly-Ser3-Gly2-Ser3-Lys the C-terminal lysine is esterified to the resin and other amino acids are added as N-α-Fmoc protected derivatives. DIC/hydroxybenzotriazol activated amino acids are coupled for 90 minutes after which the N-α-Fmoc protected groups are deprotected with 20% piperidine in dimethylformamide (DMF). Upon completion of the synthesis, the peptide is cleaved from the resin and deprotected with 95% trifluoroacetic acid (TFA) in water. The crude peptide then is dissolved in 0.1M phosphate buffer pH 7 and reacted overnight at room temperature with maleimidocaproic acid N-hydroxy-succinimide ester. The resulting homobifunctional peptide crosslinker can be purified by reverse phase HPLC, for example, on a Vydac 1×25 cm column using acetonitrile/water/TFA buffers.
  • With this procedure, it is possible to generate linkers having specific lengths and flexibilities. Since polypeptides having particular secondary structures and flexibilities are well documented in the art, it is possible to judiciously design the peptide couplers with optimal length and flexibility to optimize binding to two preselected antigens on a cell surface. As will be appreciated by those skilled in the art, the separation distance between, and interaction of, the sFv′ monomers in a dimeric construct of the invention also can be modulated by the judicious choice of amino acids in the tail sequences themselves.
  • Dimer Considerations.
  • Using the approaches described above, (sFv′)2 dimers readily can be prepared wherein the resulting dimers either can be homodimeric, where the CDR sequences define the same epitope binding site, or heterodimeric, where the CDR sequences of each sFv′ monomer define different epitope binding sites.
  • The dimeric constructs of this invention preferably target a pharmacologically active drug (or other ancillary protein) to a site of interest utilizing the bivalent capability of the dimer. Examples of pharmacolcogically active drugs include molecules that inhibit cell proliferation and cytotoxic agents that kill cells. Other, useful molecules include toxins, for instance, the toxic portion of the Pseudomonas exotoxin, phytolaccin, ricin, ricin A chain, or diptheria toxin, or other related proteins known as ricin A chain-like ribosomal inhibiting proteins, i.e., proteins capable of inhibiting protein synthesis at the level of the ribosome, such as pokeweed antiviral protein, gelonin, and barley ribosomal protein inhibitor.
  • In such cases, one sFv′ can be immunologically reactive with a binding site on an antigen at the site of interest, and the second sFv′ in the dimer can be immunologically reactive with a binding site on the drug to be targeted. For example, the (sFv′)2 dimers may have specificity for both c-erbB-2 and a pharmacologically active drug or cytotoxic agent. The resulting dimer can thus target the agent or drug to tissues expressing the c-erbB-2 antigen in vivo. Alternatively, the construct may bind one or more antigens at the the site of interest and the drug to be targeted is otherwise associated with the dimer, for example, by crosslinking to the chemical bridge itself.
  • Other bispecific (sFv′)2 constructs having particular utility in targeting malignant cells, include constructs wherein one has specificity for a c-erbB-2 or related tumor antigen, and the second determinant has specificity for a different cell surface protein, such as the CD3 antigen found on cytotoxic T-lymphocytes. The heterodimeric (sFv′)2 construct then could mediate antibody dependent cellular cytotoxicity (ADCC) or cytotoxic T-lymphocyte-induced lysis of the tumor cells expressing the c-erbB-2 antigen.
  • Still another bispecific dimeric construct having cytotoxic properties is a bispecific construct with one sFv′ capable of targeting a tumor cell and the second being a catalytic sFv′ that binds an inactive drug, and subsequently converts it into an active compound (see for example, U.S. Pat. No. 5,219,732). Such a construct would be capable of inducing the formation of a toxic substance in situ. For example, a catalytic sFv′ molecule having β-lactamase-like activity can be designed to bind and catalyze the conversion of an inactive lactam derivative of doxorubicin into the active, cytotoxic form. Here the bispecific dimer, having binding affinities for both the preselected antigen and the cytotoxic-lactam derivative, is administered to an individual and allowed to accumulate at the desired location. The inactive, nontoxic cytotoxin-lactam derivative then is administered to the individual. When the derivative is complexed with the bispecific (sFv′)2 heterodimer in situ the active form of the drug is released, enhancing both the cytotoxicity and specificity of the drug.
  • Hybrid sFv′ Considerations.
  • In a preferred aspect of the invention a humanized single-chain Fv is envisioned whereby the recombinant sFv′ contains CDRS of the murine 741F8 antibody interposed between human FR sequences to generate a humanized c-erbB-2 binding protein. The humanized Fv would be capable of binding c-erbB-2 while eliciting little or no immune response when administered to a patient. A nucleic acid sequence encoding a humanized sFv may be designed and constructed as follows.
  • FR regions identified by homology searches of the GenBank database can be introduced into an sFv of interest by site-directed mutagenesis to reproduce the corresponding human sequence. Alternatively, homologous human VH and VL sequences can be derived from a collection of PCR-cloned human V regions, after which the human FR sequences can be ligated with murine CDR regions to create humanized VL and VH genes. A humanized sFv hybrid thus can be created, for instance, where the human FR regions of the human myeloma antibody are introduced between the murine CDR sequences of the murine monoclonal antibody 741F8. The resulting sFv, containing the sequences FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4, contains a murine binding site in a human framework.
  • By directly sequencing the DNA or RNA in v hybridoma secreting an antibody to a preselected antigen, or by obtaining the sequence from the literature, one skilled in the art can essentially produce any desired CDR and FR sequence. Expressed sequences subsequently may be tested for binding and empirically refined by exchanging selected amino acids in relatively conserved regions, based on observations of trends of amino acid sequences in data bases and/or by using computer-assisted modeling techniques. Significant flexibility in VH and VL design is possible because alterations in amino acid sequences may be made at the DNA level.
  • Of course, the processes for manipulating, amplifying, and recombining DNAs that encode amino acid sequences of interest are generally well known in the art (see, for example, Sambrook et al., 1989, Molecular Cloning A Laboratory Manual, 2nd ed. Cold Spring Harbor Press), and therefore, are not described in detail herein. Similarly, methods for identifying the isolated V genes encoding antibody Fv regions of interest are well understood and are described in the patent and other literature.
  • Expression of Recombinant sFv Proteins.
  • The resulting sFv DNA constructs then are integrated into expression vectors and transfected into appropriate host cells for protein expression. After being translated, the protein may be purified from the cells themselves or recovered from the culture medium.
  • The expression vectors also may include various sequences to promote correct expression of the recombinant protein. Typical sequences include transcription promoters and termination sequences, enhancer sequences, preferred ribosome binding site sequences, preferred mRNA leader sequences, preferred protein processing sequences, preferred signal sequences for protein secretion, and the like. The DNA sequence encoding the gene of interest also may be manipulated to remove potentially inhibiting sequences or to minimize unwanted secondary structure formation. The resulting synthetic genes can be expressed in appropriate prokaryotic hosts such as various strains of E. coli, or in eucaryotic hosts such as Chinese hamster ovary cells (CHO), mouse myeloma, hybridoma, transfectoma, and human myeloma cells. The currently preferred expression system for the present invention is E. coli, as disclosed herein.
  • When the gene is to be expressed in E. coli, it is cloned into an expression vector downstream of a strong promoter sequence, such as Trp or Tac, and optionally also may include a gene coding for a leader polypeptide, such as the fragment B (FB) of staphylococcal protein A. The resulting fusion protein, when expressed, accumulates in retractile bodies (also known as inclusion bodies) in the cytoplasm, and may be harvested after disruption of the cells by French press or sonication. The proteins then are solubilized, and refolded in vitro, as described herein. Where the construct is engineered as a fusion protein, the protein is solubilized and the leader sequence preferably cleaved before renaturation. The cleavage site for the leader sequence preferably is immediately adjacent to the sFv polypeptide chain and includes one amino acid or a sequence of amino acids exclusive of any one amino acid or amino acid sequence found in the amino acid structure of the single polypeptide chain.
  • The cleavage site preferably is designed for specific cleavage by a selected agent. Endopeptidases are preferred, although non-enzymatic (e.g., chemical) cleavage agents may be used. Many useful cleavage agents, for instance, cyanogen bromide (CNBr), dilute acid, trypsin, Staphylococcus aureus V-8 protease, post-proline cleaving enzyme, blood coagulation Factor Xa, enterokinase, and renin, recognize and preferentially or exclusively cleave at particular cleavage sites. One currently preferred peptide sequence cleavage agent is V-8 protease. The currently preferred cleavage site is at a Glu residue. Other useful enzymes recognize multiple residues as a cleavage site, e.g., factor Xa (Ile-Glu-Gly-Arg) or enterokinase (Asp-Asp-Asp-Asp-Lys). Dilute acid preferentially cleaves the peptide bond between Asp-Pro residues, and CNBr in acid cleaves after Met, unless it is followed by Tyr.
  • Alternatively, the engineered gene may be incorporated into a vector without a sequence encoding a leader polypeptide, and the engineered gene expressed to produce a polypeptide chain that is secreted into the E. coli periplasmic space. The secreted protein then can be isolated and, optionally, purified further using standard methodologies. (See, for example, Pack et al. (1992) Biochem 31:1579-1584.)
  • If the engineered gene is to be expressed in eucaryotic hybridoma cells, the conventional expression host for immunoglobulins, the gene preferably is inserted into an expression vector containing, for example, the immunoglobulin promoter, a secretion signal, and immunoglobulin enhancers. This plasmid also may contain sequences encoding other polypeptide chains, including part or all of a toxin, enzyme, cytokine, or hormone. The gene then is transfected into myeloma cells via established electroporation or protoplast fusion methods. The transfected cells then may express VH-linker-VL-tail or VL-linker-VH-tail single-chain Fv′ polypeptide chains.
  • The sFv polypeptide chains can be expressed as either inactive or active polypeptide chains.
  • Spontaneously refolded sFv polypeptide chains can be obtained from either prokaryotic or eukaryotic expression systems when the polypeptide chains are secreted for instance, either into the E. coli periplasmic space or the mammalian cell culture medium. These spontaneously refolded polypeptide chains readily can be purified by affinity chromatography. Where the sFv polypeptide chains are obtained in an unfolded, inactive sFv form, for instance, when overexpression of the sFv polypeptide chain in E. coli results in the formation of inclusion bodies, the proteins can be refolded in vitro. Briefly, inclusion bodies are harvested by centrifugation, the sFv, solubilized with denaturants such as guanidine hydrochloride (GuHCl) or urea, and then refolded by dilution of the denaturant under appropriate redox (reduction/oxidation) conditions (see below). The refolded sFv polypeptide chains then can be purified by affinity chromatography. Details for the isolation of inclusion bodies, solubilization and renaturation of the sFv polypeptide chains are well known in the art (see for example, U.S. Pat. No. 5,091,513 and Huston et al., 1988, supra).
  • Dimerization and Purification of the sFv Polypeptides
  • The sFv′ monomers of the present invention can be dimerized in vivo or in vitro. In the in vivo approach, two sFv′ genes can be cotransfected into the host cell wherein the coexpressed sFv′ polypeptide chains spontaneously dimerize. Alternatively, the refolded, secreted sFv′ polypeptide chain monomers can be isolated from two expression hosts and subsequently dimerized in vitro.
  • In a preferred aspect of the invention, the sFv′ polypeptide chains comprising a single cysteine C-terminal tail residue are expressed in E. coli and form inclusion bodies. The resulting sFv′ polypeptide chains are solubilized with denaturants and renatured in vitro, either in the presence or absence of exogenously added glutathione. Surprisingly, the additional C-terminal cysteine residues apparently do not interfere with the refolding process. In some cases however, sFv and sFv′ constructs may refold poorly in vitro. These constructs can be “preoxidized prior” to refolding as taught in Huston et al., (1991) Meth. Enzymol. 203:46-88, or, alternatively, the polypeptide chains can be secreted across a membrane bilayer. The latter process spontaneously separates the extra C-terminal cysteine residue from the cysteine residues normally found in the Fv domain, minimizing inappropriate disulfide bond formation. Secretion is the preferred method if the sFv′ constructs refold poorly in vitro.
  • Following renaturation of the sFv′ monomers, (sFv′)2 dimers readily can be prepared in vitro by air oxidation if cysteine amino acids are present in the C-terminal tail sequences. Alternatively, sulfhydryl specific crosslinking reagents, for instance, the BMH crosslinker or the MCA-peptide-MCA bridge may be used to covalently couple two sFv′ chains. The resultant homo or heterodimers, then can be purified by standard size exclusion chromatography. However, when (sFv)2 heterodimers are required, then a preferred purification protocol uses a sequential two step affinity chromatography procedure. Briefly, the heterodimer is exposed to a first chromatographic system having an epitope that interacts specifically with one sFv of the heterodimer. The eluant containing the heterodimer is then exposed to a second system having an epitope that interacts specifically with the other sFv′. For specific details of the dimerization and purification procedures, see Example 2.
  • Considerations for In Vivo Administration.
  • The dimeric constructs may be administered either by intravenous or intramuscular injection. Effective dosages for the single-chain Fv constructs in antitumor therapies or in effective tumor imaging can be determined by routine experimentation, keeping in mind the objective of the treatment.
  • The pharmaceutical forms suitable for injection include sterile aqueous solutions or dispersions. In all cases, the form must be sterile and must be fluid so as to be easily administered by syringe. It must be stable under the conditions of manufacture and storage, and must be preserved against the contaminating action of,microorganisms. This may, for example, be achieved by filtration through a sterile 0.22 micron filter and/or lyophilization followed by sterilization with a gamma ray source.
  • Sterile injectable solutions are prepared by incorporating the desirable amount of the constructs, disclosed herein, into an appropriate solvent, such as sodium phosphate buffered saline (PBS), followed by filter sterilization. As used herein, “a physiologically acceptable carriers” includes any and all solvents, dispersion media, antibacterial and antifungal agents that are non-toxic to humans, and the like. The use of such media and agents as pharmaceutically active substances are well known in the art. The media or agent must be compatible with maintenance of proper conformation of the single-chain Fv polypeptide chains, and its use in the therapeutic compositions. Supplementary active ingredients can also be incorporated into the compositions.
  • A preferred remotely detectable moiety for in vivo imaging includes the radioactive atom Technetium−99m (99mTc), a gamma emitter with a half-life of about 6 hours. Non-radioactive moieties also useful in imaging include nitroxide spin labels as well as lanthanide and transition metal ions all of which induce proton relaxation in situ. In addition to immunoimaging, the complexed radioactive moieties may be used in standard radioimmunotherapy protocols to destroy the targeted cell. Preferred nucleotides for high dose radioimmunotherapy include the radioactive atoms 90Yttrium (90Yt), 131Iodine (131I) and 111Indium (111In).
  • Either the single polypeptide chain sFv′ itself, or the spacer groups for linking the sFv′ constructs can be labeled with radioisotopes such as 131I, 111In and 99mTc. 99mTc and 111In are preferred because they can be detected with gamma cameras and have favorable half-lives for in vivo imaging applications. The single polypeptide chains can be labeled, for example, with radioactive atoms such as 90Ty, 99MTc or 111I via a conjugated metal chelator (see, e.g., Khaw et al., 1980, Science 209: 295; U.S. Pat. No. 4,472,509; U.S. Patent No. 4,479,930), or by other standard means of linking isotopes to proteins, known to those with skill in the art (see for example, Thankur et al., 1991, J. Immuniol. Methods 237: 217-224).
  • The invention is illustrated by the following Examples, which are not intended to be limiting in any way.
  • EXAMPLES Example 1 Synthesis and Expression of the sFv Constructs (741F8, 26-10 and 520C9)
  • The construction of several sFv genes using different but standard recombinant DNA technology, well known to those having ordinary skill in the art, is described below. These procedures include the amplification of the VH and VL gene sequences by PCR, the ligation of appropriate synthetic DNA duplexes and the cloning of VH or VL genes by colony hybridization.
  • A. 741F8 sFv′.
  • The VH and VL genes of the 741F8 anti-c-erbB-2 monoclonal antibody were isolated from the cDNA of the parental 741F8 hybridoma line by PCR using primers homologous to the N-terminal coding regions of VH, VL, C H1, and CL. The PCR-amplified VH and VL genes were isolated by polyacrylamide gel electrophoresis and cloned into a pUC cloning vector. The first FR region of the 741F8 VH gene however contained spurious mutations due to the PCR procedure. Errors were rectified by the replacement of the first 70 nucleotides of 741F8 VH with a similar sequence from 520C9 VH, another c-erbB-2 specific monoclonal antibody.
  • Restriction sites then were introduced into the ends of the heavy and light chain variable gene segments by site-directed mutagenesis (Kunkel et al., 1985, Proc. Natl. Acad. Sci. USA 82: 488-492). A Nco I site encoding methionine was positioned at the N-terminus of VH for expression in E. coli. A Sac I site was created at the 3′ end of VH gene. A Xho I site, together with an adjacent Eco RV site, were created at the N-terminus of VL. A stop codon and a Pst I site were placed at the. C-terminal end of VL.
  • The single-chain Fv gene was constructed by connecting the VH and VL genes together with a DNA sequence encoding the 14 residue polypeptide linker, (Ser4Gly)2Ser4′ as set forth as amino acids 122 through 135 in the Sequence Listing as SEQ. ID. NOS.: 1 and 2.
  • A synthetic DNA duplex encoding the C-terminal amino acid sequence, (Gly)4-Cys was inserted into a Hpa I site located near the stop codon at the 3′ end of the 741F8 sFv gene. The resulting 741F8 anti-c-erbB-2 sFv′ gene was excised from the pUC cloning vector, with the restriction enzymes Nco I and Bam HI (a Bam HI site is located 3′ to the C-terminal Pst I site), and inserted into the same sites of a commercial T7 expression vector pET-3d (In-vitrogen, Inc.). The resulting gene, set forth in the Sequence Listing as SEQ. ID. NOS.: 1 and 2, was transformed into E. coli BL21-DE (In-vitrogen, Inc.). Protein expression was induced by the addition of IPTG to the culture medium.
  • B. 26-10 sFv′
  • Construction of the anti-digoxin 26-10 sFv has been described previously (Huston et al., 1988, Proc.
  • Natl. Acad. Sci. USA 85; 5879-5883, and U.S. Pat. No. 5,091,513, both of which are hereby incorporated by reference). Briefly, the synthetic gene was constructed by ligating multiple synthetic DNA duplexes together. The C-terminal DNA duplex coding for the amino acid sequence (Gly)4-Cys subsequently was ligated into a Hpa I restriction site close to the 3′ end of the 26-10 sFv gene. The resulting sFv′ gene, set forth in the Sequence Listing as SEQ. ID. NOS.: 3 and 4, was then inserted into the E. coli expression vector pET-3d. This plasmid was subsequently transformed into E. coli BL21-DE (In-vitrogen, Inc.) and protein expression induced by the addition of IPTG to the culture medium.
  • C. 520C9 sFv′.
  • The 520C9 sFv was generated by linking together the VH and VL genes, cloned from a 520C9 hybridoma cDNA library, with a serine rich linker. Briefly, the VH and VL genes were cloned from the 520C9 hybridoma cDNA library using probes directed toward the antibody constant (C) and joining (J) regions. Appropriate restriction sites were introduced at the ends of each gene by site-directed mutagenesis (Kunkel et al., 1985, Proc. Natl. Acad. Sci. USA 82: 488-492). The VH and VL genes were then ligated together with a serine rich linker. The resulting 520C9 sFv gene, set forth in the Sequence Listing as SEQ. ID. NOS.: 5 and 6, was transformed into the E. coli expression vector and expressed as described above and in co-pending U.S. Ser. No. 831,967, incorporated therein by reference.
  • Example 2 Renaturation, Dimerization and Purification of sFv Proteins
  • A. Renaturation and Purification of sFv Monomers.
  • Protocols for renaturing sFv monomers derived from E. coli inclusion bodies are described below. In separate experiments the 7418, 26-10 and 520C9 sFv polypeptides were expressed in E. coli. The unfolded sFv proteins were solubilized from inclusion bodies and refolded under appropriate redox conditions. The refolded sFv polypeptide chains were purified by affinity chromatography or by a combination of ion-exchange and size exclusion chromatography when affinity chromatography was not feasible or expedient.
  • Renaturation of 741F8 sFv′.
  • Inclusion bodies containing the 741F8 sFv′ proteins were washed in a buffer containing 25 mM Tris, 10 mM EDTA, 1.5 M GuHCl, pH 8.0 and solubilized in 25 mM Tris, 10 mM EDTA, 7 M GuHCl, pH 9.0 to an OD280 nm of about 25-50. The sample was reduced overnight at room temperature by the addition of dithiothreitol (DTT) to a final concentration of 10 mM. The thiol groups were converted into mixed disulfides with qlutathione by the addition of solid oxidized glutathione to a final concentration of 100 mM. The solution was adjusted to pH 9.0 and incubated for 4 hr at room temperature. The 741F8 sFv′ polypeptide chains then were refolded in vitro to generate stable monomers with their C-terminal cysteines remaining blocked with glutathione. The 741F8 sFv′ mixed disulfide preparation was diluted to an OD280 of about 0.15 by the addition of 10 mM Tris, 4 mM EDTA, 6 M urea, pH 8.5 at 4° C. After two hours an equal volume of 10 mM Tris, 4 mM EDTA, 1 mM reduced glutathione, pH 8.5, precooled to 4° C., was added with rapid mixing to reduce the urea concentration to 3 M. After dilution, the samples were allowed to renature for 72 hr at 4° C.
  • Renaturation of 26-10 sFv′.
  • Inclusion bodies containing the 26-10 sFv′ proteins were washed with 25 mM Tris, 10 mM EDTA and solubilized in 6 M GuHCl, 25 mM Tris, 10 mM EDTA, pH 8.7 to an OD280 nm of about 10 to 20. The dissolved proteins were reduced by overnight incubation at room temperature after the addition of DTT to 10 mM. The reduced protein could also be blocked with oxidized glutathione as noted above for the 741F8 sFv′ polypeptide. The reduced, denatured 26-10 sFv′ polypeptides were refolded in a manner similar to that for the 741F8 sFv′ by diluting the preparation into a buffer containing 3 M urea, 0.1 mM oxidized and 0.01 mM reduced glutathione to give a final protein concentration of about 0.15 mg/ml. After overnight incubation at 4° C, the mixture was dialyzed against PBS containing 0.05 M KH2PO4, 0.15 M NaCl, pH 7 for two days at 4° C.
  • Renaturation of 520C9 sFv.
  • The inclusion bodies containing the 520C9 sFv were washed with 25 mM Tris, 10 mM EDTA, pH 8.0, 1 M GuHCl and solubilized in 25 mM Tris, 10 mM EDTA, 6 M GuHCl, 10 mM dithiothreitol (DTT), pH 9.0. The material was ethanol precipitated and resuspended in 25 mM Tris, 10 mM EDTA, 6M urea, 10 mM DTT, pH 8.0 and fractionated by ion exchange chromatography to remove contaminating nucleic acids and E. coli proteins before renaturation of the sFv′. The material that did not bind to a DEAE Sepharose Fast Flow (FF) column was precipitated by lowering the pH to 5.5 with 1 M acetic acid. The pellet was resolubilized in 25 mM Tris, 10 mM EDTA, 6 M GuHCl, 10 mM DTT, pH 9.0 and oxidized by overnight incubation at room temperature following dilution into a buffer containing 25 mM Tris, 10 mM EDTA 6 M GuHCl, 1 mM oxidized glutathione, 0.1 mM reduced glutathione, pH 9.0. After overnight oxidation the sample was dialyzed against 10 mM NaH2PO4, 1 mM EDTA, 150 mM NaCl, 500 mM urea, pH 8.0 and the sample clarified by filtration through a membrane with a 100 kD mol. wt. cut-off prior to purification on a c-erbB-2 affinity column.
  • Purification of the Refolded sFv Polypeptides.
  • The refolded 26-10 sFv′ polypeptide chains were purified by ouabain-Sepharose affinity chromatography, as described for the 26-10 sFv constructs (Huston, et. al., 1988, Proc. Natl Acad. Sci. USA 85; 5879-5883 and Tai, et al., 1990, Biochem. 29, 8024-3080, both of which are hereby incorporated by reference). The refolded 520C9 sFv polypeptide chain was similarly purified using a c-erbB-2-agarose affinity column. In this case, the refolded samples were loaded onto a c-erbB-2 affinity column, the column washed with PBS, and the 520C9 sFv polypeptides eluted with PBS pH 641 containing 3 M LiCl. The buffer was then exchanged by dialysis. The c-erbB-2 affinity column preferably was prepared by linking the extracellular domain of c-erbB-2 onto agarose beads.
  • Briefly, the c-erbB-2 sequence coding for its extracellular domain (ECD) was derived from the baculovirus expression vector described previously (Ring et al., 1992, Mol. Immunol. 28; 915-917). A DNA duplex encoding the His6 peptide was ligated to the 3′ end of the ECD gene, and the construct expressed in CHO cells. The ECD polypeptide was purified from the CHO cell culture medium on an IMAC metal affinity column (Pharmacia, Piscataway, N.J.), as described in Skerra, et al., 1991, Bio/Technology 9: 273-278, and the eluted ECD proteins attached onto agarose beads to generate the c-erbB-2-agarose affinity resin.
  • The renatured 741F8 sFv′ polypeptides were purified by a combination of ion exchange and size exclusion chromatography. Briefly, the renatured 741F8 sFv′ preparation was passed through a DEAE-cellulose column and the 741F8 sFv′ in the unbound fraction adjusted to pH.5.0 before loading on an S-Sepharose FF column. The 741F8 sFv′ polypeptide chains were eluted with PBS containing 2 mM EDTA and 3 M urea, and dialyzed against 10 mM Tris, 2 mM EDTA, 20 mM NaCl, pH 7.5 at 20° C. The precipitate was harvested by centrifugation, dissolved in a suitable buffer, and passed through a Q-Sepharose FF column. The unbound material was adjusted to pH 5.5 and reloaded onto a S-Sepharose FF column. The 741F8 sFv′ polypeptides were eluted with a PBS, 2 mM EDTA, 100 mM NaCl, 3 M urea buffer and dialyzed against PBS, 2 mM EDTA. The precipitate was harvested again by centrifugation, dissolved in a suitable buffer, sucrose added to 5% (w/v), and the 741F8 sFv′ concentrated to 5 mg/ml in a YM10 membrane concentrator (Amicon). The 741F8 sFv′ polypeptide chains were fractionated by gel filtration chromatography using a S-200 HR column (Pharmacia LKB Biotechnology) and a PBS, 2 mM EDTA buffer.
  • B. Dimerization of the sFv′ Constructs
  • Dimerization of sFv′ monomers can be induced using standard crosslinking conditions. Where disulfide bond formation is desired, the monovalent sFv′ polypeptide chains initially are deblocked by mild reduction and (sFv′)2 dimers formed by crosslinking the sFv′ polypeptides either by disulfide linkages or by thioether linkages with the BMH or MCA-peptide-MCA crosslinking reagents.
  • In order to generate disulfide linked constructs the purified 741F8 and 26-10 sFv′ preparations were dialyzed against 50 mM Tris, 150 mM NaCl, pH 8.5. The C-terminal glutathionyl blocking groups were removed by the addition DTT to a concentration of 2 mM followed by overnight incubation at room temperature. Excess reducing agent was removed by extensive dialysis against 50 mM Tris, 150 mM NaCl, pH 8.5, during which the majority of the sFv′ polypeptides oxidized into the homodimeric form.
  • In order to generate BMH and MCA-peptide-MCA crosslinked constructs, sFv′ polypeptide chains in PBS first were reduced for two hours at room temperature by the addition of DTT to a final concentration of 1 mM. The samples were desalted by gel filtration chromatography using a PBS, 1 mM EDTA buffer. A 4-5 fold molar excess of either the BMH or MCA-peptide-MCA linkers, both dissolved in dimethylsulfoxide, were added to the reduced protein and incubated for at least 12 hours at room temperature. The resulting dimers were then purified by HPLC gel filtration chromatography.
  • A modification of the procedure of Brennan, et al. (1985, Science 229: 81-83) may be used to generate disulfide linked sFv′ heterodimers. For example, in order to link the 741F8 and 26-10 sFv′ polypeptides a thionitrobenzoate (TNB) derivative of the 26-10 sFv′ (26-10 sFv′-TNB) was mixed with mildly reduced 741F8 sFv′. The 26-10 sFv′-TNB was prepared by reducing the 26-10 sFv′ in PBS with 15 mM 2-mercaptoethylamine for 30 minutes at room temperature. The reducing agent was removed by gel filtration and the reduced 26-10 sFv′ reacted with 2.2 mM dithionitrobenzoate (DTNB) for 3 hours. The active 26-10 sFv′-TNB was adsorbed onto onto ouabain-Sepharose. The glutathionyl blocked 741F8 sFv′ monomer in 25 mM Tris, 150 mM NaCl, pH 8.2 was reduced for 2 hours at room temperature by the addition of DTT to a final concentration of 1 mM. The excess DTT was removed by gel filtration and the reduced 741F8 sFv′ reacted overnight at room temperature with the 26-10 sFv′-TNB complexed to ouabain-Sepharose. The progress of the reaction was monitored spectroscopically at 412 nm, the absorbance maximum of the TNB anion.
  • C. Purification of (sFv′)2 Dimers.
  • The (sFv′)2 homodimers may be separated from the sFv′ monomers by gel filtration chromatography. Following dimerization, the sFv′ preparations are dialyzed against PBS containing 1 mM EDTA, 3M urea, 0.03% azide, to disrupt any non-covalent homodimers and fractionated by HPLC on a TSK-G20000SW column using the same buffer. The procedure requires two passes for purification of the (sFv′)2 homodimers to homogeneity. The purified hoinodimers may be dialyzed either against PBS or any other suitable buffer prior to use.
  • The (sFv′)2 heterodimers can be separated by a two step affinity chromatography procedure taking advantage of the bivalent nature of the dimer. For instance, during the the purification of the 741F8/26-10 heterodimer the mixture initially was loaded onto an ouabain-Sepharose column, washed with a PBS, 1 M NaCl buffer, to remove any non-specifically adsorbed material, and rewashed with PBS to reduce the salt concentration. The reactive 26-10 sFv′ species bound to the resin were eluted with 20 mM ouabain in PBS and the eluate dialyzed against PBS to remove the cardiac glycoside. The 741F8/26-10 heterodimers were then repurified on a c-erbB-2-agarose affinity column taking advantage of the ECD binding site in the heterodimer. After the preparation was loaded onto the c-erbB-2 affinity column, it is washed with PBS and the (sFv′)2 heterodimer eluted with 25 mM Tris, 10 mM EDTA, 5M LiCl, pH 6.8. Prior to use, the buffer was exchanged with PBS by dialysis.
  • Example 3 Immunoreactivity of the Monomeric and Dimeric sFv Polypeptides
  • A. Radiolabeling of the sFv′ Constructs.
  • The sFv′ polypeptides may be labeled by the chloramine-T method as described (DeNardo, et al., 1986, Nucl. Med. Biol. 13: 303-310). Briefly, 1.0-2.0 mg of sFv′ was combined with 125I [14-17 mCi/μg] (Amersham, Arlington Heights, Ill.) at an iodine to protein ratio of 1:10 in a 12×75 mm plastic test tube. 10 μl (1 mg/ml] of chloramine-T (Sigma, St. Louis, Mo.) per 100 μg of protein was added and the mixture incubated for three minutes at room temperature. After the reaction was terminated, unincorporated 125I was separated from the labeled sFv′ by the spun-column method of Meares, et al., 1984, Anal. Biochem. 142: 68-78. Specific activities of 0.2-1.0 mCi/mg for the 125I-labeled products may be routinely obtained.
  • B. Competition ELISA
  • In order to prepare c-erbB-2, SK-Br-3 breast cancer cells (Ring et al., 1989, Cancer Res. 49: 3070-3080), were harvested and resuspended in 10 mM NaCl, 0.5% Nonidet-P40, pH 8. Insoluble debris was removed by centrifugation and the extract filtered through 0.45 Millex HA and 0.2 Millex GV filters. 40 μl of the extract was added to each well of a 96 well plate and incubated overnight at 37° C. The plates then were washed with PBS and non-specific binding sites blocked following the addition of PBS containing 1% skim milk by incubation for one hour at room temperature. The sFv and 520C9 Fab samples, diluted in PBS, were added to the wells and incubated for 30 mins at room temperature. A control containing only dilution buffer was also included.
  • In order to quantitate the reaction, 20 μl of a 520C9-horseradish peroxidase (HRP) probe (Zymed Labs., South San Francisco, Calif.), diluted to 14 μl/ml in PBS containing 1% skim milk, was added to each well and incubated for one hour at room temperature. The plate was then washed four times with PBS, the peroxidase substrate added and incubated for 30 minutes at room temperature. The reaction was quenched with H2SO4 and the OD150nm values measured.
  • FIG. 3 compares the binding ability of the parental 520C9 Fab fragment, together with the 520C9 sFv single-chain binding protein. The 520C9 sFv samples included the material obtained following renaturation of the polypeptide in vitro, a sample purified on a c-erbB-2 agarose affinity column, and the material that did not bind to the column. The fully purified 520C9 sFv polypeptide exhibits an affinity for c-erbB-2 indistinguishable from the parent 520C9 Fab fragment.
  • C. Biodistribution Studies.
  • In vivo immunotargeting tissue imaging studies were performed using standard procedures. Approximately 2.5×106 SK-OV-3 cells (a human ovarian cancer cell line that expresses c-erbB-2 on the cell surface) in log phase were implanted subcutaneously onto the hips of four to six week old CB17/ICI-scid mice. Three days after Lugol's solution was placed in the drinking water to block the accumulation of radioiodine in the thyroid, the mice were used in the biodistribution assays.
  • The radiolabeled sFv′ and Fab preparations were diluted in PBS for these studies. The biodistribution of the glutathionyl-blocked 741F8 sFv′ monomers, and the 741F8 and 26-10 (sFv′)2 constructs were compared after identical doses of the radiolabeled protein was administered by injection in each case. The total injected doses were determined by counting each animal on a Series 30 multichannel analyzer/probe system (probe model #2007, Canaberra, Meridian, Conn.). Groups of 3-6 mice were sacrificed twenty four hours after injection, the tumors and organs were removed, weighed and counted in a gamma counter to determine the amount of radiolabel incorporated into the tissues. From these measurements, the percentage of the initial injected dose incorporated per gram of tissue (% ID/gram) or the amount of label incorporated into the tumor relative to the amount of radiolabel incorporated into the other organs (T:O ratio) were determined. For specific details see DeNardo, et al., 1977, Cancer, 40: 2923-2929, or Adams, et al., 1992, Antibody, Immunoconjugates, and Radiopharmaceuticals 5: 81-95, both of which are hereby incorporated by reference. Specificity indices also can be determined by dividing the T:O ratios of the 125I-741F8 sFv′ by the corresponding T:O ratios of the 125I-26-10 sFv′. The results of the biodistribution studies 24 hours post administration are summarized in FIGS. 4 and 5. The mean standard error (SEM) for each value is less than 30%, except where indicated.
  • The disulfide linked 741F8 (sFv′)2 homodimers exhibit identical tumor specificities when compared to the monomeric 741F8 sFv′ polypeptide chains. The T:O ratios of the 741F8 sFv′ constructs consistently exceed those for the 26-10 sFv′ constructs, demonstrating the binding specificity of the 741F8 constructs for the tumors (FIG. 4). In addition, the 741F8 (sFv′ 2 dimers generally exhibit higher T:O ratios relative to that of the monomeric species, particularly for the disulfide bonded sFv′ 741F8 (sFv′-(Gly)4Cys)2 and the MCA linked 741F8 (sFv′ )2 homodimers. In addition, the 741F8 (sFv′ )2 homodimers localize in greater amounts in the tumors relative to the monomeric sFv′ species (FIG. 5).
  • In a separate comparative study with 125I-labeled 26-10 (sFv′ )2 and the following species of 125I-labeled 741F8: sFv′ monomers, Fab, disulfide linked (sFv′-Gly4Cys)2 homodimers, and MCA- and BMH-linked (sFv′)2 homodimers, the in vivo tumor localization properties of these molecules were compared (%ID/gram tumor tissue, see FIG. 6). As is evident from the figure, the tumor localization properties of all of the dimeric 741F8 (sFv′ )2 constructs are significantly greater than those observed with the 741F8 Fab, the 741F8 sFv′ monomer and the 26-10 (sFv′)2 dimer (FIG. 6). The results demonstrate that the increased apparent avidity and enhanced in vivo imaging of the (sFv′)2 dimer is due, at least in part, to its improved retention in tumor tissue.
  • Embodiments
  • The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The present embodiments are therefore to be considered in all respects as illustrative and not restrictive, the scope of the invention being indicated by the appended claims rather than by the foregoing description, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.

Claims (33)

1. A formulation for targeting an epitope on an antigen expressed in a mammal, the formulation comprising a pharmaceutically acceptable carrier in combination with,
a dimeric biosynthetic construct for binding at least one preselected antigen, the construct comprising:
(a) two polypeptide chains, each of which have:
an amino acid sequence defining at least two polypeptide domains, connected by a polypeptide linker spanning the distance between the C-terminus of one domain and the N-terminus of the other, the amino acid sequence of each said domain comprising complementarity determining regions (CDRs) interposed between framework regions (FRs), the CDRs and FRs of each polypeptide chain together defining a binding site immunologically reactive with a said preselected antigen, and
and a C-terminal tail having a non-self-associating structure under physiological conditions and comprising at least a crosslinking means, and
(b) a linkage coupling said crosslinking means on said two polypeptide chains,
said dimeric construct having a conformation permitting binding of a said preselected antigen by the binding site of each said polypeptide chain when administered to said mammal.
2. A formulation for targeting an epitope on an antigen expressed in a mammal, the formulation comprising a pharmaceutically acceptable carrier in combination with,
a dimeric biosynthetic construct for binding preferentially to a preselected antigen, the construct comprising:
(a) two polypeptide chains, each of which have:
an amino acid sequence defining at least two polypeptide domains, connected by a polypeptide linker spanning the distance between the C-terminus of one domain and the N-terminus of the other, the amino acid sequence of each said domain comprising complementarity determining regions (CDRs) interposed between framework regions.(FRs), the CDRs and FRs of each polypeptide chain together defining a binding site immunologically reactive with a said preselected antigen, and
a C-terminal tail having a non-self-associating structure under physiological conditions and comprising at least a crosslinking means, and
(b) a linkage coupling said crosslinking means to form a homodimeric construct,
said homodimeric construct having a conformation permitting binding to said preselected antigen in said mammal with an avidity greater than the avidity of either of said polypeptide chains individually.
3. A polypeptide chain for binding preferentially to a preselected antigen, the polypeptide chain comprising:
an amino acid sequence defining at least two polypeptide domains, connected by a polypeptide linker spanning the distance between the C-terminus of one domain and the N-terminus of the other, the amino acid sequence of each said domain comprising complementarity determining regions (CDRS) interposed between framework regions (FRs), the CDRs and FRs of each polypeptide chain together defining a binding site immunologically reactive with said preselected antigen, and
a C-terminal tail having a non-self-associating structure under physiological conditions and comprising at least a crosslinking means.
4. The polypeptide chain of claim 1, 2 or 3 wherein said C-terminal tail comprises the amino acid sequence Ser-Cys.
5. The polypeptide chain of claim 1, 2 or 3 wherein said C-terminal tail comprises the amino acid sequence (Gly)4-Cys.
6. The polypeptide chain of claim 1, 2 or 3 wherein said C-terminal tail comprises the amino acid sequence (His)6-(Gly)4-Cys.
7. The polypeptide chain of claim 1, 2 or 3 wherein said C-terminal tail can chelate one or more ions.
8. The polypeptide chain of claim 7 wherein said ion is a metal ion.
9. The polypeptide chain of claim 1, 2 or 3 wherein said crosslinking means is a derivatizable amino acid side chain.
10. The polypeptide chain of claim 9 wherein said derivatizable amino acid is selected from the group consisting of lysine, arginine and histidine.
11. The polypeptide chain of claim 9 wherein said derivatizable amino acid is a cysteine amino acid.
12. The polypeptide chain of claim 1, 2 or 3 wherein said crosslinking means comprises a posttranslationally modified amino acid.
13. The polypeptide chain of claim 12 wherein said posttranslationally modified amino acid is the Asn residue located in the amino acid sequence selected form group of Asn-Xaa-Ser and Asn-Xaa-Thr.
14. The formulation of claim 1 or 2 wherein said linkage is a chemical bridge.
15. The formulation of claim 1 or 2 wherein said linkage comprises a disulfide bond.
16. The formulation of claim 1 or 2 wherein said linkage comprises a bismaleimidohexane cross-linker.
17. The formulation of claim 1 or 2 wherein said linkage comprises a bismaleimidocaproyl amino acid linker.
18. The formulation of claim 1 or 2 wherein said linkage comprises a peptidyl linker.
19. The formulation of claim 1 or 2 wherein said linkage forms a substantially inflexible structure under physiological conditions.
20. The formulation of claim 1 or 2 wherein said linkage has a length and composition optimized for binding of two preselected antigens expressed on a tissue surface in a mammal.
21. The formulation of claim 1 or 2 wherein said linkage comprises a detectable moiety.
22. The formulation of claim 21 wherein said detectable moiety comprises Technetium−99m.
23. The formulation of claim 21 wherein said detectable moiety comprises means for inducing proton relaxation in vivo.
24. The formulation of claim 1 or 2 wherein said dimeric biosynthetic construct targets said epitope on said antigen with an avidity greater than that of a monoclonal antibody having the same antigenic determinant as said construct, or a fragment thereof.
25. The formulation of claim 1 or 2 wherein said dimeric biosynthetic construct targets said epitope on said antigen with an avidity greater than that of either of said polypeptide chains individually.
26. The formulation of claim 1 or 2 wherein said preselected antigen is expressed on the surface of a cell.
27. The formulation of claim 1 or 2 wherein said antigen is an intracellular component exposed upon cell lysis.
28. The formulation of claim 1 or 2 wherein said dimeric construct binds two different epitopes.
29. The formulation of claim 1 or 2 wherein one of said binding sites further comprises a catalytic site.
30. The formulation of claim 1 or 2 wherein one of said binding sites binds an epitope on a therapeutic agent to be targeted to a cell surface.
31. The formulation of claim 30 wherein said therapeutic agent is a cytotoxic agent.
32. The formulation of claim 1, 2 or 3 wherein said construct has improved in vivo imaging characteristics.
33-49. (canceled)
US10/684,237 1992-02-06 2003-10-10 Biosynthetic binding proteins for immuno-targeting Abandoned US20060147444A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/684,237 US20060147444A1 (en) 1992-02-06 2003-10-10 Biosynthetic binding proteins for immuno-targeting

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US83196792A 1992-02-06 1992-02-06
US08/133,804 US5534254A (en) 1992-02-06 1993-10-07 Biosynthetic binding proteins for immuno-targeting
US46264195A 1995-06-05 1995-06-05
US55874100A 2000-04-26 2000-04-26
US10/684,237 US20060147444A1 (en) 1992-02-06 2003-10-10 Biosynthetic binding proteins for immuno-targeting

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US55874100A Continuation 1987-05-21 2000-04-26

Publications (1)

Publication Number Publication Date
US20060147444A1 true US20060147444A1 (en) 2006-07-06

Family

ID=25260318

Family Applications (6)

Application Number Title Priority Date Filing Date
US08/133,804 Expired - Lifetime US5534254A (en) 1992-02-06 1993-10-07 Biosynthetic binding proteins for immuno-targeting
US08/356,786 Expired - Lifetime US5877305A (en) 1992-02-06 1994-12-12 DNA encoding biosynthetic binding protein for cancer marker
US08/461,386 Expired - Lifetime US5837846A (en) 1992-02-06 1995-06-05 Biosynthetic binding proteins for immuno-targeting
US08/461,838 Expired - Lifetime US5753204A (en) 1992-02-06 1995-06-05 Biosynthetic binding proteins for immunotargeting
US09/887,853 Expired - Fee Related US7138497B2 (en) 1992-02-06 2001-06-21 Biosynthetic binding proteins for immuno-targeting
US10/684,237 Abandoned US20060147444A1 (en) 1992-02-06 2003-10-10 Biosynthetic binding proteins for immuno-targeting

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US08/133,804 Expired - Lifetime US5534254A (en) 1992-02-06 1993-10-07 Biosynthetic binding proteins for immuno-targeting
US08/356,786 Expired - Lifetime US5877305A (en) 1992-02-06 1994-12-12 DNA encoding biosynthetic binding protein for cancer marker
US08/461,386 Expired - Lifetime US5837846A (en) 1992-02-06 1995-06-05 Biosynthetic binding proteins for immuno-targeting
US08/461,838 Expired - Lifetime US5753204A (en) 1992-02-06 1995-06-05 Biosynthetic binding proteins for immunotargeting
US09/887,853 Expired - Fee Related US7138497B2 (en) 1992-02-06 2001-06-21 Biosynthetic binding proteins for immuno-targeting

Country Status (8)

Country Link
US (6) US5534254A (en)
EP (3) EP1514934B1 (en)
JP (8) JPH08500962A (en)
AT (3) ATE503496T1 (en)
AU (1) AU675929B2 (en)
CA (2) CA2129663C (en)
DE (3) DE69334351D1 (en)
WO (1) WO1993016185A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180117153A1 (en) * 2015-04-22 2018-05-03 Ucb Biopharma Sprl Method of monomerisation of recombinant antibody molecules

Families Citing this family (1314)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7838216B1 (en) * 1986-03-05 2010-11-23 The United States Of America, As Represented By The Department Of Health And Human Services Human gene related to but distinct from EGF receptor gene
US5869620A (en) * 1986-09-02 1999-02-09 Enzon, Inc. Multivalent antigen-binding proteins
US20050058638A1 (en) * 1987-05-21 2005-03-17 Huston James S. Biosynthetic binding proteins for immuno-targeting
KR0184860B1 (en) * 1988-11-11 1999-04-01 메디칼 리써어치 카운실 Single domain ligands receptors comprising said ligands methods for their production and use of said ligands
CA2100671C (en) * 1991-02-27 1999-02-02 James S. Huston Serine rich peptide linkers
JPH06510529A (en) * 1991-08-05 1994-11-24 イゲン,インコーポレーテッド Prodrugs activated by targeted catalytic proteins
US6027725A (en) * 1991-11-25 2000-02-22 Enzon, Inc. Multivalent antigen-binding proteins
US20070031931A1 (en) * 1992-02-06 2007-02-08 Chiron Corporation Biosynthetic binding proteins for immuno-targeting
AU675929B2 (en) * 1992-02-06 1997-02-27 Curis, Inc. Biosynthetic binding protein for cancer marker
US7754211B2 (en) * 1992-04-10 2010-07-13 Research Development Foundation Immunotoxins directed against c-erbB-2(HER-2/neu) related surface antigens
US20080152586A1 (en) * 1992-09-25 2008-06-26 Avipep Pty Limited High avidity polyvalent and polyspecific reagents
AU5670194A (en) * 1992-11-20 1994-06-22 Enzon, Inc. Linker for linked fusion polypeptides
GB9225453D0 (en) 1992-12-04 1993-01-27 Medical Res Council Binding proteins
JP3720353B2 (en) * 1992-12-04 2005-11-24 メディカル リサーチ カウンシル Multivalent and multispecific binding proteins, their production and use
AU680685B2 (en) 1993-09-22 1997-08-07 Medical Research Council Retargeting antibodies
US5753225A (en) * 1993-12-03 1998-05-19 The Regents Of The University Of California Antibodies that mimic actions of neurotrophins
IT1264083B1 (en) * 1993-12-10 1996-09-10 Enea Ente Nuove Tec PROCEDURE FOR THE PRODUCTION IN PLANTS OF ENGINEERED ANTIBODY, PRODUCED ANTIBODY AND THEIR USE IN DIAGNOSIS AND THERAPY.
US5763733A (en) * 1994-10-13 1998-06-09 Enzon, Inc. Antigen-binding fusion proteins
US6514942B1 (en) * 1995-03-14 2003-02-04 The Board Of Regents, The University Of Texas System Methods and compositions for stimulating T-lymphocytes
AUPO591797A0 (en) * 1997-03-27 1997-04-24 Commonwealth Scientific And Industrial Research Organisation High avidity polyvalent and polyspecific reagents
US6685940B2 (en) * 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US5968511A (en) 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
PT896586E (en) * 1996-03-27 2007-01-31 Genentech Inc Erbb3 antibodies
US6136311A (en) 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
JP2000515735A (en) * 1996-07-03 2000-11-28 ジェネンテック インコーポレーテッド Hepatocyte growth factor receptor agonist
DE69731836T2 (en) * 1996-07-23 2005-12-01 Pangenetics B.V. INDUCTION OF T CELL TOLERANCE USING A SOLUBLE MOLECULAR THAT CAN CREATE TWO COSTIMULATION PATHS AT THE SAME TIME
WO1998005684A2 (en) * 1996-08-05 1998-02-12 President And Fellows Of Havard College Mhc binding peptide oligomers and methods of use
US7371376B1 (en) * 1996-10-18 2008-05-13 Genentech, Inc. Anti-ErbB2 antibodies
DE69841217D1 (en) * 1997-01-13 2009-11-19 Univ Emory GLUTATHION FOR THE TREATMENT OF INFLUENZA INFECTIONS
WO1998033809A1 (en) * 1997-01-31 1998-08-06 The General Hospital Corporation Compositions and methods for imaging gene expression
US20060235209A9 (en) * 1997-03-10 2006-10-19 Jin-An Jiao Use of anti-tissue factor antibodies for treating thromboses
US7749498B2 (en) * 1997-03-10 2010-07-06 Genentech, Inc. Antibodies for inhibiting blood coagulation and methods of use thereof
US5986065A (en) * 1997-03-10 1999-11-16 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US20030109680A1 (en) * 2001-11-21 2003-06-12 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
AU7266898A (en) 1997-04-30 1998-11-24 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
US20040009166A1 (en) * 1997-04-30 2004-01-15 Filpula David R. Single chain antigen-binding polypeptides for polymer conjugation
AU735883C (en) * 1997-06-13 2002-02-07 Genentech Inc. Protein recovery by chromatography followed by filtration upon a charged layer
US6172213B1 (en) 1997-07-02 2001-01-09 Genentech, Inc. Anti-IgE antibodies and method of improving polypeptides
US5994511A (en) 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
FR2766826B1 (en) * 1997-08-04 2001-05-18 Pasteur Institut VECTORS DERIVED FROM ANTIBODIES FOR TRANSFERRING SUBSTANCES IN CELLS
US6737249B1 (en) * 1997-08-22 2004-05-18 Genentech, Inc. Agonist antibodies
EP1947119A3 (en) 1997-12-12 2012-12-19 Genentech, Inc. Treatment of cancer with anti-erb2 antibodies in combination with a chemotherapeutic agent
ZA9811162B (en) 1997-12-12 2000-06-07 Genentech Inc Treatment with anti-ERBB2 antibodies.
EP1941905A1 (en) 1998-03-27 2008-07-09 Genentech, Inc. APO-2 Ligand-anti-her-2 antibody synergism
DE19819846B4 (en) * 1998-05-05 2016-11-24 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Multivalent antibody constructs
AU4009899A (en) * 1998-05-23 1999-12-13 Tanox, Inc. Molecules targeting cd40 and tumor cells
GB9812545D0 (en) * 1998-06-10 1998-08-05 Celltech Therapeutics Ltd Biological products
US7138103B2 (en) * 1998-06-22 2006-11-21 Immunomedics, Inc. Use of bi-specific antibodies for pre-targeting diagnosis and therapy
ATE460946T1 (en) * 1998-06-22 2010-04-15 Immunomedics Inc USE OF BISPECIFIC ANTIBODIES IN DIAGNOSIS AND THERAPY
US7387772B1 (en) 1999-06-22 2008-06-17 Immunimedics, Inc. Chimeric, human and humanized anti-CSAP monoclonal antibodies
US6962702B2 (en) * 1998-06-22 2005-11-08 Immunomedics Inc. Production and use of novel peptide-based agents for use with bi-specific antibodies
US7405320B2 (en) 1998-06-22 2008-07-29 Immunomedics, Inc. Therapeutic and diagnostic conjugates for use with multispecific antibodies
US7833528B2 (en) * 1998-06-22 2010-11-16 Immunomedics, Inc. Use of multispecific, non-covalent complexes for targeted delivery of therapeutics
DE19831429A1 (en) * 1998-07-07 2000-04-27 Jerini Biotools Gmbh Preparation and identification of polytopic peptide, useful e.g. therapeutically and for detecting antigens or antibodies, by linking fragments of a protein and testing for specific binding
US6228360B1 (en) * 1998-08-19 2001-05-08 Ajinomoto Co., Inc. Antithrombotic agent and humanized anti-von Willebrand factor monoclonal antibody
US7067144B2 (en) * 1998-10-20 2006-06-27 Omeros Corporation Compositions and methods for systemic inhibition of cartilage degradation
US6333396B1 (en) 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
US6818749B1 (en) * 1998-10-31 2004-11-16 The United States Of America As Represented By The Department Of Health And Human Services Variants of humanized anti carcinoma monoclonal antibody cc49
WO2000029431A1 (en) * 1998-11-17 2000-05-25 Tanox, Inc. Bispecific molecules cross-linking itim and itam for therapy
US7118743B2 (en) 1998-11-17 2006-10-10 Tanox, Inc. Bispecific molecules cross-linking ITIM and ITAM for therapy
EP1950300A3 (en) 1998-11-18 2011-03-23 Genentech, Inc. Antibody variants with higher binding affinity compared to parent antibodies
US7041292B1 (en) 1999-06-25 2006-05-09 Genentech, Inc. Treating prostate cancer with anti-ErbB2 antibodies
DK2283866T3 (en) 1999-06-25 2015-05-18 Genentech Inc METHODS OF TREATMENT USING ANTI-ERBB ANTIBODY-MAYTANSINOID CONJUGATES
EP1189641B1 (en) 1999-06-25 2009-07-29 Genentech, Inc. HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
US20040013667A1 (en) * 1999-06-25 2004-01-22 Genentech, Inc. Treatment with anti-ErbB2 antibodies
US20030086924A1 (en) * 1999-06-25 2003-05-08 Genentech, Inc. Treatment with anti-ErbB2 antibodies
JP4579471B2 (en) * 1999-06-25 2010-11-10 ジェネンテック, インコーポレイテッド Treatment of prostate cancer with anti-ErbB2 antibody
US6949245B1 (en) * 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
ES2330301T3 (en) 1999-08-27 2009-12-09 Genentech, Inc. DOSAGES FOR TREATMENT WITH ANTI-ERBB2 ANTIBODIES.
CA2385528C (en) 1999-10-01 2013-12-10 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
US6794494B1 (en) * 2003-04-14 2004-09-21 Arius Research, Inc. Cancerous disease modifying antibodies
UA74803C2 (en) 1999-11-11 2006-02-15 Осі Фармасьютікалз, Інк. A stable polymorph of n-(3-ethynylphenyl)-6,7-bis(2-methoxyetoxy)-4-quinazolinamine hydrochloride, a method for producing thereof (variants) and pharmaceutical use
WO2001056603A1 (en) 2000-02-01 2001-08-09 Tanox, Inc. Cd40-binding apc-activating molecules
WO2001062800A1 (en) * 2000-02-24 2001-08-30 Eidgenössische Technische Hochschule Zürich Antibody specific for the ed-b domain of fibronectin, conjugates comprising said antibody, and their use for the detection and treatment of angiogenesis
US6319694B1 (en) * 2000-03-03 2001-11-20 Genopsys, Inc. Random truncation and amplification of nucleic acid
US7097840B2 (en) * 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
ES2528794T3 (en) 2000-04-11 2015-02-12 Genentech, Inc. Multivalent antibodies and uses thereof
WO2001080883A1 (en) * 2000-04-26 2001-11-01 Elusys Therapeutics, Inc. Bispecific molecules and uses thereof
AU2001259271A1 (en) * 2000-04-28 2001-11-12 Millennium Pharmaceuticals, Inc. 14094, a novel human trypsin family member and uses thereof
CN102698265A (en) 2000-05-19 2012-10-03 杰南技术公司 Gene detection assay for improving the likelihood of an effective response to an erbb antagonist cancer therapy
WO2001091793A1 (en) * 2000-05-26 2001-12-06 Smithkline Beecham Corporation Anti-rank ligand monoclonal antibodies useful in treatment of rank ligand mediated disorders
WO2001097858A2 (en) 2000-06-20 2001-12-27 Idec Pharmaceuticals Corporation Cold anti-cd20 antibody/radiolabeled anti-cd22 antibody combination
GB0027328D0 (en) * 2000-06-23 2000-12-27 Aventis Pharma Inc Bioengineered vehicles for targeted nucleic acid delivery
PL364358A1 (en) * 2000-09-07 2004-12-13 Schering Ag Receptor in the edb fibronectin domain (ii)
US6631283B2 (en) * 2000-11-15 2003-10-07 Virginia Tech Intellectual Properties, Inc. B/B-like fragment targeting for the purposes of photodynamic therapy and medical imaging
AR032028A1 (en) 2001-01-05 2003-10-22 Pfizer ANTIBODIES AGAINST THE RECEIVER OF THE SIMILAR TO INSULIN GROWTH FACTOR
US20020159996A1 (en) 2001-01-31 2002-10-31 Kandasamy Hariharan Use of CD23 antagonists for the treatment of neoplastic disorders
RU2316337C2 (en) 2001-04-24 2008-02-10 Мерк Патент Гмбх Combined therapy by using antiangiogenic agents and tnf-alpha
CN1195779C (en) 2001-05-24 2005-04-06 中国科学院遗传与发育生物学研究所 Double-specificity antibody resisting human ovary cancer and human CD3
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
EP1992643A3 (en) 2001-06-20 2008-12-10 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2003000194A2 (en) 2001-06-21 2003-01-03 Pfizer Inc. Thienopyridine and thienopyrimidine anticancer agents
WO2003024392A2 (en) 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20050123925A1 (en) 2002-11-15 2005-06-09 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2003088808A2 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
AR039067A1 (en) * 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40
EP1575571A4 (en) 2002-01-02 2008-06-25 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
IL162201A0 (en) * 2002-01-03 2005-11-20 Schering Ag New methods for diagnosis and treatment of tumours
AU2003210802B2 (en) 2002-02-05 2009-09-10 Genentech Inc. Protein purification
US6869787B2 (en) * 2002-02-27 2005-03-22 The United States Of America As Represented By The Secretary Of The Army Ricin vaccine and methods of making and using thereof
CA2481515C (en) 2002-04-10 2013-10-01 Genentech, Inc. Anti-her2 antibody variants
WO2003087338A2 (en) * 2002-04-11 2003-10-23 Amgen, Inc. Her-2 receptor tyrosine kinase molecules and uses thereof
UA77303C2 (en) * 2002-06-14 2006-11-15 Pfizer Derivatives of thienopyridines substituted by benzocondensed heteroarylamide useful as therapeutic agents, pharmaceutical compositions and methods for their use
US7569673B2 (en) * 2002-06-28 2009-08-04 The United States Of America As Represented By The Department Of Health And Human Services Humanized anti-tag 72 cc49 for diagnosis and therapy of human tumors
PT1585966E (en) 2002-07-15 2012-02-20 Hoffmann La Roche Treatment of cancer with the anti-erbb2 antibody rhumab 2c4
ES2629602T5 (en) 2002-09-11 2021-06-08 Genentech Inc Protein purification
AU2002951853A0 (en) * 2002-10-04 2002-10-24 Commonwealth Scientific And Industrial Research Organisation Crystal structure of erbb2 and uses thereof
US9701754B1 (en) 2002-10-23 2017-07-11 City Of Hope Covalent disulfide-linked diabodies and uses thereof
WO2004048525A2 (en) * 2002-11-21 2004-06-10 Genentech, Inc. Therapy of non-malignant diseases or disorders with anti-erbb2 antibodies
US20040186160A1 (en) * 2002-12-13 2004-09-23 Sugen, Inc. Hexahydro-cyclohepta-pyrrole oxindole as potent kinase inhibitors
JP3814285B2 (en) * 2002-12-19 2006-08-23 ファイザー・インク 2- (1H-indazol-6-ylamino) -benzamide compounds as protein kinase inhibitors useful in the treatment of eye diseases
ES2566778T3 (en) 2003-02-01 2016-04-15 Tanox, Inc. High affinity anti-human IgE antibodies
GEP20084341B (en) 2003-02-26 2008-03-25 Sugen Inc Aminoheteroaryl compounds as protein kinase inhibitors
BRPI0409230A (en) * 2003-04-03 2006-03-28 Pfizer dosage forms comprising ag013736
BRPI0403964B8 (en) 2003-04-04 2021-05-25 Genentech Inc stable liquid formulations, article of manufacture and use of these formulations for the treatment of ige-mediated dysfunction
ES2322267T3 (en) 2003-04-09 2009-06-18 Genentech, Inc. THERAPY OF AN AUTOINMUNOLOGICAL DISEASE IN A PATIENT THAT PRESENTS AN INAPPROPRIATE RESPONSE TO A TNF-ALFA INHIBITOR.
US7425328B2 (en) * 2003-04-22 2008-09-16 Purdue Pharma L.P. Tissue factor antibodies and uses thereof
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
MXPA05012723A (en) 2003-05-30 2006-02-08 Genentech Inc Treatment with anti-vegf antibodies.
KR20060027801A (en) 2003-06-05 2006-03-28 제넨테크, 인크. Combination therapy for b cell disorders
KR100912381B1 (en) * 2003-06-19 2009-08-19 타녹스 인코퍼레이티드 Compositions and methods for treating coagulation related disorders
SI1641822T1 (en) 2003-07-08 2013-08-30 Genentech, Inc. Il-17 a/f heterologous polypeptides and therapeutic uses thereof
ES2786568T3 (en) 2003-07-28 2020-10-13 Genentech Inc Reduction of protein A leaching during protein A affinity chromatography
HN2004000285A (en) * 2003-08-04 2006-04-27 Pfizer Prod Inc ANTIBODIES DIRECTED TO c-MET
CA2536788A1 (en) * 2003-08-29 2005-03-10 Pfizer Inc. Naphthalene carboxamides and their derivatives useful as new anti-angiogenic agents
DE602004017479D1 (en) * 2003-08-29 2008-12-11 Pfizer THIENOPYRIDINPHENYLACETAMIDES SUITED AS NEW ANTIANGIOGENIC AGENTS AND DERIVATIVES THEREOF
US7589181B2 (en) * 2003-08-29 2009-09-15 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Minimally immunogenic variants of SDR-grafted humanized antibody CC49 and their use
AR045563A1 (en) * 2003-09-10 2005-11-02 Warner Lambert Co ANTIBODIES DIRECTED TO M-CSF
KR101520209B1 (en) 2003-11-06 2015-05-13 시애틀 지네틱스, 인크. Monomethylvaline compounds capable of conjugation to ligands
EP1689432B1 (en) 2003-11-17 2009-12-30 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
WO2005051919A1 (en) * 2003-11-26 2005-06-09 Pfizer Products Inc. Aminopyrazole derivatives as gsk-3 inhibitors
US8298532B2 (en) 2004-01-16 2012-10-30 Regeneron Pharmaceuticals, Inc. Fusion polypeptides capable of activating receptors
EP2168986A3 (en) 2004-02-19 2010-07-28 Genentech, Inc. CDR-repaired antibodies
BRPI0509580A (en) * 2004-03-30 2007-11-27 Pfizer Prod Inc signal transduction inhibitor combinations
MXPA06011199A (en) 2004-03-31 2007-04-16 Genentech Inc Humanized anti-tgf-beta antibodies.
US20150017671A1 (en) 2004-04-16 2015-01-15 Yaping Shou Methods for detecting lp-pla2 activity and inhibition of lp-pla2 activity
NZ551180A (en) 2004-06-01 2009-10-30 Genentech Inc Antibody drug conjugates and methods
US20060051345A1 (en) 2004-06-04 2006-03-09 Genentech, Inc. Method for treating multiple sclerosis
SV2006002143A (en) 2004-06-16 2006-01-26 Genentech Inc USE OF AN ANTIBODY FOR THE TREATMENT OF CANCER RESISTANT TO PLATINUM
CA2573821A1 (en) * 2004-07-16 2006-01-26 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-igf-1r antibody
US8604185B2 (en) 2004-07-20 2013-12-10 Genentech, Inc. Inhibitors of angiopoietin-like 4 protein, combinations, and their use
ZA200701183B (en) 2004-07-20 2008-05-28 Genentech Inc Inhibitors of angiopoietin-like 4 protein, combinations, an their use
CN101018780B (en) * 2004-08-26 2012-01-11 辉瑞大药厂 Pyrazole-substituted aminoheteroaryl compounds as protein kinase inhibitors
ATE463486T1 (en) 2004-08-26 2010-04-15 Pfizer ENANTIOMER PURE AMINOHETEROARYL COMPOUNDS AS PROTEIN KINASE INHIBITORS
JP2008510792A (en) * 2004-08-26 2008-04-10 ファイザー・インク Amino heteroaryl compounds as protein tyrosine kinase inhibitors
US7785814B2 (en) * 2004-09-21 2010-08-31 The United States Of America As Represented By The Department Of Health And Human Services Method of detecting cancer based on immune reaction to BORIS
EP1791565B1 (en) 2004-09-23 2016-04-20 Genentech, Inc. Cysteine engineered antibodies and conjugates
US20100111856A1 (en) 2004-09-23 2010-05-06 Herman Gill Zirconium-radiolabeled, cysteine engineered antibody conjugates
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US20060107555A1 (en) * 2004-11-09 2006-05-25 Curtis Marc D Universal snow plow adapter
US7964195B2 (en) 2005-01-07 2011-06-21 Diadexus, Inc. Ovr110 antibody compositions and methods of use
EP3698807A1 (en) 2005-01-21 2020-08-26 Genentech, Inc. Fixed dosing of her antibodies
CA2595395A1 (en) * 2005-02-09 2006-08-17 Genentech, Inc. Inhibiting her2 shedding with matrix metalloprotease antagonists
US20060233791A1 (en) 2005-02-15 2006-10-19 Duke University Anti-CD19 antibodies and uses in oncology
AU2006216732C1 (en) * 2005-02-23 2017-07-20 Genentech, Inc. Extending time to disease progression or survival in cancer patients using a HER dimerization inhibitor
WO2006096861A2 (en) * 2005-03-08 2006-09-14 Genentech, Inc. METHODS FOR IDENTIFYING TUMORS RESPONSIVE TO TREATMENT WITH HER DIMERIZATION INHIBITORS (HDIs)
NZ562453A (en) 2005-03-31 2010-04-30 Agensys Inc Antibodies and related molecules that bind to 161P2F10B proteins
BRPI0608096A2 (en) 2005-04-26 2009-11-10 Pfizer p-cadherin antibodies
EP1885755A4 (en) 2005-05-05 2009-07-29 Univ Duke Anti-cd19 antibody therapy for autoimmune disease
JP2006316040A (en) 2005-05-13 2006-11-24 Genentech Inc Herceptin(r) adjuvant treatment
US7858843B2 (en) 2005-06-06 2010-12-28 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
CA2614436C (en) 2005-07-07 2016-05-17 Seattle Genetics, Inc. Monomethylvaline compounds having phenylalanine side-chain modifications at the c-terminus
AU2006280321A1 (en) 2005-08-15 2007-02-22 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
UA94060C2 (en) 2005-09-07 2011-04-11 Эмджен Фримонт Инк. Monoclonal antibodies that specifically binds alk-1
WO2007035744A1 (en) 2005-09-20 2007-03-29 Osi Pharmaceuticals, Inc. Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
PL2500030T5 (en) 2005-11-04 2019-02-28 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
AU2006335053A1 (en) 2005-11-21 2007-07-19 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
CN105859886A (en) 2005-12-02 2016-08-17 健泰科生物技术公司 Compositions and methods associated with antibodies that bind to IL-22 and IL-22R
US7763245B2 (en) 2005-12-15 2010-07-27 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US7625759B2 (en) 2005-12-19 2009-12-01 Genentech, Inc. Method for using BOC/CDO to modulate hedgehog signaling
RU2450020C2 (en) 2006-01-05 2012-05-10 Дженентек, Инк. ANTI-EphB4 ANTIBODIES AND METHODS OF USING SAID ANTIBODIES
US8097705B2 (en) * 2006-01-13 2012-01-17 Irm Llc Methods and compositions for treating allergic diseases
ES2521679T3 (en) 2006-01-18 2014-11-13 Merck Patent Gmbh Specific therapy using integrin ligands for cancer treatment
CA2638821A1 (en) 2006-02-17 2007-10-11 Genentech, Inc. Gene disruptons, compositions and methods relating thereto
EP2540741A1 (en) 2006-03-06 2013-01-02 Aeres Biomedical Limited Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
AR059851A1 (en) 2006-03-16 2008-04-30 Genentech Inc ANTIBODIES OF EGFL7 AND METHODS OF USE
EP1996716B1 (en) 2006-03-20 2011-05-11 The Regents of the University of California Engineered anti-prostate stem cell antigen (psca) antibodies for cancer targeting
EP2389948A1 (en) 2006-03-23 2011-11-30 Novartis AG Anti-tumor cell antigen antibody therapeutics
EP2614839A3 (en) 2006-04-05 2015-01-28 Genentech, Inc. Method for using BOC/CDO to modulate hedgehog signaling
EP2082645A1 (en) 2006-04-19 2009-07-29 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
EA200802058A1 (en) 2006-05-09 2009-06-30 Пфайзер Продактс Инк. DERIVATIVES OF CYCLO-ALKYLAMINO ACIDS AND THEIR PHARMACEUTICAL COMPOSITIONS
NL2000613C2 (en) 2006-05-11 2007-11-20 Pfizer Prod Inc Triazole pyrazine derivatives.
AR060978A1 (en) 2006-05-30 2008-07-23 Genentech Inc ANTIBODIES AND IMMUNOCATE PLAYERS AND THEIR USES
AR061170A1 (en) 2006-06-02 2008-08-06 Aveo Pharmaceuticals Inc PROTEINS THAT JOIN THE HEPATOCITE GROWTH FACTOR (HGF)
JP4686634B2 (en) 2006-06-02 2011-05-25 アベオ ファーマシューティカルズ, インコーポレイテッド Hepatocyte growth factor (HGF) binding protein
FR2902799B1 (en) 2006-06-27 2012-10-26 Millipore Corp METHOD AND UNIT FOR PREPARING A SAMPLE FOR THE MICROBIOLOGICAL ANALYSIS OF A LIQUID
RU2499001C2 (en) 2006-06-30 2013-11-20 Ново Нордиск А/С Antibodies to nkg2a and their applications
ES2612383T3 (en) 2006-07-19 2017-05-16 The Trustees Of The University Of Pennsylvania WSX-1 / IL-27 as a target for anti-inflammatory responses
US7862813B2 (en) 2006-07-29 2011-01-04 Bjork Jr Robert Lamar Bi-specific monoclonal antibody (specific for both CD3 and CD11b) therapeutic drug
US8586006B2 (en) 2006-08-09 2013-11-19 Institute For Systems Biology Organ-specific proteins and methods of their use
JP2010501172A (en) 2006-08-25 2010-01-21 オンコセラピー・サイエンス株式会社 Prognostic markers and therapeutic targets for lung cancer
WO2008031051A2 (en) 2006-09-08 2008-03-13 United Therapeutics Corporation Clinical diagnosis of hepatic fibrosis using a novel panel of human serum protein biomarkers
MX2009002414A (en) 2006-09-08 2009-05-20 Medimmune Llc Humanized anti-cd19 antibodies and their use in treatment of oncology, transplantation and autoimmune disease.
WO2011008990A1 (en) 2009-07-15 2011-01-20 Prometheus Laboratories Inc. Drug selection for gastric cancer therapy using antibody-based arrays
US20080076139A1 (en) 2006-09-21 2008-03-27 Sharat Singh Methods and compositions for detecting the activation states of multiple signal transducers in rare circulating cells
SI2502938T1 (en) 2006-10-27 2015-05-29 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
MX2009004757A (en) 2006-11-14 2009-05-21 Genentech Inc Modulators of neuronal regeneration.
AU2007325838B2 (en) 2006-11-22 2013-09-19 Bristol-Myers Squibb Company Targeted therapeutics based on engineered proteins for tyrosine kinases receptors, including IGF-IR
JP5391073B2 (en) 2006-11-27 2014-01-15 ディアデクサス インコーポレーテッド Ovr110 antibody compositions and methods of use
US7935791B2 (en) 2006-12-18 2011-05-03 Genentech, Inc. Antagonist anti-Notch3 antibodies and their use in the prevention and treatment of Notch3-related diseases
WO2008079280A1 (en) 2006-12-21 2008-07-03 Millipore Corporation Purification of proteins
US8362217B2 (en) 2006-12-21 2013-01-29 Emd Millipore Corporation Purification of proteins
US8569464B2 (en) 2006-12-21 2013-10-29 Emd Millipore Corporation Purification of proteins
WO2008077171A1 (en) * 2006-12-22 2008-07-03 Novelix Therapeutics Gmbh Treatment of diabetes by at least one epidermal growth factor receptor specific antibody or a derivative thereof
MX2009007597A (en) 2007-01-18 2009-07-22 Merck Patent Gmbh Specific therapy and medicament using integrin ligands for treating cancer.
CA2676766A1 (en) 2007-02-09 2008-08-21 Genentech, Inc. Anti-robo4 antibodies and uses therefor
US7875431B2 (en) 2007-02-22 2011-01-25 Genentech, Inc. Methods for detecting inflammatory bowel disease
WO2008109440A2 (en) 2007-03-02 2008-09-12 Genentech, Inc. Predicting response to a her dimerisation inhibitor based on low her3 expression
US7960139B2 (en) 2007-03-23 2011-06-14 Academia Sinica Alkynyl sugar analogs for the labeling and visualization of glycoconjugates in cells
JP5926487B2 (en) 2007-04-13 2016-05-25 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Method for treating cancer resistant to ErbB therapy
RU2549701C2 (en) 2007-05-07 2015-04-27 Медиммун, Ллк Anti-icos antibodies and their application in treatment of oncological, transplantation-associated and autoimmune diseases
US20090163698A1 (en) * 2007-05-11 2009-06-25 John Joseph Grigsby Method for Preparing Antibody Conjugates
WO2008154249A2 (en) * 2007-06-08 2008-12-18 Genentech, Inc. Gene expression markers of tumor resistance to her2 inhibitor treatment
US9551033B2 (en) 2007-06-08 2017-01-24 Genentech, Inc. Gene expression markers of tumor resistance to HER2 inhibitor treatment
SI3597659T1 (en) 2007-07-09 2023-04-28 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
TW200918089A (en) 2007-07-16 2009-05-01 Genentech Inc Humanized anti-CD79b antibodies and immunoconjugates and methods of use
JP5469600B2 (en) 2007-07-16 2014-04-16 ジェネンテック, インコーポレイテッド Anti-CD79b antibody and immunoconjugate and method of use thereof
EP2198021A4 (en) 2007-08-24 2011-01-19 Oncotherapy Science Inc Ebi3, dlx5, nptx1 and cdkn3 for target genes of lung cancer therapy and diagnosis
EP2190478B1 (en) 2007-08-24 2016-03-23 Oncotherapy Science, Inc. Dkk1 oncogene as therapeutic target for cancer and a diagnosing marker
BRPI0815757A2 (en) 2007-08-24 2015-02-18 Oncotherapy Science Inc PKIB AND NAALAD2 GENES AS TARGETS OF PROSTATE CANCER TREATMENT AND DIAGNOSIS
CA2698203C (en) 2007-08-29 2018-09-11 Sanofi-Aventis Humanized anti-cxcr5 antibodies, derivatives thereof and their use
JP6126773B2 (en) 2007-09-04 2017-05-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア High affinity anti-prostatic stem cell antigen (PSCA) antibody for cancer targeting and detection
PT2185574E (en) 2007-09-07 2013-08-26 Agensys Inc Antibodies and related molecules that bind to 24p4c12 proteins
CL2008002886A1 (en) 2007-09-26 2009-12-04 Chugai Pharmaceutical Co Ltd Constant region of a human antibody; anti-interleukin-6 (yl-6) receptor antibody and pharmaceutical composition comprising it.
US8691222B2 (en) 2007-10-02 2014-04-08 Genentech, Inc. NLRR-1 antagonists and uses thereof
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
EP2200631A1 (en) 2007-10-16 2010-06-30 Zymogenetics, Inc. Combination of blys inhibition and anti-cd 20 agents for treatment of autoimmune disease
RS53850B2 (en) 2007-10-30 2018-07-31 Genentech Inc Antibody purification by cation exchange chromatography
SI2514436T1 (en) 2007-11-07 2018-04-30 Genentech, Inc. Il-22 for use in treating microbial disorders
KR20100097691A (en) 2007-11-12 2010-09-03 테라클론 사이언시스, 아이엔씨. Compositions and methods for the therapy and diagnosis of influenza
KR20110015409A (en) 2007-11-29 2011-02-15 제넨테크, 인크. Gene expression markers for inflammatory bowel disease
AR069501A1 (en) 2007-11-30 2010-01-27 Genentech Inc ANTI-VEGF ANTIBODIES (VASCULAR ENDOTELIAL GROWTH FACTOR)
CN105001333B (en) 2007-12-14 2019-05-17 诺沃—诺迪斯克有限公司 Anti-human NKG2D antibody and application thereof
US10416162B2 (en) 2007-12-20 2019-09-17 Monogram Biosciences, Inc. Her2 diagnostic methods
US8253725B2 (en) * 2007-12-28 2012-08-28 St. Jude Medical, Atrial Fibrillation Division, Inc. Method and system for generating surface models of geometric structures
US7914785B2 (en) 2008-01-02 2011-03-29 Bergen Teknologieverforing As B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
EP2077281A1 (en) 2008-01-02 2009-07-08 Bergen Teknologioverforing AS Anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
AR070141A1 (en) 2008-01-23 2010-03-17 Glenmark Pharmaceuticals Sa SPECIFIC HUMANIZED ANTIBODIES FOR VON WILLEBRAND FACTOR
EP2247619A1 (en) 2008-01-24 2010-11-10 Novo Nordisk A/S Humanized anti-human nkg2a monoclonal antibody
WO2009094561A1 (en) * 2008-01-24 2009-07-30 The Government Of The United States Of America As Represented By The Secretary Of The Departmentof Induced internalization of surface receptors
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
SI2657253T1 (en) 2008-01-31 2017-10-30 Genentech, Inc. Anti-CD79b antibodies and immunoconjugates and methods of use
MX2010008874A (en) 2008-02-14 2010-09-22 Bristol Myers Squibb Co Targeted therapeutics based on engineered proteins that bind egfr.
CA2923248A1 (en) 2008-02-25 2009-09-03 Nestec S.A. Methods for detecting truncated receptors
AU2009223688B2 (en) 2008-03-10 2014-12-11 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of cytomegalovirus infections
DK2644194T3 (en) 2008-03-18 2017-07-03 Genentech Inc Combinations of an anti-HER2 antibody-drug conjugate and docetaxel
ES2354661B1 (en) 2008-05-08 2012-02-01 Consejo Superior De Investigaciones Científicas (Csic) METHOD FOR GENERATING MONOCLONAL ANTIBODIES RECOGNIZING MEMBRANE ANTIGENS FROM NEURAL PROGENITING CELLS, ANTIBODIES PRODUCED BY SUCH METHOD, AND USES.
US8093018B2 (en) 2008-05-20 2012-01-10 Otsuka Pharmaceutical Co., Ltd. Antibody identifying an antigen-bound antibody and an antigen-unbound antibody, and method for preparing the same
AR071874A1 (en) 2008-05-22 2010-07-21 Bristol Myers Squibb Co ARMAZON DOMAIN PROTEINS BASED ON MULTIVALENT FIBRONECTINE
US8999702B2 (en) 2008-06-11 2015-04-07 Emd Millipore Corporation Stirred tank bioreactor
BRPI0812682A2 (en) 2008-06-16 2010-06-22 Genentech Inc metastatic breast cancer treatment
JP5986745B2 (en) 2008-07-15 2016-09-06 アカデミア シニカAcademia Sinica Glycan arrays on PTFE-like aluminum-coated glass slides and related methods
WO2010011349A2 (en) * 2008-07-25 2010-01-28 Supergen, Inc. Pyrimidine-2,4-diamine jak2 kinase inhibiting anti-inflammation use
HUE047316T2 (en) 2008-08-14 2020-04-28 Genentech Inc Methods for removing a contaminant using indigenous protein displacement ion exchange membrane chromatography
CA2735433C (en) 2008-09-07 2016-02-16 Glyconex Inc. Anti-extended type i glycosphingolipid antibody, derivatives thereof and use
TW201014605A (en) 2008-09-16 2010-04-16 Genentech Inc Methods for treating progressive multiple sclerosis
CA2737597C (en) 2008-10-16 2017-03-14 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Fully human antibodies to high molecular weight-melanoma associated antigen and uses thereof
AU2009308293B2 (en) 2008-10-22 2015-02-05 Genentech, Inc. Modulation of axon degeneration
AU2009308707A1 (en) 2008-10-31 2010-05-06 Biogen Idec Ma Inc. LIGHT targeting molecules and uses thereof
US20110293605A1 (en) 2008-11-12 2011-12-01 Hasige Sathish Antibody formulation
EP3693014A1 (en) 2008-11-13 2020-08-12 The General Hospital Corporation Methods and compositions for regulating iron homeostasis by modulation bmp-6
WO2010059315A1 (en) 2008-11-18 2010-05-27 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
EP2752189B1 (en) 2008-11-22 2016-10-26 F. Hoffmann-La Roche AG Use of anti-vegf antibody in combination with chemotherapy for treating breast cancer
WO2010062857A1 (en) 2008-11-26 2010-06-03 Allergan, Inc. Klk-13 antibody inhibitor for treating dry eye
WO2010065568A2 (en) 2008-12-01 2010-06-10 Laboratory Corporation Of America Holdings METHODS AND ASSAYS FOR MEASURING p95 AND/OR p95 IN A SAMPLE AND ANTIBODIES SPECIFIC FOR p95
SI2376535T1 (en) 2008-12-09 2017-07-31 F. Hoffmann-La Roche Ag Anti-pd-l1 antibodies and their use to enhance t-cell function
JP2012512244A (en) 2008-12-16 2012-05-31 イー・エム・デイー・ミリポア・コーポレイシヨン Protein purification
CN105037535A (en) 2008-12-16 2015-11-11 Emd密理博公司 Stirred tank reactor and method
KR20110101212A (en) 2008-12-17 2011-09-15 제넨테크, 인크. Hepatitis c virus combination therapy
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
ES2535723T3 (en) 2009-01-15 2015-05-14 Laboratory Corporation Of America Holdings Methods for determining the patient's response by measuring Her-3
SG172857A1 (en) 2009-02-09 2011-08-29 Supergen Inc Pyrrolopyrimidinyl axl kinase inhibitors
WO2010096486A1 (en) 2009-02-17 2010-08-26 Cornell Research Foundation, Inc. Methods and kits for diagnosis of cancer and prediction of therapeutic value
MX2011008843A (en) 2009-02-23 2011-12-14 Glenmark Pharmaceuticals Sa Humanized antibodies that bind to cd19 and their uses.
US20120189641A1 (en) 2009-02-25 2012-07-26 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
WO2010099137A2 (en) 2009-02-26 2010-09-02 Osi Pharmaceuticals, Inc. In situ methods for monitoring the emt status of tumor cells in vivo
WO2010099363A1 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
WO2010099138A2 (en) 2009-02-27 2010-09-02 Osi Pharmaceuticals, Inc. Methods for the identification of agents that inhibit mesenchymal-like tumor cells or their formation
JP2012519282A (en) 2009-02-27 2012-08-23 オーエスアイ・ファーマシューティカルズ,エルエルシー Methods for identifying mesenchymal tumor cells or agents that inhibit their production
US20100222381A1 (en) 2009-02-27 2010-09-02 Hariprasad Vankayalapati Cyclopentathiophene/cyclohexathiophene DNA methyltransferase inhibitors
EP2409155A1 (en) 2009-03-15 2012-01-25 Technion Research and Development Foundation, Ltd. Soluble hla complexes for use in disease diagnosis
SI3260136T1 (en) 2009-03-17 2021-05-31 Theraclone Sciences, Inc. Human immunodeficiency virus (hiv) -neutralizing antibodies
ES2572728T3 (en) 2009-03-20 2016-06-02 F. Hoffmann-La Roche Ag Bispecific anti-HER antibodies
CA2754163C (en) 2009-03-25 2019-04-09 Genentech, Inc. Anti-fgfr3 antibodies and methods using same
UA105384C2 (en) 2009-04-01 2014-05-12 Дженентек, Инк. Treatment of insulin-resistant disorders
AU2010236787A1 (en) 2009-04-01 2011-11-10 Genentech, Inc. Anti-FcRH5 antibodies and immunoconjugates and methods of use
EP2417156B1 (en) 2009-04-07 2015-02-11 Roche Glycart AG Trivalent, bispecific antibodies
WO2010118243A2 (en) 2009-04-08 2010-10-14 Genentech, Inc. Use of il-27 antagonists to treat lupus
US8753631B2 (en) * 2009-05-01 2014-06-17 New York University Therapeutic agents for inducing platelet fragmentation and treating thromboembolic disorders
WO2010133967A1 (en) 2009-05-14 2010-11-25 University Of Oxford Clinical diagnosis of hepatic fibrosis using a novel panel of low abundant human plasma protein biomarkers
AU2010249787A1 (en) 2009-05-20 2011-12-22 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
EA201101651A1 (en) 2009-05-25 2012-08-30 Мерк Патент Гмбх CONTINUOUS INTRODUCTION OF INTEGRINE LIGANDS FOR CANCER TREATMENT
JP5705836B2 (en) 2009-05-29 2015-04-22 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Modulators for HER2 signaling in gastric cancer patients expressing HER2
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
CN102597005A (en) 2009-06-23 2012-07-18 阿雷克森制药公司 Bispecific antibodies that bind to complement proteins
MX2012000417A (en) 2009-07-07 2012-02-08 Genentech Inc Diagnosis and treatment of autoimmune demyelinating diseases.
EP2584049A3 (en) 2009-07-20 2013-08-28 Genentech, Inc. Gene expression markers for Crohn's disease
TW201106972A (en) 2009-07-27 2011-03-01 Genentech Inc Combination treatments
NZ597531A (en) 2009-07-31 2014-05-30 Genentech Inc Inhibition of tumor metastasis using bv8- or g-csf-antagonists
WO2011014775A1 (en) 2009-07-31 2011-02-03 The Brigham And Women's Hospital, Inc. Modulation of sgk1 expression in th17 cells to modulate th17-mediated immune responses
US20110039300A1 (en) 2009-08-10 2011-02-17 Robert Bayer Antibodies with enhanced adcc functions
US20110038871A1 (en) 2009-08-11 2011-02-17 Veena Viswanth Ccr2 inhibitors for treating conditions of the eye
HUE048980T2 (en) 2009-08-11 2020-08-28 Hoffmann La Roche Production of proteins in glutamine-free cell culture media
US20110053223A1 (en) 2009-08-14 2011-03-03 Robert Bayer Cell culture methods to make antibodies with enhanced adcc function
WO2011022264A1 (en) 2009-08-15 2011-02-24 Genentech, Inc. Anti-angiogenesis therapy for the treatment of previously treated breast cancer
WO2011027249A2 (en) 2009-09-01 2011-03-10 Pfizer Inc. Benzimidazole derivatives
US9428548B2 (en) 2009-09-01 2016-08-30 Genentech, Inc. Enhanced protein purification through a modified protein A elution
US9321823B2 (en) 2009-09-02 2016-04-26 Genentech, Inc. Mutant smoothened and methods of using the same
RU2015153109A (en) 2009-09-16 2019-01-15 Дженентек, Инк. SUPERSPIRAL AND / OR BINDING PROTEIN COMPLEXES AND THEIR APPLICATIONS
JP5819307B2 (en) 2009-10-20 2015-11-24 ネステク ソシエテ アノニム Proximity-mediated assay to detect oncogenic fusion proteins
BR112012009409A2 (en) 2009-10-22 2017-02-21 Genentech Inc method of identifying an inhibitory substance, antagonist molecule, isolated nucleic acid, vector, host cell, method of making the molecule, composition, article of manufacture, method of inhibiting a biological activity, method of treating a pathological condition, method for detect msp in a sample and method to detect hepsin in a sample
CN104043126A (en) 2009-10-22 2014-09-17 霍夫曼-拉罗奇有限公司 Modulation of axon degeneration
NZ599337A (en) 2009-10-22 2013-05-31 Genentech Inc Anti-hepsin antibodies and methods using same
WO2011056497A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor type iib compositions and methods of use
WO2011056502A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Bone morphogenetic protein receptor type ii compositions and methods of use
WO2011056494A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor-like kinase-1 antagonist and vegfr3 antagonist combinations
AU2010315101B2 (en) 2009-11-04 2016-01-28 Fabrus Llc Methods for affinity maturation-based antibody optimization
RU2585488C2 (en) 2009-11-05 2016-05-27 Дженентек, Инк. Methods and composition for secretion of heterologous polypeptides
AR079217A1 (en) 2009-11-30 2012-01-04 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSIS AND TUMOR TREATMENT
US11377485B2 (en) 2009-12-02 2022-07-05 Academia Sinica Methods for modifying human antibodies by glycan engineering
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
CA2782333C (en) 2009-12-02 2019-06-04 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
TWI505836B (en) 2009-12-11 2015-11-01 Genentech Inc Anti-vegf-c antibodies and methods using same
KR101989628B1 (en) 2009-12-21 2019-06-14 제넨테크, 인크. Antibody formulation
SI2516465T1 (en) 2009-12-23 2016-08-31 F. Hoffmann-La Roche Ag Anti-bv8 antibodies and uses thereof
EP3450459B1 (en) 2009-12-28 2021-05-26 OncoTherapy Science, Inc. Anti-cdh3 antibodies and uses thereof
US20110159588A1 (en) 2009-12-30 2011-06-30 Kui Lin Methods for Modulating a PDGF-AA Mediated Biological Response
CN102933231B (en) 2010-02-10 2015-07-29 伊缪诺金公司 CD20 antibody and uses thereof
WO2011098971A1 (en) 2010-02-12 2011-08-18 Pfizer Inc. Salts and polymorphs of 8-fluoro-2-{4-[(methylamino}methyl]phenyl}-1,3,4,5-tetrahydro-6h-azepino[5,4,3-cd]indol-6-one
JP5981853B2 (en) 2010-02-18 2016-08-31 ジェネンテック, インコーポレイテッド Neuregulin antagonists and their use in the treatment of cancer
MX2012009215A (en) 2010-02-23 2012-11-23 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
AU2011221229B2 (en) 2010-02-23 2015-06-18 F. Hoffmann-La Roche Ag Anti-angiogenesis therapy for the treatment of ovarian cancer
JP6025256B2 (en) * 2010-02-23 2016-11-16 コーニング インコーポレイテッド Modified substrate for protecting peptide-immobilized surface from gamma ray degradation
US20110217309A1 (en) 2010-03-03 2011-09-08 Buck Elizabeth A Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
JP2013527748A (en) 2010-03-03 2013-07-04 オーエスアイ・ファーマシューティカルズ,エルエルシー Biological markers useful for predicting anticancer responses to insulin-like growth factor 1 receptor kinase inhibitors
US9556248B2 (en) 2010-03-19 2017-01-31 The Board Of Trustees Of The Leland Stanford Junior University Hepatocyte growth factor fragments that function as potent met receptor agonists and antagonists
NZ602040A (en) 2010-03-24 2014-12-24 Genentech Inc Anti-lrp6 antibodies
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
WO2011130332A1 (en) 2010-04-12 2011-10-20 Academia Sinica Glycan arrays for high throughput screening of viruses
WO2011133931A1 (en) 2010-04-22 2011-10-27 Genentech, Inc. Use of il-27 antagonists for treating inflammatory bowel disease
MA34291B1 (en) 2010-05-03 2013-06-01 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSING AND TREATING A TUMOR
CA2835489C (en) 2010-05-10 2018-03-06 Chi-Huey Wong Zanamivir phosphonate congeners with anti-influenza activity and determining oseltamivir susceptibility of influenza viruses
EP2571903B1 (en) 2010-05-17 2019-09-04 EMD Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
ES2637613T5 (en) 2010-05-25 2022-07-20 Hoffmann La Roche Polypeptide purification procedures
WO2011147834A1 (en) 2010-05-26 2011-12-01 Roche Glycart Ag Antibodies against cd19 and uses thereof
JP6050226B2 (en) 2010-05-28 2016-12-21 ジェネンテック, インコーポレイテッド Lowering lactic acid levels and increasing polypeptide production by downregulating LDH and PDHK expression
WO2011153243A2 (en) 2010-06-02 2011-12-08 Genentech, Inc. Anti-angiogenesis therapy for treating gastric cancer
BR112012029866A2 (en) 2010-06-03 2017-03-07 Genentech Inc method for determining the presence of a steap-1 protein
CA3220104A1 (en) 2010-06-08 2011-12-15 Genentech, Inc. Cysteine engineered antibodies and conjugates
ES2611479T3 (en) 2010-06-16 2017-05-09 University Of Pittsburgh- Of The Commonwealth System Of Higher Education Endoplasmin antibodies and their use
US20110311527A1 (en) 2010-06-16 2011-12-22 Allergan, Inc. IL23p19 ANTIBODY INHIBITOR FOR TREATING OCULAR AND OTHER CONDITIONS
RU2577986C2 (en) 2010-06-18 2016-03-20 Дженентек, Инк. Antibodies against axl and their application
WO2011161119A1 (en) 2010-06-22 2011-12-29 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2011161189A1 (en) 2010-06-24 2011-12-29 F. Hoffmann-La Roche Ag Anti-hepsin antibodies and methods of use
NZ605449A (en) 2010-07-09 2015-03-27 Genentech Inc Anti-neuropilin antibodies and methods of use
KR20130092561A (en) 2010-07-16 2013-08-20 메르크 파텐트 게엠베하 Peptide for use in the treatment of breast cancer and/or bone metastases
WO2012010582A1 (en) 2010-07-21 2012-01-26 Roche Glycart Ag Anti-cxcr5 antibodies and methods of use
WO2012012750A1 (en) 2010-07-23 2012-01-26 Trustees Of Boston University ANTI-DEsupR INHIBITORS AS THERAPEUTICS FOR INHIBITION OF PATHOLOGICAL ANGIOGENESIS AND TUMOR CELL INVASIVENESS AND FOR MOLECULAR IMAGING AND TARGETED DELIVERY
CN103153341B (en) 2010-08-03 2015-05-27 霍夫曼-拉罗奇有限公司 Chronic lymphocytic leukemia (Cll) biomarkers
CA2805564A1 (en) 2010-08-05 2012-02-09 Stefan Jenewein Anti-mhc antibody anti-viral cytokine fusion protein
JP2013540701A (en) 2010-08-12 2013-11-07 セラクローン サイエンシーズ, インコーポレイテッド Anti-hemagglutinin antibody composition and method of use thereof
EP2420250A1 (en) 2010-08-13 2012-02-22 Universitätsklinikum Münster Anti-Syndecan-4 antibodies
SG187746A1 (en) 2010-08-13 2013-03-28 Roche Glycart Ag Anti-fap antibodies and methods of use
BR112013002444A2 (en) 2010-08-13 2016-05-24 Roche Glycart Ag isolated antibody, polynucleotide and polypeptide, composition, vector, host cell, antibody conjugate, pharmaceutical formulation, use of the antibody, methods of producing an antibody, treating an individual, inducing cell lysis of a tumor cell and diagnosing a disease in an individual
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
CA2805054A1 (en) 2010-08-25 2012-03-01 F. Hoffmann-La Roche Ag Antibodies against il-18r1 and uses thereof
PT3556396T (en) 2010-08-31 2022-07-04 Scripps Research Inst Human immunodeficiency virus (hiv)-neutralizing antibodies
SG10201408229WA (en) 2010-08-31 2015-02-27 Genentech Inc Biomarkers and methods of treatment
WO2012028683A1 (en) 2010-09-02 2012-03-08 Novartis Ag Antibody gel system for sustained drug delivery
WO2012030512A1 (en) 2010-09-03 2012-03-08 Percivia Llc. Flow-through protein purification process
WO2012033953A1 (en) 2010-09-08 2012-03-15 Halozyme, Inc. Methods for assessing and identifying or evolving conditionally active therapeutic proteins
WO2012047968A2 (en) 2010-10-05 2012-04-12 Genentech, Inc. Mutant smoothened and methods of using the same
EP2630134B9 (en) 2010-10-20 2018-04-18 Pfizer Inc Pyridine-2- derivatives as smoothened receptor modulators
WO2012061129A1 (en) 2010-10-25 2012-05-10 Genentech, Inc Treatment of gastrointestinal inflammation and psoriasis a
WO2012064836A1 (en) 2010-11-10 2012-05-18 Genentech, Inc. Methods and compositions for neural disease immunotherapy
EP2643353A1 (en) 2010-11-24 2013-10-02 Novartis AG Multispecific molecules
WO2012071436A1 (en) 2010-11-24 2012-05-31 Genentech, Inc. Method of treating autoimmune inflammatory disorders using il-23r loss-of-function mutants
WO2012075333A2 (en) 2010-12-02 2012-06-07 Prometheus Laboratories Inc. Her2delta16 peptides
AU2011343570B2 (en) 2010-12-16 2016-11-03 Genentech, Inc. Diagnosis and treatments relating to TH2 inhibition
MX345519B (en) 2010-12-20 2017-02-01 Genentech Inc Anti-mesothelin antibodies and immunoconjugates.
MA34818B1 (en) 2010-12-22 2014-01-02 Genentech Inc ANTI-PCSK9 ANTIBODIES AND METHODS OF USE
CN103384681B (en) 2010-12-23 2018-05-18 霍夫曼-拉罗奇有限公司 Bonding agent
CN103384831B (en) 2010-12-23 2016-02-10 霍夫曼-拉罗奇有限公司 Polypeptide dimer is detected by bivalent binders
SG191153A1 (en) 2010-12-23 2013-07-31 Hoffmann La Roche Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
CA2822283A1 (en) 2010-12-23 2012-06-28 Nestec S.A. Drug selection for malignant cancer therapy using antibody-based arrays
EP2659269B1 (en) 2010-12-23 2016-10-26 Roche Diagniostics GmbH Detection of a posttranslationally modified polypeptide by a bi-valent binding agent
WO2012092539A2 (en) 2010-12-31 2012-07-05 Takeda Pharmaceutical Company Limited Antibodies to dll4 and uses thereof
CA2824252A1 (en) 2011-02-10 2012-08-16 Roche Glycart Ag Improved immunotherapy
PE20140303A1 (en) 2011-02-10 2014-03-22 Roche Glycart Ag MUTANT INTERLEUQUIN-2 POLYPEPTIDES
AU2012217867A1 (en) 2011-02-14 2013-09-05 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
US20120214830A1 (en) 2011-02-22 2012-08-23 OSI Pharmaceuticals, LLC Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors in hepatocellular carcinoma
ES2549638T3 (en) 2011-02-28 2015-10-30 F. Hoffmann-La Roche Ag Antigen binding proteins
CN103403025B (en) 2011-02-28 2016-10-12 弗·哈夫曼-拉罗切有限公司 Monovalent antigen binding protein
AU2012222833B2 (en) 2011-03-03 2017-03-16 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
WO2012119989A2 (en) 2011-03-04 2012-09-13 Oryzon Genomics, S.A. Methods and antibodies for the diagnosis and treatment of cancer
BR112013023050A8 (en) 2011-03-09 2018-09-25 G Pestell Richard ccr5 antagonist, method for determining whether a human individual having prostate cancer is suffering from or at risk of metastasis, to identify a ccr5 antagonist, method for determining whether a human individual having prostate cancer is suffering from or under risk of developing metastasis, to identify a candidate compound, to produce in vitro primary epithelial cells, to diagnose prostate cancer, to select a treatment for an individual having a prostate cancer / tumor, cell line, and, animal model
EP2685968A1 (en) 2011-03-15 2014-01-22 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
EP3235508B1 (en) 2011-03-16 2020-12-30 Sanofi Compositions comprising a dual v region antibody-like protein
PT2691417T (en) 2011-03-29 2018-10-31 Roche Glycart Ag Antibody fc variants
CA2831732C (en) 2011-03-31 2019-12-31 Genentech, Inc. Use of a monoclonal anti-beta7 antibody for treating gastrointestinal inflammatory disorders
JP2014516511A (en) 2011-04-07 2014-07-17 ジェネンテック, インコーポレイテッド Anti-FGFR4 antibody and method of use
US9150644B2 (en) 2011-04-12 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies that bind insulin-like growth factor (IGF) I and II
EP2699598B1 (en) 2011-04-19 2019-03-06 Pfizer Inc Combinations of anti-4-1bb antibodies and adcc-inducing antibodies for the treatment of cancer
JP2014514313A (en) 2011-04-20 2014-06-19 ロシュ グリクアート アクチェンゲゼルシャフト Methods and constructs for pH-dependent passage of the blood brain barrier
WO2012149014A1 (en) 2011-04-25 2012-11-01 OSI Pharmaceuticals, LLC Use of emt gene signatures in cancer drug discovery, diagnostics, and treatment
EA201892619A1 (en) 2011-04-29 2019-04-30 Роше Гликарт Аг IMMUNOCONJUGATES CONTAINING INTERLEUKIN-2 MUTANT POLYPETIPS
JP5987053B2 (en) 2011-05-12 2016-09-06 ジェネンテック, インコーポレイテッド Multiple reaction monitoring LC-MS / MS method for detecting therapeutic antibodies in animal samples using framework signature peptides
WO2012158704A1 (en) 2011-05-16 2012-11-22 Genentech, Inc. Fgfr1 agonists and methods of use
AR086924A1 (en) 2011-06-15 2014-01-29 Hoffmann La Roche HUMAN EPO ANTI-RECEIVER ANTIBODIES AND THE METHODS FOR USE
WO2013003680A1 (en) 2011-06-30 2013-01-03 Genentech, Inc. Anti-c-met antibody formulations
JP2013040160A (en) 2011-07-01 2013-02-28 Genentech Inc Use of anti-cd83 agonist antibody for treating autoimmune disease
LT2731677T (en) 2011-07-11 2018-07-10 Glenmark Pharmaceuticals S.A. Antibodies that bind to ox40 and their uses
ES2671748T3 (en) 2011-07-21 2018-06-08 Tolero Pharmaceuticals, Inc. Heterocyclic protein kinase inhibitors
WO2013015821A1 (en) 2011-07-22 2013-01-31 The Research Foundation Of State University Of New York Antibodies to the b12-transcobalamin receptor
US9120858B2 (en) 2011-07-22 2015-09-01 The Research Foundation Of State University Of New York Antibodies to the B12-transcobalamin receptor
US20130022551A1 (en) 2011-07-22 2013-01-24 Trustees Of Boston University DEspR ANTAGONISTS AND AGONISTS AS THERAPEUTICS
MX2014001766A (en) 2011-08-17 2014-05-01 Genentech Inc Neuregulin antibodies and uses thereof.
CN103890008A (en) 2011-08-17 2014-06-25 霍夫曼-拉罗奇有限公司 Inhibition of angiogenesis in refractory tumors
US9309306B2 (en) 2011-08-23 2016-04-12 Roche Glycart Ag Anti-MCSP antibodies
SI2748202T1 (en) 2011-08-23 2018-10-30 Roche Glycart Ag Bispecific antigen binding molecules
KR101870555B1 (en) 2011-08-23 2018-06-22 로슈 글리카트 아게 Bispecific antibodies specific for t-cell activating antigens and a tumor antigen and methods of use
US20130078250A1 (en) 2011-08-23 2013-03-28 Oliver Ast Bispecific t cell activating antigen binding molecules
CN103748114B (en) 2011-08-23 2017-07-21 罗切格利卡特公司 T cell activation bispecific antigen binding molecules
US8822651B2 (en) 2011-08-30 2014-09-02 Theraclone Sciences, Inc. Human rhinovirus (HRV) antibodies
GB201212550D0 (en) 2012-07-13 2012-08-29 Novartis Ag B cell assay
WO2013033623A1 (en) 2011-09-02 2013-03-07 Nestec S.A. Profiling of signal pathway proteins to determine therapeutic efficacy
KR20140068062A (en) 2011-09-15 2014-06-05 제넨테크, 인크. Methods of promoting differentiation
GB201116092D0 (en) 2011-09-16 2011-11-02 Bioceros B V Antibodies and uses thereof
MX2014002990A (en) 2011-09-19 2014-05-21 Genentech Inc Combination treatments comprising c-met antagonists and b-raf antagonists.
WO2013042006A1 (en) 2011-09-22 2013-03-28 Pfizer Inc. Pyrrolopyrimidine and purine derivatives
EP3275902A1 (en) 2011-10-04 2018-01-31 IGEM Therapeutics Limited Ige anti-hmw-maa antibody
US9663573B2 (en) 2011-10-05 2017-05-30 Genentech, Inc. Methods of treating liver conditions using Notch2 antagonists
KR102102862B1 (en) 2011-10-14 2020-04-22 제넨테크, 인크. ANTI-HtrA1 ANTIBODIES AND METHODS OF USE
KR20140084164A (en) 2011-10-15 2014-07-04 제넨테크, 인크. Scd1 antagonists for treating cancer
WO2013059531A1 (en) 2011-10-20 2013-04-25 Genentech, Inc. Anti-gcgr antibodies and uses thereof
WO2013063229A1 (en) 2011-10-25 2013-05-02 The Regents Of The University Of Michigan Her2 targeting agent treatment in non-her2-amplified cancers having her2 expressing cancer stem cells
AU2012328980A1 (en) 2011-10-28 2014-04-24 Genentech, Inc. Therapeutic combinations and methods of treating melanoma
JP6356607B2 (en) 2011-11-02 2018-07-11 ジェネンテック, インコーポレイテッド Overload / elution chromatography
BR112014011115A2 (en) 2011-11-08 2017-06-13 Pfizer Methods for treating inflammatory disorders using anti-csf antibodies
BR112014012005A2 (en) 2011-11-21 2017-12-19 Genentech Inc compositions, methods, pharmaceutical formulation and article
SG11201402619VA (en) 2011-11-23 2014-10-30 Igenica Biotherapeutics Inc Anti-cd98 antibodies and methods of use thereof
SG11201402711SA (en) 2011-11-29 2014-06-27 Genentech Inc Compositions and methods for prostate cancer analysis
CA2857114A1 (en) 2011-11-30 2013-06-06 Genentech, Inc. Erbb3 mutations in cancer
EP2788024A1 (en) 2011-12-06 2014-10-15 F.Hoffmann-La Roche Ag Antibody formulation
WO2013083810A1 (en) 2011-12-09 2013-06-13 F. Hoffmann-La Roche Ag Identification of non-responders to her2 inhibitors
TR201815709T4 (en) 2011-12-22 2018-11-21 Hoffmann La Roche Ion exchange membrane chromatography.
PL2794651T3 (en) 2011-12-22 2022-12-27 F.Hoffmann-La Roche Ag Expression vector element combinations, novel production cell generation methods and their use for the recombinant production of polypeptides
WO2013091903A1 (en) 2011-12-22 2013-06-27 Novo Nordisk A/S Anti-crac channel antibodies
KR102280111B1 (en) 2011-12-22 2021-07-21 에프. 호프만-라 로슈 아게 Expression vector organization, novel production cell generation methods and their use for the recombinant production of polypeptides
AR089434A1 (en) 2011-12-23 2014-08-20 Genentech Inc PROCEDURE TO PREPARE FORMULATIONS WITH HIGH CONCENTRATION OF PROTEINS
WO2013101771A2 (en) 2011-12-30 2013-07-04 Genentech, Inc. Compositions and method for treating autoimmune diseases
WO2013106485A2 (en) 2012-01-09 2013-07-18 The Scripps Research Institute Ultralong complementarity determining regions and uses thereof
WO2013106489A1 (en) 2012-01-09 2013-07-18 The Scripps Research Institute Humanized antibodies with ultralong cdr3s
JP6242813B2 (en) 2012-01-18 2017-12-06 ジェネンテック, インコーポレイテッド Anti-LRP5 antibody and method of use
CN104168920A (en) 2012-01-18 2014-11-26 霍夫曼-拉罗奇有限公司 Methods of using FGF19 modulators
WO2013116287A1 (en) 2012-01-31 2013-08-08 Genentech, Inc. Anti-ig-e m1' antibodies and methods using same
WO2013113641A1 (en) 2012-01-31 2013-08-08 Roche Glycart Ag Use of nkp46 as a predictive biomarker for cancer treatment with adcc- enhanced antibodies
KR20140127854A (en) 2012-02-10 2014-11-04 제넨테크, 인크. Single-chain antibodies and other heteromultimers
BR112014019741A2 (en) 2012-02-11 2020-12-22 Genentech, Inc USES OF AN ANTAGONIST OF THE WNT VIA, USE OF ANTI-CANCER THERAPY, METHOD OF IDENTIFICATION OF AN INDIVIDUAL WITH CANCER, METHODS FOR PREVENTING, METHOD OF INHIBITION OF A CANCER CELL PROLIFERATION, USE OF AN ANGONIST ANTAGONIST TRANSLOCATION OF ISOLATED R-SPONDINA
JP6152120B2 (en) 2012-02-15 2017-06-21 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Affinity chromatography based on Fc receptors
JP2015511702A (en) 2012-03-02 2015-04-20 ロシュ グリクアート アーゲー Predictive biomarkers for cancer treatment using ADCC-enhanced antibodies
EP3296320A1 (en) 2012-03-08 2018-03-21 Halozyme, Inc. Conditionally active anti-epidermal growth factor receptor antibodies and methods of use thereof
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
JP2015514710A (en) 2012-03-27 2015-05-21 ジェネンテック, インコーポレイテッド Diagnosis and treatment of HER3 inhibitors
AR090549A1 (en) 2012-03-30 2014-11-19 Genentech Inc ANTI-LGR5 AND IMMUNOCATE PLAYERS
WO2013152252A1 (en) 2012-04-06 2013-10-10 OSI Pharmaceuticals, LLC Combination anti-cancer therapy
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
RU2014148162A (en) 2012-05-01 2016-06-20 Дженентек, Инк. ANTI-PMEL17 ANTIBODIES AND THEIR IMMUNO CONJUGATES
US20130336973A1 (en) 2012-05-10 2013-12-19 Zymeworks Inc. Heteromultimer Constructs of Immunoglobulin Heavy Chains with Mutations in the Fc Domain
WO2013170191A1 (en) 2012-05-11 2013-11-14 Genentech, Inc. Methods of using antagonists of nad biosynthesis from nicotinamide
CA2873743C (en) 2012-05-14 2022-12-06 Prostagene, Llc Using modulators of ccr5 for treating cancer
WO2013177470A1 (en) 2012-05-23 2013-11-28 Genentech, Inc. Selection method for therapeutic agents
US20130344064A1 (en) 2012-06-08 2013-12-26 Glenmark Pharmaceuticals S.A. Anti-trka antibodies with enhanced inhibitory properties and derivatives thereof
CN104364266A (en) 2012-06-15 2015-02-18 霍夫曼-拉罗奇有限公司 Anti-PCSK9 antibodies, formulations, dosing, and methods of use
RU2015100656A (en) 2012-06-27 2016-08-20 Ф. Хоффманн-Ля Рош Аг METHOD FOR PRODUCING ANTIBODY FC-FRAGMENT CONNECTING, INCLUDING AT LEAST ONE CONNECTING GROUP, WHICH SPECIALLY RELATED TO THE TARGET, AND THEIR APPLICATION
BR112014029888A2 (en) 2012-06-27 2020-05-12 Hoffmann La Roche METHODS OF PRODUCTION OF AN ANTIBODY, DETERMINATION OF A COMBINATION OF BINDING SITES AND TREATMENT OF AN INDIVIDUAL WITH CANCER, PHARMACEUTICAL FORMULATION, ANTIBODY AND USE OF AN ANTIBODY
WO2014001324A1 (en) 2012-06-27 2014-01-03 Hoffmann-La Roche Ag Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
CA2872192A1 (en) 2012-07-04 2014-01-09 F. Hoffmann-La Roche Ag Anti-biotin antibodies and methods of use
MX353951B (en) 2012-07-04 2018-02-07 Hoffmann La Roche Anti-theophylline antibodies and methods of use.
ES2604012T3 (en) 2012-07-04 2017-03-02 F. Hoffmann-La Roche Ag Covalently bound antigen-antibody conjugates
CN110042114A (en) 2012-07-05 2019-07-23 弗·哈夫曼-拉罗切有限公司 Expression and excretory system
SG11201500087VA (en) 2012-07-09 2015-02-27 Genentech Inc Immunoconjugates comprising anti-cd22 antibodies
TW201408698A (en) 2012-07-09 2014-03-01 Genentech Inc Anti-CD79b antibodies and immunoconjugates
SG11201500096YA (en) 2012-07-09 2015-02-27 Genentech Inc Immunoconjugates comprising anti - cd79b antibodies
CA2874904A1 (en) 2012-07-09 2014-01-16 Genentech, Inc. Immunoconjugates comprising anti-cd22 antibodies
HUE056217T2 (en) 2012-07-13 2022-02-28 Roche Glycart Ag Bispecific anti-vegf/anti-ang-2 antibodies and their use in the treatment of ocular vascular diseases
WO2014012082A2 (en) 2012-07-13 2014-01-16 Zymeworks Inc. Multivalent heteromultimer scaffold design an constructs
CN112587658A (en) 2012-07-18 2021-04-02 博笛生物科技有限公司 Targeted immunotherapy for cancer
SG10201800535XA (en) 2012-08-07 2018-02-27 Roche Glycart Ag Composition comprising two antibodies engineered to have reduced and increased effector function
BR112015002085A2 (en) 2012-08-08 2017-12-19 Roche Glycart Ag protein, polynucleotide, vector, host cell, method for producing protein, pharmaceutical composition, protein use, method of treatment and invention
JP2015525792A (en) 2012-08-09 2015-09-07 ロシュ グリクアート アクチェンゲゼルシャフト ASGPR antibody and use thereof
AU2013306098A1 (en) 2012-08-18 2015-02-12 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
US9345766B2 (en) 2012-08-30 2016-05-24 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-ERBB3 agents
WO2014041072A1 (en) 2012-09-14 2014-03-20 F. Hoffmann-La Roche Ag Method for the production and selection of molecules comprising at least two different entities and uses thereof
KR101850591B1 (en) 2012-10-05 2018-04-19 제넨테크, 인크. Methods for diagnosing and treating inflammatory bowel disease
WO2014056783A1 (en) 2012-10-08 2014-04-17 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
WO2014059028A1 (en) 2012-10-09 2014-04-17 Igenica, Inc. Anti-c16orf54 antibodies and methods of use thereof
US9394257B2 (en) 2012-10-16 2016-07-19 Tolero Pharmaceuticals, Inc. PKM2 modulators and methods for their use
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
CA2884431A1 (en) 2012-11-08 2014-05-15 F. Hoffmann-La Roche Ag Her3 antigen binding proteins binding to the beta-hairpin of her3
WO2014078268A2 (en) 2012-11-13 2014-05-22 Genentech, Inc. Anti-hemagglutinin antibodies and methods of use
CN105051528A (en) 2012-11-15 2015-11-11 弗·哈夫曼-拉罗切有限公司 Ionic strength-mediated ph gradient ion exchange chromatography
MX363188B (en) 2012-11-30 2019-03-13 Hoffmann La Roche Identification of patients in need of pd-l1 inhibitor cotherapy.
US9260426B2 (en) 2012-12-14 2016-02-16 Arrien Pharmaceuticals Llc Substituted 1H-pyrrolo [2, 3-b] pyridine and 1H-pyrazolo [3, 4-b] pyridine derivatives as salt inducible kinase 2 (SIK2) inhibitors
PE20151289A1 (en) 2013-01-02 2015-10-05 Glenmark Pharmaceuticals Sa ANTIBODIES THAT JOIN THE TL1A AND ITS USES
CA3150658A1 (en) 2013-01-18 2014-07-24 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
WO2014116749A1 (en) 2013-01-23 2014-07-31 Genentech, Inc. Anti-hcv antibodies and methods of using thereof
WO2014114595A1 (en) 2013-01-23 2014-07-31 Roche Glycart Ag Predictive biomarker for cancer treatment with adcc-enhanced antibodies
JP2016509045A (en) 2013-02-22 2016-03-24 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト How to treat cancer and prevent drug resistance
RU2015140915A (en) 2013-02-26 2017-04-03 Роше Гликарт Аг BSPECIFIC ANTI-BINDING MOLECULES ACTIVATING T-CELLS
CA2896259A1 (en) 2013-02-26 2014-09-04 Roche Glycart Ag Anti-mcsp antibodies
MY192312A (en) 2013-02-26 2022-08-17 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
EP2961770A1 (en) 2013-02-26 2016-01-06 Roche Glycart AG Bispecific t cell activating antigen binding molecules
US9925240B2 (en) 2013-03-06 2018-03-27 Genentech, Inc. Methods of treating and preventing cancer drug resistance
AR095398A1 (en) 2013-03-13 2015-10-14 Genentech Inc FORMULATIONS WITH REDUCED OXIDATION
CN110075293A (en) 2013-03-13 2019-08-02 霍夫曼-拉罗奇有限公司 Aoxidize reduced preparaton
BR112015022210A8 (en) 2013-03-13 2018-01-23 Genentech Inc antibody formulations
HUE049707T2 (en) 2013-03-13 2020-11-30 Hoffmann La Roche Formulations with reduced oxidation
US10653779B2 (en) 2013-03-13 2020-05-19 Genentech, Inc. Formulations with reduced oxidation
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
CA2905070A1 (en) 2013-03-14 2014-09-25 Genentech, Inc. Methods of treating cancer and preventing cancer drug resistance
EP2968540A2 (en) 2013-03-14 2016-01-20 Genentech, Inc. Combinations of a mek inhibitor compound with an her3/egfr inhibitor compound and methods of use
US10202356B2 (en) 2013-03-14 2019-02-12 Tolero Pharmaceuticals, Inc. JAK2 and ALK2 inhibitors and methods for their use
EP2972373B1 (en) 2013-03-15 2019-10-09 F.Hoffmann-La Roche Ag Biomarkers and methods of treating pd-1 and pd-l1 related conditions
US20140283157A1 (en) 2013-03-15 2014-09-18 Diadexus, Inc. Lipoprotein-associated phospholipase a2 antibody compositions and methods of use
US9441035B2 (en) 2013-03-15 2016-09-13 Genentech, Inc. Cell culture media and methods of antibody production
JP6527132B2 (en) 2013-03-15 2019-06-05 ジェネンテック, インコーポレイテッド Compositions and methods for diagnosis and treatment of liver cancer
BR112015021521A2 (en) 2013-03-15 2017-10-10 Genentech Inc anti-crth2 antibodies and methods for their use
MX2015011899A (en) 2013-03-15 2016-05-05 Genentech Inc Methods of treating cancer and preventing cancer drug resistance.
MX2015012872A (en) 2013-03-15 2016-02-03 Ac Immune Sa Anti-tau antibodies and methods of use.
WO2014145098A1 (en) 2013-03-15 2014-09-18 Genentech, Inc. Cell culture compositions with antioxidants and methods for polypeptide production
RS57840B1 (en) 2013-03-18 2018-12-31 Biocerox Prod Bv Humanized anti-cd134 (ox40) antibodies and uses thereof
RU2015145610A (en) 2013-03-27 2017-05-04 Дженентек, Инк. APPLICATION OF BIOMARKERS FOR THE EVALUATION OF TREATMENT OF GASTROINTESTINAL INFLAMMATORY DISORDERS BY BETA7 INTEGRIN ANTAGONISTS
WO2014169076A1 (en) 2013-04-09 2014-10-16 Annexon,,Inc. Methods of treatment for neuromyelitis optica
US9206188B2 (en) 2013-04-18 2015-12-08 Arrien Pharmaceuticals Llc Substituted pyrrolo[2,3-b]pyridines as ITK and JAK inhibitors
EP2994164B1 (en) 2013-05-08 2020-08-05 Zymeworks Inc. Bispecific her2 and her3 antigen binding constructs
EP2999716A2 (en) 2013-05-20 2016-03-30 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
CN105392780A (en) 2013-05-24 2016-03-09 雀巢产品技术援助有限公司 Pathway specific assays for predicting irritable bowel syndrome diagnosis
US10086054B2 (en) 2013-06-26 2018-10-02 Academia Sinica RM2 antigens and use thereof
EP3013347B1 (en) 2013-06-27 2019-12-11 Academia Sinica Glycan conjugates and use thereof
JP6462680B2 (en) 2013-07-09 2019-01-30 アネクソン,インコーポレーテッド Anti-complement factor C1q antibody and use thereof
WO2015006686A1 (en) 2013-07-12 2015-01-15 Genentech, Inc. Elucidation of ion exchange chromatography input optimization
AU2014290044B2 (en) * 2013-07-17 2020-10-29 Foundation Medicine, Inc. Methods of treating urothelial carcinomas
CN105814074B (en) 2013-07-18 2020-04-21 图鲁斯生物科学有限责任公司 Humanized antibodies with ultralong complementarity determining regions
EP3022224A2 (en) 2013-07-18 2016-05-25 Fabrus, Inc. Antibodies with ultralong complementarity determining regions
US10093978B2 (en) 2013-08-12 2018-10-09 Genentech, Inc. Compositions for detecting complement factor H (CFH) and complement factor I (CFI) polymorphisms
WO2015035180A1 (en) 2013-09-05 2015-03-12 Genentech, Inc. Method for chromatography reuse
CN105682666B (en) 2013-09-06 2021-06-01 中央研究院 Activation of human iNKT cells using glycolipids
MX2016003256A (en) 2013-09-12 2016-06-07 Halozyme Inc Modified anti-epidermal growth factor receptor antibodies and methods of use thereof.
RU2016107435A (en) 2013-09-13 2017-10-18 Дженентек, Инк. COMPOSITIONS AND METHODS FOR DETECTING AND QUANTITATIVE DETERMINATION OF THE PROTEIN OF CELLS-OWNERS IN CELL LINES AND RECOMBINANT POLYPEPTIDE PRODUCTS
NZ756749A (en) 2013-09-13 2022-05-27 Genentech Inc Methods and compositions comprising purified recombinant polypeptides
CN105518027A (en) 2013-09-17 2016-04-20 豪夫迈·罗氏有限公司 Methods of using anti-LGR5 antibodies
EP3049437A1 (en) 2013-09-27 2016-08-03 F. Hoffmann-La Roche AG Thermus thermophilus slyd fkbp domain specific antibodies
SG11201602283UA (en) 2013-09-27 2016-04-28 Genentech Inc Anti-pdl1 antibody formulations
DK3052102T3 (en) 2013-10-04 2020-03-09 Aptose Biosciences Inc CANCER TREATMENT COMPOSITIONS
MX2016003593A (en) 2013-10-11 2016-06-02 Hoffmann La Roche Multispecific domain exchanged common variable light chain antibodies.
KR20160068855A (en) 2013-10-11 2016-06-15 제넨테크, 인크. Nsp4 inhibitors and methods of use
MX2016004802A (en) 2013-10-18 2016-07-18 Genentech Inc Anti-rsp02 and/or anti-rsp03 antibodies and their uses.
RU2016119425A (en) 2013-10-23 2017-11-28 Дженентек, Инк. METHODS FOR DIAGNOSIS AND TREATMENT OF EOSINOPHILIC DISEASES
WO2015073721A1 (en) 2013-11-13 2015-05-21 Zymeworks Inc. Monovalent antigen binding constructs targeting egfr and/or her2 and uses thereof
AU2014351996B2 (en) 2013-11-21 2020-01-02 F. Hoffmann-La Roche Ag Anti-alpha-synuclein antibodies and methods of use
UA115388C2 (en) 2013-11-21 2017-10-25 Пфайзер Інк. 2,6-substituted purine derivatives and their use in the treatment of proliferative disorders
MX2016006572A (en) 2013-11-27 2016-12-09 Zymeworks Inc Bispecific antigen-binding constructs targeting her2.
CN113861293A (en) 2013-12-09 2021-12-31 爱乐科斯公司 anti-Siglec-8 antibodies and methods of use thereof
MA39095A1 (en) 2013-12-13 2018-08-31 Genentech Inc Anti-cd33 antibodies and immunoconjugates
DK3083689T3 (en) 2013-12-17 2020-08-03 Genentech Inc Anti-CD3 antibodies and methods of use
CA2934028A1 (en) 2013-12-17 2015-06-25 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
WO2015095410A1 (en) 2013-12-17 2015-06-25 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and an anti-cd20 antibody
EP3083692B1 (en) 2013-12-17 2020-02-19 F.Hoffmann-La Roche Ag Methods of treating her2-positive cancers using pd-1 axis binding antagonists and anti-her2 antibodies
PL3083680T3 (en) 2013-12-20 2020-06-29 F. Hoffmann-La Roche Ag Humanized anti-tau(ps422) antibodies and methods of use
TWI728373B (en) 2013-12-23 2021-05-21 美商建南德克公司 Antibodies and methods of use
BR112016012666A2 (en) 2014-01-03 2017-09-26 Hoffmann La Roche conjugate, antibodies, pharmaceutical formulation and uses of conjugate
EP3089996B1 (en) 2014-01-03 2021-07-28 F. Hoffmann-La Roche AG Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
JP6521464B2 (en) 2014-01-03 2019-05-29 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Covalently linked polypeptide toxin-antibody conjugates
EP3092251B1 (en) 2014-01-06 2021-01-20 F. Hoffmann-La Roche AG Monovalent blood brain barrier shuttle modules
US10548985B2 (en) 2014-01-10 2020-02-04 Birdie Biopharmaceuticals, Inc. Compounds and compositions for treating EGFR expressing tumors
US9982041B2 (en) 2014-01-16 2018-05-29 Academia Sinica Compositions and methods for treatment and detection of cancers
US10150818B2 (en) 2014-01-16 2018-12-11 Academia Sinica Compositions and methods for treatment and detection of cancers
WO2016114819A1 (en) 2015-01-16 2016-07-21 Academia Sinica Compositions and methods for treatment and detection of cancers
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
WO2015112909A1 (en) 2014-01-24 2015-07-30 Genentech, Inc. Methods of using anti-steap1 antibodies and immunoconjugates
BR112016017248A8 (en) 2014-01-24 2018-04-17 Ngm Biopharmaceuticals Inc antibody or fragment thereof, binding agent, transgenic animal, hybridoma, vector, pharmaceutical composition, fgf19 and / or fgf21-like signaling induction method, method for activating a klotho beta / fgf receptor complex, method for improving metabolism of glucose in an individual, method of detecting klotho beta, use of antibody or fragment thereof, use of pharmaceutical composition, treatment method and method for improving metabolic parameters
WO2015116902A1 (en) 2014-01-31 2015-08-06 Genentech, Inc. G-protein coupled receptors in hedgehog signaling
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
WO2015120075A2 (en) 2014-02-04 2015-08-13 Genentech, Inc. Mutant smoothened and methods of using the same
EP3718563A1 (en) 2014-02-08 2020-10-07 F. Hoffmann-La Roche AG Methods of treating alzheimer's disease
TW202239429A (en) 2014-02-08 2022-10-16 美商建南德克公司 Methods of treating alzheimer’s disease
AU2015217271B2 (en) 2014-02-12 2018-10-25 Genentech, Inc. Anti-Jagged1 antibodies and methods of use
BR112016018980A2 (en) 2014-02-21 2017-10-10 Genentech Inc method of treating a disorder, multispecific antibody, isolated nucleic acid, host cell, methods of producing an antibody, producing an antibody half or multispecific antibody, and producing a multispecific, immunoconjugate antibody and pharmaceutical formulation
JP6825909B2 (en) 2014-02-28 2021-02-03 アラコス インコーポレイテッド Methods and Compositions for Treating SIGLEC-8 Related Diseases
CN106103484B (en) 2014-03-14 2021-08-20 诺华股份有限公司 Antibody molecules against LAG-3 and uses thereof
MA39746A (en) 2014-03-14 2021-04-28 Hoffmann La Roche HETEROLOGICAL POLYPEPTIDE SECRETION COMPOSITIONS AND ASSOCIATED PROCESSES
EP3593812A3 (en) 2014-03-15 2020-05-27 Novartis AG Treatment of cancer using chimeric antigen receptor
WO2015140591A1 (en) 2014-03-21 2015-09-24 Nordlandssykehuset Hf Anti-cd14 antibodies and uses thereof
BR112016021383A2 (en) 2014-03-24 2017-10-03 Genentech Inc METHOD TO IDENTIFY A PATIENT WITH CANCER WHO IS LIKE OR LESS LIKELY TO RESPOND TO TREATMENT WITH A CMET ANTAGONIST, METHOD TO IDENTIFY A PATIENT WITH PREVIOUSLY TREATED CANCER, METHOD TO DETERMINE THE EXPRESSION OF THE HGF BIOMARKER, ANTI-C-MET ANTAGONIST AND ITS USE, DIAGNOSTIC KIT AND ITS PREPARATION METHOD
TWI687428B (en) 2014-03-27 2020-03-11 中央研究院 Reactive labelling compounds and uses thereof
MX2016012282A (en) 2014-03-27 2017-01-06 Genentech Inc Methods for diagnosing and treating inflammatory bowel disease.
WO2015153514A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
EP3632934A1 (en) 2014-03-31 2020-04-08 F. Hoffmann-La Roche AG Anti-ox40 antibodies and methods of use
WO2015155624A1 (en) 2014-04-10 2015-10-15 Pfizer Inc. Dihydropyrrolopyrimidine derivatives
ES2845650T3 (en) 2014-04-18 2021-07-27 Acceleron Pharma Inc Procedures to increase red blood cell levels and treat sickle cell disease
WO2015164615A1 (en) 2014-04-24 2015-10-29 University Of Oslo Anti-gluten antibodies and uses thereof
WO2015164364A2 (en) 2014-04-25 2015-10-29 The Brigham And Women's Hospital, Inc. Methods to manipulate alpha-fetoprotein (afp)
PT3137460T (en) 2014-04-30 2019-12-30 Pfizer Cycloalkyl-linked diheterocycle derivatives
BR112016025312A2 (en) 2014-05-01 2017-10-17 Genentech Inc antibody variants, anti-d-factor antibody, pharmaceutical formulation, dispensing device, use of the formulation and a composition, composition and method of treating a disorder
RU2744836C2 (en) 2014-05-08 2021-03-16 Новодиакс, Инк. Direct immunohistochemical analysis
MX2016015162A (en) 2014-05-22 2017-03-03 Genentech Inc Anti-gpc3 antibodies and immunoconjugates.
CN106661622B (en) 2014-05-23 2020-08-21 豪夫迈·罗氏有限公司 MIT biomarkers and methods of using the same
CN106573971A (en) 2014-05-27 2017-04-19 中央研究院 Anti-CD20 glycoantibodies and uses thereof
EP3149161B1 (en) 2014-05-27 2021-07-28 Academia Sinica Fucosidase from bacteroides and methods using the same
US10118969B2 (en) 2014-05-27 2018-11-06 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
AU2015267045B2 (en) 2014-05-27 2021-02-25 Academia Sinica Anti-HER2 glycoantibodies and uses thereof
TWI732738B (en) 2014-05-28 2021-07-11 中央研究院 Anti-tnf-alpha glycoantibodies and uses thereof
JP2017526618A (en) 2014-06-11 2017-09-14 ジェネンテック, インコーポレイテッド Anti-LgR5 antibody and use thereof
JP2017517552A (en) 2014-06-13 2017-06-29 ジェネンテック, インコーポレイテッド Treatment and prevention of anticancer drug resistance
BR122023023170A2 (en) 2014-06-13 2024-02-20 Acceleron Pharma Inc. USE OF AN ACTRII ANTAGONIST IN THE TREATMENT OR PREVENTION OF SKIN ULCERS ASSOCIATED WITH BETA-THALASSEMIA
AR100978A1 (en) 2014-06-26 2016-11-16 Hoffmann La Roche ANTI-Tau HUMANIZED ANTIBODY BRAIN LAUNCHERS (pS422) AND USES OF THE SAME
KR20170026362A (en) 2014-06-26 2017-03-08 에프. 호프만-라 로슈 아게 Anti-brdu antibodies and methods of use
WO2016001789A1 (en) 2014-06-30 2016-01-07 Pfizer Inc. Pyrimidine derivatives as pi3k inhibitors for use in the treatment of cancer
EP3164492B1 (en) 2014-07-03 2019-10-23 F. Hoffmann-La Roche AG Polypeptide expression systems
JP6789124B2 (en) 2014-07-03 2020-11-25 イエール ユニバーシティ Inhibition of Dicopf2 (Dkk2) that suppresses tumorigenesis
CN105233291A (en) 2014-07-09 2016-01-13 博笛生物科技有限公司 Combined therapy composition and combined therapy method for treating cancers
DK3166976T3 (en) 2014-07-09 2022-04-11 Birdie Biopharmaceuticals Inc ANTI-PD-L1 COMBINATIONS FOR TREATMENT OF TUMORS
EP3166974A1 (en) 2014-07-11 2017-05-17 Genentech, Inc. Anti-pd-l1 antibodies and diagnostic uses thereof
EP3166627A1 (en) 2014-07-11 2017-05-17 Genentech, Inc. Notch pathway inhibition
CN106687483B (en) 2014-07-21 2020-12-04 诺华股份有限公司 Treatment of cancer using humanized anti-BCMA chimeric antigen receptors
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
JP2017528433A (en) 2014-07-21 2017-09-28 ノバルティス アーゲー Low immunoenhancing dose of mTOR inhibitor and CAR combination
CN112481283A (en) 2014-07-21 2021-03-12 诺华股份有限公司 Treatment of cancer using CD33 chimeric antigen receptor
US20170209492A1 (en) 2014-07-31 2017-07-27 Novartis Ag Subset-optimized chimeric antigen receptor-containing t-cells
PE20170263A1 (en) 2014-08-04 2017-03-30 Hoffmann La Roche T-CELL ACTIVATING ANTIGEN-BINDING BI-SPECIFIC MOLECULES
JP6669749B2 (en) 2014-08-08 2020-03-18 アレクトル エルエルシー Anti-TREM2 antibody and method of using the same
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
MY189028A (en) 2014-08-19 2022-01-20 Novartis Ag Anti-cd123 chimeric antigen receptor (car) for use in cancer treatment
AU2015308818B2 (en) 2014-08-28 2021-02-25 Bioatla Llc Conditionally active chimeric antigen receptors for modified T-cells
EP3186281B1 (en) 2014-08-28 2019-04-10 Halozyme, Inc. Combination therapy with a hyaluronan-degrading enzyme and an immune checkpoint inhibitor
CN112587672A (en) 2014-09-01 2021-04-02 博笛生物科技有限公司 anti-PD-L1 conjugates for the treatment of tumors
WO2016040369A2 (en) 2014-09-08 2016-03-17 Academia Sinica HUMAN iNKT CELL ACTIVATION USING GLYCOLIPIDS
EA201790545A1 (en) 2014-09-12 2017-07-31 Дженентек, Инк. ANTIBODIES AND IMMUNOCONJUGATES AGAINST HER2
US9751946B2 (en) 2014-09-12 2017-09-05 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
EP3191518B1 (en) 2014-09-12 2020-01-15 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
CA2957354A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
BR112017004393A2 (en) 2014-09-15 2018-02-27 Genentech Inc antibody formulations
JP6730261B2 (en) 2014-09-17 2020-07-29 ジェネンテック, インコーポレイテッド Immune complex containing anti-HER2 antibody
DK3194443T3 (en) 2014-09-17 2021-09-27 Novartis Ag TARGETING OF CYTOTOXIC CELLS WITH CHIMARY RECEPTORS IN CONNECTION WITH ADOPTIVE IMMUNTERAPHY
PL3262071T3 (en) 2014-09-23 2020-08-10 F. Hoffmann-La Roche Ag Method of using anti-cd79b immunoconjugates
CU20170052A7 (en) 2014-10-14 2017-11-07 Dana Farber Cancer Inst Inc ANTIBODY MOLECULES THAT JOIN PD-L1
WO2016061389A2 (en) 2014-10-16 2016-04-21 Genentech, Inc. Anti-alpha-synuclein antibodies and methods of use
CA2966523A1 (en) 2014-11-03 2016-05-12 Genentech, Inc. Assays for detecting t cell immune subsets and methods of use thereof
CN114381521A (en) 2014-11-03 2022-04-22 豪夫迈·罗氏有限公司 Methods and biomarkers for efficacy prediction and assessment of OX40 agonist treatment
RU2020141422A (en) 2014-11-05 2021-01-13 Дженентек, Инк. METHODS FOR OBTAINING TWO-STRAIN PROTEINS IN BACTERIA
KR102626877B1 (en) 2014-11-05 2024-01-19 애넥슨, 인코포레이티드 Humanized Anti-Complement Factor C1Q Antibodies and Uses Thereof
CA2961439A1 (en) 2014-11-05 2016-05-12 Genentech, Inc. Anti-fgfr2/3 antibodies and methods using same
CA2966558C (en) 2014-11-05 2024-03-12 Genentech, Inc. Methods of producing two chain proteins in bacteria
WO2016073157A1 (en) 2014-11-06 2016-05-12 Genentech, Inc. Anti-ang2 antibodies and methods of use thereof
RU2017119428A (en) 2014-11-06 2018-12-06 Дженентек, Инк. COMBINED THERAPY, INCLUDING THE USE OF OX40-CONNECTING AGONISTS AND TIGIT INHIBITORS
CR20170240A (en) 2014-11-10 2018-04-03 Genentech Inc ANTI-INTERLEUCINA-33 ANTIBODIES AND THEIR USES
EP3552488A1 (en) 2014-11-10 2019-10-16 F. Hoffmann-La Roche AG Animal model for nephropathy and agents for treating the same
EP3224275B1 (en) 2014-11-14 2020-03-04 F.Hoffmann-La Roche Ag Antigen binding molecules comprising a tnf family ligand trimer
WO2016081384A1 (en) 2014-11-17 2016-05-26 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
US10508151B2 (en) 2014-11-19 2019-12-17 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
US11008403B2 (en) 2014-11-19 2021-05-18 Genentech, Inc. Anti-transferrin receptor / anti-BACE1 multispecific antibodies and methods of use
WO2016081639A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Antibodies against bace1 and use thereof for neural disease immunotherapy
AU2015348940A1 (en) 2014-11-19 2017-05-25 Société des Produits Nestlé S.A. Antibodies against serotonin, tryptophan and kynurenine metabolites and uses thereof
PL3221357T3 (en) 2014-11-20 2020-11-02 F. Hoffmann-La Roche Ag Common light chains and methods of use
BR112017010324A2 (en) 2014-11-20 2018-05-15 F. Hoffmann-La Roche Ag method for treating or slowing cancer progression in an individual, molecules, methods for enhancing immune function in an individual and for selecting a patient for treatment, kits, pharmaceutical composition and uses of a combination of one molecule
EP3227341A1 (en) 2014-12-02 2017-10-11 CeMM - Forschungszentrum für Molekulare Medizin GmbH Anti-mutant calreticulin antibodies and their use in the diagnosis and therapy of myeloid malignancies
MA41119A (en) 2014-12-03 2017-10-10 Acceleron Pharma Inc METHODS OF TREATMENT OF MYELODYSPLASIC SYNDROMES AND SIDEROBLASTIC ANEMIA
EP3227332B1 (en) 2014-12-03 2019-11-06 F.Hoffmann-La Roche Ag Multispecific antibodies
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
US9975949B2 (en) 2014-12-05 2018-05-22 Genentech, Inc. Anti-CD79b antibodies and methods of use
RU2017120039A (en) 2014-12-10 2019-01-10 Дженентек, Инк. ANTIBODIES TO HEMATOENCEPHALIC BARRIER RECEPTORS AND METHODS OF APPLICATION
US20160168237A1 (en) 2014-12-12 2016-06-16 Alexion Pharmaceuticals, Inc. Method for treating a complement mediated disorder caused by an infectious agent in a patient
ES2746839T3 (en) 2014-12-18 2020-03-09 Pfizer Pyrimidine and triazine derivatives and their use as AXL inhibitors
EP3034130B1 (en) 2014-12-19 2019-12-04 Althia Health, S.L. Monoclonal antibody for the diagnosis, treatment and/or prevention of brain tumors and brain lesions
RU2746356C2 (en) 2014-12-19 2021-04-12 Чугаи Сейяку Кабусики Кайся C5 antibodies and their application methods
CA2973978A1 (en) 2015-01-14 2016-07-21 The Brigham And Women's Hospital, Inc. Treatment of cancer with anti-lap monoclonal antibodies
US9975965B2 (en) 2015-01-16 2018-05-22 Academia Sinica Compositions and methods for treatment and detection of cancers
US10495645B2 (en) 2015-01-16 2019-12-03 Academia Sinica Cancer markers and methods of use thereof
WO2016117346A1 (en) 2015-01-22 2016-07-28 Chugai Seiyaku Kabushiki Kaisha A combination of two or more anti-c5 antibodies and methods of use
EP3248005B1 (en) 2015-01-24 2020-12-09 Academia Sinica Novel glycan conjugates and methods of use thereof
WO2016118961A1 (en) 2015-01-24 2016-07-28 Academia Sinica Cancer markers and methods of use thereof
CN107407677B (en) 2015-01-28 2020-07-17 豪夫迈·罗氏有限公司 Gene expression markers and treatment of multiple sclerosis
AU2016211176B2 (en) 2015-01-30 2021-01-28 Academia Sinica; Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
US10330683B2 (en) 2015-02-04 2019-06-25 Genentech, Inc. Mutant smoothened and methods of using the same
KR20170110129A (en) 2015-02-05 2017-10-10 추가이 세이야쿠 가부시키가이샤 Antibodies comprising ionic concentration dependent antigen binding domains, Fc region variants, antibodies that bind to IL-8, and their use
AU2016223584B2 (en) 2015-02-23 2020-11-19 Enrico GIRAUDO Non-natural Semaphorins 3 and their medical use
AU2016222683A1 (en) 2015-02-26 2017-07-13 Genentech, Inc. Integrin beta7 antagonists and methods of treating crohn's disease
EP3636749B1 (en) 2015-03-06 2022-04-27 F. Hoffmann-La Roche AG Ultrapurified dsbc and methods of making and using the same
MX2017011486A (en) 2015-03-16 2018-06-15 Genentech Inc Methods of detecting and quantifying il-13 and uses in diagnosing and treating th2-associated diseases.
WO2016145536A1 (en) 2015-03-18 2016-09-22 Immunobiochem Corporation Conjugates for the treatment of cancer targeted at intracellular tumor-associated antigens
WO2016146833A1 (en) 2015-03-19 2016-09-22 F. Hoffmann-La Roche Ag Biomarkers for nad(+)-diphthamide adp ribosyltransferase resistance
AU2016246708B2 (en) 2015-04-06 2020-12-24 Acceleron Pharma Inc. ALK7:actRIIB heteromultimers and uses thereof
MA41919A (en) 2015-04-06 2018-02-13 Acceleron Pharma Inc ALK4 HETEROMULTIMERS: ACTRIIB AND THEIR USES
EP3280441B1 (en) 2015-04-07 2021-09-29 Alector LLC Anti-sortilin antibodies and methods of use thereof
CN107709364A (en) 2015-04-07 2018-02-16 豪夫迈·罗氏有限公司 Antigen binding complex and application method with agonist activity
SI3280729T1 (en) 2015-04-08 2022-09-30 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
MX2017013383A (en) 2015-04-20 2017-12-07 Tolero Pharmaceuticals Inc Predicting response to alvocidib by mitochondrial profiling.
CN108064343B (en) 2015-04-21 2021-07-09 基因泰克公司 Compositions and methods for prostate cancer analysis
US20180298068A1 (en) 2015-04-23 2018-10-18 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
JP7044553B2 (en) 2015-04-24 2022-03-30 ジェネンテック, インコーポレイテッド How to identify bacteria containing bound polypeptides
US10011629B2 (en) 2015-05-01 2018-07-03 Cocrystal Pharma, Inc. Nucleoside analogs for treatment of the flaviviridae family of viruses and cancer
JP2018520642A (en) 2015-05-01 2018-08-02 ジェネンテック, インコーポレイテッド Mask anti-CD3 antibody and method of use thereof
EP4238994A3 (en) 2015-05-11 2024-02-07 F. Hoffmann-La Roche AG Compositions and methods of treating lupus nephritis
ES2835866T3 (en) 2015-05-12 2021-06-23 Hoffmann La Roche Therapeutic and diagnostic procedures for cancer
KR102608921B1 (en) 2015-05-18 2023-12-01 스미토모 파마 온콜로지, 인크. Albocidip prodrug with increased bioavailability
WO2016191750A1 (en) 2015-05-28 2016-12-01 Genentech, Inc. Cell-based assay for detecting anti-cd3 homodimers
EP3302563A1 (en) 2015-05-29 2018-04-11 H. Hoffnabb-La Roche Ag Humanized anti-ebola virus glycoprotein antibodies and methods of use
KR20180012753A (en) 2015-05-29 2018-02-06 제넨테크, 인크. Treatment and Diagnosis Methods for Cancer
WO2016196381A1 (en) 2015-05-29 2016-12-08 Genentech, Inc. Pd-l1 promoter methylation in cancer
CN107613980A (en) 2015-05-31 2018-01-19 源生公司 combination composition for immunotherapy
CN107810012A (en) 2015-06-02 2018-03-16 豪夫迈·罗氏有限公司 Use the composition and method of the anti-Antybody therapy sacred diseases of IL 34
CN107849124B (en) 2015-06-05 2021-09-24 基因泰克公司 anti-TAU antibodies and methods of use
CA2988420A1 (en) 2015-06-08 2016-12-15 Genentech, Inc. Methods of treating cancer using anti-ox40 antibodies and pd-1 axis binding antagonists
EP3303399A1 (en) 2015-06-08 2018-04-11 H. Hoffnabb-La Roche Ag Methods of treating cancer using anti-ox40 antibodies
EP3307779A2 (en) 2015-06-12 2018-04-18 Alector LLC Anti-cd33 antibodies and methods of use thereof
JP7376977B2 (en) 2015-06-12 2023-11-09 アレクトル エルエルシー Anti-CD33 antibody and method of use thereof
JP2018524295A (en) 2015-06-15 2018-08-30 ジェネンテック, インコーポレイテッド Antibodies and immune complexes
CN107847568B (en) 2015-06-16 2022-12-20 豪夫迈·罗氏有限公司 anti-CLL-1 antibodies and methods of use
WO2016204966A1 (en) 2015-06-16 2016-12-22 Genentech, Inc. Anti-cd3 antibodies and methods of use
TWI731861B (en) 2015-06-16 2021-07-01 美商建南德克公司 HUMANIZED AND AFFINITY MATURED ANTIBODIES TO FcRH5 AND METHODS OF USE
JP6846362B2 (en) 2015-06-17 2021-03-24 アラコス インコーポレイテッド Methods and Compositions for Treating Fibrous Diseases
JP2018524312A (en) 2015-06-17 2018-08-30 ジェネンテック, インコーポレイテッド Anti-HER2 antibody and method of use
MX2017016353A (en) 2015-06-17 2018-05-02 Genentech Inc Methods of treating locally advanced or metastatic breast cancers using pd-1 axis binding antagonists and taxanes.
EP3313877B1 (en) 2015-06-24 2020-06-03 H. Hoffnabb-La Roche Ag Humanized anti-tau(ps422) antibodies and methods of use
ES2898065T3 (en) 2015-06-29 2022-03-03 Ventana Med Syst Inc Materials and Procedures for Performing Histochemical Assays for Human Proepiregulin and Amphiregulin
JP2018520153A (en) 2015-06-29 2018-07-26 ジェネンテック, インコーポレイテッド Type II anti-CD20 antibody for use in organ transplantation
WO2017009751A1 (en) 2015-07-15 2017-01-19 Pfizer Inc. Pyrimidine derivatives
EP3316902A1 (en) 2015-07-29 2018-05-09 Novartis AG Combination therapies comprising antibody molecules to tim-3
SI3317301T1 (en) 2015-07-29 2021-10-29 Novartis Ag Combination therapies comprising antibody molecules to lag-3
MX2018001289A (en) 2015-08-03 2018-04-30 Tolero Pharmaceuticals Inc Combination therapies for treatment of cancer.
TWI797060B (en) 2015-08-04 2023-04-01 美商再生元醫藥公司 Taurine supplemented cell culture medium and methods of use
WO2017024171A1 (en) 2015-08-04 2017-02-09 Acceleron Pharma Inc. Methods for treating myeloproliferative disorders
NZ739721A (en) 2015-08-07 2019-09-27 Imaginab Inc Antigen binding constructs to target molecules
CN105384825B (en) 2015-08-11 2018-06-01 南京传奇生物科技有限公司 A kind of bispecific chimeric antigen receptor and its application based on single domain antibody
CA2996059A1 (en) 2015-08-28 2017-03-09 Alector Llc Anti-siglec-7 antibodies and methods of use thereof
EP3341415B1 (en) 2015-08-28 2021-03-24 H. Hoffnabb-La Roche Ag Anti-hypusine antibodies and uses thereof
WO2017041027A1 (en) 2015-09-04 2017-03-09 Obi Pharma, Inc. Glycan arrays and method of use
JP2018528218A (en) 2015-09-11 2018-09-27 アンドリアン、ブルース Recombinant glycosylated eculizumab and eculizumab variants
CR20180217A (en) 2015-09-18 2018-05-03 Chugai Pharmaceutical Co Ltd ANTIBODIES THAT JOIN INTERLEUCINE 8 (IL-8) AND ITS USES
CA2999369C (en) 2015-09-22 2023-11-07 Spring Bioscience Corporation Anti-ox40 antibodies and diagnostic uses thereof
WO2017053807A2 (en) 2015-09-23 2017-03-30 Genentech, Inc. Optimized variants of anti-vegf antibodies
US10954300B2 (en) 2015-09-28 2021-03-23 The Trustees Of Columbia University In The City Of New York Use of pentoxifylline with immune checkpoint-blockade therapies for the treatment of melanoma
EA201890790A1 (en) 2015-09-29 2018-10-31 Селджин Корпорейшн CONNECTING PD-1 PROTEINS AND METHODS OF THEIR APPLICATION
AR106188A1 (en) 2015-10-01 2017-12-20 Hoffmann La Roche ANTI-CD19 HUMANIZED HUMAN ANTIBODIES AND METHODS OF USE
WO2017055388A2 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific t cell activating antigen binding molecules
BR112018002570A2 (en) 2015-10-02 2018-10-16 Hoffmann La Roche bispecific antigen binding molecule, bispecific antibody, polynucleotides, ox40-specific binding antibody, pharmaceutical composition and method for inhibiting tumor cell growth in an individual
WO2017055393A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-cd3xtim-3 bispecific t cell activating antigen binding molecules
JP6937746B2 (en) 2015-10-02 2021-09-22 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Bispecific anti-CD19 × CD3T cell-activating antigen-binding molecule
WO2017055395A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-cd3xrob04 bispecific t cell activating antigen binding molecules
US20170096485A1 (en) 2015-10-02 2017-04-06 Hoffmann-La Roche Inc. Bispecific t cell activating antigen binding molecules
WO2017055385A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-cd3xgd2 bispecific t cell activating antigen binding molecules
AU2016329251B2 (en) 2015-10-02 2023-02-02 F. Hoffmann-La Roche Ag Anti-PD1 antibodies and methods of use
WO2017055392A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-cd3xcd44v6 bispecific t cell activating antigen binding molecules
CN107849137B (en) 2015-10-02 2021-11-26 豪夫迈·罗氏有限公司 Bispecific anti-CEAXCD 3T cell activating antigen binding molecules
MA43345A (en) 2015-10-02 2018-08-08 Hoffmann La Roche PYRROLOBENZODIAZEPINE ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
KR102146319B1 (en) 2015-10-02 2020-08-25 에프. 호프만-라 로슈 아게 Bispecific antibodies specific for PD1 and TIM3
IL293708A (en) 2015-10-06 2022-08-01 Genentech Inc Method for treating multiple sclerosis
CN117069841A (en) 2015-10-06 2023-11-17 艾利妥 anti-TREM 2 antibodies and methods of use thereof
AU2016334623A1 (en) 2015-10-07 2018-02-15 F. Hoffmann-La Roche Ag Bispecific antibodies with tetravalency for a costimulatory TNF receptor
WO2017062649A1 (en) 2015-10-07 2017-04-13 Alexion Pharmaceuticals, Inc. A method for treating age-related macular degeneration in a patient
MA43354A (en) 2015-10-16 2018-08-22 Genentech Inc CONJUGATE DRUG CONJUGATES WITH CLOUDY DISULPHIDE
MA45326A (en) 2015-10-20 2018-08-29 Genentech Inc CALICHEAMICIN-ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
US10604577B2 (en) 2015-10-22 2020-03-31 Allakos Inc. Methods and compositions for treating systemic mastocytosis
CA3002903A1 (en) 2015-10-23 2017-04-27 Fred Hutchinson Cancer Research Center Methods to create chemically-induced dimerizing protein systems for regulation of cellular events
US11267875B2 (en) 2015-10-28 2022-03-08 Yale University Humanized anti-DKK2 antibody and uses thereof
EP3184547A1 (en) 2015-10-29 2017-06-28 F. Hoffmann-La Roche AG Anti-tpbg antibodies and methods of use
JP7060502B2 (en) 2015-10-29 2022-04-26 アレクトル エルエルシー Anti-Sigma-9 antibody and its usage
CN114891102A (en) 2015-10-29 2022-08-12 豪夫迈·罗氏有限公司 Anti-variant Fc region antibodies and methods of use
HUE054093T2 (en) 2015-10-30 2021-08-30 Hoffmann La Roche Anti-htra1 antibodies and methods of use thereof
EP3368090A1 (en) 2015-10-30 2018-09-05 H. Hoffnabb-La Roche Ag Anti-factor d antibody variant conjugates and uses thereof
EP3368074A2 (en) 2015-10-30 2018-09-05 Hoffmann-La Roche AG Anti-factor d antibodies and conjugates
JP6998869B2 (en) 2015-11-08 2022-02-04 ジェネンテック, インコーポレイテッド Screening method for multispecific antibody
CN106729743B (en) 2015-11-23 2021-09-21 四川科伦博泰生物医药股份有限公司 anti-ErbB 2 antibody-drug conjugate, and composition, preparation method and application thereof
KR20180096645A (en) 2015-11-23 2018-08-29 악셀레론 파마 인코포레이티드 How to treat eye diseases
JP6877429B2 (en) 2015-12-03 2021-05-26 アジオス ファーマシューティカルズ, インコーポレイテッド MAT2A inhibitor for treating MTAP null cancer
EP3178848A1 (en) 2015-12-09 2017-06-14 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody for reducing formation of anti-drug antibodies
JP7325186B2 (en) 2015-12-09 2023-08-14 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Type II anti-CD20 antibody for reducing the formation of anti-drug antibodies
SG11201804787UA (en) 2015-12-15 2018-07-30 Gilead Sciences Inc Human immunodeficiency virus neutralizing antibodies
JP2019502695A (en) 2015-12-17 2019-01-31 ノバルティス アーゲー Combination of antibody molecule against PD-1 and C-Met inhibitor and use thereof
CA3007671A1 (en) 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
PL3390442T3 (en) 2015-12-18 2024-03-18 Chugai Seiyaku Kabushiki Kaisha Anti-c5 antibodies and methods of use
CN108430455A (en) 2015-12-30 2018-08-21 豪夫迈·罗氏有限公司 Tryptophan derivative is used for the purposes of protein preparaton
EP3397287A1 (en) 2015-12-30 2018-11-07 Genentech, Inc. Formulations with reduced degradation of polysorbate
EP3401336A4 (en) 2016-01-05 2020-01-22 Jiangsu Hengrui Medicine Co., Ltd. Pcsk9 antibody, antigen-binding fragment thereof, and medical uses thereof
MX2018008347A (en) 2016-01-08 2018-12-06 Hoffmann La Roche Methods of treating cea-positive cancers using pd-1 axis binding antagonists and anti-cea/anti-cd3 bispecific antibodies.
WO2017127764A1 (en) 2016-01-20 2017-07-27 Genentech, Inc. High dose treatments for alzheimer's disease
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
CN109073635A (en) 2016-01-25 2018-12-21 豪夫迈·罗氏有限公司 Method for measuring T cell dependence bispecific antibody
CA3019952A1 (en) 2016-02-04 2017-08-10 Curis, Inc. Mutant smoothened and methods of using the same
US10982136B2 (en) 2016-02-26 2021-04-20 The Regents Of The University Of California Ligand-sensitized lanthanide nanocrystals as ultraviolet downconverters
CN109196121B (en) 2016-02-29 2022-01-04 基因泰克公司 Methods for treatment and diagnosis of cancer
EP3423482A1 (en) 2016-03-04 2019-01-09 Novartis AG Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
US11472877B2 (en) 2016-03-04 2022-10-18 Alector Llc Anti-TREM1 antibodies and methods of use thereof
EP3216458A1 (en) 2016-03-07 2017-09-13 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Modified vascular endothelial growth factor a (vegf-a) and its medical use
TW201808978A (en) 2016-03-08 2018-03-16 中央研究院 Methods for modular synthesis of N-glycans and arrays thereof
TW202248213A (en) 2016-03-15 2022-12-16 日商中外製藥股份有限公司 Methods of treating cancers using pd-1 axis binding antagonists and anti-gpc3 antibodies
WO2017161206A1 (en) 2016-03-16 2017-09-21 Halozyme, Inc. Conjugates containing conditionally active antibodies or antigen-binding fragments thereof, and methods of use
SG11201808085WA (en) 2016-03-22 2018-10-30 Hoffmann La Roche Protease-activated t cell bispecific molecules
FI3433280T3 (en) 2016-03-22 2023-06-06 Hoffmann La Roche Protease-activated t cell bispecific molecules
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
JP6943872B2 (en) 2016-03-25 2021-10-06 ジェネンテック, インコーポレイテッド Multiple whole antibody and antibody complex drug quantification assay
KR20180121786A (en) 2016-03-29 2018-11-08 오비아이 파머 인코퍼레이티드 Antibodies, pharmaceutical compositions and methods
US10980894B2 (en) 2016-03-29 2021-04-20 Obi Pharma, Inc. Antibodies, pharmaceutical compositions and methods
EP3231813A1 (en) 2016-03-29 2017-10-18 F. Hoffmann-La Roche AG Trimeric costimulatory tnf family ligand-containing antigen binding molecules
WO2017180864A1 (en) 2016-04-14 2017-10-19 Genentech, Inc. Anti-rspo3 antibodies and methods of use
ES2850428T3 (en) 2016-04-15 2021-08-30 Hoffmann La Roche Cancer monitoring and treatment procedures
MX2018012615A (en) 2016-04-15 2019-05-30 Novartis Ag Compositions and methods for selective protein expression.
CA3021086C (en) 2016-04-15 2023-10-17 Bioatla, Llc Anti-axl antibodies, antibody fragments and their immunoconjugates and uses thereof
JP2019515670A (en) 2016-04-15 2019-06-13 ジェネンテック, インコーポレイテッド Methods for monitoring and treating cancer
KR20230110820A (en) 2016-04-22 2023-07-25 오비아이 파머 인코퍼레이티드 Cancer immunotherapy by immune activation or immune modulation via globo series antigens
UA123323C2 (en) 2016-05-02 2021-03-17 Ф. Хоффманн-Ля Рош Аг The contorsbody - a single chain target binder
EP3454863A1 (en) 2016-05-10 2019-03-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Combinations therapies for the treatment of cancer
EP3243836A1 (en) 2016-05-11 2017-11-15 F. Hoffmann-La Roche AG C-terminally fused tnf family ligand trimer-containing antigen binding molecules
WO2017194442A1 (en) 2016-05-11 2017-11-16 F. Hoffmann-La Roche Ag Antigen binding molecules comprising a tnf family ligand trimer and a tenascin binding moiety
EP3455252B1 (en) 2016-05-11 2022-02-23 F. Hoffmann-La Roche AG Modified anti-tenascin antibodies and methods of use
SI3455261T1 (en) 2016-05-13 2023-01-31 Bioatla, Inc. Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
EP3243832A1 (en) 2016-05-13 2017-11-15 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer and pd1 binding moiety
JP7359547B2 (en) 2016-05-17 2023-10-11 ジェネンテック, インコーポレイテッド Stromal gene signatures for diagnosis and use in immunotherapy
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use
JP7022080B2 (en) 2016-05-27 2022-02-17 ジェネンテック, インコーポレイテッド Biochemical analytical methods for the characterization of site-specific antibody-drug conjugates
US20210177896A1 (en) 2016-06-02 2021-06-17 Novartis Ag Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
WO2017207694A1 (en) 2016-06-02 2017-12-07 Kohlmann Angelica Antibodies that bind to human anti-müllerian hormone (amh) and their uses
EP3252078A1 (en) 2016-06-02 2017-12-06 F. Hoffmann-La Roche AG Type ii anti-cd20 antibody and anti-cd20/cd3 bispecific antibody for treatment of cancer
TW201902512A (en) 2016-06-02 2019-01-16 瑞士商赫孚孟拉羅股份公司 treatment method
EP3464280B1 (en) 2016-06-06 2021-10-06 F. Hoffmann-La Roche AG Silvestrol antibody-drug conjugates and methods of use
CN109563124A (en) 2016-06-17 2019-04-02 豪夫迈·罗氏有限公司 The purifying of multi-specificity antibody
CN109563160B (en) 2016-06-24 2023-02-28 豪夫迈·罗氏有限公司 Anti-polyubiquitin multispecific antibodies
WO2018011691A1 (en) 2016-07-12 2018-01-18 Nestec S.A. Competitive immunoassay methods
CA3030841A1 (en) 2016-07-14 2018-01-18 Fred Hutchinson Cancer Research Center Multiple bi-specific binding domain constructs with different epitope binding to treat cancer
JP7219376B2 (en) 2016-07-15 2023-02-08 ノバルティス アーゲー Treatment and prevention of cytokine release syndrome using chimeric antigen receptors in combination with kinase inhibitors
ES2875905T3 (en) 2016-07-15 2021-11-11 Acceleron Pharma Inc Compositions comprising ActRIIA polypeptides for use in the treatment of pulmonary hypertension
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
BR112019001615A2 (en) 2016-07-27 2019-04-30 Acceleron Pharma Inc. methods and compositions for treating myelofibrosis
CN110072545A (en) 2016-07-27 2019-07-30 台湾浩鼎生技股份有限公司 Immunogenicity/therapeutic glycan pool object and application thereof
AU2017302668B9 (en) 2016-07-28 2023-06-22 Novartis Ag Combination therapies of chimeric antigen receptors and PD-1 inhibitors
KR102528998B1 (en) 2016-07-29 2023-05-03 오비아이 파머 인코퍼레이티드 Human Antibodies, Pharmaceutical Compositions and Methods
RU2019104730A (en) 2016-07-29 2020-08-28 Чугаи Сейяку Кабусики Кайся BISPECIFIC ANTIBODY WITH INCREASED ACTIVITY, ALTERNATIVE FUNCTION OF COFACTOR FVIII
CA3032581A1 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
EP3494139B1 (en) 2016-08-05 2022-01-12 F. Hoffmann-La Roche AG Multivalent and multiepitopic anitibodies having agonistic activity and methods of use
CN116251182A (en) 2016-08-05 2023-06-13 中外制药株式会社 Compositions for preventing or treating IL-8 related diseases
JP7250674B2 (en) 2016-08-08 2023-04-03 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト CANCER TREATMENT AND DIAGNOSTIC METHOD
WO2018031662A1 (en) 2016-08-11 2018-02-15 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
WO2018035025A1 (en) 2016-08-15 2018-02-22 Genentech, Inc. Chromatography method for quantifying a non-ionic surfactant in a composition comprising the non-ionic surfactant and a polypeptide
JP7213549B2 (en) 2016-08-22 2023-01-27 シーエイチオー ファーマ インコーポレイテッド Antibodies, Binding Fragments, and Methods of Use
EP3503930A4 (en) 2016-08-29 2020-08-05 Fred Hutchinson Cancer Research Center Chelating platform for delivery of radionuclides
BR112019003408A2 (en) 2016-08-31 2019-06-25 Oncotherapy Science Inc monoclonal antibody against melk and its use
US11684614B2 (en) 2016-09-06 2023-06-27 The Regents Of The University Of California Formulations of hydroxypyridonate actinide/lanthanide decorporation agents
EP3510046A4 (en) 2016-09-07 2020-05-06 The Regents of the University of California Antibodies to oxidation-specific epitopes
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
CN109689682B (en) 2016-09-19 2022-11-29 豪夫迈·罗氏有限公司 Complement factor-based affinity chromatography
WO2018053405A1 (en) 2016-09-19 2018-03-22 Celgene Corporation Methods of treating immune disorders using pd-1 binding proteins
US10766958B2 (en) 2016-09-19 2020-09-08 Celgene Corporation Methods of treating vitiligo using PD-1 binding antibodies
JOP20190009A1 (en) 2016-09-21 2019-01-27 Alx Oncology Inc Antibodies against signal-regulatory protein alpha and methods of use
KR102557643B1 (en) 2016-09-23 2023-07-20 제넨테크, 인크. Use of IL-13 antagonists to treat atopic dermatitis
EP3519437B1 (en) 2016-09-30 2021-09-08 F. Hoffmann-La Roche AG Bispecific antibodies against p95her2
CN110139674B (en) 2016-10-05 2023-05-16 豪夫迈·罗氏有限公司 Method for preparing antibody drug conjugates
WO2018067740A1 (en) 2016-10-05 2018-04-12 Acceleron Pharma, Inc. Compositions and method for treating kidney disease
AU2017339517B2 (en) 2016-10-06 2024-03-14 Foundation Medicine, Inc. Therapeutic and diagnostic methods for cancer
TWI762516B (en) 2016-10-06 2022-05-01 日商腫瘤療法 科學股份有限公司 Monoclonal antibodies against FZD10 and their uses
AR110676A1 (en) 2016-10-07 2019-04-24 Novartis Ag TREATMENT OF CANCER USING CHEMERIC ANTIGENS RECEPTORS
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
EP3529618B1 (en) 2016-10-19 2020-12-02 Alexion Pharmaceuticals, Inc. A method of quantitating unbound c5 in a sample
WO2018075761A1 (en) 2016-10-19 2018-04-26 Alexion Pharmaceuticals, Inc. A method of quantitating unbound c5a in a sample
EP3532496A1 (en) 2016-10-28 2019-09-04 Banyan Biomarkers, Inc. Antibodies to ubiquitin c-terminal hydrolase l1 (uch-l1) and glial fibrillary acidic protein (gfap) and related methods
EP3532091A2 (en) 2016-10-29 2019-09-04 H. Hoffnabb-La Roche Ag Anti-mic antibidies and methods of use
AU2017361081A1 (en) 2016-11-15 2019-05-23 Genentech, Inc. Dosing for treatment with anti-CD20/anti-CD3 bispecific antibodies
TW201829463A (en) 2016-11-18 2018-08-16 瑞士商赫孚孟拉羅股份公司 Anti-hla-g antibodies and use thereof
US11279694B2 (en) 2016-11-18 2022-03-22 Sumitomo Dainippon Pharma Oncology, Inc. Alvocidib prodrugs and their use as protein kinase inhibitors
JOP20190100A1 (en) 2016-11-19 2019-05-01 Potenza Therapeutics Inc Anti-gitr antigen-binding proteins and methods of use thereof
JP2019535731A (en) 2016-11-21 2019-12-12 オービーアイ ファーマ,インコーポレイテッド Conjugated biological molecules, pharmaceutical compositions and methods
CR20230163A (en) 2016-12-07 2023-07-06 Genentech Inc Anti-tau antibodies and methods of use
AU2017373884A1 (en) 2016-12-07 2019-05-30 Ac Immune Sa Anti-tau antibodies and methods of their use
WO2018108759A1 (en) 2016-12-13 2018-06-21 F. Hoffmann-La Roche Ag Method to determine the presence of a target antigen in a tumor sample
US10132797B2 (en) 2016-12-19 2018-11-20 Tolero Pharmaceuticals, Inc. Profiling peptides and methods for sensitivity profiling
CN110087682B (en) 2016-12-19 2023-12-15 豪夫迈·罗氏有限公司 Combination therapy with targeted 4-1BB (CD 137) agonists
EP3559034B1 (en) 2016-12-20 2020-12-02 H. Hoffnabb-La Roche Ag Combination therapy of anti-cd20/anti-cd3 bispecific antibodies and 4-1bb (cd137) agonists
JOP20190134A1 (en) 2016-12-23 2019-06-02 Potenza Therapeutics Inc Anti-neuropilin antigen-binding proteins and methods of use thereof
BR112019013189A2 (en) 2017-01-03 2019-12-10 Hoffmann La Roche bispecific antigen binding molecules, polynucleotide, host cell, bispecific antigen binding molecule production method, pharmaceutical composition, use, methods for inhibiting tumor cell growth in an individual and for treating cancer or an infectious disease
TW201825515A (en) 2017-01-04 2018-07-16 美商伊繆諾金公司 Met antibodies and immunoconjugates and uses thereof
EP3565845A4 (en) 2017-01-06 2020-10-07 Biosion, Inc. Erbb2 antibodies and uses therefore
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
WO2018147960A1 (en) 2017-02-08 2018-08-16 Imaginab, Inc. Extension sequences for diabodies
AR110873A1 (en) 2017-02-10 2019-05-08 Genentech Inc ANTIBODIES AGAINST TRIPTASE, COMPOSITIONS OF THESE AND USES OF THESE
WO2018152496A1 (en) 2017-02-17 2018-08-23 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Compositions and methods for the diagnosis and treatment of zika virus infection
EP3589647A1 (en) 2017-02-28 2020-01-08 Novartis AG Shp inhibitor compositions and uses for chimeric antigen receptor therapy
TW201837467A (en) 2017-03-01 2018-10-16 美商建南德克公司 Diagnostic and therapeutic methods for cancer
WO2018158719A1 (en) 2017-03-02 2018-09-07 Novartis Ag Engineered heterodimeric proteins
SG10202107829YA (en) 2017-03-22 2021-08-30 Genentech Inc Hydrogel cross-linked hyaluronic acid prodrug compositions and methods
TWI808963B (en) 2017-03-22 2023-07-21 法商賽諾菲公司 Treatment of lupus using humanized anti-cxcr5 antibodies
MX2019011141A (en) 2017-03-22 2019-11-05 Genentech Inc Optimized antibody compositions for treatment of ocular disorders.
CR20190440A (en) 2017-03-27 2019-11-12 Hoffmann La Roche Improved antigen binding receptors
SG11201908784TA (en) 2017-03-27 2019-10-30 Hoffmann La Roche Improved antigen binding receptor formats
TW202400231A (en) 2017-03-28 2024-01-01 美商建南德克公司 Methods of treating neurodegenerative diseases
WO2018178074A1 (en) 2017-03-29 2018-10-04 F. Hoffmann-La Roche Ag Trimeric antigen binding molecules specific for a costimulatory tnf receptor
CN110382542B (en) 2017-03-29 2023-06-09 豪夫迈·罗氏有限公司 Bispecific antigen binding molecules to costimulatory TNF receptors
CN110573528B (en) 2017-03-29 2023-06-09 豪夫迈·罗氏有限公司 Bispecific antigen binding molecules to costimulatory TNF receptors
JOP20190203A1 (en) 2017-03-30 2019-09-03 Potenza Therapeutics Inc Anti-tigit antigen-binding proteins and methods of use thereof
BR112019018767A2 (en) 2017-04-03 2020-05-05 Hoffmann La Roche antibodies, bispecific antigen binding molecule, one or more isolated polynucleotides, one or more vectors, host cell, method for producing an antibody, pharmaceutical composition, uses, method for treating a disease in an individual and invention
WO2018184965A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of il-2 with an anti-pd-1 and tim-3 bispecific antibody
LT3606946T (en) 2017-04-03 2022-10-25 F. Hoffmann-La Roche Ag Immunoconjugates of an anti-pd-1 antibody with a mutant il-2 or with il-15
BR112019017753A2 (en) 2017-04-04 2020-04-07 Hoffmann La Roche bispecific molecule, polynucleotide, vector, cell, methods for the production of a molecule and for the treatment of an individual, composition and use of the bispecific molecule
KR102346336B1 (en) 2017-04-05 2022-01-04 에프. 호프만-라 로슈 아게 Bispecific antibodies that specifically bind to PD1 and LAG3
MX2019011916A (en) 2017-04-05 2020-01-09 Hoffmann La Roche Anti-lag3 antibodies.
RU2665790C1 (en) 2017-04-17 2018-09-04 Закрытое Акционерное Общество "Биокад" Monoclonal pd-l1 antibody
CN108728444A (en) 2017-04-18 2018-11-02 长春华普生物技术股份有限公司 Immunoregulation polynucleotide and its application
JP7248588B2 (en) 2017-04-21 2023-03-29 ジェネンテック, インコーポレイテッド Use of KLK5 antagonists for the treatment of disease
EP3615569A1 (en) 2017-04-25 2020-03-04 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Antibodies and methods for the diagnosis and treatment of epstein barr virus infection
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
MX2019013137A (en) 2017-05-05 2020-07-14 Allakos Inc Methods and compositions for treating allergic ocular diseases.
CN111094335B (en) 2017-05-15 2022-08-23 罗切斯特大学 Broadly neutralizing anti-influenza monoclonal antibodies and uses thereof
US11359014B2 (en) 2017-05-16 2022-06-14 Alector Llc Anti-siglec-5 antibodies and methods of use thereof
CA3063344A1 (en) 2017-05-23 2018-11-29 Helmholtz Zentrum Munchen - Deutsches Forschungszentrum Fur Gesundheit Und Umwelt (Gmbh) Novel cd73 antibody, preparation and uses thereof
EP3409322A1 (en) 2017-06-01 2018-12-05 F. Hoffmann-La Roche AG Treatment method
JP7433910B2 (en) 2017-06-22 2024-02-20 ノバルティス アーゲー Antibody molecules against CD73 and uses thereof
WO2019006007A1 (en) 2017-06-27 2019-01-03 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof
WO2019000223A1 (en) 2017-06-27 2019-01-03 Nanjing Legend Biotech Co., Ltd. Chimeric antibody immune effctor cell engagers and methods of use thereof
BR112019025105A2 (en) 2017-06-30 2020-10-20 Zymeworks Inc. stabilized chimeric fabs
MX2020000342A (en) 2017-07-11 2020-08-17 Compass Therapeutics Llc Agonist antibodies that bind human cd137 and uses thereof.
CN111094334A (en) 2017-07-19 2020-05-01 美国卫生与公众服务部 Antibodies and methods for diagnosis and treatment of hepatitis B virus infection
JP2020527572A (en) 2017-07-20 2020-09-10 ノバルティス アーゲー Anti-LAG-3 antibody dosage regimen and its use
JP2020527351A (en) 2017-07-21 2020-09-10 ジェネンテック, インコーポレイテッド Cancer treatment and diagnosis
EP3658589B1 (en) 2017-07-26 2023-09-27 Forty Seven, Inc. Anti-sirp-alpha antibodies and related methods
CA3069073A1 (en) 2017-07-28 2019-01-31 F. Hoffmann-La Roche Ag Bispecific antibody formulation
CN117700548A (en) 2017-08-03 2024-03-15 艾利妥 anti-CD 33 antibodies and methods of use thereof
ES2952982T3 (en) 2017-08-03 2023-11-07 Alector Llc Anti-TREM2 antibodies and methods of their use
WO2019033043A2 (en) 2017-08-11 2019-02-14 Genentech, Inc. Anti-cd8 antibodies and uses thereof
WO2019055579A1 (en) 2017-09-12 2019-03-21 Tolero Pharmaceuticals, Inc. Treatment regimen for cancers that are insensitive to bcl-2 inhibitors using the mcl-1 inhibitor alvocidib
EP3684413A1 (en) 2017-09-20 2020-07-29 Chugai Seiyaku Kabushiki Kaisha Dosage regimen for combination therapy using pd-1 axis binding antagonists and gpc3 targeting agent
MX2020002710A (en) 2017-09-29 2020-07-20 Chugai Pharmaceutical Co Ltd Multispecific antigen-binding molecule having blood coagulation factor viii (fviii) cofactor function-substituting activity, and pharmaceutical formulation containing said molecule as active ingredient.
RU2698048C2 (en) 2017-10-03 2019-08-21 Закрытое Акционерное Общество "Биокад" Monoclonal antibody to il-5rα
EA039662B1 (en) 2017-10-03 2022-02-24 Закрытое Акционерное Общество "Биокад" Antibodies specific to cd47 and pd-l1
US20200237766A1 (en) 2017-10-13 2020-07-30 Tolero Pharmaceuticals, Inc. Pkm2 activators in combination with reactive oxygen species for treatment of cancer
EP3697441B1 (en) 2017-10-20 2023-06-07 F. Hoffmann-La Roche AG Method for generating multispecific antibodies from monospecific antibodies
CA3078676A1 (en) 2017-10-30 2019-05-09 F. Hoffmann-La Roche Ag Method for in vivo generation of multispecific antibodies from monospecific antibodies
WO2019089753A2 (en) 2017-10-31 2019-05-09 Compass Therapeutics Llc Cd137 antibodies and pd-1 antagonists and uses thereof
CA3079036A1 (en) 2017-11-01 2019-05-09 F. Hoffmann-La Roche Ag Combination therapy with targeted ox40 agonists
PL3704146T3 (en) 2017-11-01 2022-03-07 F. Hoffmann-La Roche Ag Trifab-contorsbody
EP3703821A2 (en) 2017-11-01 2020-09-09 F. Hoffmann-La Roche AG Bispecific 2+1 contorsbodies
JP2021500902A (en) 2017-11-01 2021-01-14 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト New TNF family ligand trimer-containing antigen-binding molecule
EP3704150A1 (en) 2017-11-01 2020-09-09 F. Hoffmann-La Roche AG The compbody - a multivalent target binder
JP2021502066A (en) 2017-11-06 2021-01-28 ジェネンテック, インコーポレイテッド Cancer diagnosis and therapy
JP2021503478A (en) 2017-11-16 2021-02-12 ノバルティス アーゲー Combination treatment
WO2019100052A2 (en) 2017-11-20 2019-05-23 Compass Therapeutics Llc Cd137 antibodies and tumor antigen-targeting antibodies and uses thereof
MX2020006125A (en) 2017-12-14 2020-08-24 Hoffmann La Roche Use of a cea cd3 bispecific antibody and a pd-1 axis binding antagonist in a dosage regime to treat cancer.
CN111247429A (en) 2017-12-21 2020-06-05 豪夫迈·罗氏有限公司 Universal reporter cell assay for specific testing of novel antigen binding modules
CN111492243A (en) 2017-12-21 2020-08-04 豪夫迈·罗氏有限公司 CAR-T cell assay for specific testing of novel antigen binding modules
MX2020006119A (en) 2017-12-21 2020-08-24 Hoffmann La Roche Antibodies binding to hla-a2/wt1.
EP3502140A1 (en) 2017-12-21 2019-06-26 F. Hoffmann-La Roche AG Combination therapy of tumor targeted icos agonists with t-cell bispecific molecules
EP3728321A1 (en) 2017-12-22 2020-10-28 F. Hoffmann-La Roche AG Use of pilra binding agents for treatment of a disease
KR20200104886A (en) 2017-12-28 2020-09-04 난징 레전드 바이오테크 씨오., 엘티디. Antibodies and variants against PD-L1
KR20200104333A (en) 2017-12-28 2020-09-03 난징 레전드 바이오테크 씨오., 엘티디. Single-domain antibodies to TIGIT and variants thereof
US11440957B2 (en) 2017-12-29 2022-09-13 Alector Llc Anti-TMEM106B antibodies and methods of use thereof
CN111479588A (en) 2017-12-29 2020-07-31 豪夫迈·罗氏有限公司 Methods for improving VEGF receptor blocking selectivity of anti-VEGF antibodies
US11713353B2 (en) 2018-01-15 2023-08-01 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against PD-1
EP3740505A1 (en) 2018-01-16 2020-11-25 Lakepharma Inc. Bispecific antibody that binds cd3 and another target
CN111630063A (en) 2018-01-31 2020-09-04 豪夫迈·罗氏有限公司 Stabilized immunoglobulin domains
US20210038659A1 (en) 2018-01-31 2021-02-11 Novartis Ag Combination therapy using a chimeric antigen receptor
WO2019149716A1 (en) 2018-01-31 2019-08-08 F. Hoffmann-La Roche Ag Bispecific antibodies comprising an antigen-binding site binding to lag3
US11472874B2 (en) 2018-01-31 2022-10-18 Alector Llc Anti-MS4A4A antibodies and methods of use thereof
AU2019218959A1 (en) 2018-02-08 2020-09-03 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
SG11202007564VA (en) 2018-02-09 2020-09-29 Genentech Inc Therapeutic and diagnostic methods for mast cell-mediated inflammatory diseases
TWI829667B (en) 2018-02-09 2024-01-21 瑞士商赫孚孟拉羅股份公司 Antibodies binding to gprc5d
WO2019165434A1 (en) 2018-02-26 2019-08-29 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
WO2019166453A1 (en) 2018-03-01 2019-09-06 F. Hoffmann-La Roche Ag Specificity assay for novel target antigen binding moieties
TW202003561A (en) 2018-03-13 2020-01-16 瑞士商赫孚孟拉羅股份公司 Combination therapy with targeted 4-1BB (CD137) agonists
WO2019175071A1 (en) 2018-03-13 2019-09-19 F. Hoffmann-La Roche Ag Therapeutic combination of 4-1 bb agonists with anti-cd20 antibodies
US20200040103A1 (en) 2018-03-14 2020-02-06 Genentech, Inc. Anti-klk5 antibodies and methods of use
BR112020014591A2 (en) 2018-03-14 2020-12-01 Beijing Xuanyi Pharmasciences Co., Ltd. anticlaudin antibodies 18.2
CA3093729A1 (en) 2018-03-15 2019-09-19 Chugai Seiyaku Kabushiki Kaisha Anti-dengue virus antibodies having cross-reactivity to zika virus and methods of use
CA3094098A1 (en) 2018-03-21 2019-09-26 ALX Oncology Inc. Antibodies against signal-regulatory protein alpha and methods of use
AU2019245243A1 (en) 2018-03-29 2020-09-03 Genentech, Inc Modulating lactogenic activity in mammalian cells
WO2019192432A1 (en) 2018-04-02 2019-10-10 上海博威生物医药有限公司 Lymphocyte activation gene-3 (lag-3) binding antibody and use thereof
TW202011029A (en) 2018-04-04 2020-03-16 美商建南德克公司 Methods for detecting and quantifying FGF21
WO2019192972A1 (en) 2018-04-04 2019-10-10 F. Hoffmann-La Roche Ag Diagnostic assays to detect tumor antigens in cancer patients
CN111742220A (en) 2018-04-04 2020-10-02 豪夫迈·罗氏有限公司 Diagnostic assay for detecting tumor antigens in cancer patients
US10870691B2 (en) 2018-04-05 2020-12-22 Gilead Sciences, Inc. Antibodies and fragments thereof that bind hepatitis B virus protein X
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
AR114284A1 (en) 2018-04-13 2020-08-12 Hoffmann La Roche HER2 TARGETING ANTIGEN BINDING MOLECULES COMPOSING 4-1BBL
AR115052A1 (en) 2018-04-18 2020-11-25 Hoffmann La Roche MULTI-SPECIFIC ANTIBODIES AND THE USE OF THEM
AR114789A1 (en) 2018-04-18 2020-10-14 Hoffmann La Roche ANTI-HLA-G ANTIBODIES AND THE USE OF THEM
US20210047405A1 (en) 2018-04-27 2021-02-18 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213416A1 (en) 2018-05-02 2019-11-07 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies and methods for the diagnosis, prevention, and treatment of epstein barr virus infection
CA3096703A1 (en) 2018-05-03 2019-11-07 University Of Rochester Anti-influenza neuraminidase monoclonal antibodies and uses thereof
ES2955511T3 (en) 2018-05-14 2023-12-04 Werewolf Therapeutics Inc Activatable interleukin 2 polypeptides and methods of use thereof
JP2021524756A (en) 2018-05-14 2021-09-16 ウェアウルフ セラピューティクス, インコーポレイテッド Activateable cytokine polypeptides and how to use them
AU2019269066B2 (en) 2018-05-18 2022-10-06 F. Hoffmann-La Roche Ag Targeted intracellular delivery of large nucleic acids
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
WO2019226617A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Compositions and methods for enhancing the killing of target cells by nk cells
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
CA3098710A1 (en) 2018-05-25 2019-11-28 Alector Llc Anti-sirpa antibodies and methods of use thereof
US20210214459A1 (en) 2018-05-31 2021-07-15 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2019235426A1 (en) 2018-06-04 2019-12-12 中外製薬株式会社 Antigen-binding molecule showing changed half-life in cytoplasm
WO2019236965A1 (en) 2018-06-08 2019-12-12 Alector Llc Anti-siglec-7 antibodies and methods of use thereof
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
KR20210035173A (en) 2018-06-19 2021-03-31 아타르가, 엘엘씨 Antibody molecule against complement component 5 and use thereof
WO2019246557A1 (en) 2018-06-23 2019-12-26 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
US11203645B2 (en) 2018-06-27 2021-12-21 Obi Pharma, Inc. Glycosynthase variants for glycoprotein engineering and methods of use
CN112384532A (en) 2018-06-29 2021-02-19 艾利妥 anti-SIRP-beta 1 antibodies and methods of use thereof
EP3818082A1 (en) 2018-07-04 2021-05-12 F. Hoffmann-La Roche AG Novel bispecific agonistic 4-1bb antigen binding molecules
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
AR116109A1 (en) 2018-07-10 2021-03-31 Novartis Ag DERIVATIVES OF 3- (5-AMINO-1-OXOISOINDOLIN-2-IL) PIPERIDINE-2,6-DIONA AND USES OF THE SAME
CA3060547A1 (en) 2018-07-13 2020-01-13 Alector Llc Anti-sortilin antibodies and methods of use thereof
CA3104147A1 (en) 2018-07-18 2020-01-23 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
AU2019306628A1 (en) 2018-07-20 2021-02-11 Surface Oncology, Inc. Anti-CD112R compositions and methods
WO2020021465A1 (en) 2018-07-25 2020-01-30 Advanced Accelerator Applications (Italy) S.R.L. Method of treatment of neuroendocrine tumors
CA3103995A1 (en) 2018-07-26 2020-01-30 Sumitomo Dainippon Pharma Oncology, Inc. Methods for treating diseases associated with abnormal acvr1 expression and acvr1 inhibitors for use in the same
WO2020023920A1 (en) 2018-07-27 2020-01-30 Alector Llc Anti-siglec-5 antibodies and methods of use thereof
WO2020027330A1 (en) 2018-08-03 2020-02-06 中外製薬株式会社 Antigen-binding molecule containing two antigen-binding domains that are linked to each other
BR112021002130A2 (en) 2018-08-08 2021-05-04 Genentech, Inc. liquid formulation, article of manufacture or kit and method for reducing oxidation of a polypeptide
WO2020032230A1 (en) 2018-08-10 2020-02-13 中外製薬株式会社 Anti-cd137 antigen-binding molecule and utilization thereof
TW202021618A (en) 2018-08-17 2020-06-16 美商23與我有限公司 Anti-il1rap antibodies and methods of use thereof
US11548938B2 (en) 2018-08-21 2023-01-10 Quidel Corporation DbpA antibodies and uses thereof
US11401339B2 (en) 2018-08-23 2022-08-02 Seagen Inc. Anti-TIGIT antibodies
EP3843851A1 (en) 2018-08-31 2021-07-07 Alector LLC Anti-cd33 antibodies and methods of use thereof
GB201814281D0 (en) 2018-09-03 2018-10-17 Femtogenix Ltd Cytotoxic agents
TW202023542A (en) 2018-09-18 2020-07-01 瑞士商赫孚孟拉羅股份公司 Use of a cathepsin s inhibitor against the formation of anti-drug antibodies
AU2019342099A1 (en) 2018-09-19 2021-04-08 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
EP4249917A3 (en) 2018-09-21 2023-11-08 F. Hoffmann-La Roche AG Diagnostic methods for triple-negative breast cancer
JP2022502088A (en) 2018-09-27 2022-01-11 エクシリオ デベロップメント, インコーポレイテッド Masked cytokine polypeptide
AU2019349958A1 (en) 2018-09-28 2021-05-06 Kyowa Kirin Co., Ltd. IL-36 antibodies and uses thereof
EP3861025A1 (en) 2018-10-01 2021-08-11 F. Hoffmann-La Roche AG Bispecific antigen binding molecules with trivalent binding to cd40
JP7221379B2 (en) 2018-10-01 2023-02-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Bispecific antigen-binding molecule comprising anti-FAP clone 212
US20200109200A1 (en) 2018-10-09 2020-04-09 Genentech, Inc. Methods and systems for determining synapse formation
MX2021004348A (en) 2018-10-18 2021-05-28 Genentech Inc Diagnostic and therapeutic methods for sarcomatoid kidney cancer.
JP2022505450A (en) 2018-10-24 2022-01-14 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Conjugated chemical decomposition inducers and usage
RU2724469C2 (en) 2018-10-31 2020-06-23 Закрытое Акционерное Общество "Биокад" Monoclonal antibody which specifically binds to cd20
AU2019375413A1 (en) 2018-11-05 2021-05-27 Genentech, Inc. Methods of producing two chain proteins in prokaryotic host cells
MX2021005594A (en) 2018-11-13 2021-10-22 Compass Therapeutics Llc Multispecific binding constructs against checkpoint molecules and uses thereof.
EP3884276A2 (en) 2018-11-23 2021-09-29 Katholieke Universiteit Leuven Predicting a treatment response in inflammatory bowel disease
MX2021006544A (en) 2018-12-04 2021-07-07 Sumitomo Pharma Oncology Inc Cdk9 inhibitors and polymorphs thereof for use as agents for treatment of cancer.
MX2021006573A (en) 2018-12-06 2021-07-15 Genentech Inc Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody.
CN113227119A (en) 2018-12-10 2021-08-06 基因泰克公司 Photocrosslinked peptides for site-specific conjugation to Fc-containing proteins
AR117327A1 (en) 2018-12-20 2021-07-28 23Andme Inc ANTI-CD96 ANTIBODIES AND METHODS OF USE OF THEM
AU2019402189B2 (en) 2018-12-20 2023-04-13 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2020128927A1 (en) 2018-12-20 2020-06-25 Kyowa Kirin Co., Ltd. Fn14 antibodies and uses thereof
JP2022514017A (en) 2018-12-20 2022-02-09 ノバルティス アーゲー Combination of medicines
CN113631714A (en) 2018-12-21 2021-11-09 豪夫迈·罗氏有限公司 Methods of producing polypeptides using apoptosis-resistant cell lines
AR117728A1 (en) 2018-12-21 2021-08-25 Hoffmann La Roche CD28 ANTIGEN BINDING SUPERAGONIST MOLECULES WITH TUMOR TARGET
MY198034A (en) 2018-12-21 2023-07-27 Hoffmann La Roche Tumor-targeted agonistic cd28 antigen binding molecules
TWI829831B (en) 2018-12-21 2024-01-21 瑞士商赫孚孟拉羅股份公司 Antibodies binding to cd3
JP2022514950A (en) 2018-12-21 2022-02-16 23アンドミー・インコーポレイテッド Anti-IL-36 antibody and how to use it
EP3902833A2 (en) 2018-12-26 2021-11-03 City of Hope Activatable masked anti-ctla4 binding proteins
CN113272327A (en) 2018-12-30 2021-08-17 豪夫迈·罗氏有限公司 Anti-rabbit CD19 antibodies and methods of use thereof
AU2020208193A1 (en) 2019-01-14 2021-07-29 BioNTech SE Methods of treating cancer with a PD-1 axis binding antagonist and an RNA vaccine
JPWO2020153467A1 (en) 2019-01-24 2021-12-02 中外製薬株式会社 New cancer antigens and antibodies against those antigens
GB201901197D0 (en) 2019-01-29 2019-03-20 Femtogenix Ltd G-A Crosslinking cytotoxic agents
EP3924351A4 (en) 2019-02-12 2022-12-21 Sumitomo Pharma Oncology, Inc. Formulations comprising heterocyclic protein kinase inhibitors
AU2020222346B2 (en) 2019-02-15 2021-12-09 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
EA202192019A1 (en) 2019-02-15 2021-11-02 Новартис Аг DERIVATIVES OF 3- (1-OXO-5- (PIPERIDIN-4-YL) ISOINDOLIN-2-YL) PIPERIDINE-2,6-DIONE AND ROUTES OF THEIR APPLICATION
US10871640B2 (en) 2019-02-15 2020-12-22 Perkinelmer Cellular Technologies Germany Gmbh Methods and systems for automated imaging of three-dimensional objects
WO2020169698A1 (en) 2019-02-21 2020-08-27 F. Hoffmann-La Roche Ag Sensitization of cancer cells to tnf by bet inhibition
EP3927371A1 (en) 2019-02-22 2021-12-29 Novartis AG Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
CN113710706A (en) 2019-02-27 2021-11-26 豪夫迈·罗氏有限公司 Administration for anti-TIGIT antibody and anti-CD 20 antibody or anti-CD 38 antibody treatment
MX2021010565A (en) 2019-03-08 2021-10-13 Genentech Inc Methods for detecting and quantifying membrane-associated proteins on extracellular vesicles.
JP2022524537A (en) 2019-03-13 2022-05-06 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Process for the preparation of lipid-added proteinaceous structures
MA55296A (en) 2019-03-14 2022-03-23 Hoffmann La Roche CANCER TREATMENT WITH BISPECIFIC ANTIBODIES TO HER2XCD3 IN COMBINATION WITH AN ANTI-HER2 MAB
WO2020191326A1 (en) 2019-03-20 2020-09-24 Sumitomo Dainippon Pharma Oncology, Inc. Treatment of acute myeloid leukemia (aml) with venetoclax failure
AU2020245437A1 (en) 2019-03-22 2021-09-30 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same
TW202102261A (en) 2019-03-29 2021-01-16 美商艾特加有限責任公司 Antibody molecules to fgf23 and uses thereof
WO2020208124A1 (en) 2019-04-12 2020-10-15 F. Hoffmann-La Roche Ag Treatment of cancer using a cea cd3 bispecific antibody and a wnt signaling inhibitor
CN113677403A (en) 2019-04-12 2021-11-19 豪夫迈·罗氏有限公司 Bispecific antigen binding molecules comprising lipocalin muteins
CN114364703A (en) 2019-04-19 2022-04-15 豪夫迈·罗氏有限公司 Anti-merk antibodies and methods of use thereof
TW202106876A (en) 2019-04-19 2021-02-16 日商中外製藥股份有限公司 Chimeric receptor recognizing modification site of antibody
RU2734432C1 (en) 2019-04-23 2020-10-16 Закрытое Акционерное Общество "Биокад" Monoclonal antibody which specifically binds gitr
AU2020270376A1 (en) 2019-05-03 2021-10-07 Genentech, Inc. Methods of treating cancer with an anti-PD-L1 antibody
JP2022532192A (en) 2019-05-10 2022-07-13 武田薬品工業株式会社 Antibody drug conjugate
JP2022532217A (en) 2019-05-14 2022-07-13 ウェアウルフ セラピューティクス, インコーポレイテッド Separation part and how to use it
JP2022536602A (en) 2019-05-14 2022-08-18 ジェネンテック, インコーポレイテッド Methods of using anti-CD79B immunoconjugates to treat follicular lymphoma
WO2020232295A1 (en) 2019-05-16 2020-11-19 Procisedx Inc. An assay method for the detection of vcam-1 and alpha-2-macroglobulin in blood
WO2020232262A1 (en) 2019-05-16 2020-11-19 Procisedx Inc. Assay detection methods for vcam-1 and calprotectin
WO2020236528A1 (en) 2019-05-23 2020-11-26 Procisedx Inc. Assay methods for the detection of human serum albumin, vitamin d, c-reactive protein, and anti-transglutaminase autoantibody
AR119264A1 (en) 2019-06-05 2021-12-09 Genentech Inc METHOD FOR REUSE OF CHROMATOGRAPHY
WO2020247159A1 (en) 2019-06-06 2020-12-10 Procisedx Inc. DETECTION OF HEMOGLOBIN A1C (HbA1c) IN BLOOD
US20200392229A1 (en) 2019-06-11 2020-12-17 Alector Llc Methods of use of anti-sortilin antibodies
WO2020263450A1 (en) 2019-06-25 2020-12-30 Procisedx Inc. Detection of anti-tnf alpha drug biologics and anti-drug antibodies
TW202115124A (en) 2019-06-26 2021-04-16 瑞士商赫孚孟拉羅股份公司 Novel antigen binding molecules binding to cea
WO2020260326A1 (en) 2019-06-27 2020-12-30 F. Hoffmann-La Roche Ag Novel icos antibodies and tumor-targeted antigen binding molecules comprising them
MX2021015694A (en) 2019-06-28 2022-03-11 Genentech Inc Composition and methods for stabilizing liquid protein formulations.
EP3994169A1 (en) 2019-07-02 2022-05-11 F. Hoffmann-La Roche AG Immunoconjugates comprising a mutant interleukin-2 and an anti-cd8 antibody
AR119382A1 (en) 2019-07-12 2021-12-15 Hoffmann La Roche PRE-TARGETING ANTIBODIES AND METHODS OF USE
JPWO2021010326A1 (en) 2019-07-12 2021-01-21
AR119393A1 (en) 2019-07-15 2021-12-15 Hoffmann La Roche ANTIBODIES THAT BIND NKG2D
MX2022000726A (en) 2019-07-19 2022-02-10 Oncoresponse Inc Immunomodulatory antibodies and methods of use thereof.
JP2022543553A (en) 2019-07-31 2022-10-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibody that binds to GPRC5D
CA3145885A1 (en) 2019-07-31 2021-02-04 Jeonghoon Sun Anti-ms4a4a antibodies and methods of use thereof
JP2022543551A (en) 2019-07-31 2022-10-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Antibody that binds to GPRC5D
KR20220062304A (en) 2019-09-12 2022-05-16 제넨테크, 인크. Compositions and methods for treating lupus nephritis
US20210130492A1 (en) 2019-09-18 2021-05-06 Genentech, Inc. Anti-klk7 antibodies, anti-klk5 antibodies, multispecific anti-klk5/klk7 antibodies, and methods of use
KR20220066295A (en) 2019-09-20 2022-05-24 제넨테크, 인크. Dosing of Anti-Tryptase Antibodies
CR20220127A (en) 2019-09-27 2022-05-27 Genentech Inc Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
CN114829401A (en) 2019-09-27 2022-07-29 南京金斯瑞生物科技有限公司 anti-VHH domain antibodies and uses thereof
JP7413519B2 (en) 2019-10-18 2024-01-15 ジェネンテック, インコーポレイテッド Methods of using anti-CD79B immunoconjugates to treat diffuse large B-cell lymphoma
MX2022004769A (en) 2019-10-21 2022-05-16 Novartis Ag Tim-3 inhibitors and uses thereof.
CN114786679A (en) 2019-10-21 2022-07-22 诺华股份有限公司 Combination therapy with Vernetork and TIM-3 inhibitors
AU2020381349A1 (en) 2019-11-05 2022-04-28 F. Hoffmann-La Roche Ag Treatment of cancer using a HLA-A2/WT1 x CD3 bispecific antibody and lenalidomide
US20220389103A1 (en) 2019-11-06 2022-12-08 Genentech, Inc. Diagnostic and therapeutic methods for treatment of hematologic cancers
EP4065158A2 (en) 2019-11-26 2022-10-05 Novartis AG Chimeric antigen receptors binding bcma and cd19 and uses thereof
CN115003699A (en) 2019-12-05 2022-09-02 艾利妥 Methods of use of anti-TREM 2 antibodies
AU2020403021A1 (en) 2019-12-12 2022-06-23 Alector Llc Methods of use of anti-CD33 antibodies
PE20221511A1 (en) 2019-12-13 2022-10-04 Genentech Inc ANTI-LY6G6D ANTIBODIES AND METHODS OF USE
BR112022011570A2 (en) 2019-12-13 2022-12-13 Alector Llc ANTI-MERTK ANTIBODIES AND METHODS OF THEIR USE
PE20221282A1 (en) 2019-12-18 2022-09-05 Hoffmann La Roche ANTIBODIES THAT BIND HLA-A2/MAGE-A4
WO2021123996A1 (en) 2019-12-20 2021-06-24 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
KR20220118527A (en) 2019-12-23 2022-08-25 제넨테크, 인크. Apolipoprotein L1-specific antibodies and methods of use
US20230058982A1 (en) 2019-12-27 2023-02-23 Chugai Seiyaku Kabushiki Kaisha Anti-ctla-4 antibody and use thereof
CA3162009A1 (en) 2020-01-09 2021-07-15 F. Hoffmann-La Roche Ag New 4-1bbl trimer-containing antigen binding molecules
CN110818795B (en) 2020-01-10 2020-04-24 上海复宏汉霖生物技术股份有限公司 anti-TIGIT antibodies and methods of use
CN114980902A (en) 2020-01-17 2022-08-30 诺华股份有限公司 Combination comprising a TIM-3 inhibitor and a hypomethylated drug for the treatment of myelodysplastic syndrome or chronic myelomonocytic leukemia
EP4090762A1 (en) 2020-01-17 2022-11-23 Becton, Dickinson and Company Methods and compositions for single cell secretomics
WO2021194481A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
WO2022050954A1 (en) 2020-09-04 2022-03-10 Genentech, Inc. Dosing for treatment with anti-tigit and anti-pd-l1 antagonist antibodies
TW202142257A (en) 2020-01-31 2021-11-16 美商建南德克公司 Methods of inducing neoepitope-specific t cells with a pd-1 axis binding antagonist and an rna vaccine
CN115768800A (en) 2020-01-31 2023-03-07 克利夫兰诊所基金会 Anti-mullerian hormone receptor 2 antibodies and methods of use
WO2021155006A1 (en) 2020-01-31 2021-08-05 Les Laboratoires Servier Sas Inhibitors of cyclin-dependent kinases and uses thereof
CN115427453A (en) 2020-02-10 2022-12-02 上海诗健生物科技有限公司 CLDN18.2 antibodies and uses thereof
EP4105238A4 (en) 2020-02-10 2024-03-27 Shanghai Escugen Biotechnology Co Ltd Claudin 18.2 antibody and use thereof
TW202144395A (en) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 Anti-CD137 antigen-binding molecule for use in cancer treatment
WO2021173565A1 (en) 2020-02-24 2021-09-02 Alector Llc Methods of use of anti-trem2 antibodies
EP4110377A2 (en) 2020-02-27 2023-01-04 Novartis AG Methods of making chimeric antigen receptor-expressing cells
WO2021183849A1 (en) 2020-03-13 2021-09-16 Genentech, Inc. Anti-interleukin-33 antibodies and uses thereof
EP4121163A1 (en) 2020-03-19 2023-01-25 Genentech, Inc. Isoform-selective anti-tgf-beta antibodies and methods of use
US11365239B2 (en) 2020-03-20 2022-06-21 Tsb Therapeutics (Beijing) Co., Ltd. Anti-SARS-COV-2 antibodies and uses thereof
CA3169967A1 (en) 2020-03-24 2021-09-30 Genentech, Inc. Tie2-binding agents and methods of use
JP2023518841A (en) 2020-03-26 2023-05-08 ジェネンテック, インコーポレイテッド Modified mammalian cells with reduced host cell proteins
JP2023519776A (en) 2020-03-31 2023-05-15 中外製薬株式会社 Methods for Producing Multispecific Antigen-Binding Molecules
EP4126937A1 (en) 2020-03-31 2023-02-08 Alector LLC Anti-mertk antibodies and methods of use thereof
AR121706A1 (en) 2020-04-01 2022-06-29 Hoffmann La Roche OX40 AND FAP-TARGETED BSPECIFIC ANTIGEN-BINDING MOLECULES
CA3170570A1 (en) 2020-04-01 2021-10-07 James J. KOBIE Monoclonal antibodies against the hemagglutinin (ha) and neuraminidase (na) of influenza h3n2 viruses
CN115698717A (en) 2020-04-03 2023-02-03 基因泰克公司 Methods of treatment and diagnosis of cancer
CA3172451A1 (en) 2020-04-03 2021-10-07 Robert Paul Methods of use of anti-trem2 antibodies
JP2023521785A (en) 2020-04-09 2023-05-25 テクニッシェ ウニベルシタット ミュンヘン Targeted delivery of miR-21 inhibitors to macrophages for treatment of pulmonary fibrosis
KR20230004494A (en) 2020-04-15 2023-01-06 에프. 호프만-라 로슈 아게 immunoconjugate
BR112022020753A2 (en) 2020-04-15 2022-12-20 Voyager Therapeutics Inc TAU-BINDING COMPOUNDS
MX2022013173A (en) 2020-04-24 2022-11-30 Hoffmann La Roche Enzyme and pathway modulation with sulfhydryl compounds and their derivatives.
TW202206111A (en) 2020-04-24 2022-02-16 美商建南德克公司 Methods of using anti-cd79b immunoconjugates
US20230167198A1 (en) 2020-04-27 2023-06-01 The Regents Of The University Of California Isoform-independent antibodies to lipoprotein(a)
EP4143345A1 (en) 2020-04-28 2023-03-08 Genentech, Inc. Methods and compositions for non-small cell lung cancer immunotherapy
US11634477B2 (en) 2020-04-28 2023-04-25 The Rockefeller University Neutralizing anti-SARS-CoV-2 antibodies and methods of use thereof
WO2021222533A1 (en) 2020-04-30 2021-11-04 Procisedx Inc. Methods of detecting antibodies to sars-cov-2
TW202200212A (en) 2020-05-03 2022-01-01 中國大陸商聯寧(蘇州)生物製藥有限公司 Antibody-drug conjugates comprising an anti-trop-2 antibody
US20230212260A1 (en) 2020-05-15 2023-07-06 Apogenix Ag Multi-specific immune modulators
BR112022024339A2 (en) 2020-05-29 2022-12-27 23Andme Inc ANTI CD200R1 ANTIBODIES AND METHODS OF THEIR USE
US20230340081A1 (en) 2020-06-08 2023-10-26 Hoffmann-La Roche Inc. Anti-hbv antibodies and methods of use
MX2022015651A (en) 2020-06-11 2023-01-16 Genentech Inc Nanolipoprotein-polypeptide conjugates and compositions, systems, and methods using same.
EP4165415A1 (en) 2020-06-12 2023-04-19 Genentech, Inc. Methods and compositions for cancer immunotherapy
KR20230025691A (en) 2020-06-16 2023-02-22 제넨테크, 인크. Methods and compositions for treating triple negative breast cancer
EP4168118A1 (en) 2020-06-18 2023-04-26 Genentech, Inc. Treatment with anti-tigit antibodies and pd-1 axis binding antagonists
WO2021255146A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and cea
AU2021291011A1 (en) 2020-06-19 2023-01-05 F. Hoffmann-La Roche Ag Antibodies binding to CD3 and CD19
AU2021291407A1 (en) 2020-06-19 2022-09-29 F. Hoffmann-La Roche Ag Antibodies binding to CD3
WO2021255143A1 (en) 2020-06-19 2021-12-23 F. Hoffmann-La Roche Ag Antibodies binding to cd3 and folr1
AR122659A1 (en) 2020-06-19 2022-09-28 Hoffmann La Roche BISPECIFIC ANTIBODIES TO PROTEASE-ACTIVATED T-LYMPHOCYTES
CN116234824A (en) 2020-06-22 2023-06-06 阿尔米雷尔有限公司 anti-IL-36 antibodies and methods of use thereof
US20220017637A1 (en) 2020-06-23 2022-01-20 Hoffmann-La Roche Inc. Agonistic cd28 antigen binding molecules targeting her2
WO2021260528A1 (en) 2020-06-23 2021-12-30 Novartis Ag Dosing regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
WO2021262783A1 (en) 2020-06-24 2021-12-30 Genentech, Inc. Apoptosis resistant cell lines
CN115916830A (en) 2020-06-25 2023-04-04 豪夫迈·罗氏有限公司 anti-CD 3/anti-CD 28 bispecific antigen binding molecules
CR20230076A (en) 2020-07-10 2023-03-13 Hoffmann La Roche Antibodies which bind to cancer cells and target radionuclides to said cells
WO2022015726A1 (en) 2020-07-13 2022-01-20 Genentech, Inc. Cell-based methods for predicting polypeptide immunogenicity
TW202216761A (en) 2020-07-16 2022-05-01 瑞士商諾華公司 Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
AU2021308653A1 (en) 2020-07-17 2023-02-16 Genentech, Inc. Anti-Notch2 antibodies and methods of use
BR112023001143A2 (en) 2020-07-21 2023-02-14 Genentech Inc CONJUGATE, COMPOUND, PHARMACEUTICAL COMPOSITION, METHODS TO TREAT A DISEASE AND REDUCE THE LEVEL OF A TARGET BRM PROTEIN IN AN INDIVIDUAL
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
GB2597532A (en) 2020-07-28 2022-02-02 Femtogenix Ltd Cytotoxic compounds
CN116134027A (en) 2020-08-03 2023-05-16 诺华股份有限公司 Heteroaryl-substituted 3- (1-oxo-isoindolin-2-yl) piperidine-2, 6-dione derivatives and uses thereof
JP2023537683A (en) 2020-08-07 2023-09-05 ジェネンテック, インコーポレイテッド T cell-based methods for predicting polypeptide immunogenicity
KR20230056766A (en) 2020-08-28 2023-04-27 제넨테크, 인크. CRISPR/Cas9 multiple knockout of host cell proteins
WO2022043557A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
WO2022043558A1 (en) 2020-08-31 2022-03-03 Advanced Accelerator Applications International Sa Method of treating psma-expressing cancers
MX2023002901A (en) 2020-09-14 2023-06-01 Ichnos Sciences SA Antibodies that bind to il1rap and uses thereof.
CN116322691A (en) 2020-09-24 2023-06-23 豪夫迈·罗氏有限公司 Prevention or alleviation of T cell bispecific antibody related adverse reactions
CN116406291A (en) 2020-10-05 2023-07-07 基因泰克公司 Administration of treatment with anti-FCRH 5/anti-CD 3 bispecific antibodies
JP2023545566A (en) 2020-10-20 2023-10-30 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy with PD-1 axis binding antagonist and LRRK2 inhibitor
TW202233671A (en) 2020-10-20 2022-09-01 美商建南德克公司 Peg-conjugated anti-mertk antibodies and methods of use
TW202233672A (en) 2020-10-22 2022-09-01 美商健生生物科技公司 Proteins comprising delta-like ligand 3 (dll3) antigen binding regions and their uses
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
EP4237450A1 (en) 2020-10-30 2023-09-06 F. Hoffmann-La Roche AG Treatment of cancer using a cea cd3 bispecific antibody and a tgfbeta signaling inhibitor
US20220162329A1 (en) 2020-11-04 2022-05-26 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
JP2023548064A (en) 2020-11-04 2023-11-15 ジェネンテック, インコーポレイテッド Administration for treatment with anti-CD20/anti-CD3 bispecific antibody and anti-CD79B antibody drug conjugate
EP4240766A2 (en) 2020-11-04 2023-09-13 Genentech, Inc. Subcutaneous dosing of anti-cd20/anti-cd3 bispecific antibodies
AR123997A1 (en) 2020-11-04 2023-02-01 Univ Rockefeller NEUTRALIZING ANTIBODIES AGAINST SARS-CoV-2
EP4240765A2 (en) 2020-11-06 2023-09-13 Novartis AG Antibody fc variants
TW202233248A (en) 2020-11-08 2022-09-01 美商西健公司 Combination therapy
WO2022097117A1 (en) 2020-11-09 2022-05-12 Takeda Pharmaceutical Company Ltd. Antibody drug conjugates
AU2021380807A1 (en) 2020-11-10 2023-04-20 F. Hoffmann-La Roche Ag Prevention or mitigation of T-cell engaging agent-related adverse effects
MX2023005609A (en) 2020-11-13 2023-05-29 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells.
IL302740A (en) 2020-11-16 2023-07-01 Hoffmann La Roche Fab high mannose glycoforms
EP4244254A1 (en) 2020-11-16 2023-09-20 F. Hoffmann-La Roche AG Combination therapy with fap-targeted cd40 agonists
AU2021392039A1 (en) 2020-12-02 2023-06-29 Alector Llc Methods of use of anti-sortilin antibodies
CN116635403A (en) 2020-12-04 2023-08-22 豪夫迈·罗氏有限公司 PH-dependent mutant interleukin-2 polypeptides
BR112023008265A2 (en) 2020-12-07 2024-02-06 UCB Biopharma SRL ANTIBODIES AGAINST INTERLEUKIN-22
MX2023006650A (en) 2020-12-07 2023-06-21 UCB Biopharma SRL Multi-specific antibodies and antibody combinations.
KR20230120665A (en) 2020-12-17 2023-08-17 에프. 호프만-라 로슈 아게 Anti-HLA-G Antibodies and Uses Thereof
JP2024502832A (en) 2020-12-31 2024-01-23 アラマー バイオサイエンシーズ, インコーポレイテッド Binding agent molecules with high affinity and/or specificity and methods for their production and use
MX2023007846A (en) 2021-01-06 2023-07-07 F Hoffmann La Roche Ag Combination therapy employing a pd1-lag3 bispecific antibody and a cd20 t cell bispecific antibody.
AU2022207615A1 (en) 2021-01-12 2023-07-13 F. Hoffmann-La Roche Ag Split antibodies which bind to cancer cells and target radionuclides to said cells
EP4277668A1 (en) 2021-01-13 2023-11-22 F. Hoffmann-La Roche AG Combination therapy
US20220227844A1 (en) 2021-01-15 2022-07-21 The Rockefeller University Neutralizing anti-sars-cov-2 antibodies
JP2024503724A (en) 2021-01-20 2024-01-26 オンコレスポンス,インク. Immunomodulatory antibodies and their uses
KR20230147092A (en) 2021-01-22 2023-10-20 바이원큐어 테라퓨틱스, 인크. Anti-HER-2/TROP-2 constructs and uses thereof
JP2024505049A (en) 2021-01-29 2024-02-02 ノバルティス アーゲー Administration modes for anti-CD73 and anti-ENTPD2 antibodies and their uses
TW202317612A (en) 2021-03-01 2023-05-01 美商艾希利歐發展股份有限公司 Combination of ctla4 and pd1/pdl1 antibodies for treating cancer
EP4301781A1 (en) 2021-03-01 2024-01-10 Xilio Development, Inc. Combination of masked ctla4 and pd1/pdl1 antibodies for treating cancer
JP2024509169A (en) 2021-03-03 2024-02-29 ソレント・セラピューティクス・インコーポレイテッド Antibody-drug conjugates including anti-BCMA antibodies
AR125074A1 (en) 2021-03-12 2023-06-07 Genentech Inc ANTI-KLK7 ANTIBODIES, ANTI-KLK5 ANTIBODIES, ANTI-KLK5/KLK7 MULTI-SPECIFIC ANTIBODIES AND METHODS OF USE
IL305283A (en) 2021-03-15 2023-10-01 Genentech Inc Compositions and methods of treating lupus nephritis
WO2022197947A1 (en) 2021-03-18 2022-09-22 Alector Llc Anti-tmem106b antibodies and methods of use thereof
WO2022197877A1 (en) 2021-03-19 2022-09-22 Genentech, Inc. Methods and compositions for time delayed bio-orthogonal release of cytotoxic agents
EP4314063A1 (en) 2021-03-23 2024-02-07 Alector LLC Anti-tmem106b antibodies for treating and preventing coronavirus infections
GB202104104D0 (en) 2021-03-24 2021-05-05 Liliumx Ltd Platform and method
AU2022246275A1 (en) 2021-03-26 2023-11-09 Janssen Biotech, Inc. Humanized antibodies against paired helical filament tau and uses thereof
CN117157312A (en) 2021-03-30 2023-12-01 豪夫迈·罗氏有限公司 Protease-activated polypeptides
TW202304979A (en) 2021-04-07 2023-02-01 瑞士商諾華公司 USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
WO2022217026A1 (en) 2021-04-09 2022-10-13 Seagen Inc. Methods of treating cancer with anti-tigit antibodies
AR125344A1 (en) 2021-04-15 2023-07-05 Chugai Pharmaceutical Co Ltd ANTI-C1S ANTIBODY
TW202305122A (en) 2021-04-19 2023-02-01 美商建南德克公司 Modified mammalian cells
CN117222412A (en) 2021-04-23 2023-12-12 豪夫迈·罗氏有限公司 Prevention or alleviation of NK cell binding agent-related adverse reactions
EP4330282A1 (en) 2021-04-30 2024-03-06 F. Hoffmann-La Roche AG Dosing for combination treatment with anti-cd20/anti-cd3 bispecific antibody and anti-cd79b antibody drug conjugate
AU2021443863A1 (en) 2021-04-30 2023-10-26 F. Hoffmann-La Roche Ag Dosing for treatment with anti-cd20/anti-cd3 bispecific antibody
AU2022268545A1 (en) 2021-05-03 2023-11-02 UCB Biopharma SRL Antibodies
WO2022235867A2 (en) 2021-05-06 2022-11-10 The Rockefeller University Neutralizing anti-sars- cov-2 antibodies and methods of use thereof
IL308351A (en) 2021-05-12 2024-01-01 Genentech Inc Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
BR112023023777A2 (en) 2021-05-14 2024-01-30 Genentech Inc ISOLATED ANTIBODIES, PHARMACEUTICAL COMPOSITION, ISOLATED NUCLEIC ACID, ISOLATED VECTOR, ISOLATED HOST CELL, METHOD FOR PRODUCING AN ANTIBODY, METHOD FOR TREATING A CONDITION ASSOCIATED WITH LOSS OF TREM2 FUNCTION, METHOD FOR REDUCING STREM2 LEVELS AND USE OF AN ANTIBODY
AR125874A1 (en) 2021-05-18 2023-08-23 Novartis Ag COMBINATION THERAPIES
WO2022243261A1 (en) 2021-05-19 2022-11-24 F. Hoffmann-La Roche Ag Agonistic cd40 antigen binding molecules targeting cea
KR20240010469A (en) 2021-05-21 2024-01-23 제넨테크, 인크. Modified cells for production of recombinant products of interest
AR126009A1 (en) 2021-06-02 2023-08-30 Hoffmann La Roche CD28 ANTIGEN-BINDING AGONIST MOLECULES THAT TARGET EPCAM
TW202306994A (en) 2021-06-04 2023-02-16 日商中外製藥股份有限公司 Anti-ddr2 antibodies and uses thereof
KR20240019109A (en) 2021-06-09 2024-02-14 에프. 호프만-라 로슈 아게 Combination of a specific BRAF inhibitor (Paradox Break) and a PD-1 axis binding antagonist for use in the treatment of cancer
WO2022266223A1 (en) 2021-06-16 2022-12-22 Alector Llc Bispecific anti-mertk and anti-pdl1 antibodies and methods of use thereof
EP4355783A1 (en) 2021-06-16 2024-04-24 Alector LLC Monovalent anti-mertk antibodies and methods of use thereof
WO2022270612A1 (en) 2021-06-25 2022-12-29 中外製薬株式会社 Use of anti-ctla-4 antibody
JP7472405B2 (en) 2021-06-25 2024-04-22 中外製薬株式会社 Anti-CTLA-4 antibody
BR112023026966A2 (en) 2021-07-02 2024-03-12 Hoffmann La Roche METHODS FOR TREATING AN INDIVIDUAL WITH MELANOMA, FOR ACHIEVING A CLINICAL RESPONSE, FOR TREATING AN INDIVIDUAL WITH NON-HODGKIN LYMPHOMA, FOR TREATING A POPULATION OF INDIVIDUALS WITH NON-HODGKIN LYMPHOMA, AND FOR TREATING AN INDIVIDUAL WITH METASTATIC COLORECTAL CANCER
TW202309097A (en) 2021-07-14 2023-03-01 美商建南德克公司 Anti-c-c motif chemokine receptor 8 (ccr8) antibodies and methods of use
WO2023004386A1 (en) 2021-07-22 2023-01-26 Genentech, Inc. Brain targeting compositions and methods of use thereof
WO2023001884A1 (en) 2021-07-22 2023-01-26 F. Hoffmann-La Roche Ag Heterodimeric fc domain antibodies
AU2022317820A1 (en) 2021-07-28 2023-12-14 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
WO2023012147A1 (en) 2021-08-03 2023-02-09 F. Hoffmann-La Roche Ag Bispecific antibodies and methods of use
WO2023019239A1 (en) 2021-08-13 2023-02-16 Genentech, Inc. Dosing for anti-tryptase antibodies
AU2022332285A1 (en) 2021-08-23 2024-02-15 Immunitas Therapeutics, Inc. Anti-cd161 antibodies and uses thereof
WO2023028591A1 (en) 2021-08-27 2023-03-02 Genentech, Inc. Methods of treating tau pathologies
TW202325727A (en) 2021-08-30 2023-07-01 美商建南德克公司 Anti-polyubiquitin multispecific antibodies
WO2023044483A2 (en) 2021-09-20 2023-03-23 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
WO2023056069A1 (en) 2021-09-30 2023-04-06 Angiex, Inc. Degrader-antibody conjugates and methods of using same
TW202321308A (en) 2021-09-30 2023-06-01 美商建南德克公司 Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
AU2022362681A1 (en) 2021-10-14 2024-04-04 F. Hoffmann-La Roche Ag New interleukin-7 immunoconjugates
WO2023062048A1 (en) 2021-10-14 2023-04-20 F. Hoffmann-La Roche Ag Alternative pd1-il7v immunoconjugates for the treatment of cancer
WO2023069919A1 (en) 2021-10-19 2023-04-27 Alector Llc Anti-cd300lb antibodies and methods of use thereof
TW202325742A (en) 2021-11-01 2023-07-01 瑞士商赫孚孟拉羅股份公司 Treatment of cancer using a hla-a2/wt1 x cd3 bispecific antibody and a 4-1bb (cd137) agonist
WO2023081898A1 (en) 2021-11-08 2023-05-11 Alector Llc Soluble cd33 as a biomarker for anti-cd33 efficacy
WO2023086807A1 (en) 2021-11-10 2023-05-19 Genentech, Inc. Anti-interleukin-33 antibodies and uses thereof
TW202337494A (en) 2021-11-16 2023-10-01 美商建南德克公司 Methods and compositions for treating systemic lupus erythematosus (sle) with mosunetuzumab
WO2023088889A1 (en) 2021-11-16 2023-05-25 Apogenix Ag CD137 ligands
WO2023088876A1 (en) 2021-11-16 2023-05-25 Apogenix Ag Multi-specific immune modulators
WO2023092004A1 (en) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions and methods for the treatment of tau-related disorders
US20230312724A1 (en) 2021-12-01 2023-10-05 Kadmon Corporation, Llc B7-H4 Antibodies and Anti-B7-H4 Antibody/IL-15 Fusion Proteins
AR127887A1 (en) 2021-12-10 2024-03-06 Hoffmann La Roche ANTIBODIES THAT BIND CD3 AND PLAP
TW202339797A (en) 2021-12-14 2023-10-16 瑞士商赫孚孟拉羅股份公司 Treatment of cancer using a hla-a2/mage-a4 x cd3 bispecific antibody and a 4-1bb (cd137) agonist
WO2023114544A1 (en) 2021-12-17 2023-06-22 Dana-Farber Cancer Institute, Inc. Antibodies and uses thereof
WO2023114543A2 (en) 2021-12-17 2023-06-22 Dana-Farber Cancer Institute, Inc. Platform for antibody discovery
AR128031A1 (en) 2021-12-20 2024-03-20 Hoffmann La Roche ANTI-LTBR AGONIST ANTIBODIES AND BISPECIFIC ANTIBODIES THAT COMPRISE THEM
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
US20230322958A1 (en) 2022-01-19 2023-10-12 Genentech, Inc. Anti-Notch2 Antibodies and Conjugates and Methods of Use
WO2023147329A1 (en) 2022-01-26 2023-08-03 Genentech, Inc. Antibody-conjugated chemical inducers of degradation and methods thereof
WO2023147328A1 (en) 2022-01-26 2023-08-03 Genentech, Inc. Antibody-conjugated chemical inducers of degradation with hydolysable maleimide linkers and methods thereof
WO2023150778A1 (en) 2022-02-07 2023-08-10 Visterra, Inc. Anti-idiotype antibody molecules and uses thereof
WO2023164516A1 (en) 2022-02-23 2023-08-31 Alector Llc Methods of use of anti-trem2 antibodies
WO2023173026A1 (en) 2022-03-10 2023-09-14 Sorrento Therapeutics, Inc. Antibody-drug conjugates and uses thereof
TW202345899A (en) 2022-03-11 2023-12-01 比利時商健生藥品公司 Multispecific antibodies and uses thereof
WO2023170291A1 (en) 2022-03-11 2023-09-14 Janssen Pharmaceutica Nv Multispecific antibodies and uses thereof
WO2023170290A1 (en) 2022-03-11 2023-09-14 Janssen Pharmaceutica Nv Multispecific antibodies and uses thereof
TW202346365A (en) 2022-03-23 2023-12-01 瑞士商赫孚孟拉羅股份公司 Combination treatment of an anti-cd20/anti-cd3 bispecific antibody and chemotherapy
TW202402790A (en) 2022-03-25 2024-01-16 英商梅迪繆思有限公司 Methods for reducing respiratory infections
WO2023186756A1 (en) 2022-03-28 2023-10-05 F. Hoffmann-La Roche Ag Interferon gamma variants and antigen binding molecules comprising these
WO2023192622A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Hydroxypropyl methyl cellulose derivatives to stabilize polypeptides
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023198727A1 (en) 2022-04-13 2023-10-19 F. Hoffmann-La Roche Ag Pharmaceutical compositions of anti-cd20/anti-cd3 bispecific antibodies and methods of use
US20230406930A1 (en) 2022-04-13 2023-12-21 Genentech, Inc. Pharmaceutical compositions of therapeutic proteins and methods of use
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023215737A1 (en) 2022-05-03 2023-11-09 Genentech, Inc. Anti-ly6e antibodies, immunoconjugates, and uses thereof
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023220695A2 (en) 2022-05-13 2023-11-16 Voyager Therapeutics, Inc. Compositions and methods for the treatment of her2 positive cancer
US20230416412A1 (en) 2022-05-31 2023-12-28 Hoffmann-La Roche Inc. Prevention or mitigation of t-cell engaging agent-related adverse effects
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2023239803A1 (en) 2022-06-08 2023-12-14 Angiex, Inc. Anti-tm4sf1 antibody-drug conjugates comprising cleavable linkers and methods of using same
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024015993A1 (en) 2022-07-15 2024-01-18 Fibrogen, Inc. Modified anti-galectin-9 antibody and uses thereof
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020579A1 (en) 2022-07-22 2024-01-25 Bristol-Myers Squibb Company Antibodies binding to human pad4 and uses thereof
WO2024020564A1 (en) 2022-07-22 2024-01-25 Genentech, Inc. Anti-steap1 antigen-binding molecules and uses thereof
WO2024026472A2 (en) 2022-07-29 2024-02-01 Alector Llc Transferrin receptor antigen-binding domains and uses therefor
WO2024026471A1 (en) 2022-07-29 2024-02-01 Alector Llc Cd98hc antigen-binding domains and uses therefor
WO2024026447A1 (en) 2022-07-29 2024-02-01 Alector Llc Anti-gpnmb antibodies and methods of use thereof
WO2024030976A2 (en) 2022-08-03 2024-02-08 Voyager Therapeutics, Inc. Compositions and methods for crossing the blood brain barrier
WO2024040216A2 (en) 2022-08-19 2024-02-22 Fibrogen, Inc. Anti-ccr8 antibodies and uses thereof
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024059739A1 (en) 2022-09-15 2024-03-21 Voyager Therapeutics, Inc. Tau binding compounds
WO2024056861A1 (en) 2022-09-15 2024-03-21 Avidicure Ip B.V. Multispecific antigen binding proteins for stimulating nk cells and use thereof
WO2024069180A2 (en) 2022-09-28 2024-04-04 LiliumX Ltd. Multivalent proteins and screening methods
WO2024068705A1 (en) 2022-09-29 2024-04-04 F. Hoffmann-La Roche Ag Protease-activated polypeptides
WO2024074649A1 (en) 2022-10-05 2024-04-11 Alcea Therapeutics, Inc. Notch4 antibodies, compositions, and methods for treating airway inflammation
WO2024077239A1 (en) 2022-10-07 2024-04-11 Genentech, Inc. Methods of treating cancer with anti-c-c motif chemokine receptor 8 (ccr8) antibodies
WO2024079010A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and cd38 antibodies
WO2024079015A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and imids
WO2024079009A1 (en) 2022-10-10 2024-04-18 F. Hoffmann-La Roche Ag Combination therapy of a gprc5d tcb and proteasome inhibitors
WO2024079310A1 (en) 2022-10-14 2024-04-18 Ebbil, Ltd. Sil-6r and ctgf binding proteins and methods of use thereof

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4132405A (en) * 1977-02-16 1979-01-02 Asher Nathan F Balanced swivel exercising device
US4472509A (en) * 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4479930A (en) * 1982-07-26 1984-10-30 Trustees Of The University Of Massachusetts Amines coupled wth dicyclic dianhydrides capable of being radiolabeled product
US4753894A (en) * 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5258498A (en) * 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5260203A (en) * 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US5534254A (en) * 1992-02-06 1996-07-09 Chiron Corporation Biosynthetic binding proteins for immuno-targeting
US5571894A (en) * 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) * 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US6054561A (en) * 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
US6207804B1 (en) * 1987-05-21 2001-03-27 Curis, Inc. Genetically engineered antibody analogues and fusion proteins thereof

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169774A (en) 1984-02-08 1992-12-08 Cetus Oncology Corporation Monoclonal anti-human breast cancer antibodies
US4938948A (en) * 1985-10-07 1990-07-03 Cetus Corporation Method for imaging breast tumors using labeled monoclonal anti-human breast cancer antibodies
JPS6360942A (en) * 1986-08-01 1988-03-17 シタス コ−ポレイシヨン Combination remedy using immunotoxin or antibody together with interleukin-2
US5132405A (en) * 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
IL162181A (en) * 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
DE69027210T2 (en) * 1989-04-21 1997-01-23 Us Health RECOMBINANT ANTIBODY TOXIN FUSION PROTEIN
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
AU662311B2 (en) * 1991-02-05 1995-08-31 Novartis Ag Recombinant antibodies specific for a growth factor receptor
CA2100671C (en) * 1991-02-27 1999-02-02 James S. Huston Serine rich peptide linkers
US7018809B1 (en) * 1991-09-19 2006-03-28 Genentech, Inc. Expression of functional antibody fragments
JPH07501451A (en) * 1991-11-25 1995-02-16 エンゾン・インコーポレイテッド Multivalent antigen binding protein
US5932448A (en) * 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
DE69309472T2 (en) * 1992-01-23 1997-10-23 Merck Patent Gmbh FUSION PROTEINS OF MONOMERS AND DIMERS OF ANTIBODY FRAGMENTS
AU4025193A (en) * 1992-04-08 1993-11-18 Cetus Oncology Corporation Humanized C-erbB-2 specific antibodies

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4132405A (en) * 1977-02-16 1979-01-02 Asher Nathan F Balanced swivel exercising device
US4472509A (en) * 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4479930A (en) * 1982-07-26 1984-10-30 Trustees Of The University Of Massachusetts Amines coupled wth dicyclic dianhydrides capable of being radiolabeled product
US4753894A (en) * 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US6054561A (en) * 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
US5260203A (en) * 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US5258498A (en) * 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US6207804B1 (en) * 1987-05-21 2001-03-27 Curis, Inc. Genetically engineered antibody analogues and fusion proteins thereof
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5571894A (en) * 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) * 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5534254A (en) * 1992-02-06 1996-07-09 Chiron Corporation Biosynthetic binding proteins for immuno-targeting
US5877305A (en) * 1992-02-06 1999-03-02 Chiron Corporation DNA encoding biosynthetic binding protein for cancer marker

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180117153A1 (en) * 2015-04-22 2018-05-03 Ucb Biopharma Sprl Method of monomerisation of recombinant antibody molecules
US10828366B2 (en) * 2015-04-22 2020-11-10 Ucb Biopharma Sprl Method of monomerisation of recombinant antibody molecules
US11786593B2 (en) 2015-04-22 2023-10-17 UCB Biopharma SRL Method of monomerisation of recombinant antibody molecules

Also Published As

Publication number Publication date
EP0625200B1 (en) 2005-05-11
ATE503496T1 (en) 2011-04-15
EP1997894B1 (en) 2011-03-30
AU3612293A (en) 1993-09-03
US20020168375A1 (en) 2002-11-14
US5837846A (en) 1998-11-17
DE69334255D1 (en) 2009-02-12
US5753204A (en) 1998-05-19
CA2129663A1 (en) 1993-08-19
WO1993016185A2 (en) 1993-08-19
CA2129663C (en) 2005-07-05
EP1997894A2 (en) 2008-12-03
DE69333807D1 (en) 2005-06-16
JPH08500962A (en) 1996-02-06
EP0625200A1 (en) 1994-11-23
ATE419355T1 (en) 2009-01-15
US5877305A (en) 1999-03-02
US5534254A (en) 1996-07-09
JP2007228979A (en) 2007-09-13
JP2005104965A (en) 2005-04-21
AU675929B2 (en) 1997-02-27
DE69334351D1 (en) 2011-05-12
JP2003284577A (en) 2003-10-07
EP1514934A2 (en) 2005-03-16
EP1514934B1 (en) 2008-12-31
DE69333807T2 (en) 2006-02-02
WO1993016185A3 (en) 1993-09-30
JP2007231027A (en) 2007-09-13
JP2005168489A (en) 2005-06-30
JP2008289500A (en) 2008-12-04
JP4236493B2 (en) 2009-03-11
ATE295420T1 (en) 2005-05-15
EP1997894A3 (en) 2009-11-04
US7138497B2 (en) 2006-11-21
EP1514934A3 (en) 2006-04-19
CA2372813A1 (en) 1993-08-19
JP2009035558A (en) 2009-02-19

Similar Documents

Publication Publication Date Title
US5534254A (en) Biosynthetic binding proteins for immuno-targeting
Cumber et al. Comparative stabilities in vitro and in vivo of a recombinant mouse antibody FvCys fragment and a bisFvCys conjugate.
Pavlinkova et al. Pharmacokinetics and biodistribution of engineered single-chain antibody constructs of MAb CC49 in colon carcinoma xenografts
Hu et al. Minibody: a novel engineered anti-carcinoembryonic antigen antibody fragment (single-chain Fv-CH3) which exhibits rapid, high-level targeting of xenografts
Whitlow et al. An improved linker for single-chain Fv with reduced aggregation and enhanced proteolytic stability
RU2464277C2 (en) Antibody and fragments of human anti-folate receptor alpha antibodies for radioimmunotherapy of ovarian carcinoma
US6071515A (en) Dimer and multimer forms of single chain polypeptides
US20180155449A1 (en) Covalent disulfide-linked diabodies and uses thereof
JP2009268470A (en) Selective targeting of tumor vasculature using antibody molecule
JPH07504334A (en) Humanized antibody against A33 antigen
CN114106190A (en) anti-VEGF/PD-L1 bispecific antibody and application thereof
Pavlinkova et al. Pharmacokinetics and biodistribution of a light-chain-shuffled CC49 single-chain Fv antibody construct
WO2023025304A1 (en) Polypeptide fusion molecule close to natural molecule
US5728821A (en) Mutant BR96 antibodies reactive with human carcinomas
US20070031931A1 (en) Biosynthetic binding proteins for immuno-targeting
US20050058638A1 (en) Biosynthetic binding proteins for immuno-targeting
CN117964777A (en) Polypeptide fusion molecule close to natural molecule
CZ2008606A3 (en) Recombinant protein adapted for direct radioiodination, preparation and use thereof
Pavlinkova et al. Pharmacokinetics and Biodistribution

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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