WO2011056983A1 - Zirconium-radiolabeled, cysteine engineered antibody conjugates - Google Patents

Zirconium-radiolabeled, cysteine engineered antibody conjugates Download PDF

Info

Publication number
WO2011056983A1
WO2011056983A1 PCT/US2010/055465 US2010055465W WO2011056983A1 WO 2011056983 A1 WO2011056983 A1 WO 2011056983A1 US 2010055465 W US2010055465 W US 2010055465W WO 2011056983 A1 WO2011056983 A1 WO 2011056983A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
cysteine
frac acc
zirconium
engineered antibody
Prior art date
Application number
PCT/US2010/055465
Other languages
French (fr)
Inventor
Herman Gill
Jagath R. Junutula
Henry B. Lowman
Jan Marik
Jeff Tinianow
Simon Williams
Original Assignee
Genentech, Inc.
F. Hoffmann-La Roche Ag
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 Genentech, Inc., F. Hoffmann-La Roche Ag filed Critical Genentech, Inc.
Priority to CA2780216A priority Critical patent/CA2780216A1/en
Priority to JP2012537224A priority patent/JP5850843B2/en
Priority to KR1020177034613A priority patent/KR20170136652A/en
Priority to EP10776064.7A priority patent/EP2496270B1/en
Priority to RU2012123007/10A priority patent/RU2562862C2/en
Priority to CN201080050134.4A priority patent/CN102596260B/en
Priority to BR112012007774A priority patent/BR112012007774A2/en
Priority to MX2012005211A priority patent/MX340674B/en
Publication of WO2011056983A1 publication Critical patent/WO2011056983A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1093Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody conjugates with carriers being antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • A61K49/14Peptides, e.g. proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • A61K49/14Peptides, e.g. proteins
    • A61K49/16Antibodies; Immunoglobulins; Fragments thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1051Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from breast, e.g. the antibody being herceptin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C259/00Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups
    • C07C259/04Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids
    • C07C259/06Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids having carbon atoms of hydroxamic groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/60Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton with the carbon atom of at least one of the carboxyl groups bound to nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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
    • C07K2317/624Disulfide-stabilized antibody (dsFv)

Definitions

  • the invention relates generally to antibodies engineered with reactive cysteine residues and more specifically to antibodies with therapeutic or diagnostic applications.
  • the cysteine engineered antibodies may be conjugated with chemotherapeutic drugs, toxins, affinity ligands such as biotin, and detection labels such as radioisotopes and fluorophores.
  • the invention also relates to methods of using antibodies and antibody-drug conjugate compounds for in vitro, in situ, and in vivo diagnosis or treatment of mammalian cells, or associated pathological conditions.
  • Immuno-positron emission tomography is a rapidly emerging method for tracking and quantifying monoclonal antibodies (mAbs) in vivo as it efficiently combines the high sensitivity of PET with the high specificity of mAbs.
  • ImmunoPET aspires to be the clinical method of choice for non-invasive diagnosis providing "comprehensive immunohistochemical staining in vivo" (van Dongen GA, et al. "ImmunoPET: a navigator in monoclonal antibody development and applications" Oncologist
  • PET Positron Emission Tomographic
  • the isotopes are typically administered to a patient by injection of probe molecules that comprise a positron-emitting isotope, such as F-18, C-l 1, N-13, or 0-15, covalently attached to a molecule that is readily metabolized or localized in the body (e.g., glucose) or that chemically binds to receptor sites within the body.
  • a positron-emitting isotope such as F-18, C-l 1, N-13, or 0-15
  • covalently attached to a molecule that is readily metabolized or localized in the body e.g., glucose
  • the isotope is administered to the patient as an ionic solution or by inhalation.
  • Small immuno-PET imaging agents such as Fab antibody fragments (50 kDa) or diabodies, paired dimers of the covalently associated V H -V L region of Mab, 55 kDa (Shively et al (2007) J Nucl Med 48: 170- 2), may be particularly useful since they exhibit a short circulation half-life, high tissue permeability, and reach an optimal tumor to background ratio between two to four hours after
  • Iodine 124 ( I) was coupled to antibody 3F9 and used to estimate the dosimetry for radioimmunotherapy of neuroblastoma (Larson SM, et al "PET scanning of iodine- 124-3 F9 as an approach to tumor dosimetry during treatment planning for radioimmunotherapy in a child with neuroblastoma" J Nucl Med 1992;33:2020-3). Later, as more sophisticated PET
  • the complex decay scheme involves energetic positrons ( ⁇ + max. 1.5 and 2.1 MeV) which negatively affects the resolution of small animal microPET.
  • a label such as a radioisotope, fluorescent dye, or drug moiety
  • cytotoxic drugs have typically been conjugated to antibodies through the often-numerous lysine residues of an antibody, generating a heterogeneous antibody-drug conjugate mixture.
  • the heterogeneous mixture typically contains a distribution of antibodies with from 0 to about 8, or more, attached drug moieties.
  • Antibodies are large, complex and structurally diverse biomolecules, often with many reactive functional groups. Their reactivities with linker reagents and drug-linker
  • Cysteine thiols are reactive at neutral pH, unlike most amines which are protonated and less nucleophilic near pH 7. Since free thiol (RSH, sulfhydryl) groups are relatively reactive, proteins with cysteine residues often exist in their oxidized form as disulfide-linked oligomers or have internally bridged disulfide groups. Extracellular proteins generally do not have free thiols (Garman, 1997, Non-Radioactive Labelling: A Practical Approach,
  • the amount of free thiol in a protein may be estimated by the standard Ellman's assay.
  • Immunoglobulin M is an example of a disulfide- linked pentamer
  • immunoglobulin G is an example of a protein with internal disulfide bridges bonding the subunits together.
  • DTT dithiothreitol
  • selenol Singh et al (2002) Anal.
  • Biochem. 304: 147-1566 is required to generate the reactive free thiol. This approach may result in loss of antibody tertiary structure and antigen binding specificity.
  • Antibody cysteine thiol groups are generally more reactive, i.e. more nucleophilic, towards electrophilic conjugation reagents than antibody amine or hydroxyl groups.
  • Cysteine residues have been introduced into proteins by genetic engineering techniques to form covalent attachments to ligands or to form new intramolecular disulfide bonds (Better et al (1994) J. Biol. Chem. 269(13):9644-9650; Bernhard et al (1994) Bioconjugate Chem. 5: 126- 132; Greenwood et al (1994) Therapeutic Immunology 1 :247-255; Tu et al (1999) Proc. Natl. Acad.
  • cysteine thiol groups by the mutation of various amino acid residues of a protein to cysteine amino acids is potentially problematic, particularly in the case of unpaired (free Cys) residues or those which are relatively accessible for reaction or oxidation.
  • Site-specific conjugation is preferred over random amino modification as it enables chemical modification of a site away from the binding site, promoting complete retention of biological activity and allowing control over the possible number of prosthetic groups added.
  • Cysteine-engineered antibodies have been designed as FAB antibody fragments (thioFab) and expressed as full-length, IgG monoclonal (thioMab) antibodies. See: US 7521541; Junutula JR et al. "Rapid identification of reactive cysteine residues for site-specific labeling of antibody-Fabs" J Immunol Methods 2008;332:41-52; Junutula JR et al. "Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index” (2008) Nat Biotechnol.
  • ThioFab and ThioMab antibodies have been conjugated through linkers at the newly introduced cysteine thiols with thiol-reactive linker reagents and drug-linker reagents to prepare cysteine- engineered antibody drug conjugates (Thio ADC) with anti-cancer properties, including anti- MUC16 (US 2008/0311134), anti-CD22 (US 2008/0050310), anti-ROB04 (US)
  • the compounds of the invention include cysteine engineered antibodies where one or more amino acids of a parent antibody are replaced with a free cysteine amino acid.
  • a cysteine engineered antibody comprises one or more free cysteine amino acids having a thiol reactivity value in the range of 0.6 to l .O.
  • a free cysteine amino acid is a cysteine residue which has been engineered into the parent antibody and is not part of a disulfide bridge.
  • Cysteine engineered antibodies may be useful in the diagnosis and treatment of cancer and include antibodies specific for cell surface and transmembrane receptors, and tumor-associated antigens (TAA). Such antibodies may be used as naked antibodies
  • Embodiments of the methods for preparing and screening a cysteine engineered antibody include where the parent antibody is an antibody fragment, such as hu4D5Fabv8.
  • the parent antibody may also be a fusion protein comprising an albumin-binding peptide sequence (ABP).
  • the parent antibody may also be a humanized antibody selected from huMAb4D5-l, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (trastuzumab).
  • Cysteine engineered antibodies of the invention may be site-specifically and efficiently coupled with a thiol-reactive reagent.
  • the thiol-reactive reagent may be a radioisotope reagent, multifunctional linker reagent, a capture label reagent, a fluorophore reagent, or a drug-linker intermediate.
  • the cysteine engineered antibody may be labeled with a detectable label, immobilized on a solid phase support and/or conjugated with a drug moiety.
  • Another aspect of the invention is a zirconium-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a zirconium complex (Z), having Formula I: where p is 1 to 4.
  • Ab cysteine engineered antibody
  • L linker
  • Z zirconium complex
  • Another aspect of the invention is a desferrioxamine-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
  • p 1 to 4.
  • Another aspect of the invention is a method of making a desferrioxamine-labelled, cysteine-engineered antibody comprising comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
  • L-Df is selected from:
  • p 1 to 4.
  • R is selected from:
  • cysteine-engineered antibody having one or more free cysteine amino acids, whereby the desferrioxamine-labelled, cysteine-engineered antibody is formed.
  • Another aspect of the invention is a method of making a zirconium-labelled, cysteine- engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a zirconium complex (Z), having Formula I: where p is 1 to 4;
  • the method comprising complexing a zirconium reagent with a desferrioxamine- labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
  • p 1 to 4.
  • Another aspect of the invention is a method of imaging comprising:
  • zirconium-labelled, cysteine-engineered antibody comprises a cysteine engineered antibody (Ab) having one or more free cysteine amino acids conjugated with one or more zirconium complex (Z) through a linker (L), and having Formula I:
  • Another aspect of the invention includes diagnostic uses for the compounds and compositions disclosed herein. BRIEF DESCRIPTION OF THE DRAWINGS
  • Figure 1 A shows a three-dimensional representation of the hu4D5Fabv7 antibody fragment derived by X-ray crystal coordinates.
  • the structure positions of the exemplary engineered Cys residues of the heavy and light chains are numbered (according to a sequential numbering system).
  • Figure IB shows a sequential numbering scheme (top row), starting at the N-terminus in comparison with the Kabat numbering scheme (bottom row) for 4D5v7fabH. Kabat numbering insertions are noted by a,b,c.
  • Figures 2 A and 2B show binding measurements with detection of absorbance at 450nm of hu4D5Fabv8 and hu4D5Fabv8 Cys mutant (ThioFab) phage variants: (A) non- biotinylated phage-hu4D5Fabv8 and (B) biotinylated phage-hu4D5Fabv8 (B) by the PHESELECTOR assay for interactions with BSA (open bar), HER2 (striped bar) or streptavidin (solid bar).
  • FIG. 4A shows Fractional Surface Accessibility values of residues on wild type
  • hu4D5Fabv8 Light chain sites are on the left side and heavy chain sites are on the right side.
  • Figure 4B shows binding measurements with detection of absorbance at 450nm of
  • Phage-hu4D5Fabv8 Cys variants were isolated and stored at 4 °C. Biotin conjugation was carried out either at day 2 or day 4 followed by PHESELECTOR analyses to monitor their interaction with Her2 and streptavidin as described in Example 2, and probe the stability of reactive thiol groups on engineered ThioFab variants.
  • Figure 5 shows binding measurements with detection of absorbance at 450nm of biotin- maleimide conjugated-hu4D5Fabv8 (A121C) and non-biotinylated wild type hu4D5Fabv8 for binding to streptavidin and HER2. Each Fab was tested at 2 ng and 20 ng.
  • Figure 6 shows ELISA analysis with detection of absorbance at 450nm of biotinylated ABP- hu4D5Fabv8 wild type (wt), and ABP-hu4D5Fabv8 cysteine mutants VI IOC and A121C for binding with rabbit albumin, streptavidin (SA), and HER2.
  • Figure 7 shows ELISA analysis with detection of absorbance at 450nm of biotinylated ABP- hu4D5Fabv8 cysteine mutants (ThioFab variants): (left to right) single Cys variants ABP-V1 IOC, ABP-A121C, and double Cys variants ABP-V110C-A88C and ABP- VI 10C-A121C for binding with rabbit albumin, HER2 and streptavidin (SA), and probing with Fab-HRP or SA-HRP.
  • ThioFab variants biotinylated ABP- hu4D5Fabv8 cysteine mutants
  • Figure 8 shows binding of biotinylated ThioFab phage and an anti-phage HRP antibody to HER2 (top) and Streptavidin (bottom).
  • Figure 13A shows a cartoon depiction of biotinylated antibody binding to immobilized HER2 with binding of HRP labeled secondary antibody for absorbance detection.
  • Figure 13B shows binding measurements with detection of absorbance at 450nm of biotin- maleimide conjugated thio-trastuzumab variants and non-biotinylated wild type trastuzumab in binding to immobilized HER2. From left to right: VI IOC (single cys), A121C (single cys), VI 10C/A121C (double cys), and trastuzumab. Each thio IgG variant and trastuzumab was tested at 1, 10, and 100 ng.
  • Figure 14A shows a cartoon depiction of biotinylated antibody binding to immobilized HER2 with binding of biotin to anti-IgG-HRP for absorbance detection.
  • Figure 14B shows binding measurements with detection of absorbance at 450nm of biotin- maleimide conjugated-thio trastuzumab variants and non-biotinylated wild type trastuzumab in binding to immobilized streptavidin. From left to right: VI IOC (single cys), A121C (single cys), VI 10C/A121C (double cys), and trastuzumab. Each thio IgG variant and trastuzumab was tested at 1, 10, and 100 ng.
  • Figure 15 shows the general process to prepare a cysteine engineered antibody (ThioMab) expressed from cell culture for conjugation.
  • Figure 16 shows non-reducing (top) and reducing (bottom) denaturing polyacrylamide gel electrophoresis analysis of 2H9 ThioMab Fc variants (left to right, lanes 1-9): A339C; S337C; S324C; A287C; V284C; V282C; V279C; V273C, and 2H9 wild type after purification on immobilized Protein A.
  • the lane on the right is a size marker ladder, indicating the intact proteins are about 150 kDa, heavy chain fragments about 50 kDa, and light chain fragments about 25 kDa.
  • Figure 17A shows non-reducing (left) and reducing (+DTT) (right) denaturing polyacrylamide gel electrophoresis analysis of 2H9 ThioMab variants (left to right, lanes 1-4): L-V15C; S179C; S375C; S400C, after purification on immobilized Protein A.
  • Figure 17B shows non-reducing (left) and reducing (+DTT) (right) denaturing
  • Figure 18 shows western blot analysis of biotinylated Thio-IgG variants. 2H9 and 3A5
  • ThioMab variants were analyzed on reduced denaturing polyacrylamide gel electrophoresis, the proteins were transferred to nitrocellulose membrane. The presence of antibody and conjugated biotin were probed with anti-IgG-HRP (top) and streptavidin-HRP (bottom), respectively. Lane 1 : 3A5 H-A121C. Lane 2: 3A5 L- VI IOC. Lane 3: 2H9 H-A121C. Lane 4: 2H9 L-V110C. Lane 5: 2H9 wild type.
  • Figure 19 shows ELISA analysis for the binding of biotinylated 2H9 variants to streptavidin by probing with anti-IgG-HRP and measuring the absorbance at 450 nm of (top bar diagram).
  • Bottom schematic diagram depicts the experimental design used in the ELISA analysis.
  • Figure 20 shows bifunctional reagents for coupling chelator of 89 Zr desferoxamine B (Df, top) with proteins using amino reactive linkers, TFP-N-SucDf and Df-Bz-NCS (center) and thiol reactive linkers, Df-Chx-Mal, Df-Bac, and Df-lac (bottom).
  • Figure 21 shows the preparation of Df-Chx-Mal, Df-Bac, Df-lac and conjugation to thio- trastuzumab via Cys residues incorporated into the heavy chain of Fab.
  • Reaction conditions i. DIEA, DMF/H 2 0 (10: 1), RT, 0.5-1 h; ii. DIEA, DMF, 0°C, 4 h; iii. pH 7.5, RT, 1 h; iv. pH 9, RT, 5 h; v. pH 9, RT, 2 h.
  • Figure 22 shows chelation of zirconium-89 oxalate with a desferrioxamine-labelled, cysteine- engineered antibody, such as variants of Df-linker-trastuzumab containing four linkers: N-Suc, Bz-SCN, Chx-maleimide (CHx-Mal), or acetyl (Ac).
  • a desferrioxamine-labelled, cysteine- engineered antibody such as variants of Df-linker-trastuzumab containing four linkers: N-Suc, Bz-SCN, Chx-maleimide (CHx-Mal), or acetyl (Ac).
  • Figure 23 shows mass spectrometry analysis of reduced antibodies showing separate signals from light and heavy chains.
  • Figure 25 shows representative full-body images (maximum intensity projection) acquired 96 hours after the tail vein bolus injection of 100 ⁇ of Zr-Trasuzumab prepared using four different linkers (Bz-SCN, N-Suc, Chx-Mal, and Ac).
  • Figure 26 shows In vivo uptake in selected tissues at 24, 96 and 144 h post injection as
  • antibody herein is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments, so long as they exhibit the desired biological activity (Miller et al (2003) Jour, of Immunology 170:4854-4861). Antibodies may be murine, human, humanized, chimeric, or derived from other species. An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen. (Janeway, C, Travers, P., Walport, M., Shlomchik (2001) Immuno Biology, 5th Ed., Garland Publishing, New York).
  • a target antigen generally has numerous binding sites, also called epitopes, recognized by CDRs on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody.
  • An antibody includes a full-length immunoglobulin molecule or an immunologically active portion of a full-length
  • immunoglobulin molecule i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease.
  • the immunoglobulin disclosed herein can be of any type (e.g., IgG, IgE, IgM, IgD, and IgA), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and
  • immunoglobulins can be derived from any species. In one aspect, however, the immunoglobulin is of human, murine, or rabbit origin.
  • Antibody fragments comprise a portion of a full length antibody, generally the antigen binding or variable region thereof.
  • Examples of antibody fragments include Fab, Fab, and fragments thereof.
  • Fab', F(ab') 2 , and Fv fragments diabodies; linear antibodies; minibodies (Olafsen et al (2004) Protein Eng. Design & Sel. 17(4):315-323), fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, CDR (complementary determining region), and epitope- binding fragments of any of the above which immunospecifically bind to cancer cell antigens, viral antigens or microbial antigens, single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies.
  • the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al (1975) Nature 256:495, or may be made by recombinant DNA methods (see for example: US 4816567; US 5807715).
  • the monoclonal antibodies may also be isolated from phage antibody libraries using the techniques described in Clackson et al (1991) Nature, 352:624-628; Marks et al (1991) J. Mol. Biol, 222:581-597; for example.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (US 4816567; and Morrison et al (1984) Proc. Natl. Acad. Sci. USA, 81 :6851-6855).
  • Chimeric antibodies of interest herein include "primatized" antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape etc) and human constant region sequences.
  • an “intact antibody” herein is one comprising a VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CHI , CH2 and CH3.
  • the constant domains may be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variant thereof.
  • the intact antibody may have one or more "effector functions" which refer to those biological activities attributable to the Fc constant region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include Clq binding;
  • immunoglobulin antibodies can be assigned to different "classes.” There are five major classes of intact immunoglobulin antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into “subclasses” (isotypes), e.g., IgGl , IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • Ig forms include hinge-modifications or hingeless forms (Roux et al (1998) J. Immunol. 161 :4083-4090; Lund et al (2000) Eur. J. Biochem. 267:7246-7256; US 2005/0048572; US 2004/0229310).
  • An "ErbB receptor” is a receptor protein tyrosine kinase which belongs to the ErbB receptor family whose members are important mediators of cell growth, differentiation and survival.
  • the ErbB receptor family includes four distinct members including epidermal growth factor receptor (EGFR, ErbBl, HER1), HER2 (ErbB2 or pl85neu), HER3 (ErbB3) and HER4 (ErbB4 or tyro2).
  • a panel of anti-ErbB2 antibodies has been characterized using the human breast tumor cell line SKBR3 (Hudziak et al (1989) Mol. Cell. Biol. 9(3): 1165- 1172. Maximum inhibition was obtained with the antibody called 4D5 which inhibited cellular proliferation by 56%. Other antibodies in the panel reduced cellular proliferation to a lesser extent in this assay. The antibody 4D5 was further found to sensitize ErbB2- overexpressing breast tumor cell lines to the cytotoxic effects of TNF-a (US 5677171).
  • the anti-ErbB2 antibodies discussed in Hudziak et al. are further characterized in Fendly et al
  • the ErbB receptor will generally comprise an extracellular domain, which may bind an ErbB ligand; a lipophilic transmembrane domain; a conserved intracellular tyrosine kinase domain; and a carboxyl-terminal signaling domain harboring several tyrosine residues which can be phosphorylated.
  • the ErbB receptor may be a "native sequence” ErbB receptor or an "amino acid sequence variant" thereof.
  • the ErbB receptor is native sequence human ErbB receptor. Accordingly, a "member of the ErbB receptor family" is EGFR (ErbBl), ErbB2, ErbB3, ErbB4 or any other ErbB receptor currently known or to be identified in the future.
  • ErbBl "epidermal growth factor receptor", “EGFR” and “HER1” are used interchangeably herein and refer to EGFR as disclosed, for example, in Carpenter et al (1987) Ann. Rev. Biochem., 56:881-914, including naturally occurring mutant forms thereof (e.g., a deletion mutant EGFR as in Humphrey et al (1990) Proc. Nat. Acad. Sci. (USA) 87:4207-4211).
  • the term erbBl refers to the gene encoding the EGFR protein product. Antibodies against HER1 are described, for example, in Murthy et al (1987) Arch. Biochem.
  • ERRP epidermal growth factor receptor
  • EGF epidermal growth factor receptor
  • ErbB2 and "HER2” are used interchangeably herein and refer to human HER2 protein described, for example, in Semba et al (1985) Proc. Nat. Acad. Sci. (USA) 82:6497-6501 and Yamamoto et al (1986) Nature, 319:230-234 (Genebank accession number X03363).
  • the term “erbB2” refers to the gene encoding human ErbB2 and "neu” refers to the gene encoding rat pl85neu.
  • Preferred ErbB2 is native sequence human ErbB2.
  • ErbB3 and "HER3” refer to the receptor polypeptide as disclosed, for example, in U.S. Patent Nos. 5183884 and 5480968 as well as Kraus et al (1989) Proc. Nat. Acad. Sci. (USA) 86:9193-9197.
  • Antibodies against ErbB3 are known in the art and are described, for example, in U.S. Patent Nos. 5183884, 5480968 and in WO 97/35885.
  • ErbB4 and HER4 herein refer to the receptor polypeptide as disclosed, for example, in EP Pat Application No 599,274; Plowman et al (1993) Proc. Natl. Acad. Sci. USA 90: 1746-1750; and Plowman et al (1993) Nature 366:473-475, including isoforms thereof, e.g., as disclosed in WO 99/19488.
  • Antibodies against HER4 are described, for example, in WO 02/18444.
  • Antibodies to ErbB receptors are available commercially from a number of sources, including, for example, Santa Cruz Biotechnology, Inc., California, USA.
  • amino acid sequence variant refers to polypeptides having amino acid sequences that differ to some extent from a native sequence polypeptide. Ordinarily, amino acid sequence variants will possess at least about 70% sequence identity with at least one receptor binding domain of a native ErbB ligand or with at least one ligand binding domain of a native ErbB receptor, and preferably, they will be at least about 80%, more preferably, at least about 90%> homologous by sequence with such receptor or ligand binding domains. The amino acid sequence variants possess substitutions, deletions, and/or insertions at certain positions within the amino acid sequence of the native amino acid sequence. Amino acids are designated by the conventional names, one-letter and three-letter codes.
  • Sequence identity is defined as the percentage of residues in the amino acid sequence variant that are identical after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Methods and computer programs for the alignment are well known in the art. One such computer program is "Align 2,” authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, DC 20559, on December 10, 1991.
  • Antibody-dependent cell-mediated cytotoxicity and “ADCC” refer to a cell- mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g., Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcRs Fc receptors
  • FcR expression on hematopoietic cells in summarized is Table 3 on page 464 of Ravetch and Kinet, (1991) "Annu. Rev. Immunol.” 9:457-92.
  • ADCC activity of a molecule of interest may be assessed in vitro, such as that described in US 5500362 and US 5821337.
  • useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al (1998) PROC. NAT. ACAD. SCI. (USA) (USA) 95:652-656.
  • Human effector cells are leukocytes which express one or more constant region receptors (FcRs) and perform effector functions. Preferably, the cells express at least FcyRIII and perform ADCC effector function. Examples of human leukocytes which mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being preferred.
  • the effector cells may be isolated from a native source thereof, e.g., from blood or PBMCs as described herein.
  • Fc receptor or “FcR” are used to describe a receptor that binds to the Fc constant region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII, and Fey RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • FcyRII receptors include FcyRIIA (anadvant), FcyRIIB (anadvant), and othersadvant.
  • activating receptor and FcyRIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor FcyRIIA contains an immunoreceptor tyrosine-based activation motif (IT AM) in its cytoplasmic domain.
  • Inhibiting receptor FcyRIIB contains an immunoreceptor tyrosine - based inhibition motif (ITIM) in its cytoplasmic domain.
  • ITIM immunoreceptor tyrosine - based inhibition motif
  • FcR neonatal receptor
  • “Complement dependent cytotoxicity” or “CDC” refers to the ability of a molecule to lyse a target in the presence of complement.
  • the complement activation pathway is initiated by the binding of the first component of the complement system (Clq) to a molecule (e.g., an antibody) complexed with a cognate antigen.
  • a CDC assay e.g., as described in Gazzano-Santoro et al J. Immunol. Methods, 202: 163 (1996), may be performed.
  • “Native antibodies” are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (V H ) followed by a number of constant domains. Each light chain has a variable domain at one end (V L ) and a constant domain at its other end. The constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light-chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains.
  • variable refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FRs).
  • the variable domains of native heavy and light chains each comprise four FRs, largely adopting a ⁇ -sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen- binding site of antibodies (see Kabat et al (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD).
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody dependent cellular cytotoxicity (ADCC).
  • hypervariable region when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region generally comprises amino acid residues from a "complementarity determining region" or "CDR" (e.g., residues 24-34 (LI), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (HI), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al supra) and/or those residues from a "hypervariable loop” (e.g., residues 26-32 (LI), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (HI), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk (1987) J. Mol. Biol, 196:901-917).
  • CDR complementarity determining region
  • Framework Region or "FR” residues are those variable domain residues other than the hypervariable region residues as herein defined.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual "Fc” fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen.
  • Fv is the minimum antibody fragment which contains a complete antigen- recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to define an antigen- binding site on the surface of the V H -V L dimer. Collectively, the six hypervariable regions confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • the Fab fragment also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain.
  • Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CHI domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear at least one free thiol group.
  • F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • the "light chains" of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequences of their constant domains.
  • Single-chain Fv or “scFv” antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a variable heavy domain (VH) connected to a variable light domain (VL) in the same polypeptide chain (VH - VL).
  • VH variable heavy domain
  • VL variable light domain
  • Diabodies are described more fully in, for example, EP 404,097; WO 93/11 161 ; and
  • “Humanized” forms of non-human (e.g., rodent) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. Humanization is a method to transfer the murine antigen binding information to a non-immunogenic human antibody acceptor, and has resulted in many therapeutically useful drugs. The method of humanization generally begins by transferring all six murine complementarity determining regions (CDRs) onto a human antibody framework (Jones et al, (1986) Nature 321 :522-525). These CDR-grafted antibodies generally do not retain their original affinity for antigen binding, and in fact, affinity is often severely impaired. Besides the CDRs, select non-human antibody framework residues must also be incorporated to maintain proper CDR
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • a “free cysteine amino acid” refers to a cysteine amino acid residue which has been engineered into a parent antibody, has a thiol functional group (-SH), and is not paired as an intramolecular or intermolecular disulfide bridge.
  • thiol reactivity value is a quantitative characterization of the reactivity of free cysteine amino acids.
  • the thiol reactivity value is the percentage of a free cysteine amino acid in a cysteine engineered antibody which reacts with a thiol-reactive reagent, and converted to a maximum value of 1.
  • a free cysteine amino acid on a cysteine engineered antibody which reacts in 100% yield with a thiol-reactive reagent, such as a biotin-maleimide reagent, to form a biotin-labelled antibody has a thiol reactivity value of 1.0.
  • Another cysteine amino acid engineered into the same or different parent antibody which reacts in 80% yield with a thiol-reactive reagent has a thiol reactivity value of 0.8.
  • Another cysteine amino acid engineered into the same or different parent antibody which fails totally to react with a thiol-reactive reagent has a thiol reactivity value of 0.
  • Determination of the thiol reactivity value of a particular cysteine may be conducted by ELISA assay, mass spectroscopy, liquid chromatography, autoradiography, or other quantitative analytical tests.
  • a “parent antibody” is an antibody comprising an amino acid sequence from which one or more amino acid residues are replaced by one or more cysteine residues.
  • the parent antibody may comprise a native or wild type sequence.
  • the parent antibody may have preexisting amino acid sequence modifications (such as additions, deletions and/or substitutions) relative to other native, wild type, or modified forms of an antibody.
  • a parent antibody may be directed against a target antigen of interest, e.g. a biologically important polypeptide.
  • Antibodies directed against nonpolypeptide antigens are also contemplated.
  • Exemplary parent antibodies include antibodies having affinity and selectivity for cell surface and transmembrane receptors and tumor-associated antigens (TAA).
  • TAA tumor-associated antigens
  • exemplary parent antibodies include those selected from, and without limitation, anti-estrogen receptor antibody, anti-progesterone receptor antibody, anti-p53 antibody, anti-HER-2/neu antibody, anti-EGFR antibody, anti-cathepsin D antibody, anti- Bcl-2 antibody, anti-E-cadherin antibody, anti-CA125 antibody, anti-CA15-3 antibody, anti- CA19-9 antibody, anti-c-erbB-2 antibody, anti-P-glycoprotein antibody, anti-CEA antibody, anti-retinoblastoma protein antibody, anti-ras oncoprotein antibody, anti-Lewis X antibody, anti-Ki-67 antibody, anti-PCNA antibody, anti-CD3 antibody, anti-CD4 antibody, anti-CD5 antibody, anti-CD7 antibody, anti-CD8 antibody, anti-CD9/p24 antibody, anti-CD 10 antibody, anti-CDl lc antibody, anti-CD 13 antibody, anti-CD 14 antibody, anti-CD 15 antibody, anti-CD 19 antibody, anti-CD20 antibody, anti
  • an “isolated” antibody is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or
  • the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • an antibody "which binds" a molecular target or an antigen of interest is one capable of binding that antigen with sufficient affinity such that the antibody is useful in targeting a cell expressing the antigen.
  • the antibody is one which binds ErbB2
  • it will usually preferentially bind ErbB2 as opposed to other ErbB receptors, and may be one which does not significantly cross-react with other proteins such as EGFR, ErbB3 or ErbB4.
  • the extent of binding of the antibody to these non-ErbB2 proteins e.g., cell surface binding to endogenous receptor
  • FACS fluorescence activated cell sorting
  • RIA radioimmunoprecipitation
  • CD proteins and their ligands such as, but not limited to: (i) CD3, CD4, CD8, CD19, CD20, CD22, CD34, CD40, CD79a (CD79a), and CD79p (CD79b); (ii) members of the ErbB receptor family such as the EGF receptor, HER2, HER3 or HER4 receptor; (iii) cell adhesion molecules such as LFA-1, Macl, pl50,95, VLA-4, ICAM-1, VCAM and ⁇ / ⁇ 3 integrin, including either alpha or beta subunits thereof (e.g.
  • anti-CD 1 la, anti-CD 18 or anti-CD 1 lb antibodies growth factors such as VEGF; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C, BR3, c-met, tissue factor, ⁇ 7 etc; and (v) cell surface and transmembrane tumor-associated antigens (TAA).
  • growth factors such as VEGF; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C, BR3, c-met, tissue factor, ⁇ 7 etc; and TAA).
  • the term “monoclonal antibody 4D5" refers to an antibody that has antigen binding residues of, or derived from, the murine 4D5 antibody (ATCC CRL 10463).
  • the monoclonal antibody 4D5 may be murine
  • monoclonal antibody 4D5 or a variant thereof, such as a humanized 4D5.
  • exemplary humanized 4D5 antibodies include huMAb4D5-l, huMAb4D5-2, huMAb4D5-3,
  • huMAb4D5-4 huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (trastuzumab, HERCEPTIN®) as in US Patent No. 5821337.
  • Phage display is a technique by which variant polypeptides are displayed as fusion proteins to a coat protein on the surface of phage, e.g., filamentous phage, particles.
  • phage display One utility of phage display lies in the fact that large libraries of randomized protein variants can be rapidly and efficiently sorted for those sequences that bind to a target molecule with high affinity. Display of peptide and protein libraries on phage has been used for screening millions of polypeptides for ones with specific binding properties. Polyvalent phage display methods have been used for displaying small random peptides and small proteins, typically through fusions to either pill or pVIII of filamentous phage (Wells and Lowman, (1992) Curr. Opin. Struct.
  • phagemid vectors are used, which simplify DNA manipulations. Lowman and Wells, Methods: A companion to Methods in Enzymology, 3:205-0216 (1991).
  • Phage display includes techniques for producing antibody- like molecules (Janeway, C, Travers, P., Walport, M., Shlomchik (2001) Immunobiology, 5th Ed., Garland Publishing, New York, p627-628; Lee et al ).
  • a "phagemid” is a plasmid vector having a bacterial origin of replication, e.g., ColEl, and a copy of an intergenic region of a bacteriophage.
  • the phagemid may be used on any known bacteriophage, including filamentous bacteriophage and lambdoid bacteriophage.
  • the plasmid will also generally contain a selectable marker for antibiotic resistance. Segments of DNA cloned into these vectors can be propagated as plasmids. When cells harboring these vectors are provided with all genes necessary for the production of phage particles, the mode of replication of the plasmid changes to rolling circle replication to generate copies of one strand of the plasmid DNA and package phage particles.
  • the phagemid may form infectious or non-infectious phage particles.
  • This term includes phagemids which contain a phage coat protein gene or fragment thereof linked to a heterologous polypeptide gene as a gene fusion such that the heterologous polypeptide is displayed on the surface of the phage particle.
  • Linker means a chemical moiety comprising a covalent bond or a chain of atoms that covalently attaches an antibody to a drug moiety.
  • a linker is specified as L.
  • Linkers include a divalent radical such as an alkyldiyl, an arylene, a heteroarylene, moieties such as: -(CR 2 ) n O(CR 2 ) n -, repeating units of alkyloxy (e.g. polyethylenoxy, PEG, polymethyleneoxy) and alkylamino (e.g.
  • polyethyleneamino, JeffamineTM polyethyleneamino, JeffamineTM
  • diacid ester and amides including succinate, succinamide, diglycolate, malonate, and caproamide.
  • label means any moiety which can be covalently attached to an antibody and that functions to: (i) provide a detectable signal; (ii) interact with a second label to modify the detectable signal provided by the first or second label, e.g. FRET (fluorescence resonance energy transfer); (iii) stabilize interactions or increase affinity of binding, with antigen or ligand; (iv) affect mobility, e.g. electrophoretic mobility, or cell-permeability, by charge, hydrophobicity, shape, or other physical parameters, or (v) provide a capture moiety, to modulate ligand affinity, antibody/antigen binding, or ionic complexation.
  • FRET fluorescence resonance energy transfer
  • d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • these stereoisomers are identical except that they are mirror images of one another.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • racemic mixture and racemate refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • organic or inorganic salts of an AZC include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, /?-toluenesulfonate, and pamoate (i.e., 1,1 ' -methyl ene-bis -(2-hydroxy-3- naphthoate)) salts.
  • exemplary salts include, but are not limited
  • a pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counterion.
  • the counterion may be any organic or inorganic moiety that stabilizes the charge on the parent compound.
  • a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counterion.
  • “Pharmaceutically acceptable solvate” refers to an association of one or more solvent molecules and an AZC.
  • solvents that form pharmaceutically acceptable solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • BME beta-mercaptoethanol
  • Boc N-(t-butoxycarbonyl)
  • cit citrulline (2-amino-5-ureido pentanoic acid)
  • dap is dolaproine
  • DCC 1,3-dicyclohexylcarbodiimide
  • DCM dichloromethane
  • DEA diethylamine
  • DEAD diethylazodicarboxylate
  • DEPC diethylphosphorylcyanidate
  • DIAD diisopropylazodicarboxylate
  • DIEA N,N- diisopropylethylamine
  • dil dolaisoleucine
  • DMA is dimethylacetamide
  • DMAP 4- dimethylaminopyridine
  • DME is ethyleneglycol dimethyl ether (or 1 ,2-dimethoxyethane)
  • DMF is N,N-dimethylformamide
  • DMSO dimethylsulfoxide
  • doe is
  • DTT dithiothreitol
  • EDCI l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • EEDQ 2-ethoxy-l- ethoxycarbonyl-l,2-dihydroquinoline
  • ES-MS electrospray mass spectrometry
  • EtOAc ethyl acetate
  • Fmoc is N-(9-fluorenylmethoxycarbonyl)
  • gly glycine
  • HATU 0-(7- azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • HOBt 1- hydroxybenzotriazole
  • HPLC high pressure liquid chromatography
  • ile is isoleucine
  • lys is lysine
  • MeCN (CH 3 CN) is acetonitrile
  • MeOH MeOH
  • the compounds of the invention include cysteine engineered antibodies where one or more amino acids of a wild-type or parent antibody are replaced with a cysteine amino acid. Any form of antibody may be so engineered, i.e. mutated.
  • a parent Fab antibody fragment may be engineered to form a cysteine engineered Fab, referred to herein as "ThioFab.”
  • a parent monoclonal antibody may be engineered to form a
  • ThioMab “ThioMab.” It should be noted that a single site mutation yields a single engineered cysteine residue in a ThioFab, while a single site mutation yields two engineered cysteine residues in a ThioMab, due to the dimeric nature of the IgG antibody. Mutants with replaced
  • cysteine (Cys) residues are evaluated for the reactivity of the newly introduced, engineered cysteine thiol groups.
  • the thiol reactivity value is a relative, numerical term in the range of 0 to 1.0 and can be measured for any cysteine engineered antibody.
  • Thiol reactivity values of cysteine engineered antibodies of the invention are in the ranges of 0.6 to 1.0; 0.7 to 1.0; or 0.8 to 1.0.
  • the design, selection, and preparation methods of the invention enable cysteine engineered antibodies which are reactive with electrophilic functionality. These methods further enable antibody conjugate compounds such as antibody-zirconium conjugate (AZC) compounds with zirconium atoms at designated, designed, selective sites. Reactive cysteine residues on an antibody surface allow specifically conjugating a zirconium moiety through a thiol reactive group such as maleimide or haloacetyl.
  • a thiol reactive group such as maleimide or haloacetyl.
  • the nucleophilic reactivity of the thiol functionality of a Cys residue to a maleimide group is about 1000 times higher compared to any other amino acid functionality in a protein, such as amino group of lysine residues or the N-terminal amino group.
  • Thiol specific functionality in iodoacetyl and maleimide reagents may react with amine groups, but higher pH (>9.0) and longer reaction times are required (Garman, 1997, Non-Radioactive Labelling: A Practical Approach, Academic Press, London).
  • cysteine engineered antibodies of the invention preferably retain the antigen binding capability of their wild type, parent antibody counterparts.
  • cysteine engineered antibodies are capable of binding, preferably specifically, to antigens.
  • antigens include, for example, tumor-associated antigens (TAA), cell surface receptor proteins and other cell surface molecules, transmembrane proteins, signalling proteins, cell survival regulatory factors, cell proliferation regulatory factors, molecules associated with (for e.g., known or suspected to contribute functionally to) tissue development or differentiation, lymphokines, cytokines, molecules involved in cell cycle regulation, molecules involved in vasculogenesis and molecules associated with (for e.g., known or suspected to contribute functionally to) angiogenesis.
  • TAA tumor-associated antigens
  • cell surface receptor proteins and other cell surface molecules include, for example, tumor-associated antigens (TAA), cell surface receptor proteins and other cell surface molecules, transmembrane proteins, signalling proteins, cell survival regulatory factors, cell proliferation regulatory factors, molecules associated with (for e.g., known or
  • the tumor-associated antigen may be a cluster differentiation factor (i.e., a CD protein).
  • An antigen to which a cysteine engineered antibody is capable of binding may be a member of a subset of one of the above-mentioned categories, wherein the other subset(s) of said category comprise other molecules/antigens that have a distinct characteristic (with respect to the antigen of interest).
  • the parent antibody may also be a humanized antibody selected from huMAb4D5-l, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (Trastuzumab, HERCEPTIN®) as described in Table 3 of US 5821337, expressly incorporated herein by reference; humanized 520C9 (WO 93/21319) and humanized 2C4 antibodies as described herein.
  • Cysteine engineered antibodies of the invention may be site-specifically and efficiently coupled with a thiol-reactive reagent.
  • the thiol-reactive reagent may be a multifunctional linker reagent, a capture, i.e. affinity, label reagent (e.g. a biotin-linker reagent), a detection label (e.g. a fluorophore reagent), a solid phase immobilization reagent (e.g. SEPHAROSETM, polystyrene, or glass), or a zirconium-linker intermediate.
  • label reagent e.g. a biotin-linker reagent
  • detection label e.g. a fluorophore reagent
  • solid phase immobilization reagent e.g. SEPHAROSETM, polystyrene, or glass
  • zirconium-linker intermediate e.g. SEPHAROSETM, polystyrene, or glass
  • reaction of a ThioFab with a biotin-linker reagent provides a biotinylated ThioFab by which the presence and reactivity of the engineered cysteine residue may be detected and measured.
  • Reaction of a ThioFab with a multifunctional linker reagent provides a ThioFab with a functionalized linker which may be further reacted with a zirconium moiety reagent or other label.
  • Reaction of a ThioFab with a zirconium-linker intermediate provides a ThioFab zirconium conjugate.
  • the exemplary methods described here may be applied generally to the identification and production of antibodies, and more generally, to other proteins through application of the design and screening steps described herein.
  • Such an approach may be applied to the conjugation of other thiol-reactive agents in which the reactive group is, for example, a maleimide, an iodoacetamide, a pyridyl disulfide, or other thiol-reactive conjugation partner (Haugland, 2003, Molecular Probes Handbook of Fluorescent Probes and Research Chemicals, Molecular Probes, Inc.; Brinkley, 1992, Bioconjugate Chem. 3:2; Garman, 1997, Non-Radioactive Labelling: A Practical Approach, Academic Press, London; Means (1990) Bioconjugate Chem. 1 :2; Hermanson, G. in
  • the partner may be a cytotoxic agent (e.g. a toxin such as doxorubicin or pertussis toxin), a fluorophore such as a fluorescent dye like fluorescein or rhodamine, a chelating agent for an imaging or radiotherapeutic metal, a peptidyl or non-peptidyl label or detection tag, or a clearance-modifying agent such as various isomers of polyethylene glycol, a peptide that binds to a third component, or another carbohydrate or lipophilic agent.
  • a cytotoxic agent e.g. a toxin such as doxorubicin or pertussis toxin
  • a fluorophore such as a fluorescent dye like fluorescein or rhodamine
  • a chelating agent for an imaging or radiotherapeutic metal e.g. a chelating agent for an imaging or radiotherapeutic metal
  • a peptidyl or non-peptidyl label or detection tag
  • the sites identified on the exemplary antibody fragment, hu4D5Fabv8, herein are primarily in the constant domain of an antibody which is well conserved across all species of antibodies. These sites should be broadly applicable to other antibodies, without further need of structural design or knowledge of specific antibody structures, and without interference in the antigen binding properties inherent to the variable domains of the antibody.
  • Cysteine engineered antibodies which may be useful in the treatment of cancer include, but are not limited to, antibodies against cell surface receptors and tumor-associated antigens (TAA). Such antibodies may be used as naked antibodies (unconjugated to a label moiety) or as Formula I antibody-zirconium conjugates (AZC). Tumor-associated antigens are known in the art, and can prepared for use in generating antibodies using methods and information which are well known in the art. In attempts to discover effective cellular targets for cancer diagnosis and therapy, researchers have sought to identify transmembrane or otherwise tumor-associated polypeptides that are specifically expressed on the surface of one or more particular type(s) of cancer cell as compared to on one or more normal non- cancerous cell(s).
  • tumor-associated polypeptides are more abundantly expressed on the surface of the cancer cells as compared to on the surface of the non-cancerous cells.
  • the identification of such tumor-associated cell surface antigen polypeptides has given rise to the ability to specifically target cancer cells for destruction via antibody-based therapies.
  • TAA examples include, but are not limited to, TAA (l)-(36) listed below.
  • TAA examples include, but are not limited to, TAA (l)-(36) listed below.
  • information relating to these antigens is listed below and includes names, alternative names, Genbank accession numbers and primary reference(s), following nucleic acid and protein sequence identification conventions of the National Center for Biotechnology Information (NCBI).
  • NCBI National Center for Biotechnology Information
  • Nucleic acid and protein sequences corresponding to TAA (l)-(36) are available in public databases such as GenBank.
  • Tumor- associated antigens targeted by antibodies include all amino acid sequence variants and isoforms possessing at least about 70%, 80%>, 85%, 90%>, or 95% sequence identity relative to the sequences identified in the cited references, or which exhibit substantially the same biological properties or characteristics as a TAA having a sequence found in the cited references.
  • a TAA having a variant sequence generally is able to bind specifically to an antibody that binds specifically to the TAA with the corresponding sequence listed.
  • the sequences and disclosure in the reference specifically recited herein are expressly incorporated by reference.
  • BMPR1B bone morphogenetic protein receptor-type IB, Genbank accession
  • NP 001194 bone morphogenetic protein receptor, type IB /pid NP_001194.1 - Cross-references: MIM:603248; NP 001194.1; AY065994
  • WO2004048938 (Example 2); WO2004032842 (Example IV); WO2003042661 (Claim 12); WO2003016475 (Claim 1); WO200278524 (Example 2); WO200299074 (Claim 19; Page 127-129); WO200286443 (Claim 27; Pages 222, 393); WO2003003906 (Claim 10; Page 293); WO200264798 (Claim 33; Page 93-95); WO200014228 (Claim 5; Page 133-136); US2003224454 (Fig 3); WO2003025138 (Claim 12; Page 150);
  • member 5 /pid NP_003477.3 - Homo sapiens
  • EP1394274 (Example 11); WO2004016225 (Claim 2); WO2003042661 (Claim 12);
  • WO200289747 (Example 5; Page 618-619); WO2003022995 (Example 9; Fig 13A, Example 53; Page 173, Example 2; Fig 2A); NP 036581 six transmembrane epithelial antigen of the prostate
  • WO200292836 (Claim 6; Fig 12); WO200283866 (Claim 15; Page 116-121);
  • MPF MPF
  • MSLN MSLN
  • SMR megakaryocyte potentiating factor
  • mesothelin mesothelin
  • Napi3b (NAPI-3B, NPTIIb, SLC34A2, solute carrier family 34 (sodium
  • Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thrombospondin repeats (type 1 and type 1 -like), transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 5B, Genbank accession no. AB040878) Nagase T., et al (2000) DNA Res. 7 (2): 143-150); WO2004000997 (Claim 1);
  • WO2003003984 (Claim 1); WO200206339 (Claim 1; Page 50); WO200188133 (Claim 1; Page 41-43, 48-58); WO2003054152 (Claim 20); WO2003101400 (Claim 11);
  • PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genbank accession no. AY358628); Ross et al (2002) Cancer Res. 62:2546-2553; US2003129192 (Claim 2); US2004044180 (Claim 12); US2004044179 (Claim 11); US2003096961 (Claim 11); US2003232056 (Example 5); WO2003105758 (Claim 12); US2003206918 (Example 5); EP1347046 (Claim 1); WO2003025148 (Claim 20);
  • ETBR Endothelin type B receptor, Genbank accession no. AY275463
  • WO2004048938 (Example 2); WO2004040000 (Claim 151); WO2003087768 (Claim 1); WO2003016475 (Claim 1); WO2003016475 (Claim 1); WO200261087 (Fig 1);
  • WO2003016494 (Fig 6); WO2003025138 (Claim 12; Page 144); WO200198351 (Claim 1; Page 124-125); EP522868 (Claim 8; Fig 2); WO200177172 (Claim 1; Page 297-299);
  • WO2003104275 (Claim 1); WO2004046342 (Example 2); WO2003042661 (Claim 12); WO2003083074 (Claim 14; Page 61); WO2003018621 (Claim 1); WO2003024392 (Claim 2; Fig 93); WO200166689 (Example 6);
  • LocusID 54894; NP_060233.2; NM_017763_1
  • STEAP2 (HGNC 8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, prostate cancer associated gene 1 , prostate cancer associated protein 1 , six transmembrane epithelial antigen of prostate 2, six transmembrane prostate protein, Genbank accession no.
  • TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, subfamily M, member 4, Genbank accession no. NM 017636)
  • WO200040614 (Claim 14; Page 100-103); WO200210382 (Claim 1; Fig 9A);
  • WO2003042661 (Claim 12); WO200230268 (Claim 27; Page 391); US2003219806 (Claim 4); WO200162794 (Claim 14; Fig 1A-D);
  • CRIPTO (CR, CRl, CRGF, CRIPTO, TDGFl, teratocarcinoma-derived growth factor, Genbank accession no. NP 003203 or NM 003212)
  • WO200216413 (Claim 1; Page 94-95, 105); WO200222808 (Claim 2; Fig 1); US5854399 (Example 2; Col 17-18); US5792616 (Fig 2); Cross-references: MIM: 187395; NP_003203.1; NM_003212_1
  • CD21 CR2 (Complement receptor 2) or C3DR (C3d/Epstein Barr virus
  • WO9102536 (Fig 9.1-9.9); WO2004020595 (Claim 1);
  • CD79b (CD79B, CD79 , IGb (immunoglobulin-associated beta), B29, Genbank accession no. NM 000626 or 11038674)
  • WO2003048202 (claim 1, pages 306 and 309); WO 99/558658, US6534482 (claim 13, Fig 17A/B); WO200055351 (claim 11, pages 1145-1146);
  • FcRH2 (IFGP4, IRTA4, SPAPIA (SH2 domain containing phosphatase anchor protein la), SPAP1B, SPAP1C, Genbank accession no. NM 030764, AY358130)
  • WO2003055439 (Claim 29; Fig 1A-B); WO2003025228 (Claim 37; Fig 5C);
  • WO200222636 (Example 13; Page 95-107); WO200212341 (Claim 68; Fig 7);
  • NCA (CEACAM6, Genbank accession no. M18728);
  • WO200222153 (Page 45-47); US2002042366 (Page 20-21); WO200146261 (Page
  • EphB2R (DRT, ERK, Hek5, EPHT3, Tyro5, Genbank accession no. NM 004442) Chan,J. and Watt, V.M., Oncogene 6 (6), 1057-1061 (1991) Oncogene 10 (5):897-905 (1995), Annu. Rev. Neurosci. 21 :309-345 (1998), Int. Rev. Cytol.
  • WO2002102235 (Claim 13; Page 299); US2003091580 (Example 2); WO200210187 (Claim 6; Fig 10); WO200194641 (Claim 12; Fig 7b); WO200202624 (Claim 13; Fig 1A-1B); US2002034749 (Claim 54; Page 45-46); WO200206317 (Example 2; Page 320-321, Claim 34; Page 321-322); WO200271928 (Page 468-469); WO200202587 (Example 1; Fig 1); WO200140269 (Example 3; Pages 190-192); WO200036107 (Example 2; Page 205-207); WO2004053079 (Claim 12); WO2003004989 (Claim 1); WO200271928 (Page 233-234, 452-453); WO 0116318;
  • PSCA Prostate stem cell antigen precursor, Genbank accession no. AJ297436
  • WO2003003906 (Claim 10; Page 288); WO200140309 (Example 1; Fig 17);
  • WO9851805 (Claim 17; Page 97); W09851824 (Claim 10; Page 94); WO9840403 (Claim 2; Fig IB);
  • AAP14954 lipoma HMGIC fusion-partner-like protein /pid AAP14954.1 - Homo sapiens Species: Homo sapiens (human)
  • WO2003054152 (Claim 20); WO2003000842 (Claim 1); WO2003023013 (Example 3,
  • WO2004011611; WO2003045422 (Example; Page 32-33); WO2003014294 (Claim 35; Fig 6B); WO2003035846 (Claim 70; Page 615-616); WO200294852 (Col 136-137);
  • WO200238766 (Claim 3; Page 133); WO200224909 (Example 3; Fig 3);
  • CD22 B-cell receptor CD22-B isoform, BL-CAM, Lyb-8, Lyb8, SIGLEC-2, FLJ22814, Genbank accession No. AK026467); Wilson et al (1991) J. Exp. Med. 173: 137-146; WO2003072036 (Claim 1; Fig 1);
  • CD79a (CD79A, CD79a, immunoglobulin-associated alpha, a B cell-specific protein that covalently interacts with Ig beta (CD79B) and forms a complex on the surface with Ig M molecules, transduces a signal involved in B-cell differentiation), pi: 4.84, MW: 25028 TM: 2 [P] Gene Chromosome: 19ql3.2, Genbank accession No. NP 001774.10)
  • CXCR5 Burkitt's lymphoma receptor 1, a G protein-coupled receptor that is activated by the CXCL13 chemokine, functions in lymphocyte migration and humoral defense, plays a role in HIV-2 infection and perhaps development of AIDS, lymphoma, myeloma, and leukemia); 372 aa, pi: 8.54 MW: 41959 TM: 7 [P] Gene Chromosome: l lq23.3, Genbank accession No. NP_001707.1)
  • HLA-DOB Beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4+ T lymphocytes); 273 aa, pi: 6.56 MW: 30820 TM: 1 [P] Gene Chromosome: 6p21.3, Genbank accession No. NP 002111.1)
  • CD72 B-cell differentiation antigen CD72, Lyb-2) PROTEIN SEQUENCE Full maeaity...tafrfpd (1..359; 359 aa), pi: 8.66, MW: 40225 TM: 1 [P] Gene Chromosome: 9pl3.3, Genbank accession No. NP_001773.1)
  • WO2004042346 (claim 65); WO2003026493 (pages 51-52, 57-58); WO200075655 (pages 105-106); Von Hoegen et al (1990) J. Immunol. 144(12):4870-4877; Strausberg et al (2002) Proc. Natl. Acad.
  • LY64 Lymphocyte antigen 64 (RP105), type I membrane protein of the leucine rich repeat (LRR) family, regulates B-cell activation and apoptosis, loss of function is associated with increased disease activity in patients with systemic lupus erythematosis); 661 aa, pi: 6.20, MW: 74147 TM: 1 [P] Gene Chromosome: 5ql2, Genbank accession No.
  • FcRHl Fc receptor- like protein 1, a putative receptor for the immunoglobulin Fc domain that contains C2 type Ig-like and IT AM domains, may have a role in B-lymphocyte differentiation
  • WO2003089624 (claim 7); (35) IRTA2 (Immunoglobulin superfamily receptor translocation associated 2, a putative immunoreceptor with possible roles in B cell development and lymphomagenesis;
  • TENB2 tomoregulin, TPEF, HPP 1 , TR, putative transmembrane
  • proteoglycan related to the EGF/heregulin family of growth factors and follistatin
  • 374 aa NCBI Accession: AAD55776, AAF91397, AAG49451, NCBI RefSeq: NP 057276; NCBI Gene: 23671; OMIM: 605734; SwissProt Q9UIK5; Genbank accession No. AF179274; AY358907, CAF85723, CQ782436
  • WO2004074320 (SEQ ID NO 810); JP2004113151 (SEQ ID NOS 2, 4, 8); WO2003042661 (SEQ ID NO 580); WO2003009814 (SEQ ID NO 411); EP1295944 (pages 69-70);
  • WO200230268 page 329
  • WO200190304 SEQ ID NO 2706
  • US2004249130
  • PMEL17 (silver homolog; SILV; D12S53E; PMEL17; (SI); (SIL); ME20; gplOO) BC001414; BT007202; M32295; M77348; NM 006928; McGlinchey,R.P. et al (2009) Proc. Natl. Acad. Sci. U.S.A. 106 (33), 13731-13736; Kummer,M.P. et al (2009) J. Biol. Chem. 284 (4), 2296-2306;
  • TMEFF1 transmembrane protein with EGF-like and two follistatin-like domains 1; Tomoregulin- 1; H7365; C9orf2; C90RF2; U19878; X83961) NM 080655; NM 003692; Harms, P.W. (2003) Genes Dev. 17 (21), 2624-2629; Gery, S. et al (2003) Oncogene 22 (18):2723-2727;
  • GDNF-Ral GDNF family receptor alpha 1 ; GFRA1; GDNFR; GDNFRA; RETL1; TRNR1; RET1L; GDNFR-alphal; GFR-ALPHA-1; U95847; BC014962; NM 145793)
  • Ly6E lymphocyte antigen 6 complex, locus E; Ly67,RIG-E,SCA-2,TSA-l
  • TMEM46 shisa homolog 2 (Xenopus laevis); SHISA2) NP 001007539.1;
  • Ly6G6D lymphocyte antigen 6 complex, locus G6D; Ly6-D, MEGT1
  • NP_067079.2 NP_067079.2
  • NM_021246.2 Mallya, M. et al (2002) Genomics 80 (1): 113-123; Ribas,G. et al (1999) J. Immunol. 163 (l):278-287;
  • LGR5 leucme-rich repeat-containing G protein-coupled receptor 5; GPR49, GPR67
  • NP 003658.1 NM 003667.2
  • Salanti,G. et al (2009) Am. J. Epidemiol. 170 (5):537-545; Yamamoto,Y. et al (2003) Hepatology 37 (3):528-533
  • RET ret proto-oncogene; MEN2A; HSCR1; MEN2B; MTC1; (PTC); CDHF12;
  • LY6K lymphocyte antigen 6 complex, locus K; LY6K; HSJ001348; FLJ35226) NP_059997.3; NM_017527.3; Ishikawa,N. et al (2007) Cancer Res. 67 (24): 11601-11611; de Nooij-van Dalen,A.G. et al (2003) Int. J. Cancer 103 (6):768-774; (46) GPR19 (G protein-coupled receptor 19; Mm.4787) NP_006134.1; NM_006143.2;
  • Tyrosinase (TYR; OCAIA; OCAIA; tyrosinase; SHEP3) NP 000363.1; NM 000372.4; Bishop,D.T. et al (2009) Nat. Genet. 41 (8):920-925; Nan, H. et al (2009) Int. J. Cancer 125 (4):909-917;
  • TMEM118 ring finger protein, transmembrane 2; RNFT2; FLJ14627
  • GPR172A G protein-coupled receptor 172A; GPCR41; FLJ11856; D15Ertd747e
  • the parent antibody may also be a fusion protein comprising an albumin-binding peptide (ABP) sequence (Dennis et al. (2002) “Albumin Binding As A General Strategy For Improving The Pharmacokinetics Of Proteins” J Biol Chem. 277:35035-35043; WO
  • ABSP albumin-binding peptide
  • Antibodies of the invention include fusion proteins with ABP sequences taught by: (i) Dennis et al (2002) J Biol Chem. 277:35035-35043 at Tables III and IV, page 35038; (ii) US 20040001827 at [0076] SEQ ID NOS: 9-22; and (iii) WO 01/45746 at pages 12-13, SEQ ID NOS: zl-zl4, and all of which are incorporated herein by reference.
  • DNA encoding an amino acid sequence variant of the starting polypeptide is prepared by a variety of methods known in the art. These methods include, but are not limited to, preparation by site-directed (or oligonucleotide-mediated) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared DNA encoding the polypeptide. Variants of recombinant antibodies may be constructed also by restriction fragment manipulation or by overlap extension PCR with synthetic oligonucleotides. Mutagenic primers encode the cysteine codon replacement(s). Standard mutagenesis techniques can be employed to generate DNA encoding such mutant cysteine engineered antibodies.
  • Site-directed mutagenesis is one method for preparing substitution variants, i.e.
  • mutant proteins This technique is well known in the art (see for example, Carter (1985) et al Nucleic Acids Res. 13:4431-4443; Ho et al (1989) Gene (Amst.) 77:51-59; and Kunkel et al (1987) Proc. Natl. Acad. Sci. USA 82:488). Briefly, in carrying out site-directed mutagenesis of DNA, the starting DNA is altered by first hybridizing an oligonucleotide encoding the desired mutation to a single strand of such starting DNA.
  • a DNA polymerase is used to synthesize an entire second strand, using the hybridized oligonucleotide as a primer, and using the single strand of the starting DNA as a template.
  • the oligonucleotide encoding the desired mutation is incorporated in the resulting double-stranded DNA.
  • Site-directed mutagenesis may be carried out within the gene expressing the protein to be mutagenized in an expression plasmid and the resulting plasmid may be sequenced to confirm the introduction of the desired cysteine replacement mutations (Liu et al (1998) J. Biol. Chem. 273:20252-20260).
  • Site-directed of protocols and formats including those commercially available, e.g. QuikChange® Multi Site-Directed Mutagenesis Kit (Stratagene, La Jolla, CA).
  • PCR mutagenesis is also suitable for making amino acid sequence variants of the starting polypeptide. See Higuchi, (1990) in PCR Protocols, pp.177-183, Academic Press; Ito et al (1991) Gene 102:67-70; Bernhard et al (1994) Bioconjugate Chem. 5: 126-132; and Vallette et al (1989) Nuc. Acids Res. 17:723-733. Briefly, when small amounts of template DNA are used as starting material in a PCR, primers that differ slightly in sequence from the corresponding region in a template DNA can be used to generate relatively large quantities of a specific DNA fragment that differs from the template sequence only at the positions where the primers differ from the template.
  • the starting material is the plasmid (or other vector) comprising the starting polypeptide DNA to be mutated.
  • the codon(s) in the starting DNA to be mutated are identified. There must be a unique restriction endonuclease site on each side of the identified mutation site(s). If no such restriction sites exist, they may be generated using the above described oligonucleotide -mediated
  • oligonucleotide encoding the sequence of the DNA between the restriction sites but containing the desired mutation(s) is synthesized using standard procedures, wherein the two strands of the oligonucleotide are synthesized separately and then hybridized together using standard techniques.
  • This double-stranded oligonucleotide is referred to as the cassette.
  • This cassette is designed to have 5' and 3' ends that are compatible with the ends of the linearized plasmid, such that it can be directly ligated to the plasmid.
  • This plasmid now contains the mutated DNA sequence. Mutant DNA containing the encoded cysteine replacements can be confirmed by DNA sequencing.
  • Single mutations are also generated by oligonucleotide directed mutagenesis using double stranded plasmid DNA as template by PCR based mutagenesis (Sambrook and Russel, (2001) Molecular Cloning: A Laboratory Manual, 3rd edition; Zoller et al (1983) Methods Enzymol. 100:468-500; Zoller, M.J. and Smith, M. (1982) Nucl. Acids Res.
  • hu4D5Fabv8 displayed on M13 phage (Gerstner et al (2002) "Sequence Plasticity In The Antigen-Binding Site Of A Therapeutic Anti-HER2 Antibody", J Mol Biol. 321 : 851-62) was used for experiments as a model system. Cysteine mutations were introduced in hu4D5Fabv8-phage, hu4D5Fabv8, and ABP-hu4D5Fabv8 constructs. The hu4D5-ThioFab-Phage preps were carried out using the polyethylene glycol (PEG) precipitation method as described earlier (Lowman, Henry B. (1998) Methods in Molecular Biology (Totowa, New Jersey) 87 (Combinatorial Peptide Library Protocols) 249-264).
  • PEG polyethylene glycol
  • Oligonucleotides are prepared by the phosphoramidite synthesis method (US
  • the phosphoramidite method entails cyclical addition of nucleotide monomer units with a reactive 3 ' phosphoramidite moiety to an oligonucleotide chain growing on a solid-support comprised of controlled-pore glass or highly crosslinked polystyrene, and most commonly in the 3' to 5' direction in which the 3' terminus nucleoside is attached to the solid-support at the beginning of synthesis (US 5047524; US 5262530).
  • Oligonucleotides can be chemically labelled with non-isotopic moieties for detection, capture, stabilization, or other purposes (Andrus, A. "Chemical methods for 5' non-isotopic labelling of PCR probes and primers” (1995) in PCR 2: A Practical Approach, Oxford University Press, Oxford, pp. 39-54; Hermanson, G. in Bioconjugate Techniques (1996) Academic Press, San Diego, pp. 40-55, 643-671; Keller, G. and Manak, M. in DNA Probes Second Edition (1993), Stockton Press, New York, pp. 121-23).
  • PHESELECTOR ASSAY Applied Biosystems, Foster City, CA.
  • the PHESELECTOR Phage ELISA for Selection of Reactive Thiols assay allows for detection of reactive cysteine groups in antibodies in an ELISA phage format (US 7521541; Junutula JR et al. "Rapid identification of reactive cysteine residues for site- specific labeling of antibody-Fabs" J Immunol Methods 2008;332:41-52).
  • the process of coating the protein (e.g. antibody) of interest on well surfaces, followed incubation with phage particles and then HRP labeled secondary antibody with absorbance detection is detailed in Example 2. Mutant proteins displayed on phage may be screened in a rapid, robust, and high-throughput manner. Libraries of cysteine engineered antibodies can be produced and subjected to binding selection using the same approach to identify
  • FIG. 8 illustrates the PHESELECTOR Assay by a schematic representation depicting the binding of Fab or ThioFab to HER2 (top) and biotinylated ThioFab to streptavidin (bottom).
  • DNA encoding the cysteine engineered antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells serve as a source of such DNA.
  • the DNA may be placed into expression vectors, which are then trans fected into host cells such as E.
  • coli cells simian COS cells, Chinese Hamster Ovary (CHO) cells, or other mammalian host cells, such as myeloma cells (US 5807715; US 2005/0048572; US 2004/0229310) that do not otherwise produce the antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • myeloma cells US 5807715; US 2005/0048572; US 2004/0229310
  • the yields of hu4D5Fabv8 cysteine engineered antibodies were similar to wild type hu4D5Fabv8.
  • Review articles on recombinant expression in bacteria of DNA encoding the antibody include Skerra et al (1993) Curr. Opinion in Immunol. 5:256- 262 and Pluckthun (1992) Immunol. Revs. 130: 151-188.
  • cysteine engineered antibodies e.g. ThioFabs, with highly reactive unpaired Cys residues
  • a bacterial e.g. E. coli
  • a mammalian cell culture system e.g. Chinese Hamster Ovary cells (CHO)
  • CHO Chinese Hamster Ovary cells
  • ThioFabs were expressed upon induction in 34B8, a non-suppressor E. coli strain (Baca et al (1997) Journal Biological Chemistry 272(16): 10678-84). See Example 3a.
  • the harvested cell pellet was resuspended in PBS (phosphate buffered saline), total cell lysis was performed by passing through a microfluidizer and the ThioFabs were purified by affinity chromatography with protein G SEPHAROSETM (Amersham).
  • ThioFabs were conjugated with biotin-PEO-maleimide as described above and the biotinylated-ThioFabs were further purified by Superdex-200TM (Amersham) gel filtration chromatography, which eliminated the free biotin-PEO-maleimide and the oligomeric fraction of ThioFabs.
  • LC-ESI-MS Liquid chromatography electrospray ionization mass spectrometric analysis was employed for the accurate molecular weight determination of biotin conjugated Fab (Cole, R.B. Electro Spray Ionization Mass Spectrometry: Fundamentals, Instrumentation And Applications. (1997) Wiley, New York).
  • the antibody Fab fragment hu4D5Fabv8 contains about 445 amino acid residues, including 10 Cys residues (five on the light and five on the heavy chain).
  • the high-resolution structure of the humanized 4D5 variable fragment (Fv4D5) has been established, see:
  • Figure 1 A shows a three-dimensional representation of the hu4D5Fabv8 antibody fragment derived by X-ray crystal coordinates.
  • the structure positions of the engineered Cys residues of the heavy and light chains are numbered according to a sequential numbering system.
  • This sequential numbering system is correlated to the Kabat numbering system (Kabat et al., (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD) for the 4d5v7fabH variant of trastuzumab according to Figure IB which shows the sequential numbering scheme (top row), starting at the N-terminus, differs from the Kabat numbering scheme (bottom row) by insertions noted by a,b,c.
  • the actual linear amino acid sequence may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the variable domain.
  • the cysteine engineered heavy chain variant sites are identified by the sequential numbering and Kabat numbering schemes in the following chart:
  • Ml 3 phagemid-Cys mutant Fabs ( Figures 3 A and 3B) can be rapidly screened compared to Fab proteins.
  • Phagemid-ThioFab binding to antigen and to streptavidin can be tested by coating HER2 and streptavidin, respectively, onto ELISA plates followed by probing with anti-Fab-HRP (Horse radish peroxidase) as described in Example 2 and depicted in Figure 8.
  • This method allowed simultaneous monitoring of the effect on the antigen binding and the reactivity of the thiol group by the engineered Cys
  • the method can be applied to screen the reactive thiol groups for any protein displayed on Ml 3 phage.
  • Conjugated or unconjugated phagemid-ThioFabs are purified by simple PEG precipitation.
  • FIG. 8 is a graphical representation of the PHESELECTOR assay, depicting binding of a biotinylated ThioFab phage and an anti-phage HRP antibody to HER2 (top) and Streptavidin (bottom).
  • Biotin conjugated and unconjugated variants were tested for HER2 and streptavidin binding using an ELISA based PHESELECTOR assay ( Figure 8, Example 2) with an HRP (horseradish peroxidase)-conjugated anti-phage antibody.
  • HRP horseradish peroxidase
  • the ThioFabs-phage samples showed varying levels of streptavidin binding activity. From all the tested phage-ThioFabs, the A121C cysteine engineered antibody exhibited maximal thiol reactivity. Even though wild type hu4D5Fabv8- phage was incubated with the same amounts of biotin-maleimide, these phage had little streptavidin binding indicating that preexisting cysteine residues (involved in disulfide bond formation) from the hu4D5Fabv8 and Ml 3 phage coat proteins did not interfere with the site- specific conjugation of biotin-maleimide. These results demonstrate that the phage ELISA assay can be used successfully to screen reactive thiol groups on the Fab surface.
  • the PHESELECTOR assay allows screening of reactive thiol groups in antibodies.
  • a parameter, fractional surface accessibility was employed to identify and quantitate the accessibility of solvent to the amino acid residues in a polypeptide.
  • the surface accessibility can be expressed as the surface area (A 2 ) that can be contacted by a solvent molecule, e.g. water.
  • a solvent molecule e.g. water.
  • the occupied space of water is approximated as a 1.4 A radius sphere.
  • Software is freely available or licensable (Secretary to CCP4, Daresbury Laboratory, Warrington, WA4 4 AD, United Kingdom, Fax: (+44) 1925 603825, or by internet:
  • AREAIMOL defines the solvent accessible surface of a protein as the locus of the centre of a probe sphere (representing a solvent molecule) as it rolls over the Van der Waals surface of the protein.
  • AREAIMOL calculates the solvent accessible surface area by generating surface points on an extended sphere about each atom (at a distance from the atom centre equal to the sum of the atom and probe radii), and eliminating those that lie within equivalent spheres associated with neighboring atoms.
  • AREAIMOL finds the solvent accessible area of atoms in a PDB coordinate file, and summarizes the accessible area by residue, by chain and for the whole molecule. Accessible areas (or area differences) for individual atoms can be written to a pseudo-PDB output file.
  • AREAIMOL assumes a single radius for each element, and only recognizes a limited number of different elements. Unknown atom types (i.e. those not in AREAIMOL's internal database) will be assigned the default radius of 1.8 A. The list of recognized atoms is:
  • AREAIMOL and SURFACE report absolute accessibilities, i.e. the number of square Angstroms (A).
  • Fractional surface accessibility is calculated by reference to a standard state relevant for an amino acid within a polypeptide.
  • the reference state is tripeptide Gly-X-Gly, where X is the amino acid of interest, and the reference state should be an 'extended' conformation, i.e. like those in beta-strands.
  • the extended conformation maximizes the accessibility of X.
  • a calculated accessible area is divided by the accessible area in a Gly-X- Gly tripeptide reference state and reports the quotient, which is the fractional accessibility. Percent accessibility is fractional accessibility multiplied by 100.
  • Another exemplary algorithm for calculating surface accessibility is based on the SOLV module of the program xsae (Broger, C, F. Hoffman-LaRoche, Basel) which calculates fractional accessibility of an amino acid residue to a water sphere based on the X- ray coordinates of the polypeptide.
  • Residues are sorted based on their role in functional and structural interactions of
  • Thiol reactivity may be generalized to any antibody where substitution of amino acids with reactive cysteine amino acids may be made within the ranges in the light chain selected from: L-10 to L-20; L-38 to L-48; L-105 to L-l 15; L-139 to L-149; L-163 to L-173; and within the ranges in the heavy chain selected from: H-35 to H-45; H-83 to H-93; H-l 14 to H- 127; and H-l 70 to H-l 84, and in the Fc region within the ranges selected from H-268 to H- 291; H-319 to H-344; H-370 to H-380; and H-395 to H-405.
  • Thiol reactivity may also be generalized to certain domains of an antibody, such as the light chain constant domain (CL) and heavy chain constant domains, CHI, CH2 and CH3. Cysteine replacements resulting in thiol reactivity values of 0.6 and higher may be made in the heavy chain constant domains ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ of intact antibodies: IgA, IgD, IgE, IgG, and IgM, respectively, including the IgG subclasses: IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
  • Thiol reactivity data is shown in Figures 3A and 3B for amino acid residues of 4D5 ThioFab Cys mutants: (3 A) non-biotinylated (control) and (3B) biotinylated phage-ThioFabs. Reactive thiol groups on antibody/Fab surface were identified by PHESELECTOR assay analyses for the interaction of non-biotinylated phage-hu4D5Fabv8 (3 A) and biotinylated phage-hu4D5Fabv8 (3B) with BSA (open box), HER2 (grey box) or streptavidin (solid box). The assay was carried out as described in Example 2.
  • Light chain variants are on the left side and heavy chain variants are on the right side.
  • the binding of non-biotinylated 4D5 ThioFab Cys mutants is low as expected, but strong binding to HER2 is retained.
  • the ratio of binding to streptavidin and to HER2 of the biotinylated 4D5 ThioFab Cys mutants gives the thiol reactivity values in Table 2.
  • Background absorbance at 450 nm or small amounts of nonspecific protein binding of the biotinylated 4D5 ThioFab Cys mutants to BSA is also evident in Figure 3B. Fractional Surface Accessibility values of the selected amino acid residues that were replaced with a Cys residue are shown in Figure 4A.
  • Amino acids at positions L-15, L-43, L-l 10, L-144, L-168, H-40, H-88, H-l 19, H- 121, H-l 22, H-l 75, and H-l 79 of an antibody may generally be mutated (replaced) with free cysteine amino acids. Ranges within about 5 amino acid residues on each side of these positions may also be replaced with free cysteine acids, i.e.
  • L light chain
  • H heavy chain
  • A alanine
  • S serine
  • V valine
  • C cysteine
  • Thiol reactivity is measured as the ratio of OD 450 nm for streptavidin binding to OD 450 ancient for HER2 (antibody) binding (Example 2). Thiol reactivity value of 1 indicates complete biotinylation of the cysteine thiol.
  • Fab preparation may require 2-3 days, depending on the scale of production. During this time, thiol groups may lose reactivity due to oxidation.
  • stability of the thiol reactivity of phage-thioFabs was measured ( Figure 4B).
  • ThioFab-phage purification on day 1, day 2 and day 4, all the samples were conjugated with biotin-PEO-maleimide and probed with phage ELISA assay (PHESELECTOR) to test HER2 and streptavidin binding.
  • PESELECTOR phage ELISA assay
  • the compounds of the invention include cysteine engineered antibodies where one or more amino acids of a parent antibody are replaced with a free cysteine amino acid.
  • a cysteine engineered antibody comprises one or more free cysteine amino acids having a thiol reactivity value in the range of 0.6 to l .O.
  • a free cysteine amino acid is a cysteine residue which has been engineered into the parent antibody and is not part of a disulfide bridge.
  • cysteine engineered antibody is prepared by a process comprising:
  • the cysteine engineered antibody may be more reactive than the parent antibody with the thiol-reactive reagent.
  • the free cysteine amino acid residues may be located in the heavy or light chains, or in the constant or variable domains.
  • Antibody fragments e.g. Fab, may also be engineered with one or more cysteine amino acids replacing amino acids of the antibody fragment, to form cysteine engineered antibody fragments.
  • Another aspect of the invention provides a method of preparing (making) a cysteine engineered antibody, comprising:
  • cysteine engineered antibody is more reactive than the parent antibody with the thiol-reactive reagent.
  • Step (a) of the method of preparing a cysteine engineered antibody may comprise:
  • Step (b) of the method of preparing a cysteine engineered antibody may comprise expressing the cysteine engineered antibody on a viral particle selected from a phage or a phagemid particle.
  • Step (b) of the method of preparing a cysteine engineered antibody may also comprise:
  • Another aspect of the invention is a method of screening cysteine engineered antibodies with highly reactive, unpaired cysteine amino acids for thiol reactivity
  • Step (a) of the method of screening cysteine engineered antibodies may comprise:
  • Step (b) of the method of screening cysteine engineered antibodies may comprise expressing the cysteine engineered antibody on a viral particle selected from a phage or a phagemid particle.
  • Step (b) of the method of screening cysteine engineered antibodies may also comprise:
  • the cysteine engineered antibodies of the invention may be conjugated with any label moiety which can be covalently attached to the antibody through a reactive cysteine thiol group (Singh et al (2002) Anal. Biochem. 304: 147-15; Harlow E. and Lane, D. (1999) Using Antibodies: A Laboratory Manual, Cold Springs Harbor Laboratory Press, Cold Spring Harbor, NY; Lundblad R.L. (1991) Chemical Reagents for Protein Modification, 2nd ed. CRC Press, Boca Raton, FL).
  • the attached label may function to: (i) provide a detectable signal; (ii) interact with a second label to modify the detectable signal provided by the first or second label, e.g.
  • FRET fluorescence resonance energy transfer
  • Labelled cysteine engineered antibodies may be useful in diagnostic assays, e.g., for detecting expression of an antigen of interest in specific cells, tissues, or serum.
  • the antibody will typically be labeled with a detectable moiety.
  • Radioisotopes such as 3 H, n C, 14 C, 18 F, 32 P, 35 S, 64 Cu, 68 Ga,
  • the antibody can be labeled with ligand reagents that bind, chelate or otherwise complex a radioisotope metal where the reagent is reactive with the engineered cysteine thiol of the antibody, using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley- Interscience, New York, NY, Pubs. (1991).
  • Chelating ligands which may complex a metal ion include DOT A, DOPA, DOTP, DOTMA, DTP A and TETA (Macrocyclics, Dallas, TX).
  • Radionuclides can be targetted via complexation with cysteine-engineered antibodies as antibody-zirconium conjugates of the invention (Wu et al (2005) Nature Biotechnology 23(9): 1137-1146).
  • Metal-chelate complexes suitable as antibody labels for imaging experiments are disclosed: US 5342606; US 5428155; US 5316757; US 5480990; US 5462725; US 5428139; US 5385893; US 5739294; US 5750660; US 5834456; Hnatowich et al (1983) J. Immunol. Methods 65: 147-157; Meares et al (1984) Anal. Biochem. 142:68-78; Mirzadeh et al (1990) Bioconjugate Chem. 1 :59-65; Meares et al (1990) J. Cancerl990, Suppl. 10:21-26; Izard et al (1992) Bioconjugate Chem.
  • Fluorescent labels such as rare earth chelates (europium chelates), fluorescein types including FITC, 5-carboxyfluorescein, 6-carboxy fluorescein; rhodamine types including TAMRA; dansyl; Lissamine; cyanines; phycoerythrins; Texas Red; and analogs thereof.
  • the fluorescent labels can be conjugated to antibodies using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescent dyes and fluorescent label reagents include those which are commercially available from
  • the enzyme generally catalyzes a chemical alteration of a chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above.
  • the chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
  • enzymatic labels include luciferases ⁇ e.g., firefly luciferase and bacterial luciferase; US 4737456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRP), alkaline phosphatase (AP), ⁇ - galactosidase, glucoamylase, lysozyme, saccharide oxidases ⁇ e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
  • luciferases ⁇ e.g., firefly luciferase and bacterial luciferase;
  • enzyme-substrate combinations include, for example:
  • HRP Horseradish peroxidase
  • OPD orthophenylene diamine
  • TMB 3,3',5,5'-tetramethylbenzidine hydrochloride
  • ⁇ -D-galactosidase ( ⁇ -D-Gal) with a chromogenic substrate ⁇ e.g., p- nitrophenyl- -D-galactosidase) or fluorogenic substrate 4-methylumbelliferyl- -D- galactosidase.
  • a chromogenic substrate ⁇ e.g., p- nitrophenyl- -D-galactosidase
  • fluorogenic substrate 4-methylumbelliferyl- -D- galactosidase 4-methylumbelliferyl- -D- galactosidase.
  • a label may be indirectly conjugated with a cysteine engineered antibody.
  • the antibody can be conjugated with biotin and any of the three broad categories of labels mentioned above can be conjugated with avidin or streptavidin, or vice versa. Biotin binds selectively to streptavidin and thus, the label can be conjugated with the antibody in this indirect manner.
  • the polypeptide variant is conjugated with a small hapten (e.g., digoxin) and one of the different types of labels mentioned above is conjugated with an anti-hapten polypeptide variant ⁇ e.g., anti-digoxin antibody).
  • a small hapten e.g., digoxin
  • an anti-hapten polypeptide variant e.g., anti-digoxin antibody
  • polypeptide variant of the present invention may be employed in any known assay method, such as ELISA, competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays (Zola, (1987) Monoclonal Antibodies: A Manual of Techniques, pp.147-158, CRC Press, Inc.).
  • a detection label may be useful for localizing, visualizing, and quantitating a binding or recognition event.
  • the labelled antibodies of the invention can detect cell-surface receptors.
  • Another use for detectably labelled antibodies is a method of bead-based immunocapture comprising conjugating a bead with a fluorescent labelled antibody and detecting a fluorescence signal upon binding of a ligand. Similar binding detection methodologies utilize the surface plasmon resonance (SPR) effect to measure and detect antibody-antigen interactions.
  • SPR surface plasmon resonance
  • Detection labels such as fluorescent dyes and chemiluminescent dyes (Briggs et al (1997) "Synthesis of Functionalised Fluorescent Dyes and Their Coupling to Amines and
  • the labels preferably (iii) have good water-solubility to achieve effective conjugate concentration and detection sensitivity and (iv) are non-toxic to living cells so as not to disrupt the normal metabolic processes of the cells or cause premature cell death.
  • fluorescently labelled events e.g. cell surface binding of peptide-dye conjugates may be conducted on an system (FMAT® 8100 HTS System, Applied Biosystems, Foster City, Calif.) that automates mix-and-read, non-radioactive assays with live cells or beads (Miraglia, "Homogeneous cell- and bead-based assays for high throughput screening using fluorometric microvolume assay technology", (1999) J. of Biomolecular Screening 4: 193-204).
  • FMAT® 8100 HTS System Applied Biosystems, Foster City, Calif.
  • labelled antibodies also include cell surface receptor binding assays, inmmunocapture assays, fluorescence linked immunosorbent assays (FLISA), caspase-cleavage (Zheng, "Caspase-3 controls both cytoplasmic and nuclear events associated with Fas-mediated apoptosis in vivo", (1998) Proc. Natl. Acad. Sci. USA 95:618-23; US 6372907), apoptosis (Vermes, "A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V" (1995) J. Immunol. Methods 184:39-51) and cytotoxicity assays. Fluorometric microvolume assay technology can be used to identify the up or down regulation by a molecule that is targeted to the cell surface
  • Labelled cysteine engineered antibodies of the invention are useful as imaging biomarkers and probes by the various methods and techniques of biomedical and molecular imaging such as: (i) MRI (magnetic resonance imaging); (ii) MicroCT (computerized tomography); (iii) SPECT (single photon emission computed tomography); (iv) PET
  • Imaging biomarkers may be objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention.
  • Biomarkers may be of several types: Type 0 are natural history markers of a disease and correlate longitudinally with known clinical indices, e.g.
  • Imaging biomarkers thus can provide pharmacodynamic (PD) therapeutic information about: (i) expression of a target protein, (ii) binding of a therapeutic to the target protein, i.e. selectivity, and (iii) clearance and half-life pharmacokinetic data.
  • PD pharmacodynamic
  • in vivo imaging biomarkers relative to lab-based biomarkers include: non-invasive treatment, quantifiable, whole body assessment, repetitive dosing and assessment, i.e. multiple time points, and potentially transferable effects from preclinical (small animal) to clinical (human) results. For some applications, bioimaging supplants or minimizes the number of animal experiments in preclinical studies.
  • Radionuclide imaging labels include radionuclides such as H, C, C, F, P, S,
  • radionuclide metal ion can be complexed with a chelating linker such as DOTA.
  • Linker reagents such as DOTA-maleimide (4-maleimidobutyramidobenzyl-DOTA) can be prepared by the reaction of aminobenzyl-DOTA with 4-maleimidobutyric acid (Fluka) activated with isopropylchloro formate (Aldrich), following the procedure of Axworthy et al (2000) Proc. Natl. Acad. Sci. USA 97(4): 1802-1807). DOTA-maleimide reagents react with the free cysteine amino acids of the cysteine engineered antibodies and provide a metal complexing ligand on the antibody (Lewis et al (1998) Bioconj. Chem. 9:72-86).
  • Chelating linker labelling reagents such as DOTA-NHS (l,4,7,10-tetraazacyclododecane-l,4,7,10-tetraacetic acid mono (N-hydroxysuccinimide ester) are commercially available (Macrocyclics, Dallas, TX).
  • Receptor target imaging with radionuclide labelled antibodies can provide a marker of pathway activation by detection and quantitation of progressive accumulation of antibodies in tumor tissue (Albert et al (1998) Bioorg. Med. Chem. Lett. 8: 1207-1210).
  • the conjugated radio-metals may remain intracellular following lysosomal degradation.
  • FRET fluorescence resonance energy transfer
  • Reporter groups are typically fluorescent dyes that are excited by light at a certain wavelength and transfer energy to an acceptor, or quencher, group, with the appropriate Stokes shift for emission at maximal brightness.
  • Fluorescent dyes include molecules with extended aromaticity, such as fluorescein and rhodamine, and their derivatives.
  • the fluorescent reporter may be partially or significantly quenched by the quencher moiety in an intact peptide. Upon cleavage of the peptide by a peptidase or protease, a detectable increase in fluorescence may be measured (Knight, C. (1995) "Fluorimetric Assays of Proteolytic Enzymes", Methods in Enzymology, Academic Press, 248: 18-34).
  • the labelled antibodies of the invention may also be used as an affinity purification agent.
  • the labelled antibody is immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art.
  • the immobilized antibody is contacted with a sample containing the antigen to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the antigen to be purified, which is bound to the immobilized polypeptide variant. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the antigen from the polypeptide variant.
  • Labelling reagents typically bear reactive functionality which may react (i) directly with a cysteine thiol of a cysteine engineered antibody to form the labelled antibody, (ii) with a linker reagent to form a linker-label intermediate, or (iii) with a linker antibody to form the labelled antibody.
  • Reactive functionality of labelling reagents include: maleimide, haloacetyl, iodoacetamide succinimidyl ester (e.g.
  • An exemplary reactive functional group is N-hydroxysuccinimidyl ester (NHS) of a carboxyl group substituent of a detectable label, e.g. biotin or a fluorescent dye.
  • the NHS ester of the label may be preformed, isolated, purified, and/or characterized, or it may be formed in situ and reacted with a nucleophilic group of an antibody.
  • the carboxyl form of the label is activated by reacting with some combination of a carbodiimide reagent, e.g. dicyclohexylcarbodiimide, diisopropylcarbodiimide, or a uranium reagent, e.g.
  • TSTU (0-(N-Succinimidyl)-N,N,N',N'-tetramethyluronium tetrafluoroborate
  • HBTU O- benzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • HATU (0-(7- azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • an activator such as 1-hydroxybenzotriazole (HOBt)
  • HOBt 1-hydroxybenzotriazole
  • N-hydroxysuccinimide to give the NHS ester of the label.
  • the label and the antibody may be coupled by in situ activation of the label and reaction with the antibody to form the label-antibody conjugate in one step.
  • Other activating and coupling reagents include TBTU (2-(lH-benzotriazo-l-yl)-l-l,3,3- tetramethyluronium hexafluorophosphate), TFFH (N,N',N",N"'-tetramethyluronium 2- fluoro-hexafluorophosphate), PyBOP (benzotriazole- 1 -yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate, EEDQ (2-ethoxy-l-ethoxycarbonyl-l,2-dihydro-quinoline), DCC (dicyclohexylcarbodiimide); DIPCDI (diisopropylcarbodiimide), MSNT (l-(mesitylene-2- sulfonyl)-3-nitro
  • ThioFab constructs were cloned into an expression vector under alkaline phosphatase promoter (Chang et al (1987) Gene 55: 189-196) and the ThioFab expression was induced by growing E. coli cells in the phosphate-free medium.
  • ThioFabs were purified on a Protein G SEPHAROSETM column and analyzed on reducing and non-reducing SDS-PAGE gels.
  • ThioFabs L-V15C, L-Vl IOC, H-A88C, and H-A121C were expressed and purified by Protein-G SEPHAROSETM column chromatography (see methods sections for details). Purified proteins were analyzed on SDS-PAGE gel in reducing (with DTT) and non-reducing (without DTT) conditions. Other reducing agents such as BME (beta-mercaptoethanol) can used in the gel to cleave interchain disulfide groups. It is evident from SDS-PAGE gel analysis that the major (-90%) fraction of ThioFab is in the monomeric form, while wild type hu4D5Fabv8 is essentially in the monomeric form (47 kDa).
  • ThioFab (A121C) and wild type hu4D5Fabv8 were incubated with 100 fold excess of biotin-maleimide for 3 hours at room temperature and the biotinylated Fabs were loaded onto a Superdex-200TM gel filtration column. This purification step was useful in separating monomeric Fab from oligomeric Fab and also from excess free biotin-maleimide (or free zirconium reagent).
  • FIG. 5 shows validation of the properties of ThioFab variants in the absence of the phage context.
  • the proteins without phage fusion, hu4D5Fabv8 and hu4D5Fabv8-A121C (ThioFab-A121C) were expressed and purified using protein-G agarose beads followed by incubation with 100 fold molar excess of biotin-maleimide.
  • Streptavidin and HER2 binding of a biotinylated cys engineered ThioFab and a non-biotinylated wild type Fab was compared.
  • the extent of biotin conjugation (interaction with streptavidin) and their binding ability to HER2 were monitored by ELISA analyses. Each Fab was tested at 2ng and 20ng.
  • Biotinylated A121C ThioFab retained comparable HER2 binding to that of wild type hu4D5Fabv8 (Figure 5).
  • Wild type Fab and A121C-ThioFab were purified by gel filtration column chromatography. The two samples were tested for HER2 and streptavidin binding by ELISA using goat anti-Fab-HRP as secondary antibody. Both wild type (open box) and
  • ThioFab (dotted box) have similar binding to HER2 but only ThioFab retained streptavidin binding. Only a background level of interaction with streptavidin was observed with non- biotinylated wild type hu4D5Fabv8 ( Figure 5).
  • Plasma-protein binding can be an effective means of improving the pharmacokinetic properties of short lived molecules.
  • Albumin is the most abundant protein in plasma.
  • Serum albumin binding peptides can alter the pharmacodynamics of fused active domain proteins, including alteration of tissue uptake, penetration, and diffusion.
  • albumin binding peptide sequence US 20040001827.
  • a series of albumin binding peptides were identified by phage display screening (Dennis et al. (2002) “Albumin Binding As A General Strategy For Improving The Pharmacokinetics Of Proteins” J Biol Chem. 277:35035-35043; WO 01/45746).
  • Compounds of the invention include ABP sequences taught by: (i) Dennis et al (2002) J Biol Chem.
  • Albumin Binding (ABP)-Fabs were engineered by fusing an albumin binding peptide to the C-terminus of Fab heavy chain in 1 : 1 stoichiometric ratio (1 ABP / 1 Fab). It was shown that association of these ABP-Fabs with albumin increased their half life by more than 25 fold in rabbits and mice. The above described reactive Cys residues can therefore be introduced in these ABP-Fabs and used for site-specific conjugation with zirconium reagents followed by in vivo animal studies.
  • Exemplary albumin binding peptide sequences include, but are not limited to the amino acid sequences listed in SEQ ID NOS: 1-5:
  • the albumin binding peptide does not compete with ligands known to bind albumin and has a half life (T1 ⁇ 2) in rabbit of 2.3 hr.
  • ABP-ThioFab proteins were purified on BSA-SEPHAROSETM followed by biotin-maleimide conjugation and purification on Superdex-S200 column chromatography as described in previous sections. Purified biotinylated proteins were homogeneous and devoid of any oligomeric forms (Example 4).
  • FIG. 6 shows the properties of Albumin Binding Peptide (ABP)-ThioFab variants.
  • ELISA analyses were carried out to test the binding ability of ABP-hu4D5Fabv8-wt, ABP- hu4D5Fabv8-Vl IOC and ABP-hu4D5Fabv8-A121C with rabbit albumin, streptavidin and HER2.
  • Biotinylated ABP-ThioFabs are capable of binding to albumin and HER2 with similar affinity to that of wild type ABP-hu4D5Fabv8 as confirmed by ELISA ( Figure 6) and BIAcore binding kinetics analysis (Table 3).
  • An ELISA plate was coated with albumin, HER2 and SA as described.
  • Biotinylated ABP-ThioFabs were capable of binding to streptavidin compared to non biotinylated control ABP-hu4D5Fabv8-wt indicating that ABP- ThioFabs were conjugated with biotin maleimide like ThioFabs in a site specific manner as the same Cys mutants were used for both the variants (Figure 6).
  • Table 3 BIAcore kinetic analysis for HER2 and rabbit albumin binding to biotinylated
  • ABP albumin binding peptide
  • an albumin-binding peptide may be linked to the antibody by covalent attachment through a linker moiety.
  • HER2/Fab, SA/Fab and SA/SA indicate that their interactions were monitored by anti-Fab-HRP, SA- HRP, respectively.
  • SA/Fab monitors the presence of single biotin per Fab and more than one biotin per Fab is monitored by SA/SA analysis. Binding of HER2 with double cys mutants is similar to that of single Cys variants ( Figure 7). However the extent of biotinylation on double Cys mutants was higher compared to single Cys variants due to more than one free thiol group per Fab molecule ( Figure 7).
  • the single cys mutants H-A88C, H- A121C and L-Vl IOC of trastuzumab, and double cys mutants VI 10C-A121C and VI 10C- A121C of trastuzumab were expressed in CHO (Chinese Hamster Ovary) cells by transient fermentation in media containing 1 mM cysteine.
  • the A88C mutant heavy chain sequence (450 aa) is SEQ ID NO:6.
  • the A121C mutant heavy chain sequence (450 aa) is SEQ ID NO:7.
  • the VI IOC mutant light chain sequence (214 aa) is SEQ ID NO:8.
  • the cysteine engineered thio-trastuzumab antibodies comprise one or more of the following variable region heavy chain sequences with a free cysteine amino acid (SEQ ID NOS: 9-16). Mutant Sequence SEQ ID NO
  • cysteine engineered thio-trastuzumab antibodies comprise one or more of the following variable region light chain sequences with a free cysteine amino acid (SEQ ID NOS: 17-27).
  • FIG. 13 A shows a cartoon depiction of biotinylated antibody binding to immobilized HER2 and HRP labeled secondary antibody for absorbance detection.
  • Figure 13B shows binding measurements to immobilized HER2 with detection of absorbance at 450 nm of (left to right): non-biotinylated wild type trastuzumab (Wt), biotin- maleimide conjugated thio-trastuzumab variants VI IOC (single cys), A121C (single cys), and VI 10C-A121C (double cys).
  • Wt non-biotinylated wild type trastuzumab
  • VI IOC sino cys
  • A121C single cys
  • VI 10C-A121C double cys
  • Figure 14A shows a cartoon depiction of a biotinylated antibody binding to immobilized HER2 with binding of biotin to anti-IgG-HRP for absorbance detection.
  • Figure 14B shows binding measurements with detection of absorbance at 450 nm of biotin- maleimide conjugated thio-trastuzumab variants and non-biotinylated wild type trastuzumab in binding to streptavidin. From left to right: VI IOC (single cys), A121C (single cys), VI 10C/A121C (double cys), and trastuzumab. Each thio IgG trastuzumab variant and parent trastuzumab was tested at 1, 10, and 100 ng. The measurements show that the HER2
  • ThioMabs have high thiol reactivity.
  • Cysteine was introduced into the full-length 2H9 anti-EphB2R antibody at certain residues.
  • the single cys mutant H-A121C of 2H9 was expressed in CHO (Chinese Hamster Ovary) cells by transient fermentation in media containing 1 mM cysteine.
  • the A121C 2H9 mutant heavy chain sequence (450 aa) is SEQ ID NO:28.
  • Cysteine engineered thio-2H9 antibodies comprise the following Fc constant region heavy chain sequences with a free cysteine amino acid (SEQ ID NOS: 29-38).
  • Figure 16 shows non-reducing (top) and reducing (bottom) denaturing SDS-PAGE (polyacrylamide gel electrophoresis) analysis of 2H9 ThioMab Fc variants (left to right, lanes 1-9): A339C; S337C; S324C; A287C; V284C; V282C; V279C; and V273C, with 2H9 wild type, after purification on immobilized Protein A.
  • the lane on the right is a size marker ladder, indicating the intact proteins are about 150 kDa, heavy chain fragments about 50 kDa, and light chain fragments about 25 kDa.
  • Figure 17A shows non-reducing (left) and reducing (right) denaturing polyacrylamide gel electrophoresis analysis of 2H9 ThioMab variants (left to right, lanes 1-4): L-V15C; S179C; S375C; S400C, after purification on immobilized Protein A.
  • Figure 17B shows non-reducing (left) and reducing (+DTT) (right) denaturing polyacrylamide gel electrophoresis analysis of additional 2H9 and 3A5 ThioMab variants after purification on immobilized Protein A.
  • the 2H9 ThioMab variants (in the Fab as well as Fc region) were expressed and purified as described.
  • all the proteins are homogenous on SDS-PAGE followed by the reduction and oxidation procedure of Example 11 to prepare reactive ThioMabs for conjugation (Example 12).
  • Cysteine was introduced into the full-length 3A5 anti-MUC16 antibody at certain residues.
  • the single cys mutant H-A121C of 3A5 was expressed in CHO (Chinese Hamster Ovary) cells by transient fermentation in media containing 1 mM cysteine.
  • the A121C 3A5 mutant heavy chain sequence (446 aa) comprises SEQ ID NO:39.
  • Cysteine engineered thio-3A5 anti-MUC16 antibodies comprise the following variable region heavy chain sequences with a free cysteine amino acid (SEQ ID NOS: 40-44). Mutant Sequence SEQ ID NO:
  • Cysteine engineered thio-3A5 anti-MUC16 antibodies comprise the following variable region light chain sequences with a free cysteine amino acid (SEQ ID NOS: 45-49).
  • the thiol reactivity of full length, IgG cysteine engineered antibodies was measured by biotinylation and streptavidin binding.
  • a western blot assay was set up to screen the ThioMab that is specifically conjugated with biotin-maleimide.
  • the antibodies are analyzed on reducing SDS-PAGE and the presence of Biotin is specifically probed by incubating with streptavidin-HRP.
  • streptavidin-HRP interaction is either observed in heavy chain or light chain depending on which engineered cys variant is being used and no interaction is seen with wild type, indicating that ThioMab variants specifically conjugated the biotin at engineered Cys residue.
  • Figure 18 shows denaturing gel analysis of reduced, biotinylated Thio-IgG variants after capture on immobilized anti-IgG-HRP (top gel) and streptavidin-HRP (bottom gel).
  • Lane 1 3A5 H- A121C.
  • Lane 2 3A5 L-V110C.
  • Lane 3 2H9 H-A121C.
  • Lane 4 2H9 L-V110C.
  • Lane 5 anti-EphB2R 2H9 parent, wild type.
  • Each mutant (lanes 1-4) was captured by anti-IgG with HRP detection (top) indicating that selectivity and affinity were retained.
  • Capture by immobilized streptavidin with HRP detection (bottom) confirmed the location of biotin on heavy and light chains.
  • the location of cysteine mutation on the cysteine engineered antibodies in lanes 1 and 3 is the heavy chain.
  • the location of cysteine mutation on the cysteine engineered antibodies in lanes 2 and 4 is the light chain.
  • the cysteine mutation site undergoes conjugation with the biotin-maleimide reagent.
  • Analysis of the ThioMab cysteine engineered antibodies of Figure 18 and a 2H9 V15C variant by LC/MS gave quantitative indication of thiol reactivity (Table 5).
  • Cysteine engineering was conducted in the constant domain, i.e. Fc region, of IgG antibodies. A variety of amino acid sites were converted to cysteine sites and the expressed mutants, i.e. cysteine engineered antibodies, were assessed for their thiol reactivity.
  • FIG. 19 schematic diagram, the streptavidin-biotin interaction is monitored by probing with anti-IgG-HRP followed by measuring absorbance at 450 nm.
  • These results confirmed 2H9- ThioFc variants V282C, A287C, A339C, S375C and S400C had moderate to highest Thiol reactivity.
  • the extent of biotin conjugation of 2H9 ThioMab Fc variants was quantitated by LS/MS analysis as reported in Table 6.
  • the LS/MS analysis confirmed that the A282C, S375C and S400C variants had 100% biotin conjugation and V284C and A339C had 50% conjugation, indicating the presence of a reactive cysteine thiol group.
  • the other ThioFc variants, and the parent, wild type 2H9 had either very little biotinylation or none.
  • Desferoxamine B ( ⁇ '- ⁇ 5-[acetyl(hydroxy)amino]pentyl ⁇ -N-[5-( ⁇ 4-[(5- aminopentyl)(hydroxy)amino]-4-oxobutanoyl ⁇ amino)pentyl]-N-hydroxysuccinamide (CAS Reg. No. 70-51-9); and also known as Deferoxamine, desferoxamine B, DFO-B, DFOA, DFB or desferal) is a bacterial siderophore produced by the actinobacter Streptomyces pilosus ( Figure 20 top). Desferoxamine B has medical applications as a chelating agent used to remove excess iron from the body (Miller, Marvin J.
  • TFP-N-Suc-Df Figure 20 center
  • mAb-N-Suc-Df was chelated with 89 Zr.
  • Embodiments of zirconium complexes also include zirconium-binding (chelating) ligands such as DTP A (CAS Reg. No. 67-43-6), DOP A(l, 4,7,10-tetraazacyclododecane- 1,4,7,10-tetraacetic acid) (Liu, Shuang (2008) Advanced Drug Delivery Reviews 60(12): 1347-1370), cyclopentadienyl, and allyl groups (Erker, G. (1991) Pure and Applied Chemistry 63(6):797-806; Erker, G. (1990) Jour, of Organometallic Chem. 400(1-2): 185- 203), each of which are incorporated by reference herein.
  • DTP A CAS Reg. No. 67-43-6
  • DOP A(l, 4,7,10-tetraazacyclododecane- 1,4,7,10-tetraacetic acid) Liu, Shuang (2008) Advanced Drug Delivery Reviews 60(12): 1347-1370
  • Zirconium complexes (Z) and other radionuclides may be conjugated to antibodies (Ab), including monoclonal antibodies (mAbs) through ⁇ -amino group in lysine side chain or through thiol group of cysteine. Since approximately 40 lysine side chains (Wang L et al "Structural characterization of the maytansinoid-monoclonal antibody immunoconjugate, huN901-DMl, by mass spectrometry” (2005) Protein Sci. 14:2436-46) or 8 cysteines (Hamblett KJ et al. "Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate" (2004) Clin Cancer Res.
  • THlOMABs site-specifically radiolabeled cysteine- engineered antibodies
  • One aspect of the present invention is a method for site-specific radiolabeling of THIOMABs using novel Df-based thiol reactive bifunctional reagents
  • trastuzumab trastuzumab
  • Zr zinc-reacted calcium
  • One metastable isomer of zirconium is 89 Zr with a half-life of 78.4 hours with decay modes of beta (electron emission), positron (beta plus), and gamma radiation.
  • radioisotope or other labels may be incorporated in the conjugate in known ways (Fraker et al (1978) Biochem. Biophys. Res. Commun. 80: 49-57; "Monoclonal Antibodies in Immunoscintigraphy” Chatal, CRC Press 1989).
  • Carbon- 14-labeled 1-isothiocyanatobenzyl- 3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of a radionuclide to the antibody (WO 94/11026).
  • a “Linker” (L) is a bifunctional or multifunctional moiety which can be used to link one or more zirconium complex moieties (Z) and an antibody unit (Ab) to form antibody- zirconium conjugates (AZC) of Formula I.
  • Antibody-zirconium conjugates (AZC) can be conveniently prepared using a Linker having reactive functionality for binding to zirconium and to the Antibody.
  • a cysteine thiol of a cysteine engineered antibody (Ab) can form a bond with a functional group of a linker reagent, a zirconium label moiety or zirconium- linker intermediate.
  • a Linker has a reactive site which has an electrophilic group that is reactive to a nucleophilic cysteine present on an antibody.
  • the cysteine thiol of the antibody is reactive with an electrophilic group on a Linker and forms a covalent bond to a Linker.
  • Useful electrophilic groups include, but are not limited to, maleimide and haloacetamide groups.
  • Cysteine engineered antibodies may react with linker reagents or zirconium-linker intermediates, with electrophilic functional groups such as maleimide or a-halo carbonyl, according to the conjugation method at page 766 of Klussman, et al (2004), Bioconjugate Chemistry 15(4):765-773, and according to the protocols of Examples 17-19.
  • the Z moieties are the same.
  • Z moieties are different.
  • Exemplary embodiments of the Formula I antibody-zirconium conjugate (AZC) compounds include:
  • R is independently H or Ci-C 6 alkyl; and n is 1 to 12.
  • a Linker has a reactive functional group which has a nucleophilic group that is reactive to an electrophilic group present on an antibody.
  • Useful electrophilic groups on an antibody include, but are not limited to, aldehyde and ketone carbonyl groups.
  • the heteroatom of a nucleophilic group of a Linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit.
  • Useful nucleophilic groups on a Linker include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide.
  • the electrophilic group on an antibody provides a convenient site for attachment to a Linker.
  • the Linker may be substituted with groups which modulated solubility or reactivity.
  • a charged substituent such as sulfonate (-SO 3 ) or ammonium, may increase water solubility of the reagent and facilitate the coupling reaction of the linker reagent with the antibody or the zirconium moiety, or facilitate the coupling reaction of Ab-L (antibody-linker intermediate) with Z, or Z-L (zirconium-linker
  • the compounds of the invention expressly contemplate, but are not limited to, AZC prepared with linker reagents: BMPEO, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4- vinylsulfone)benzoate), and including bis-maleimide reagents: DTME, BMB, BMDB, BMH, BMOE, BM(PEO) 2 , and BM(PEO) 4 3, which are commercially available from Pierce
  • Bis-maleimide reagents allow the attachment of the thiol group of a cysteine engineered antibody to a thiol-containing zirconium moiety, label, or linker intermediate, in a sequential or concurrent fashion.
  • Other functional groups besides maleimide, which are reactive with a thiol group of a cysteine engineered antibody, zirconium moiety, label, or linker intermediate include iodoacetamide, bromoacetamide, vinyl pyridine, disulfide, pyridyl disulfide isocyanate, and isothiocyanate.
  • linker reagents can also be obtained via other commercial sources, such as Molecular Biosciences Inc. (Boulder, CO), or synthesized in accordance with procedures described in Toki et al (2002) J. Org. Chem. 67: 1866-1872; Walker, M.A. (1995) J. Org. Chem. 60:5352-5355; Frisch et al (1996) Bioconjugate Chem. 7: 180-186; US 6214345; WO 02/088172; US 2003130189; US2003096743; WO 03/026577; WO 03/043583; and WO 04/032828.
  • Exemplary linker reagents include:
  • n is an integer ranging from 1-10 and T is -H or -S0 3 Na;
  • n is an integer ranging from 0-3;
  • linker L may be a dendritic type linker for covalent attachment of more than one zirconium moiety through a branching, multifunctional linker moiety to an antibody (Sun et al (2002) Bioorganic & Medicinal Chemistry Letters 12:2213- 2215; Sun et al (2003) Bioorganic & Medicinal Chemistry 11 : 1761-1768).
  • Dendritic linkers can increase the molar ratio of zirconium to antibody, i.e. loading of the AZC.
  • a cysteine engineered antibody bears only one reactive cysteine thiol group, a multitude of zirconium moieties may be attached through a dendritic linker.
  • branched, dendritic linkers include those with self-immolative 2,6-bis(hydroxymethyl)-p-cresol and 2,4,6-tris(hydroxymethyl)-phenol dendrimer units (WO 2004/01993; Szalai et al (2003) J. Amer. Chem. Soc. 125: 15688-15689; Shamis et al (2004) J. Amer. Chem. Soc. 126: 1726-1731; Amir et al (2003) Angew. Chem. Int. Ed. 42:4494- 4499).
  • An aspect of the invention is a desferrioxamine-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
  • L-Df is selected from:
  • p 1 to 4.
  • the antibody-zirconium conjugates (AZC) of Formula I may be prepared by several routes, employing organic chemistry reactions, conditions, and reagents known to those skilled in the art, including: (1) reaction of a cysteine group of a cysteine engineered antibody with a linker reagent, to form antibody-linker intermediate Ab-L, via a covalent bond, followed by reaction with an activated zirconium label moiety Z; and (2) reaction of a nucleophilic group of a zirconium moiety with a linker reagent, to form zirconium label- linker intermediate Z-L, via a covalent bond, followed by reaction with a cysteine group of a cysteine engineered antibody. Conjugation methods (1) and (2) may be employed with a variety of cysteine engineered antibodies, zirconium label moieties, and linkers to prepare the antibody-zirconium conjugates of Formula I.
  • Antibody cysteine thiol groups are nucleophilic and capable of reacting to form covalent bonds with electrophilic groups on linker reagents and zirconium-linker
  • intermediates including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides, such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups; and (iv) disulfides, including pyridyl disulfides, via sulfide exchange.
  • active esters such as NHS esters, HOBt esters, haloformates, and acid halides
  • alkyl and benzyl halides such as haloacetamides
  • aldehydes ketones
  • carboxyl and maleimide groups
  • disulfides including pyridyl disulfides, via sulfide exchange.
  • Nucleophilic groups on a zirconium lable moiety include, but are not limited to: amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide groups capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents.
  • the cysteine engineered antibodies may be made reactive for conjugation with linker reagents by treatment with a reducing agent such as DTT (Cleland's reagent, dithiothreitol) or TCEP (tris(2-carboxyethyl)phosphine hydrochloride; Getz et al (1999) Anal. Biochem. Vol 273:73-80; Soltec Ventures, Beverly, MA).
  • a reducing agent such as DTT (Cleland's reagent, dithiothreitol) or TCEP (tris(2-carboxyethyl)phosphine hydrochloride; Getz et al (1999) Anal. Biochem. Vol 273:73-80; Soltec Ventures, Beverly, MA).
  • a reducing agent such as DTT (Cleland's reagent, dithiothreitol) or TCEP (tris(2-carboxyethyl)phosphine hydroch
  • the reduced ThioMab was diluted and loaded onto HiTrap S column in 10 mM sodium acetate, pH 5, and eluted with PBS containing 0.3M sodium chloride. Disulfide bonds were reestablished between cysteine residues present in the parent Mab with dilute (200 nM) aqueous copper sulfate (CuS0 4 ) at room temperature, overnight.
  • Other oxidants i.e.
  • oxidizing agents and oxidizing conditions, which are known in the art may be used.
  • Ambient air oxidation is also effective. This mild, partial reoxidation step forms intrachain disulfides efficiently with high fidelity.
  • An approximate 10 fold excess of zirconium-linker intermediate is added, mixed, and let stand for about an hour at room temperature to effect conjugation and form the ThioMab antibody-zirconium conjugate.
  • the conjugation mixture is gel filtered and loaded and eluted through a HiTrap S column to remove excess zirconium- linker intermediate and other impurities.
  • Figure 15 shows the general process to prepare a cysteine engineered antibody expressed from cell culture for conjugation.
  • Cysteine adducts presumably along with various interchain disulfide bonds, are reductively cleaved to give a reduced form of the antibody.
  • the interchain disulfide bonds between paired cysteine residues are reformed under partial oxidation conditions, such as exposure to ambient oxygen.
  • the newly introduced, engineered, and unpaired cysteine residues remain available for reaction with linker reagents or zirconium-linker intermediates to form the antibody conjugates of the invention.
  • the ThioMabs expressed in mammalian cell lines result in externally conjugated Cys adduct to an engineered Cys through -S-S- bond formation. Hence the purified
  • ThioMabs have to be treated with reduction and oxidation procedures as described in Example 11 to produce reactive ThioMabs. These ThioMabs are used to conjugate with maleimide containing radiolabels, cytotoxic drugs, fluorophores, and other labels.
  • TFP-N-SucDf-Fe The protected active ester TFP-N-SucDf-Fe was prepared according to the previously described procedure (Verel I et al "89Zr Immuno-PET: Comprehensive Procedures For The Production Of 89Zr-Labeled Monoclonal Antibodies” (2003) J Nucl Med 44: 1271-81) and conjugated to trastuzumab using a 5-fold molar excess of TFP-N-SucDf-Fe to yield N-SucDf- trastuzumab with an average of 1.6 molecules of desferoxamine (Table 8).
  • Df-Bz-SCN- trastuzumab was obtained by coupling an 8-fold molar excess of Df-Bz-SCN at pH 8.5 (Perk LR, et al. "Facile radiolabeling of monoclonal antibodies and other proteins with zirconium- 89 or gallium-68 for PET Imaging using p-isothiocyanatobenzyl-desferrioxamine” (2008) Nature Protocols; published online:DOI: 10.1038/nprot.2008.22).
  • the reaction provided Df- Bz-SCN-trastuzumab decorated in average with 2.4 molecules of desferoxamine (Table 1).
  • novel maleimide based thiol reactive bifunctional linker Df-Chx-Mal was prepared from equimolar amounts of desferoxamine mesylate and SMCC ( Figure 21,
  • Example 13 The reaction was complete within 30 min at room temperature and the product was isolated by precipitation upon addition of water in 45% yield and more than 95% purity.
  • the reaction of an 8.5-fold molar excess of Df-Chx-Mal with freshly prepared thio- trastuzumab (Figure 21 Example 17) provided Df-Chx-Mal-thio-trastuzumab conjugate with exactly 2 molecules of desferoxamine in 1 h (Table 1, Figure 21). Bromoacetyl
  • BDf-Bac desferoxamine
  • Df-Iac desferoxamine mesylate and bromoacetyl bromide at 0°C
  • the product was obtained in 14% yield after HPLC purification.
  • the alkylation of freshly prepared thio- trastuzumab ( Figure 21, Example 16) with a 12-fold molar excess of Df-Bac provided the conjugate (Df-Ac-thio-trastuzumab) with 1.8 molecules of Df per antibody within 5 h (Table 8, Figure 21, Example 18).
  • the low reactivity of bromide prompted us to explore the more reactive iodoacetyl derivative (Df-Iac).
  • Df-Iac was prepared in 53% yield by the reaction of desferoxamine mesylate with a slight excess of N-hydroxysuccinimidyl iodoacetate ( Figure 21, Example 15). The product was obtained in more than 95% purity by precipitation from the reaction mixture. The subsequent reaction of an 11 -fold excess of Df-Iac provided Df-Ac- thio-trastuzumab decorated with 1.8 molecules of Df within 2 h (Table 1, Figure 21, Example 19). Based on our experience, Df-Chx-Mal is the preferred reagent of the three compounds investigated.
  • the Zr was chelated as 89-zirconium oxalatewith all four variants of Df- trastuzumab using previously described experimental procedure (Verel I et al "89Zr immuno- PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies” (2003) J Nucl Med. 44: 1271-81).
  • the radiolabeled proteins were purified on a desalting column and the final solution was concentrated to the required volume by membrane
  • the yield, purity, and final specific activity of the Zr conjugates are summarized in Table 9.
  • the chelation yield was over 80% with the exception of the Df-N-Suc linker obtained in lower yield presumably due to the lower amount of Df per antibody molecule and/or incomplete removal of Fe(III) used to protect the chelator during activation and conjugation.
  • the product purity was over 90% with a small amount (1-6%) of high molecular weight
  • Df-Chx-Mal-thio-trastuzumab provided the Zr complex in 99% purity (Table 9) as opposed to Df-Ac conjugate which was contaminated with approximately 8% of a low molecular weight impurity and 2% of high molecular weight aggregates.
  • the contaminant resisted removal using NAP- 10 column but the removal was possible using repeated buffer exchange on Amicon filter.
  • the obtained K D values were compared to non-modified trastuzumab (0.91 ⁇ 0.20 nM).
  • the K D for the thio-trastuzumab conjugate containing Chx-Mal linker was 0.93 ⁇ 0.15 nM and the values for the conjugates containing Ac linker were 1.22 ⁇ 0.22 nM for conjugate prepared using Df-Bac and 0.87 ⁇ 0.15 nM prepared using Df-Iac.
  • the results of the biological activity analyses indicate that the modification of thio-trastuzumab did not affect the binding affinity of the antibody to HER2.
  • Fig. 3 The Zr-trastuzumab uptake in selected tissues is summarized in Fig 4.
  • the images at 1 h (not shown) were dominated by the high blood pool uptake with the exception of Df-Bac where rapid hepatobiliary excretion of the lipophilic impurity resulted in an elevated uptake in intestine.
  • the impurity was totally cleared within the first 24 h and elevated small and large intestine uptake was not detected at 24 h or later time after tracer injection.
  • the tissue uptake of Df-Ac conjugate was resultantly slightly (-8%) lower, the tumor to blood ratios (Table 10) were not affected by the loss of injected radioactivity.
  • the images at 96 h were dominated by the high tumor uptake
  • BT474 (3+ expression level of HER2) xenografts exhibited lower absolute uptake of the tracer (15 %ID/g) than measured previously by Dijkers et al in SKOV3 (3+ expression level of HER2) 33.4 ⁇ 7.7%ID/g (Dijkers EC, et al. "Development and Characterization of Clinical-Grade 89Zr-Trastuzumab for HER2/neu ImmunoPET Imaging" (2009) J Nucl Med 50(6):974-981).
  • the tumor to blood ratio of 5.7-7.1 (Table 10) is comparable to the value obtained with SKOV3 (tumor to blood of 7.6).
  • the difference in tumor uptake may be attributed to the tumor model and total dose of trastuzumab.
  • a material with higher specific activity was used hence so significantly less antibody was injected (35 ⁇ g, 1.4 mg/kg) compared to the Dijkers et al study with SKOV3 (100 ⁇ g, 4 mg/kg).
  • the difference in trastuzumab was attributed to the tumor model and total dose of trastuzumab.
  • the bone uptake may originate from the breakdown of the Zr-
  • thiol specific reagents Three thiol specific reagents are exemplified herein for the chemoselective conjugation of desferrioxamine (Df) to monoclonal antibodies through the thiol group of cysteine of cysteine-engineered antibodies.
  • the thiol-specific Df-reagents were obtained by the acylation of the amino group of desferrioxamine B in 14% (Df-Bac), 53% (Df-Iac) and 45% (Df-Chx-Mal) yields and conjugated to thio-trastuzumab resulting in site-specific modification on both engineered cysteines within 1-5 h.
  • the binding activities of site-specific thio-trastuzumab conjugates to HER2 were identical to the activity of non-modified trastuzumab.
  • the Df-modified thio-trastuzumabs (Df-Ac-thio-trastuzumab and Df-Chx-Mal-
  • Df-Chx-Mal is a useful reagent for conjugation of Df to antibodies through cysteine side chain and showed several advantages over Df-Bac and Df-Iac.
  • moderate pH 7.5 was required for complete conjugation of Df-Chx-Mal within 1 h as compared to pH 9 and 2 or 5 h required for Df-Bac
  • the site specifically Zr labeled engineered THIOMAB conjugates can be used similarly as 18 F labeled THIOFAB conjugates (Gill HS, et al. "A modular platform for the rapid site-specific radiolabeling of proteins with 18F exemplified by quantitative positron emission tomography of human epidermal growth factor receptor 2" (2009) Jour, of Med. Chem. 52:5816-25) as valuable tools for PET imaging applications in biomedical research.
  • the antibody-zirconium conjugates (AZC) of the invention may be administered by any route appropriate to the condition to be treated.
  • the AZC will typically be administered parenterally, i.e. infusion, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural.
  • compositions of diagnostic antibody-zirconium conjugates (AZC) of the invention are typically prepared for parenteral administration, i.e. bolus, intravenous, intratumor injection with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form.
  • An antibody-zirconium conjugate (AZC) having the desired degree of purity is optionally mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation or an aqueous solution.
  • Acceptable diluents, carriers, excipients, and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparag
  • Zn-protein complexes Zn-protein complexes
  • non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol (PEG).
  • TWEENTM TWEENTM
  • PLURONICSTM polyethylene glycol
  • PEG polyethylene glycol
  • the active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in
  • sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi permeable matrices of solid hydrophobic polymers containing the AZC, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or polyvinyl alcohol)), polylactides (US 3773919), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid.
  • polyesters for example, poly(2-hydroxyethyl-methacrylate), or polyvinyl alcohol
  • polylactides US 3773919
  • copolymers of L-glutamic acid and gamma-ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • the formulations to be used for in vivo administration must be sterile, which is readily accomplished by filtration through sterile filtration membranes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Optics & Photonics (AREA)
  • Physics & Mathematics (AREA)
  • Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Inorganic Chemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Biotechnology (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)

Abstract

Antibodies are engineered by replacing one or more amino acids of a parent antibody with non cross-linked, highly reactive cysteine amino acids. Antibody fragments may also be engineered with one or more cysteine amino acids to form cysteine engineered antibody fragments (ThioFab). Methods of design, preparation, screening, and selection of the cysteine engineered antibodies are provided. Cysteine engineered antibodies (Ab) are conjugated with one or more zirconium complex (Z) labels through a linker (L) to form cysteine engineered zirconium-labeled antibody conjugates having Formula I: where p is 1 to 4. Imaging methods and diagnostic uses for zirconium-radiolabeled, cysteine engineered antibody conjugate compositions are disclosed.

Description

ZIRCONIUM-RADIOLABELED, CYSTEINE ENGINEERED ANTIBODY
CONJUGATES
This application claims priority to US Ser. No. 12/612,912 filed November 5, 2009 which is a continuation-in-part of US Ser. No. 12/399,241 filed on March 6, 2009 which is a continuation of US Ser. No. 11/233,258 filed on September 22, 2005, now US 7,521,541 issued April 21, 2009, and also claims the benefit of priority under 35 USC § 119(e) of US Provisional Application Ser. No. 60/612,468 filed on September 23, 2004 and US Provisional Application Ser. No. 60/696,353 filed on June 30, 2005, each of which are incorporated by reference in their entirety.
FIELD OF THE INVENTION
The invention relates generally to antibodies engineered with reactive cysteine residues and more specifically to antibodies with therapeutic or diagnostic applications. The cysteine engineered antibodies may be conjugated with chemotherapeutic drugs, toxins, affinity ligands such as biotin, and detection labels such as radioisotopes and fluorophores. The invention also relates to methods of using antibodies and antibody-drug conjugate compounds for in vitro, in situ, and in vivo diagnosis or treatment of mammalian cells, or associated pathological conditions.
BACKGROUND OF THE INVENTION
Molecular imaging is an important tool in the development and evaluation of novel pharmaceuticals. Immuno-positron emission tomography (ImmunoPET) is a rapidly emerging method for tracking and quantifying monoclonal antibodies (mAbs) in vivo as it efficiently combines the high sensitivity of PET with the high specificity of mAbs.
ImmunoPET aspires to be the clinical method of choice for non-invasive diagnosis providing "comprehensive immunohistochemical staining in vivo" (van Dongen GA, et al. "ImmunoPET: a navigator in monoclonal antibody development and applications" Oncologist
2007;12: 1379-89). Since ImmunoPET requires a positron-emitting radioisotope to be coupled to a target specific molecule it is essential to match the biological half-life of the molecule with the half-life of the radionuclide (Verel I, et al. "The promise of immuno-PET in radioimmunotherapy" J Nucl Med 2005;46 Suppl 1 : 164S-71S). Although antibodies (-150 kDa) have plasma half-life ranging from days to weeks, imaging typically provides maximum target-to-background ratios 2-6 days after antibody-based tracer administration demanding
89 124
the use of radioisotopes such as Zr and I with half-life of 3.3 days and 4.2 days, respectively. Unfortunately, the half-life of readily available 64Cu (12.7 h) is too short to provide images with good contrast in this time frame.
The development of Positron Emission Tomographic (PET) imaging agents from a Mab template (Immuno-PET) holds promise as a tool for localizing and quantifying molecular targets and may enhance the non-invasive clinical diagnosis of pathological conditions (van Dongen et al (2007) Oncologist 12;1379-89; Williams et a (2001) Cancer Biother Radiopharm 16:25-35; Holliger et al (2005) Nat Biotechnol 23: 1126-36). PET is a molecular imaging technology that is increasingly used for detection of disease. PET imaging systems create images based on the distribution of positron-emitting isotopes in the tissue of a patient. The isotopes are typically administered to a patient by injection of probe molecules that comprise a positron-emitting isotope, such as F-18, C-l 1, N-13, or 0-15, covalently attached to a molecule that is readily metabolized or localized in the body (e.g., glucose) or that chemically binds to receptor sites within the body. In some cases, the isotope is administered to the patient as an ionic solution or by inhalation. Small immuno-PET imaging agents, such as Fab antibody fragments (50 kDa) or diabodies, paired dimers of the covalently associated VH-VL region of Mab, 55 kDa (Shively et al (2007) J Nucl Med 48: 170- 2), may be particularly useful since they exhibit a short circulation half-life, high tissue permeability, and reach an optimal tumor to background ratio between two to four hours after
18 injection facilitating the use of short half-life isotopes such as the widely available F (109.8 min).
124
Iodine 124 ( I) was coupled to antibody 3F9 and used to estimate the dosimetry for radioimmunotherapy of neuroblastoma (Larson SM, et al "PET scanning of iodine- 124-3 F9 as an approach to tumor dosimetry during treatment planning for radioimmunotherapy in a child with neuroblastoma" J Nucl Med 1992;33:2020-3). Later, as more sophisticated PET
124
instrumentation and improved techniques for radioiodination emerged, I was employed in numerous immunoPET studies (Verel I, et al "High-quality 1241-labelled monoclonal antibodies for use as PET scouting agents prior to 13 ll-radioimmunotherapy" European journal of nuclear medicine and molecular imaging 2004;31 : 1645-52; Lee FT et al
"Immuno-PET of human colon xenograft- bearing BALB/c nude mice using 124I-CDR- grafted humanized A33 monoclonal antibody" J Nucl Med 2001;42:764-9; Sundaresan G, et al. "1241-labeled engineered anti-CEA minibodies and diabodies allow high-contrast, antigen-specific small-animal PET imaging of xenografts in athymic mice" J Nucl Med 2003;44: 1962-9; Jain M and Batra SK. "Genetically engineered antibody fragments and PET imaging: a new era of radioimmunodiagnosis" J Nucl Med 2003;44: 1970-2; Gonzalez Trotter DE et al. "Quantitation of small-animal (124)1 activity distributions using a clinical PET/CT scanner" J Nucl Med 2004;45 : 1237-44; Robinson MK, et al. "Quantitative immuno- positron emission tomography imaging of HER2 -positive tumor xenografts with an iodine- 124 labeled anti-HER2 diabody" Cancer Res 2005;65: 1471-8; Jayson GC et al. "Molecular imaging and biological evaluation of HuMV833 anti-VEGF antibody: implications for trial design of antiangiogenic antibodies" J Natl Cancer Inst 2002;94: 1484-93; Divgi CR, et al. "Preoperative characterisation of clear-cell renal carcinoma using iodine- 124-labelled antibody chimeric G250 (124I-cG250) and PET in patients with renal masses: a phase I trial" Lancet Oncol 2007;8:304-10). Despite the relatively simple radioiodination techniques
124
available for coupling I onto mAbs, important limitations slow a widespread pre-clinical use of this radionuclide. Notably, the complex decay scheme involves energetic positrons (β+ max. 1.5 and 2.1 MeV) which negatively affects the resolution of small animal microPET.
Additionally, internalized iodinated proteins undergo enzymatic deiodination with free iodide rapidly cleared from the target cells providing PET images not reflective of the actual mAb uptake "Perera RM et al. "Internalization, intracellular trafficking, and biodistribution of monoclonal antibody 806: a novel anti-epidermal growth factor receptor antibody" Neoplasia (New York, N.Y 2007;9: 1099-110). The use of 89Zr overcomes these drawbacks as the
89 "
positrons emitted in Zr decay (β max. 897 keV) provide microPET resolution comparable
18 11 89
to F and C (around 1 mm). Also, the metabolites of internalized Zr-mAbs are
intracellularly trapped in lysosomes, providing better correlation of actual mAb uptake with PET imaging (van Dongen GA, et al. "Immuno-PET: a navigator in monoclonal antibody development and applications" Oncologist 2007;12: 1379-89).
Conventional means of attaching, i.e. linking through covalent bonds, a label, such as a radioisotope, fluorescent dye, or drug moiety, to an antibody generally leads to a heterogeneous mixture of molecules where the label moieties are attached at a number of sites on the antibody. For example, cytotoxic drugs have typically been conjugated to antibodies through the often-numerous lysine residues of an antibody, generating a heterogeneous antibody-drug conjugate mixture. Depending on reaction conditions, the heterogeneous mixture typically contains a distribution of antibodies with from 0 to about 8, or more, attached drug moieties. In addition, within each subgroup of conjugates with a particular integer ratio of drug moieties to antibody, is a potentially heterogeneous mixture where the drug moiety is attached at various sites on the antibody. Analytical and preparative methods are inadequate to separate and characterize the antibody-drug conjugate species molecules within the heterogeneous mixture resulting from a conjugation reaction.
Antibodies are large, complex and structurally diverse biomolecules, often with many reactive functional groups. Their reactivities with linker reagents and drug-linker
intermediates are dependent on factors such as pH, concentration, salt concentration, and co- solvents. Furthermore, the multistep conjugation process may be nonreproducible due to difficulties in controlling the reaction conditions and characterizing reactants and
intermediates.
Cysteine thiols are reactive at neutral pH, unlike most amines which are protonated and less nucleophilic near pH 7. Since free thiol (RSH, sulfhydryl) groups are relatively reactive, proteins with cysteine residues often exist in their oxidized form as disulfide-linked oligomers or have internally bridged disulfide groups. Extracellular proteins generally do not have free thiols (Garman, 1997, Non-Radioactive Labelling: A Practical Approach,
Academic Press, London , at page 55). The amount of free thiol in a protein may be estimated by the standard Ellman's assay. Immunoglobulin M is an example of a disulfide- linked pentamer, while immunoglobulin G is an example of a protein with internal disulfide bridges bonding the subunits together. In proteins such as this, reduction of the disulfide bonds with a reagent such as dithiothreitol (DTT) or selenol (Singh et al (2002) Anal.
Biochem. 304: 147-156) is required to generate the reactive free thiol. This approach may result in loss of antibody tertiary structure and antigen binding specificity.
Antibody cysteine thiol groups are generally more reactive, i.e. more nucleophilic, towards electrophilic conjugation reagents than antibody amine or hydroxyl groups. Cysteine residues have been introduced into proteins by genetic engineering techniques to form covalent attachments to ligands or to form new intramolecular disulfide bonds (Better et al (1994) J. Biol. Chem. 269(13):9644-9650; Bernhard et al (1994) Bioconjugate Chem. 5: 126- 132; Greenwood et al (1994) Therapeutic Immunology 1 :247-255; Tu et al (1999) Proc. Natl. Acad. Sci USA 96:4862-4867; Kanno et al (2000) J. of Biotechnology, 76:207-214; Chmura et al (2001) Proc. Nat. Acad. Sci. USA 98(15):8480-8484; US 6248564). However, designing in cysteine thiol groups by the mutation of various amino acid residues of a protein to cysteine amino acids is potentially problematic, particularly in the case of unpaired (free Cys) residues or those which are relatively accessible for reaction or oxidation. In
concentrated solutions of the protein, whether in the periplasm of E. coli, culture supernatants, or partially or completely purified protein, unpaired Cys residues on the surface of the protein can pair and oxidize to form intermolecular disulfides, and hence protein dimers or multimers. Disulfide dimer formation renders the new Cys unreactive for conjugation to a drug, ligand, or other label. Furthermore, if the protein oxidatively forms an intramolecular disulfide bond between the newly engineered Cys and an existing Cys residue, both Cys groups are unavailable for active site participation and interactions. Furthermore, the protein may be rendered inactive or non-specific, by misfolding or loss of tertiary structure (Zhang et al (2002) Anal. Biochem. 311 : 1-9).
Site-specific conjugation is preferred over random amino modification as it enables chemical modification of a site away from the binding site, promoting complete retention of biological activity and allowing control over the possible number of prosthetic groups added. Cysteine-engineered antibodies have been designed as FAB antibody fragments (thioFab) and expressed as full-length, IgG monoclonal (thioMab) antibodies. See: US 7521541; Junutula JR et al. "Rapid identification of reactive cysteine residues for site-specific labeling of antibody-Fabs" J Immunol Methods 2008;332:41-52; Junutula JR et al. "Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index" (2008) Nat Biotechnol. 26:925-32, the contents of which are incorporated by reference. ThioFab and ThioMab antibodies have been conjugated through linkers at the newly introduced cysteine thiols with thiol-reactive linker reagents and drug-linker reagents to prepare cysteine- engineered antibody drug conjugates (Thio ADC) with anti-cancer properties, including anti- MUC16 (US 2008/0311134), anti-CD22 (US 2008/0050310), anti-ROB04 (US
2008/0247951), anti-TENB2 (US 2009/0117100), anti-CD79B (US 2009/0028856; US 2009/0068202) Thio ADC.
SUMMARY
The compounds of the invention include cysteine engineered antibodies where one or more amino acids of a parent antibody are replaced with a free cysteine amino acid. A cysteine engineered antibody comprises one or more free cysteine amino acids having a thiol reactivity value in the range of 0.6 to l .O. A free cysteine amino acid is a cysteine residue which has been engineered into the parent antibody and is not part of a disulfide bridge.
Cysteine engineered antibodies may be useful in the diagnosis and treatment of cancer and include antibodies specific for cell surface and transmembrane receptors, and tumor-associated antigens (TAA). Such antibodies may be used as naked antibodies
(unconjugated to a drug or label moiety) or as antibody-zirconium conjugates (AZC). Embodiments of the methods for preparing and screening a cysteine engineered antibody include where the parent antibody is an antibody fragment, such as hu4D5Fabv8. The parent antibody may also be a fusion protein comprising an albumin-binding peptide sequence (ABP). The parent antibody may also be a humanized antibody selected from huMAb4D5-l, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (trastuzumab).
Cysteine engineered antibodies of the invention may be site-specifically and efficiently coupled with a thiol-reactive reagent. The thiol-reactive reagent may be a radioisotope reagent, multifunctional linker reagent, a capture label reagent, a fluorophore reagent, or a drug-linker intermediate.
The cysteine engineered antibody may be labeled with a detectable label, immobilized on a solid phase support and/or conjugated with a drug moiety.
Another aspect of the invention is a zirconium-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a zirconium complex (Z), having Formula I:
Figure imgf000008_0001
where p is 1 to 4.
Another aspect of the invention is a desferrioxamine-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
Ab-(L-Df)p II wherein L-Df is selected from:
Figure imgf000009_0001
where the wavy line indicates the attachment to the antibody (Ab); and
p is 1 to 4.
Another aspect of the invention is a desferrioxamine-labelling reagent selected from the stru tures:
Figure imgf000009_0002
Another aspect of the invention is a method of making a desferrioxamine-labelled, cysteine-engineered antibody comprising comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
Ab-(L-Df)p II
wherein L-Df is selected from:
Figure imgf000010_0001
where the wavy line indicates the attachment to the antibody (Ab); and
p is 1 to 4;
the method comprising reacting a composition selected from the structures:
Figure imgf000011_0001
wherein R is selected from:
Figure imgf000011_0002
with a cysteine-engineered antibody having one or more free cysteine amino acids, whereby the desferrioxamine-labelled, cysteine-engineered antibody is formed.
Another aspect of the invention is a method of making a zirconium-labelled, cysteine- engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a zirconium complex (Z), having Formula I:
Figure imgf000011_0003
where p is 1 to 4;
the method comprising complexing a zirconium reagent with a desferrioxamine- labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
Ab-(L-Df)p II wherein L-Df is selected from:
Figure imgf000012_0001
where the wavy line indicates the attachment to the antibody (Ab); and
p is 1 to 4;
whereby a desferrioxamine-labelled, cysteine-engineered antibody is formed.
Another aspect of the invention is a method of imaging comprising:
administering a zirconium-labelled, cysteine-engineered antibody to an animal; and detecting in vivo the presence of the zirconium-labelled, cysteine-engineered antibody by imaging,
wherein the zirconium-labelled, cysteine-engineered antibody comprises a cysteine engineered antibody (Ab) having one or more free cysteine amino acids conjugated with one or more zirconium complex (Z) through a linker (L), and having Formula I:
Figure imgf000012_0002
where p is 1 to 4.
Another aspect of the invention includes diagnostic uses for the compounds and compositions disclosed herein. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 A shows a three-dimensional representation of the hu4D5Fabv7 antibody fragment derived by X-ray crystal coordinates. The structure positions of the exemplary engineered Cys residues of the heavy and light chains are numbered (according to a sequential numbering system).
Figure IB shows a sequential numbering scheme (top row), starting at the N-terminus in comparison with the Kabat numbering scheme (bottom row) for 4D5v7fabH. Kabat numbering insertions are noted by a,b,c.
Figures 2 A and 2B show binding measurements with detection of absorbance at 450nm of hu4D5Fabv8 and hu4D5Fabv8 Cys mutant (ThioFab) phage variants: (A) non- biotinylated phage-hu4D5Fabv8 and (B) biotinylated phage-hu4D5Fabv8 (B) by the PHESELECTOR assay for interactions with BSA (open bar), HER2 (striped bar) or streptavidin (solid bar).
Figures 3A and 3B show binding measurements with detection of absorbance at 450nm of hu4D5Fabv8 (left) and hu4D5Fabv8 Cys mutant (ThioFab) variants: (A) non- biotinylated phage-hu4D5Fabv8 and (B) biotinylated phage-hu4D5Fabv8 by the PHESELECTOR assay for interactions with: BSA (open bar), HER2 (striped bar) and streptavidin (solid bar). Light chain variants are on the left side and heavy chain variants are on the right side. Thiol reactivity = OD450„ for streptavidin binding ÷ OD450 nm for HER2 (antibody) binding
Figure 4A shows Fractional Surface Accessibility values of residues on wild type
hu4D5Fabv8. Light chain sites are on the left side and heavy chain sites are on the right side.
Figure 4B shows binding measurements with detection of absorbance at 450nm of
biotinylated hu4D5Fabv8 (left) and hu4D5Fabv8 Cys mutant (ThioFab) variants for interactions with HER2 (day 2), streptavidin (SA) (day 2), HER2 (day 4), and SA (day 4). Phage-hu4D5Fabv8 Cys variants were isolated and stored at 4 °C. Biotin conjugation was carried out either at day 2 or day 4 followed by PHESELECTOR analyses to monitor their interaction with Her2 and streptavidin as described in Example 2, and probe the stability of reactive thiol groups on engineered ThioFab variants.
Figure 5 shows binding measurements with detection of absorbance at 450nm of biotin- maleimide conjugated-hu4D5Fabv8 (A121C) and non-biotinylated wild type hu4D5Fabv8 for binding to streptavidin and HER2. Each Fab was tested at 2 ng and 20 ng.
Figure 6 shows ELISA analysis with detection of absorbance at 450nm of biotinylated ABP- hu4D5Fabv8 wild type (wt), and ABP-hu4D5Fabv8 cysteine mutants VI IOC and A121C for binding with rabbit albumin, streptavidin (SA), and HER2.
Figure 7 shows ELISA analysis with detection of absorbance at 450nm of biotinylated ABP- hu4D5Fabv8 cysteine mutants (ThioFab variants): (left to right) single Cys variants ABP-V1 IOC, ABP-A121C, and double Cys variants ABP-V110C-A88C and ABP- VI 10C-A121C for binding with rabbit albumin, HER2 and streptavidin (SA), and probing with Fab-HRP or SA-HRP.
Figure 8 shows binding of biotinylated ThioFab phage and an anti-phage HRP antibody to HER2 (top) and Streptavidin (bottom).
Figure 13A shows a cartoon depiction of biotinylated antibody binding to immobilized HER2 with binding of HRP labeled secondary antibody for absorbance detection.
Figure 13B shows binding measurements with detection of absorbance at 450nm of biotin- maleimide conjugated thio-trastuzumab variants and non-biotinylated wild type trastuzumab in binding to immobilized HER2. From left to right: VI IOC (single cys), A121C (single cys), VI 10C/A121C (double cys), and trastuzumab. Each thio IgG variant and trastuzumab was tested at 1, 10, and 100 ng.
Figure 14A shows a cartoon depiction of biotinylated antibody binding to immobilized HER2 with binding of biotin to anti-IgG-HRP for absorbance detection.
Figure 14B shows binding measurements with detection of absorbance at 450nm of biotin- maleimide conjugated-thio trastuzumab variants and non-biotinylated wild type trastuzumab in binding to immobilized streptavidin. From left to right: VI IOC (single cys), A121C (single cys), VI 10C/A121C (double cys), and trastuzumab. Each thio IgG variant and trastuzumab was tested at 1, 10, and 100 ng.
Figure 15 shows the general process to prepare a cysteine engineered antibody (ThioMab) expressed from cell culture for conjugation.
Figure 16 shows non-reducing (top) and reducing (bottom) denaturing polyacrylamide gel electrophoresis analysis of 2H9 ThioMab Fc variants (left to right, lanes 1-9): A339C; S337C; S324C; A287C; V284C; V282C; V279C; V273C, and 2H9 wild type after purification on immobilized Protein A. The lane on the right is a size marker ladder, indicating the intact proteins are about 150 kDa, heavy chain fragments about 50 kDa, and light chain fragments about 25 kDa. Figure 17A shows non-reducing (left) and reducing (+DTT) (right) denaturing polyacrylamide gel electrophoresis analysis of 2H9 ThioMab variants (left to right, lanes 1-4): L-V15C; S179C; S375C; S400C, after purification on immobilized Protein A.
Figure 17B shows non-reducing (left) and reducing (+DTT) (right) denaturing
polyacrylamide gel electrophoresis analysis of 2H9 and 3 A5 ThioMab variants after purification on immobilized Protein A.
Figure 18 shows western blot analysis of biotinylated Thio-IgG variants. 2H9 and 3A5
ThioMab variants were analyzed on reduced denaturing polyacrylamide gel electrophoresis, the proteins were transferred to nitrocellulose membrane. The presence of antibody and conjugated biotin were probed with anti-IgG-HRP (top) and streptavidin-HRP (bottom), respectively. Lane 1 : 3A5 H-A121C. Lane 2: 3A5 L- VI IOC. Lane 3: 2H9 H-A121C. Lane 4: 2H9 L-V110C. Lane 5: 2H9 wild type.
Figure 19 shows ELISA analysis for the binding of biotinylated 2H9 variants to streptavidin by probing with anti-IgG-HRP and measuring the absorbance at 450 nm of (top bar diagram). Bottom schematic diagram depicts the experimental design used in the ELISA analysis.
Figure 20 shows bifunctional reagents for coupling chelator of 89 Zr desferoxamine B (Df, top) with proteins using amino reactive linkers, TFP-N-SucDf and Df-Bz-NCS (center) and thiol reactive linkers, Df-Chx-Mal, Df-Bac, and Df-lac (bottom).
Figure 21 shows the preparation of Df-Chx-Mal, Df-Bac, Df-lac and conjugation to thio- trastuzumab via Cys residues incorporated into the heavy chain of Fab. Reaction conditions: i. DIEA, DMF/H20 (10: 1), RT, 0.5-1 h; ii. DIEA, DMF, 0°C, 4 h; iii. pH 7.5, RT, 1 h; iv. pH 9, RT, 5 h; v. pH 9, RT, 2 h.
Figure 22 shows chelation of zirconium-89 oxalate with a desferrioxamine-labelled, cysteine- engineered antibody, such as variants of Df-linker-trastuzumab containing four linkers: N-Suc, Bz-SCN, Chx-maleimide (CHx-Mal), or acetyl (Ac).
Figure 23 shows mass spectrometry analysis of reduced antibodies showing separate signals from light and heavy chains. A: thio-trasuzumab, B: Df-Ac-thio-trastuzumab (using Df-Bac), and C: Df-Ac-thio-trastuzumab (using Df-lac), and D: Df-Chx-Mal-thio- tratsuzumab.
89 89
Figure 24 shows stability of Zr-Chx-Mal-thio-trastuzumab (open circle) and Zr-Df-Ac- thio-trastuzumab (full circle) in mouse serum at 37 °C (n = 3).
Figure 25 shows representative full-body images (maximum intensity projection) acquired 96 hours after the tail vein bolus injection of 100 μθ of Zr-Trasuzumab prepared using four different linkers (Bz-SCN, N-Suc, Chx-Mal, and Ac).
Figure 26 shows In vivo uptake in selected tissues at 24, 96 and 144 h post injection as
measured by PET. DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulas. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents, which may be included within the scope of the present invention as defined by the claims.
One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described.
Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs, and are consistent with: Singleton et al (1994) Dictionary of Microbiology and Molecular Biology, 2nd Ed., J. Wiley & Sons, New York, NY; and Janeway, C, Travers, P., Walport, M., Shlomchik (2001) Immunobiology, 5th Ed., Garland Publishing, New York. DEFINITIONS
Unless stated otherwise, the following terms and phrases as used herein are intended to have the following meanings:
When trade names are used herein, applicants intend to independently include the trade name product formulation, the generic drug, and the active pharmaceutical ingredient(s) of the trade name product.
The term "antibody" herein is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, dimers, multimers, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments, so long as they exhibit the desired biological activity (Miller et al (2003) Jour, of Immunology 170:4854-4861). Antibodies may be murine, human, humanized, chimeric, or derived from other species. An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen. (Janeway, C, Travers, P., Walport, M., Shlomchik (2001) Immuno Biology, 5th Ed., Garland Publishing, New York). A target antigen generally has numerous binding sites, also called epitopes, recognized by CDRs on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody. An antibody includes a full-length immunoglobulin molecule or an immunologically active portion of a full-length
immunoglobulin molecule, i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease. The immunoglobulin disclosed herein can be of any type (e.g., IgG, IgE, IgM, IgD, and IgA), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and
IgA2) or subclass of immunoglobulin molecule. The immunoglobulins can be derived from any species. In one aspect, however, the immunoglobulin is of human, murine, or rabbit origin.
"Antibody fragments" comprise a portion of a full length antibody, generally the antigen binding or variable region thereof. Examples of antibody fragments include Fab,
Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; minibodies (Olafsen et al (2004) Protein Eng. Design & Sel. 17(4):315-323), fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, CDR (complementary determining region), and epitope- binding fragments of any of the above which immunospecifically bind to cancer cell antigens, viral antigens or microbial antigens, single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies
comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al (1975) Nature 256:495, or may be made by recombinant DNA methods (see for example: US 4816567; US 5807715). The monoclonal antibodies may also be isolated from phage antibody libraries using the techniques described in Clackson et al (1991) Nature, 352:624-628; Marks et al (1991) J. Mol. Biol, 222:581-597; for example.
The monoclonal antibodies herein specifically include "chimeric" antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (US 4816567; and Morrison et al (1984) Proc. Natl. Acad. Sci. USA, 81 :6851-6855). Chimeric antibodies of interest herein include "primatized" antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g., Old World Monkey, Ape etc) and human constant region sequences.
An "intact antibody" herein is one comprising a VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CHI , CH2 and CH3. The constant domains may be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variant thereof. The intact antibody may have one or more "effector functions" which refer to those biological activities attributable to the Fc constant region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include Clq binding;
complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.
Depending on the amino acid sequence of the constant domain of their heavy chains, intact antibodies can be assigned to different "classes." There are five major classes of intact immunoglobulin antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into "subclasses" (isotypes), e.g., IgGl , IgG2, IgG3, IgG4, IgA, and IgA2. The heavy-chain constant domains that correspond to the different classes of antibodies are called α, δ, ε, γ, and μ, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. Ig forms include hinge-modifications or hingeless forms (Roux et al (1998) J. Immunol. 161 :4083-4090; Lund et al (2000) Eur. J. Biochem. 267:7246-7256; US 2005/0048572; US 2004/0229310). An "ErbB receptor" is a receptor protein tyrosine kinase which belongs to the ErbB receptor family whose members are important mediators of cell growth, differentiation and survival. The ErbB receptor family includes four distinct members including epidermal growth factor receptor (EGFR, ErbBl, HER1), HER2 (ErbB2 or pl85neu), HER3 (ErbB3) and HER4 (ErbB4 or tyro2). A panel of anti-ErbB2 antibodies has been characterized using the human breast tumor cell line SKBR3 (Hudziak et al (1989) Mol. Cell. Biol. 9(3): 1165- 1172. Maximum inhibition was obtained with the antibody called 4D5 which inhibited cellular proliferation by 56%. Other antibodies in the panel reduced cellular proliferation to a lesser extent in this assay. The antibody 4D5 was further found to sensitize ErbB2- overexpressing breast tumor cell lines to the cytotoxic effects of TNF-a (US 5677171). The anti-ErbB2 antibodies discussed in Hudziak et al. are further characterized in Fendly et al
(1990) Cancer Research 50: 1550-1558; Kotts et al. (1990) In Vitro 26(3):59A; Sarup et al.
(1991) Growth Regulation 1 :72-82; Shepard et al. J. (1991) Clin. Immunol. 11(3): 117-127; Kumar et al. (1991) Mol. Cell. Biol. 11(2):979-986; Lewis et al. (1993) Cancer Immunol. Immunother. 37:255-263; Pietras et al. (1994) Oncogene 9: 1829-1838; Vitetta et al. (1994) Cancer Research 54:5301-5309; Sliwkowski et al. (1994) J. Biol. Chem. 269(20): 14661 - 14665; Scott et al. (1991) J. Biol. Chem. 266: 14300-5; D'souza et al. Proc. Natl. Acad. Sci. (1994) 91 :7202-7206; Lewis et al. (1996) Cancer Research 56: 1457-1465; and Schaefer et al. (1997) Oncogene 15: 1385-1394.
The ErbB receptor will generally comprise an extracellular domain, which may bind an ErbB ligand; a lipophilic transmembrane domain; a conserved intracellular tyrosine kinase domain; and a carboxyl-terminal signaling domain harboring several tyrosine residues which can be phosphorylated. The ErbB receptor may be a "native sequence" ErbB receptor or an "amino acid sequence variant" thereof. Preferably, the ErbB receptor is native sequence human ErbB receptor. Accordingly, a "member of the ErbB receptor family" is EGFR (ErbBl), ErbB2, ErbB3, ErbB4 or any other ErbB receptor currently known or to be identified in the future.
The terms "ErbBl", "epidermal growth factor receptor", "EGFR" and "HER1" are used interchangeably herein and refer to EGFR as disclosed, for example, in Carpenter et al (1987) Ann. Rev. Biochem., 56:881-914, including naturally occurring mutant forms thereof (e.g., a deletion mutant EGFR as in Humphrey et al (1990) Proc. Nat. Acad. Sci. (USA) 87:4207-4211). The term erbBl refers to the gene encoding the EGFR protein product. Antibodies against HER1 are described, for example, in Murthy et al (1987) Arch. Biochem. Biophys., 252:549-560 and in WO 95/25167. The term "ERRP", "EGF-Receptor Related Protein", "EGFR Related Protein" and "epidermal growth factor receptor related protein" are used interchangeably herein and refer to ERRP as disclosed, for example in US 6399743 and US Publication No. 2003/0096373.
The expressions "ErbB2" and "HER2" are used interchangeably herein and refer to human HER2 protein described, for example, in Semba et al (1985) Proc. Nat. Acad. Sci. (USA) 82:6497-6501 and Yamamoto et al (1986) Nature, 319:230-234 (Genebank accession number X03363). The term "erbB2" refers to the gene encoding human ErbB2 and "neu" refers to the gene encoding rat pl85neu. Preferred ErbB2 is native sequence human ErbB2.
"ErbB3" and "HER3" refer to the receptor polypeptide as disclosed, for example, in U.S. Patent Nos. 5183884 and 5480968 as well as Kraus et al (1989) Proc. Nat. Acad. Sci. (USA) 86:9193-9197. Antibodies against ErbB3 are known in the art and are described, for example, in U.S. Patent Nos. 5183884, 5480968 and in WO 97/35885.
The terms "ErbB4" and "HER4" herein refer to the receptor polypeptide as disclosed, for example, in EP Pat Application No 599,274; Plowman et al (1993) Proc. Natl. Acad. Sci. USA 90: 1746-1750; and Plowman et al (1993) Nature 366:473-475, including isoforms thereof, e.g., as disclosed in WO 99/19488. Antibodies against HER4 are described, for example, in WO 02/18444.
Antibodies to ErbB receptors are available commercially from a number of sources, including, for example, Santa Cruz Biotechnology, Inc., California, USA.
The term "amino acid sequence variant" refers to polypeptides having amino acid sequences that differ to some extent from a native sequence polypeptide. Ordinarily, amino acid sequence variants will possess at least about 70% sequence identity with at least one receptor binding domain of a native ErbB ligand or with at least one ligand binding domain of a native ErbB receptor, and preferably, they will be at least about 80%, more preferably, at least about 90%> homologous by sequence with such receptor or ligand binding domains. The amino acid sequence variants possess substitutions, deletions, and/or insertions at certain positions within the amino acid sequence of the native amino acid sequence. Amino acids are designated by the conventional names, one-letter and three-letter codes.
"Sequence identity" is defined as the percentage of residues in the amino acid sequence variant that are identical after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Methods and computer programs for the alignment are well known in the art. One such computer program is "Align 2," authored by Genentech, Inc., which was filed with user documentation in the United States Copyright Office, Washington, DC 20559, on December 10, 1991. "Antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to a cell- mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g., Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. The primary cells for mediating ADCC, NK cells, express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII. FcR expression on hematopoietic cells in summarized is Table 3 on page 464 of Ravetch and Kinet, (1991) "Annu. Rev. Immunol." 9:457-92. To assess ADCC activity of a molecule of interest, an in vitro ADCC assay, such as that described in US 5500362 and US 5821337 may be performed. Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al (1998) PROC. NAT. ACAD. SCI. (USA) (USA) 95:652-656.
"Human effector cells" are leukocytes which express one or more constant region receptors (FcRs) and perform effector functions. Preferably, the cells express at least FcyRIII and perform ADCC effector function. Examples of human leukocytes which mediate ADCC include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being preferred. The effector cells may be isolated from a native source thereof, e.g., from blood or PBMCs as described herein.
The terms "Fc receptor" or "FcR" are used to describe a receptor that binds to the Fc constant region of an antibody. The preferred FcR is a native sequence human FcR.
Moreover, a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII, and Fey RIII subclasses, including allelic variants and alternatively spliced forms of these receptors. FcyRII receptors include FcyRIIA (an
"activating receptor") and FcyRIIB (an "inhibiting receptor"), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof. Activating receptor FcyRIIA contains an immunoreceptor tyrosine-based activation motif (IT AM) in its cytoplasmic domain. Inhibiting receptor FcyRIIB contains an immunoreceptor tyrosine - based inhibition motif (ITIM) in its cytoplasmic domain. (See review M. in Daeron, "Annu. Rev. Immunol." 15:203-234 (1997)). FcRs are reviewed in Ravetch and Kinet, "Annu. Rev. Immunol"., 9:457-92 (1991); Capel et al (1994) Immunomethods 4:25-34; and de Haas et al (1995) J. Lab. Clin. Med. 126:330-41. Other FcRs, including those to be identified in the future, are encompassed by the term "FcR" herein. The term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al (1976) J. Immunol, 117:587 and Kim et al (1994) J. Immunol. 24:249).
"Complement dependent cytotoxicity" or "CDC" refers to the ability of a molecule to lyse a target in the presence of complement. The complement activation pathway is initiated by the binding of the first component of the complement system (Clq) to a molecule (e.g., an antibody) complexed with a cognate antigen. To assess complement activation, a CDC assay, e.g., as described in Gazzano-Santoro et al J. Immunol. Methods, 202: 163 (1996), may be performed.
"Native antibodies" are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end. The constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light-chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable domains.
The term "variable" refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FRs). The variable domains of native heavy and light chains each comprise four FRs, largely adopting a β-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the β-sheet structure. The hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen- binding site of antibodies (see Kabat et al (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody dependent cellular cytotoxicity (ADCC).
The term "hypervariable region" when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding. The hypervariable region generally comprises amino acid residues from a "complementarity determining region" or "CDR" (e.g., residues 24-34 (LI), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (HI), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al supra) and/or those residues from a "hypervariable loop" (e.g., residues 26-32 (LI), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (HI), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk (1987) J. Mol. Biol, 196:901-917).
"Framework Region" or "FR" residues are those variable domain residues other than the hypervariable region residues as herein defined.
Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments, each with a single antigen-binding site, and a residual "Fc" fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen.
"Fv" is the minimum antibody fragment which contains a complete antigen- recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to define an antigen- binding site on the surface of the VH-VL dimer. Collectively, the six hypervariable regions confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
The Fab fragment also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CHI domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear at least one free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known. The "light chains" of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (κ) and lambda (λ), based on the amino acid sequences of their constant domains.
"Single-chain Fv" or "scFv" antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain. Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding. For a review of scFv, see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 1 13, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994). Anti-ErbB2 antibody scFv fragments are described in WO 93/16185; US Patent Nos. 5571894; and 5587458.
The term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a variable heavy domain (VH) connected to a variable light domain (VL) in the same polypeptide chain (VH - VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
Diabodies are described more fully in, for example, EP 404,097; WO 93/11 161 ; and
Hollinger et al (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448.
"Humanized" forms of non-human (e.g., rodent) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. Humanization is a method to transfer the murine antigen binding information to a non-immunogenic human antibody acceptor, and has resulted in many therapeutically useful drugs. The method of humanization generally begins by transferring all six murine complementarity determining regions (CDRs) onto a human antibody framework (Jones et al, (1986) Nature 321 :522-525). These CDR-grafted antibodies generally do not retain their original affinity for antigen binding, and in fact, affinity is often severely impaired. Besides the CDRs, select non-human antibody framework residues must also be incorporated to maintain proper CDR
conformation (Chothia et al (1989) Nature 342:877). The transfer of key mouse framework residues to the human acceptor in order to support the structural conformation of the grafted CDRs has been shown to restore antigen binding and affinity (Riechmann et al (1992) J. Mol. Biol. 224, 487-499; Foote and Winter, (1992) J. Mol. Biol. 224:487-499; Presta et al (1993) J. Immunol. 151 , 2623-2632; Werther et al (1996) J. Immunol. Methods 157:4986-4995; and Presta et al (2001) Thromb. Haemost. 85 :379-389). For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see US 6407213; Jones et al (1986) Nature, 321 :522- 525; Riechmann et al (1988) Nature 332:323-329; and Presta, (1992) Curr. Op. Struct. Biol, 2:593-596.
A "free cysteine amino acid" refers to a cysteine amino acid residue which has been engineered into a parent antibody, has a thiol functional group (-SH), and is not paired as an intramolecular or intermolecular disulfide bridge.
The term "thiol reactivity value" is a quantitative characterization of the reactivity of free cysteine amino acids. The thiol reactivity value is the percentage of a free cysteine amino acid in a cysteine engineered antibody which reacts with a thiol-reactive reagent, and converted to a maximum value of 1. For example, a free cysteine amino acid on a cysteine engineered antibody which reacts in 100% yield with a thiol-reactive reagent, such as a biotin-maleimide reagent, to form a biotin-labelled antibody has a thiol reactivity value of 1.0. Another cysteine amino acid engineered into the same or different parent antibody which reacts in 80% yield with a thiol-reactive reagent has a thiol reactivity value of 0.8. Another cysteine amino acid engineered into the same or different parent antibody which fails totally to react with a thiol-reactive reagent has a thiol reactivity value of 0.
Determination of the thiol reactivity value of a particular cysteine may be conducted by ELISA assay, mass spectroscopy, liquid chromatography, autoradiography, or other quantitative analytical tests.
A "parent antibody" is an antibody comprising an amino acid sequence from which one or more amino acid residues are replaced by one or more cysteine residues. The parent antibody may comprise a native or wild type sequence. The parent antibody may have preexisting amino acid sequence modifications (such as additions, deletions and/or substitutions) relative to other native, wild type, or modified forms of an antibody. A parent antibody may be directed against a target antigen of interest, e.g. a biologically important polypeptide. Antibodies directed against nonpolypeptide antigens (such as tumor-associated glycolipid antigens; see US 5091178) are also contemplated.
Exemplary parent antibodies include antibodies having affinity and selectivity for cell surface and transmembrane receptors and tumor-associated antigens (TAA).
Other exemplary parent antibodies include those selected from, and without limitation, anti-estrogen receptor antibody, anti-progesterone receptor antibody, anti-p53 antibody, anti-HER-2/neu antibody, anti-EGFR antibody, anti-cathepsin D antibody, anti- Bcl-2 antibody, anti-E-cadherin antibody, anti-CA125 antibody, anti-CA15-3 antibody, anti- CA19-9 antibody, anti-c-erbB-2 antibody, anti-P-glycoprotein antibody, anti-CEA antibody, anti-retinoblastoma protein antibody, anti-ras oncoprotein antibody, anti-Lewis X antibody, anti-Ki-67 antibody, anti-PCNA antibody, anti-CD3 antibody, anti-CD4 antibody, anti-CD5 antibody, anti-CD7 antibody, anti-CD8 antibody, anti-CD9/p24 antibody, anti-CD 10 antibody, anti-CDl lc antibody, anti-CD 13 antibody, anti-CD 14 antibody, anti-CD 15 antibody, anti-CD 19 antibody, anti-CD20 antibody, anti-CD22 antibody, anti-CD23 antibody, anti-CD30 antibody, anti-CD31 antibody, anti-CD33 antibody, anti-CD34 antibody, anti-CD35 antibody, anti-CD38 antibody, anti-CD41 antibody, anti-LCA/CD45 antibody, anti-CD45RO antibody, anti-CD45RA antibody, anti-CD39 antibody, anti-CD 100 antibody, anti-CD95/Fas antibody, anti-CD99 antibody, anti-CD 106 antibody, anti-ubiquitin antibody, anti-CD71 antibody, anti-c-myc antibody, anti-cytokeratins antibody, anti- vimentins antibody, anti-HPV proteins antibody, anti-kappa light chains antibody, anti- lambda light chains antibody, anti-melanosomes antibody, anti-prostate specific antigen antibody, anti-S-100 antibody, anti-tau antigen antibody, anti-fibrin antibody, anti-keratins antibody and anti-Tn-antigen antibody.
An "isolated" antibody is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or
nonproteinaceous solutes. In preferred embodiments, the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain. Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
An antibody "which binds" a molecular target or an antigen of interest, e.g., ErbB2 antigen, is one capable of binding that antigen with sufficient affinity such that the antibody is useful in targeting a cell expressing the antigen. Where the antibody is one which binds ErbB2, it will usually preferentially bind ErbB2 as opposed to other ErbB receptors, and may be one which does not significantly cross-react with other proteins such as EGFR, ErbB3 or ErbB4. In such embodiments, the extent of binding of the antibody to these non-ErbB2 proteins (e.g., cell surface binding to endogenous receptor) will be less than 10% as determined by fluorescence activated cell sorting (FACS) analysis or
radioimmunoprecipitation (RIA). Sometimes, the anti-ErbB2 antibody will not significantly cross-react with the rat neu protein, e.g., as described in Schecter et al. (1984) Nature 312:513 and Drebin et al (1984) Nature 312:545-548.
Molecular targets for antibodies encompassed by the present invention include CD proteins and their ligands, such as, but not limited to: (i) CD3, CD4, CD8, CD19, CD20, CD22, CD34, CD40, CD79a (CD79a), and CD79p (CD79b); (ii) members of the ErbB receptor family such as the EGF receptor, HER2, HER3 or HER4 receptor; (iii) cell adhesion molecules such as LFA-1, Macl, pl50,95, VLA-4, ICAM-1, VCAM and ν/β3 integrin, including either alpha or beta subunits thereof (e.g. anti-CD 1 la, anti-CD 18 or anti-CD 1 lb antibodies); (iv) growth factors such as VEGF; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C, BR3, c-met, tissue factor, β7 etc; and (v) cell surface and transmembrane tumor-associated antigens (TAA).
Unless indicated otherwise, the term "monoclonal antibody 4D5" refers to an antibody that has antigen binding residues of, or derived from, the murine 4D5 antibody (ATCC CRL 10463). For example, the monoclonal antibody 4D5 may be murine
monoclonal antibody 4D5 or a variant thereof, such as a humanized 4D5. Exemplary humanized 4D5 antibodies include huMAb4D5-l, huMAb4D5-2, huMAb4D5-3,
huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (trastuzumab, HERCEPTIN®) as in US Patent No. 5821337.
"Phage display" is a technique by which variant polypeptides are displayed as fusion proteins to a coat protein on the surface of phage, e.g., filamentous phage, particles. One utility of phage display lies in the fact that large libraries of randomized protein variants can be rapidly and efficiently sorted for those sequences that bind to a target molecule with high affinity. Display of peptide and protein libraries on phage has been used for screening millions of polypeptides for ones with specific binding properties. Polyvalent phage display methods have been used for displaying small random peptides and small proteins, typically through fusions to either pill or pVIII of filamentous phage (Wells and Lowman, (1992) Curr. Opin. Struct. Biol, 3:355-362, and references cited therein). In monovalent phage display, a protein or peptide library is fused to a phage coat protein or a portion thereof, and expressed at low levels in the presence of wild type protein. Avidity effects are reduced relative to polyvalent phage so that sorting is on the basis of intrinsic ligand affinity, and phagemid vectors are used, which simplify DNA manipulations. Lowman and Wells, Methods: A companion to Methods in Enzymology, 3:205-0216 (1991). Phage display includes techniques for producing antibody- like molecules (Janeway, C, Travers, P., Walport, M., Shlomchik (2001) Immunobiology, 5th Ed., Garland Publishing, New York, p627-628; Lee et al ).
A "phagemid" is a plasmid vector having a bacterial origin of replication, e.g., ColEl, and a copy of an intergenic region of a bacteriophage. The phagemid may be used on any known bacteriophage, including filamentous bacteriophage and lambdoid bacteriophage. The plasmid will also generally contain a selectable marker for antibiotic resistance. Segments of DNA cloned into these vectors can be propagated as plasmids. When cells harboring these vectors are provided with all genes necessary for the production of phage particles, the mode of replication of the plasmid changes to rolling circle replication to generate copies of one strand of the plasmid DNA and package phage particles. The phagemid may form infectious or non-infectious phage particles. This term includes phagemids which contain a phage coat protein gene or fragment thereof linked to a heterologous polypeptide gene as a gene fusion such that the heterologous polypeptide is displayed on the surface of the phage particle.
"Linker", "Linker Unit", or "link" means a chemical moiety comprising a covalent bond or a chain of atoms that covalently attaches an antibody to a drug moiety. In various embodiments, a linker is specified as L. Linkers include a divalent radical such as an alkyldiyl, an arylene, a heteroarylene, moieties such as: -(CR2)nO(CR2)n-, repeating units of alkyloxy (e.g. polyethylenoxy, PEG, polymethyleneoxy) and alkylamino (e.g.
polyethyleneamino, Jeffamine™); and diacid ester and amides including succinate, succinamide, diglycolate, malonate, and caproamide.
The term "label" means any moiety which can be covalently attached to an antibody and that functions to: (i) provide a detectable signal; (ii) interact with a second label to modify the detectable signal provided by the first or second label, e.g. FRET (fluorescence resonance energy transfer); (iii) stabilize interactions or increase affinity of binding, with antigen or ligand; (iv) affect mobility, e.g. electrophoretic mobility, or cell-permeability, by charge, hydrophobicity, shape, or other physical parameters, or (v) provide a capture moiety, to modulate ligand affinity, antibody/antigen binding, or ionic complexation.
Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
The phrase "pharmaceutically acceptable salt," as used herein, refers to
pharmaceutically acceptable organic or inorganic salts of an AZC. Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, /?-toluenesulfonate, and pamoate (i.e., 1,1 ' -methyl ene-bis -(2-hydroxy-3- naphthoate)) salts. A pharmaceutically acceptable salt may involve the inclusion of another molecule such as an acetate ion, a succinate ion or other counterion. The counterion may be any organic or inorganic moiety that stabilizes the charge on the parent compound. Furthermore, a pharmaceutically acceptable salt may have more than one charged atom in its structure. Instances where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counter ions. Hence, a pharmaceutically acceptable salt can have one or more charged atoms and/or one or more counterion.
"Pharmaceutically acceptable solvate" refers to an association of one or more solvent molecules and an AZC. Examples of solvents that form pharmaceutically acceptable solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
The following abbreviations are used herein and have the indicated definitions: BME is beta-mercaptoethanol, Boc is N-(t-butoxycarbonyl), cit is citrulline (2-amino-5-ureido pentanoic acid), dap is dolaproine, DCC is 1,3-dicyclohexylcarbodiimide, DCM is dichloromethane, DEA is diethylamine, DEAD is diethylazodicarboxylate, DEPC is diethylphosphorylcyanidate, DIAD is diisopropylazodicarboxylate, DIEA is N,N- diisopropylethylamine, dil is dolaisoleucine, DMA is dimethylacetamide, DMAP is 4- dimethylaminopyridine, DME is ethyleneglycol dimethyl ether (or 1 ,2-dimethoxyethane), DMF is N,N-dimethylformamide, DMSO is dimethylsulfoxide, doe is dolaphenine, dov is N,N-dimethylvaline, DTNB is 5,5'-dithiobis(2-nitrobenzoic acid), DTP A is
diethylenetriaminepentaacetic acid, DTT is dithiothreitol, EDCI is l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, EEDQ is 2-ethoxy-l- ethoxycarbonyl-l,2-dihydroquinoline, ES-MS is electrospray mass spectrometry, EtOAc is ethyl acetate, Fmoc is N-(9-fluorenylmethoxycarbonyl), gly is glycine, HATU is 0-(7- azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate, HOBt is 1- hydroxybenzotriazole, HPLC is high pressure liquid chromatography, ile is isoleucine, lys is lysine, MeCN (CH3CN) is acetonitrile, MeOH is methanol, Mtr is 4-anisyldiphenylmethyl (or 4-methoxytrityl),nor is (IS, 2i?)-(+)-norephedrine, PAB is p-aminobenzylcarbamoyl, PBS is phosphate-buffered saline (pH 7), PEG is polyethylene glycol, Ph is phenyl, Pnp is p- nitrophenyl, MC is 6-maleimidocaproyl, phe is L-phenylalanine, PyBrop is bromo tris- pyrrolidino phosphonium hexafluorophosphate, SEC is size-exclusion chromatography, Su is succinimide, TFA is trifluoroacetic acid, TLC is thin layer chromatography, UV is ultraviolet, and val is valine.
CYSTEINE ENGINEERED ANTIBODIES
The compounds of the invention include cysteine engineered antibodies where one or more amino acids of a wild-type or parent antibody are replaced with a cysteine amino acid. Any form of antibody may be so engineered, i.e. mutated. For example, a parent Fab antibody fragment may be engineered to form a cysteine engineered Fab, referred to herein as "ThioFab." Similarly, a parent monoclonal antibody may be engineered to form a
"ThioMab." It should be noted that a single site mutation yields a single engineered cysteine residue in a ThioFab, while a single site mutation yields two engineered cysteine residues in a ThioMab, due to the dimeric nature of the IgG antibody. Mutants with replaced
("engineered") cysteine (Cys) residues are evaluated for the reactivity of the newly introduced, engineered cysteine thiol groups. The thiol reactivity value is a relative, numerical term in the range of 0 to 1.0 and can be measured for any cysteine engineered antibody. Thiol reactivity values of cysteine engineered antibodies of the invention are in the ranges of 0.6 to 1.0; 0.7 to 1.0; or 0.8 to 1.0.
The design, selection, and preparation methods of the invention enable cysteine engineered antibodies which are reactive with electrophilic functionality. These methods further enable antibody conjugate compounds such as antibody-zirconium conjugate (AZC) compounds with zirconium atoms at designated, designed, selective sites. Reactive cysteine residues on an antibody surface allow specifically conjugating a zirconium moiety through a thiol reactive group such as maleimide or haloacetyl. The nucleophilic reactivity of the thiol functionality of a Cys residue to a maleimide group is about 1000 times higher compared to any other amino acid functionality in a protein, such as amino group of lysine residues or the N-terminal amino group. Thiol specific functionality in iodoacetyl and maleimide reagents may react with amine groups, but higher pH (>9.0) and longer reaction times are required (Garman, 1997, Non-Radioactive Labelling: A Practical Approach, Academic Press, London).
Cysteine engineered antibodies of the invention preferably retain the antigen binding capability of their wild type, parent antibody counterparts. Thus, cysteine engineered antibodies are capable of binding, preferably specifically, to antigens. Such antigens include, for example, tumor-associated antigens (TAA), cell surface receptor proteins and other cell surface molecules, transmembrane proteins, signalling proteins, cell survival regulatory factors, cell proliferation regulatory factors, molecules associated with (for e.g., known or suspected to contribute functionally to) tissue development or differentiation, lymphokines, cytokines, molecules involved in cell cycle regulation, molecules involved in vasculogenesis and molecules associated with (for e.g., known or suspected to contribute functionally to) angiogenesis. The tumor-associated antigen may be a cluster differentiation factor (i.e., a CD protein). An antigen to which a cysteine engineered antibody is capable of binding may be a member of a subset of one of the above-mentioned categories, wherein the other subset(s) of said category comprise other molecules/antigens that have a distinct characteristic (with respect to the antigen of interest).
The parent antibody may also be a humanized antibody selected from huMAb4D5-l, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8 (Trastuzumab, HERCEPTIN®) as described in Table 3 of US 5821337, expressly incorporated herein by reference; humanized 520C9 (WO 93/21319) and humanized 2C4 antibodies as described herein.
Cysteine engineered antibodies of the invention may be site-specifically and efficiently coupled with a thiol-reactive reagent. The thiol-reactive reagent may be a multifunctional linker reagent, a capture, i.e. affinity, label reagent (e.g. a biotin-linker reagent), a detection label (e.g. a fluorophore reagent), a solid phase immobilization reagent (e.g. SEPHAROSE™, polystyrene, or glass), or a zirconium-linker intermediate. One example of a thiol-reactive reagent is N-ethyl maleimide (NEM). In an exemplary embodiment, reaction of a ThioFab with a biotin-linker reagent provides a biotinylated ThioFab by which the presence and reactivity of the engineered cysteine residue may be detected and measured. Reaction of a ThioFab with a multifunctional linker reagent provides a ThioFab with a functionalized linker which may be further reacted with a zirconium moiety reagent or other label. Reaction of a ThioFab with a zirconium-linker intermediate provides a ThioFab zirconium conjugate.
The exemplary methods described here may be applied generally to the identification and production of antibodies, and more generally, to other proteins through application of the design and screening steps described herein.
Such an approach may be applied to the conjugation of other thiol-reactive agents in which the reactive group is, for example, a maleimide, an iodoacetamide, a pyridyl disulfide, or other thiol-reactive conjugation partner (Haugland, 2003, Molecular Probes Handbook of Fluorescent Probes and Research Chemicals, Molecular Probes, Inc.; Brinkley, 1992, Bioconjugate Chem. 3:2; Garman, 1997, Non-Radioactive Labelling: A Practical Approach, Academic Press, London; Means (1990) Bioconjugate Chem. 1 :2; Hermanson, G. in
Bioconjugate Techniques (1996) Academic Press, San Diego, pp. 40-55, 643-671). The partner may be a cytotoxic agent (e.g. a toxin such as doxorubicin or pertussis toxin), a fluorophore such as a fluorescent dye like fluorescein or rhodamine, a chelating agent for an imaging or radiotherapeutic metal, a peptidyl or non-peptidyl label or detection tag, or a clearance-modifying agent such as various isomers of polyethylene glycol, a peptide that binds to a third component, or another carbohydrate or lipophilic agent. The sites identified on the exemplary antibody fragment, hu4D5Fabv8, herein are primarily in the constant domain of an antibody which is well conserved across all species of antibodies. These sites should be broadly applicable to other antibodies, without further need of structural design or knowledge of specific antibody structures, and without interference in the antigen binding properties inherent to the variable domains of the antibody.
Cysteine engineered antibodies which may be useful in the treatment of cancer include, but are not limited to, antibodies against cell surface receptors and tumor-associated antigens (TAA). Such antibodies may be used as naked antibodies (unconjugated to a label moiety) or as Formula I antibody-zirconium conjugates (AZC). Tumor-associated antigens are known in the art, and can prepared for use in generating antibodies using methods and information which are well known in the art. In attempts to discover effective cellular targets for cancer diagnosis and therapy, researchers have sought to identify transmembrane or otherwise tumor-associated polypeptides that are specifically expressed on the surface of one or more particular type(s) of cancer cell as compared to on one or more normal non- cancerous cell(s). Often, such tumor-associated polypeptides are more abundantly expressed on the surface of the cancer cells as compared to on the surface of the non-cancerous cells. The identification of such tumor-associated cell surface antigen polypeptides has given rise to the ability to specifically target cancer cells for destruction via antibody-based therapies.
Examples of TAA include, but are not limited to, TAA (l)-(36) listed below. For convenience, information relating to these antigens, all of which are known in the art, is listed below and includes names, alternative names, Genbank accession numbers and primary reference(s), following nucleic acid and protein sequence identification conventions of the National Center for Biotechnology Information (NCBI). Nucleic acid and protein sequences corresponding to TAA (l)-(36) are available in public databases such as GenBank. Tumor- associated antigens targeted by antibodies include all amino acid sequence variants and isoforms possessing at least about 70%, 80%>, 85%, 90%>, or 95% sequence identity relative to the sequences identified in the cited references, or which exhibit substantially the same biological properties or characteristics as a TAA having a sequence found in the cited references. For example, a TAA having a variant sequence generally is able to bind specifically to an antibody that binds specifically to the TAA with the corresponding sequence listed. The sequences and disclosure in the reference specifically recited herein are expressly incorporated by reference. TUMOR- ASSOCIATED ANTIGENS (l)-(36):
(1) BMPR1B (bone morphogenetic protein receptor-type IB, Genbank accession
no. NM_001203)
ten Dijke,P., et al Science 264 (5155): 101-104 (1994), Oncogene 14 (11): 1377- 1382 (1997)); WO2004063362 (Claim 2); WO2003042661 (Claim 12);
US2003134790-A1 (Page 38-39); WO2002102235 (Claim 13; Page 296);
WO2003055443 (Page 91-92); WO200299122 (Example 2; Page 528-530);
WO2003029421 (Claim 6); WO2003024392 (Claim 2; Fig 112); WO200298358
(Claim 1; Page 183); WO200254940 (Page 100-101); WO200259377(Page 349- 350); WO200230268 (Claim 27; Page 376); WO200148204 (Example; Fig 4)
NP 001194 bone morphogenetic protein receptor, type IB /pid=NP_001194.1 - Cross-references: MIM:603248; NP 001194.1; AY065994
(2) E16 (LAT1, SLC7A5, Genbank accession no. NM 003486)
Biochem. Biophys. Res. Commun. 255 (2), 283-288 (1999), Nature 395 (6699):288-291 (1998), Gaugitsch, H.W., et al (1992) J. Biol. Chem. 267 (16): 11267-11273);
WO2004048938 (Example 2); WO2004032842 (Example IV); WO2003042661 (Claim 12); WO2003016475 (Claim 1); WO200278524 (Example 2); WO200299074 (Claim 19; Page 127-129); WO200286443 (Claim 27; Pages 222, 393); WO2003003906 (Claim 10; Page 293); WO200264798 (Claim 33; Page 93-95); WO200014228 (Claim 5; Page 133-136); US2003224454 (Fig 3); WO2003025138 (Claim 12; Page 150);
NP 003477 solute carrier family 7 (cationic amino acid transporter, y+
system), member 5 /pid=NP_003477.3 - Homo sapiens
Cross-references: MIM:600182; NP_003477.3; NM_015923; NM_003486_1 (3) STEAP1 (six transmembrane epithelial antigen of prostate, Genbank accession no. NM_012449)
Cancer Res. 61 (15), 5857-5860 (2001), Hubert, R.S., et al (1999) Proc. Natl. Acad. Sci. U.S.A. 96 (25): 14523-14528); WO2004065577 (Claim 6); WO2004027049 (Fig 1L);
EP1394274 (Example 11); WO2004016225 (Claim 2); WO2003042661 (Claim 12);
US2003157089 (Example 5); US2003185830 (Example 5); US2003064397 (Fig 2);
WO200289747 (Example 5; Page 618-619); WO2003022995 (Example 9; Fig 13A, Example 53; Page 173, Example 2; Fig 2A); NP 036581 six transmembrane epithelial antigen of the prostate
Cross-references: MIM:604415; NP_036581.1; NM_012449_1
(4) 0772P (CA125, MUC16, Genbank accession no. AF361486)
J. Biol. Chem. 276 (29):27371-27375 (2001)); WO2004045553 (Claim 14);
WO200292836 (Claim 6; Fig 12); WO200283866 (Claim 15; Page 116-121);
US2003124140 (Example 16); Cross-references: GL34501467; AAK74120.3;
AF361486 1 (5) MPF (MPF, MSLN, SMR, megakaryocyte potentiating factor, mesothelin,
Genbank accession no. NM_005823) Yamaguchi, N., et al Biol. Chem. 269 (2),
805-808 (1994), Proc. Natl. Acad. Sci. U.S.A. 96 (20): 11531-11536 (1999), Proc.
Natl. Acad. Sci. U.S.A. 93 (1): 136-140 (1996), J. Biol. Chem. 270 (37):21984- 21990 (1995)); WO2003101283 (Claim 14); (WO2002102235 (Claim 13; Page
287-288); WO2002101075 (Claim 4; Page 308-309); WO200271928 (Page 320- 321); WO9410312 (Page 52-57); Cross-references: MIM:601051; NP_005814.2;
NM_005823_1
(6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, solute carrier family 34 (sodium
phosphate), member 2, type II sodium-dependent phosphate transporter
3b,Genbank accession no. NM 006424)
J. Biol. Chem. 277 (22): 19665-19672 (2002), Genomics 62 (2):281-284 (1999), Feild, J.A., et al (1999) Biochem. Biophys. Res. Commun. 258 (3):578-582); WO2004022778 (Claim 2); EP1394274 (Example 11); WO2002102235 (Claim 13; Page 326); EP875569 (Claim 1; Page 17-19); WO200157188 (Claim 20; Page 329); WO2004032842 (Example IV);
WO200175177 (Claim 24; Page 139-140);
Cross-references: MIM:604217; NP_006415.1; NM_006424_1
(7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thrombospondin repeats (type 1 and type 1 -like), transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 5B, Genbank accession no. AB040878) Nagase T., et al (2000) DNA Res. 7 (2): 143-150); WO2004000997 (Claim 1);
WO2003003984 (Claim 1); WO200206339 (Claim 1; Page 50); WO200188133 (Claim 1; Page 41-43, 48-58); WO2003054152 (Claim 20); WO2003101400 (Claim 11);
Accession: Q9P283; EMBL; AB040878; BAA95969.1. Genew; HGNC: 10737;
(8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genbank accession no. AY358628); Ross et al (2002) Cancer Res. 62:2546-2553; US2003129192 (Claim 2); US2004044180 (Claim 12); US2004044179 (Claim 11); US2003096961 (Claim 11); US2003232056 (Example 5); WO2003105758 (Claim 12); US2003206918 (Example 5); EP1347046 (Claim 1); WO2003025148 (Claim 20);
Cross-references: GL37182378; AAQ88991.1; AY358628J
(9) ETBR (Endothelin type B receptor, Genbank accession no. AY275463);
Nakamuta M., et al Biochem. Biophys. Res. Commun. 177, 34-39, 1991; Ogawa Y., et al Biochem. Biophys. Res. Commun. 178, 248-255, 1991; Arai H., et al Jpn. Circ. J. 56, 1303- 1307, 1992; Arai H., et al J. Biol. Chem. 268, 3463-3470, 1993; Sakamoto A., Yanagisawa M., et al Biochem. Biophys. Res. Commun. 178, 656-663, 1991; Elshourbagy N.A., et al J. Biol. Chem. 268, 3873-3879, 1993; Haendler B., et al J. Cardiovasc. Pharmacol. 20, sl-S4, 1992; Tsutsumi M., et al Gene 228, 43-49, 1999; Strausberg R.L., et al Proc. Natl. Acad. Sci. U.S.A. 99, 16899-16903, 2002; Bourgeois C, et al J. Clin. Endocrinol. Metab. 82, 3116- 3123, 1997; Okamoto Y., et al Biol. Chem. 272, 21589-21596, 1997; Verheij J.B., et al Am. J. Med. Genet. 108, 223-225, 2002; Hofstra R.M.W., et al Eur. J. Hum. Genet. 5, 180-185, 1997; Puffenberger E.G., et al Cell 79, 1257-1266, 1994; Attie T., et al, Hum. Mol. Genet. 4, 2407-2409, 1995; Auricchio A., et al Hum. Mol. Genet. 5:351-354, 1996; Amiel J., et al Hum. Mol. Genet. 5, 355-357, 1996; Hofstra R.M.W., et al Nat. Genet. 12, 445-447, 1996; Svensson P.J., et al Hum. Genet. 103, 145-148, 1998; Fuchs S., et al Mol. Med. 7, 115-124, 2001; Pingault V., et al (2002) Hum. Genet. I l l, 198-206; WO2004045516 (Claim 1);
WO2004048938 (Example 2); WO2004040000 (Claim 151); WO2003087768 (Claim 1); WO2003016475 (Claim 1); WO2003016475 (Claim 1); WO200261087 (Fig 1);
WO2003016494 (Fig 6); WO2003025138 (Claim 12; Page 144); WO200198351 (Claim 1; Page 124-125); EP522868 (Claim 8; Fig 2); WO200177172 (Claim 1; Page 297-299);
US2003109676; US6518404 (Fig 3); US5773223 (Claim la; Col 31-34); WO2004001004; (10) MSG783 (RNF124, hypothetical protein FLJ20315, Genbank accession no.
NM_017763);
WO2003104275 (Claim 1); WO2004046342 (Example 2); WO2003042661 (Claim 12); WO2003083074 (Claim 14; Page 61); WO2003018621 (Claim 1); WO2003024392 (Claim 2; Fig 93); WO200166689 (Example 6);
Cross-references: LocusID:54894; NP_060233.2; NM_017763_1
(11) STEAP2 (HGNC 8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, prostate cancer associated gene 1 , prostate cancer associated protein 1 , six transmembrane epithelial antigen of prostate 2, six transmembrane prostate protein, Genbank accession no.
AF455138)
Lab. Invest. 82 (11): 1573-1582 (2002)); WO2003087306; US2003064397 (Claim 1; Fig 1); WO200272596 (Claim 13; Page 54-55); WO200172962 (Claim 1; Fig 4B); WO2003104270 (Claim 11); WO2003104270 (Claim 16); US2004005598 (Claim 22); WO2003042661 (Claim 12); US2003060612 (Claim 12; Fig 10); WO200226822 (Claim 23; Fig 2);
WO200216429 (Claim 12; Fig 10);
Cross-references: GL22655488; AAN04080.1; AF455138J
(12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, subfamily M, member 4, Genbank accession no. NM 017636)
Xu, X.Z., et al Proc. Natl. Acad. Sci. U.S.A. 98 (19): 10692-10697 (2001), Cell 109 (3):397- 407 (2002), J. Biol. Chem. 278 (33):30813-30820 (2003)); US2003143557 (Claim 4);
WO200040614 (Claim 14; Page 100-103); WO200210382 (Claim 1; Fig 9A);
WO2003042661 (Claim 12); WO200230268 (Claim 27; Page 391); US2003219806 (Claim 4); WO200162794 (Claim 14; Fig 1A-D);
Cross-references: MIM:606936; NP 060106.2; NM 017636 1
(13) CRIPTO (CR, CRl, CRGF, CRIPTO, TDGFl, teratocarcinoma-derived growth factor, Genbank accession no. NP 003203 or NM 003212)
Ciccodicola, A., et al EMBO J. 8 (7): 1987-1991 (1989), Am. J. Hum. Genet. 49 (3):555-565 (1991)); US2003224411 (Claim 1); WO2003083041 (Example 1); WO2003034984 (Claim 12); WO200288170 (Claim 2; Page 52-53); WO2003024392 (Claim 2; Fig 58);
WO200216413 (Claim 1; Page 94-95, 105); WO200222808 (Claim 2; Fig 1); US5854399 (Example 2; Col 17-18); US5792616 (Fig 2); Cross-references: MIM: 187395; NP_003203.1; NM_003212_1
(14) CD21 (CR2 (Complement receptor 2) or C3DR (C3d/Epstein Barr virus
receptor) or Hs.73792 Genbank accession no. M26004)
Fujisaku et al (1989) J. Biol. Chem. 264 (4):2118-2125); Weis J.J., et al J. Exp.
Med. 167, 1047-1066, 1988; Moore M., et al Proc. Natl. Acad. Sci. U.S.A. 84,
9194-9198, 1987; Barel M., et al Mol. Immunol. 35, 1025-1031, 1998; Weis J.J., et al Proc. Natl. Acad. Sci. U.S.A. 83, 5639-5643, 1986; Sinha S.K., et al (1993) J.
Immunol. 150, 5311-5320; WO2004045520 (Example 4); US2004005538
(Example 1); WO2003062401 (Claim 9); WO2004045520 (Example 4);
WO9102536 (Fig 9.1-9.9); WO2004020595 (Claim 1);
Accession: P20023; Q13866; Q14212; EMBL; M26004; AAA35786.1.
(15) CD79b (CD79B, CD79 , IGb (immunoglobulin-associated beta), B29, Genbank accession no. NM 000626 or 11038674)
Proc. Natl. Acad. Sci. U.S.A. (2003) 100 (7):4126-4131, Blood (2002) 100 (9):3068-3076, MuUer et al (1992) Eur. J. Immunol. 22 (6): 1621-1625); WO2004016225 (claim 2, Fig 140); WO2003087768, US2004101874 (claim 1, page 102); WO2003062401 (claim 9);
WO200278524 (Example 2); US2002150573 (claim 5, page 15); US5644033;
WO2003048202 (claim 1, pages 306 and 309); WO 99/558658, US6534482 (claim 13, Fig 17A/B); WO200055351 (claim 11, pages 1145-1146);
Cross-references: MIM: 147245; NP_000617.1; NM_000626_1
(16) FcRH2 (IFGP4, IRTA4, SPAPIA (SH2 domain containing phosphatase anchor protein la), SPAP1B, SPAP1C, Genbank accession no. NM 030764, AY358130)
Genome Res. 13 (10):2265-2270 (2003), Immunogenetics 54 (2):87-95 (2002), Blood 99 (8):2662-2669 (2002), Proc. Natl. Acad. Sci. U.S.A. 98 (17):9772-9777 (2001), Xu, M.J., et al (2001) Biochem. Biophys. Res. Commun. 280 (3):768-775; WO2004016225 (Claim 2); WO2003077836; WO200138490 (Claim 5; Fig 18D-1-18D-2); WO2003097803 (Claim 12); WO2003089624 (Claim 25);
Cross-references: MIM:606509; NP l 10391.2; NM 030764 1 (17) HER2 (ErbB2, Genbank accession no. Ml 1730)
Coussens L., et al Science (1985) 230(4730): 1132-1139); Yamamoto T., et al
Nature 319, 230-234, 1986; Semba K., et al Proc. Natl. Acad. Sci. U.S.A. 82, 6497- 6501, 1985; Swiercz J.M., et al J. Cell Biol. 165, 869-880, 2004; Kuhns J.J., et al J.
Biol. Chem. 274, 36422-36427, 1999; Cho H.-S., et al Nature 421, 756-760, 2003;
Ehsani A., et al (1993) Genomics 15, 426-429; WO2004048938 (Example 2);
WO2004027049 (Fig II); WO2004009622; WO2003081210; WO2003089904
(Claim 9); WO2003016475 (Claim 1); US2003118592; WO2003008537 (Claim 1);
WO2003055439 (Claim 29; Fig 1A-B); WO2003025228 (Claim 37; Fig 5C);
WO200222636 (Example 13; Page 95-107); WO200212341 (Claim 68; Fig 7);
WO200213847 (Page 71-74); WO200214503 (Page 114-117); WO200153463
(Claim 2; Page 41-46); WO200141787 (Page 15); WO200044899 (Claim 52; Fig
7); WO200020579 (Claim 3; Fig 2); US5869445 (Claim 3; Col 31-38);
WO9630514 (Claim 2; Page 56-61); EP1439393 (Claim 7); WO2004043361
(Claim 7); WO2004022709; WO200100244 (Example 3; Fig 4);
Accession: P04626; EMBL; Ml 1767; AAA35808.1. EMBL; Ml 1761;
AAA35808.1.
(18) NCA (CEACAM6, Genbank accession no. M18728);
Barnett T., et al Genomics 3, 59-66, 1988; Tawaragi Y., et al Biochem. Biophys. Res.
Commun. 150, 89-96, 1988; Strausberg R.L., et al Proc. Natl. Acad. Sci. U.S.A. 99: 16899-
16903, 2002; WO2004063709; EP1439393 (Claim 7); WO2004044178 (Example 4);
WO2004031238; WO2003042661 (Claim 12); WO200278524 (Example 2); WO200286443
(Claim 27; Page 427); WO200260317 (Claim 2);
Accession: P40199; Q14920; EMBL; M29541; AAA59915.1. EMBL; M18728;
(19) MDP (DPEP1, Genbank accession no. BC017023)
Proc. Natl. Acad. Sci. U.S.A. 99 (26): 16899-16903 (2002)); WO2003016475
(Claim 1); WO200264798 (Claim 33; Page 85-87); JP05003790 (Fig 6-8);
W09946284 (Fig 9);
Cross-references: MIM: 179780; AAH17023.1; BC017023 1 (20) IL20Ra (IL20Ra, ZCYTOR7, Genbank accession no. AF184971);
Clark H.F., et al Genome Res. 13, 2265-2270, 2003; Mungall A.J., et al Nature 425, 805-811, 2003; Blumberg H., et al Cell 104, 9-19, 2001; Dumoutier L., et al J.
Immunol. 167, 3545-3549, 2001; Parrish-Novak J., et al J. Biol. Chem. 277, 47517- 47523, 2002; Pletnev S., et al (2003) Biochemistry 42: 12617-12624; Sheikh F., et al (2004) J. Immunol. 172, 2006-2010; EP1394274 (Example 11); US2004005320
(Example 5); WO2003029262 (Page 74-75); WO2003002717 (Claim 2; Page 63);
WO200222153 (Page 45-47); US2002042366 (Page 20-21); WO200146261 (Page
57-59); WO200146232 (Page 63-65); W09837193 (Claim 1; Page 55-59);
Accession: Q9UHF4; Q6UWA9; Q96SH8; EMBL; AF184971; AAF01320.1.
(21) Brevican (BCAN, BEHAB, Genbank accession no. AF229053)
Gary S.C., et al Gene 256, 139-147, 2000; Clark H.F., et al Genome Res. 13, 2265- 2270, 2003; Strausberg R.L., et al Proc. Natl. Acad. Sci. U.S.A. 99, 16899-16903,
2002; US2003186372 (Claim 11); US2003186373 (Claim 11); US2003119131
(Claim 1; Fig 52); US2003119122 (Claim 1; Fig 52); US2003119126 (Claim 1);
US2003119121 (Claim 1; Fig 52); US2003119129 (Claim 1); US2003119130
(Claim 1); US2003119128 (Claim 1; Fig 52); US2003119125 (Claim 1);
WO2003016475 (Claim 1); WO200202634 (Claim 1);
(22) EphB2R (DRT, ERK, Hek5, EPHT3, Tyro5, Genbank accession no. NM 004442) Chan,J. and Watt, V.M., Oncogene 6 (6), 1057-1061 (1991) Oncogene 10 (5):897-905 (1995), Annu. Rev. Neurosci. 21 :309-345 (1998), Int. Rev. Cytol. 196: 177-244 (2000)); WO2003042661 (Claim 12); WO200053216 (Claim 1; Page 41); WO2004065576 (Claim 1); WO2004020583 (Claim 9); WO2003004529 (Page 128-132); WO200053216 (Claim 1; Page 42);
Cross-references: MIM:600997; NP_004433.2; NM_004442_1
(23) ASLG659 (B7h, Genbank accession no. AX092328)
US20040101899 (Claim 2); WO2003104399 (Claim 11); WO2004000221 (Fig 3);
US2003165504 (Claim 1); US2003124140 (Example 2); US2003065143 (Fig 60);
WO2002102235 (Claim 13; Page 299); US2003091580 (Example 2); WO200210187 (Claim 6; Fig 10); WO200194641 (Claim 12; Fig 7b); WO200202624 (Claim 13; Fig 1A-1B); US2002034749 (Claim 54; Page 45-46); WO200206317 (Example 2; Page 320-321, Claim 34; Page 321-322); WO200271928 (Page 468-469); WO200202587 (Example 1; Fig 1); WO200140269 (Example 3; Pages 190-192); WO200036107 (Example 2; Page 205-207); WO2004053079 (Claim 12); WO2003004989 (Claim 1); WO200271928 (Page 233-234, 452-453); WO 0116318;
(24) PSCA (Prostate stem cell antigen precursor, Genbank accession no. AJ297436) Reiter R.E., et al Proc. Natl. Acad. Sci. U.S.A. 95, 1735-1740, 1998; Gu Z., et al
Oncogene 19, 1288-1296, 2000; Biochem. Biophys. Res. Commun. (2000)
275(3):783-788; WO2004022709; EP1394274 (Example 11); US2004018553
(Claim 17); WO2003008537 (Claim 1); WO200281646 (Claim 1; Page 164);
WO2003003906 (Claim 10; Page 288); WO200140309 (Example 1; Fig 17);
US2001055751 (Example 1; Fig lb); WO200032752 (Claim 18; Fig 1);
WO9851805 (Claim 17; Page 97); W09851824 (Claim 10; Page 94); WO9840403 (Claim 2; Fig IB);
Accession: 043653; EMBL; AF043498; AAC39607.1.
(25) GEDA (Genbank accession No. AY260763);
AAP14954 lipoma HMGIC fusion-partner-like protein /pid=AAP14954.1 - Homo sapiens Species: Homo sapiens (human)
WO2003054152 (Claim 20); WO2003000842 (Claim 1); WO2003023013 (Example 3,
Claim 20); US2003194704 (Claim 45);
Cross-references: GL30102449; AAP14954.1; AY260763 1 (26) BAFF-R (B cell -activating factor receptor, BLyS receptor 3, BR3, Genbank accession No. AF116456); BAFF receptor /pid=NP_443177.1 - Homo sapiens
Thompson, J.S., et al Science 293 (5537), 2108-2111 (2001); WO2004058309;
WO2004011611; WO2003045422 (Example; Page 32-33); WO2003014294 (Claim 35; Fig 6B); WO2003035846 (Claim 70; Page 615-616); WO200294852 (Col 136-137);
WO200238766 (Claim 3; Page 133); WO200224909 (Example 3; Fig 3);
Cross-references: MIM:606269; NP_443177.1; NM_052945_1; AF 132600
(27) CD22 (B-cell receptor CD22-B isoform, BL-CAM, Lyb-8, Lyb8, SIGLEC-2, FLJ22814, Genbank accession No. AK026467); Wilson et al (1991) J. Exp. Med. 173: 137-146; WO2003072036 (Claim 1; Fig 1);
Cross-references: MIM: 107266; NP_001762.1; NM_001771_1
(28) CD79a (CD79A, CD79a, immunoglobulin-associated alpha, a B cell-specific protein that covalently interacts with Ig beta (CD79B) and forms a complex on the surface with Ig M molecules, transduces a signal involved in B-cell differentiation), pi: 4.84, MW: 25028 TM: 2 [P] Gene Chromosome: 19ql3.2, Genbank accession No. NP 001774.10)
WO2003088808, US20030228319; WO2003062401 (claim 9); US2002150573 (claim 4, pages 13-14); W09958658 (claim 13, Fig 16); WO9207574 (Fig 1); US5644033; Ha et al (1992) J. Immunol. 148(5): 1526-1531; Mueller et al (1992) Eur. J. Biochem. 22: 1621-1625; Hashimoto et al (1994) Immunogenetics 40(4):287-295; Preud'homme et al (1992) Clin. Exp. Immunol. 90(1): 141-146; Yu et al (1992) J. Immunol. 148(2) 633-637; Sakaguchi et al (1988) EMBO J. 7(11):3457-3464; (29) CXCR5 (Burkitt's lymphoma receptor 1, a G protein-coupled receptor that is activated by the CXCL13 chemokine, functions in lymphocyte migration and humoral defense, plays a role in HIV-2 infection and perhaps development of AIDS, lymphoma, myeloma, and leukemia); 372 aa, pi: 8.54 MW: 41959 TM: 7 [P] Gene Chromosome: l lq23.3, Genbank accession No. NP_001707.1)
WO2004040000; WO2004015426; US2003105292 (Example 2); US6555339 (Example 2); WO200261087 (Fig 1); WO200157188 (Claim 20, page 269); WO200172830 (pages 12- 13); WO200022129 (Example 1, pages 152-153, Example 2, pages 254-256); W09928468 (claim 1, page 38); US5440021 (Example 2, col 49-52); W09428931 (pages 56-58);
W09217497 (claim 7, Fig 5); Dobner et al (1992) Eur. J. Immunol. 22:2795-2799; Barella et al (1995) Biochem. J. 309:773-779;
(30) HLA-DOB (Beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4+ T lymphocytes); 273 aa, pi: 6.56 MW: 30820 TM: 1 [P] Gene Chromosome: 6p21.3, Genbank accession No. NP 002111.1)
Tonnelle et al (1985) EMBO J. 4(11):2839-2847; Jonsson et al (1989) Immunogenetics 29(6):411-413; Beck et al (1992) J. Mol. Biol. 228:433-441; Strausberg et al (2002) Proc. Natl. Acad. Sci USA 99: 16899-16903; Servenius et al (1987) J. Biol. Chem. 262:8759-8766; Beck et al (1996) J. Mol. Biol. 255: 1-13; Naruse et al (2002) Tissue Antigens 59:512-519; W09958658 (claim 13, Fig 15); US6153408 (Col 35-38); US5976551 (col 168-170);
US6011146 (col 145-146); Kasahara et al (1989) Immunogenetics 30(l):66-68; Larhammar et al (1985) J. Biol. Chem. 260(26): 14111-14119; (31) P2X5 (Purinergic receptor P2X ligand-gated ion channel 5, an ion channel gated by extracellular ATP, may be involved in synaptic transmission and neurogenesis, deficiency may contribute to the pathophysiology of idiopathic detrusor instability); 422 aa), pi: 7.63, MW: 47206 TM: 1 [P] Gene Chromosome: 17pl3.3, Genbank accession No. NP_002552.2) Le et al (1997) FEBS Lett. 418(1-2): 195-199; WO2004047749; WO2003072035 (claim 10); Touchman et al (2000) Genome Res. 10: 165-173; WO200222660 (claim 20);
WO2003093444 (claim 1); WO2003087768 (claim 1); WO2003029277 (page 82);
(32) CD72 (B-cell differentiation antigen CD72, Lyb-2) PROTEIN SEQUENCE Full maeaity...tafrfpd (1..359; 359 aa), pi: 8.66, MW: 40225 TM: 1 [P] Gene Chromosome: 9pl3.3, Genbank accession No. NP_001773.1)
WO2004042346 (claim 65); WO2003026493 (pages 51-52, 57-58); WO200075655 (pages 105-106); Von Hoegen et al (1990) J. Immunol. 144(12):4870-4877; Strausberg et al (2002) Proc. Natl. Acad. Sci USA 99: 16899-16903; (33) LY64 (Lymphocyte antigen 64 (RP105), type I membrane protein of the leucine rich repeat (LRR) family, regulates B-cell activation and apoptosis, loss of function is associated with increased disease activity in patients with systemic lupus erythematosis); 661 aa, pi: 6.20, MW: 74147 TM: 1 [P] Gene Chromosome: 5ql2, Genbank accession No.
NP_005573.1)
US2002193567; WO9707198 (claim 11, pages 39-42); Miura et al (1996) Genomics 38(3):299-304; Miura et al (1998) Blood 92:2815-2822; WO2003083047; W09744452 (claim 8, pages 57-61); WO200012130 (pages 24-26);
(34) FcRHl (Fc receptor- like protein 1, a putative receptor for the immunoglobulin Fc domain that contains C2 type Ig-like and IT AM domains, may have a role in B-lymphocyte differentiation); 429 aa, pi: 5.28, MW: 46925 TM: 1 [P] Gene Chromosome: Iq21-lq22, Genbank accession No. NP_443170.1) WO2003077836; WO200138490 (claim 6, Fig 18E-1-18-E-2); Davis et al (2001) Proc. Natl. Acad. Sci USA 98(17):9772-9777; WO2003089624 (claim 8); EP1347046 (claim 1);
WO2003089624 (claim 7); (35) IRTA2 (Immunoglobulin superfamily receptor translocation associated 2, a putative immunoreceptor with possible roles in B cell development and lymphomagenesis;
deregulation of the gene by translocation occurs in some B cell malignancies); 977 aa, pi: 6.88 MW: 106468 TM: 1 [P] Gene Chromosome: lq21, Genbank accession No.
Human:AF343662, AF343663, AF343664, AF343665, AF369794, AF397453, AK090423, AK090475, AL834187, AY358085; Mouse:AK089756, AY158090, AY506558;
NP_112571.1
WO2003024392 (claim 2, Fig 97); Nakayama et al (2000) Biochem. Biophys. Res.
Commun. 277(1): 124-127; WO2003077836; WO200138490 (claim 3, Fig 18B-1-18B-2); (36) TENB2 (TMEFF2, tomoregulin, TPEF, HPP 1 , TR, putative transmembrane
proteoglycan, related to the EGF/heregulin family of growth factors and follistatin); 374 aa, NCBI Accession: AAD55776, AAF91397, AAG49451, NCBI RefSeq: NP 057276; NCBI Gene: 23671; OMIM: 605734; SwissProt Q9UIK5; Genbank accession No. AF179274; AY358907, CAF85723, CQ782436
WO2004074320 (SEQ ID NO 810); JP2004113151 (SEQ ID NOS 2, 4, 8); WO2003042661 (SEQ ID NO 580); WO2003009814 (SEQ ID NO 411); EP1295944 (pages 69-70);
WO200230268 (page 329); WO200190304 (SEQ ID NO 2706); US2004249130;
US2004022727; WO2004063355; US2004197325; US2003232350; US2004005563;
US2003124579; Horie et al (2000) Genomics 67: 146-152; Uchida et al (1999) Biochem. Biophys. Res. Commun. 266:593-602; Liang et al (2000) Cancer Res. 60:4907-12; Glynne- Jones et al (2001) Int J Cancer. Oct 15;94(2): 178-84;
(37) PMEL17 (silver homolog; SILV; D12S53E; PMEL17; (SI); (SIL); ME20; gplOO) BC001414; BT007202; M32295; M77348; NM 006928; McGlinchey,R.P. et al (2009) Proc. Natl. Acad. Sci. U.S.A. 106 (33), 13731-13736; Kummer,M.P. et al (2009) J. Biol. Chem. 284 (4), 2296-2306;
(38) TMEFF1 (transmembrane protein with EGF-like and two follistatin-like domains 1; Tomoregulin- 1; H7365; C9orf2; C90RF2; U19878; X83961) NM 080655; NM 003692; Harms, P.W. (2003) Genes Dev. 17 (21), 2624-2629; Gery, S. et al (2003) Oncogene 22 (18):2723-2727;
(39) GDNF-Ral (GDNF family receptor alpha 1 ; GFRA1; GDNFR; GDNFRA; RETL1; TRNR1; RET1L; GDNFR-alphal; GFR-ALPHA-1; U95847; BC014962; NM 145793)
NM 005264; Kim,M.H. et al (2009) Mol. Cell. Biol. 29 (8), 2264-2277; Treanor, J.J. et al (1996) Nature 382 (6586): 80-83;
(40) Ly6E (lymphocyte antigen 6 complex, locus E; Ly67,RIG-E,SCA-2,TSA-l)
NP 002337.1; NM 002346.2; de Nooij-van Dalen,A.G. et al (2003) Int. J. Cancer 103 (6), 768-774; Zammit,D.J. et al (2002) Mol. Cell. Biol. 22 (3):946-952;
(41) TMEM46 (shisa homolog 2 (Xenopus laevis); SHISA2) NP 001007539.1;
NM 001007538.1; Furushima,K. et al (2007) Dev. Biol. 306 (2), 480-492; Clark,H.F. et al (2003) Genome Res. 13 (10):2265-2270;
(42) Ly6G6D (lymphocyte antigen 6 complex, locus G6D; Ly6-D, MEGT1) NP_067079.2; NM_021246.2; Mallya, M. et al (2002) Genomics 80 (1): 113-123; Ribas,G. et al (1999) J. Immunol. 163 (l):278-287;
(43) LGR5 (leucme-rich repeat-containing G protein-coupled receptor 5; GPR49, GPR67) NP 003658.1; NM 003667.2; Salanti,G. et al (2009) Am. J. Epidemiol. 170 (5):537-545; Yamamoto,Y. et al (2003) Hepatology 37 (3):528-533; (44) RET (ret proto-oncogene; MEN2A; HSCR1; MEN2B; MTC1; (PTC); CDHF12;
Hs.168114; RET51; RET-ELEl) NP 066124.1; NM 020975.4; Tsukamoto,H. et al (2009) Cancer Sci. 100 (10): 1895-1901; Narita,N. et al (2009) Oncogene 28 (34):3058-3068;
(45) LY6K (lymphocyte antigen 6 complex, locus K; LY6K; HSJ001348; FLJ35226) NP_059997.3; NM_017527.3; Ishikawa,N. et al (2007) Cancer Res. 67 (24): 11601-11611; de Nooij-van Dalen,A.G. et al (2003) Int. J. Cancer 103 (6):768-774; (46) GPR19 (G protein-coupled receptor 19; Mm.4787) NP_006134.1; NM_006143.2;
Montpetit, A. and Sinnett,D. (1999) Hum. Genet. 105 (1-2): 162-164; O'Dowd, B.F. et al (1996) FEBS Lett. 394 (3):325-329; (47) GPR54 (KISS1 receptor; KISS1R; GPR54; HOT7T175; AXOR12) NP l 15940.2;
NM 032551.4; Navenot, J.M. et al (2009) Mol. Pharmacol. 75 (6): 1300-1306; Hata, K. et al (2009) Anticancer Res. 29 (2):617-623;
(48) ASPHDl (aspartate beta-hydroxylase domain containing 1 ; LOC253982) NP_859069.2; NM_181718.3; Gerhard, D.S. et al (2004) Genome Res. 14 (10B):2121-2127;
(49) Tyrosinase (TYR; OCAIA; OCAIA; tyrosinase; SHEP3) NP 000363.1; NM 000372.4; Bishop,D.T. et al (2009) Nat. Genet. 41 (8):920-925; Nan, H. et al (2009) Int. J. Cancer 125 (4):909-917;
(50) TMEM118 (ring finger protein, transmembrane 2; RNFT2; FLJ14627)
NP 001103373.1; NM 001109903.1; Clark, H.F. et al (2003) Genome Res. 13 (10):2265- 2270; Scherer,S.E. et al (2006) Nature 440 (7082):346-351 (51) GPR172A (G protein-coupled receptor 172A; GPCR41; FLJ11856; D15Ertd747e)
NP_078807.1; NM 024531.3; Ericsson, T.A. et al (2003) Proc. Natl. Acad. Sci. U.S.A. 100 (l l):6759-6764; Takeda, S. et al (2002) FEBS Lett. 520 (l-3):97-101.
The parent antibody may also be a fusion protein comprising an albumin-binding peptide (ABP) sequence (Dennis et al. (2002) "Albumin Binding As A General Strategy For Improving The Pharmacokinetics Of Proteins" J Biol Chem. 277:35035-35043; WO
01/45746). Antibodies of the invention include fusion proteins with ABP sequences taught by: (i) Dennis et al (2002) J Biol Chem. 277:35035-35043 at Tables III and IV, page 35038; (ii) US 20040001827 at [0076] SEQ ID NOS: 9-22; and (iii) WO 01/45746 at pages 12-13, SEQ ID NOS: zl-zl4, and all of which are incorporated herein by reference.
MUTAGENESIS
DNA encoding an amino acid sequence variant of the starting polypeptide is prepared by a variety of methods known in the art. These methods include, but are not limited to, preparation by site-directed (or oligonucleotide-mediated) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared DNA encoding the polypeptide. Variants of recombinant antibodies may be constructed also by restriction fragment manipulation or by overlap extension PCR with synthetic oligonucleotides. Mutagenic primers encode the cysteine codon replacement(s). Standard mutagenesis techniques can be employed to generate DNA encoding such mutant cysteine engineered antibodies. General guidance can be found in Sambrook et al Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989; and Ausubel et al Current Protocols in Molecular Biology, Greene Publishing and Wiley-Interscience, New York, N.Y., 1993.
Site-directed mutagenesis is one method for preparing substitution variants, i.e.
mutant proteins. This technique is well known in the art (see for example, Carter (1985) et al Nucleic Acids Res. 13:4431-4443; Ho et al (1989) Gene (Amst.) 77:51-59; and Kunkel et al (1987) Proc. Natl. Acad. Sci. USA 82:488). Briefly, in carrying out site-directed mutagenesis of DNA, the starting DNA is altered by first hybridizing an oligonucleotide encoding the desired mutation to a single strand of such starting DNA. After hybridization, a DNA polymerase is used to synthesize an entire second strand, using the hybridized oligonucleotide as a primer, and using the single strand of the starting DNA as a template. Thus, the oligonucleotide encoding the desired mutation is incorporated in the resulting double-stranded DNA. Site-directed mutagenesis may be carried out within the gene expressing the protein to be mutagenized in an expression plasmid and the resulting plasmid may be sequenced to confirm the introduction of the desired cysteine replacement mutations (Liu et al (1998) J. Biol. Chem. 273:20252-20260). Site-directed of protocols and formats, including those commercially available, e.g. QuikChange® Multi Site-Directed Mutagenesis Kit (Stratagene, La Jolla, CA).
PCR mutagenesis is also suitable for making amino acid sequence variants of the starting polypeptide. See Higuchi, (1990) in PCR Protocols, pp.177-183, Academic Press; Ito et al (1991) Gene 102:67-70; Bernhard et al (1994) Bioconjugate Chem. 5: 126-132; and Vallette et al (1989) Nuc. Acids Res. 17:723-733. Briefly, when small amounts of template DNA are used as starting material in a PCR, primers that differ slightly in sequence from the corresponding region in a template DNA can be used to generate relatively large quantities of a specific DNA fragment that differs from the template sequence only at the positions where the primers differ from the template. Another method for preparing variants, cassette mutagenesis, is based on the technique described by Wells et al (1985) Gene 34:315-323. The starting material is the plasmid (or other vector) comprising the starting polypeptide DNA to be mutated. The codon(s) in the starting DNA to be mutated are identified. There must be a unique restriction endonuclease site on each side of the identified mutation site(s). If no such restriction sites exist, they may be generated using the above described oligonucleotide -mediated
mutagenesis method to introduce them at appropriate locations in the starting polypeptide DNA. The plasmid DNA is cut at these sites to linearize it. A double-stranded
oligonucleotide encoding the sequence of the DNA between the restriction sites but containing the desired mutation(s) is synthesized using standard procedures, wherein the two strands of the oligonucleotide are synthesized separately and then hybridized together using standard techniques. This double-stranded oligonucleotide is referred to as the cassette. This cassette is designed to have 5' and 3' ends that are compatible with the ends of the linearized plasmid, such that it can be directly ligated to the plasmid. This plasmid now contains the mutated DNA sequence. Mutant DNA containing the encoded cysteine replacements can be confirmed by DNA sequencing.
Single mutations are also generated by oligonucleotide directed mutagenesis using double stranded plasmid DNA as template by PCR based mutagenesis (Sambrook and Russel, (2001) Molecular Cloning: A Laboratory Manual, 3rd edition; Zoller et al (1983) Methods Enzymol. 100:468-500; Zoller, M.J. and Smith, M. (1982) Nucl. Acids Res.
10:6487-6500).
In the present invention, hu4D5Fabv8 displayed on M13 phage (Gerstner et al (2002) "Sequence Plasticity In The Antigen-Binding Site Of A Therapeutic Anti-HER2 Antibody", J Mol Biol. 321 : 851-62) was used for experiments as a model system. Cysteine mutations were introduced in hu4D5Fabv8-phage, hu4D5Fabv8, and ABP-hu4D5Fabv8 constructs. The hu4D5-ThioFab-Phage preps were carried out using the polyethylene glycol (PEG) precipitation method as described earlier (Lowman, Henry B. (1998) Methods in Molecular Biology (Totowa, New Jersey) 87 (Combinatorial Peptide Library Protocols) 249-264).
Oligonucleotides are prepared by the phosphoramidite synthesis method (US
4415732; US 4458066; Beaucage, S. and Iyer, R. (1992) "Advances in the synthesis of oligonucleotides by the phosphoramidite approach", Tetrahedron 48:2223-2311). The phosphoramidite method entails cyclical addition of nucleotide monomer units with a reactive 3 ' phosphoramidite moiety to an oligonucleotide chain growing on a solid-support comprised of controlled-pore glass or highly crosslinked polystyrene, and most commonly in the 3' to 5' direction in which the 3' terminus nucleoside is attached to the solid-support at the beginning of synthesis (US 5047524; US 5262530). The method is usually practiced using automated, commercially available synthesizers (Applied Biosystems, Foster City, CA). Oligonucleotides can be chemically labelled with non-isotopic moieties for detection, capture, stabilization, or other purposes (Andrus, A. "Chemical methods for 5' non-isotopic labelling of PCR probes and primers" (1995) in PCR 2: A Practical Approach, Oxford University Press, Oxford, pp. 39-54; Hermanson, G. in Bioconjugate Techniques (1996) Academic Press, San Diego, pp. 40-55, 643-671; Keller, G. and Manak, M. in DNA Probes Second Edition (1993), Stockton Press, New York, pp. 121-23). PHESELECTOR ASSAY
The PHESELECTOR (Phage ELISA for Selection of Reactive Thiols) assay allows for detection of reactive cysteine groups in antibodies in an ELISA phage format (US 7521541; Junutula JR et al. "Rapid identification of reactive cysteine residues for site- specific labeling of antibody-Fabs" J Immunol Methods 2008;332:41-52). The process of coating the protein (e.g. antibody) of interest on well surfaces, followed incubation with phage particles and then HRP labeled secondary antibody with absorbance detection is detailed in Example 2. Mutant proteins displayed on phage may be screened in a rapid, robust, and high-throughput manner. Libraries of cysteine engineered antibodies can be produced and subjected to binding selection using the same approach to identify
appropriately reactive sites of free Cys incorporation from random protein-phage libraries of antibodies or other proteins. This technique includes reacting cysteine mutant proteins displayed on phage with an affinity reagent or reporter group which is also thiol-reactive. Figure 8 illustrates the PHESELECTOR Assay by a schematic representation depicting the binding of Fab or ThioFab to HER2 (top) and biotinylated ThioFab to streptavidin (bottom). PROTEIN EXPRESSION AND PURIFICATION
DNA encoding the cysteine engineered antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells serve as a source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then trans fected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or other mammalian host cells, such as myeloma cells (US 5807715; US 2005/0048572; US 2004/0229310) that do not otherwise produce the antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The yields of hu4D5Fabv8 cysteine engineered antibodies were similar to wild type hu4D5Fabv8. Review articles on recombinant expression in bacteria of DNA encoding the antibody include Skerra et al (1993) Curr. Opinion in Immunol. 5:256- 262 and Pluckthun (1992) Immunol. Revs. 130: 151-188.
After design and selection, cysteine engineered antibodies, e.g. ThioFabs, with highly reactive unpaired Cys residues, may be produced by: (i) expression in a bacterial, e.g. E. coli, system or a mammalian cell culture system (WO 01/00245), e.g. Chinese Hamster Ovary cells (CHO); and (ii) purification using common protein purification techniques (Lowman et al (1991) J. Biol. Chem. 266(17): 10982-10988).
ThioFabs were expressed upon induction in 34B8, a non-suppressor E. coli strain (Baca et al (1997) Journal Biological Chemistry 272(16): 10678-84). See Example 3a. The harvested cell pellet was resuspended in PBS (phosphate buffered saline), total cell lysis was performed by passing through a microfluidizer and the ThioFabs were purified by affinity chromatography with protein G SEPHAROSE™ (Amersham). ThioFabs were conjugated with biotin-PEO-maleimide as described above and the biotinylated-ThioFabs were further purified by Superdex-200™ (Amersham) gel filtration chromatography, which eliminated the free biotin-PEO-maleimide and the oligomeric fraction of ThioFabs.
MASS SPECTROSCOPY ANALYSIS
Liquid chromatography electrospray ionization mass spectrometric (LC-ESI-MS) analysis was employed for the accurate molecular weight determination of biotin conjugated Fab (Cole, R.B. Electro Spray Ionization Mass Spectrometry: Fundamentals, Instrumentation And Applications. (1997) Wiley, New York). The amino acid sequence of biotinylated hu4D5Fabv8 (A121C) peptide was determined by tryptic digestion followed by LC-ESI- Tandem MS analysis (Table 4, Example 3b).
The antibody Fab fragment hu4D5Fabv8 contains about 445 amino acid residues, including 10 Cys residues (five on the light and five on the heavy chain). The high-resolution structure of the humanized 4D5 variable fragment (Fv4D5) has been established, see:
Eigenbrot et al "X-Ray Structures Of The Antigen-Binding Domains From Three Variants Of Humanized Anti-P185her2 Antibody 4D5 And Comparison With Molecular Modeling" (1993) J Mol Biol. 229:969-995). All the Cys residues are present in the form of disulfide bonds, therefore these residues do not have any reactive thiol groups to conjugate with zirconium-maleimide (unless treated with a reducing agent). Hence, the newly engineered Cys residue, can remain unpaired, and able to react with, i.e. conjugate to, an electrophilic linker reagent or zirconium-linker intermediate, such as a zirconium-maleimide. Figure 1 A shows a three-dimensional representation of the hu4D5Fabv8 antibody fragment derived by X-ray crystal coordinates. The structure positions of the engineered Cys residues of the heavy and light chains are numbered according to a sequential numbering system. This sequential numbering system is correlated to the Kabat numbering system (Kabat et al., (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD) for the 4d5v7fabH variant of trastuzumab according to Figure IB which shows the sequential numbering scheme (top row), starting at the N-terminus, differs from the Kabat numbering scheme (bottom row) by insertions noted by a,b,c. Using the Kabat numbering system, the actual linear amino acid sequence may contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the variable domain. The cysteine engineered heavy chain variant sites are identified by the sequential numbering and Kabat numbering schemes in the following chart:
Figure imgf000051_0001
Ml 3 phagemid-Cys mutant Fabs (Figures 3 A and 3B) can be rapidly screened compared to Fab proteins. Phagemid-ThioFab binding to antigen and to streptavidin can be tested by coating HER2 and streptavidin, respectively, onto ELISA plates followed by probing with anti-Fab-HRP (Horse radish peroxidase) as described in Example 2 and depicted in Figure 8. This method allowed simultaneous monitoring of the effect on the antigen binding and the reactivity of the thiol group by the engineered Cys
residue/conjugated biotin molecule. Also, the method can be applied to screen the reactive thiol groups for any protein displayed on Ml 3 phage. Conjugated or unconjugated phagemid-ThioFabs are purified by simple PEG precipitation.
The antigen-binding fragment of humanized 4D5 (hu4D5Fab) is well expressed in E. Coli and has been displayed on bacteriophage (Garrard et al (1993) Gene 128: 103-109). The antibody Fab fragment hu4D5Fabv8 was displayed on M13 phage as a model system in the ELISA based assay to probe thiol reactivity. Figure 8 is a graphical representation of the PHESELECTOR assay, depicting binding of a biotinylated ThioFab phage and an anti-phage HRP antibody to HER2 (top) and Streptavidin (bottom). Five amino acid residues (L-Ala43, H-Ala40, H-Serl 19, H-Alal21 and H-Serl22) were initially selected from crystal structure information as remote from the antigen binding surface (Eigenbrot et al. (1993) J Mol Biol. 229:969-995). The Protein Database X-ray crystal structure was designated as 1FVC. Cys residues were engineered at these positions by site directed mutagenesis. ThioFab-phage preparations were isolated and reacted with the biotinylation reagent.
Biotin conjugated and unconjugated variants were tested for HER2 and streptavidin binding using an ELISA based PHESELECTOR assay (Figure 8, Example 2) with an HRP (horseradish peroxidase)-conjugated anti-phage antibody. The interaction of non-biotinylated phage-hu4D5Fabv8 (Figure 2A) and biotinylated phage-hu4D5Fabv8 (Figure 2B) with BSA (open box), HER2 (grey box) or streptavidin (solid box) were monitored through anti-M13- horseradish peroxidase (HRP) antibody by developing a standard HRP reaction and measuring absorbance at 450 nm. The absorbance produced by turnover of a colorimetric substrate was measured at 450 nm. The reactivity of ThioFab with HER2 measures antigen binding. The reactivity of ThioFab with streptavidin measures the extent of biotinylation. The reactivity of ThioFab with BSA is a negative control for nonspecific interaction. As seen in Figure 2A, all the ThioFab-phage variants have similar binding to HER2 compared to that of wild type hu4D5Fabv8-phage. Furthermore, conjugation with biotin did not interfere in the ThioFab binding to HER2 (Figure 2B).
Surprisingly and unexpectedly, the ThioFabs-phage samples showed varying levels of streptavidin binding activity. From all the tested phage-ThioFabs, the A121C cysteine engineered antibody exhibited maximal thiol reactivity. Even though wild type hu4D5Fabv8- phage was incubated with the same amounts of biotin-maleimide, these phage had little streptavidin binding indicating that preexisting cysteine residues (involved in disulfide bond formation) from the hu4D5Fabv8 and Ml 3 phage coat proteins did not interfere with the site- specific conjugation of biotin-maleimide. These results demonstrate that the phage ELISA assay can be used successfully to screen reactive thiol groups on the Fab surface.
The PHESELECTOR assay allows screening of reactive thiol groups in antibodies.
Identification of the A121C variant by this method is exemplary. The entire Fab molecule may be effectively searched to identify more ThioFab variants with reactive thiol groups. A parameter, fractional surface accessibility, was employed to identify and quantitate the accessibility of solvent to the amino acid residues in a polypeptide. The surface accessibility can be expressed as the surface area (A2) that can be contacted by a solvent molecule, e.g. water. The occupied space of water is approximated as a 1.4 A radius sphere. Software is freely available or licensable (Secretary to CCP4, Daresbury Laboratory, Warrington, WA4 4 AD, United Kingdom, Fax: (+44) 1925 603825, or by internet:
www.ccp4.ac.uk/dist/html/INDEX.html) as the CCP4 Suite of crystallography programs which employ algorithms to calculate the surface accessibility of each amino acid of a protein with known x-ray crystallography derived coordinates ("The CCP4 Suite: Programs for Protein Crystallography" (1994) Acta. Cryst. D50:760-763). Two exemplary software modules that perform surface accessibility calculations are "AREAIMOL" and "SURFACE", based on the algorithms of B.Lee and F.M.Richards (1971) J.Mol.Biol. 55:379-400.
AREAIMOL defines the solvent accessible surface of a protein as the locus of the centre of a probe sphere (representing a solvent molecule) as it rolls over the Van der Waals surface of the protein. AREAIMOL calculates the solvent accessible surface area by generating surface points on an extended sphere about each atom (at a distance from the atom centre equal to the sum of the atom and probe radii), and eliminating those that lie within equivalent spheres associated with neighboring atoms. AREAIMOL finds the solvent accessible area of atoms in a PDB coordinate file, and summarizes the accessible area by residue, by chain and for the whole molecule. Accessible areas (or area differences) for individual atoms can be written to a pseudo-PDB output file. AREAIMOL assumes a single radius for each element, and only recognizes a limited number of different elements. Unknown atom types (i.e. those not in AREAIMOL's internal database) will be assigned the default radius of 1.8 A. The list of recognized atoms is:
Atom Atomic no. Van der Waals rad. (A)
C 6 1.80
N 7 1.65
O 8 1.60
Mg 12 1.60
S 16 1.85
P 15 1.90
CI 17 1.80
Co 27 1.80
AREAIMOL and SURFACE report absolute accessibilities, i.e. the number of square Angstroms (A). Fractional surface accessibility is calculated by reference to a standard state relevant for an amino acid within a polypeptide. The reference state is tripeptide Gly-X-Gly, where X is the amino acid of interest, and the reference state should be an 'extended' conformation, i.e. like those in beta-strands. The extended conformation maximizes the accessibility of X. A calculated accessible area is divided by the accessible area in a Gly-X- Gly tripeptide reference state and reports the quotient, which is the fractional accessibility. Percent accessibility is fractional accessibility multiplied by 100.
Another exemplary algorithm for calculating surface accessibility is based on the SOLV module of the program xsae (Broger, C, F. Hoffman-LaRoche, Basel) which calculates fractional accessibility of an amino acid residue to a water sphere based on the X- ray coordinates of the polypeptide.
The fractional surface accessibility for every amino acid in hu4D5Fabv7 was calculated using the crystal structure information (Eigenbrot et al. (1993) J Mol Biol.
229:969-995). The fractional surface accessibility values for the amino acids of the light chain and heavy chain of hu4D5Fabv7 are shown in descending order in Table 1.
Table 1.
hu4D5Fabv7-light chain
SER A 202 frac acc= 101. 236 ASP A 151 frac acc= 41. 586
GLY A 41 frac acc= 90. 775 SER A 12 frac acc= 40. 633
GLY A 157 frac acc= 88. 186 ASN A 210 frac acc= 40. 158
ASP A 1 frac acc= 87. 743 SER A 63 frac acc= 39. 872
SER A 156 frac acc= 83. 742 ARG A 66 frac acc= 39. 669
GLY A 57 frac acc= 81. 611 PRO A 8 frac acc= 39. 297
SER A 168 frac acc= 79. 680 SER A 65 frac acc= 39. 219
SER A 56 frac acc= 79. 181 SER A 77 frac acc= 38. 820
LYS A 169 frac acc= 77. .591 THR A 180 frac acc= 38. , 296
SER A 60 frac acc= 75. .291 ASP A 185 frac acc= 38. , 234
THR A 109 frac acc= 74. .603 THR A 31 frac acc= 38. , 106
CYS A 214 frac acc= 72. .021 THR A 94 frac acc= 37. , 452
LYS A 126 frac acc= 71. .002 THR A 93 frac acc= 37. ,213
SER A 67 frac acc= 66. , 694 THR A 197 frac acc= 36. , 709
ARG A 18 frac acc= 66. , 126 SER A 182 frac acc= 36. , 424
ASN A 152 frac acc= 65. ,415 GLY A 128 frac acc= 35. , 779
SER A 127 frac acc= 65. , 345 LYS A 207 frac acc= 35. , 638
LYS A 190 frac acc= 65. ,189 ASP A 17 frac acc= 35. ,413
LYS A 145 frac acc= 63. , 342 GLY A 200 frac acc= 35. , 274
GLN A 199 frac acc= 62. , 470 GLU A 165 frac acc= 35. ,067
GLU A 143 frac acc= 61. .681 ALA A 112 frac acc= 34. .912
GLN A 3 frac acc= 59. .976 GLN A 79 frac acc= 34. .601
LYS A 188 frac acc= 59. .680 VAL A 191 frac acc= 33. .935
ARG A 24 frac acc= 59. .458 SER A 208 frac acc= 33. .525
PHE A 53 frac acc= 58. .705 LYS A 39 frac acc= 33. .446
SER A 9 frac acc= 58. .446 GLU A 123 frac acc= 32. .486
GLN A 27 frac acc= 57. .247 THR A 69 frac acc= 32. .276
ALA A 153 frac acc= 56. .538 SER A 76 frac acc= 32. .108
SER A 203 frac acc= 55. .864 HIS A 189 frac acc= 31. .984
LYS A 42 frac acc= 54. .730 ARG A 108 frac acc= 31. .915
GLY A 16 frac acc= 54. .612 ASN A 158 frac acc= 31. .447
LYS A 45 frac acc= 54. .464 VAL A 205 frac acc= 31. .305
PRO A 204 frac acc= 53. .172 SER A 14 frac acc= 31 , .094
GLU A 213 frac acc= 53. .084 GLN A 155 frac acc= 30 , .630
ALA A 184 frac acc= 52. .556 GLU A 187 frac acc= 30 , .328
VAL A 15 frac acc= 52. .460 ARG A 211 frac acc= 30 , .027
SER A 7 frac acc= 51. .936 LYS A 183 frac acc= 29, .751
LEU A 154 frac acc= 51 , .525 ASN A 138 frac acc= 29, .306
GLN A 100 frac acc= 51 , .195 ASP A 170 frac acc= 29, .041
SER A 10 frac acc= 49 , .907 SER A 159 frac acc= 27 , .705
THR A 5 frac acc= 48. .879 GLN A 147 frac acc= 27 , .485
THR A 206 frac acc= 48. .853 THR A 22 frac acc= 27 , .121
ASP A 28 frac acc= 48. .758 ALA A 43 frac acc= 26, .801
GLY A 68 frac acc= 48. .690 ARG A 142 frac acc= 26, .447
THR A 20 frac acc= 48 .675 LEU A 54 frac acc= 25 .882
ASP A 122 frac acc= 47 .359 ASP A 167 frac acc= 25 .785
PRO A 80 frac acc= 46 .984 THR A 129 frac acc= 23 .880
SER A 52 frac acc= 46 .917 ALA A 144 frac acc= 23 .652
SER A 26 frac acc= 46 .712 VAL A 163 frac acc= 22 .261
TYR A 92 frac acc= 46 .218 PRO A 95 frac acc= 20 .607
LYS A 107 frac acc= 45 .912 ALA A 111 frac acc= 19 .942
GLU A 161 frac acc= 45 .100 LYS A 103 frac acc= 18 .647
VAL A 110 frac acc= 44 .844 LEU A 181 frac acc= 18 .312
GLU A 81 frac acc= 44 .578 THR A 72 frac acc= 18 .226
PRO A 59 frac acc= 44 .290 GLU A 195 frac acc= 18 .006
ASN A 30 frac acc= 42 .721 THR A 178 frac acc= 17 .499
GLN A 160 frac acc= 42 .692 THR A 85 frac acc= 17 .343 SER A 114 frac acc= 42.374 ASP A 70 frac acc= 17.194
PRO A 40 frac acc= 41. 928 LEU A 11 frac acc= 16. 568
PHE A 116 frac acc= 16. 406 LEU A 125 frac acc= 2. 398
THR A 97 frac acc= 16. 204 PRO A 96 frac acc= 2. 387
ARG A 61 frac acc= 16. 192 LEU A 47 frac acc= 2. 180
TYR A 49 frac acc= 16. 076 ALA A 51 frac acc= 1. 837
SER A 50 frac acc= 15. 746 PHE A 118 frac acc= 1. 779
LYS A 149 frac acc= 15. 510 PHE A 62 frac acc= 1. 581
GLU A 55 frac acc= 14. 927 ALA A 25 frac acc= 1. 538
LEU A 201 frac acc= 14. 012 VAL A 133 frac acc= 1. 315
GLY A 64 frac acc= 13. .735 ASP A 82 frac acc= 1. .141
GLY A 212 frac acc= 13. , 396 LEU A 179 frac acc= 0. .872
PHE A 98 frac acc= 12. , 852 GLN A 124 frac acc= 0. ,787
THR A 74 frac acc= 12. ,169 MET A 4 frac acc= 0. , 778
SER A 171 frac acc= 11. , 536 SER A 177 frac acc= 0. , 693
PRO A 141 frac acc= 11. , 073 SER A 131 frac acc= 0. , 693
PHE A 83 frac acc= 10. , 871 LEU A 135 frac acc= 0. , 654
THR A 164 frac acc= 10. , 325 PHE A 71 frac acc= 0. , 593
ALA A 32 frac acc= 9. , 971 TRP A 35 frac acc= 0. , 448
HIS A 198 frac acc= 9. .958 PHE A 209 frac acc= 0. , 395
VAL A 146 frac acc= 9. .861 TYR A 186 frac acc= 0. .259
SER A 121 frac acc= 9. .833 LEU A 78 frac acc= 0. .157
ALA A 13 frac acc= 9. .615 VAL A 196 frac acc= 0. .000
GLU A 105 frac acc= 9. .416 VAL A 132 frac acc= 0. .000
SER A 162 frac acc= 9. .304 VAL A 104 frac acc= 0. .000
ILE A 117 frac acc= 8. .780 VAL A 33 frac acc= 0. .000
HIS A 91 frac acc= 8. .557 VAL A 29 frac acc= 0. .000
ALA A 193 frac acc= 8. .547 TYR A 192 frac acc= 0. .000
GLN A 37 frac acc= 8. .442 TYR A 86 frac acc= 0. .000
VAL A 58 frac acc= 8. .281 TYR A 36 frac acc= 0. .000
PRO A 120 frac acc= 8. .095 THR A 102 frac acc= 0. .000
GLN A 38 frac acc= 6. .643 SER A 174 frac acc= 0. .000
PRO A 113 frac acc= 6, .594 PHE A 139 frac acc= 0 , .000
GLY A 101 frac acc= 6, .558 LEU A 136 frac acc= 0 , .000
TYR A 140 frac acc= 5, .894 LEU A 73 frac acc= 0 , .000
VAL A 115 frac acc= 5, .712 ILE A 75 frac acc= 0 , .000
TYR A 87 frac acc= 4 , .539 ILE A 48 frac acc= 0 , .000
SER A 176 frac acc= 4 , .106 ILE A 21 frac acc= 0 , .000
ILE A 2 frac acc= 4 , .080 GLN A 90 frac acc= 0 , .000
ASN A 137 frac acc= 3, .906 GLN A 89 frac acc= 0. .000
TRP A 148 frac acc= 3. .676 CYS A 194 frac acc= 0. .000
GLY A 99 frac acc= 3 .550 CYS A 134 frac acc= 0 .000
PRO A 44 frac acc= 3 .543 CYS A 88 frac acc= 0 .000
LEU A 175 frac acc= 3 .488 CYS A 23 frac acc= 0 .000
VAL A 19 frac acc= 3 .420 ALA A 130 frac acc= 0 .000
ILE A 106 frac acc= 3 .337 ALA A 84 frac acc= 0 .000
PRO A 119 frac acc= 2 .953 ALA A 34 frac acc= 0 .000
LEU A 46 frac acc= 2 .887
GLN A 6 frac acc= 2 .860
TYR A 173 frac acc= 2 .825
VAL A 150 frac acc= 2 .525
GLN A 166 frac acc= 2 .525
THR A 172 frac acc= 2 .436 hu4D5Fabv7-heavy chain
SER B 179 frac acc= 99.479 PRO B 14 frac acc= 45. .729 GLY B 42 frac acc= 95.850 THR B 54 frac acc= 45. .503 GLU B 1 frac acc= 87.276 THR B 200 frac acc= 45. .369 GLY B 66 frac acc= 84.541 LEU B 177 frac acc= 45.337
ASP B 102 frac acc= 83. 794 GLY B 8 frac acc= 44. 898
SER B 75 frac acc= 80. 567 SER B 7 frac acc= 43. 530
GLY B 140 frac acc= 80. 344 THR B 69 frac acc= 43. 503
ASN B 211 frac acc= 79. 588 PRO B 220 frac acc= 43. 378
GLY B 197 frac acc= 78. 676 LYS B 208 frac acc= 43. 138
ASP B 62 frac acc= 77. ,716 LYS B 30 frac acc= 42. 380
GLY B 103 frac acc= 77. , 176 ALA B 23 frac acc= 41. 952
SER B 163 frac acc= 76. , 664 GLU B 46 frac acc= 41. 430
SER B 139 frac acc= 74. , 946 SER B 25 frac acc= 41. 323
LYS B 213 frac acc= 74. , 442 ARG B 87 frac acc= 41. 282
ALA B 165 frac acc= 74. , 339 LYS B 124 frac acc= 40. 888
THR B 167 frac acc= 73. , 934 ASN B 28 frac acc= 40. 529
SER B 122 frac acc= 72. , 870 GLN B 3 frac acc= 39. 824
SER B 194 frac acc= 71. , 959 THR B 123 frac acc= 39. 306
PRO B 41 frac acc= 71. , 540 SER B 63 frac acc= 38. 867
THR B 198 frac acc= 68. .668 GLY B 56 frac acc= 38. , 582
SER B 222 frac acc= 68. .128 GLY B 169 frac acc= 38. ,469
LYS B 43 frac acc= 67. .782 THR B 172 frac acc= 38. , 421
GLY B 26 frac acc= 67. .782 PRO B 209 frac acc= 38. , 309
THR B 138 frac acc= 65. .826 GLY B 101 frac acc= 38. , 040
ASP B 31 frac acc= 64. .222 TYR B 109 frac acc= 36. , 829
GLY B 15 frac acc= 64. .172 LYS B 221 frac acc= 36. , 520
SER B 168 frac acc= 62. .100 GLY B 44 frac acc= 35. , 147
SER B 120 frac acc= 61. .332 GLY B 181 frac acc= 34. , 735
LYS B 76 frac acc= 61. .092 THR B 58 frac acc= 34. , 457
GLY B 141 frac acc= 59. .419 GLY B 9 frac acc= 34. .254
SER B 137 frac acc= 59, .179 VAL B 5 frac acc= 34. .198
TYR B 57 frac acc= 58 , .916 ALA B 121 frac acc= 33. .049
GLU B 89 frac acc= 58 , .483 SER B 127 frac acc= 32. .390
SER B 180 frac acc= 56, .289 GLY B 10 frac acc= 32. .230
LYS B 65 frac acc= 55, .044 SER B 71 frac acc= 30. .659
ASP B 215 frac acc= 54 , .656 ASP B 73 frac acc= 30. .245
GLN B 13 frac acc= 53, .719 LEU B 115 frac acc= 29. .867
GLN B 112 frac acc= 53, .215 LEU B 11 frac acc= 29. .825
TYR B 105 frac acc= 51, .940 ASN B 84 frac acc= 29. .765
ALA B 88 frac acc= 51 .602 SER B 210 frac acc= 28. .656
GLY B 164 frac acc= 50 .259 GLU B 155 frac acc= 28 , .162
PRO B 192 frac acc= 49 .826 SER B 160 frac acc= 26, .526
THR B 158 frac acc= 49 .694 CYS B 223 frac acc= 26, .270
THR B 142 frac acc= 48 .896 GLY B 16 frac acc= 26, .158
ASN B 55 frac acc= 48 .344 ILE B 202 frac acc= 26, .068
LYS B 136 frac acc= 48 .312 GLN B 82 frac acc= 25, .836
ARG B 19 frac acc= 48 .082 SER B 193 frac acc= 25, .550
PRO B 156 frac acc= 47 .366 ASN B 77 frac acc= 25, .418
PRO B 174 frac acc= 47 .157 ARG B 59 frac acc= 25 .301
LYS B 217 frac acc= 47 .102 VAL B 93 frac acc= 25 .254
GLN B 199 frac acc= 46 .650 THR B 74 frac acc= 24 .902
SER B 17 frac acc= 45 .980 GLU B 219 frac acc= 24 .778
SER B 85 frac acc= 45 .824 ASN B 206 frac acc= 24 .647
VAL B 170 frac acc= 24 .549 PRO B 154 frac acc= 6 .767
TYR B 52 frac acc= 24 .298 PRO B 133 frac acc= 6 .767 ALA B 175 frac acc= 23.804 TRP B 99 frac acc= 6.502
LYS B 216 frac acc= 23. 277 THR B 32 frac acc= 6. 291
VAL B 214 frac acc= 23. 150 LEU B 45 frac acc= 4. 649
GLY B 125 frac acc= 22. 802 VAL B 128 frac acc= 4. 515
ASN B 162 frac acc= 22. 245 ILE B 51 frac acc= 4. 307
ALA B 72 frac acc= 22. 166 SER B 186 frac acc= 4. 084
ALA B 40 frac acc= 21. 974 PHE B 173 frac acc= 3. 969
LEU B 18 frac acc= 20. 273 ARG B 38 frac acc= 3. 734
THR B 212 frac acc= 20. 170 TRP B 47 frac acc= 3. 561
LEU B 182 frac acc= 19. 619 VAL B 118 frac acc= 3. 409
TYR B 33 frac acc= 19. 398 ALA B 24 frac acc= 3. 376
THR B 190 frac acc= 19. 365 TYR B 95 frac acc= 3. 242
VAL B 176 frac acc= 18. 941 GLU B 6 frac acc= 3. 216
SER B 21 frac acc= 18. .929 ALA B 144 frac acc= 3. .167
SER B 119 frac acc= 18. , 877 ILE B 70 frac acc= 1. .958
THR B 91 frac acc= 18. , 237 GLY B 111 frac acc= 1. .868
ASP B 151 frac acc= 17. , 849 LEU B 4 frac acc= 1. .808
THR B 114 frac acc= 17. , 601 TYR B 201 frac acc= 1. , 758
SER B 134 frac acc= 17. , 571 LEU B 148 frac acc= 1. , 744
LEU B 196 frac acc= 17. ,090 PHE B 68 frac acc= 1. , 708
TYR B 60 frac acc= 16. , 575 VAL B 188 frac acc= 1. , 315
TYR B 183 frac acc= 15. , 968 CYS B 22 frac acc= 0. , 935
VAL B 2 frac acc= 15. .901 TRP B 161 frac acc= 0. .876
PRO B 130 frac acc= 15. .342 LEU B 131 frac acc= 0. .654
LEU B 166 frac acc= 15. .268 VAL B 205 frac acc= 0. .495
GLY B 100 frac acc= 15. .003 ALA B 92 frac acc= 0. .356
PHE B 27 frac acc= 14. .383 ALA B 79 frac acc= 0. .356
ASN B 204 frac acc= 13. .873 VAL B 64 frac acc= 0. .263
PHE B 104 frac acc= 13. .836 ILE B 29 frac acc= 0. .227
TYR B 80 frac acc= 13. .490 VAL B 218 frac acc= 0. .000
VAL B 159 frac acc= 12. .782 VAL B 189 frac acc= 0. .000
ARG B 67 frac acc= 12. .362 VAL B 149 frac acc= 0. .000
GLN B 178 frac acc= 12 , .131 VAL B 116 frac acc= 0. .000
HIS B 171 frac acc= 11 , .412 VAL B 48 frac acc= 0. .000
SER B 184 frac acc= 11 , .255 VAL B 37 frac acc= 0 , .000
ARG B 98 frac acc= 11 , .115 TYR B 152 frac acc= 0. .000
PRO B 53 frac acc= 11 , .071 TYR B 94 frac acc= 0. .000
GLN B 39 frac acc= 11 , .037 TRP B 36 frac acc= 0. .000
SER B 195 frac acc= 10. .909 SER B 187 frac acc= 0. .000
ASP B 108 frac acc= 10 .525 SER B 97 frac acc= 0 .000
LEU B 185 frac acc= 10 .464 MET B 107 frac acc= 0 .000
GLY B 113 frac acc= 10 .406 MET B 83 frac acc= 0 .000
THR B 78 frac acc= 10 .213 LEU B 145 frac acc= 0 .000
THR B 117 frac acc= 9 .990 LEU B 86 frac acc= 0 .000
LYS B 150 frac acc= 9 .447 LEU B 81 frac acc= 0 .000
VAL B 157 frac acc= 9 .323 LEU B 20 frac acc= 0 .000
VAL B 12 frac acc= 9 .207 ILE B 34 frac acc= 0 .000
TRP B 110 frac acc= 9 .069 HIS B 207 frac acc= 0 .000
ALA B 143 frac acc= 8 .903 HIS B 35 frac acc= 0 .000
SER B 135 frac acc= 8 .897 GLY B 146 frac acc= 0 .000
PHE B 129 frac acc= 8 .895 CYS B 203 frac acc= 0 .000
ARG B 50 frac acc= 8 .639 CYS B 147 frac acc= 0 .000
ALA B 61 frac acc= 8 .547 CYS B 96 frac acc= 0 .000
ALA B 132 frac acc= 7 .882 ASP B 90 frac acc= 0 .000
VAL B 191 frac acc= 7 .366 ALA B 106 frac acc= 0 .000
PRO B 126 frac acc= 7 .258 ALA B 49 frac acc= 0 .000
PHE B 153 frac acc= 6 .918 The following two criteria were applied to identify the residues of hu4D5Fabv8 that can be engineered to replace with Cys residues:
1. Amino acid residues that are completely buried are eliminated, i.e. less than 10% fractional surface accessibility. Table 1 shows there are 134 (light chain) and 151 (heavy chain) residues of hu4D5Fabv8 that are more than 10% accessible (fractional surface accessibility). The top ten most accessible Ser, Ala and Val residues were selected due to their close structural similarity to Cys over other amino acids, introducing only minimal structural constraints in the antibody by newly engineered Cys. Other cysteine replacement sites can also be screened, and may be useful for conjugation.
2. Residues are sorted based on their role in functional and structural interactions of
Fab. The residues which are not involved in antigen interactions and distant from the existing disulfide bonds were further selected. The newly engineered Cys residues should be distinct from, and not interfere with, antigen binding nor mispair with cysteines involved in disulfide bond formation.
The following residues of hu4D5Fabv8 possessed the above criteria and were selected to be replaced with Cys: L-V15, L-A43, L-V110, L-A144, L-S168, H-A88, H-A121, H-S122, H-A175 and H-S179 (shown in Figure 1).
Thiol reactivity may be generalized to any antibody where substitution of amino acids with reactive cysteine amino acids may be made within the ranges in the light chain selected from: L-10 to L-20; L-38 to L-48; L-105 to L-l 15; L-139 to L-149; L-163 to L-173; and within the ranges in the heavy chain selected from: H-35 to H-45; H-83 to H-93; H-l 14 to H- 127; and H-l 70 to H-l 84, and in the Fc region within the ranges selected from H-268 to H- 291; H-319 to H-344; H-370 to H-380; and H-395 to H-405.
Thiol reactivity may also be generalized to certain domains of an antibody, such as the light chain constant domain (CL) and heavy chain constant domains, CHI, CH2 and CH3. Cysteine replacements resulting in thiol reactivity values of 0.6 and higher may be made in the heavy chain constant domains α, δ, ε, γ, and μ of intact antibodies: IgA, IgD, IgE, IgG, and IgM, respectively, including the IgG subclasses: IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
It is evident from the crystal structure data that the selected 10 Cys mutants are far away from the antigen-combining site, such as the interface with HER2 in this case. These mutants can be tested experimentally for indirect effects on functional interactions. The thiol reactivities of all the Cys Fab variants were measured and calculated as described in
Examples 1 and 2, and presented in Table 2. The residues L-V15C, L-Vl IOC, H-A88C and H-A121C have reactive and stable thiol groups (Figures 3A and 3B). Mutants V15C, VI IOC, A144C, S168C are light chain Cys variants. Mutants A88C, A121C, A175C, S179C are heavy chain Cys variants. It was surprising and unexpected that the sites with high fractional surface accessibility did not have the highest thiol reactivity as calculated by the PHESELECTOR assay (Table 2). In other words, fractional surface accessibility (Tables 1, 2) did not correlate with thiol reactivity (Table 2). In fact, the Cys residues engineered at the sites with moderate surface accessibility of 20% to 80% (Figure 4A, Table 1), or partially exposed sites, like Ala or Val residues, exhibited better thiol reactivity, i.e. >0.6, (Figure 3B, Table 2) than the Cys introduced at Ser residues, thus necessitating the use of
PHESELECTOR assay in the screening of thiol reactive sites since the crystal structure information alone is not sufficient to select these sites (Figure 3B and 4A).
Thiol reactivity data is shown in Figures 3A and 3B for amino acid residues of 4D5 ThioFab Cys mutants: (3 A) non-biotinylated (control) and (3B) biotinylated phage-ThioFabs. Reactive thiol groups on antibody/Fab surface were identified by PHESELECTOR assay analyses for the interaction of non-biotinylated phage-hu4D5Fabv8 (3 A) and biotinylated phage-hu4D5Fabv8 (3B) with BSA (open box), HER2 (grey box) or streptavidin (solid box). The assay was carried out as described in Example 2. Light chain variants are on the left side and heavy chain variants are on the right side. The binding of non-biotinylated 4D5 ThioFab Cys mutants is low as expected, but strong binding to HER2 is retained. The ratio of binding to streptavidin and to HER2 of the biotinylated 4D5 ThioFab Cys mutants gives the thiol reactivity values in Table 2. Background absorbance at 450 nm or small amounts of nonspecific protein binding of the biotinylated 4D5 ThioFab Cys mutants to BSA is also evident in Figure 3B. Fractional Surface Accessibility values of the selected amino acid residues that were replaced with a Cys residue are shown in Figure 4A. Fractional surface accessibility was calculated from the available hu4D5Fabv7 structure and shown on Table 1 (Eigenbrot et al. (1993) J Mol Biol. 229:969-995). The conformational parameters of the hu4D5Fabv7 and hu4D5Fabv8 structures are highly consistent and allow for determination of any correlation between fractional surface accessibility calculations of hu4D5Fabv7 and thiol reactivity of hu4D5Fabv8 cysteine mutants. The measured thiol reactivity of phage ThioFab Cys residues introduced at partially exposed residues (Ala or Val) have better thiol reactivity compared to the ones introduced at Ser residues (Table 2). It can be seen from the ThioFab Cys mutants of Table 2 that there is little or no correlation between thio reactivity values and fractional surface accessibility.
Amino acids at positions L-15, L-43, L-l 10, L-144, L-168, H-40, H-88, H-l 19, H- 121, H-l 22, H-l 75, and H-l 79 of an antibody may generally be mutated (replaced) with free cysteine amino acids. Ranges within about 5 amino acid residues on each side of these positions may also be replaced with free cysteine acids, i.e. L-10 to L-20; L-38 to L-48; L- 105 to L-l 15; L-139 to L-149; L-163 to L-173; H-35 to H-45; H-83 to H-93; H-l 14 to H- 127; and H-l 70 to H-l 84, as well as the ranges in the Fc region selected from H-268 to H- 291; H-319 to H-344; H-370 to H-380; and H-395 to H-405, to yield the cysteine engineered antibodies of the invention.
Table 2. Thiol reactivity of phage -ThioFabs
Figure imgf000061_0001
L = light chain, H = heavy chain, A = alanine, S = serine, V = valine, C = cysteine
Thiol reactivity is measured as the ratio of OD450 nm for streptavidin binding to OD450„ for HER2 (antibody) binding (Example 2). Thiol reactivity value of 1 indicates complete biotinylation of the cysteine thiol.
Two Cys variants from light chain (L-V15C and L-Vl IOC) and two from heavy chain
(H-A88C and H-A121C) were selected for further analysis as these variants showed the highest thiol reactivity (Table 2).
Unlike phage purification, Fab preparation may require 2-3 days, depending on the scale of production. During this time, thiol groups may lose reactivity due to oxidation. To probe the stability of thiol groups on hu4D5Fabv8-phage, stability of the thiol reactivity of phage-thioFabs was measured (Figure 4B). After ThioFab-phage purification, on day 1, day 2 and day 4, all the samples were conjugated with biotin-PEO-maleimide and probed with phage ELISA assay (PHESELECTOR) to test HER2 and streptavidin binding. L-V15C, L- VI IOC, H-A88C and H-A121C retain significant amounts of thiol reactivity compared to other ThioFab variants (Figure 4B).
METHODS TO PREPARE CYSTEINE ENGINEERED ANTIBODIES
The compounds of the invention include cysteine engineered antibodies where one or more amino acids of a parent antibody are replaced with a free cysteine amino acid. A cysteine engineered antibody comprises one or more free cysteine amino acids having a thiol reactivity value in the range of 0.6 to l .O. A free cysteine amino acid is a cysteine residue which has been engineered into the parent antibody and is not part of a disulfide bridge.
In one aspect, the cysteine engineered antibody is prepared by a process comprising:
(a) replacing one or more amino acid residues of a parent antibody by cysteine; and
(b) determining the thiol reactivity of the cysteine engineered antibody by reacting the cysteine engineered antibody with a thiol-reactive reagent.
The cysteine engineered antibody may be more reactive than the parent antibody with the thiol-reactive reagent.
The free cysteine amino acid residues may be located in the heavy or light chains, or in the constant or variable domains. Antibody fragments, e.g. Fab, may also be engineered with one or more cysteine amino acids replacing amino acids of the antibody fragment, to form cysteine engineered antibody fragments.
Another aspect of the invention provides a method of preparing (making) a cysteine engineered antibody, comprising:
(a) introducing one or more cysteine amino acids into a parent antibody in order to generate the cysteine engineered antibody; and
(b) determining the thiol reactivity of the cysteine engineered antibody with a thiol-reactive reagent;
wherein the cysteine engineered antibody is more reactive than the parent antibody with the thiol-reactive reagent.
Step (a) of the method of preparing a cysteine engineered antibody may comprise:
(i) mutagenizing a nucleic acid sequence encoding the cysteine engineered antibody;
(ii) expressing the cysteine engineered antibody; and (iii) isolating and purifying the cysteine engineered antibody. Step (b) of the method of preparing a cysteine engineered antibody may comprise expressing the cysteine engineered antibody on a viral particle selected from a phage or a phagemid particle.
Step (b) of the method of preparing a cysteine engineered antibody may also comprise:
(i) reacting the cysteine engineered antibody with a thiol-reactive affinity reagent to generate an affinity labelled, cysteine engineered antibody; and
(ii) measuring the binding of the affinity labelled, cysteine engineered antibody to a capture media.
Another aspect of the invention is a method of screening cysteine engineered antibodies with highly reactive, unpaired cysteine amino acids for thiol reactivity
comprising:
(a) introducing one or more cysteine amino acids into a parent antibody in order to generate a cysteine engineered antibody;
(b) reacting the cysteine engineered antibody with a thiol-reactive affinity reagent to generate an affinity labelled, cysteine engineered antibody; and
(c) measuring the binding of the affinity labelled, cysteine engineered antibody to a capture media; and
(d) determining the thiol reactivity of the cysteine engineered antibody with the thiol-reactive reagent.
Step (a) of the method of screening cysteine engineered antibodies may comprise:
(i) mutagenizing a nucleic acid sequence encoding the cysteine engineered antibody;
(ii) expressing the cysteine engineered antibody; and
(iii) isolating and purifying the cysteine engineered antibody.
Step (b) of the method of screening cysteine engineered antibodies may comprise expressing the cysteine engineered antibody on a viral particle selected from a phage or a phagemid particle.
Step (b) of the method of screening cysteine engineered antibodies may also comprise:
(i) reacting the cysteine engineered antibody with a thiol-reactive affinity reagent to generate an affinity labelled, cysteine engineered antibody; and (ii) measuring the binding of the affinity labelled, cysteine engineered antibody to a capture media.
LABELLED CYSTEINE ENGINEERED ANTIBODIES
The cysteine engineered antibodies of the invention may be conjugated with any label moiety which can be covalently attached to the antibody through a reactive cysteine thiol group (Singh et al (2002) Anal. Biochem. 304: 147-15; Harlow E. and Lane, D. (1999) Using Antibodies: A Laboratory Manual, Cold Springs Harbor Laboratory Press, Cold Spring Harbor, NY; Lundblad R.L. (1991) Chemical Reagents for Protein Modification, 2nd ed. CRC Press, Boca Raton, FL). The attached label may function to: (i) provide a detectable signal; (ii) interact with a second label to modify the detectable signal provided by the first or second label, e.g. to give FRET (fluorescence resonance energy transfer); (iii) stabilize interactions or increase affinity of binding, with antigen or ligand; (iv) affect mobility, e.g. electrophoretic mobility or cell-permeability, by charge, hydrophobicity, shape, or other physical parameters, or (v) provide a capture moiety, to modulate ligand affinity,
antibody/antigen binding, or ionic complex ation.
Labelled cysteine engineered antibodies may be useful in diagnostic assays, e.g., for detecting expression of an antigen of interest in specific cells, tissues, or serum. For diagnostic applications, the antibody will typically be labeled with a detectable moiety.
Numerous labels are available which can be generally grouped into the following categories:
(a) Radioisotopes (radionuclides), such as 3H, nC, 14C, 18F, 32P, 35 S, 64Cu, 68Ga,
Figure imgf000064_0001
antibodies are useful in receptor targeted imaging experiments. The antibody can be labeled with ligand reagents that bind, chelate or otherwise complex a radioisotope metal where the reagent is reactive with the engineered cysteine thiol of the antibody, using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley- Interscience, New York, NY, Pubs. (1991). Chelating ligands which may complex a metal ion include DOT A, DOPA, DOTP, DOTMA, DTP A and TETA (Macrocyclics, Dallas, TX). Radionuclides can be targetted via complexation with cysteine-engineered antibodies as antibody-zirconium conjugates of the invention (Wu et al (2005) Nature Biotechnology 23(9): 1137-1146).
Metal-chelate complexes suitable as antibody labels for imaging experiments are disclosed: US 5342606; US 5428155; US 5316757; US 5480990; US 5462725; US 5428139; US 5385893; US 5739294; US 5750660; US 5834456; Hnatowich et al (1983) J. Immunol. Methods 65: 147-157; Meares et al (1984) Anal. Biochem. 142:68-78; Mirzadeh et al (1990) Bioconjugate Chem. 1 :59-65; Meares et al (1990) J. Cancerl990, Suppl. 10:21-26; Izard et al (1992) Bioconjugate Chem. 3:346-350; Nikula et al (1995) Nucl. Med. Biol. 22:387-90; Camera et al (1993) Nucl. Med. Biol. 20:955-62; Kukis et al (1998) J. Nucl. Med. 39:2105- 2110; Verel et al (2003) J. Nucl. Med. 44: 1663-1670; Camera et al (1994) J. Nucl. Med. 21 :640-646; Ruegg et al (1990) Cancer Res. 50:4221-4226; Verel et al (2003) J. Nucl. Med. 44: 1663-1670; Lee et al (2001) Cancer Res. 61 :4474-4482; Mitchell, et al (2003) J. Nucl. Med. 44: 1105-1112; Kobayashi et al (1999) Bioconjugate Chem. 10: 103-11 1; Miederer et al (2004) J. Nucl. Med. 45: 129-137; DeNardo et al (1998) Clinical Cancer Research 4:2483-90; Blend et al (2003) Cancer Biotherapy & Radiopharmaceuticals 18:355-363; Nikula et al (1999) J. Nucl. Med. 40: 166-76; Kobayashi et al (1998) J. Nucl. Med. 39:829-36;
Mardirossian et al (1993) Nucl. Med. Biol. 20:65-74; Roselli et al (1999) Cancer Biotherapy & Radiopharmaceuticals, 14:209-20.
(b) Fluorescent labels such as rare earth chelates (europium chelates), fluorescein types including FITC, 5-carboxyfluorescein, 6-carboxy fluorescein; rhodamine types including TAMRA; dansyl; Lissamine; cyanines; phycoerythrins; Texas Red; and analogs thereof. The fluorescent labels can be conjugated to antibodies using the techniques disclosed in Current Protocols in Immunology, supra, for example. Fluorescent dyes and fluorescent label reagents include those which are commercially available from
Invitrogen/Molecular Probes (Eugene, OR) and Pierce Biotechnology, Inc. (Rockford, IL).
(c) Various enzyme-substrate labels are available or disclosed (US 4275149). The enzyme generally catalyzes a chemical alteration of a chromogenic substrate that can be measured using various techniques. For example, the enzyme may catalyze a color change in a substrate, which can be measured spectrophotometrically. Alternatively, the enzyme may alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above. The chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (using a chemiluminometer, for example) or donates energy to a fluorescent acceptor.
Examples of enzymatic labels include luciferases {e.g., firefly luciferase and bacterial luciferase; US 4737456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRP), alkaline phosphatase (AP), β- galactosidase, glucoamylase, lysozyme, saccharide oxidases {e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like. Techniques for conjugating enzymes to antibodies are described in O'Sullivan et al (1981) "Methods for the Preparation of Enzyme- Antibody Conjugates for use in Enzyme Immunoassay", in Methods in Enzym. (ed J. Langone & H. Van Vunakis), Academic Press, New York, 73: 147-166.
Examples of enzyme-substrate combinations include, for example:
(i) Horseradish peroxidase (HRP) with hydrogen peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye precursor (e.g., orthophenylene diamine (OPD) or 3,3',5,5'-tetramethylbenzidine hydrochloride (TMB));
(ii) alkaline phosphatase (AP) with para-nitrophenyl phosphate as chromogenic substrate; and
(iii) β-D-galactosidase (β-D-Gal) with a chromogenic substrate {e.g., p- nitrophenyl- -D-galactosidase) or fluorogenic substrate 4-methylumbelliferyl- -D- galactosidase.
Numerous other enzyme-substrate combinations are available to those skilled in the art. For a general review, see US 4275149 and US 4318980.
A label may be indirectly conjugated with a cysteine engineered antibody. For example, the antibody can be conjugated with biotin and any of the three broad categories of labels mentioned above can be conjugated with avidin or streptavidin, or vice versa. Biotin binds selectively to streptavidin and thus, the label can be conjugated with the antibody in this indirect manner. Alternatively, to achieve indirect conjugation of the label with the polypeptide variant, the polypeptide variant is conjugated with a small hapten (e.g., digoxin) and one of the different types of labels mentioned above is conjugated with an anti-hapten polypeptide variant {e.g., anti-digoxin antibody). Thus, indirect conjugation of the label with the polypeptide variant can be achieved (Hermanson, G. (1996) in Bioconjugate Techniques Academic Press, San Diego).
The polypeptide variant of the present invention may be employed in any known assay method, such as ELISA, competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays (Zola, (1987) Monoclonal Antibodies: A Manual of Techniques, pp.147-158, CRC Press, Inc.).
A detection label may be useful for localizing, visualizing, and quantitating a binding or recognition event. The labelled antibodies of the invention can detect cell-surface receptors. Another use for detectably labelled antibodies is a method of bead-based immunocapture comprising conjugating a bead with a fluorescent labelled antibody and detecting a fluorescence signal upon binding of a ligand. Similar binding detection methodologies utilize the surface plasmon resonance (SPR) effect to measure and detect antibody-antigen interactions.
Detection labels such as fluorescent dyes and chemiluminescent dyes (Briggs et al (1997) "Synthesis of Functionalised Fluorescent Dyes and Their Coupling to Amines and
Amino Acids," J. Chem. Soc, Perkin-Trans. 1 : 1051-1058) provide a detectable signal and are generally applicable for labelling antibodies, preferably with the following properties: (i) the labelled antibody should produce a very high signal with low background so that small quantities of antibodies can be sensitively detected in both cell-free and cell-based assays; and (ii) the labelled antibody should be photostable so that the fluorescent signal may be observed, monitored and recorded without significant photo bleaching. For applications involving cell surface binding of labelled antibody to membranes or cell surfaces, especially live cells, the labels preferably (iii) have good water-solubility to achieve effective conjugate concentration and detection sensitivity and (iv) are non-toxic to living cells so as not to disrupt the normal metabolic processes of the cells or cause premature cell death.
Direct quantification of cellular fluorescence intensity and enumeration of
fluorescently labelled events, e.g. cell surface binding of peptide-dye conjugates may be conducted on an system (FMAT® 8100 HTS System, Applied Biosystems, Foster City, Calif.) that automates mix-and-read, non-radioactive assays with live cells or beads (Miraglia, "Homogeneous cell- and bead-based assays for high throughput screening using fluorometric microvolume assay technology", (1999) J. of Biomolecular Screening 4: 193-204). Uses of labelled antibodies also include cell surface receptor binding assays, inmmunocapture assays, fluorescence linked immunosorbent assays (FLISA), caspase-cleavage (Zheng, "Caspase-3 controls both cytoplasmic and nuclear events associated with Fas-mediated apoptosis in vivo", (1998) Proc. Natl. Acad. Sci. USA 95:618-23; US 6372907), apoptosis (Vermes, "A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V" (1995) J. Immunol. Methods 184:39-51) and cytotoxicity assays. Fluorometric microvolume assay technology can be used to identify the up or down regulation by a molecule that is targeted to the cell surface
(Swartzman, "A homogeneous and multiplexed immunoassay for high-throughput screening using fluorometric microvolume assay technology", (1999) Anal. Biochem. 271 : 143-51).
Labelled cysteine engineered antibodies of the invention are useful as imaging biomarkers and probes by the various methods and techniques of biomedical and molecular imaging such as: (i) MRI (magnetic resonance imaging); (ii) MicroCT (computerized tomography); (iii) SPECT (single photon emission computed tomography); (iv) PET
(positron emission tomography) Chen et al (2004) Bioconjugate Chem. 15:41-49; (v) bioluminescence; (vi) fluorescence; and (vii) ultrasound. Immunoscintigraphy is an imaging procedure in which antibodies labeled with radioactive substances are administered to an animal or human patient and a picture is taken of sites in the body where the antibody localizes (US 6528624). Imaging biomarkers may be objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention. Biomarkers may be of several types: Type 0 are natural history markers of a disease and correlate longitudinally with known clinical indices, e.g. MRI assessment of synovial inflammation in rheumatoid arthritis; Type I markers capture the effect of an intervention in accordance with a mechanism-of-action, even though the mechanism may not be associated with clinical outcome; Type II markers function as surrogate endpoints where the change in, or signal from, the biomarker predicts a clinical benefit to "validate" the targeted response, such as measured bone erosion in rheumatoid arthritis by CT. Imaging biomarkers thus can provide pharmacodynamic (PD) therapeutic information about: (i) expression of a target protein, (ii) binding of a therapeutic to the target protein, i.e. selectivity, and (iii) clearance and half-life pharmacokinetic data. Advantages of in vivo imaging biomarkers relative to lab-based biomarkers include: non-invasive treatment, quantifiable, whole body assessment, repetitive dosing and assessment, i.e. multiple time points, and potentially transferable effects from preclinical (small animal) to clinical (human) results. For some applications, bioimaging supplants or minimizes the number of animal experiments in preclinical studies.
3 1 1 14 18 32 35
Radionuclide imaging labels include radionuclides such as H, C, C, F, P, S,
64^ Cu, 68^ Ga, 86v Y, Zr, 99^ Tc, 1 11 T In, 123T I, 124T I, 125T I, 131 T I, 133 Xve, 177T Lu, 21 1 At,, or 213d Bi-. r T,he radionuclide metal ion can be complexed with a chelating linker such as DOTA. Linker reagents such as DOTA-maleimide (4-maleimidobutyramidobenzyl-DOTA) can be prepared by the reaction of aminobenzyl-DOTA with 4-maleimidobutyric acid (Fluka) activated with isopropylchloro formate (Aldrich), following the procedure of Axworthy et al (2000) Proc. Natl. Acad. Sci. USA 97(4): 1802-1807). DOTA-maleimide reagents react with the free cysteine amino acids of the cysteine engineered antibodies and provide a metal complexing ligand on the antibody (Lewis et al (1998) Bioconj. Chem. 9:72-86). Chelating linker labelling reagents such as DOTA-NHS (l,4,7,10-tetraazacyclododecane-l,4,7,10-tetraacetic acid mono (N-hydroxysuccinimide ester) are commercially available (Macrocyclics, Dallas, TX). Receptor target imaging with radionuclide labelled antibodies can provide a marker of pathway activation by detection and quantitation of progressive accumulation of antibodies in tumor tissue (Albert et al (1998) Bioorg. Med. Chem. Lett. 8: 1207-1210). The conjugated radio-metals may remain intracellular following lysosomal degradation.
Peptide labelling methods are well known. See Haugland, 2003, Molecular Probes Handbook of Fluorescent Probes and Research Chemicals, Molecular Probes, Inc.; Brinkley, 1992, Bioconjugate Chem. 3:2; Garman, (1997) Non-Radioactive Labelling: A Practical Approach, Academic Press, London; Means (1990) Bioconjugate Chem. 1 :2; Glazer et al (1975) Chemical Modification of Proteins. Laboratory Techniques in Biochemistry and Molecular Biology (T. S. Work and E. Work, Eds.) American Elsevier Publishing Co., New York; Lundblad, R. L. and Noyes, C. M. (1984) Chemical Reagents for Protein Modification, Vols. I and II, CRC Press, New York; Pfleiderer, G. (1985) "Chemical Modification of Proteins", Modern Methods in Protein Chemistry, H. Tschesche, Ed., Walter DeGryter, Berlin and New York; and Wong (1991) Chemistry of Protein Conjugation and Cross- linking, CRC Press, Boca Raton, Fla.); De Leon-Rodriguez et al (2004) Chem.Eur. J.
10: 1149-1155; Lewis et al (2001) Bioconjugate Chem. 12:320-324; Li et al (2002)
Bioconjugate Chem. 13: 110-115; Mier et al (2005) Bioconjugate Chem. 16:240-237.
Peptides and proteins labelled with two moieties, a fluorescent reporter and quencher in sufficient proximity undergo fluorescence resonance energy transfer (FRET). Reporter groups are typically fluorescent dyes that are excited by light at a certain wavelength and transfer energy to an acceptor, or quencher, group, with the appropriate Stokes shift for emission at maximal brightness. Fluorescent dyes include molecules with extended aromaticity, such as fluorescein and rhodamine, and their derivatives. The fluorescent reporter may be partially or significantly quenched by the quencher moiety in an intact peptide. Upon cleavage of the peptide by a peptidase or protease, a detectable increase in fluorescence may be measured (Knight, C. (1995) "Fluorimetric Assays of Proteolytic Enzymes", Methods in Enzymology, Academic Press, 248: 18-34).
The labelled antibodies of the invention may also be used as an affinity purification agent. In this process, the labelled antibody is immobilized on a solid phase such a Sephadex resin or filter paper, using methods well known in the art. The immobilized antibody is contacted with a sample containing the antigen to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the antigen to be purified, which is bound to the immobilized polypeptide variant. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the antigen from the polypeptide variant. Labelling reagents typically bear reactive functionality which may react (i) directly with a cysteine thiol of a cysteine engineered antibody to form the labelled antibody, (ii) with a linker reagent to form a linker-label intermediate, or (iii) with a linker antibody to form the labelled antibody. Reactive functionality of labelling reagents include: maleimide, haloacetyl, iodoacetamide succinimidyl ester (e.g. NHS, N-hydroxysuccinimide), isothiocyanate, sulfonyl chloride, 2,6-dichlorotriazinyl, pentafluorophenyl ester, and phosphoramidite, although other functional groups can also be used.
An exemplary reactive functional group is N-hydroxysuccinimidyl ester (NHS) of a carboxyl group substituent of a detectable label, e.g. biotin or a fluorescent dye. The NHS ester of the label may be preformed, isolated, purified, and/or characterized, or it may be formed in situ and reacted with a nucleophilic group of an antibody. Typically, the carboxyl form of the label is activated by reacting with some combination of a carbodiimide reagent, e.g. dicyclohexylcarbodiimide, diisopropylcarbodiimide, or a uranium reagent, e.g. TSTU (0-(N-Succinimidyl)-N,N,N',N'-tetramethyluronium tetrafluoroborate, HBTU (O- benzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate), or HATU (0-(7- azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate), an activator, such as 1-hydroxybenzotriazole (HOBt), and N-hydroxysuccinimide to give the NHS ester of the label. In some cases, the label and the antibody may be coupled by in situ activation of the label and reaction with the antibody to form the label-antibody conjugate in one step. Other activating and coupling reagents include TBTU (2-(lH-benzotriazo-l-yl)-l-l,3,3- tetramethyluronium hexafluorophosphate), TFFH (N,N',N",N"'-tetramethyluronium 2- fluoro-hexafluorophosphate), PyBOP (benzotriazole- 1 -yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate, EEDQ (2-ethoxy-l-ethoxycarbonyl-l,2-dihydro-quinoline), DCC (dicyclohexylcarbodiimide); DIPCDI (diisopropylcarbodiimide), MSNT (l-(mesitylene-2- sulfonyl)-3-nitro-lH-l,2,4-triazole, and aryl sulfonyl halides, e.g.
triisopropylbenzenesulfonyl chloride.
CONJUGATION OF BIOTIN-MALEIMIDE TO THIOFABS
The above-described ThioFab properties were established in the presence of phage because fusion of the Fab to the phage coat protein could potentially alter Cys thiol accessibility or reactivity. Therefore, the ThioFab constructs were cloned into an expression vector under alkaline phosphatase promoter (Chang et al (1987) Gene 55: 189-196) and the ThioFab expression was induced by growing E. coli cells in the phosphate-free medium. ThioFabs were purified on a Protein G SEPHAROSE™ column and analyzed on reducing and non-reducing SDS-PAGE gels. These analyses allow assessment of whether ThioFabs retained their reactive thiol group or were rendered inactive by forming intramolecular or intermolecular disulfide bonds. ThioFabs L-V15C, L-Vl IOC, H-A88C, and H-A121C were expressed and purified by Protein-G SEPHAROSE™ column chromatography (see methods sections for details). Purified proteins were analyzed on SDS-PAGE gel in reducing (with DTT) and non-reducing (without DTT) conditions. Other reducing agents such as BME (beta-mercaptoethanol) can used in the gel to cleave interchain disulfide groups. It is evident from SDS-PAGE gel analysis that the major (-90%) fraction of ThioFab is in the monomeric form, while wild type hu4D5Fabv8 is essentially in the monomeric form (47 kDa).
ThioFab (A121C) and wild type hu4D5Fabv8 were incubated with 100 fold excess of biotin-maleimide for 3 hours at room temperature and the biotinylated Fabs were loaded onto a Superdex-200™ gel filtration column. This purification step was useful in separating monomeric Fab from oligomeric Fab and also from excess free biotin-maleimide (or free zirconium reagent).
Figure 5 shows validation of the properties of ThioFab variants in the absence of the phage context. The proteins without phage fusion, hu4D5Fabv8 and hu4D5Fabv8-A121C (ThioFab-A121C), were expressed and purified using protein-G agarose beads followed by incubation with 100 fold molar excess of biotin-maleimide. Streptavidin and HER2 binding of a biotinylated cys engineered ThioFab and a non-biotinylated wild type Fab was compared. The extent of biotin conjugation (interaction with streptavidin) and their binding ability to HER2 were monitored by ELISA analyses. Each Fab was tested at 2ng and 20ng.
Biotinylated A121C ThioFab retained comparable HER2 binding to that of wild type hu4D5Fabv8 (Figure 5). Wild type Fab and A121C-ThioFab were purified by gel filtration column chromatography. The two samples were tested for HER2 and streptavidin binding by ELISA using goat anti-Fab-HRP as secondary antibody. Both wild type (open box) and
ThioFab (dotted box) have similar binding to HER2 but only ThioFab retained streptavidin binding. Only a background level of interaction with streptavidin was observed with non- biotinylated wild type hu4D5Fabv8 (Figure 5). Mass spectral (LC-ESI-MS) analysis of biotinylated-ThioFab (A121C) resulted in a major peak with 48294.5 daltons compared to the wild type hu4D5Fabv8 (47737 daltons). The 537.5 daltons difference between the two molecules exactly corresponds to a single biotin-maleimide conjugated to the ThioFab. Mass spec protein sequencing (LC-ESI-Tandem mass spec analysis) results further confirmed that the conjugated biotin molecule was at the newly engineered Cys residue (Table 4, Example 3). SITE SPECIFIC CONJUGATION OF BIOTIN-MALEIMIDE TO ALBUMIN BINDING PEPTIDE (ABP)-THIOFABS
Plasma-protein binding can be an effective means of improving the pharmacokinetic properties of short lived molecules. Albumin is the most abundant protein in plasma. Serum albumin binding peptides (ABP) can alter the pharmacodynamics of fused active domain proteins, including alteration of tissue uptake, penetration, and diffusion. These
pharmacodynamic parameters can be modulated by specific selection of the appropriate serum albumin binding peptide sequence (US 20040001827). A series of albumin binding peptides were identified by phage display screening (Dennis et al. (2002) "Albumin Binding As A General Strategy For Improving The Pharmacokinetics Of Proteins" J Biol Chem. 277:35035-35043; WO 01/45746). Compounds of the invention include ABP sequences taught by: (i) Dennis et al (2002) J Biol Chem. 277:35035-35043 at Tables III and IV, page 35038; (ii) US 20040001827 at [0076] SEQ ID NOS: 9-22; and (iii) WO 01/45746 at pages 12-13, SEQ ID NOS: zl-zl4, and all of which are incorporated herein by reference.
Albumin Binding (ABP)-Fabs were engineered by fusing an albumin binding peptide to the C-terminus of Fab heavy chain in 1 : 1 stoichiometric ratio (1 ABP / 1 Fab). It was shown that association of these ABP-Fabs with albumin increased their half life by more than 25 fold in rabbits and mice. The above described reactive Cys residues can therefore be introduced in these ABP-Fabs and used for site-specific conjugation with zirconium reagents followed by in vivo animal studies.
Exemplary albumin binding peptide sequences include, but are not limited to the amino acid sequences listed in SEQ ID NOS: 1-5:
CDKTHTGGGSQRLMEDICLPRWGCLWEDDF SEQ ID NO: l
QRLMEDICLPRWGCLWEDDF SEQ ID NO:2 QRLIEDICLPRWGCLWEDDF SEQ ID NO:3
RLIEDICLPRWGCLWEDD SEQ ID NO :4
DICLPRWGCLW SEQ ID NO: 5
The albumin binding peptide (ABP) sequences bind albumin from multiple species (mouse, rat, rabbit, bovine, rhesus, baboon, and human) with Kd (rabbit) = 0.3 μΜ. The albumin binding peptide does not compete with ligands known to bind albumin and has a half life (T½) in rabbit of 2.3 hr. ABP-ThioFab proteins were purified on BSA-SEPHAROSE™ followed by biotin-maleimide conjugation and purification on Superdex-S200 column chromatography as described in previous sections. Purified biotinylated proteins were homogeneous and devoid of any oligomeric forms (Example 4).
Figure 6 shows the properties of Albumin Binding Peptide (ABP)-ThioFab variants. ELISA analyses were carried out to test the binding ability of ABP-hu4D5Fabv8-wt, ABP- hu4D5Fabv8-Vl IOC and ABP-hu4D5Fabv8-A121C with rabbit albumin, streptavidin and HER2. Biotinylated ABP-ThioFabs are capable of binding to albumin and HER2 with similar affinity to that of wild type ABP-hu4D5Fabv8 as confirmed by ELISA (Figure 6) and BIAcore binding kinetics analysis (Table 3). An ELISA plate was coated with albumin, HER2 and SA as described. Binding of biotinylated ABP-ThioFabs to albumin, HER2 and SA was probed with anti-Fab HRP. Biotinylated ABP-ThioFabs were capable of binding to streptavidin compared to non biotinylated control ABP-hu4D5Fabv8-wt indicating that ABP- ThioFabs were conjugated with biotin maleimide like ThioFabs in a site specific manner as the same Cys mutants were used for both the variants (Figure 6). Table 3. BIAcore kinetic analysis for HER2 and rabbit albumin binding to biotinylated
ABP- hu4D5Fabv8 wild type and ThioFabs
Figure imgf000073_0001
ABP = albumin binding peptide
Alternatively, an albumin-binding peptide may be linked to the antibody by covalent attachment through a linker moiety. ENGINEERING OF ABP-THIOFABS WITH TWO FREE THIOL GROUPS PER FAB
The above results indicate that all four (L-V15C, L-Vl IOC, H-A88C and H-A121C) thioFab (cysteine engineered Fab antibodies) variants have reactive thiol groups that can be used for site specific conjugation with a label reagent, linker reagent, or zirconium-linker intermediate. L-V15C can be expressed and purified but with relatively low yields. However the expression and purification yields of L-Vl IOC, H-A88C and H-A121C variants were similar to that of hu4D5Fabv8. Therefore these mutants can be used for further analysis and recombined to get more than one thiol group per Fab. Towards this objective, one thiol group on the light and one on heavy chain were constructed to obtain two thiol groups per Fab molecule (L-Vl 10C/H-A88C and L-Vl 10C/H-A121C). These two double Cys variants were expressed in an E. coli expression system and purified. The homogeneity of purified biotinylated ABP-ThioFabs was found to be similar to that of single Cys variants.
The effects of engineering two reactive Cys residues per Fab was investigated (Figure 7). The presence of a second biotin was tested by probing the binding of biotinylated ABP- ThioFab to SA using streptavidin-HRP (Figure 7). For HER2/Fab analysis, an ELISA plate was coated with HER2 and probed with anti-Fab HRP. For SA/Fab analysis, an ELISA plate was coated with SA and probed with anti-Fab HRP. For SA/SA analysis, an ELISA plate was coated with SA and probed with SA-HRP. Figure 7. ELISA analyses for the interaction of biotinylated ABP-hu4D5Fabv8 cys variants with HER2, streptavidin (SA). HER2/Fab, SA/Fab and SA/SA indicate that their interactions were monitored by anti-Fab-HRP, SA- HRP, respectively. SA/Fab monitors the presence of single biotin per Fab and more than one biotin per Fab is monitored by SA/SA analysis. Binding of HER2 with double cys mutants is similar to that of single Cys variants (Figure 7). However the extent of biotinylation on double Cys mutants was higher compared to single Cys variants due to more than one free thiol group per Fab molecule (Figure 7).
ENGINEERING OF THIO IgG VARIANTS OF TRASTUZUMAB
Cysteine was introduced into the full-length monoclonal antibody, trastuzumab
(HERCEPTIN®, Genentech Inc.) at certain residues. The single cys mutants H-A88C, H- A121C and L-Vl IOC of trastuzumab, and double cys mutants VI 10C-A121C and VI 10C- A121C of trastuzumab were expressed in CHO (Chinese Hamster Ovary) cells by transient fermentation in media containing 1 mM cysteine. The A88C mutant heavy chain sequence (450 aa) is SEQ ID NO:6. The A121C mutant heavy chain sequence (450 aa) is SEQ ID NO:7. The VI IOC mutant light chain sequence (214 aa) is SEQ ID NO:8. EVQLVESGGGLVQPGGSLRLSCAASGF IKDTYIHWVRQAPGKGLEWVARIYPTNGYTRY ADSVKGRF I SAD SKNTAYLQMNSLRCEDTAVYYCSRWGGDGFYAMDYWGQGTLVTVSS ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGG PSVFLFPPKPKDTLMI SR PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN S YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEK I SKAKGQPREPQVYTLPPSREE MTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGK
SEQ ID N0:6
EVQLVESGGGLVQPGGSLRLSCAASGF IKDTYIHWVRQAPGKGLEWVARIYPTNGYTRY ADSVKGRFTI SADTSKNTAYLQMNSLRAEDTAVYYCSRWGGDGFYAMDYWGQGTLVTVSS CSTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGG PSVFLFPPKPKDTLMI SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSREE MTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGK
SEQ ID N0:7
DIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAPKLLIYSASFLYSGVPS RFSGSRSGTDFTLTI SSLQPEDFATYYCQQHYTTPPTFGQGTKVEIKRTCAAPSVFIFPP SDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLT LSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC SEQ ID NO: 8
According to one embodiment, the cysteine engineered thio-trastuzumab antibodies comprise one or more of the following variable region heavy chain sequences with a free cysteine amino acid (SEQ ID NOS: 9-16). Mutant Sequence SEQ ID NO
A40C WVRQCPGKGL SEQ ID NO 9
A88C NSLRCEDTAV SEQ ID NO 10
S119C LVTVCSASTKGPS SEQ ID NO 11
S120C LVTVSCASTKGPS SEQ ID NO 12
A121C LVTVSSCSTKGPS SEQ ID NO 13
S122C LVTVSSACTKGPS SEQ ID NO 14
A175C HTFPCVLQSSGLYS SEQ ID NO 15
S179C HTFPAVLQCSGLYS SEQ ID NO 16
According to another embodiment, the cysteine engineered thio-trastuzumab antibodies comprise one or more of the following variable region light chain sequences with a free cysteine amino acid (SEQ ID NOS: 17-27).
Figure imgf000076_0001
The resulting full-length, thio-trastuzumab IgG variants were assayed for thiol reactivity and HER2 binding activity. Figure 13 A shows a cartoon depiction of biotinylated antibody binding to immobilized HER2 and HRP labeled secondary antibody for absorbance detection. Figure 13B shows binding measurements to immobilized HER2 with detection of absorbance at 450 nm of (left to right): non-biotinylated wild type trastuzumab (Wt), biotin- maleimide conjugated thio-trastuzumab variants VI IOC (single cys), A121C (single cys), and VI 10C-A121C (double cys). Each thio IgG variant and trastuzumab was tested at 1, 10, and 100 ng. The measurements show that biotinylated anti-HER2 ThioMabs retain HER2 binding activity.
Figure 14A shows a cartoon depiction of a biotinylated antibody binding to immobilized HER2 with binding of biotin to anti-IgG-HRP for absorbance detection. Figure 14B shows binding measurements with detection of absorbance at 450 nm of biotin- maleimide conjugated thio-trastuzumab variants and non-biotinylated wild type trastuzumab in binding to streptavidin. From left to right: VI IOC (single cys), A121C (single cys), VI 10C/A121C (double cys), and trastuzumab. Each thio IgG trastuzumab variant and parent trastuzumab was tested at 1, 10, and 100 ng. The measurements show that the HER2
ThioMabs have high thiol reactivity.
Cysteine was introduced into the full-length 2H9 anti-EphB2R antibody at certain residues. The single cys mutant H-A121C of 2H9 was expressed in CHO (Chinese Hamster Ovary) cells by transient fermentation in media containing 1 mM cysteine. The A121C 2H9 mutant heavy chain sequence (450 aa) is SEQ ID NO:28.
EVQLVESGGGLVQPGGSLRLSCAASGYTFTSYWMHWVRQAPGKGLEWVGFINPSTGYTDY NQKFKDRF I SAD SKNTAYLQMNSLRAEDTAVYYCTRRPKI PRHANVFWGQGTLVTVSS CSTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGG PSVFLFPPKPKDTLMI SR PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN S YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEK I SKAKGQPREPQVYTLPPSREE MTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGK
SEQ ID NO:28
Cysteine engineered thio-2H9 antibodies comprise the following Fc constant region heavy chain sequences with a free cysteine amino acid (SEQ ID NOS: 29-38).
Figure imgf000077_0001
Figure 16 shows non-reducing (top) and reducing (bottom) denaturing SDS-PAGE (polyacrylamide gel electrophoresis) analysis of 2H9 ThioMab Fc variants (left to right, lanes 1-9): A339C; S337C; S324C; A287C; V284C; V282C; V279C; and V273C, with 2H9 wild type, after purification on immobilized Protein A. The lane on the right is a size marker ladder, indicating the intact proteins are about 150 kDa, heavy chain fragments about 50 kDa, and light chain fragments about 25 kDa. Figure 17A shows non-reducing (left) and reducing (right) denaturing polyacrylamide gel electrophoresis analysis of 2H9 ThioMab variants (left to right, lanes 1-4): L-V15C; S179C; S375C; S400C, after purification on immobilized Protein A. Figure 17B shows non-reducing (left) and reducing (+DTT) (right) denaturing polyacrylamide gel electrophoresis analysis of additional 2H9 and 3A5 ThioMab variants after purification on immobilized Protein A. The 2H9 ThioMab variants (in the Fab as well as Fc region) were expressed and purified as described. As seen in Figures 16, 17A and 17B, all the proteins are homogenous on SDS-PAGE followed by the reduction and oxidation procedure of Example 11 to prepare reactive ThioMabs for conjugation (Example 12).
Cysteine was introduced into the full-length 3A5 anti-MUC16 antibody at certain residues. The single cys mutant H-A121C of 3A5 was expressed in CHO (Chinese Hamster Ovary) cells by transient fermentation in media containing 1 mM cysteine. The A121C 3A5 mutant heavy chain sequence (446 aa) comprises SEQ ID NO:39.
DVQLQESGPGLVNPSQSLSLTCTVTGYS ITNDYAWNWIRQFPGNKLEWMGYINYSGYTTY NPSLKSRI S ITRD SKNQFFLHLNSV ED A YYCARWDGGLTYWGQGTLVTVSACS K GPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYS LSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVF LFPPKPKDTLMI SR PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS YR VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEK I SKAKGQPREPQVYTLPPSREEMTKN QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGK
SEQ ID NO:39
Cysteine engineered thio-3A5 anti-MUC16 antibodies comprise the following variable region heavy chain sequences with a free cysteine amino acid (SEQ ID NOS: 40-44). Mutant Sequence SEQ ID NO:
F45C NWIRQCPGNK SEQ ID NO:40
A90C LNSCTTEDTAT SEQ ID NO:41
A121C GQGTLVTVSACSTKGPSVFPL SEQ ID NO:42
A175C HTFPCVLQSSGLYS SEQ ID NO:43
V176C HTFPACLQSSGLYS SEQ ID NO:44
Cysteine engineered thio-3A5 anti-MUC16 antibodies comprise the following variable region light chain sequences with a free cysteine amino acid (SEQ ID NOS: 45-49).
Figure imgf000079_0001
THIOL REACTIVITY OF THIOMABS
The thiol reactivity of full length, IgG cysteine engineered antibodies (ThioMabs) was measured by biotinylation and streptavidin binding. A western blot assay was set up to screen the ThioMab that is specifically conjugated with biotin-maleimide. In this assay, the antibodies are analyzed on reducing SDS-PAGE and the presence of Biotin is specifically probed by incubating with streptavidin-HRP. As seen from figure 18, the streptavidin-HRP interaction is either observed in heavy chain or light chain depending on which engineered cys variant is being used and no interaction is seen with wild type, indicating that ThioMab variants specifically conjugated the biotin at engineered Cys residue. Figure 18 shows denaturing gel analysis of reduced, biotinylated Thio-IgG variants after capture on immobilized anti-IgG-HRP (top gel) and streptavidin-HRP (bottom gel). Lane 1 : 3A5 H- A121C. Lane 2: 3A5 L-V110C. Lane 3: 2H9 H-A121C. Lane 4: 2H9 L-V110C. Lane 5: anti-EphB2R 2H9 parent, wild type. Each mutant (lanes 1-4) was captured by anti-IgG with HRP detection (top) indicating that selectivity and affinity were retained. Capture by immobilized streptavidin with HRP detection (bottom) confirmed the location of biotin on heavy and light chains. The location of cysteine mutation on the cysteine engineered antibodies in lanes 1 and 3 is the heavy chain. The location of cysteine mutation on the cysteine engineered antibodies in lanes 2 and 4 is the light chain. The cysteine mutation site undergoes conjugation with the biotin-maleimide reagent. Analysis of the ThioMab cysteine engineered antibodies of Figure 18 and a 2H9 V15C variant by LC/MS gave quantitative indication of thiol reactivity (Table 5).
Table 5 LC/MS quantitation of biotinylation of ThioMabs - Thiol reactivity
Figure imgf000080_0001
Cysteine engineering was conducted in the constant domain, i.e. Fc region, of IgG antibodies. A variety of amino acid sites were converted to cysteine sites and the expressed mutants, i.e. cysteine engineered antibodies, were assessed for their thiol reactivity.
Biotinylated 2H9 ThioMab Fc variants were assessed for thiol reactivity by HRP quantitation by capture on immobilized streptavidin in an ELISA assay (Figure 19). An ELISA assay was established to rapidly screen the Cys residues with reactive Thiol groups. As depicted in
Figure 19 schematic diagram, the streptavidin-biotin interaction is monitored by probing with anti-IgG-HRP followed by measuring absorbance at 450 nm. These results confirmed 2H9- ThioFc variants V282C, A287C, A339C, S375C and S400C had moderate to highest Thiol reactivity. The extent of biotin conjugation of 2H9 ThioMab Fc variants was quantitated by LS/MS analysis as reported in Table 6. The LS/MS analysis confirmed that the A282C, S375C and S400C variants had 100% biotin conjugation and V284C and A339C had 50% conjugation, indicating the presence of a reactive cysteine thiol group. The other ThioFc variants, and the parent, wild type 2H9, had either very little biotinylation or none.
Table 6 LC/MS quantitation of biotinylation of 2H9 Fc ThioMabs
Figure imgf000081_0001
THIOL REACTIVITY OF THIO-4D5 FAB LIGHT CHAIN VARIANTS
Screening of a variety of cysteine engineered light chain variant Fabs of the antiErbB2 antibody 4D5 gave a number of variants with a thiol reactivity value of 0.6 and higher (Table 7), as measured by the PHESELECTOR assay of Figure 8. The thiol reactivity values of Table 7 are normalized to the heavy chain 4D5 ThioFab variant (HC-A121C) which is set at 100%, assuming complete biotinylation of HC-A121C variant, and represented as per cent values.
Table 7 Thiol reactivity per cent values of 4D5 ThioFab light chain variants
Figure imgf000081_0002
ZIRCONIUM-LABELLING REAGENTS
Exemplary bifunctional reagents based on desferoxamine B (Df) are employed for
89
the complexation of Zr to antibodies, including monoclonal antibodies (mAbs).
Desferoxamine B (Ν'- {5-[acetyl(hydroxy)amino]pentyl} -N-[5-( {4-[(5- aminopentyl)(hydroxy)amino]-4-oxobutanoyl} amino)pentyl]-N-hydroxysuccinamide (CAS Reg. No. 70-51-9); and also known as Deferoxamine, desferoxamine B, DFO-B, DFOA, DFB or desferal) is a bacterial siderophore produced by the actinobacter Streptomyces pilosus (Figure 20 top). Desferoxamine B has medical applications as a chelating agent used to remove excess iron from the body (Miller, Marvin J. "Syntheses and therapeutic potential of hydroxamic acid based siderophores and analogs" (1989) Chemical Reviews 89 (7): 1563-1579). The mesylate salt of DFO-B is commercially available. Initial experiments were conducted with N-^-acety^thioacetyl-Df (SATA-Df) and mAb decorated with maleimide groups, 4-[N-maleimidomethyl]cyclohexane-l-carboxylate (mAb-SMCC) attached to ε-amino group in lysine side chain (Meijs WE et al. "Zirconium-labeled monoclonal antibodies and their distribution in tumor-bearing nude mice" (1997) J. Nucl.
Med. 38: 112-8; Meijs WE et al. "A facile method for the labeling of proteins with zirconium isotopes" (1996) Nucl Med Biol. 23:439-48). However, the resulting thioether conjugates (mAb-SMCC-SATA-Df) were unstable in human serum at 37 °C (Verel I et al "89Zr immuno-PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies" (2003) J Nucl Med 44: 1271-81). Another exemplary amino reactive bifunctional chelators, based on Df modified with succinic anhydride (Sue), was used to convert the amino group of Df to carboxylic acid and subsequently activated as 2,3,5,6-tetrafluorophenyl ester (TFP). TFP-N-Suc-Df (Figure 20 center) was coupled to lysine ε-amino groups of mAb and the purified mAb-N-Suc-Df was chelated with 89 Zr. The resulting 89 Zr-mAb-N-Suc-Df was stable at physiological conditions and its biodistribution was compared to mAb-SMCC- SATA-Df in mice (Verel I et al "89Zr immuno-PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies" (2003) J Nucl Med. 44: 1271-81).
However, the preparation of TFP-N-Suc-Df requires protection of hydroxamide groups as
Fe(III) complex. The iron is removed by treatment with EDTA prior to chelation with 89 Zr, but the multistep method is tedious and possesses a danger of incomplete removal of the iron from the desferoxamine and/or incomplete removal of EDTA from the conjugation buffer which may negatively impact the 89 Zr-chelation yield. Therefore, a heterobifunctional amino reactive reagent, /?-isothiocyanatobenzyl-desfemoxaimine (Df-Bz-NCS) was recently developed for incorporation of Df into proteins via thiourea linkage, Figure 20 center (Perk LR et al. "Facile radiolabeling of monoclonal antibodies and other proteins with zirconium- 89 or gallium-68 for PET Imaging using p-isothiocyanatobenzyl-desferrioxamine" (2008) Nature Protocols, published online:DOI: 10.1038/nprot.2008.22; Perk LR et al. "p- Isothiocyanatobenzyl-desferrioxamine: a new bifunctional chelate for facile radiolabeling of monoclonal antibodies with zirconium-89 for immuno-PET imaging" (2009) European Journal Of Nuclear Medicine And Molecular Imaging). The antibody conjugates prepared using Df-Bz-NCS showed comparable stability and imaging properties to the reference
89 conjugates prepared using TFP-N-Suc-Df. Since reliable methods for coupling of Zr with antibodies through lysine ε-amino groups were developed the number of reported pre-clinical
89
and clinical immunoPET studies with Zr labeled antibodies has been rapidly increasing (Verel I, et al. "Long-lived positron emitters zirconium-89 and iodine- 124 for scouting of therapeutic radioimmunoconjugates with PET" (2003) Cancer Biother Radiopharm. 18:655- 61; Nagengast WB et al. "In vivo VEGF imaging with radiolabeled bevacizumab in a human ovarian tumor xenograft" (2007) J Nucl Med. 48 : 1313-9; Perk LR, et al. "(89)Zr as a PET surrogate radioisotope for scouting biodistribution of the therapeutic radiometals (90)Y and (177)Lu in tumor-bearing nude mice after coupling to the internalizing antibody cetuximab" (2005) J Nucl Med. 46: 1898-906; Perk LR et al. "Quantitative PET imaging of Met- expressing human cancer xenografts with (89)Zr-labelled monoclonal antibody DN30" (2008) European Journal Of Nuclear Medicine And Molecular Imaging 35: 1857-67; Perk LR et al. "Preparation and evaluation of (89)Zr-Zevalin for monitoring of (90)Y-Zevalin biodistribution with positron emission tomography" (2006) European Journal Of Nuclear Medicine And Molecular Imaging 33: 1337-45; Borjesson PK et al. "Performance of immuno- positron emission tomography with zirconium-89-labeled chimeric monoclonal antibody U36 in the detection of lymph node metastases in head and neck cancer patients" (2006) Clin Cancer Res. 12:2133-40; Aerts HJ et al. "Disparity between in vivo EGFR expression and 89Zr-labeled cetuximab uptake assessed with PET" (2009) J Nucl Med. 50: 123-31; Dijkers EC et al. "Development and Characterization of Clinical-Grade 89Zr-Trastuzumab for HER2/neu ImmunoPET Imaging" (2009) J Nucl Med 50(6):974-981).
Embodiments of zirconium complexes also include zirconium-binding (chelating) ligands such as DTP A (CAS Reg. No. 67-43-6), DOP A(l, 4,7,10-tetraazacyclododecane- 1,4,7,10-tetraacetic acid) (Liu, Shuang (2008) Advanced Drug Delivery Reviews 60(12): 1347-1370), cyclopentadienyl, and allyl groups (Erker, G. (1991) Pure and Applied Chemistry 63(6):797-806; Erker, G. (1990) Jour, of Organometallic Chem. 400(1-2): 185- 203), each of which are incorporated by reference herein.
Zirconium complexes (Z) and other radionuclides may be conjugated to antibodies (Ab), including monoclonal antibodies (mAbs) through ε-amino group in lysine side chain or through thiol group of cysteine. Since approximately 40 lysine side chains (Wang L et al "Structural characterization of the maytansinoid-monoclonal antibody immunoconjugate, huN901-DMl, by mass spectrometry" (2005) Protein Sci. 14:2436-46) or 8 cysteines (Hamblett KJ et al. "Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate" (2004) Clin Cancer Res. 10:7063-70) are available for conjugation in a mAb, both approaches provide heterogeneity with respect to mAb conjugate ratios and the site of conjugation. The modification of a lysine residue within the binding site may decrease the biological activity of the conjugate (Cai W et al. "PET imaging of colorectal cancer in xenograft-bearing mice by use of an 18F-labeled T84.66 anti-carcinoembryonic antigen diabody" (2007) J Nucl Med. 48:304-10; Shively JE. "18F labeling for immuno-PET: where speed and contrast meet" (2007) J Nucl Med. 48: 170-2; Tait JF et al "Improved detection of cell death in vivo with annexin V radiolabeled by site-specific methods" (2006) J Nucl Med. 47: 1546-53; Schellenberger EA et al "Optical imaging of apoptosis as a biomarker of tumor response to chemotherapy" (2003) Neoplasia (New York, N.Y) 5: 187-92), while the modification of cysteines in the hinge region provides a reduced plasma half-life (Hamblett KJ et al. "Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate" (2004) Clin Cancer Res. 10:7063-70). These limitations can be avoided by using mAbs engineered to contain cysteine selectively positioned for the purpose of site-specific conjugation with a biochemical assay, PHESELECTOR (US 7521541; Junutula JR et al. "Rapid identification of reactive cysteine residues for site-specific labeling of antibody-Fabs" J Immunol Methods 2008;332:41-52) for the rapid identification of preferred amino acids in an antibody for mutation to cysteine. The resulting antibody (THIOMAB) is subsequently chemoselectively and site-specifically conjugated to cytotoxic drugs without any loss of binding affinity or detrimental effect on the antibody scaffold stability (Junutula JR et al. "Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index" (2008) Nat Biotechnol. 26:925-32).
From an imaging point of view, a high target affinity and minimal non-specific uptake are required for optimal image quality. Accordingly, site-specifically radiolabeled cysteine- engineered antibodies (THlOMABs) could provide tracers with unaltered binding affinity and scaffold stability which may minimize the non-specific uptake of metabolites outside the target tissue. One aspect of the present invention is a method for site-specific radiolabeling of THIOMABs using novel Df-based thiol reactive bifunctional reagents
maleimidocyclohexyl-desferrioxamine (Df-Chx-Mal), bromoacetyl-desferrioxamine (Df-Bac) and iodoacetyl-desferrioxamine (Df-Iac) (Figure 20). Exemplary embodiments include where these reagents were site-specifically conjugated to trastuzumab THIOMAB (thio-
89
trastuzumab), chelated with Zr, and evaluated in vitro and in vivo.
One metastable isomer of zirconium is 89 Zr with a half-life of 78.4 hours with decay modes of beta (electron emission), positron (beta plus), and gamma radiation.
The radioisotope or other labels may be incorporated in the conjugate in known ways (Fraker et al (1978) Biochem. Biophys. Res. Commun. 80: 49-57; "Monoclonal Antibodies in Immunoscintigraphy" Chatal, CRC Press 1989). Carbon- 14-labeled 1-isothiocyanatobenzyl- 3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of a radionuclide to the antibody (WO 94/11026).
LINKERS
A "Linker" (L) is a bifunctional or multifunctional moiety which can be used to link one or more zirconium complex moieties (Z) and an antibody unit (Ab) to form antibody- zirconium conjugates (AZC) of Formula I. Antibody-zirconium conjugates (AZC) can be conveniently prepared using a Linker having reactive functionality for binding to zirconium and to the Antibody. A cysteine thiol of a cysteine engineered antibody (Ab) can form a bond with a functional group of a linker reagent, a zirconium label moiety or zirconium- linker intermediate.
In one aspect, a Linker has a reactive site which has an electrophilic group that is reactive to a nucleophilic cysteine present on an antibody. The cysteine thiol of the antibody is reactive with an electrophilic group on a Linker and forms a covalent bond to a Linker. Useful electrophilic groups include, but are not limited to, maleimide and haloacetamide groups.
Cysteine engineered antibodies may react with linker reagents or zirconium-linker intermediates, with electrophilic functional groups such as maleimide or a-halo carbonyl, according to the conjugation method at page 766 of Klussman, et al (2004), Bioconjugate Chemistry 15(4):765-773, and according to the protocols of Examples 17-19.
In another embodiment, the Z moieties are the same.
In yet another embodiment, the Z moieties are different. Exemplary embodiments of the Formula I antibody-zirconium conjugate (AZC) compounds include:
Figure imgf000086_0001
R is independently H or Ci-C6 alkyl; and n is 1 to 12.
In another embodiment, a Linker has a reactive functional group which has a nucleophilic group that is reactive to an electrophilic group present on an antibody. Useful electrophilic groups on an antibody include, but are not limited to, aldehyde and ketone carbonyl groups. The heteroatom of a nucleophilic group of a Linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit. Useful nucleophilic groups on a Linker include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide. The electrophilic group on an antibody provides a convenient site for attachment to a Linker. In another embodiment, the Linker may be substituted with groups which modulated solubility or reactivity. For example, a charged substituent such as sulfonate (-SO3 ) or ammonium, may increase water solubility of the reagent and facilitate the coupling reaction of the linker reagent with the antibody or the zirconium moiety, or facilitate the coupling reaction of Ab-L (antibody-linker intermediate) with Z, or Z-L (zirconium-linker
intermediate) with Ab, depending on the synthetic route employed to prepare the AZC.
The compounds of the invention expressly contemplate, but are not limited to, AZC prepared with linker reagents: BMPEO, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4- vinylsulfone)benzoate), and including bis-maleimide reagents: DTME, BMB, BMDB, BMH, BMOE, BM(PEO)2, and BM(PEO)43, which are commercially available from Pierce
Biotechnology, Inc., Customer Service Department, P.O. Box 117, Rockford, IL. 61105 U.S.A. Bis-maleimide reagents allow the attachment of the thiol group of a cysteine engineered antibody to a thiol-containing zirconium moiety, label, or linker intermediate, in a sequential or concurrent fashion. Other functional groups besides maleimide, which are reactive with a thiol group of a cysteine engineered antibody, zirconium moiety, label, or linker intermediate include iodoacetamide, bromoacetamide, vinyl pyridine, disulfide, pyridyl disulfide isocyanate, and isothiocyanate.
Figure imgf000087_0001
BM(PEO)2 BM(PEO)3
Useful linker reagents can also be obtained via other commercial sources, such as Molecular Biosciences Inc. (Boulder, CO), or synthesized in accordance with procedures described in Toki et al (2002) J. Org. Chem. 67: 1866-1872; Walker, M.A. (1995) J. Org. Chem. 60:5352-5355; Frisch et al (1996) Bioconjugate Chem. 7: 180-186; US 6214345; WO 02/088172; US 2003130189; US2003096743; WO 03/026577; WO 03/043583; and WO 04/032828. Exemplary linker reagents include:
Figure imgf000088_0001
where n is an integer ranging from 1-10 and T is -H or -S03Na;
Figure imgf000088_0002
where n is an integer ranging from 0-3;
Figure imgf000088_0003
In another embodiment, linker L may be a dendritic type linker for covalent attachment of more than one zirconium moiety through a branching, multifunctional linker moiety to an antibody (Sun et al (2002) Bioorganic & Medicinal Chemistry Letters 12:2213- 2215; Sun et al (2003) Bioorganic & Medicinal Chemistry 11 : 1761-1768). Dendritic linkers can increase the molar ratio of zirconium to antibody, i.e. loading of the AZC. Thus, where a cysteine engineered antibody bears only one reactive cysteine thiol group, a multitude of zirconium moieties may be attached through a dendritic linker.
The following exemplary embodiments of dendritic linker reagents allow up to nine nucleophilic zirconium moiety reagents to be conjugated by reaction with the chloroethyl nitrogen mustard functional groups:
Figure imgf000088_0004
CH2- )— N(CH2CH2CI)2
Figure imgf000089_0001
or
O II
CH2OCH2CH2CNHCH2CY3
Other embodiments of branched, dendritic linkers include those with self-immolative 2,6-bis(hydroxymethyl)-p-cresol and 2,4,6-tris(hydroxymethyl)-phenol dendrimer units (WO 2004/01993; Szalai et al (2003) J. Amer. Chem. Soc. 125: 15688-15689; Shamis et al (2004) J. Amer. Chem. Soc. 126: 1726-1731; Amir et al (2003) Angew. Chem. Int. Ed. 42:4494- 4499).
DESFERRIOXAMINE-LABELLED, CYSTEINE -ENGINEERED ANTIBODIES
An aspect of the invention is a desferrioxamine-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
Ab-(L-Df)p 11
wherein L-Df is selected from:
Figure imgf000090_0001
where the wavy line indicates the attachment to the antibody (Ab); and
p is 1 to 4.
PREPARATION OF ANTIBODY-ZIRCONIUM CONJUGATES
The antibody-zirconium conjugates (AZC) of Formula I may be prepared by several routes, employing organic chemistry reactions, conditions, and reagents known to those skilled in the art, including: (1) reaction of a cysteine group of a cysteine engineered antibody with a linker reagent, to form antibody-linker intermediate Ab-L, via a covalent bond, followed by reaction with an activated zirconium label moiety Z; and (2) reaction of a nucleophilic group of a zirconium moiety with a linker reagent, to form zirconium label- linker intermediate Z-L, via a covalent bond, followed by reaction with a cysteine group of a cysteine engineered antibody. Conjugation methods (1) and (2) may be employed with a variety of cysteine engineered antibodies, zirconium label moieties, and linkers to prepare the antibody-zirconium conjugates of Formula I.
Antibody cysteine thiol groups are nucleophilic and capable of reacting to form covalent bonds with electrophilic groups on linker reagents and zirconium-linker
intermediates including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides, such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups; and (iv) disulfides, including pyridyl disulfides, via sulfide exchange. Nucleophilic groups on a zirconium lable moiety include, but are not limited to: amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide groups capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents.
Under certain conditions, the cysteine engineered antibodies may be made reactive for conjugation with linker reagents by treatment with a reducing agent such as DTT (Cleland's reagent, dithiothreitol) or TCEP (tris(2-carboxyethyl)phosphine hydrochloride; Getz et al (1999) Anal. Biochem. Vol 273:73-80; Soltec Ventures, Beverly, MA). Full length, cysteine engineered monoclonal antibodies (ThioMabs) expressed in CHO cells were reduced with about a 50 fold excess of TCEP for 3 hrs at 37 °C to reduce disulfide bonds which may form between the newly introduced cysteine residues and the cysteine present in the culture media. The reduced ThioMab was diluted and loaded onto HiTrap S column in 10 mM sodium acetate, pH 5, and eluted with PBS containing 0.3M sodium chloride. Disulfide bonds were reestablished between cysteine residues present in the parent Mab with dilute (200 nM) aqueous copper sulfate (CuS04) at room temperature, overnight. Other oxidants, i.e.
oxidizing agents, and oxidizing conditions, which are known in the art may be used.
Ambient air oxidation is also effective. This mild, partial reoxidation step forms intrachain disulfides efficiently with high fidelity. An approximate 10 fold excess of zirconium-linker intermediate is added, mixed, and let stand for about an hour at room temperature to effect conjugation and form the ThioMab antibody-zirconium conjugate. The conjugation mixture is gel filtered and loaded and eluted through a HiTrap S column to remove excess zirconium- linker intermediate and other impurities.
Figure 15 shows the general process to prepare a cysteine engineered antibody expressed from cell culture for conjugation. Cysteine adducts, presumably along with various interchain disulfide bonds, are reductively cleaved to give a reduced form of the antibody. The interchain disulfide bonds between paired cysteine residues are reformed under partial oxidation conditions, such as exposure to ambient oxygen. The newly introduced, engineered, and unpaired cysteine residues remain available for reaction with linker reagents or zirconium-linker intermediates to form the antibody conjugates of the invention. The ThioMabs expressed in mammalian cell lines result in externally conjugated Cys adduct to an engineered Cys through -S-S- bond formation. Hence the purified
ThioMabs have to be treated with reduction and oxidation procedures as described in Example 11 to produce reactive ThioMabs. These ThioMabs are used to conjugate with maleimide containing radiolabels, cytotoxic drugs, fluorophores, and other labels.
PREPARATION AND ANALYSIS OF 89Zr-Df-TRASTUZUMAB CONJUGATES
The protected active ester TFP-N-SucDf-Fe was prepared according to the previously described procedure (Verel I et al "89Zr Immuno-PET: Comprehensive Procedures For The Production Of 89Zr-Labeled Monoclonal Antibodies" (2003) J Nucl Med 44: 1271-81) and conjugated to trastuzumab using a 5-fold molar excess of TFP-N-SucDf-Fe to yield N-SucDf- trastuzumab with an average of 1.6 molecules of desferoxamine (Table 8). Df-Bz-SCN- trastuzumab was obtained by coupling an 8-fold molar excess of Df-Bz-SCN at pH 8.5 (Perk LR, et al. "Facile radiolabeling of monoclonal antibodies and other proteins with zirconium- 89 or gallium-68 for PET Imaging using p-isothiocyanatobenzyl-desferrioxamine" (2008) Nature Protocols; published online:DOI: 10.1038/nprot.2008.22). The reaction provided Df- Bz-SCN-trastuzumab decorated in average with 2.4 molecules of desferoxamine (Table 1).
The novel maleimide based thiol reactive bifunctional linker Df-Chx-Mal was prepared from equimolar amounts of desferoxamine mesylate and SMCC (Figure 21,
Example 13). The reaction was complete within 30 min at room temperature and the product was isolated by precipitation upon addition of water in 45% yield and more than 95% purity. The reaction of an 8.5-fold molar excess of Df-Chx-Mal with freshly prepared thio- trastuzumab (Figure 21 Example 17) provided Df-Chx-Mal-thio-trastuzumab conjugate with exactly 2 molecules of desferoxamine in 1 h (Table 1, Figure 21). Bromoacetyl
desferoxamine (BDf-Bac) was prepared by the reaction of equimolar amounts of desferoxamine mesylate and bromoacetyl bromide at 0°C (Example 14). The product was obtained in 14% yield after HPLC purification. The alkylation of freshly prepared thio- trastuzumab (Figure 21, Example 16) with a 12-fold molar excess of Df-Bac provided the conjugate (Df-Ac-thio-trastuzumab) with 1.8 molecules of Df per antibody within 5 h (Table 8, Figure 21, Example 18). The low reactivity of bromide prompted us to explore the more reactive iodoacetyl derivative (Df-Iac). Df-Iac was prepared in 53% yield by the reaction of desferoxamine mesylate with a slight excess of N-hydroxysuccinimidyl iodoacetate (Figure 21, Example 15). The product was obtained in more than 95% purity by precipitation from the reaction mixture. The subsequent reaction of an 11 -fold excess of Df-Iac provided Df-Ac- thio-trastuzumab decorated with 1.8 molecules of Df within 2 h (Table 1, Figure 21, Example 19). Based on our experience, Df-Chx-Mal is the preferred reagent of the three compounds investigated. Notably, the reaction of Df-Chx-Mal was complete at a mild pH within 1 h as oppose to higher pH and longer reaction time required for the alkylation of thiol groups with Df-Bac and Df-Iac. Additionally, the lower reactivity of haloacetamides might have caused the incomplete loading of both available cysteines of thio-trastuzumab
Table 8. Reaction conditions and yields of Df-linker-trastuzumab conjugates prepared using various reagents
Reagent Temp. pH Reagent Reaction Loading
[°C] Excess Time [hr] [Df/Mab]
Fe-Df-N-Suc-TFP 37 8.5 5 1.5 1.6
Df-Bz-SCN 37 9 8 0.5 2.4
Df-Chx-Mal 25 7.5 8.5 1 2.0
Df-Bac 25 9 12 5 1.8
Df-Iac 25 9 11 2 1.8
The Zr was chelated as 89-zirconium oxalatewith all four variants of Df- trastuzumab using previously described experimental procedure (Verel I et al "89Zr immuno- PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies" (2003) J Nucl Med. 44: 1271-81). The radiolabeled proteins were purified on a desalting column and the final solution was concentrated to the required volume by membrane
89
filtration. The yield, purity, and final specific activity of the Zr conjugates are summarized in Table 9. In general, the chelation yield was over 80% with the exception of the Df-N-Suc linker obtained in lower yield presumably due to the lower amount of Df per antibody molecule and/or incomplete removal of Fe(III) used to protect the chelator during activation and conjugation. After purification of the Df-trastuzumab variants using desalting column, the product purity was over 90% with a small amount (1-6%) of high molecular weight
89 aggregates detected in each sample. Df-Chx-Mal-thio-trastuzumab provided the Zr complex in 99% purity (Table 9) as opposed to Df-Ac conjugate which was contaminated with approximately 8% of a low molecular weight impurity and 2% of high molecular weight aggregates. The contaminant resisted removal using NAP- 10 column but the removal was possible using repeated buffer exchange on Amicon filter.
89
Table 9. Yields, specific activity and purity of radiolabeled Zr-Df-linker-trastuzumab
Linker Radiochemical Specific activity Purity [%]
yield [%] [mCi/mg]
N-Suc 60 2.2 98
Bz-SCN 81 2.9 94
Chx-Mal 87 3.4 99 Ac 84 3.2 90
BIOLOGICAL ACTIVITY OF Zr-Df-TRASTUZUMAB CONJUGATES
The biological activities of newly prepared site-specific Df-/m er-thio-trastuzumab conjugates were determined using binding assay to BT474 breast cancer cell line by
Scatchard analysis. The obtained KD values were compared to non-modified trastuzumab (0.91 ± 0.20 nM). The KD for the thio-trastuzumab conjugate containing Chx-Mal linker was 0.93 ± 0.15 nM and the values for the conjugates containing Ac linker were 1.22 ± 0.22 nM for conjugate prepared using Df-Bac and 0.87 ± 0.15 nM prepared using Df-Iac. The results of the biological activity analyses indicate that the modification of thio-trastuzumab did not affect the binding affinity of the antibody to HER2.
IN VITRO SERUM STABILITY
The previously reported Df-antibody conjugate with N-Suc and Bz-SCN linkers containing amide or thiourea linkages were stable in vitro over a 6 day period in serum at 37 °C (Verel I et al "89Zr immuno-PET: comprehensive procedures for the production of 89Zr- labeled monoclonal antibodies" J Nucl Med 2003;44: 1271-81; Perk LR et al, (2009)
European Journal Of Nuclear Medicine And Molecular Imaging 35(10): 1857-1867). The
89
stability of Zr-thio-trastuzumab conjugates with Chx-Mal and Ac linkers was determined in mouse serum at 37 °C. A significant loss of antibody bound 89Zr was not observed within a 5 day period. Both thio conjugates were stable with an average loss of antibody bound 89Zr 1.8% per day for 89Zr-Df-Chx-Mal-thio-trastuzumab and 1.4% per day for 89Zr-Df-Ac-thio- trastuzumab (Figure 24). Slow formation of high molecular weight species, presumably aggregates, was observed for both Df-Chx-Mal and Df-Ac linkers.
IN VIVO MICROPET IMAGING
Twenty animals (5 animals per group) bearing subcutaneous BT474M1 xenografts
3 89
(size -200 mm ) were injected intravenously with Zr-trastuzumab. The amount of antibody injected per animal was 1.4 ± 0.29 mg/kg. The maximum intensity projection images of
89
representative animals (at 96 h p.i.) are shown in Fig. 3. The Zr-trastuzumab uptake in selected tissues is summarized in Fig 4. The images at 1 h (not shown) were dominated by the high blood pool uptake with the exception of Df-Bac where rapid hepatobiliary excretion of the lipophilic impurity resulted in an elevated uptake in intestine. The impurity was totally cleared within the first 24 h and elevated small and large intestine uptake was not detected at 24 h or later time after tracer injection. Although the tissue uptake of Df-Ac conjugate was resultantly slightly (-8%) lower, the tumor to blood ratios (Table 10) were not affected by the loss of injected radioactivity. The images at 96 h were dominated by the high tumor uptake
89
with minor differences observed among the four different Zr-trastuzumab variants (Figure 25). The tumor uptake was identical for each tracer reaching maximum values at 24 h post injection and maximum tumor-to-blood ratios at 144 h due to blood clearance (Table 10). The
89
thiol based conjugate Zr-Df-Chx-Mal-thio-trastuzumab exhibited elevated bone uptake (P < 0.05) compared to the amine based conjugates (Df-Bz-SCN and Df-N-Suc) at 96 and 144 h p.i. The bone uptake of Df-Ac-thio-trastuzumab was not significantly elevated (P = 0.20) compared to the Df-Bz-SCN and Df-N-Suc linkers but may become significant when corrected for the 8% loss of radioactivity during the first 24 h. The kidney uptake of each
89
tracer was low (Figure 26) as expected for antibody based tracers but Zr-Df-Chx-Mal-thio- trastuzumab was slightly higher compared to other linkers at 24, 96 and 144 h (P < 0.05).
Table 10. Average tumor to blood ratios at 24, 96 and 144 h post injection
Linker 24 h 96 h 144 h
N-Suc 1.8 3.8 6.0
Bz-SCN 2.0 4.0 5.7
Chx-Mal 2.0 4.9 7.1
Ac (Bac) 2.0 4.7 6.1
BT474 (3+ expression level of HER2) xenografts exhibited lower absolute uptake of the tracer (15 %ID/g) than measured previously by Dijkers et al in SKOV3 (3+ expression level of HER2) 33.4 ± 7.7%ID/g (Dijkers EC, et al. "Development and Characterization of Clinical-Grade 89Zr-Trastuzumab for HER2/neu ImmunoPET Imaging" (2009) J Nucl Med 50(6):974-981). However, the tumor to blood ratio of 5.7-7.1 (Table 10) is comparable to the value obtained with SKOV3 (tumor to blood of 7.6). The difference in tumor uptake may be attributed to the tumor model and total dose of trastuzumab. A material with higher specific activity was used hence so significantly less antibody was injected (35 μg, 1.4 mg/kg) compared to the Dijkers et al study with SKOV3 (100 μg, 4 mg/kg). The difference in
89
specific activity may have also contributed to lower bone uptake of free Zr in the experiment herein of 2-3 %ID/g compared to SKOV3 model (5-10 %ID/g). Unfortunately, no teaching regarding the elevated bone uptake is provided by Dijkers et al. Zirconium is known to bind plasma proteins (Mealey J, Jr. "Turn-over of carrier-free zirconium-89 in man" (1957) Nature 179:673-4 and is later deposited in mineral bone (Fletcher CR. "The radiological hazards of zirconium-95 and niobium-95" (1969) Health Phys. 16:209-20; Shiraishi Y and Ichikawa R. "Absorption and retention of 144 Ce and 95 Zr- 95 Nb in newborn, juvenile and adult rats" (1972) Health Phys. 22:373-8). Since the injected material
89 89 did not contain free Zr, the bone uptake may originate from the breakdown of the Zr-
89
antibody or from Zr non-specifically associated with antibody which could then trans-
89
chelate to plasma proteins compared to Zr bound to Df.
Three thiol specific reagents are exemplified herein for the chemoselective conjugation of desferrioxamine (Df) to monoclonal antibodies through the thiol group of cysteine of cysteine-engineered antibodies. The thiol-specific Df-reagents were obtained by the acylation of the amino group of desferrioxamine B in 14% (Df-Bac), 53% (Df-Iac) and 45% (Df-Chx-Mal) yields and conjugated to thio-trastuzumab resulting in site-specific modification on both engineered cysteines within 1-5 h. The binding activities of site-specific thio-trastuzumab conjugates to HER2 were identical to the activity of non-modified trastuzumab. The Df-modified thio-trastuzumabs (Df-Ac-thio-trastuzumab and Df-Chx-Mal-
89
thio-trastuzumab) were chelated with Zr (Figure 22) in yields exceeding 80% within 1 h comparable to lysine conjugates prepared using previously described Df-Bz-SCN and Df-N-
89 89
Sue linkers. Both Zr-Df-Ac-thio-trastuzumab and Zr-Df-Chx-Mal-thio-trastuzumab showed comparable stability in mouse serum. Both compounds also showed PET imaging capabilities in BT474M1 breast cancer model comparable to lysine conjugates reaching 10- 15 %ID/g of tumor uptake with a tumor to blood ratio in the range 6.1-7.1. Overall, the novel reagents are readily available, demonstrated good reactivity with thiol groups of the protein,
89 89
and exhibited very good chelation properties with Zr. The Zr-labeled antibodies were stable in serum and showed excellent PET imaging properties. Df-Chx-Mal is a useful reagent for conjugation of Df to antibodies through cysteine side chain and showed several advantages over Df-Bac and Df-Iac. First, moderate pH 7.5 was required for complete conjugation of Df-Chx-Mal within 1 h as compared to pH 9 and 2 or 5 h required for Df-Bac
89
and Df-Iac. Additionally, the site specifically Zr labeled engineered THIOMAB conjugates can be used similarly as 18 F labeled THIOFAB conjugates (Gill HS, et al. "A modular platform for the rapid site-specific radiolabeling of proteins with 18F exemplified by quantitative positron emission tomography of human epidermal growth factor receptor 2" (2009) Jour, of Med. Chem. 52:5816-25) as valuable tools for PET imaging applications in biomedical research.
ADMINISTRATION OF ANTIBODY-ZIRCONIUM CONJUGATES
The antibody-zirconium conjugates (AZC) of the invention may be administered by any route appropriate to the condition to be treated. The AZC will typically be administered parenterally, i.e. infusion, subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural.
PHARMACEUTICAL FORMULATIONS
Pharmaceutical formulations of diagnostic antibody-zirconium conjugates (AZC) of the invention are typically prepared for parenteral administration, i.e. bolus, intravenous, intratumor injection with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form. An antibody-zirconium conjugate (AZC) having the desired degree of purity is optionally mixed with pharmaceutically acceptable diluents, carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation or an aqueous solution.
Acceptable diluents, carriers, excipients, and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). For example, lyophilized anti-ErbB2 antibody formulations are described in WO 97/04801, expressly incorporated herein by reference.
The active pharmaceutical ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in
macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980). Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semi permeable matrices of solid hydrophobic polymers containing the AZC, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or polyvinyl alcohol)), polylactides (US 3773919), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid.
The formulations to be used for in vivo administration must be sterile, which is readily accomplished by filtration through sterile filtration membranes.
The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington's Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g.,
polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin. The pharmaceutical compositions of AZC may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may likewise be used in the preparation of injectables.
The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 μg of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
Although oral administration of protein therapeutics are disfavored due to hydrolysis or denaturation in the gut, formulations of AZC suitable for oral administration may be prepared as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the AZC.
The formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient. The invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore. Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
LABELLED ANTIBODY IMAGING METHODS
In another embodiment of the invention, cysteine engineered antibodies may be labelled through the cysteine thiol with radionuclides, fluorescent dyes, bioluminescence- triggering substrate moieties, chemiluminescence-triggering substrate moieties, enzymes, and other detection labels for imaging experiments with diagnostic, pharmacodynamic, and therapeutic applications. Generally, the labelled cysteine engineered antibody, i.e.
"biomarker" or "probe", is administered by injection, perfusion, or oral ingestion to a living organism, e.g. human, rodent, or other small animal, a perfused organ, or tissue sample. The distribution of the probe is detected over a time course and represented by an image.
EXAMPLES
Preparation of Solvents and chemicals were purchased from Aldrich (Milwaukee, WI) unless stated otherwise. The following reversed-phase HPLC systems were used to analyze and purify the products. System A: Phenomenex BioSep-SEC-S 3000 (300 x 4.60 mm, 5 μπι) 50 mM PBS 0.5 ml/min equipped with UV absorbance and radioactivity detector (PMT);
System B Altima C-18 (100 χ 22.0 mm, 5 μπι) 0.05% TFA + 10-50% acetonitrile, 0-30 min, 24 mL/min, equipped with UV detector. Mass spectrometry analysis of low molecular weight products was performed on a PE Sciex API 150EX LCMS system equipped with an Onyx Monolithic C18 column. LCMS analysis of proteins was performed on a TSQ Quantum Triple quadrupole mass spectrometer with extended mass range (Thermo Electron, Thermo Fisher Scientific Inc., USA). The protein samples for LCMS analysis were reduced by treatment with 20 mM dithiothreitol (DTT) at 37°C for 1 h to separate heavy and light chains. Samples were chromato graphed on a PRLP-S 1000 A microbore column (50 mm x 2.1 mm, Polymer Laboratories, Varian Inc., USA) heated to 75°C. A linear gradient from 30-40% B (solvent A, 0.05% TFA in water; solvent B, 0.04% TFA in acetonitrile) was used and the eluant was directly ionized using the electrospray source. Data were collected by the Xcalibur data system and deconvolution was performed using ProMass software (Novatia, Monmouth Junction, NJ). NMR spectra were recorded on Bruker Avance II 400 spectrometer at 298K and the chemical shifts are reported relative to TMS. Protein concentrations were measured at
89
280 nm using Eppendorf BioPhotometer (Westbury, NY). Zr was obtained from Memorial
89
Sloan-Kettering Cancer Center (New York, NY) as Zr(IV) oxalate in 1M oxalic acid solution with specific activity 470-1 195 Ci/mmol (Holland JP, et al (2009) "Standardized methods for the production of high specific-activity zirconium-89" Nucl Med Biol. 36:729- 39). Heterobifunctional linker succinimidyl 4-[N-maleimidomethyl]cyclohexane-l- carboxylate (SMCC) was purchased from Pierce (Rockford, IL) and N-hydroxysuccinimidyl iodoacetate was obtained from Indofine Chemical Company (Hillsborough, NJ). NAP- 10 columns were obtained from (GE Healthcare, USA) and Amicon Ultra-4 centrifugal filters (10,000 MWCO) from Millipore (Billerica, MA). Df-Bz-SCN was purchased Macrocyclics (Dallas, TX).
Example 1 Preparation of Biotinylated ThioFab Phage
ThioFab-phage (5 x 1012 phage particles) were reacted with 150 fold excess of biotin- PEO-maleimide ((+)-biotinyl-3-maleimidopropionamidyl-3,6-dioxaoctainediamine, Oda et al (2001) Nature Biotechnology 19:379-382, Pierce Biotechnology, Inc.) for 3 hours at room temperature. Excess biotin-PEO-maleimide was removed from biotin-conjugated phage by repeated PEG precipitations (3-4 times). Other commercially available biotinylation reagents with electrophilic groups which are reactive with cysteine thiol groups may be used, including Biotin-BMCC, PEO-Iodoacetyl Biotin, Iodoacetyl-LC-Biotin, and Biotin-HPDP (Pierce Biotechnology, Inc.), and N°-(3- maleimidylpropionyl)biocytin (MPB, Molecular Probes, Eugene, OR). Other commercial sources for biotinylation, bifunctional and multifunctional linker reagents include Molecular Probes, Eugene, OR, and Sigma, St. Louis, MO.
Figure imgf000101_0001
Biotin-PEO-maleimide Example 2 PHESELECTOR Assay
Bovine serum albumin (BSA), erbB2 extracellular domain (HER2) and streptavidin (100 μΐ of 2 μ§/ι 1) were separately coated on Maxisorp 96 well plates. After blocking with 0.5% Tween-20 (in PBS), biotinylated and non-biotinylated hu4D5Fabv8-ThioFab-Phage (2xl010 phage particles) were incubated for 1 hour at room temperature followed by incubation with horseradish peroxidase (HRP) labeled secondary antibody (anti-M13 phage coat protein, pVIII protein antibody). Figure 8 illustrates the PHESELECTOR Assay by a schematic representation depicting the binding of Fab or ThioFab to HER2 (top) and biotinylated ThioFab to streptavidin (bottom).
Standard HRP reaction was carried out and the absorbance was measured at 450 nm.
Thiol reactivity was measured by calculating the ratio between OD450 for streptavidin/OD45o for HER2. A thiol reactivity value of 1 indicates complete biotinylation of the cysteine thiol. In the case of Fab protein binding measurements, hu4D5Fabv8 (2-20 ng) was used followed by incubation with HRP labeled goat polyclonal anti-Fab antibodies. Example 3a Expression and Purification of ThioFabs
ThioFabs were expressed upon induction in 34B8, a non-suppressor E. coli strain (Baca et al (1997) Journal Biological Chemistry 272(16): 10678-84). The harvested cell pellet was resuspended in PBS (phosphate buffered saline), total cell lysis was performed by passing through a microfluidizer and the ThioFabs were purified by affinity chromatography with protein G SEPHAROSE™ (Amersham).
ThioFabs L-V15C, L-Vl IOC, H-A88C, and H-A121C were expressed and purified by Protein-G SEPHAROSE™ column chromatography. Oligomeric-Fab was present in fractions 26 to 30, and most of the monomeric form was in fractions 31-34. Fractions consisting of the monomeric form were pooled and analyzed by SDS-PAGE along with wild type hu4D5Fabv8and analyzed on SDS-PAGE gel in reducing (with DTT or BME) and non- reducing (without DTT or BME) conditions. Gel filtration fractions of A121C-ThioFab were analyzed on non-reducing SDS-PAGE.
ThioFabs were conjugated with biotin-PEO-maleimide as described above and the biotinylated-ThioFabs were further purified by Superdex-200™ (Amersham) gel filtration chromatography, which eliminated the free biotin-PEO-maleimide and the oligomeric fraction of ThioFabs. Wild type hu4D5Fabv8 and hu4D5Fabv8 A121C-ThioFab (0.5 mg in quantity) were each and separately incubated with 100 fold molar excess of biotin-PEO- maleimide for 3 hours at room temperature and loaded onto a Superdex-200 gel filtration column to separate free biotin as well as oligomeric Fabs from the monomeric form.
Example 3b Analysis of ThioFabs
Enzymatic digest fragments of biotinylated hu4D5Fabv8 (A121C) ThioFab and wild type hu4D5Fabv8 were analyzed by liquid chromatography electrospray ionization mass spectroscopy (LS-ESI-MS) The difference between the 48294.5 primary mass of biotinylated hu4D5Fabv8 (A121C) and the 47737.0 primary mass of wild type hu4D5Fabv8 was 557.5 mass units. This fragment indicates the presence of a single biotin-PEO-maleimide moiety (C23H36N5O7S2). Table 4 shows assignment of the fragmentation values which confirms the sequence.
Table 4. LC-ESI-Mass spec analysis of biotinylated hu4D5Fabv8 ThioFab A121C after tryptic digestion
Figure imgf000104_0001
Before and after Superdex-200 gel filtration, SDS-PAGE gel analyses, with and without reduction by DTT or BME, of biotinylated ABP- hu4D5Fabv8-A121C, biotinylated ABP- hu4D5Fabv8-Vl IOC, biotinylated double Cys ABP-hu4D5Fabv8-(Vl 10C-A88C), and biotinylated double Cys ABP-hu4D5Fabv8-(Vl 10C-A121C) were conducted.
Mass spectroscopy analysis (MS/MS) of of hu4D5Fabv8-(Vl 10C)-BMPEO-DM1 (after Superdex-200 gel filtration purification): Fab+1 51607.5, Fab 50515.5. This data shows 91.2% conjugation. MS/MS analysis of hu4D5Fabv8-(Vl 10C)-BMPEO-DM1
(reduced): LC 23447.2, LC+1 24537.3, HC (Fab) 27072.5. This data shows that all DM1 conjugation is on the light chain of the Fab. Example 11 Reduction/Oxidation of ThioMabs for Conjugation
Full length, cysteine engineered monoclonal antibodies (ThioMabs) expressed in CHO cells were reduced with about a 50 fold excess of TCEP (tris(2-carboxyethyl)phosphine hydrochloride; Getz et al (1999) Anal. Biochem. Vol 273:73-80; Soltec Ventures, Beverly, MA) for 3 hrs at 37 °C. The reduced ThioMab (Figure 15) was diluted and loaded onto a HiTrap S column in 10 mM sodium acetate, pH 5, and eluted with PBS containing 0.3M sodium chloride. The eluted reduced ThioMab was treated with 200 nM aqueous copper sulfate (CuS04) at room temperature, overnight. Ambient air oxidation was also effective.
Example 13 N-[4-(N-maleimidomethyl)cyclohexane-l-carboxyl]desferrioxamine (Df-Chx- Mai)
Figure imgf000105_0001
4-[N-Maleimidomethyl]cyclohexane-l-carboxylate (SMCC, 40 mg, 0.12 mmol), desferoxamine mesylate (78 mg, 0.12 mmol) and N,N-diisopropylethylamine (22 μί, 0.13 mmol) were dissolved in a mixture of DMF (2.0 mL) and 0.2 mL water (Figure 21). The resulting turbid solution was stirred at room temperature for 30 min. Water (8 mL) was added and the precipitated product was isolated by filtration, washed with water, and dried at reduced pressure to yield 42 mg (45%) of N-[4-(N-maleimidomethyl)cyclohexane-l- carboxyl]desferrioxamine (Df-Chx-Mal) as a white solid (Figure 20 bottom). lH NMR (400 MHz, de-DMSO) δ 0.87-0.90 (m, 2H), 1.20-1.26 (m, 8H), 1.35-1.41 (m, 6H), 1.45-1.55 (m, 8H), 1.60-1.70 (m, 4H), 1.97 (s, 3H, acetyl), 2.26-2.29 (m, 4H), 2.56-2.60 (m, 4H), 2.95-3.05 (m, 6H), 3.24 (d, J = 7.0 Hz, 2H), 3.44-3.48 (m, 6H), 7.00 (s, 2H, maleimide), 7.62 (t, J = 5.4 Hz, IH, amide), 7.75 (m, 2H, amide), 9.59 (s, 2H, hydroxyl), 9.64 (s, IH, hydroxyl). MS ESI (m/z): [M+H]+ calculated for C37H62N7O11 780.44; found 780.6. Example 14 N-bromoacetyldesferrioxamine (Df-Bac)
Figure imgf000106_0001
A solution of bromoacetyl bromide (27 μί, 0.30 mmol) in DMF (1 mL) was added dropwise in 5 min into a cooled (0°C) mixture of desferoxamine mesylate (200 mg, 0.30 mmol) and N,N-diisopropylethylamine (106 μί, 0.60 mmol) in DMF (5 mL) after which the reaction mixture was stirred at 0°C for 4 h (Figure 21). Water (10 mL) was added and the product was isolated using HPLC (System B, retention time 7.5 min) to yield 29 mg (14%) of N-bromoacetyldesferrioxamine (Df-Bac) as a white solid (Figure 20 bottom). lH NMR (400 MHz, dg-DMSO) δ 1.18-1.26 (m, 6H), 1.35-1.42 (m, 6H), 1.45-1.55 (m, 6H), 1.97 (s, 3H, acetyl), 2.24-2.30 (m, 4H), 2.54-2.59 (m, 4H), 2.96-3.07 (m, 6H), 3.44-3.47 (m, 6H), 3.82 (s, 2H, bromoacetyl), 7.74 (m, 2H, amide), 8.21 (t, 1H, amide), 9.59 (s, 2H, hydroxyl), 9.63 (s, 1H, hydroxyl). MS ESI (m/z): [M+H]+ calculated for C27H5oBrN609 681.27, 683.27; found 681.1, 683.0.
Example 15 N-iodoacetyldesferrioxamine (Df-lac)
Figure imgf000106_0002
Desferoxamine mesylate (200 mg, 0.30 mmol) and N,N-diisopropylethylamine (53 μί, 0.30 mmol) were mixed in DMF (4 mL) and water (0.4 mL). N-hydroxysuccinimidyl iodoacetate (93 mg, 0.33 mmol) was added and the resulting mixture was stirred at room temperature for 1 h (Figure 21). Water (8 mL) was added and the precipitated product was separated, washed with water, and dried at reduced pressure to yield 115 mg (53%) of N- iodoacetyldesferrioxamine (Df-Iac) as a white solid (Figure 20 bottom). lH NMR (400 MHz, dg-DMSO) δ 1.20-1.25 (m, 6H), 1.35-1.42 (m, 6H), 1.47-1.54 (m, 6H), 1.97 (s, 3H, acetyl), 2.25-2.29 (m, 4H), 2.56-2.59 (m, 4H), 2.98-3.03 (m, 6H), 3.45 (m, 6H), 3.61 (s, 2H, iodoacetyl), 7.75 (m, 2H, amide), 8.17 (t, 1H, amide), 9.57 (s, 2H, hydroxyl), 9.61 (s, 1H, hydroxyl). MS ESI (m/z): [M+H]+ calculated for C27H5oIN609 729.26; found 729.1.
Example 16 Thio-trastuzumab
The construction, expression, and purification of THIOMAB with Cys substitution at Ala114 (Kabat numbering) in heavy chain was described previously (Junutula JR, et al "Site- specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index" (2008) Nat Biotechnol 26:925-32). The isolated thio-trastuzumab was prepared for conjugation by a reduction and re-oxidation procedure to remove disulfide adducts bound to Cys114. First, the protein was reduced for 24 h by treatment with a 40-fold molar excess of DTT and 2 mM EDTA in 88 mM Tris buffer pH 7.5. To remove DTT prior to re-oxidation, the thio-trastuzumab solution was adjusted to pH 5 by the addition of 10 mM sodium succinate buffer. The solution was then loaded on an ion exchange column (HiTrap SP FF, GE Healthcare) that had been sterilized and equilibrated with 10 mM sodium succinate buffer pH 5. The column was washed with 10 mM sodium succinate buffer (10 mL) and the thio- trastuzumab was then eluted with 3 mL of 50 mM Tris, 150 mM NaCl buffer with pH 7.5. thio-trastuzumab re-oxidization was achieved by treatment with a 25-fold molar excess of dehydroascobric acid (100 mM in N,N-dimethylacetamide (DMA)) in 75 mM Tris, 150 mM NaCl pH 7.5 buffer at 25°C for 3.5 h. After re-oxidation, the thio-trastuzumab was conjugated to desferoxamine without further purification. MS ESI (m/z): found light chain 23440.0, heavy chain 50627.3.
Example 17 Df-Chx-Mal-thio-trastuzumab
Figure imgf000107_0001
Df-Chx-Mal-ThioMab The 2 mM stock solution of the bifunctional chelator was prepared by dissolving Df- Chx-Mal (1.5 mg, 2 μπιοΐ) in a 1 : 1 mixture (1 mL) of DMF and DMA by heating to 44°C for 30 min, the stock solution was then aliquoted and stored at -80 °C (Figure 21). An aliquot of the stock solution (220 μί, 0.440 μπιοΐ) was then added to the solution of thio-trastuzumab (7.5 mg, 52 nmol) in 50 mM Tris, 150 mM NaCl buffer pH 7.5 (1.5 mL) and incubated at room temperature for 1 h. The solution was then buffer exchanged on an Amicon Ultra-4 filter into 0.25 M sodium acetate buffer to obtain 1 mL of Df-Chx-Mal-thio-trastuzumab conjugate solution at a concentration of 6 mg/mL. MS ESI (m/z): found light chain 23440.2, heavy chain 51407.3 (Figure 23, D). Example 18 Df-Ac-thio-trastuzumab using Df-Bac
Figure imgf000108_0001
Df-Ac-ThioMab
The 12 mM stock solution of the bifunctional chelator was prepared by dissolving Df- Bac (8 mg, 12 μπιοΐ) in 1 mL DMA (Figure 21). The stock solution was then aliquoted and stored at -80 °C. The re -oxidized thio-trastuzumab was buffer exchanged on an Amicon Ultra-4 filter into 0.05 M sodium borate buffer pH 9. An aliquot of the Df-Bac stock solution (35 μί,, 0.410 μπιοΐ) was added to the solution of thio-trastuzumab (4.9 mg, 34 nmol) in 0.05 M sodium borate buffer pH 9 (1 mL) and incubated at room temperature for 5 h. The reaction mixture was loaded on a NAP- 10 column, and the Df-Ac-thio-trastuzumab was eluted with 1.5 mL of 0.25 M sodium acetate buffer to obtain the product at a concentration of 3.2 mg/mL. MS ESI (m/z): found light chain 23440.1 , heavy chain 51228.1 (Figure 23, B).
Example 19 Df-Ac-thio-trastuzumab using Df-Iac
Figure imgf000108_0002
Df-Ac-ThioMab
The 11 mM stock solution of the bifunctional chelator was prepared by dissolving Df- Iac (8 mg, 11 μπιοΐ) in DMSO (1 mL), the stock solution was then aliquoted and stored at - 80°C (Figure 21). The thio-trastuzumab solution (3.2 mL) was adjusted to pH 9 with the addition of 0.5 mL of 0.1 M sodium carbonate. An aliquot of the stock solution (110 μί, 1.20 μπιοΐ) was then added to the solution of thio-trastuzumab (16 mg, 110 nmol) in 50 mM Tris, 150 mM NaCl, 0.0125 M sodium carbonate buffer with pH 9 (4 mL) and incubated at room temperature for 2 h. The solution was then buffer exchanged on an Amicon Ultra-4 filter into 0.25 M sodium acetate buffer to obtain 1 mL of Df-Ac-thio-trastuzumab conjugate solution at a concentration of 8 mg/mL. MS ESI (m/z): found light chain 23440.1, heavy chain 51228.3 (Figure 23, C).
89
Example 20 General procedure for preparation of Zr chelates
89
The solution of Zr(IV) oxalate (2-4 mCi, 100 μί) in 1 M oxalic acid was mixed with
2 M solution of Na2C03 (45 μί) and incubated at room temperature for 3 min after which 0.5 M HEPES buffer (0.15 mL) was added (Figure 22). The Df-thio-trastuzumab conjugate (1 mg, 7 nmol) was diluted with 0.25 M sodium acetate/0.5% gentisic acid to a final volume of
89
0.356 mL and added to the Zr solution. Finally, a second portion of HEPES buffer (0.350 mL) was added resulting in 1 mL of total volume. The mixture was incubated at room
89
temperature for 1 h. To remove free Zr the radiolabeled protein was purified using a NAP- 10 desalting column. The NAP-10 column was equilibrated with 20 mL of 0.25M sodium acetate/0.5% gentisic acid. The reaction mixture was loaded on the NAP-10 column, and the
89
Zr-Df-thio-trastuzumab was eluted with 1.5 mL of 0.25M sodium acetate/0.5% gentisic acid
89
buffer (1.5 mL). If needed, the Zr-Df-thio-trastuzumab was concentrated using Amicon Ultra-4 filter to the desired volume. The product was analyzed by SEC HPLC (System A).
Example 21 In vitro serum stability
89
A solution of Zr-Df-thio-trastuzumab conjugate 0.5-1.5 mCi (1 mg) in 0.25 M sodium acetate/0.5% gentisic acid buffer (0.1 mL) was added to fresh mouse serum (0.9 mL) and incubated at 37°C for 0-96 h. Samples (20 μί) of the serum solution were analyzed using SEC HPLC (System A), with results shown in Figure 24.
Example 22 Animal models
Beige nude XID mice of age 6-8 weeks were obtained from Harlan Sprague Dawley (Livermore, CA). Three days prior to cell inoculation, the mice were implanted (s. c, left flank) with a 0.36 mg 60-day sustained release 17 -estradiol pellets (Innovative Research of America) to maintain serum estrogen level. Mice were inoculated in the mammary fat pad with 5 x 106 BT474M1 cells in 50% phenol red-free matrigel. BT474M1 is a subclone of human breast tumor cell line BT474 that was obtained from California Pacific Medical Center. Animal care and treatment followed protocols approved by Genentech's Institutioned Animal Care and Use Committee which is accredited by the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC).
Example 23 MicroPET Imaging
Mice were anesthetized with approx. 3% sevoflurane to effect and injected i.v. via the
89
tail vein with approximately 0.1 mCi of Zr-radio labeled trastuzumab in isotonic solution (100-130 μί) and returned to the cage for recovery. The PET imaging (Figure 25) was performed on an Inveon PET/CT scanner at 1, 24, 96 and 144 h post tracer injection as follows. Animals anesthetized with sevoflurane were placed head-first, prone position on the scanner bed and static 15 or 30 min scans were acquired. Body temperature was measured by a rectal probe and maintained with warm air. Full-body iterative image reconstructions were obtained using maximum a posteriori algorithm (MAP, hyperparameter beta (β) 0.05) and corrected for signal attenuation using the tissue density obtained from CT. Projections were created with ASIPro software (Siemens Preclinical Solutions) and used to obtain quantitative activity levels in each organ of interest using region-of-interest analysis.
Statistical analysis: The plots of Figure 26 were constructed with R software version 2.4.1 (R Foundation for Statistical Computing, Vienna, Austria). Statistical significance was determined using a two-tailed Student's t-test or ANOVA and P values of less than 0.05 were considered significant; data are presented as mean ± s.d. if not stated otherwise.
The present invention is not to be limited in scope by the specific embodiments disclosed in the examples which are intended as illustrations of a few aspects of the invention and any embodiments that are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art and are intended to fall within the scope of the appended claims.
All patents, patent applications, and references cited throughout the specification are expressly incorporated by reference.

Claims

We claim:
1. A zirconium-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a zirconium complex (Z), having Formula I:
Ab-(L-Z)p where p is 1 to 4.
2. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein p is
2.
3. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein the cysteine engineered antibody comprises a sequence in the heavy chain selected from SEQ ID NOS: 11, 12, 13, and 15:
LVTVCSASTKGPS SEQIDNO:ll
LVTVSCASTKGPS SEQIDNO:12
LVTVSSCSTKGPS SEQIDNO:13
HTFPCVLQSSGLYS SEQIDNO:15 where the cysteine in SEQ ID NOS: 11, 12, 13, and 15 is the free cysteine amino acid.
4. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein the cysteine engineered antibody comprises a sequence in the light chain selected from SEQ ID NOS: 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 45, and 46:
SLSASCGDRVT (SEQ ID NO: 17)
QKPGKCPKLLI (SEQ ID NO: 18)
EIKRTCAAPSV (SEQ ID NO: 19)
TCAAPCVFIFPP (SEQ ID NO:20)
FIFPPCDEQLK (SEQ ID NO:21)
DEQLKCGTASV (SEQ ID NO:22)
FYPRECKVQWK (SEQ ID NO:23)
WKVDNCLQSGN (SEQ ID NO:24)
ALQSGCSQESV (SEQ ID NO:25) VTEQDCKDSTY (SEQ ID NO:26)
GLSSPCTKSFN (SEQ ID NO:27)
FLSVSCGGRVT (SEQ ID NO:45)
QKPGNCPRLLI (SEQ ID NO:46) where the cysteine in SEQ ID NOS: 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 45, and 46 is the free cysteine amino acid.
5. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein the cysteine engineered antibody is prepared by a process comprising:
(i) mutagenizing a nucleic acid sequence encoding the cysteine engineered antibody;
(ii) expressing the cysteine engineered antibody; and
(iii) isolating and purifying the cysteine engineered antibody.
6. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein the cysteine engineered antibody is selected from a monoclonal antibody, a bispecific antibody, a chimeric antibody, a human antibody, a humanized antibody, and a Fab fragment.
7. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein the cysteine engineered antibody is A121C thio-trastuzumab.
8. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein the cysteine engineered antibody is prepared by a process comprising replacing one or more amino acid residues of a parent antibody with the one or more free cysteine amino acids, where the parent antibody selectively binds to an antigen and the cysteine engineered antibody selectively binds to the same antigen as the parent antibody.
9. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein the cysteine engineered antibody or the parent antibody binds to one or more of receptors (1)-
(51):
(1) BMPR1B (bone morphogenetic protein receptor-type IB);
(2) E16 (LAT1, SLC7A5);
(3) STEAP1 (six transmembrane epithelial antigen of prostate);
(4) 0772P (CA125, MUC16);
(5) MPF (MPF, MSLN, SMR, megakaryocyte potentiating factor, mesothelin);
(6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, solute carrier family 34 (sodium phosphate), member 2, type II sodium-dependent phosphate transporter 3b); (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thrombospondin repeats (type 1 and type 1 -like),
transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 5B);
(8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C 12 gene);
(9) ETBR (Endothelin type B receptor);
(10) MSG783 (RNF124, hypothetical protein FLJ20315);
(11) STEAP2 (HGNC 8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, prostate cancer associated gene 1 , prostate cancer associated protein 1 , six transmembrane epithelial antigen of prostate 2, six transmembrane prostate protein);
(12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, subfamily M, member 4);
(13) CRIPTO (CR, CRl, CRGF, CRIPTO, TDGFl, teratocarcinoma-derived growth factor);
(14) CD21 (CR2 (Complement receptor 2) or C3DR (C3d/Epstein Barr virus receptor) or Hs.73792);
(15) CD79b (CD79B, CD79 , IGb (immunoglobulin-associated beta), B29);
(16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchor protein la), SPAP1B, SPAP1C);
(17) HER2;
(18) NCA;
(19) MDP;
(20) IL20Ra;
(21) Brevican;
(22) EphB2R;
(23) ASLG659;
(24) PSCA;
(25) GEDA;
(26) BAFF-R (B cell -activating factor receptor, BLyS receptor 3, BR3;
(27) CD22 (B-cell receptor CD22-B isoform);
(28) CD79a (CD79A, CD79a, immunoglobulin-associated alpha, a B cell-specific protein that covalently interacts with Ig beta (CD79B) and forms a complex on the surface with IgM molecules, transduces a signal involved in B-cell differentiation); (29) CXCR5 (Burkitt's lymphoma receptor 1, a G protein-coupled receptor that is activated by the CXCL13 chemokine, functions in lymphocyte migration and humoral defense, plays a role in HIV-2 infection and perhaps development of AIDS, lymphoma, myeloma, and leukemia);
(30) HLA-DOB (Beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4+ T lymphocytes);
(31) P2X5 (Purinergic receptor P2X ligand-gated ion channel 5, an ion channel gated by extracellular ATP, may be involved in synaptic transmission and neurogenesis, deficiency may contribute to the pathophysiology of idiopathic detrusor instability);
(32) CD72 (B-cell differentiation antigen CD72, Lyb-2);
(33) LY64 (Lymphocyte antigen 64 (RP105), type I membrane protein of the leucine rich repeat (LRR) family, regulates B-cell activation and apoptosis, loss of function is associated with increased disease activity in patients with systemic lupus erythematosis);
(34) FcRHl (Fc receptor-like protein 1, a putative receptor for the immunoglobulin Fc domain that contains C2 type Ig-like and IT AM domains, may have a role in B-lymphocyte differentiation);
(35) IRTA2 (Immunoglobulin superfamily receptor translocation associated 2, a putative immunoreceptor with possible roles in B cell development and lymphomagenesis; deregulation of the gene by translocation occurs in some B cell malignancies);
(36) TENB2 (putative transmembrane proteoglycan, related to the EGF/heregulin family of growth factors and follistatin);
(37) PMEL17 (silver homolog; SILV; D12S53E; PMEL17; (SI); (SIL); ME20;
gplOO);
(38) TMEFF1 (transmembrane protein with EGF-like and two follistatm-like domains 1 ; Tomoregulin-1; H7365; C9orf2; C90RF2; U19878; X83961;
(39) GDNF-Ral (GDNF family receptor alpha 1; GFRA1; GDNFR; GDNFRA;
RETL1; TRNR1; RET1L; GDNFR-alphal; GFR-ALPHA-1; U95847; BC014962);
(40) Ly6E (lymphocyte antigen 6 complex, locus E; Ly67,RIG-E,SCA-2,TSA-l);
(41) TMEM46 (shisa homolog 2 (Xenopus laevis); SHISA2);
(42) Ly6G6D (lymphocyte antigen 6 complex, locus G6D; Ly6-D, MEGT1);
(43) LGR5 (leucine-ricli repeat-containing G protein-coupled receptor 5; GPR49, GPR67);
(44) RET (ret proto-oncogene; MEN2A; HSCR1; MEN2B; MTC1; (PTC); CDHF12; Hs.168114; RET51; RET-ELE1); (45) LY6K (lymphocyte antigen 6 complex, locus K; LY6K; HSJ001348; FLJ35226);
(46) GPR19 (G protein-coupled receptor 19; Mm.4787);
(47) GPR54 (KISS1 receptor; KISS1R; GPR54; HOT7T175; AXOR12);
(48) ASPHD1 (aspartate beta-hydroxylase domain containing 1 ; LOC253982);
(49) Tyrosinase (TYR; OCAIA; OCA1A; tyrosinase; SHEP3);
(50) TMEM118 (ring finger protein, transmembrane 2; RNFT2; FLJ14627); and
(51) GPR172A (G protein-coupled receptor 172A; GPCR41; FLJ11856;
D15Ertd747e).
10. The zirconium-labelled, cysteine-engineered antibody of claim 1 wherein Z comprises zirconium complexed to desferoxamine B.
11. The zirconium-labelled, cysteine-engineered antibody of claim 10 wherein
89
zirconi m is complexed to the structure:
Figure imgf000115_0001
where the wavy line indicates the attachment to the linker (L).
12. The zirconium-labelled, cysteine-engineered antibody of claim 1 selected from the structures:
Figure imgf000115_0002
Figure imgf000116_0001
where X is selected from:
Figure imgf000116_0002
~ (CH2)n (CH2CH20)n - -(CH2CH20)n CH2CH2-
Figure imgf000116_0003
Y is selected from:
— N-(CH2)n
Figure imgf000116_0004
R is independently H or Ci-C6 alkyl; and n is 1 to 12.
13. A desferrioxamine-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II: l o Ab-(L-Df)p II
wherein L-Df is selected from:
Figure imgf000117_0001
where the wavy line indicates the attachment to the antibody (Ab); and p is 1 to 4.
14. A desferrioxamine-labelling reagent selected from the structures:
Figure imgf000117_0002
wherein R is selected from:
Figure imgf000117_0003
15. A method of making a desferrioxamine-labelled, cysteine-engineered antibody comprising comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
Ab-(L-Df) II wherein L-Df is selected from:
Figure imgf000118_0001
where the wavy line indicates the attachment to the antibody (Ab); and
p is 1 to 4;
the method comprising reacting a composition selected from the structures:
Figure imgf000118_0002
wherein R is selected from:
Figure imgf000119_0001
with a cysteine-engineered antibody having one or more free cysteine amino acids, whereby the desferrioxamine-labelled, cysteine-engineered antibody is formed.
16. A method of making a zirconium-labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a zirconium complex (Z), having Formula I:
Figure imgf000119_0002
where p is 1 to 4;
the method comprising complexing a zirconium reagent with a desferrioxamine- labelled, cysteine-engineered antibody comprising a cysteine engineered antibody (Ab) conjugated through a free cysteine amino acid to a linker (L) and a desferoxamine moiety (Df), having Formula II:
Ab-(L-Df)p II
wherein L-Df is selected from:
Figure imgf000120_0001
where the wavy line indicates the attachment to the antibody (Ab); and
p is 1 to 4;
whereby a desferrioxamine-labelled, cysteine-engineered antibody is formed.
17. The method of claim 16 wherein the zirconium reagent is 89 zirconium oxalate.
18. A method of imaging comprising:
administering a zirconium-labelled, cysteine-engineered antibody to an animal; and detecting in vivo the presence of the zirconium-labelled, cysteine-engineered antibody by imaging,
wherein the zirconium-labelled, cysteine-engineered antibody comprises a cysteine engineered antibody (Ab) having one or more free cysteine amino acids conjugated with one or more zirconium complex (Z) through a linker (L), and having Formula I:
Figure imgf000120_0002
where p is 1 to 4.
19. The method of claim 18 wherein Z comprises zirconium complexed to desferoxamine B.
20. The method of claim 18 wherein the zirconium-labelled, cysteine-engineered antibody binds an antigen.
21. The method of claim 18 wherein the animal is a tumor xenograft mouse model.
PCT/US2010/055465 2009-11-05 2010-11-04 Zirconium-radiolabeled, cysteine engineered antibody conjugates WO2011056983A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA2780216A CA2780216A1 (en) 2009-11-05 2010-11-04 Zirconium-desferrioxamine-labelled and desferrioxamine-labelled, cysteine-engineered antibodies and methods of making said antibodies
JP2012537224A JP5850843B2 (en) 2009-11-05 2010-11-04 Zirconium radiolabeled cysteine engineered antibody conjugate
KR1020177034613A KR20170136652A (en) 2009-11-05 2010-11-04 Zirconium-radiolabeled, cysteine engineered antibody conjugates
EP10776064.7A EP2496270B1 (en) 2009-11-05 2010-11-04 Zirconium-radiolabeled, cysteine engineered antibody conjugates
RU2012123007/10A RU2562862C2 (en) 2009-11-05 2010-11-04 Radioactive zirconium- labelled engineered cysteine antibodies conjugates
CN201080050134.4A CN102596260B (en) 2009-11-05 2010-11-04 Radiolabeled, the cysteine engineered antibody conjugates of zirconium
BR112012007774A BR112012007774A2 (en) 2009-11-05 2010-11-04 cysteine modified antibody, desferrioxamine labeling reagent, production method of a cysteine modified antibody production method of a zirconium labeled cysteine modified antibody and imaging method
MX2012005211A MX340674B (en) 2009-11-05 2010-11-04 Zirconium-radiolabeled, cysteine engineered antibody conjugates.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12/612,912 2009-11-05
US12/612,912 US20100111856A1 (en) 2004-09-23 2009-11-05 Zirconium-radiolabeled, cysteine engineered antibody conjugates

Publications (1)

Publication Number Publication Date
WO2011056983A1 true WO2011056983A1 (en) 2011-05-12

Family

ID=43383591

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/055465 WO2011056983A1 (en) 2009-11-05 2010-11-04 Zirconium-radiolabeled, cysteine engineered antibody conjugates

Country Status (10)

Country Link
US (2) US20100111856A1 (en)
EP (1) EP2496270B1 (en)
JP (1) JP5850843B2 (en)
KR (2) KR20170136652A (en)
CN (1) CN102596260B (en)
BR (1) BR112012007774A2 (en)
CA (1) CA2780216A1 (en)
MX (1) MX340674B (en)
RU (1) RU2562862C2 (en)
WO (1) WO2011056983A1 (en)

Cited By (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013149159A1 (en) 2012-03-30 2013-10-03 Genentech, Inc. Anti-lgr5 antibodies and immunoconjugates
WO2013165940A1 (en) 2012-05-01 2013-11-07 Genentech, Inc. Anti-pmel17 antibodies and immunoconjugates
US8772459B2 (en) 2009-12-02 2014-07-08 Imaginab, Inc. J591 minibodies and Cys-diabodies for targeting human prostate specific membrane antigen (PSMA) and methods for their use
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
US8877897B2 (en) 2010-02-23 2014-11-04 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8911732B2 (en) 2010-12-20 2014-12-16 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US8940298B2 (en) 2007-09-04 2015-01-27 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting and detection
US8940871B2 (en) 2006-03-20 2015-01-27 The Regents Of The University Of California Engineered anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting
US8951737B2 (en) 1996-05-06 2015-02-10 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
WO2015042108A1 (en) 2013-09-17 2015-03-26 Genentech, Inc. Methods of using anti-lgr5 antibodies
WO2015089344A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015112909A1 (en) 2014-01-24 2015-07-30 Genentech, Inc. Methods of using anti-steap1 antibodies and immunoconjugates
JP2015528692A (en) * 2012-05-21 2015-10-01 ジェネンテック, インコーポレイテッド Anti-Ly6E antibody, immunoconjugate and method of use
JP2015528818A (en) * 2012-08-02 2015-10-01 ジェネンテック, インコーポレイテッド Anti-ETBR antibodies and immunoconjugates
JP2015529656A (en) * 2012-08-02 2015-10-08 ジェネンテック, インコーポレイテッド Anti-ETBR antibodies and immune complexes
WO2015179658A2 (en) 2014-05-22 2015-11-26 Genentech, Inc. Anti-gpc3 antibodies and immunoconjugates
WO2015191715A1 (en) 2014-06-11 2015-12-17 Genentech, Inc. Anti-lgr5 antibodies and uses thereof
WO2016040868A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
US9290578B2 (en) 2013-10-21 2016-03-22 Genentech, Inc. Anti-Ly6E antibodies and methods of use
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2017181152A2 (en) 2016-04-15 2017-10-19 Alpine Immune Sciences, Inc. Cd80 variant immunomodulatory proteins and uses thereof
WO2017181148A2 (en) 2016-04-15 2017-10-19 Alpine Immune Sciences, Inc. Icos ligand variant immunomodulatory proteins and uses thereof
WO2017197234A1 (en) 2016-05-13 2017-11-16 Bioatla, Llc Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
WO2017223405A1 (en) 2016-06-24 2017-12-28 Genentech, Inc. Anti-polyubiquitin multispecific antibodies
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
WO2018022945A1 (en) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Cd112 variant immunomodulatory proteins and uses thereof
WO2018022946A1 (en) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Cd155 variant immunomodulatory proteins and uses thereof
US9896506B2 (en) 2008-01-31 2018-02-20 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
EP3207027A4 (en) * 2014-10-16 2018-04-18 The University of Melbourne Novel imaging composition and uses thereof
WO2018091724A1 (en) 2016-11-21 2018-05-24 Cureab Gmbh Anti-gp73 antibodies and immunoconjugates
WO2018102682A1 (en) 2016-12-01 2018-06-07 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-pd-l1 antibodies for immuno-pet imaging
WO2018102787A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
US10011658B2 (en) 2015-04-03 2018-07-03 Eureka Therapeutics, Inc. Constructs targeting AFP peptide/MHC complexes and uses thereof
WO2018148476A1 (en) 2017-02-10 2018-08-16 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-lag3 antibodies for immuno-pet imaging
US10059768B2 (en) 2014-09-12 2018-08-28 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US10058613B2 (en) 2015-10-02 2018-08-28 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US10077318B2 (en) 2014-09-12 2018-09-18 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2018170021A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l1 variant immunomodulatory proteins and uses thereof
WO2018170023A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l2 variant immunomodulatory proteins and uses thereof
WO2018170026A2 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Cd80 variant immunomodulatory proteins and uses thereof
US10137212B2 (en) * 2013-03-13 2018-11-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tetrahydroxamate chelators of zirconium89 and niobium90 for use in diagnostic applications
WO2018223098A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
US10179820B2 (en) 2014-09-12 2019-01-15 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
WO2019023148A1 (en) 2017-07-24 2019-01-31 Regeneron Pharmaceuticals, Inc. Anti-cd8 antibodies and uses thereof
WO2019074983A1 (en) 2017-10-10 2019-04-18 Alpine Immune Sciences, Inc. Ctla-4 variant immunomodulatory proteins and uses thereof
WO2019079520A2 (en) 2017-10-18 2019-04-25 Alpine Immune Sciences, Inc. Variant icos ligand immunomodulatory proteins and related compositions and methods
WO2019089858A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
WO2019089848A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019109053A1 (en) 2017-12-01 2019-06-06 Juno Therapeutics, Inc. Methods for dosing and for modulation of genetically engineered cells
US10494432B2 (en) 2007-07-16 2019-12-03 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
WO2019241758A1 (en) 2018-06-15 2019-12-19 Alpine Immune Sciences, Inc. Pd-1 variant immunomodulatory proteins and uses thereof
US10517969B2 (en) 2009-02-17 2019-12-31 Cornell University Methods and kits for diagnosis of cancer and prediction of therapeutic value
US10533058B2 (en) 2013-12-16 2020-01-14 Genentech Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US10604557B2 (en) 2010-06-08 2020-03-31 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2020077257A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Pd-1 single domain antibodies and therapeutic compositions thereof
WO2020076992A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. 5t4 single domain antibodies and therapeutic compositions thereof
WO2020076970A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. B7h3 single domain antibodies and therapeutic compositions thereof
WO2020076977A2 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Dll3 single domain antibodies and therapeutic compositions thereof
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020113141A2 (en) 2018-11-30 2020-06-04 Alpine Immune Sciences, Inc. Cd86 variant immunomodulatory proteins and uses thereof
WO2020117257A1 (en) 2018-12-06 2020-06-11 Genentech, Inc. Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody
EP3689910A2 (en) 2014-09-23 2020-08-05 F. Hoffmann-La Roche AG Method of using anti-cd79b immunoconjugates
WO2020205623A1 (en) 2019-03-29 2020-10-08 Rakuten Medical, Inc. Methods for photoimmunotherapy and related biomarkers
WO2020232169A1 (en) 2019-05-14 2020-11-19 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat follicular lymphoma
WO2020257604A1 (en) 2019-06-21 2020-12-24 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind muc16 and cd3 in combination with 4-1bb co-stimulation
WO2020257681A1 (en) 2019-06-21 2020-12-24 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind psma and cd3 in combination with 4-1bb co-stimulation
WO2021055350A1 (en) 2019-09-16 2021-03-25 Regeneron Pharmaceuticals, Inc. Radiolabeled met binding proteins for immuno-pet imaging
US10981987B2 (en) 2007-07-16 2021-04-20 Genentech, Inc. Humanized anti-CD79b antibodies and immunoconjugates and methods of use
WO2021076196A1 (en) 2019-10-18 2021-04-22 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2021113776A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
WO2021113780A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to gprc5d-targeted binding domains and related compositions and methods
WO2021155071A1 (en) 2020-01-29 2021-08-05 Inhibrx, Inc. Cd28 single domain antibodies and multivalent and multispecific constructs thereof
US11142578B2 (en) 2016-11-16 2021-10-12 Regeneron Pharmaceuticals, Inc. Anti-MET antibodies, bispecific antigen binding molecules that bind MET, and methods of use thereof
US11149088B2 (en) 2016-04-15 2021-10-19 Bioatla, Inc. Anti-Axl antibodies, antibody fragments and their immunoconjugates and uses thereof
WO2021217051A1 (en) 2020-04-24 2021-10-28 Genentech, Inc. Methods of using anti-cd79b immunoconjugates
WO2022029660A1 (en) 2020-08-05 2022-02-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
US11254744B2 (en) 2015-08-07 2022-02-22 Imaginab, Inc. Antigen binding constructs to target molecules
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies
US11274157B2 (en) 2017-01-12 2022-03-15 Eureka Therapeutics, Inc. Constructs targeting histone H3 peptide/MHC complexes and uses thereof
WO2022147463A2 (en) 2020-12-31 2022-07-07 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
US11447564B2 (en) 2017-04-26 2022-09-20 Eureka Therapeutics, Inc. Constructs specifically recognizing glypican 3 and uses thereof
US11471488B2 (en) 2016-07-28 2022-10-18 Alpine Immune Sciences, Inc. CD155 variant immunomodulatory proteins and uses thereof
WO2022241446A1 (en) 2021-05-12 2022-11-17 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023019092A1 (en) 2021-08-07 2023-02-16 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
US11584927B2 (en) 2014-08-28 2023-02-21 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified T-cells
US11596695B2 (en) 2013-02-08 2023-03-07 Novartis Ag Specific sites for modifying antibodies to make immunoconjugates
WO2023034750A1 (en) 2021-08-30 2023-03-09 Genentech, Inc. Anti-polyubiquitin multispecific antibodies
US11667724B2 (en) 2017-07-07 2023-06-06 Astellas Pharma Inc. Anti-human CEACAM5 antibody Fab fragment
US11679166B2 (en) 2016-11-18 2023-06-20 Astellas Pharma Inc. Anti-human MUC1 antibody Fab fragment
WO2023172883A1 (en) 2022-03-07 2023-09-14 Alpine Immune Sciences, Inc. Immunomodulatory proteins of variant cd80 polypeptides, cell therapies thereof and related methods and uses
US11993661B2 (en) 2018-06-18 2024-05-28 Eureka Therapeutics, Inc. Constructs targeting prostate-specific membrane antigen (PSMA) and uses thereof
WO2024151515A2 (en) 2023-01-09 2024-07-18 Odyssey Therapeutics, Inc. Anti-tnfr2 antigen-binding proteins and uses thereof
US12065503B2 (en) 2018-10-10 2024-08-20 Astellas Pharma Inc. Pharmaceutical composition containing tagged site-antihuman antibody fab fragment complex
WO2024173876A1 (en) 2023-02-17 2024-08-22 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-lag3 antibodies for immuno-pet imaging
WO2024192065A1 (en) 2023-03-14 2024-09-19 Odyssey Therapeutics, Inc. Anti-cd25 antigen-binding proteins and uses thereof

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9701754B1 (en) 2002-10-23 2017-07-11 City Of Hope Covalent disulfide-linked diabodies and uses thereof
JP5951929B2 (en) * 2007-10-03 2016-07-13 コーネル ユニヴァーシティー Treatment of proliferative disorders using PSMA antibodies
GB0803076D0 (en) 2008-02-20 2008-03-26 Univ Ghent Mucosal Membrane Receptor and uses thereof
US20100008978A1 (en) * 2008-05-09 2010-01-14 The Regents Of The University Of California Nanoparticles effective for internalization into cells
US20100009390A1 (en) * 2008-05-09 2010-01-14 The Regents Of The University Of California Mutant antibodies with high affinity for egfr
WO2010014222A1 (en) * 2008-07-30 2010-02-04 Dana-Farber Cancer Institute, Inc. Compositions for detecting cell death and methods of use thereof
EP2400992B1 (en) * 2009-02-27 2015-07-22 Genentech, Inc. Methods and compositions for protein labelling
EP2577310B1 (en) * 2010-06-03 2018-11-14 F.Hoffmann-La Roche Ag Immuno-pet imaging of antibodies and immunoconjugates and uses therefor
TW201302793A (en) 2010-09-03 2013-01-16 Glaxo Group Ltd Novel antigen binding proteins
EP2786151B1 (en) 2011-11-29 2019-07-03 F.Hoffmann-La Roche Ag Methods for prostate cancer analysis
KR102006997B1 (en) * 2012-07-03 2019-08-02 한국생명공학연구원 A site-selective binding peptide for IgG Fc and a hybrid molecule comprising the same
RU2017128512A (en) 2012-07-04 2019-02-15 Ф. Хоффманн-Ля Рош Аг ANTIBODIES TO THEOPHYLLIN AND WAYS OF THEIR APPLICATION
CN107973856B (en) 2012-07-04 2021-11-23 弗·哈夫曼-拉罗切有限公司 Covalently linked antigen-antibody conjugates
CN104411725B (en) 2012-07-04 2018-09-28 弗·哈夫曼-拉罗切有限公司 Anti-biotin antibodies and application method
WO2014011327A1 (en) * 2012-07-12 2014-01-16 Dana-Farber Cancer Institute, Inc. Radiolabeled probes for the non-invasive detection and imaging of cell death
WO2014069647A1 (en) * 2012-11-05 2014-05-08 全薬工業株式会社 Antibody and antibody composition production method
US20140147381A1 (en) * 2012-11-29 2014-05-29 Gregory David Espenan 89zr compounds, to include somatostatin, apparatus and products comprising such compounds, methods of making same, and methods of using same for radio imaging and/or treatment
CA2898146C (en) 2012-12-19 2020-12-01 Charles Andrew BOSWELL Methods and compositions for radiohalogen protein labeling
ES2699599T3 (en) * 2013-03-15 2019-02-11 Abbvie Biotherapeutics Inc Fc variants
EP3008212A4 (en) * 2013-06-10 2017-05-24 Millennium Pharmaceuticals, Inc. Methods of treatment of cancer
MX2016007958A (en) 2013-12-17 2016-08-03 Genentech Inc Anti-cd3 antibodies and methods of use.
EP3089759B1 (en) 2014-01-03 2018-12-05 F. Hoffmann-La Roche AG Covalently linked polypeptide toxin-antibody conjugates
RU2694981C2 (en) 2014-01-03 2019-07-18 Ф. Хоффманн-Ля Рош Аг Covalently linked conjugates chelicar-antibody against chelicar and use thereof
PL3089996T3 (en) 2014-01-03 2021-12-13 F. Hoffmann-La Roche Ag Bispecific anti-hapten/anti-blood brain barrier receptor antibodies, complexes thereof and their use as blood brain barrier shuttles
WO2015138615A2 (en) * 2014-03-12 2015-09-17 Irm Llc Specific sites for modifying antibodies to make immunoconjugates
ES2768762T3 (en) * 2014-11-05 2020-06-23 Illumina Cambridge Ltd Reduction of DNA damage during sample preparation and sequencing using siderophore chelating agents
CN107001447B (en) 2014-12-17 2021-07-23 豪夫迈·罗氏有限公司 Novel enzyme-mediated polypeptide conjugation method using sortase
CN104645364A (en) * 2015-01-27 2015-05-27 南京江原安迪科正电子研究发展有限公司 Mark product of <89>Zr marked denosumab, and preparation method and application thereof
WO2016179003A1 (en) 2015-05-01 2016-11-10 Genentech, Inc. Masked anti-cd3 antibodies and methods of use
EP3307780A1 (en) 2015-06-15 2018-04-18 Genentech, Inc. Antibodies and immunoconjugates
CN107849145B (en) 2015-06-16 2021-10-26 基因泰克公司 anti-CD 3 antibodies and methods of use thereof
CN108138158B (en) 2015-09-25 2021-11-09 豪夫迈·罗氏有限公司 Soluble sortase A
JP6895953B2 (en) 2015-09-25 2021-06-30 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Method for making thioester using sortase A
WO2017050889A1 (en) 2015-09-25 2017-03-30 F. Hoffmann-La Roche Ag Recombinant immunoglobulin heavy chains comprising a sortase conjugation loop and conjugates thereof
WO2017050872A1 (en) 2015-09-25 2017-03-30 F. Hoffmann-La Roche Ag Transamidation employing sortase a in deep eutectic solvents
WO2017161356A1 (en) * 2016-03-18 2017-09-21 Wake Forest University Compounds, compositions and associated methods using zirconium-89 in immuno-positron emission tomography
CN109072212B (en) 2016-03-30 2022-10-04 豪夫迈·罗氏有限公司 Improved sortase
CN109069657B (en) * 2016-04-06 2022-04-12 阿特根公司 Antibody-drug conjugates comprising modified antibodies
EP3609897A1 (en) 2017-04-12 2020-02-19 H. Hoffnabb-La Roche Ag A method for labeling of aldehyde containing target molecules
EP3652214A1 (en) 2017-07-13 2020-05-20 H. Hoffnabb-La Roche Ag New binding agent and assay for pivka
AU2018321893A1 (en) 2017-08-23 2020-03-19 Wayne State University In vivo immunoimaging of interferon-gamma
JP7403444B2 (en) 2017-10-20 2023-12-22 エフ. ホフマン-ラ ロシュ アーゲー Antibody copy protection measures
WO2019165143A1 (en) * 2018-02-21 2019-08-29 Cytomx Therapeutics, Inc. Positron emission tomography imaging of activatable binding polypeptides and related compositions thereof
WO2019175127A1 (en) 2018-03-14 2019-09-19 F. Hoffmann-La Roche Ag Novel anti-troponint antibodies
JP7333332B2 (en) 2018-03-14 2023-08-24 エフ. ホフマン-ラ ロシュ アーゲー Methods for Affinity Maturation of Antibodies
WO2019201901A1 (en) 2018-04-18 2019-10-24 F. Hoffmann-La Roche Ag Novel anti-thymidine kinase antibodies
WO2020043868A1 (en) 2018-08-31 2020-03-05 F. Hoffmann-La Roche Ag Thymidine kinase (tk-1) in prognostic indices for dlbcl
JOP20210159A1 (en) 2018-12-21 2023-01-30 Novartis Ag Antibodies to pmel17 and conjugates thereof
EP3921443A1 (en) 2019-02-08 2021-12-15 F. Hoffmann-La Roche AG Diagnostic and therapeutic methods for cancer
WO2020236679A1 (en) * 2019-05-17 2020-11-26 Cytomx Therapeutics, Inc. Methods and compositions for determining the biodistribution of activatable anti-cd166 antibody conjugates
JP2022537543A (en) * 2019-06-17 2022-08-26 タグワークス ファーマシューティカルス ビー.ブイ. Compounds for fast and efficient click release
WO2022036146A1 (en) 2020-08-12 2022-02-17 Genentech, Inc. Diagnostic and therapeutic methods for cancer
CN116457660A (en) 2020-10-30 2023-07-18 豪夫迈·罗氏有限公司 TIMP1 as a marker for cholangiocarcinoma
EP4448564A1 (en) 2021-12-17 2024-10-23 F. Hoffmann-La Roche AG A novel antibody for detection of amyloid beta 42 (a-beta42)
AU2023236386A1 (en) 2022-03-18 2024-10-10 Evolveimmune Therapeutics, Inc. Bispecific antibody fusion molecules and methods of use thereof
CN117198390B (en) * 2023-09-08 2024-03-12 中国科学院广州生物医药与健康研究院 Preparation method of SLC (SLC) membrane protein complex by designing and modifying disulfide bond crosslinking site

Citations (286)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1991002536A1 (en) 1989-08-23 1991-03-07 Scripps Clinic And Research Foundation Compositions and methods for detection and treatment of epstein-barr virus infection and immune disorders
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5091178A (en) 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
WO1992007574A1 (en) 1990-10-25 1992-05-14 Tanox Biosystems, Inc. Glycoproteins associated with membrane-bound immunoglobulins as antibody targets on b cells
WO1992017497A1 (en) 1991-03-29 1992-10-15 Genentech, Inc. Human pf4a receptors and their use
EP0522868A1 (en) 1991-07-12 1993-01-13 SHIONOGI SEIYAKU KABUSHIKI KAISHA trading under the name of SHIONOGI &amp; CO. LTD. A human endothelin receptor
JPH053790B2 (en) 1983-11-02 1993-01-18 Canon Kk
US5183884A (en) 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
WO1993011161A1 (en) 1991-11-25 1993-06-10 Enzon, Inc. Multivalent antigen-binding proteins
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1993021319A1 (en) 1992-04-08 1993-10-28 Cetus Oncology Corporation HUMANIZED C-erbB-2 SPECIFIC ANTIBODIES
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
WO1994010312A1 (en) 1992-10-23 1994-05-11 Chugai Seiyaku Kabushiki Kaisha Gene coding for megakaryocyte potentiator
WO1994011026A2 (en) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
US5316757A (en) 1984-10-18 1994-05-31 Board Of Regents, The University Of Texas System Synthesis of polyazamacrocycles with more than one type of side-chain chelating groups
EP0599274A1 (en) 1992-11-24 1994-06-01 Bristol-Myers Squibb Company HER4, a human receptor tyrosine kinase of the epidermal growth factor receptor family
US5342606A (en) 1984-10-18 1994-08-30 Board Of Regents, The University Of Texas System Polyazamacrocyclic compounds for complexation of metal ions
WO1994028931A1 (en) 1993-06-11 1994-12-22 Genentech, Inc. Methods for treating inflammatory disorders
US5385893A (en) 1993-05-06 1995-01-31 The Dow Chemical Company Tricyclopolyazamacrocyclophosphonic acids, complexes and derivatives thereof, for use as contrast agents
US5428139A (en) 1991-12-10 1995-06-27 The Dow Chemical Company Bicyclopolyazamacrocyclophosphonic acid complexes for use as radiopharmaceuticals
US5440021A (en) 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
WO1995025167A1 (en) 1994-03-17 1995-09-21 Merck Patent Gmbh Anti-egfr single-chain fvs and anti-egfr antibodies
US5462725A (en) 1993-05-06 1995-10-31 The Dow Chemical Company 2-pyridylmethylenepolyazamacrocyclophosphonic acids, complexes and derivatives thereof, for use as contrast agents
US5480990A (en) 1991-12-10 1996-01-02 The Dow Chemical Company Bicyclopolyazamacrocyclocarboxylic acid complexes for use as contrast agents
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
WO1996030514A1 (en) 1995-03-31 1996-10-03 University Of Washington Intracellular domain of the her-2/neu protein for prevention or treatment of malignancies
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
WO1997004801A1 (en) 1995-07-27 1997-02-13 Genentech, Inc. Stabile isotonic lyophilized protein formulation
WO1997007198A2 (en) 1995-08-11 1997-02-27 Genetics Institute, Inc. Dna sequences and secreted proteins encoded thereby
US5644033A (en) 1992-12-22 1997-07-01 Health Research, Inc. Monoclonal antibodies that define a unique antigen of human B cell antigen receptor complex and methods of using same for diagnosis and treatment
WO1997035885A1 (en) 1996-03-27 1997-10-02 Genentech, Inc. ErbB3 ANTIBODIES
US5677171A (en) 1988-01-12 1997-10-14 Genentech, Inc. Monoclonal antibodies directed to the HER2 receptor
WO1997044452A1 (en) 1996-05-17 1997-11-27 Schering Corporation Human b-cell antigens, related reagents
US5739294A (en) 1991-12-10 1998-04-14 The Dow Chemical Company Bicyclopol yazamacrocyclophosphonic acid complexes for use as contrast agents
US5773223A (en) 1993-09-02 1998-06-30 Chiron Corporation Endothelin B1, (ETB1) receptor polypeptide and its encoding nucleic acid methods, and uses thereof
US5792616A (en) 1990-05-29 1998-08-11 The United States Of America Antibodies to human cripto protein
WO1998037193A1 (en) 1997-02-20 1998-08-27 Zymogenetics, Inc. Zcytor7 cytokine receptor
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
WO1998040403A1 (en) 1997-03-10 1998-09-17 The Regents Of The University Of California Psca: prostate stem cell antigen
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
EP0875569A1 (en) 1997-04-28 1998-11-04 Smithkline Beecham Corporation A human sodium dependent phosphate transporter (IPT-1)
US5834456A (en) 1996-02-23 1998-11-10 The Dow Chemical Company Polyazamacrocyclofluoromonoalkylphosphonic acids, and their complexes, for use as contrast agents
WO1998051824A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting disease of the urinary tract
WO1998051805A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting diseases of the prostate
US5854399A (en) 1991-08-23 1998-12-29 The United States Of America As Represented By The Department Of Health And Human Services Antibodies specific for human cripto-related polypeptide CR-3
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
WO1999019488A1 (en) 1997-10-15 1999-04-22 Children's Medical Center Corporation Novel human egf receptors and use thereof
WO1999028468A1 (en) 1997-12-02 1999-06-10 The Regents Of The University Of California Modulating b lymphocyte chemokine/receptor interactions
WO1999046284A2 (en) 1998-03-13 1999-09-16 The Burnham Institute Molecules that home to various selected organs or tissues
US5976551A (en) 1991-11-15 1999-11-02 Institut Pasteur And Institut Nationale De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and method of using the determinant
WO1999058658A2 (en) 1998-05-13 1999-11-18 Epimmune, Inc. Expression vectors for stimulating an immune response and methods of using the same
WO2000012130A1 (en) 1998-08-27 2000-03-09 Smithkline Beecham Corporation Rp105 agonists and antagonists
WO2000014228A1 (en) 1998-09-03 2000-03-16 Japan Science And Technology Corporation Neutral amino acid transporter and gene thereof
WO2000020579A1 (en) 1998-10-02 2000-04-13 Mcmaster University Spliced form of erbb-2/neu oncogene
WO2000022129A1 (en) 1998-10-13 2000-04-20 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human g protein-coupled receptors
WO2000032752A1 (en) 1998-12-02 2000-06-08 The Regents Of The University Of California Psca: prostate stem cell antigen and uses thereof
WO2000036107A2 (en) 1998-12-17 2000-06-22 Corixa Corporation Compositions and methods for therapy and diagnosis of ovarian cancer
WO2000040614A2 (en) 1998-12-30 2000-07-13 Beth Israel Deaconess Medical Center, Inc. Characterization of the soc/crac calcium channel protein family
WO2000044899A1 (en) 1999-01-29 2000-08-03 Corixa Corporation Her-2/neu fusion proteins
WO2000053216A2 (en) 1999-03-05 2000-09-14 Smithkline Beecham Biologicals S.A. Use of casb616 polypeptides and polynucleotides for cancer treatment
WO2000055351A1 (en) 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Human colon cancer associated gene sequences and polypeptides
US6153408A (en) 1991-11-15 2000-11-28 Institut Pasteur And Institut National De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
WO2000075655A1 (en) 1999-06-03 2000-12-14 Takeda Chemical Industries, Ltd. Screening method with the use of cd100
WO2001000245A2 (en) 1999-06-25 2001-01-04 Genentech, Inc. HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
WO2001000244A2 (en) 1999-06-25 2001-01-04 Genentech, Inc. METHODS OF TREATMENT USING ANTI-ErbB ANTIBODY-MAYTANSINOID CONJUGATES
WO2001016318A2 (en) 1999-09-01 2001-03-08 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
WO2001038490A2 (en) 1999-11-29 2001-05-31 The Trustees Of Columbia University In The City Of New York ISOLATION OF FIVE NOVEL GENES CODING FOR NEW Fc RECEPTORS-TYPE MELANOMA INVOLVED IN THE PATHOGENESIS OF LYMPHOMA/MELANOMA
WO2001040269A2 (en) 1999-11-30 2001-06-07 Corixa Corporation Compositions and methods for therapy and diagnosis of breast cancer
WO2001040309A2 (en) 1999-10-29 2001-06-07 Genentech, Inc. Anti-prostate stem cell antigen (psca) antibody compositions and methods of use
WO2001041787A1 (en) 1999-12-10 2001-06-14 Epimmune Inc. INDUCING CELLULAR IMMUNE RESPONSES TO HER2/neu USING PEPTIDE AND NUCLEIC ACID COMPOSITIONS
US6248564B1 (en) 1997-08-29 2001-06-19 Harvard University Mutant MHC class I molecules
WO2001046232A2 (en) 1999-12-23 2001-06-28 Zymogenetics, Inc. Soluble interleukin-20 receptor
WO2001045746A2 (en) 1999-12-24 2001-06-28 Genentech, Inc. Methods and compositions for prolonging elimination half-times of bioactive compounds
WO2001046261A1 (en) 1999-12-23 2001-06-28 Zymogenetics, Inc. Method for treating inflammation
WO2001048204A1 (en) 1999-12-23 2001-07-05 Agresearch Limited Mutated bmp1b receptor as regulator of ovulation rate
WO2001053463A2 (en) 2000-01-21 2001-07-26 Corixa Corporation COMPOUNDS AND METHODS FOR PREVENTION AND TREATMENT OF HER-2/neu ASSOCIATED MALIGNANCIES
WO2001057188A2 (en) 2000-02-03 2001-08-09 Hyseq, Inc. Novel nucleic acids and polypeptides
WO2001062794A2 (en) 2000-02-22 2001-08-30 Millennium Pharmaceuticals, Inc. 18607, a human calcium channel
WO2001066689A2 (en) 2000-03-07 2001-09-13 Hyseq, Inc. Novel nucleic acids and polypeptides
WO2001072962A2 (en) 2000-03-24 2001-10-04 Fahri Saatcioglu Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
WO2001072830A2 (en) 2000-03-31 2001-10-04 Ipf Pharmaceuticals Gmbh Diagnostic and medicament for analysing the cell surface proteome of tumour and inflammatory cells and for treating tumorous and inflammatory diseases, preferably using a specific chemokine receptor analysis and the chemokine receptor-ligand interaction
WO2001075177A2 (en) 2000-04-03 2001-10-11 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tumor markers in ovarian cancer
WO2001077172A2 (en) 2000-04-07 2001-10-18 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated known g protein-coupled receptors
WO2001088133A2 (en) 2000-05-18 2001-11-22 Lexicon Genetics Incorporated Human semaphorin homologs and polynucleotides encoding the same
WO2001090304A2 (en) 2000-05-19 2001-11-29 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
WO2001094641A2 (en) 2000-06-09 2001-12-13 Idec Pharmaceuticals Corporation Gene targets and ligands that bind thereto for treatment and diagnosis of ovarian carcinomas
WO2001098351A2 (en) 2000-06-16 2001-12-27 Incyte Genomics, Inc. G-protein coupled receptors
US20010055751A1 (en) 1997-03-10 2001-12-27 Reiter Robert E PSCA: Prostate stem cell antigen and uses thereof
WO2002002634A2 (en) 2000-06-30 2002-01-10 Incyte Genomics, Inc. Human extracellular matrix and cell adhesion polypeptides
WO2002002624A2 (en) 2000-06-30 2002-01-10 Amgen, Inc. B7-like molecules and uses thereof
WO2002002587A1 (en) 2000-06-30 2002-01-10 Human Genome Sciences, Inc. B7-like polynucleotides, polypeptides, and antibodies
WO2002006339A2 (en) 2000-07-03 2002-01-24 Curagen Corporation Proteins and nucleic acids encoding same
WO2002006317A2 (en) 2000-07-17 2002-01-24 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2002010187A1 (en) 2000-07-27 2002-02-07 Mayo Foundation For Medical Education And Research B7-h3 and b7-h4, novel immunoregulatory molecules
WO2002010382A2 (en) 2000-07-28 2002-02-07 Ulrich Wissenbach Trp8, trp9 and trp10, markers for cancer
WO2002012341A2 (en) 2000-08-03 2002-02-14 Corixa Corporation Her-2/neu fusion proteins
WO2002013847A2 (en) 2000-08-14 2002-02-21 Corixa Corporation Methods for diagnosis and therapy of hematological and virus-associated malignancies
WO2002014503A2 (en) 2000-08-14 2002-02-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of her-2/neu-associated malignancies
WO2002016413A2 (en) 2000-08-24 2002-02-28 Glaxosmithkline Biologicals S.A. Cripto tumour polypeptide
WO2002016429A2 (en) 2000-08-24 2002-02-28 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2002018444A2 (en) 2000-09-01 2002-03-07 Genentech, Inc. Erbb4 antagonists
WO2002022808A2 (en) 2000-09-18 2002-03-21 Biogen, Inc. Cripto mutant and uses thereof
WO2002022636A1 (en) 2000-09-15 2002-03-21 Isis Pharmaceuticals, Inc. Antisense modulation of her-2 expression
WO2002022660A2 (en) 2000-09-11 2002-03-21 Hyseq, Inc. Novel nucleic acids and polypeptides
WO2002022153A2 (en) 2000-09-15 2002-03-21 Zymogenetics, Inc. Use of a polypeptide comprising the extracellular domains of il-20rb for the treatment of inflammation
US20020034749A1 (en) 1997-11-18 2002-03-21 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
WO2002024909A2 (en) 2000-09-18 2002-03-28 Biogen, Inc. Receptor nucleic acids and polypeptides
US20020042366A1 (en) 1999-12-23 2002-04-11 Penny Thompson Method for treating inflammation
US6372907B1 (en) 1999-11-03 2002-04-16 Apptera Corporation Water-soluble rhodamine dye peptide conjugates
WO2002030268A2 (en) 2000-10-13 2002-04-18 Eos Biotechnology, Inc. Methods of diagnosis of prostate cancer, compositions and methods of screening for modulators of prostate cancer
WO2002038766A2 (en) 2000-11-07 2002-05-16 Zymogenetics, Inc. Human tumor necrosis factor receptor
US6399743B1 (en) 1999-05-14 2002-06-04 Dept. Of Veterans Affairs Isolation and characterization of a rat epidermal growth factor related protein
WO2002054940A2 (en) 2001-01-12 2002-07-18 University Of Medicine & Dentistry Of New Jersey Bone morphogenetic protein-2 in the treatment and diagnosis of cancer
WO2002059377A2 (en) 2001-01-24 2002-08-01 Protein Design Labs Methods of diagnosis of breast cancer, compositions and methods of screening for modulators of breast cancer
WO2002060317A2 (en) 2001-01-30 2002-08-08 Corixa Corporation Compositions and methods for the therapy and diagnosis of pancreatic cancer
WO2002061087A2 (en) 2000-12-19 2002-08-08 Lifespan Biosciences, Inc. Antigenic peptides, such as for g protein-coupled receptors (gpcrs), antibodies thereto, and systems for identifying such antigenic peptides
WO2002064798A1 (en) 2001-02-12 2002-08-22 Bionomics Limited Dna sequences differentially expressed in tumour cell lines
WO2002072596A1 (en) 2001-03-09 2002-09-19 Incyte Genomics, Inc. Steap-related protein
WO2002071928A2 (en) 2001-03-14 2002-09-19 Millennium Pharmaceuticals, Inc. Nucleic acid molecules and proteins for the identification, assessment, prevention, and therapy of ovarian cancer
WO2002078524A2 (en) 2001-03-28 2002-10-10 Zycos Inc. Translational profiling
US20020150573A1 (en) 2000-11-10 2002-10-17 The Rockefeller University Anti-Igalpha-Igbeta antibody for lymphoma therapy
WO2002081646A2 (en) 2001-04-06 2002-10-17 Mannkind Corporation Epitope sequences
WO2002083866A2 (en) 2001-04-17 2002-10-24 The Board Of Trustees Of The University Of Arkansas Repeat sequences of the ca125 gene and their use for diagnostic and therapeutic interventions
WO2002086443A2 (en) 2001-04-18 2002-10-31 Protein Design Labs, Inc Methods of diagnosis of lung cancer, compositions and methods of screening for modulators of lung cancer
WO2002088170A2 (en) 2001-04-26 2002-11-07 Biogen, Inc. Cripto blocking antibodies and uses thereof
WO2002088172A2 (en) 2001-04-30 2002-11-07 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
WO2002089747A2 (en) 2001-05-09 2002-11-14 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
WO2002092836A2 (en) 2001-05-11 2002-11-21 Sloan-Kettering Institute For Cancer Research Nucleic acid sequence encoding ovarian antigen, ca125, and uses thereof
WO2002094852A2 (en) 2001-05-24 2002-11-28 Zymogenetics, Inc. Taci-immunoglobulin fusion proteins
WO2002099074A2 (en) 2001-06-05 2002-12-12 Exelixis, Inc. Slc7s as modifiers of the p53 pathway and methods of use
WO2002098358A2 (en) 2001-06-04 2002-12-12 Eos Biotechnology, Inc. Methods of diagnosis and treatment of androgen-dependent prostate cancer, prostate cancer undergoing androgen-withdrawal, and androgen-independent prostate cancer
WO2002099122A1 (en) 2001-06-05 2002-12-12 Exelixis, Inc. Modifiers of the p53 pathway and methods of use
WO2002101075A2 (en) 2001-06-13 2002-12-19 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
US20020193567A1 (en) 1995-08-11 2002-12-19 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
WO2002102235A2 (en) 2001-06-18 2002-12-27 Eos Biotechnology Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
WO2003000842A2 (en) 2001-06-04 2003-01-03 Curagen Corporation Novel proteins and nucleic acids encoding same
WO2003002717A2 (en) 2001-06-28 2003-01-09 Schering Corporation Biological activity of ak155
WO2003004989A2 (en) 2001-06-21 2003-01-16 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
WO2003003984A2 (en) 2001-07-05 2003-01-16 Curagen Corporation Novel proteins and nucleic acids encoding same
WO2003004529A2 (en) 2001-07-02 2003-01-16 Licentia Ltd. Ephrin-tie receptor materials and methods
WO2003003906A2 (en) 2001-07-03 2003-01-16 Eos Biotechnology, Inc. Diagnostic and screening methods for bladder cancer
WO2003009814A2 (en) 2001-07-25 2003-02-06 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of prostate cancer
US6518404B1 (en) 1994-10-17 2003-02-11 Human Genome Sciences, Inc. Human endothelin-bombesin receptor antibodies
WO2003014294A2 (en) 2001-08-03 2003-02-20 Genentech, Inc. Tacis and br3 polypeptides and uses thereof
WO2003016475A2 (en) 2001-08-14 2003-02-27 The General Hospital Corporation Nucleic acid and amino acid sequences involved in pain
WO2003016494A2 (en) 2001-08-16 2003-02-27 Vitivity, Inc. Diagnosis and treatment of vascular disease
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
WO2003018621A2 (en) 2001-08-23 2003-03-06 Oxford Biomedica (Uk) Limited Genes
WO2003023013A2 (en) 2001-09-13 2003-03-20 Nuvelo, Inc. Novel nucleic acids and polypeptides
WO2003022995A2 (en) 2001-09-06 2003-03-20 Agensys, Inc. Nucleic acid and corresponding protein entitled steap-1 useful in treatment and detection of cancer
EP1295944A2 (en) 1996-03-19 2003-03-26 Otsuka Pharmaceutical Co., Ltd. GDP dissociation stimulating protein, brain-specific nucleosome assembly protein, skeletal muscle specific ubiquitin-conjugating enzyme, cell proliferation protein, phosphatidylinositolkinase, nel related proteins
WO2003025138A2 (en) 2001-09-17 2003-03-27 Protein Design Labs, Inc. Methods of diagnosis of cancer compositions and methods of screening for modulators of cancer
WO2003025148A2 (en) 2001-09-19 2003-03-27 Nuvelo, Inc. Novel nucleic acids and polypeptides
WO2003025228A1 (en) 2001-09-18 2003-03-27 Proteologics, Inc. Methods and compositions for treating hcap associated diseases
WO2003024392A2 (en) 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20030060612A1 (en) 1997-10-28 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2003026493A2 (en) 2001-09-28 2003-04-03 Bing Yang Diagnosis and treatment of diseases caused by mutations in cd72
WO2003026577A2 (en) 2001-09-24 2003-04-03 Seattle Genetics, Inc. P-amidobenzylethers in drug delivery agents
US20030064397A1 (en) 1998-05-22 2003-04-03 Incyte Genomics, Inc. Transmembrane protein differentially expressed in prostate and lung tumors
US20030065143A1 (en) 1998-12-30 2003-04-03 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2003029262A2 (en) 2001-08-29 2003-04-10 Vanderbilt University The human mob-5 (il-24) receptors and uses thereof
WO2003029421A2 (en) 2001-10-03 2003-04-10 Origene Technologies, Inc. Regulated breast cancer genes
WO2003029277A2 (en) 2001-10-03 2003-04-10 Rigel Pharmaceuticals, Inc. Modulators of lymphocyte activation and migration
WO2003034984A2 (en) 2001-10-19 2003-05-01 Genentech, Inc. Compositions and methods for the diagnosis and treatment of inflammatory bowel disorders
WO2003035846A2 (en) 2001-10-24 2003-05-01 National Jewish Medical And Research Center Structure of tall-1 and its cognate receptor
US20030091580A1 (en) 2001-06-18 2003-05-15 Mitcham Jennifer L. Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030096373A1 (en) 1999-05-14 2003-05-22 Majumdar Adhip P. N. Antibodies to a novel EGF-receptor related protein (ERRP)
US20030096961A1 (en) 2001-06-01 2003-05-22 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030096743A1 (en) 2001-09-24 2003-05-22 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
WO2003042661A2 (en) 2001-11-13 2003-05-22 Protein Design Labs, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
WO2003043583A2 (en) 2001-11-20 2003-05-30 Seattle Genetics, Inc. Treatment of immunological disorders using anti-cd30 antibodies
WO2003045422A1 (en) 2001-11-29 2003-06-05 Genset S.A. Agonists and antagonists of prolixin for the treatment of metabolic disorders
WO2003048202A2 (en) 2001-12-03 2003-06-12 Asahi Kasei Pharma Corporation Nf-kappab activating genes
US20030119126A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119128A1 (en) 1999-07-20 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119130A1 (en) 1999-08-17 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119122A1 (en) 1999-05-11 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119129A1 (en) 1999-08-10 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119131A1 (en) 2000-01-20 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030118592A1 (en) 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20030119125A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119121A1 (en) 2000-09-15 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030124140A1 (en) 1998-12-17 2003-07-03 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030124579A1 (en) 2001-09-05 2003-07-03 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
WO2003054152A2 (en) 2001-12-10 2003-07-03 Nuvelo, Inc. Novel nucleic acids and polypeptides
WO2003055439A2 (en) 2001-07-18 2003-07-10 The Regents Of The University Of California Her2/neu target antigen and use of same to stimulate an immune response
WO2003055443A2 (en) 2001-10-31 2003-07-10 Alcon, Inc. Bone morphogenic proteins (bmp), bmp receptors and bmp binding proteins and their use in the diagnosis and treatment of glaucoma
US20030129192A1 (en) 1999-09-10 2003-07-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030134790A1 (en) 2002-01-11 2003-07-17 University Of Medicine And Dentistry Of New Jersey Bone Morphogenetic Protein-2 And Bone Morphogenetic Protein-4 In The Treatment And Diagnosis Of Cancer
WO2003062401A2 (en) 2002-01-22 2003-07-31 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US20030143557A1 (en) 2002-01-25 2003-07-31 Reinhold Penner Methods of screening for TRPM4b modulators
US20030157089A1 (en) 1997-02-25 2003-08-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
US20030165504A1 (en) 1999-09-24 2003-09-04 Retter Marc W. Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2003072036A2 (en) 2002-02-21 2003-09-04 Duke University Treatment methods using anti-cd22 antibodies
WO2003072035A2 (en) 2002-02-22 2003-09-04 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
EP1347046A1 (en) 2002-03-22 2003-09-24 Research Association for Biotechnology Full-length cDNA sequences
WO2003077836A2 (en) 2001-11-06 2003-09-25 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US20030186372A1 (en) 2000-02-11 2003-10-02 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030185830A1 (en) 1997-02-25 2003-10-02 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
WO2003081210A2 (en) 2002-03-21 2003-10-02 Sunesis Pharmaceuticals, Inc. Identification of kinase inhibitors
WO2003083041A2 (en) 2002-03-22 2003-10-09 Biogen, Inc. Cripto-specific antibodies
WO2003083074A2 (en) 2002-03-28 2003-10-09 Idec Pharmaceuticals Corporation Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
WO2003083047A2 (en) 2002-03-01 2003-10-09 Exelixis, Inc. MP53s AS MODIFIERS OF THE p53 PATHWAY AND METHODS OF USE
US20030194704A1 (en) 2002-04-03 2003-10-16 Penn Sharron Gaynor Human genome-derived single exon nucleic acid probes useful for gene expression analysis two
WO2003087768A2 (en) 2002-04-12 2003-10-23 Mitokor Targets for therapeutic intervention identified in the mitochondrial proteome
WO2003087306A2 (en) 2002-04-05 2003-10-23 Agensys, Inc. Nucleic acid and corresponding protein entitled 98p4b6 useful in treatment and detection of cancer
WO2003089624A2 (en) 2002-03-25 2003-10-30 Uab Research Foundation Fc receptor homolog, reagents, and uses thereof
WO2003088808A2 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2003089904A2 (en) 2002-04-17 2003-10-30 Baylor College Of Medicine Aib1 as a prognostic marker and predictor of resistance to encocrine therapy
US20030206918A1 (en) 1999-09-10 2003-11-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2003093444A2 (en) 2002-05-03 2003-11-13 Incyte Corporation Transporters and ion channels
WO2003097803A2 (en) 2002-05-15 2003-11-27 Avalon Pharmaceuticals Cancer-linked gene as target for chemotherapy
US20030219806A1 (en) 2000-02-22 2003-11-27 Millennium Pharmaceuticals, Inc. Novel 18607, 15603, 69318, 12303, 48000, 52920, 5433, 38554, 57301, 58324, 55063, 52991, 59914, 59921 and 33751 molecules and uses therefor
US20030224411A1 (en) 2003-03-13 2003-12-04 Stanton Lawrence W. Genes that are up- or down-regulated during differentiation of human embryonic stem cells
US20030224454A1 (en) 2002-05-30 2003-12-04 Ryseck Rolf Peter Human solute carrier family 7, member 11 (hSLC7A11)
WO2003101400A2 (en) 2002-06-04 2003-12-11 Avalon Pharmaceuticals, Inc. Cancer-linked gene as target for chemotherapy
WO2003101283A2 (en) 2002-06-04 2003-12-11 Incyte Corporation Diagnostics markers for lung cancer
US20030232056A1 (en) 1999-09-10 2003-12-18 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2003104270A2 (en) 2002-06-06 2003-12-18 Ingenium Pharmaceuticals Ag Dudulin 2 genes, expression products, non-human animal model: uses in human hematological disease
US20030232350A1 (en) 2001-11-13 2003-12-18 Eos Biotechnology, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
WO2003104399A2 (en) 2002-06-07 2003-12-18 Avalon Pharmaceuticals, Inc Cancer-linked gene as target for chemotherapy
WO2003104275A2 (en) 2002-06-06 2003-12-18 Oncotherapy Science, Inc. Genes and polypeptides relating to human colon cancers
WO2003105758A2 (en) 2002-06-12 2003-12-24 Avalon Pharmaceuticals, Inc. Cancer-linked gene as target for chemotherapy
WO2004001004A2 (en) 2002-06-21 2003-12-31 Johns Hopkins University School Of Medicine Membrane associated tumor endothelium markers
WO2004000997A2 (en) 2002-03-19 2003-12-31 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2004001993A1 (en) 2002-06-20 2003-12-31 Snaptrack Incorporated Reducing cross-interference in a combined gps receiver and communication system
WO2004000221A2 (en) 2002-06-20 2003-12-31 The Regents Of The University Of California Compositions and methods for modulating lymphocyte activity
US20040001827A1 (en) 2002-06-28 2004-01-01 Dennis Mark S. Serum albumin binding peptides for tumor targeting
US20040005538A1 (en) 2001-04-11 2004-01-08 Xiaojiang Chen Three-dimensional structure of complement receptor type 2 and uses thereof
US20040005563A1 (en) 2001-06-18 2004-01-08 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
WO2004009622A2 (en) 2002-07-19 2004-01-29 Cellzome Ag Protein complexes of cellular networks underlying the development of cancer and other diseases
US20040022727A1 (en) 2002-06-18 2004-02-05 Martin Stanton Aptamer-toxin molecules and methods for using same
WO2004011611A2 (en) 2002-07-25 2004-02-05 Genentech, Inc. Taci antibodies and uses thereof
WO2004015426A1 (en) 2002-08-06 2004-02-19 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with human cxc chemokine receptor 5(cxcr5)
WO2004016225A2 (en) 2002-08-19 2004-02-26 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1394274A2 (en) 2002-08-06 2004-03-03 Genox Research, Inc. Methods of testing for bronchial asthma or chronic obstructive pulmonary disease
US20040044179A1 (en) 2000-07-25 2004-03-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2004020595A2 (en) 2002-08-29 2004-03-11 Five Prime Therapeutics, Inc. Novel human polypeptides encoded by polynucleotides
WO2004020583A2 (en) 2002-08-27 2004-03-11 Bristol-Myers Squibb Company Polynucleotide predictor set for identifying protein tyrosine kinase modulators
WO2004022709A2 (en) 2002-09-06 2004-03-18 Mannkind Corporation Epitope sequences
WO2004022778A1 (en) 2002-09-05 2004-03-18 Garvan Institute Of Medical Research Methods of diagnosis and prognosis of ovarian cancer
WO2004027049A2 (en) 2002-09-20 2004-04-01 Astral, Inc. Methods and compositions to generate and control the effector profile of t cells by simultaneous loading and activation of selected subsets of antigen presenting cells
JP2004113151A (en) 2002-09-27 2004-04-15 Sankyo Co Ltd Oncogene and its application
WO2004031238A2 (en) 2002-10-03 2004-04-15 Mcgill Univeristy Antibodies and cyclic peptides which bind cea (carcinoembryonic antigen) and their use as cancer therapeutics
WO2004032828A2 (en) 2002-07-31 2004-04-22 Seattle Genetics, Inc. Anti-cd20 antibody-drug conjugates for the treatment of cancer and immune disorders
WO2004032842A2 (en) 2002-10-04 2004-04-22 Van Andel Research Institute Molecular sub-classification of kidney tumors and the discovery of new diagnostic markers
WO2004040000A2 (en) 2002-09-09 2004-05-13 Nura, Inc G protein coupled receptors and uses thereof
WO2004042346A2 (en) 2002-04-24 2004-05-21 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
WO2004044178A2 (en) 2002-11-13 2004-05-27 Genentech, Inc. Methods and compositions for diagnosing dysplasia
WO2004043361A2 (en) 2002-11-08 2004-05-27 Genentech, Inc. Compositions and methods for the treatment of natural killer cell related diseases
WO2004046342A2 (en) 2002-11-20 2004-06-03 Biogen Idec Inc. Novel gene targets and ligands that bind thereto for treatment and diagnosis of carcinomas
WO2004045520A2 (en) 2002-11-15 2004-06-03 Musc Foundation For Research Development Complement receptor 2 targeted complement modulators
WO2004045516A2 (en) 2002-11-15 2004-06-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2004045553A2 (en) 2002-11-15 2004-06-03 The Board Of Trustees Of The University Of Arkansas Ca125 gene and its use for diagnostic and therapeutic interventions
WO2004047749A2 (en) 2002-11-21 2004-06-10 University Of Utah Research Foundation Purinergic modulation of smell
WO2004048938A2 (en) 2002-11-26 2004-06-10 Protein Design Labs, Inc. Methods of detecting soft tissue sarcoma, compositions and methods of screening for soft tissue sarcoma modulators
WO2004053079A2 (en) 2002-12-06 2004-06-24 Diadexus, Inc. Compositions, splice variants and methods relating to ovarian specific genes and proteins
WO2004058309A1 (en) 2002-12-23 2004-07-15 Human Genome Sciences, Inc. Neutrokine-alpha conjugate, neutrokine-alpha complex, and uses thereof
EP1439393A2 (en) 2002-12-13 2004-07-21 Bayer Healthcare LLC Detection methods using TIMP 1 for colon cancer diagnosis
WO2004063709A2 (en) 2003-01-08 2004-07-29 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
WO2004063355A2 (en) 2003-01-10 2004-07-29 Protein Design Labs, Inc. Novel methods of diagnosis of metastatic cancer, compositions and methods of screening for modulators of matastatic cancer
WO2004063362A2 (en) 2003-01-10 2004-07-29 Cyclacel Limited Cell cycle progression proteins
WO2004065576A2 (en) 2003-01-15 2004-08-05 Millennium Pharmaceuticals, Inc. Methods and compositions for the treatment of urological disorder using differential expressed polypeptides
WO2004065577A2 (en) 2003-01-14 2004-08-05 Bristol-Myers Squibb Company Polynucleotides and polypeptides associated with the nf-kb pathway
WO2004074320A2 (en) 2003-02-14 2004-09-02 Sagres Discovery, Inc. Therapeutic targets in cancer
US20040197325A1 (en) 2002-12-20 2004-10-07 Debbie Law Antibodies against GPR64 and uses thereof
US20040229310A1 (en) 2003-01-23 2004-11-18 Genentech, Inc. Methods for producing humanized antibodies and improving yield of antibodies or antigen binding fragments in cell culture
US20040249130A1 (en) 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
US20050048572A1 (en) 2002-10-31 2005-03-03 Genentech, Inc. Methods and compositions for increasing antibody production
WO2006034488A2 (en) * 2004-09-23 2006-03-30 Genentech, Inc. Cysteine engineered antibodies and conjugates
US20080050310A1 (en) 2006-05-30 2008-02-28 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
US20080247951A1 (en) 2007-02-09 2008-10-09 Genentech, Inc. Anti-robo4 antibodies and uses therefor
WO2008141044A2 (en) * 2007-05-08 2008-11-20 Genentech, Inc. Cysteine engineered anti-muc16 antibodies and antibody drug conjugates
US20090028856A1 (en) 2007-07-16 2009-01-29 Genentech, Inc. Anti-CD79B Antibodies and Immunoconjugates and Methods of Use
US20090068202A1 (en) 2007-07-16 2009-03-12 Genentech, Inc. Humanized Anti-CD79B Antibodies and Immunoconjugates and Methods of Use
US20090117100A1 (en) 2007-10-19 2009-05-07 Weiguang Mao Cysteine engineered anti-TENB2 antibodies and antibody drug conjugates

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8720833D0 (en) * 1987-09-04 1987-10-14 Celltech Ltd Recombinant dna product
AU7075496A (en) * 1995-09-18 1997-04-09 Intracel Corporation Neutralizing monoclonal antibodies to respiratory syncytial virus
US6753165B1 (en) * 1999-01-14 2004-06-22 Bolder Biotechnology, Inc. Methods for making proteins containing free cysteine residues
US7097840B2 (en) * 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
US20040235068A1 (en) * 2001-09-05 2004-11-25 Levinson Arthur D. Methods for the identification of polypeptide antigens associated with disorders involving aberrant cell proliferation and compositions useful for the treatment of such disorders

Patent Citations (307)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4318980A (en) 1978-04-10 1982-03-09 Miles Laboratories, Inc. Heterogenous specific binding assay employing a cycling reactant as label
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JPH053790B2 (en) 1983-11-02 1993-01-18 Canon Kk
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5428155A (en) 1984-10-18 1995-06-27 Board Of Regents, The University Of Texas System Synthesis of polyazamacrocycles with more than one type of side-chain chelating groups
US5316757A (en) 1984-10-18 1994-05-31 Board Of Regents, The University Of Texas System Synthesis of polyazamacrocycles with more than one type of side-chain chelating groups
US5342606A (en) 1984-10-18 1994-08-30 Board Of Regents, The University Of Texas System Polyazamacrocyclic compounds for complexation of metal ions
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US5091178A (en) 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5677171A (en) 1988-01-12 1997-10-14 Genentech, Inc. Monoclonal antibodies directed to the HER2 receptor
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1991002536A1 (en) 1989-08-23 1991-03-07 Scripps Clinic And Research Foundation Compositions and methods for detection and treatment of epstein-barr virus infection and immune disorders
US5183884A (en) 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
US5480968A (en) 1989-12-01 1996-01-02 The United States Of America As Represented By The Department Of Health And Human Services Isolated polypeptide erbB-3, related to the epidermal growth factor receptor and antibody thereto
US5792616A (en) 1990-05-29 1998-08-11 The United States Of America Antibodies to human cripto protein
WO1992007574A1 (en) 1990-10-25 1992-05-14 Tanox Biosystems, Inc. Glycoproteins associated with membrane-bound immunoglobulins as antibody targets on b cells
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5440021A (en) 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
WO1992017497A1 (en) 1991-03-29 1992-10-15 Genentech, Inc. Human pf4a receptors and their use
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US6407213B1 (en) 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
EP0522868A1 (en) 1991-07-12 1993-01-13 SHIONOGI SEIYAKU KABUSHIKI KAISHA trading under the name of SHIONOGI &amp; CO. LTD. A human endothelin receptor
US5854399A (en) 1991-08-23 1998-12-29 The United States Of America As Represented By The Department Of Health And Human Services Antibodies specific for human cripto-related polypeptide CR-3
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US6153408A (en) 1991-11-15 2000-11-28 Institut Pasteur And Institut National De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
US5976551A (en) 1991-11-15 1999-11-02 Institut Pasteur And Institut Nationale De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and method of using the determinant
US6011146A (en) 1991-11-15 2000-01-04 Institut Pasteur Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
WO1993011161A1 (en) 1991-11-25 1993-06-10 Enzon, Inc. Multivalent antigen-binding proteins
US5480990A (en) 1991-12-10 1996-01-02 The Dow Chemical Company Bicyclopolyazamacrocyclocarboxylic acid complexes for use as contrast agents
US5750660A (en) 1991-12-10 1998-05-12 The Dow Chemical Company Bicyclopolyazamacrocyclophosphonic acid half esters
US5739294A (en) 1991-12-10 1998-04-14 The Dow Chemical Company Bicyclopol yazamacrocyclophosphonic acid complexes for use as contrast agents
US5428139A (en) 1991-12-10 1995-06-27 The Dow Chemical Company Bicyclopolyazamacrocyclophosphonic acid complexes for use as radiopharmaceuticals
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1993021319A1 (en) 1992-04-08 1993-10-28 Cetus Oncology Corporation HUMANIZED C-erbB-2 SPECIFIC ANTIBODIES
WO1994010312A1 (en) 1992-10-23 1994-05-11 Chugai Seiyaku Kabushiki Kaisha Gene coding for megakaryocyte potentiator
WO1994011026A2 (en) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
EP0599274A1 (en) 1992-11-24 1994-06-01 Bristol-Myers Squibb Company HER4, a human receptor tyrosine kinase of the epidermal growth factor receptor family
US5644033A (en) 1992-12-22 1997-07-01 Health Research, Inc. Monoclonal antibodies that define a unique antigen of human B cell antigen receptor complex and methods of using same for diagnosis and treatment
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
US5462725A (en) 1993-05-06 1995-10-31 The Dow Chemical Company 2-pyridylmethylenepolyazamacrocyclophosphonic acids, complexes and derivatives thereof, for use as contrast agents
US5385893A (en) 1993-05-06 1995-01-31 The Dow Chemical Company Tricyclopolyazamacrocyclophosphonic acids, complexes and derivatives thereof, for use as contrast agents
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
WO1994028931A1 (en) 1993-06-11 1994-12-22 Genentech, Inc. Methods for treating inflammatory disorders
US5773223A (en) 1993-09-02 1998-06-30 Chiron Corporation Endothelin B1, (ETB1) receptor polypeptide and its encoding nucleic acid methods, and uses thereof
WO1995025167A1 (en) 1994-03-17 1995-09-21 Merck Patent Gmbh Anti-egfr single-chain fvs and anti-egfr antibodies
US6518404B1 (en) 1994-10-17 2003-02-11 Human Genome Sciences, Inc. Human endothelin-bombesin receptor antibodies
US20030109676A1 (en) 1994-10-17 2003-06-12 Human Genome Sciences, Inc. Human endothelin-bombesin receptor
WO1996030514A1 (en) 1995-03-31 1996-10-03 University Of Washington Intracellular domain of the her-2/neu protein for prevention or treatment of malignancies
WO1997004801A1 (en) 1995-07-27 1997-02-13 Genentech, Inc. Stabile isotonic lyophilized protein formulation
WO1997007198A2 (en) 1995-08-11 1997-02-27 Genetics Institute, Inc. Dna sequences and secreted proteins encoded thereby
US20020193567A1 (en) 1995-08-11 2002-12-19 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
US5834456A (en) 1996-02-23 1998-11-10 The Dow Chemical Company Polyazamacrocyclofluoromonoalkylphosphonic acids, and their complexes, for use as contrast agents
EP1295944A2 (en) 1996-03-19 2003-03-26 Otsuka Pharmaceutical Co., Ltd. GDP dissociation stimulating protein, brain-specific nucleosome assembly protein, skeletal muscle specific ubiquitin-conjugating enzyme, cell proliferation protein, phosphatidylinositolkinase, nel related proteins
WO1997035885A1 (en) 1996-03-27 1997-10-02 Genentech, Inc. ErbB3 ANTIBODIES
WO1997044452A1 (en) 1996-05-17 1997-11-27 Schering Corporation Human b-cell antigens, related reagents
WO1998037193A1 (en) 1997-02-20 1998-08-27 Zymogenetics, Inc. Zcytor7 cytokine receptor
US20030185830A1 (en) 1997-02-25 2003-10-02 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
US20030157089A1 (en) 1997-02-25 2003-08-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
US20010055751A1 (en) 1997-03-10 2001-12-27 Reiter Robert E PSCA: Prostate stem cell antigen and uses thereof
WO1998040403A1 (en) 1997-03-10 1998-09-17 The Regents Of The University Of California Psca: prostate stem cell antigen
US20030105292A1 (en) 1997-04-14 2003-06-05 Liaw Chen W. Non-endogenous, constitutively activated human G protein-coupled receptors
US6555339B1 (en) 1997-04-14 2003-04-29 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human protein-coupled receptors
EP0875569A1 (en) 1997-04-28 1998-11-04 Smithkline Beecham Corporation A human sodium dependent phosphate transporter (IPT-1)
US20040018553A1 (en) 1997-05-15 2004-01-29 Patricia A. Billing-Medel Reagents and methods useful for detecting diseases of the prostate
WO1998051805A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting diseases of the prostate
WO1998051824A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting disease of the urinary tract
US6248564B1 (en) 1997-08-29 2001-06-19 Harvard University Mutant MHC class I molecules
WO1999019488A1 (en) 1997-10-15 1999-04-22 Children's Medical Center Corporation Novel human egf receptors and use thereof
US20030060612A1 (en) 1997-10-28 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20020034749A1 (en) 1997-11-18 2002-03-21 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
WO1999028468A1 (en) 1997-12-02 1999-06-10 The Regents Of The University Of California Modulating b lymphocyte chemokine/receptor interactions
WO1999046284A2 (en) 1998-03-13 1999-09-16 The Burnham Institute Molecules that home to various selected organs or tissues
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6534482B1 (en) 1998-05-13 2003-03-18 Epimmune, Inc. Expression vectors for stimulating an immune response and methods of using the same
WO1999058658A2 (en) 1998-05-13 1999-11-18 Epimmune, Inc. Expression vectors for stimulating an immune response and methods of using the same
US20030064397A1 (en) 1998-05-22 2003-04-03 Incyte Genomics, Inc. Transmembrane protein differentially expressed in prostate and lung tumors
WO2000012130A1 (en) 1998-08-27 2000-03-09 Smithkline Beecham Corporation Rp105 agonists and antagonists
WO2000014228A1 (en) 1998-09-03 2000-03-16 Japan Science And Technology Corporation Neutral amino acid transporter and gene thereof
WO2000020579A1 (en) 1998-10-02 2000-04-13 Mcmaster University Spliced form of erbb-2/neu oncogene
WO2000022129A1 (en) 1998-10-13 2000-04-20 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human g protein-coupled receptors
WO2000032752A1 (en) 1998-12-02 2000-06-08 The Regents Of The University Of California Psca: prostate stem cell antigen and uses thereof
WO2000036107A2 (en) 1998-12-17 2000-06-22 Corixa Corporation Compositions and methods for therapy and diagnosis of ovarian cancer
US20030124140A1 (en) 1998-12-17 2003-07-03 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2000040614A2 (en) 1998-12-30 2000-07-13 Beth Israel Deaconess Medical Center, Inc. Characterization of the soc/crac calcium channel protein family
US20030065143A1 (en) 1998-12-30 2003-04-03 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2000044899A1 (en) 1999-01-29 2000-08-03 Corixa Corporation Her-2/neu fusion proteins
WO2000053216A2 (en) 1999-03-05 2000-09-14 Smithkline Beecham Biologicals S.A. Use of casb616 polypeptides and polynucleotides for cancer treatment
WO2000055351A1 (en) 1999-03-12 2000-09-21 Human Genome Sciences, Inc. Human colon cancer associated gene sequences and polypeptides
US20030119122A1 (en) 1999-05-11 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6399743B1 (en) 1999-05-14 2002-06-04 Dept. Of Veterans Affairs Isolation and characterization of a rat epidermal growth factor related protein
US20030096373A1 (en) 1999-05-14 2003-05-22 Majumdar Adhip P. N. Antibodies to a novel EGF-receptor related protein (ERRP)
WO2000075655A1 (en) 1999-06-03 2000-12-14 Takeda Chemical Industries, Ltd. Screening method with the use of cd100
WO2001000245A2 (en) 1999-06-25 2001-01-04 Genentech, Inc. HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
WO2001000244A2 (en) 1999-06-25 2001-01-04 Genentech, Inc. METHODS OF TREATMENT USING ANTI-ErbB ANTIBODY-MAYTANSINOID CONJUGATES
US20030119128A1 (en) 1999-07-20 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119129A1 (en) 1999-08-10 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119130A1 (en) 1999-08-17 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001016318A2 (en) 1999-09-01 2001-03-08 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030129192A1 (en) 1999-09-10 2003-07-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030206918A1 (en) 1999-09-10 2003-11-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030232056A1 (en) 1999-09-10 2003-12-18 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030165504A1 (en) 1999-09-24 2003-09-04 Retter Marc W. Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2001040309A2 (en) 1999-10-29 2001-06-07 Genentech, Inc. Anti-prostate stem cell antigen (psca) antibody compositions and methods of use
US6372907B1 (en) 1999-11-03 2002-04-16 Apptera Corporation Water-soluble rhodamine dye peptide conjugates
WO2001038490A2 (en) 1999-11-29 2001-05-31 The Trustees Of Columbia University In The City Of New York ISOLATION OF FIVE NOVEL GENES CODING FOR NEW Fc RECEPTORS-TYPE MELANOMA INVOLVED IN THE PATHOGENESIS OF LYMPHOMA/MELANOMA
WO2001040269A2 (en) 1999-11-30 2001-06-07 Corixa Corporation Compositions and methods for therapy and diagnosis of breast cancer
US20040101899A1 (en) 1999-11-30 2004-05-27 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
WO2001041787A1 (en) 1999-12-10 2001-06-14 Epimmune Inc. INDUCING CELLULAR IMMUNE RESPONSES TO HER2/neu USING PEPTIDE AND NUCLEIC ACID COMPOSITIONS
WO2001046261A1 (en) 1999-12-23 2001-06-28 Zymogenetics, Inc. Method for treating inflammation
WO2001048204A1 (en) 1999-12-23 2001-07-05 Agresearch Limited Mutated bmp1b receptor as regulator of ovulation rate
WO2001046232A2 (en) 1999-12-23 2001-06-28 Zymogenetics, Inc. Soluble interleukin-20 receptor
US20020042366A1 (en) 1999-12-23 2002-04-11 Penny Thompson Method for treating inflammation
US20040005320A1 (en) 1999-12-23 2004-01-08 Penny Thompson Method for treating inflammation
WO2001045746A2 (en) 1999-12-24 2001-06-28 Genentech, Inc. Methods and compositions for prolonging elimination half-times of bioactive compounds
US20030119131A1 (en) 2000-01-20 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001053463A2 (en) 2000-01-21 2001-07-26 Corixa Corporation COMPOUNDS AND METHODS FOR PREVENTION AND TREATMENT OF HER-2/neu ASSOCIATED MALIGNANCIES
WO2001057188A2 (en) 2000-02-03 2001-08-09 Hyseq, Inc. Novel nucleic acids and polypeptides
US20030186372A1 (en) 2000-02-11 2003-10-02 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030219806A1 (en) 2000-02-22 2003-11-27 Millennium Pharmaceuticals, Inc. Novel 18607, 15603, 69318, 12303, 48000, 52920, 5433, 38554, 57301, 58324, 55063, 52991, 59914, 59921 and 33751 molecules and uses therefor
WO2001062794A2 (en) 2000-02-22 2001-08-30 Millennium Pharmaceuticals, Inc. 18607, a human calcium channel
WO2001066689A2 (en) 2000-03-07 2001-09-13 Hyseq, Inc. Novel nucleic acids and polypeptides
WO2001072962A2 (en) 2000-03-24 2001-10-04 Fahri Saatcioglu Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
WO2001072830A2 (en) 2000-03-31 2001-10-04 Ipf Pharmaceuticals Gmbh Diagnostic and medicament for analysing the cell surface proteome of tumour and inflammatory cells and for treating tumorous and inflammatory diseases, preferably using a specific chemokine receptor analysis and the chemokine receptor-ligand interaction
WO2001075177A2 (en) 2000-04-03 2001-10-11 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tumor markers in ovarian cancer
WO2001077172A2 (en) 2000-04-07 2001-10-18 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated known g protein-coupled receptors
WO2001088133A2 (en) 2000-05-18 2001-11-22 Lexicon Genetics Incorporated Human semaphorin homologs and polynucleotides encoding the same
WO2001090304A2 (en) 2000-05-19 2001-11-29 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
WO2001094641A2 (en) 2000-06-09 2001-12-13 Idec Pharmaceuticals Corporation Gene targets and ligands that bind thereto for treatment and diagnosis of ovarian carcinomas
WO2001098351A2 (en) 2000-06-16 2001-12-27 Incyte Genomics, Inc. G-protein coupled receptors
WO2002002634A2 (en) 2000-06-30 2002-01-10 Incyte Genomics, Inc. Human extracellular matrix and cell adhesion polypeptides
WO2002002624A2 (en) 2000-06-30 2002-01-10 Amgen, Inc. B7-like molecules and uses thereof
WO2002002587A1 (en) 2000-06-30 2002-01-10 Human Genome Sciences, Inc. B7-like polynucleotides, polypeptides, and antibodies
WO2002006339A2 (en) 2000-07-03 2002-01-24 Curagen Corporation Proteins and nucleic acids encoding same
WO2002006317A2 (en) 2000-07-17 2002-01-24 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20040044179A1 (en) 2000-07-25 2004-03-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2002010187A1 (en) 2000-07-27 2002-02-07 Mayo Foundation For Medical Education And Research B7-h3 and b7-h4, novel immunoregulatory molecules
WO2002010382A2 (en) 2000-07-28 2002-02-07 Ulrich Wissenbach Trp8, trp9 and trp10, markers for cancer
WO2002012341A2 (en) 2000-08-03 2002-02-14 Corixa Corporation Her-2/neu fusion proteins
WO2002013847A2 (en) 2000-08-14 2002-02-21 Corixa Corporation Methods for diagnosis and therapy of hematological and virus-associated malignancies
WO2002014503A2 (en) 2000-08-14 2002-02-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of her-2/neu-associated malignancies
WO2002016413A2 (en) 2000-08-24 2002-02-28 Glaxosmithkline Biologicals S.A. Cripto tumour polypeptide
WO2002016429A2 (en) 2000-08-24 2002-02-28 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2002018444A2 (en) 2000-09-01 2002-03-07 Genentech, Inc. Erbb4 antagonists
WO2002022660A2 (en) 2000-09-11 2002-03-21 Hyseq, Inc. Novel nucleic acids and polypeptides
US20030186373A1 (en) 2000-09-15 2003-10-02 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2002022636A1 (en) 2000-09-15 2002-03-21 Isis Pharmaceuticals, Inc. Antisense modulation of her-2 expression
US20030119121A1 (en) 2000-09-15 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2002022153A2 (en) 2000-09-15 2002-03-21 Zymogenetics, Inc. Use of a polypeptide comprising the extracellular domains of il-20rb for the treatment of inflammation
WO2002022808A2 (en) 2000-09-18 2002-03-21 Biogen, Inc. Cripto mutant and uses thereof
WO2002024909A2 (en) 2000-09-18 2002-03-28 Biogen, Inc. Receptor nucleic acids and polypeptides
WO2002026822A2 (en) 2000-09-26 2002-04-04 Genentech, Inc. Pumpcn compositions and uses thereof
US20040005598A1 (en) 2000-09-26 2004-01-08 Genentech, Inc. PUMPCn compositions and uses thereof
WO2002030268A2 (en) 2000-10-13 2002-04-18 Eos Biotechnology, Inc. Methods of diagnosis of prostate cancer, compositions and methods of screening for modulators of prostate cancer
WO2002038766A2 (en) 2000-11-07 2002-05-16 Zymogenetics, Inc. Human tumor necrosis factor receptor
US20020150573A1 (en) 2000-11-10 2002-10-17 The Rockefeller University Anti-Igalpha-Igbeta antibody for lymphoma therapy
WO2002061087A2 (en) 2000-12-19 2002-08-08 Lifespan Biosciences, Inc. Antigenic peptides, such as for g protein-coupled receptors (gpcrs), antibodies thereto, and systems for identifying such antigenic peptides
WO2002054940A2 (en) 2001-01-12 2002-07-18 University Of Medicine & Dentistry Of New Jersey Bone morphogenetic protein-2 in the treatment and diagnosis of cancer
US20030119125A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119126A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030118592A1 (en) 2001-01-17 2003-06-26 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
WO2002059377A2 (en) 2001-01-24 2002-08-01 Protein Design Labs Methods of diagnosis of breast cancer, compositions and methods of screening for modulators of breast cancer
WO2002060317A2 (en) 2001-01-30 2002-08-08 Corixa Corporation Compositions and methods for the therapy and diagnosis of pancreatic cancer
WO2002064798A1 (en) 2001-02-12 2002-08-22 Bionomics Limited Dna sequences differentially expressed in tumour cell lines
WO2002072596A1 (en) 2001-03-09 2002-09-19 Incyte Genomics, Inc. Steap-related protein
WO2002071928A2 (en) 2001-03-14 2002-09-19 Millennium Pharmaceuticals, Inc. Nucleic acid molecules and proteins for the identification, assessment, prevention, and therapy of ovarian cancer
WO2002078524A2 (en) 2001-03-28 2002-10-10 Zycos Inc. Translational profiling
WO2003008537A2 (en) 2001-04-06 2003-01-30 Mannkind Corporation Epitope sequences
WO2002081646A2 (en) 2001-04-06 2002-10-17 Mannkind Corporation Epitope sequences
US20040005538A1 (en) 2001-04-11 2004-01-08 Xiaojiang Chen Three-dimensional structure of complement receptor type 2 and uses thereof
WO2002083866A2 (en) 2001-04-17 2002-10-24 The Board Of Trustees Of The University Of Arkansas Repeat sequences of the ca125 gene and their use for diagnostic and therapeutic interventions
WO2002086443A2 (en) 2001-04-18 2002-10-31 Protein Design Labs, Inc Methods of diagnosis of lung cancer, compositions and methods of screening for modulators of lung cancer
WO2002088170A2 (en) 2001-04-26 2002-11-07 Biogen, Inc. Cripto blocking antibodies and uses thereof
WO2002088172A2 (en) 2001-04-30 2002-11-07 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
WO2002089747A2 (en) 2001-05-09 2002-11-14 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
WO2002092836A2 (en) 2001-05-11 2002-11-21 Sloan-Kettering Institute For Cancer Research Nucleic acid sequence encoding ovarian antigen, ca125, and uses thereof
WO2002094852A2 (en) 2001-05-24 2002-11-28 Zymogenetics, Inc. Taci-immunoglobulin fusion proteins
US20030096961A1 (en) 2001-06-01 2003-05-22 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20040044180A1 (en) 2001-06-01 2004-03-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2002098358A2 (en) 2001-06-04 2002-12-12 Eos Biotechnology, Inc. Methods of diagnosis and treatment of androgen-dependent prostate cancer, prostate cancer undergoing androgen-withdrawal, and androgen-independent prostate cancer
WO2003000842A2 (en) 2001-06-04 2003-01-03 Curagen Corporation Novel proteins and nucleic acids encoding same
WO2002099074A2 (en) 2001-06-05 2002-12-12 Exelixis, Inc. Slc7s as modifiers of the p53 pathway and methods of use
WO2002099122A1 (en) 2001-06-05 2002-12-12 Exelixis, Inc. Modifiers of the p53 pathway and methods of use
WO2002101075A2 (en) 2001-06-13 2002-12-19 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
US20040005563A1 (en) 2001-06-18 2004-01-08 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
US20030091580A1 (en) 2001-06-18 2003-05-15 Mitcham Jennifer L. Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2002102235A2 (en) 2001-06-18 2002-12-27 Eos Biotechnology Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
WO2003004989A2 (en) 2001-06-21 2003-01-16 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
WO2003002717A2 (en) 2001-06-28 2003-01-09 Schering Corporation Biological activity of ak155
WO2003004529A2 (en) 2001-07-02 2003-01-16 Licentia Ltd. Ephrin-tie receptor materials and methods
WO2003003906A2 (en) 2001-07-03 2003-01-16 Eos Biotechnology, Inc. Diagnostic and screening methods for bladder cancer
WO2003003984A2 (en) 2001-07-05 2003-01-16 Curagen Corporation Novel proteins and nucleic acids encoding same
WO2003055439A2 (en) 2001-07-18 2003-07-10 The Regents Of The University Of California Her2/neu target antigen and use of same to stimulate an immune response
WO2003009814A2 (en) 2001-07-25 2003-02-06 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of prostate cancer
WO2003014294A2 (en) 2001-08-03 2003-02-20 Genentech, Inc. Tacis and br3 polypeptides and uses thereof
WO2003016475A2 (en) 2001-08-14 2003-02-27 The General Hospital Corporation Nucleic acid and amino acid sequences involved in pain
WO2003016494A2 (en) 2001-08-16 2003-02-27 Vitivity, Inc. Diagnosis and treatment of vascular disease
WO2003018621A2 (en) 2001-08-23 2003-03-06 Oxford Biomedica (Uk) Limited Genes
WO2003029262A2 (en) 2001-08-29 2003-04-10 Vanderbilt University The human mob-5 (il-24) receptors and uses thereof
US20030124579A1 (en) 2001-09-05 2003-07-03 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
WO2003022995A2 (en) 2001-09-06 2003-03-20 Agensys, Inc. Nucleic acid and corresponding protein entitled steap-1 useful in treatment and detection of cancer
WO2003023013A2 (en) 2001-09-13 2003-03-20 Nuvelo, Inc. Novel nucleic acids and polypeptides
WO2003025138A2 (en) 2001-09-17 2003-03-27 Protein Design Labs, Inc. Methods of diagnosis of cancer compositions and methods of screening for modulators of cancer
WO2003024392A2 (en) 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2003025228A1 (en) 2001-09-18 2003-03-27 Proteologics, Inc. Methods and compositions for treating hcap associated diseases
WO2003025148A2 (en) 2001-09-19 2003-03-27 Nuvelo, Inc. Novel nucleic acids and polypeptides
US20030130189A1 (en) 2001-09-24 2003-07-10 Senter Peter D. P-amidobenzylethers in drug delivery agents
WO2003026577A2 (en) 2001-09-24 2003-04-03 Seattle Genetics, Inc. P-amidobenzylethers in drug delivery agents
US20030096743A1 (en) 2001-09-24 2003-05-22 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
WO2003026493A2 (en) 2001-09-28 2003-04-03 Bing Yang Diagnosis and treatment of diseases caused by mutations in cd72
WO2003029421A2 (en) 2001-10-03 2003-04-10 Origene Technologies, Inc. Regulated breast cancer genes
WO2003029277A2 (en) 2001-10-03 2003-04-10 Rigel Pharmaceuticals, Inc. Modulators of lymphocyte activation and migration
WO2003034984A2 (en) 2001-10-19 2003-05-01 Genentech, Inc. Compositions and methods for the diagnosis and treatment of inflammatory bowel disorders
WO2003035846A2 (en) 2001-10-24 2003-05-01 National Jewish Medical And Research Center Structure of tall-1 and its cognate receptor
WO2003055443A2 (en) 2001-10-31 2003-07-10 Alcon, Inc. Bone morphogenic proteins (bmp), bmp receptors and bmp binding proteins and their use in the diagnosis and treatment of glaucoma
WO2003077836A2 (en) 2001-11-06 2003-09-25 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US20030232350A1 (en) 2001-11-13 2003-12-18 Eos Biotechnology, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
WO2003042661A2 (en) 2001-11-13 2003-05-22 Protein Design Labs, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
WO2003043583A2 (en) 2001-11-20 2003-05-30 Seattle Genetics, Inc. Treatment of immunological disorders using anti-cd30 antibodies
WO2003045422A1 (en) 2001-11-29 2003-06-05 Genset S.A. Agonists and antagonists of prolixin for the treatment of metabolic disorders
WO2003048202A2 (en) 2001-12-03 2003-06-12 Asahi Kasei Pharma Corporation Nf-kappab activating genes
WO2003054152A2 (en) 2001-12-10 2003-07-03 Nuvelo, Inc. Novel nucleic acids and polypeptides
US20030134790A1 (en) 2002-01-11 2003-07-17 University Of Medicine And Dentistry Of New Jersey Bone Morphogenetic Protein-2 And Bone Morphogenetic Protein-4 In The Treatment And Diagnosis Of Cancer
WO2003062401A2 (en) 2002-01-22 2003-07-31 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US20030143557A1 (en) 2002-01-25 2003-07-31 Reinhold Penner Methods of screening for TRPM4b modulators
WO2003072036A2 (en) 2002-02-21 2003-09-04 Duke University Treatment methods using anti-cd22 antibodies
WO2003072035A2 (en) 2002-02-22 2003-09-04 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO2003083047A2 (en) 2002-03-01 2003-10-09 Exelixis, Inc. MP53s AS MODIFIERS OF THE p53 PATHWAY AND METHODS OF USE
WO2004000997A2 (en) 2002-03-19 2003-12-31 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2003081210A2 (en) 2002-03-21 2003-10-02 Sunesis Pharmaceuticals, Inc. Identification of kinase inhibitors
EP1347046A1 (en) 2002-03-22 2003-09-24 Research Association for Biotechnology Full-length cDNA sequences
WO2003083041A2 (en) 2002-03-22 2003-10-09 Biogen, Inc. Cripto-specific antibodies
WO2003089624A2 (en) 2002-03-25 2003-10-30 Uab Research Foundation Fc receptor homolog, reagents, and uses thereof
WO2003083074A2 (en) 2002-03-28 2003-10-09 Idec Pharmaceuticals Corporation Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
US20030194704A1 (en) 2002-04-03 2003-10-16 Penn Sharron Gaynor Human genome-derived single exon nucleic acid probes useful for gene expression analysis two
WO2003087306A2 (en) 2002-04-05 2003-10-23 Agensys, Inc. Nucleic acid and corresponding protein entitled 98p4b6 useful in treatment and detection of cancer
US20040101874A1 (en) 2002-04-12 2004-05-27 Mitokor Inc. Targets for therapeutic intervention identified in the mitochondrial proteome
WO2003087768A2 (en) 2002-04-12 2003-10-23 Mitokor Targets for therapeutic intervention identified in the mitochondrial proteome
US20030228319A1 (en) 2002-04-16 2003-12-11 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
WO2003089904A2 (en) 2002-04-17 2003-10-30 Baylor College Of Medicine Aib1 as a prognostic marker and predictor of resistance to encocrine therapy
WO2004042346A2 (en) 2002-04-24 2004-05-21 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
WO2003093444A2 (en) 2002-05-03 2003-11-13 Incyte Corporation Transporters and ion channels
WO2003097803A2 (en) 2002-05-15 2003-11-27 Avalon Pharmaceuticals Cancer-linked gene as target for chemotherapy
US20030224454A1 (en) 2002-05-30 2003-12-04 Ryseck Rolf Peter Human solute carrier family 7, member 11 (hSLC7A11)
WO2003101283A2 (en) 2002-06-04 2003-12-11 Incyte Corporation Diagnostics markers for lung cancer
WO2003101400A2 (en) 2002-06-04 2003-12-11 Avalon Pharmaceuticals, Inc. Cancer-linked gene as target for chemotherapy
WO2003104270A2 (en) 2002-06-06 2003-12-18 Ingenium Pharmaceuticals Ag Dudulin 2 genes, expression products, non-human animal model: uses in human hematological disease
WO2003104275A2 (en) 2002-06-06 2003-12-18 Oncotherapy Science, Inc. Genes and polypeptides relating to human colon cancers
WO2003104399A2 (en) 2002-06-07 2003-12-18 Avalon Pharmaceuticals, Inc Cancer-linked gene as target for chemotherapy
WO2003105758A2 (en) 2002-06-12 2003-12-24 Avalon Pharmaceuticals, Inc. Cancer-linked gene as target for chemotherapy
US20040249130A1 (en) 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
US20040022727A1 (en) 2002-06-18 2004-02-05 Martin Stanton Aptamer-toxin molecules and methods for using same
WO2004000221A2 (en) 2002-06-20 2003-12-31 The Regents Of The University Of California Compositions and methods for modulating lymphocyte activity
WO2004001993A1 (en) 2002-06-20 2003-12-31 Snaptrack Incorporated Reducing cross-interference in a combined gps receiver and communication system
WO2004001004A2 (en) 2002-06-21 2003-12-31 Johns Hopkins University School Of Medicine Membrane associated tumor endothelium markers
US20040001827A1 (en) 2002-06-28 2004-01-01 Dennis Mark S. Serum albumin binding peptides for tumor targeting
WO2004009622A2 (en) 2002-07-19 2004-01-29 Cellzome Ag Protein complexes of cellular networks underlying the development of cancer and other diseases
WO2004011611A2 (en) 2002-07-25 2004-02-05 Genentech, Inc. Taci antibodies and uses thereof
WO2004032828A2 (en) 2002-07-31 2004-04-22 Seattle Genetics, Inc. Anti-cd20 antibody-drug conjugates for the treatment of cancer and immune disorders
WO2004015426A1 (en) 2002-08-06 2004-02-19 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with human cxc chemokine receptor 5(cxcr5)
EP1394274A2 (en) 2002-08-06 2004-03-03 Genox Research, Inc. Methods of testing for bronchial asthma or chronic obstructive pulmonary disease
WO2004016225A2 (en) 2002-08-19 2004-02-26 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2004020583A2 (en) 2002-08-27 2004-03-11 Bristol-Myers Squibb Company Polynucleotide predictor set for identifying protein tyrosine kinase modulators
WO2004020595A2 (en) 2002-08-29 2004-03-11 Five Prime Therapeutics, Inc. Novel human polypeptides encoded by polynucleotides
WO2004022778A1 (en) 2002-09-05 2004-03-18 Garvan Institute Of Medical Research Methods of diagnosis and prognosis of ovarian cancer
WO2004022709A2 (en) 2002-09-06 2004-03-18 Mannkind Corporation Epitope sequences
WO2004040000A2 (en) 2002-09-09 2004-05-13 Nura, Inc G protein coupled receptors and uses thereof
WO2004027049A2 (en) 2002-09-20 2004-04-01 Astral, Inc. Methods and compositions to generate and control the effector profile of t cells by simultaneous loading and activation of selected subsets of antigen presenting cells
JP2004113151A (en) 2002-09-27 2004-04-15 Sankyo Co Ltd Oncogene and its application
WO2004031238A2 (en) 2002-10-03 2004-04-15 Mcgill Univeristy Antibodies and cyclic peptides which bind cea (carcinoembryonic antigen) and their use as cancer therapeutics
WO2004032842A2 (en) 2002-10-04 2004-04-22 Van Andel Research Institute Molecular sub-classification of kidney tumors and the discovery of new diagnostic markers
US20050048572A1 (en) 2002-10-31 2005-03-03 Genentech, Inc. Methods and compositions for increasing antibody production
WO2004043361A2 (en) 2002-11-08 2004-05-27 Genentech, Inc. Compositions and methods for the treatment of natural killer cell related diseases
WO2004044178A2 (en) 2002-11-13 2004-05-27 Genentech, Inc. Methods and compositions for diagnosing dysplasia
WO2004045516A2 (en) 2002-11-15 2004-06-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2004045553A2 (en) 2002-11-15 2004-06-03 The Board Of Trustees Of The University Of Arkansas Ca125 gene and its use for diagnostic and therapeutic interventions
WO2004045520A2 (en) 2002-11-15 2004-06-03 Musc Foundation For Research Development Complement receptor 2 targeted complement modulators
WO2004046342A2 (en) 2002-11-20 2004-06-03 Biogen Idec Inc. Novel gene targets and ligands that bind thereto for treatment and diagnosis of carcinomas
WO2004047749A2 (en) 2002-11-21 2004-06-10 University Of Utah Research Foundation Purinergic modulation of smell
WO2004048938A2 (en) 2002-11-26 2004-06-10 Protein Design Labs, Inc. Methods of detecting soft tissue sarcoma, compositions and methods of screening for soft tissue sarcoma modulators
WO2004053079A2 (en) 2002-12-06 2004-06-24 Diadexus, Inc. Compositions, splice variants and methods relating to ovarian specific genes and proteins
EP1439393A2 (en) 2002-12-13 2004-07-21 Bayer Healthcare LLC Detection methods using TIMP 1 for colon cancer diagnosis
US20040197325A1 (en) 2002-12-20 2004-10-07 Debbie Law Antibodies against GPR64 and uses thereof
WO2004058309A1 (en) 2002-12-23 2004-07-15 Human Genome Sciences, Inc. Neutrokine-alpha conjugate, neutrokine-alpha complex, and uses thereof
WO2004063709A2 (en) 2003-01-08 2004-07-29 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
WO2004063355A2 (en) 2003-01-10 2004-07-29 Protein Design Labs, Inc. Novel methods of diagnosis of metastatic cancer, compositions and methods of screening for modulators of matastatic cancer
WO2004063362A2 (en) 2003-01-10 2004-07-29 Cyclacel Limited Cell cycle progression proteins
WO2004065577A2 (en) 2003-01-14 2004-08-05 Bristol-Myers Squibb Company Polynucleotides and polypeptides associated with the nf-kb pathway
WO2004065576A2 (en) 2003-01-15 2004-08-05 Millennium Pharmaceuticals, Inc. Methods and compositions for the treatment of urological disorder using differential expressed polypeptides
US20040229310A1 (en) 2003-01-23 2004-11-18 Genentech, Inc. Methods for producing humanized antibodies and improving yield of antibodies or antigen binding fragments in cell culture
WO2004074320A2 (en) 2003-02-14 2004-09-02 Sagres Discovery, Inc. Therapeutic targets in cancer
US20030224411A1 (en) 2003-03-13 2003-12-04 Stanton Lawrence W. Genes that are up- or down-regulated during differentiation of human embryonic stem cells
WO2006034488A2 (en) * 2004-09-23 2006-03-30 Genentech, Inc. Cysteine engineered antibodies and conjugates
US20080050310A1 (en) 2006-05-30 2008-02-28 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
US20080247951A1 (en) 2007-02-09 2008-10-09 Genentech, Inc. Anti-robo4 antibodies and uses therefor
WO2008141044A2 (en) * 2007-05-08 2008-11-20 Genentech, Inc. Cysteine engineered anti-muc16 antibodies and antibody drug conjugates
US20080311134A1 (en) 2007-05-08 2008-12-18 Junutula Jagath R Cysteine engineered anti-muc16 antibodies and antibody drug conjugates
US20090028856A1 (en) 2007-07-16 2009-01-29 Genentech, Inc. Anti-CD79B Antibodies and Immunoconjugates and Methods of Use
US20090068202A1 (en) 2007-07-16 2009-03-12 Genentech, Inc. Humanized Anti-CD79B Antibodies and Immunoconjugates and Methods of Use
US20090117100A1 (en) 2007-10-19 2009-05-07 Weiguang Mao Cysteine engineered anti-TENB2 antibodies and antibody drug conjugates

Non-Patent Citations (368)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Immunology", vol. 1, 2, 1991, WILEY-INTERSCIENCE
"McGraw-Hill Dictionary of Chemical Terms", 1984, MCGRAW-HILL BOOK COMPANY
"The CCP4 Suite: Programs for Protein Crystallography", ACTA. CRYST., vol. D50, 1994, pages 760 - 763
AERTS HJ ET AL.: "Disparity between in vivo EGFR expression and 89Zr-labeled cetuximab uptake assessed with PET", J NUCL MED., vol. 50, 2009, pages 123 - 31, XP055026510, DOI: doi:10.2967/jnumed.108.054312
ALBERT ET AL., BIOORG. MED. CHEM. LETT., vol. 8, 1998, pages 1207 - 1210
AM. J. HUM. GENET., vol. 49, no. 3, 1991, pages 555 - 565
AMIEL J. ET AL., HUM. MOL. GENET., vol. 5, 1996, pages 355 - 357
AMIR ET AL., ANGEW. CHEM. INT. ED., vol. 42, 2003, pages 4494 - 4499
ANDRUS, A.: "PCR 2: A Practical Approach", 1995, OXFORD UNIVERSITY PRESS, article "Chemical methods for 5' non-isotopic labelling of PCR probes and primers", pages: 39 - 54
ANNU. REV. NEUROSCI., vol. 21, 1998, pages 309 - 345
ARAI H. ET AL., J. BIOL. CHEM., vol. 268, 1993, pages 3463 - 3470
ARAI H. ET AL., JPN. CIRC. J., vol. 56, 1992, pages 1303 - 1307
ATTIE T. ET AL., HUM. MOL. GENET., vol. 4, 1995, pages 2407 - 2409
AURICCHIO A. ET AL., HUM. MOL. GENET., vol. 5, 1996, pages 351 - 354
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1993, GREENE PUBLISHING AND WILEY-INTERSCIENCE
AXWORTHY ET AL., PROC. NATL. ACAD. SCI. USA, vol. 97, no. 4, 2000, pages 1802 - 1807
B.LEEF.M.RICHARDS, J.MOL.BIOL., vol. 55, 1971, pages 379 - 400
BACA ET AL., JOURNAL BIOLOGICAL CHEMISTRY, vol. 272, no. 16, 1997, pages 10678 - 84
BAREL M. ET AL., MOL. IMMUNOL., vol. 35, 1998, pages 1025 - 1031
BARELLA ET AL., BIOCHEM. J., vol. 309, 1995, pages 773 - 779
BARNETT T. ET AL., GENOMICS, vol. 3, 1988, pages 59 - 66
BEAUCAGE, S.IYER, R.: "Advances in the synthesis of oligonucleotides by the phosphoramidite approach", TETRAHEDRON, vol. 48, 1992, pages 2223 - 2311, XP000915225, DOI: doi:10.1016/S0040-4020(01)88752-4
BECK ET AL., J. MOL. BIOL., vol. 228, 1992, pages 433 - 441
BECK, J. MOL. BIOL., vol. 255, 1996, pages 1 - 13
BERNHARD ET AL., BIOCONJUGATE CHEM., vol. 5, 1994, pages 126 - 132
BETTER ET AL., J. BIOL. CHEM., vol. 269, no. 13, 1994, pages 9644 - 9650
BIOCHEM. BIOPHYS. RES. COMMUN., vol. 255, no. 2, 1999, pages 283 - 288
BIOCHEM. BIOPHYS. RES. COMMUN., vol. 275, no. 3, 2000, pages 783 - 788
BIOCONJUGATE CHEM., vol. 3, 1992, pages 2
BISHOP,D.T. ET AL., NAT. GENET., vol. 41, no. 8, 2009, pages 920 - 925
BLEND ET AL., CANCER BIOTHERAPY & RADIOPHARMACEUTICALS, vol. 18, 2003, pages 355 - 363
BLOOD, vol. 100, no. 9, 2002, pages 3068 - 3076
BLOOD, vol. 99, no. 8, 2002, pages 2662 - 2669
BLUMBERG H. ET AL., CELL, vol. 104, 2001, pages 9 - 19
BOIJESSON PK ET AL.: "Performance ofimmuno- positron emission tomography with zirconium-89-labeled chimeric monoclonal antibody U36 in the detection of lymph node metastases in head and neck cancer patients", CLIN CANCER RES., vol. 12, 2006, pages 2133 - 40
BOURGEOIS C. ET AL., J. CLIN. ENDOCRINOL. METAB., vol. 82, 1997, pages 3116 - 3123
BRIGGS ET AL.: "Synthcsis of Functionaliscd Fluorescent Dyes and Their Coupling to Amincs and Amino Acids", J. CHEM. SOC., PERKIN-TRANS., vol. 1, 1997, pages 1051 - 1058
BRINKLEY, BIOCONJUGATE CHEM., vol. 3, 1992, pages 2
CAI W ET AL.: "PET imaging of colorectal cancer in xenograft-bearing mice by use of an 18F-labeled T84.66 anti-carcinoembryonic antigen diabody", J NUCL MED., vol. 48, 2007, pages 304 - 10, XP055003542, DOI: doi:10.2967/jnumed.107.043216
CAMERA ET AL., NUCL. MED. BID., vol. 20, 1993, pages 955 - 62
CAMERA, J. NUCL. MCD., vol. 21, 1994, pages 640 - 646
CANCER RES., vol. 61, no. 15, 2001, pages 5857 - 5860
CAPEL ET AL., IMMUNOMETHODS, vol. 4, 1994, pages 25 - 34
CARPENTER ET AL., ANN. REV. BIOCHEM., vol. 56, 1987, pages 881 - 914
CARTER ET AL., NUCLEIC ACIDS RES., vol. 13, 1985, pages 4431 - 4443
CELL, vol. 109, no. 3, 2002, pages 397 - 407
CHAN,J.WATT, V.M., ONCOGENE, vol. 6, no. 6, 1991, pages 1057 - 1061
CHANG ET AL., GENE, vol. 55, 1987, pages 189 - 196
CHATAL: "Monoclonal Antibodies in Immunoscintigraphy", 1989, CRC PRESS
CHEN ET AL., BIOCONJUGATE CHEM., vol. 15, 2004, pages 41 - 49
CHMURA ET AL., PROC. NAT. ACAD. SCI. USA, vol. 98, no. 15, 2001, pages 8480 - 8484
CHO H.-S., NATURE, vol. 421, 2003, pages 756 - 760
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CICCODICOLA, A. ET AL., EMBO J., vol. 8, no. 7, 1989, pages 1987 - 1991
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLARK H.F. ET AL., GENOME RES., vol. 13, 2003, pages 2265 - 2270
CLARK, H.F. ET AL., GENOME RES., vol. 13, no. 10, 2003, pages 2265 - 2270
CLARK,H.F. ET AL., GENOME RES., vol. 13, no. 10, 2003, pages 2265 - 2270
CLYNES ET AL., PROC. NAT. ACAD. SCI., vol. 95, 1998, pages 652 - 656
COLE, R.B.: "Electro Spray Ionization Mass Spectrometry: Fundamentals, Instrumentation And Applications", 1997, WILEY
COUSSENS L. ET AL., SCIENCE, vol. 230, no. 4730, 1985, pages 1132 - 1139
DAVIS ET AL., PROC. NATL. ACAD. SCI USA, vol. 98, no. 17, 2001, pages 9772 - 9777
DE HAAS ET AL., J. LAB. CLIN. MED., vol. 126, 1995, pages 330 - 41
DE LEON-RODRIGUEZ ET AL., CHEM.EUR. J., vol. 10, 2004, pages 1149 - 1155
DE NOOIJ-VAN DALEN,A.G. ET AL., INT. J. CANCER, vol. 103, no. 6, 2003, pages 768 - 774
DENARDO ET AL., CLINICAL CANCER RESEARCH, vol. 4, 1998, pages 2483 - 90
DENNIS ET AL., J BIOL CHEM., vol. 277, 2002, pages 35035 - 35043
DENNIS ET AL.: "Albumin Binding As A General Strategy For Improving The Pharmacokinetics Of Proteins", J BIOL CHEM., vol. 277, 2002, pages 35035 - 35043
DIJKE,P. ET AL., SCIENCE, vol. 264, no. 5155, 1994, pages 101 - 104
DIJKERS EC ET AL.: "Development and Characterization of Clinical-Grade 89Zr-Trastuzumab for HER2/neu ImmunoPET Imaging", J NUCL MED, vol. 50, no. 6, 2009, pages 974 - 981, XP002615664, DOI: doi:10.2967/JNUMED.108.060392
DIJKERS ELI C F ET AL: "Development and characterization of clinical-grade 89Zr-trastuzumab for HER2/neu immunoPET imaging.", JOURNAL OF NUCLEAR MEDICINE : OFFICIAL PUBLICATION, SOCIETY OF NUCLEAR MEDICINE JUN 2009 LNKD- PUBMED:19443585, vol. 50, no. 6, June 2009 (2009-06-01), pages 974 - 981, XP002615664, ISSN: 0161-5505 *
DIVGI CR ET AL.: "Preoperative characterisation of clear-cell renal carcinoma using iodine-124-labelled antibody chimeric G250 (124I-cG250) and PET in patients with renal masses: a phase I trial", LANCET ONCOL, vol. 8, 2007, pages 304 - 10, XP002615898, DOI: doi:10.1016/S1470-2045(07)70044-X
DOBNER ET AL., EUR. J. IMMUNOL., vol. 22, 1992, pages 2795 - 2799
DREBIN ET AL., NATURE, vol. 312, 1984, pages 545 - 548
D'SOUZA ET AL., PROC. NATL. ACAD. SCI., vol. 91, 1994, pages 7202 - 7206
DUMOUTIER L. ET AL., J. IMMUNOL., vol. 167, 2001, pages 3545 - 3549
EHSANI A. ET AL., GENOMICS, vol. 15, 1993, pages 426 - 429
EIGCNBROT, J MOL BIOL., vol. 229, 1993, pages 969 - 995
EIGENBROT ET AL., J MOL BIOL., vol. 229, 1993, pages 969 - 995
EIGENBROT ET AL.: "X-Ray Structures Of The Antigen-Binding Domains From Three Variants Of Humanized Anti-P185her2 Antibody 4D5 And Comparison With Molecular Modeling", J MOL BIOL., vol. 229, 1993, pages 969 - 995, XP024009357, DOI: doi:10.1006/jmbi.1993.1099
ELIEL, E.WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC.
ELSHOURBAGY N.A. ET AL., J. BIOL. CHEM., vol. 268, 1993, pages 3873 - 3879
ERICSSON, T.A. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 100, no. 11, 2003, pages 6759 - 6764
ERKCR, G., PURE AND APPLIED CHEMISTRY, vol. 63, no. 6, 1991, pages 797 - 806
ERKER, G., JOUR. OF ORGANOMETALLIC CHEM., vol. 400, no. 1-2, 1990, pages 185 - 203
FCILD, J.A. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 258, no. 3, 1999, pages 578 - 582
FENDLY ET AL., CANCER RESEARCH, vol. 50, 1990, pages 1550 - 1558
FLETCHER CR: "The radiological hazards of zirconium-95 and niobium-95", HEALTH PHYS., vol. 16, 1969, pages 209 - 20
FOOTEWINTER, J. MOL. BIOL., vol. 224, 1992, pages 487 - 499
FRAKER ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 80, 1978, pages 49 - 57
FRISCH ET AL., BIOCONJUGATE CHEM., vol. 7, 1996, pages 180 - 186
FUCHS S. ET AL., MOL. MED., vol. 7, 2001, pages 115 - 124
FUJISAKU, J. BIOL. CHCM., vol. 264, no. 4, 1989, pages 2118 - 2125
FURUSHIMA,K. ET AL., DEV. BIOL., vol. 306, no. 2, 2007, pages 480 - 492
GARMAN: "Non-Radioactive Labelling: A Practical Approach", 1997, ACADEMIC PRESS
GARMAN: "Non-Radioactive Labelling: A Practical Approach", 1997, ACADEMIC PRESS, pages: 55
GARRARD ET AL., GENE, vol. 128, 1993, pages 103 - 109
GARY S.C. ET AL., GENE, vol. 256, 2000, pages 139 - 147
GAUGITSCH, H.W. ET AL., J. BIOL. CHEM., vol. 267, no. 16, 1992, pages 11267 - 11273
GAZZANO-SANTORO ET AL., J. IMMUNOL. METHODS, vol. 202, 1996, pages 163
GCNOMICS, vol. 62, no. 2, 1999, pages 281 - 284
GCRSTNCR: "Sequence Plasticity In The Antigen-Binding Site Of A Therapeutic Anti-HER2 Antibody", J MOL BIOL., vol. 321, 2002, pages 851 - 62, XP002972421, DOI: doi:10.1016/S0022-2836(02)00677-0
GENOME RES., vol. 13, no. 10, 2003, pages 2265 - 2270
GERHARD,D.S. ET AL., GENOME RES., vol. 14, no. 1 OB, 2004, pages 2121 - 2127
GERY, S. ET AL., ONCOGENE, vol. 22, no. 18, 2003, pages 2723 - 2727
GETZ ET AL., ANAL. BIOCHEM., vol. 273, 1999, pages 73 - 80
GILL HS ET AL.: "A modular platform for the rapid site-specific radiolabeling of proteins with 18F exemplified by quantitative positron emission tomography of human epidermal growth factor receptor 2", JOUR. OFMED. CHEM., vol. 52, 2009, pages 5816 - 25, XP055238938, DOI: doi:10.1021/jm900420c
GLAZER ET AL.: "Chemical Modification of Proteins. Laboratory Techniques in Biochemistry and Molecular Biology", 1975, AMERICAN ELSEVIER PUBLISHING CO.
GLYNNE-JONES ET AL., INT J CANCER., vol. 94, no. 2, 15 October 2001 (2001-10-15), pages 178 - 84
GONZALEZ TROTTER DE ET AL.: "Quantitation of small-animal (124)1 activity distributions using a clinical PET/CT scanncr", J NUCL MED, vol. 45, 2004, pages 1237 - 44
GOVINDAN S V ET AL: "Deferoxamine as a chelator for <67>Ga in the preparation of antibody conjugates", NUCLEAR MEDICINE AND BIOLOGY, ELSEVIER, NY, US, vol. 32, no. 5, 1 July 2005 (2005-07-01), pages 513 - 519, XP004952403, ISSN: 0969-8051, DOI: DOI:10.1016/J.NUCMEDBIO.2005.04.009 *
GREENWOOD ET AL., THERAPEUTIC IMMUNOLOGY, vol. 1, 1994, pages 247 - 255
GU Z. ET AL., ONCOGENE, vol. 19, 2000, pages 1288 - 1296
GUYER ET AL., J. IMMUNOL., vol. 117, 1976, pages 587
HA ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1526 - 1531
HAENDLER B. ET AL., J. CARDIOVASC. PHARMACOL., vol. 20, 1992, pages S1 - S4
HAMBLETT KJ ET AL.: "Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate", CLIN CANCER RES., vol. 10, 2004, pages 7063 - 70, XP002726047, DOI: doi:10.1158/1078-0432.CCR-04-0789
HARLOW E.LANE, D.: "Using Antibodies: A Laboratory Manual", 1999, COLD SPRINGS HARBOR LABORATORY PRESS
HARMS, P.W., GENES DEV., vol. 17, no. 21, 2003, pages 2624 - 2629
HASHIMOTO ET AL., IMMUNOGENETICS, vol. 40, no. 4, 1994, pages 287 - 295
HATA, K. ET AL., ANTICANCER RES., vol. 29, no. 2, 2009, pages 617 - 623
HAUGLAND: "Molecular Probes Handbook of Fluorescent Probes and Research Chemicals", 2003, MOLECULAR PROBES, INC.
HERMANSON, G.: "Bioconjugate Techniques", 1996, ACADEMIC PRESS
HERMANSON, G.: "Bioconjugate Techniques", 1996, ACADEMIC PRESS, pages: 40 - 55,643-6
HIGUCHI: "PCR Protocols", 1990, ACADEMIC PRESS, pages: 177 - 183
HNATOWICH ET AL., J. IMMUNOL. METHODS, vol. 65, 1983, pages 147 - 157
HO ET AL., GENE, vol. 77, 1989, pages 51 - 59
HOFSTRA R.M.W. ET AL., EUR. J. HUM. GENET., vol. 5, 1997, pages 180 - 185
HOFSTRA R.M.W. ET AL., NAT. GENET., vol. 12, 1996, pages 445 - 447
HOLLAND JP: "Standardizcd methods for the production of high specific-activity zirconium-89", NUCL MED BIOL., vol. 36, 2009, pages 729 - 39, XP026545224, DOI: doi:10.1016/j.nucmedbio.2009.05.007
HOLLIGER ET AL., NAT BIOTECHNOL, vol. 23, 2005, pages 1126 - 36
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HORIE ET AL., GENOMICS, vol. 67, 2000, pages 146 - 152
HUBERT, R.S. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 96, no. 25, 1999, pages 14523 - 14528
HUDZIAK ET AL., MOL. CELL. BIOL., vol. 9, no. 3, 1989, pages 1165 - 1172
HUMPHREY ET AL., PROC. NAT. ACAD. SCI., vol. 87, 1990, pages 4207 - 4211
IMMUNOGENETICS, vol. 54, no. 2, 2002, pages 87 - 95
INT. REV. CYTOL., vol. 196, 2000, pages 177 - 244
ISHIKAWA,N. ET AL., CANCER RES., vol. 67, no. 24, 2007, pages 11601 - 11611
ITO ET AL., GENE, vol. 102, 1991, pages 67 - 70
IZARD ET AL., BIOCONJUGATE CHEM., vol. 3, 1992, pages 346 - 350
J. BIOL. CHCM., vol. 276, no. 29, 2001, pages 27371 - 27375
J. BIOL. CHCM., vol. 277, no. 22, 2002, pages 19665 - 19672
J. BIOL. CHCM., vol. 278, no. 33, 2003, pages 30813 - 30820
J. BIOL. CHEM., vol. 270, no. 37, 1995, pages 21984 - 21990
JAIN MBATRA SK: "Genetically engineered antibody fragments and PET imaging: a new era of radioimmunodiagnosis", J NUCL MED, vol. 44, 2003, pages 1970 - 2
JANEWAY, C.TRAVERS, P.WALPORT, M.SHLOMCHIK. 5TH ED.: "Immunobiology", 2001, GARLAND PUBLISHING
JANEWAY, C.TRAVERS, P.WALPORT, M.SHLOMCHIK: "Immunobiology. 5th Ed.", 2001, GARLAND PUBLISHING, pages: 627 - 628
JAYSON GC ET AL.: "Molecular imaging and biological evaluation of HuMV833 anti-VEGF antibody: implications for trial design of antiangiogenic antibodies", J NATL CANCER INST, vol. 94, 2002, pages 1484 - 93, XP002472298
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
JONSSON ET AL., IMMUNOGENETICS, vol. 29, no. 6, 1989, pages 411 - 413
JUNUTULA ET AL: "Rapid identification of reactive cysteine residues for site-specific labeling of antibody-Fabs", JOURNAL OF IMMUNOLOGICAL METHODS, ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM, NL, vol. 332, no. 1-2, 14 January 2008 (2008-01-14), pages 41 - 52, XP022527824, ISSN: 0022-1759, DOI: DOI:10.1016/J.JIM.2007.12.011 *
JUNUTULA J R ET AL: "Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index", NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 26, no. 8, 1 August 2008 (2008-08-01), pages 925 - 932, XP002499771, ISSN: 1087-0156, [retrieved on 20080720], DOI: DOI:10.1038/NBT.1480 *
JUNUTULA JR ET AL.: "Rapid identification of reactive cysteine residues for site-specific labeling of antibody-Fabs", J IMMUNOL METHODS, vol. 332, 2008, pages 41 - 52, XP022527824, DOI: doi:10.1016/j.jim.2007.12.011
JUNUTULA JR ET AL.: "Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index", NAT BIOTECHNOL, vol. 26, 2008, pages 925 - 32
JUNUTULA JR ET AL.: "Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index", NAT BIOTECHNOL., vol. 26, 2008, pages 925 - 32
KABAT ET AL.: "Sequences of Proteins of Immunological Interest. 5th Ed.", 1991, NATIONAL INSTITUTES OF HEALTH
KANNO ET AL., J. OF BIOTECHNOLOGY, vol. 76, 2000, pages 207 - 214
KASAHARA ET AL., IMMUNOGENETICS, vol. 30, no. 1, 1989, pages 66 - 68
KELLER, G.MANAK, M.: "DNA Probes", 1993, STOCKTON PRESS, pages: 121 - 23
KIM ET AL., J. IMMUNOL., vol. 24, 1994, pages 249
KIM,M.H. ET AL., MOL. CELL. BIOL., vol. 29, no. 8, 2009, pages 2264 - 2277
KLUSSMAN ET AL., BIOCONJUGATE CHEMISTRY, vol. 15, no. 4, 2004, pages 765 - 773
KNIGHT, C.: "Methods in Enzymology", vol. 248, 1995, ACADEMIC PRESS, article "Fluorimetric Assays of Proteolytic Enzymes", pages: 18 - 34
KOBAYASHI ET AL., BIOCONJUGATE CHEM., vol. 10, 1999, pages 103 - 111
KOBAYASHI ET AL., J. NUCL. MED., vol. 39, 1998, pages 829 - 36
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
KOTTS ET AL., IN VITRO, vol. 26, no. 3, 1990, pages 59A
KRAUS ET AL., PROC. NAT. ACAD. SCI., vol. 86, 1989, pages 9193 - 9197
KUHNS J.J. ET AL., J. BIOL. CHCM., vol. 274, 1999, pages 36422 - 36427
KUKIS ET AL., J. NUCL. MED., vol. 39, 1998, pages 2105 - 2110
KUMAR ET AL., MOL. CELL. BIOL., vol. 11, no. 2, 1991, pages 979 - 986
KUMMER,M.P. ET AL., J. BIOL. CHEM., vol. 284, no. 4, 2009, pages 2296 - 2306
KUNKEL ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1987, pages 488
LAB. INVEST., vol. 82, no. 11, 2002, pages 1573 - 1582
LARHAMMAR ET AL., J. BIOL. CHEM., vol. 260, no. 26, 1985, pages 14111 - 14119
LARSON SM ET AL.: "PET scanning of iodine-124-3F9 as an approach to tumor dosimetry during treatment planning for radioimmunotherapy in a child with neuroblastoma", J NUCL MED, vol. 33, 1992, pages 2020 - 3
LE ET AL., FEBS LETT., vol. 418, no. 1-2, 1997, pages 195 - 199
LEE ET AL., CANCER RES., vol. 61, 2001, pages 4474 - 4482
LEE FT ET AL.: "Immuno-PET of human colon xenograft- bearing BALB/c nude mice using 124I-CDR-grafted humanized A33 monoclonal antibody", J NUCL MED, vol. 42, 2001, pages 764 - 9, XP001023782
LEWIS ET AL., BIOCONJ. CHEM., vol. 9, 1998, pages 72 - 86
LEWIS ET AL., BIOCONJUGATE CHEM., vol. 12, 2001, pages 320 - 324
LEWIS ET AL., CANCER RESEARCH, vol. 56, 1996, pages 1457 - 1465
LEWIS, CANCER IMMUNOL. IMMUNOTHER, vol. 37, 1993, pages 255 - 263
LI ET AL., BIOCONJUGATE CHEM., vol. 13, 2002, pages 110 - 115
LIANG ET AL., CANCER RES., vol. 60, 2000, pages 4907 - 12
LIU ET AL., J. BIOL. CHEM., vol. 273, 1998, pages 20252 - 20260
LIU, SHUANG, ADVANCED DRUG DELIVERY REVIEWS, vol. 60, no. 12, 2008, pages 1347 - 1370
LOWMAN ET AL., J. BIOL. CHEM., vol. 266, no. 17, 1991, pages 10982 - 10988
LOWMAN, HENRY B., METHODS IN MOLECULAR BIOLOGY, vol. 87, 1998, pages 249 - 264
LOWMANWELLS, METHODS: A COMPANION TO METHODS IN ENZYMOLOGY, vol. 3, 1991, pages 205 - 0216
LUND ET AL., EUR. J. BIOCHEM., vol. 267, 2000, pages 7246 - 7256
LUNDBLAD R.L.: "Chemical Reagents for Protein Modification. 2nd Ed.", 1991, CRC PRESS
LUNDBLAD, R. L.NOYES, C. M.: "Chemical Reagents for Protein Modification", vol. I, II, 1984, CRC PRESS
M. IN DAERON, ANNU. REV. IMMUNOL, vol. 15, 1997, pages 203 - 234
MALLYA, M. ET AL., GENOMICS, vol. 80, no. 1, 2002, pages 113 - 123
MARDIROSSIAN ET AL., NUCL. MED. BIOL., vol. 20, 1993, pages 65 - 74
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
MCGLINCHEY,R.P. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 106, no. 33, 2009, pages 13731 - 13736
MCIJS WE: "A facile method for the labeling of proteins with zirconium isotopes", NUCL MED BIOL., vol. 23, 1996, pages 439 - 48, XP004051812, DOI: doi:10.1016/0969-8051(96)00020-0
MEALEY J, JR: "Turn-over of carrier-free zirconium-89 in man", NATURE, vol. 179, 1957, pages 673 - 4
MEANS, BIOCONJUGATE CHEM., vol. 1, 1990, pages 2
MEARES ET AL., ANAL. BIOCHEM., vol. 142, 1984, pages 68 - 78
MEARES ET AL., J. CANCER, vol. 1990, 1990, pages 21 - 26
MEIJS WE ET AL.: "Zirconium-labeled monoclonal antibodies and their distribution in tumor-bearing nude mice", J. NUCL. MCD., vol. 38, 1997, pages 112 - 8
MIEDERER ET AL., J. NUCL. MED., vol. 45, 2004, pages 129 - 137
MIER ET AL., BIOCONJUGATE CHEM., vol. 16, 2005, pages 240 - 237
MILLER ET AL., JOUR. OF IMMUNOLOGY, vol. 170, 2003, pages 4854 - 4861
MILLER, MARVIN J: "Syntheses and therapeutic potential of hydroxamic acid based siderophores and analogs", CHEMICAL REVIEWS, vol. 89, no. 7, 1989, pages 1563 - 1579, XP002713430, DOI: doi:10.1021/cr00097a011
MIRAGLIA: "Homogeneous cell- and bead-based assays for high throughput screening using fluorometric microvolume assay technology", J. OF BIOMOLECULAR SCREENING, vol. 4, 1999, pages 193 - 204
MIRZADEH ET AL., BIOCONJUGATE CHEM., vol. 1, 1990, pages 59 - 65
MITCHELL ET AL., J. NUCL. MED., vol. 44, 2003, pages 1105 - 1112
MIURA ET AL., BLOOD, vol. 92, 1998, pages 2815 - 2822
MIURA ET AL., GENOMICS, vol. 38, no. 3, 1996, pages 299 - 304
MONTPETIT, A.SINNETT,D., HUM. GENET., vol. 105, no. 1-2, 1999, pages 162 - 164
MOORE M. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 84, 1987, pages 9194 - 9198
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
MUELLER ET AL., EUR. J. BIOCHEM., vol. 22, 1992, pages 1621 - 1625
MULLER ET AL., EUR. J. IMMUNOL., vol. 22, no. 6, 1992, pages 1621 - 1625
MUNGALL A.J. ET AL., NATURE, vol. 425, 2003, pages 805 - 811
MURTHY ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 252, 1987, pages 549 - 560
NAGASE T. ET AL., DNA RES., vol. 7, no. 2, 2000, pages 143 - 150
NAGENGAST WB ET AL.: "In vivo VEGF imaging with radiolabeled bevacizumab in a human ovarian tumor xenograft", J NUCL MED., vol. 48, 2007, pages 1313 - 9
NAKAMUTA M. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 177, 1991, pages 34 - 39
NAKAYAMA ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 277, no. 1, 2000, pages 124 - 127
NAN, H. ET AL., INT. J. CANCER, vol. 125, no. 4, 2009, pages 909 - 917
NARITA,N. ET AL., ONCOGENE, vol. 28, no. 34, 2009, pages 3058 - 3068
NARUSC, TISSUE ANTIGENS, vol. 59, 2002, pages 512 - 519
NATURE, vol. 395, no. 6699, 1998, pages 288 - 291
NAVENOT, J.M. ET AL., MOL. PHARMACOL., vol. 75, no. 6, 2009, pages 1300 - 1306
NIKULA ET AL., J. NUCL. MED., vol. 40, 1999, pages 166 - 76
NIKULA ET AL., NUCL. MED. BIOL., vol. 22, 1995, pages 387 - 90
ODA ET AL.: "Nature Biotechnology", vol. 19, 2001, PIERCE BIOTECHNOLOGY, INC., pages: 379 - 382
O'DOWD, B.F. ET AL., FEBS LETT., vol. 394, no. 3, 1996, pages 325 - 329
OGAWA Y. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 178, 1991, pages 248 - 255
OKAMOTO Y., BIOL. CHCM., vol. 272, 1997, pages 21589 - 21596
OLAFSEN ET AL., PROTEIN ENG. DESIGN & SEL., vol. 17, no. 4, 2004, pages 315 - 323
ONCOGENE, vol. 10, no. 5, 1995, pages 897 - 905
ONCOGENE, vol. 14, no. 11, 1997, pages 1377 - 1382
OSOL, A: "Remington's Pharmaceutical Sciences. 16th Ed.", 1980
O'SULLIVAN ET AL.: "Methods in Enzym.", vol. 73, 1981, ACADEMIC PRESS, article "Methods for the Preparation of Enzyme-Antibody Conjugates for use in Enzyme Immunoassay", pages: 147 - 166
PARRISH-NOVAK J. ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 47517 - 47523
PERERA RM ET AL.: "Internalization, intracellular trafficking, and biodistribution of monoclonal antibody 806: a novel anti-epidermal growth factor receptor antibody", NEOPLASIA, vol. 9, 2007, pages 1099 - 110
PERK LR ET AL., EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, vol. 35, no. 10, 2009, pages 1857 - 1867
PERK LR ET AL.: "(89)Zr as a PET surrogate radioisotope for scouting biodistribution of the therapeutic radiomctals (90)Y and (177)Lu in tumor-bearing nude mice after coupling to the internalizing antibody cetuximab", J NUCL MED., vol. 46, 2005, pages 1898 - 906, XP008068580
PERK LR ET AL.: "Facile radio labeling of monoclonal antibodies and other proteins with zirconium-89 or gallium-68 for PET Imaging using p-isothiocyanatobenzyl-desferrioxamine", NATURE PROTOCOLS, 2008
PERK LR ET AL.: "Facile radiolabeling of monoclonal antibodies and other proteins with zirconium-89 or gallium-68 for PET Imaging using p-isothiocyanatobenzyl-desferrioxamine", NATURE PROTOCOLS, 2008
PERK LR ET AL.: "p-Isothiocyanatobcnzyl-dcsfcrrioxaminc: a new bifunctional chclatc for facile radiolabcling of monoclonal antibodies with zirconium-89 for immuno-PET imaging", EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, 2009
PERK LR ET AL.: "Preparation and evaluation of (89)Zr-Zevalin for monitoring of (90)Y-Zevalin biodistribution with positron emission tomography", EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, vol. 33, 2006, pages 1337 - 45, XP019441788, DOI: doi:10.1007/s00259-006-0160-0
PERK LR ET AL.: "Quantitative PET imaging of Met- expressing human cancer xenografts with (89)Zr-labelled monoclonal antibody DN30", EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, vol. 35, 2008, pages 1857 - 67, XP019654427, DOI: doi:10.1007/s00259-008-0774-5
PFLEIDERER, G.: "Modern Methods in Protein Chemistry", 1985, WALTER DEGRYTER, article "Chemical Modification of Proteins"
PIETRAS ET AL., ONCOGENE, vol. 9, 1994, pages 1829 - 1838
PINGAULT V. ET AL., HUM. GENET., vol. 111, 2002, pages 198 - 206
PLETNEV S. ET AL., BIOCHEMISTRY, vol. 42, 2003, pages 12617 - 12624
PLIICKTHUN: "The Pharmacology of Monoclonal Antibodies", vol. 113, 1994, SPRINGER-VERLAG, pages: 269 - 315
PLOWMAN ET AL., NATURE, vol. 366, 1993, pages 473 - 475
PLOWMAN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 1746 - 1750
PLUCKTHUN, IMMUNOL. REVS., vol. 130, 1992, pages 151 - 188
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623 - 2632
PRESTA ET AL., THROMB. HAEMOST., vol. 85, 2001, pages 379 - 389
PRESTA, CURR. OP. STRUCT. BIOL., vol. 2, 1992, pages 593 - 596
PREUD'HOMME ET AL., CLIN. EXP. IMMUNOL., vol. 90, no. 1, 1992, pages 141 - 146
PROC. NATL. ACAD. SCI. U.S.A., vol. 100, no. 7, 2003, pages 4126 - 4131
PROC. NATL. ACAD. SCI. U.S.A., vol. 93, no. 1, 1996, pages 136 - 140
PROC. NATL. ACAD. SCI. U.S.A., vol. 96, no. 20, 1999, pages 11531 - 11536
PROC. NATL. ACAD. SCI. U.S.A., vol. 98, no. 17, 2001, pages 9772 - 9777
PROC. NATL. ACAD. SCI. U.S.A., vol. 99, no. 26, 2002, pages 16899 - 16903
PUFFENBERGER E.G. ET AL., CELL, vol. 79, 1994, pages 1257 - 1266
RAVETCHKINET, ANNU. REV. IMMUNOL, vol. 9, 1991, pages 457 - 92
REITER R.E. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 95, 1998, pages 1735 - 1740
RIBAS,G. ET AL., J. IMMUNOL., vol. 163, no. 1, 1999, pages 278 - 287
RIECHMANN ET AL., J. MOL. BIOL., vol. 224, 1992, pages 487 - 499
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 329
ROBINSON MK: "Quantitativc immuno-positron emission tomography imaging of HER2-positive tumor xenografts with an iodine-124 labeled anti-HER2 diabody", CANCER RES, vol. 65, 2005, pages 1471 - 8, XP002458366, DOI: doi:10.1158/0008-5472.CAN-04-2008
ROSELLI ET AL., CANCER BIOTHERAPY & RADIOPHARMACEUTICALS, vol. 14, 1999, pages 209 - 20
ROSS ET AL., CANCER RES., vol. 62, 2002, pages 2546 - 2553
ROUX ET AL., J. IMMUNOL., vol. 161, 1998, pages 4083 - 4090
RUEGG ET AL., CANCER RES., vol. 50, 1990, pages 4221 - 4226
SAKAGUCHI ET AL., EMBO J., vol. 7, no. 11, 1988, pages 3457 - 3464
SAKAMOTO A.YANAGISAWA M. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 178, 1991, pages 656 - 663
SALANTI,G., AM. J. EPIDCMIOL., vol. 170, no. 5, 2009, pages 537 - 545
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SAMBROOKRUSSEL: "Molecular Cloning: A Laboratory Manual. 3rd Ed.", 2001
SARUP ET AL., GROWTH REGULATION, vol. 1, 1991, pages 72 - 82
SCHAEFER ET AL., ONCOGENE, vol. 15, 1997, pages 1385 - 1394
SCHECTER ET AL., NATURE, vol. 312, 1984, pages 513
SCHELLENBERGER EA ET AL.: "Optical imaging of apoptosis as a biomarker of tumor response to chemotherapy", NEOPLASIA, vol. 5, 2003, pages 187 - 92, XP009162585, DOI: doi:10.1016/S1476-5586(03)80050-7
SCHERER,S.E. ET AL., NATURE, vol. 440, no. 7082, 2006, pages 346 - 351
SCMBA, PROC. NAT. ACAD. SCI., vol. 82, 1985, pages 6497 - 6501
SCOTT ET AL., J. BIOL. CHEM., vol. 266, 1991, pages 14300 - 5
SEMBA K. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 82, 1985, pages 6497 - 6501
SERVENIUS ET AL., J. BIOL. CHEM., vol. 262, 1987, pages 8759 - 8766
SHAMIS ET AL., J. AMER. CHEM. SOC., vol. 126, 2004, pages 1726 - 1731
SHEIKH F. ET AL., J. IMMUNOL., vol. 172, 2004, pages 2006 - 2010
SHEPARD ET AL., CLIN. IMMUNOL., vol. 11, no. 3, 1991, pages 117 - 127
SHIRAISHI YICHIKAWA R: "Absorption and retention of 144 Ce and 95 Zr- 95 Nb in newborn, juvenile and adult rats", HEALTH PHYS., vol. 22, 1972, pages 373 - 8
SHIVELY ET AL., J NUCL MED, vol. 48, 2007, pages 170 - 2
SHIVELY JE.: "18F labeling for immuno-PET: where speed and contrast meet", J NUCL MED., vol. 48, 2007, pages 170 - 2
SINGH ET AL., ANAL. BIOCHEM., vol. 304, 2002, pages 147 - 15
SINGH ET AL., ANAL. BIOCHEM., vol. 304, 2002, pages 147 - 156
SINGLETON ET AL.: "Dictionary of Microbiology and Molecular Biology. 2nd Ed.", 1994, J. WILEY & SONS
SINHA S.K. ET AL., J. IMMUNOL., vol. 150, 1993, pages 5311 - 5320
SKERRA ET AL., CURR. OPINION IN IMMUNOL., vol. 5, 1993, pages 256 - 262
SLIWKOWSKI ET AL., J. BIOL. CHEM., vol. 269, no. 20, 1994, pages 14661 - 14665
STRAUSBERG ET AL., PROC. NATL. ACAD. SCI USA, vol. 99, 2002, pages 16899 - 16903
STRAUSBERG R.L. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 99, 2002, pages 16899 - 16903
SUN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 12, 2002, pages 2213 - 2215
SUN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 11, 2003, pages 1761 - 1768
SUNDARESAN G ET AL.: "124I-labeled engineered anti-CEA minibodies and diabodies allow high-contrast, antigen-specific small-animal PET imaging ofxenografts in athymic mice", J NUCL MED, vol. 44, 2003, pages 1962 - 9
SVENSSON P.J. ET AL., HUM. GENET., vol. 103, 1998, pages 145 - 148
SWARTZMAN: "A homogeneous and multiplexed immunoassay for high-throughput screening using fluorometric microvolume assay technology", ANAL. BIOCHEM., vol. 271, 1999, pages 143 - 51, XP055271863, DOI: doi:10.1006/abio.1999.4128
SWIERCZ J.M. ET AL., J. CELL BIOL., vol. 165, 2004, pages 869 - 880
SZALAI ET AL., J. AMER. CHEM. SOC., vol. 125, 2003, pages 15688 - 15689
TAIT JF ET AL.: "Improved detection of cell death in vivo with anncxin V radiolabclcd by site-specific methods", J NUCL MED., vol. 47, 2006, pages 1546 - 53
TAKCDA, S., FEBS LCTT., vol. 520, no. 1-3, 2002, pages 97 - 101
TAWARAGI Y. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 150, 1988, pages 89 - 96
THOMPSON, J.S. ET AL., SCIENCE, vol. 293, no. 5537, 2001, pages 2108 - 2111
TINIANOW JEFF N ET AL: "Site-specifically 89Zr-labeled monoclonal antibodies for ImmunoPET.", NUCLEAR MEDICINE AND BIOLOGY APR 2010 LNKD- PUBMED:20346868, vol. 37, no. 3, April 2010 (2010-04-01), pages 289 - 297, XP002615665, ISSN: 1872-9614 *
TOKI ET AL., J. ORG. CHEM., vol. 67, 2002, pages 1866 - 1872
TONNELLE ET AL., EMBO J., vol. 4, no. 11, 1985, pages 2839 - 2847
TOUCHMAN ET AL., GENOME RES., vol. 10, 2000, pages 165 - 173
TREANOR, J.J. ET AL., NATURE, vol. 382, no. 6586, 1996, pages 80 - 83
TSUKAMOTO,H. ET AL., CANCER SCI., vol. 100, no. 10, 2009, pages 1895 - 1901
TSUTSUMI M. ET AL., GENE, vol. 228, 1999, pages 43 - 49
TU ET AL., PROC. NATL. ACAD. SCI USA, vol. 96, 1999, pages 4862 - 4867
UCHIDA ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 266, 1999, pages 593 - 602
VALLETTE ET AL., NUC. ACIDS RES., vol. 17, 1989, pages 723 - 733
VAN DONGEN ET AL., ONCOLOGIST, vol. 12, 2007, pages 1379 - 89
VAN DONGEN GA ET AL.: "Immuno- PET: a navigator in monoclonal antibody development and applications", ONCOLOGIST, vol. 12, 2007, pages 1379 - 89, XP002492359, DOI: doi:10.1634/theoncologist.12-12-1379
VAN DONGEN GA ET AL.: "Immuno-PET: a navigator in monoclonal antibody development and applications", ONCOLOGIST, vol. 12, 2007, pages 1379 - 89, XP002492359, DOI: doi:10.1634/theoncologist.12-12-1379
VCRCL, J. NUCL. MCD., vol. 44, 2003, pages 1663 - 1670
VCRHCIJ J.B., AM. J. MED. GENET., vol. 108, 2002, pages 223 - 225
VEREL ET AL., J. NUCL. MED., vol. 44, 2003, pages 1663 - 1670
VEREL I ET AL.: "89Zr immuno- PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies", J NUCL MED., vol. 44, 2003, pages 1271 - 81
VEREL I ET AL.: "89Zr Immuno-PET: Comprehensive Procedures For The Production Of 89Zr-Labeled Monoclonal Antibodies", J NUCL MED, vol. 44, 2003, pages 1271 - 81
VEREL I ET AL.: "89Zr immuno-PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies", J NUCL MED., vol. 44, 2003, pages 1271 - 81
VEREL I ET AL.: "High-quality 124I-labelled monoclonal antibodies for use as PET scouting agents prior to 131I-radioimmunotherapy", EUROPEAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING, vol. 31, 2004, pages 1645 - 52
VEREL I ET AL.: "Long-lived positron emitters zirconium-89 and iodine-124 for scouting of therapeutic radioimmunoconjugates with PET", CANCER BIOTHER RADIOPHARM., vol. 18, 2003, pages 655 - 61
VEREL I ET AL.: "The promise of immuno-PET in radioimmunothcrapy", J NUCL MED, vol. 46, no. 1, 2005, pages 164S - 71S, XP008103691
VEREL IRIS ET AL: "89Zr immuno-PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies.", JOURNAL OF NUCLEAR MEDICINE : OFFICIAL PUBLICATION, SOCIETY OF NUCLEAR MEDICINE AUG 2003 LNKD- PUBMED:12902418, vol. 44, no. 8, August 2003 (2003-08-01), pages 1271 - 1281, XP002615666, ISSN: 0161-5505 *
VERMES: "A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V", J. IMMUNOL. METHODS, vol. 184, 1995, pages 39 - 51, XP004021001, DOI: doi:10.1016/0022-1759(95)00072-I
VITETTA ET AL., CANCER RESEARCH, vol. 54, 1994, pages 5301 - 5309
VON HOEGEN ET AL., J. IMMUNOL., vol. 144, no. 12, 1990, pages 4870 - 4877
WALKER, M.A., J. ORG. CHEM., vol. 60, 1995, pages 5352 - 5355
WANG L ET AL.: "Structural characterization of the maytansinoid-monoclonal antibody immunoconjugate, huN901-DM1, by mass spectrometry", PROTEIN SCI., vol. 14, 2005, pages 2436 - 46, XP008081802, DOI: doi:10.1110/ps.051478705
WCIS J.J., J. EXP. MED., vol. 167, 1988, pages 1047 - 1066
WEIS J.J. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 83, 1986, pages 5639 - 5643
WELLS ET AL., GENE, vol. 34, 1985, pages 315 - 323
WELLSLOWMAN, CURR. OPIN. STRUCT. BIOL., vol. 3, 1992, pages 355 - 362
WERTHER ET AL., J. IMMUNOL. METHODS, vol. 157, 1996, pages 4986 - 4995
WILLIAMS, CANCER BIOTHER RADIOPHARM, vol. 16, 2001, pages 25 - 35
WILSON ET AL., J. EXP. MED., vol. 173, 1991, pages 137 - 146
WONG: "Chemistry of Protein Conjugation and Cross-linking", 1991, CRC PRESS
WU ET AL., NATURE BIOTECHNOLOGY, vol. 23, no. 9, 2005, pages 1137 - 1146
XU, M.J. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 280, no. 3, 2001, pages 768 - 775
XU, X.Z. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 98, no. 19, 2001, pages 10692 - 10697
YAMAGUCHI, N. ET AL., BIOL. CHEM., vol. 269, no. 2, 1994, pages 805 - 808
YAMAMOTO ET AL., NATURE, vol. 319, 1986, pages 230 - 234
YAMAMOTO T. ET AL., NATURE, vol. 319, 1986, pages 230 - 234
YAMAMOTO,Y. ET AL., HEPATOLOGY, vol. 37, no. 3, 2003, pages 528 - 533
YU ET AL., J. IMMUNOL., vol. 148, no. 2, 1992, pages 633 - 637
ZAMMIT,D.J. ET AL., MOL. CELL. BIOL., vol. 22, no. 3, 2002, pages 946 - 952
ZHANG ET AL., ANAL. BIOCHEM., vol. 311, 2002, pages 1 - 9
ZHENG: "Caspase-3 controls both cytoplasmic and nuclear events associated with Fas-mediated apoptosis in vivo", PROC. NATL. ACAD. SCI. USA, vol. 95, 1998, pages 618 - 23
ZOLA: "Monoclonal Antibodies: A Manual of Techniques", 1987, CRC PRESS, INC., pages: 147 - 158
ZOLLER ET AL., METHODS ENZYMOL., vol. 100, 1983, pages 468 - 500
ZOLLER, M.J.SMITH, M., NUCL. ACIDS RES., vol. 10, 1982, pages 6487 - 6500

Cited By (172)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8951737B2 (en) 1996-05-06 2015-02-10 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US8940871B2 (en) 2006-03-20 2015-01-27 The Regents Of The University Of California Engineered anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting
US10981987B2 (en) 2007-07-16 2021-04-20 Genentech, Inc. Humanized anti-CD79b antibodies and immunoconjugates and methods of use
US10494432B2 (en) 2007-07-16 2019-12-03 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
USRE48558E1 (en) 2007-07-16 2021-05-18 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US11866496B2 (en) 2007-07-16 2024-01-09 Genentech, Inc. Humanized anti-CD79B antibodies and immunoconjugates and methods of use
US9527919B2 (en) 2007-09-04 2016-12-27 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting and detection
US8940298B2 (en) 2007-09-04 2015-01-27 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting and detection
US10544218B2 (en) 2008-01-31 2020-01-28 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US9896506B2 (en) 2008-01-31 2018-02-20 Genentech, Inc. Anti-CD79B antibodies and immunoconjugates and methods of use
US10517969B2 (en) 2009-02-17 2019-12-31 Cornell University Methods and kits for diagnosis of cancer and prediction of therapeutic value
US11180570B2 (en) 2009-12-02 2021-11-23 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (PSMA) and methods for their use
US8772459B2 (en) 2009-12-02 2014-07-08 Imaginab, Inc. J591 minibodies and Cys-diabodies for targeting human prostate specific membrane antigen (PSMA) and methods for their use
US9815905B2 (en) 2010-02-23 2017-11-14 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8877897B2 (en) 2010-02-23 2014-11-04 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US11873330B2 (en) 2010-06-08 2024-01-16 Genentech, Inc. Cysteine engineered antibodies and conjugates
US10604557B2 (en) 2010-06-08 2020-03-31 Genentech, Inc. Cysteine engineered antibodies and conjugates
US8911732B2 (en) 2010-12-20 2014-12-16 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US10022452B2 (en) 2010-12-20 2018-07-17 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US9719996B2 (en) 2010-12-20 2017-08-01 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US9175089B2 (en) 2012-03-30 2015-11-03 Genentech, Inc. Anti-LGR5 antibodies and immunoconjugates
WO2013149159A1 (en) 2012-03-30 2013-10-03 Genentech, Inc. Anti-lgr5 antibodies and immunoconjugates
US9597411B2 (en) 2012-05-01 2017-03-21 Genentech, Inc. Anti-PMEL17 antibodies and immunoconjugates
WO2013165940A1 (en) 2012-05-01 2013-11-07 Genentech, Inc. Anti-pmel17 antibodies and immunoconjugates
US10196454B2 (en) 2012-05-01 2019-02-05 Genentech, Inc. Anti-PMEL17 antibodies and immunoconjugates
US9056910B2 (en) 2012-05-01 2015-06-16 Genentech, Inc. Anti-PMEL17 antibodies and immunoconjugates
US10653792B2 (en) 2012-05-21 2020-05-19 Genentech, Inc. Anti-Ly6E antibodies and immunoconjugates and methods of use
US9724427B2 (en) 2012-05-21 2017-08-08 Genentech, Inc. Anti-Ly6E antibodies and immunoconjugates and methods of use
JP2017169572A (en) * 2012-05-21 2017-09-28 ジェネンテック, インコーポレイテッド ANTI-Ly6E ANTIBODIES AND IMMUNOCONJUGATES AND METHODS OF USE
JP2015528692A (en) * 2012-05-21 2015-10-01 ジェネンテック, インコーポレイテッド Anti-Ly6E antibody, immunoconjugate and method of use
US9463251B2 (en) 2012-08-02 2016-10-11 Genentech, Inc. Anti-ETBR antibodies and immunoconjugates
US9464141B2 (en) 2012-08-02 2016-10-11 Genentech, Inc. Anti-ETBR antibodies and immunoconjugates
JP2015529656A (en) * 2012-08-02 2015-10-08 ジェネンテック, インコーポレイテッド Anti-ETBR antibodies and immune complexes
JP2015528818A (en) * 2012-08-02 2015-10-01 ジェネンテック, インコーポレイテッド Anti-ETBR antibodies and immunoconjugates
US11596695B2 (en) 2013-02-08 2023-03-07 Novartis Ag Specific sites for modifying antibodies to make immunoconjugates
US10137212B2 (en) * 2013-03-13 2018-11-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tetrahydroxamate chelators of zirconium89 and niobium90 for use in diagnostic applications
US10150813B2 (en) 2013-03-14 2018-12-11 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2014159835A1 (en) 2013-03-14 2014-10-02 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
US9562099B2 (en) 2013-03-14 2017-02-07 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
EP3299391A1 (en) 2013-03-14 2018-03-28 Genentech, Inc. Anti-b7-h4 antibodies and immunoconjugates
US11230600B2 (en) 2013-03-14 2022-01-25 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US10246515B2 (en) 2013-09-17 2019-04-02 Genentech, Inc. Methods of treating hedgehog-related diseases with an anti-LGR5 antibody
WO2015042108A1 (en) 2013-09-17 2015-03-26 Genentech, Inc. Methods of using anti-lgr5 antibodies
US9290578B2 (en) 2013-10-21 2016-03-22 Genentech, Inc. Anti-Ly6E antibodies and methods of use
US10889651B2 (en) 2013-10-21 2021-01-12 Genentech, Inc. Anti-Ly6E antibodies and methods of use
US10066022B2 (en) 2013-10-21 2018-09-04 Genentech, Inc. Anti-Ly6E antibodies and methods of use
WO2015089344A1 (en) 2013-12-13 2015-06-18 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
EP3461845A1 (en) 2013-12-13 2019-04-03 Genentech, Inc. Anti-cd33 antibodies and immunoconjugates
US10533058B2 (en) 2013-12-16 2020-01-14 Genentech Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US11692043B2 (en) 2013-12-16 2023-07-04 Medimmune Limited Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015112909A1 (en) 2014-01-24 2015-07-30 Genentech, Inc. Methods of using anti-steap1 antibodies and immunoconjugates
WO2015179658A2 (en) 2014-05-22 2015-11-26 Genentech, Inc. Anti-gpc3 antibodies and immunoconjugates
WO2015191715A1 (en) 2014-06-11 2015-12-17 Genentech, Inc. Anti-lgr5 antibodies and uses thereof
US11584927B2 (en) 2014-08-28 2023-02-21 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified T-cells
US10556966B2 (en) 2014-09-12 2020-02-11 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
US10059768B2 (en) 2014-09-12 2018-08-28 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
US10179820B2 (en) 2014-09-12 2019-01-15 Genentech, Inc. Anti-HER2 antibodies and immunoconjugates
EP3782654A1 (en) 2014-09-12 2021-02-24 Genentech, Inc. Anti-her2 antibodies and immunoconjugates
US10077318B2 (en) 2014-09-12 2018-09-18 Genentech, Inc. Cysteine engineered antibodies and conjugates
EP3693391A1 (en) 2014-09-12 2020-08-12 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
US11286302B2 (en) 2014-09-12 2022-03-29 Genentech, Inc. Anti-B7-H4 antibodies and immunoconjugates
WO2016040868A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anti-cll-1 antibodies and immunoconjugates
US11000510B2 (en) 2014-09-23 2021-05-11 Genentech, Inc. Methods of using anti-CD79b immunoconjugates
EP3689910A2 (en) 2014-09-23 2020-08-05 F. Hoffmann-La Roche AG Method of using anti-cd79b immunoconjugates
US12016842B2 (en) 2014-09-23 2024-06-25 Genentech, Inc. Methods of using anti-CD79b immunoconjugates
EP3207027A4 (en) * 2014-10-16 2018-04-18 The University of Melbourne Novel imaging composition and uses thereof
EP3543226A1 (en) * 2014-10-16 2019-09-25 The University of Melbourne Novel imaging composition and uses thereof
US10011658B2 (en) 2015-04-03 2018-07-03 Eureka Therapeutics, Inc. Constructs targeting AFP peptide/MHC complexes and uses thereof
US11254744B2 (en) 2015-08-07 2022-02-22 Imaginab, Inc. Antigen binding constructs to target molecules
US10632196B2 (en) 2015-10-02 2020-04-28 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US10639373B2 (en) 2015-10-02 2020-05-05 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US10058613B2 (en) 2015-10-02 2018-08-28 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US11078282B2 (en) 2016-04-15 2021-08-03 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
WO2017181152A2 (en) 2016-04-15 2017-10-19 Alpine Immune Sciences, Inc. Cd80 variant immunomodulatory proteins and uses thereof
US11479609B2 (en) 2016-04-15 2022-10-25 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
WO2017181148A2 (en) 2016-04-15 2017-10-19 Alpine Immune Sciences, Inc. Icos ligand variant immunomodulatory proteins and uses thereof
US11149088B2 (en) 2016-04-15 2021-10-19 Bioatla, Inc. Anti-Axl antibodies, antibody fragments and their immunoconjugates and uses thereof
US11359022B2 (en) 2016-04-15 2022-06-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11498967B2 (en) 2016-04-15 2022-11-15 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US12110339B2 (en) 2016-04-15 2024-10-08 Alpine Immune Sciences, Inc. ICOS ligand variant immunomodulatory proteins and uses thereof
US11897959B2 (en) 2016-04-15 2024-02-13 Bioatla, Inc. Anti-AXL antibodies, antibody fragments and their immunoconjugates and uses thereof
US10882914B2 (en) 2016-04-15 2021-01-05 Alpine Immune Sciences, Inc. ICOS ligand variant immunomodulatory proteins and uses thereof
EP4122958A1 (en) 2016-05-13 2023-01-25 BioAtla, Inc. Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
US11879011B2 (en) 2016-05-13 2024-01-23 Bioatla, Inc. Anti-ROR2 antibodies, antibody fragments, their immunoconjucates and uses thereof
WO2017197234A1 (en) 2016-05-13 2017-11-16 Bioatla, Llc Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
US11254742B2 (en) 2016-05-13 2022-02-22 Bioatla, Inc. Anti-Ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
WO2017223405A1 (en) 2016-06-24 2017-12-28 Genentech, Inc. Anti-polyubiquitin multispecific antibodies
WO2018022946A1 (en) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Cd155 variant immunomodulatory proteins and uses thereof
US11834490B2 (en) 2016-07-28 2023-12-05 Alpine Immune Sciences, Inc. CD112 variant immunomodulatory proteins and uses thereof
US11471488B2 (en) 2016-07-28 2022-10-18 Alpine Immune Sciences, Inc. CD155 variant immunomodulatory proteins and uses thereof
WO2018022945A1 (en) 2016-07-28 2018-02-01 Alpine Immune Sciences, Inc. Cd112 variant immunomodulatory proteins and uses thereof
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
US11142578B2 (en) 2016-11-16 2021-10-12 Regeneron Pharmaceuticals, Inc. Anti-MET antibodies, bispecific antigen binding molecules that bind MET, and methods of use thereof
US11679166B2 (en) 2016-11-18 2023-06-20 Astellas Pharma Inc. Anti-human MUC1 antibody Fab fragment
WO2018091724A1 (en) 2016-11-21 2018-05-24 Cureab Gmbh Anti-gp73 antibodies and immunoconjugates
EP4015532A1 (en) 2016-11-21 2022-06-22 cureab GmbH Anti-gp73 antibodies and immunoconjugates
WO2018102682A1 (en) 2016-12-01 2018-06-07 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-pd-l1 antibodies for immuno-pet imaging
US12053534B2 (en) 2016-12-01 2024-08-06 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-PD-L1 antibodies for immuno-PET imaging
US10736976B2 (en) 2016-12-01 2020-08-11 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-PD-L1 antibodies for immuno-PET imaging
EP4279136A2 (en) 2016-12-03 2023-11-22 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
WO2018102787A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
US11274157B2 (en) 2017-01-12 2022-03-15 Eureka Therapeutics, Inc. Constructs targeting histone H3 peptide/MHC complexes and uses thereof
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies
US10905784B2 (en) 2017-02-10 2021-02-02 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-LAG3 antibodies for immuno-PET imaging
WO2018148476A1 (en) 2017-02-10 2018-08-16 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-lag3 antibodies for immuno-pet imaging
US11511001B2 (en) 2017-02-10 2022-11-29 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-LAG3 antibodies for immuno-PET imaging
WO2018170026A2 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Cd80 variant immunomodulatory proteins and uses thereof
EP4306537A2 (en) 2017-03-16 2024-01-17 Alpine Immune Sciences, Inc. Pd-l1 variant immunomodulatory proteins and uses thereof
US11117948B2 (en) 2017-03-16 2021-09-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
WO2018170021A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l1 variant immunomodulatory proteins and uses thereof
US11117949B2 (en) 2017-03-16 2021-09-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11096988B2 (en) 2017-03-16 2021-08-24 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11117950B2 (en) 2017-03-16 2021-09-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11732022B2 (en) 2017-03-16 2023-08-22 Alpine Immune Sciences, Inc. PD-L2 variant immunomodulatory proteins and uses thereof
US11230588B2 (en) 2017-03-16 2022-01-25 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
WO2018170023A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l2 variant immunomodulatory proteins and uses thereof
US11639375B2 (en) 2017-03-16 2023-05-02 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11447564B2 (en) 2017-04-26 2022-09-20 Eureka Therapeutics, Inc. Constructs specifically recognizing glypican 3 and uses thereof
US11740231B2 (en) 2017-06-02 2023-08-29 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
WO2018223098A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
US11667724B2 (en) 2017-07-07 2023-06-06 Astellas Pharma Inc. Anti-human CEACAM5 antibody Fab fragment
US10730944B2 (en) 2017-07-24 2020-08-04 Regeneron Pharmaceuticals, Inc. Anti-CD8 antibodies and uses thereof
US12077587B2 (en) 2017-07-24 2024-09-03 Regeneron Pharmaceuticals, Inc. Anti-CD8 antibodies and uses thereof
WO2019023148A1 (en) 2017-07-24 2019-01-31 Regeneron Pharmaceuticals, Inc. Anti-cd8 antibodies and uses thereof
US11525001B2 (en) 2017-07-24 2022-12-13 Regeneron Pharmaceuticals, Inc. Anti-CD8 antibodies and uses thereof
EP4442268A2 (en) 2017-10-10 2024-10-09 Alpine Immune Sciences, Inc. Ctla-4 variant immunomodulatory proteins and uses thereof
US11753458B2 (en) 2017-10-10 2023-09-12 Alpine Immune Sciences, Inc. CTLA-4 variant immunomodulatory proteins and uses thereof
EP4219540A2 (en) 2017-10-10 2023-08-02 Alpine Immune Sciences, Inc. Ctla-4 variant immunomodulatory proteins and uses thereof
WO2019074983A1 (en) 2017-10-10 2019-04-18 Alpine Immune Sciences, Inc. Ctla-4 variant immunomodulatory proteins and uses thereof
WO2019079520A2 (en) 2017-10-18 2019-04-25 Alpine Immune Sciences, Inc. Variant icos ligand immunomodulatory proteins and related compositions and methods
US11613566B2 (en) 2017-10-18 2023-03-28 Alpine Immune Sciences, Inc. Variant ICOS ligand immunomodulatory proteins and related compositions and methods
US11564946B2 (en) 2017-11-01 2023-01-31 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
US12031975B2 (en) 2017-11-01 2024-07-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
WO2019089858A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
WO2019089848A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019109053A1 (en) 2017-12-01 2019-06-06 Juno Therapeutics, Inc. Methods for dosing and for modulation of genetically engineered cells
US12065476B2 (en) 2018-06-15 2024-08-20 Alpine Immune Sciences, Inc. PD-1 variant immunomodulatory proteins and uses thereof
WO2019241758A1 (en) 2018-06-15 2019-12-19 Alpine Immune Sciences, Inc. Pd-1 variant immunomodulatory proteins and uses thereof
US11993661B2 (en) 2018-06-18 2024-05-28 Eureka Therapeutics, Inc. Constructs targeting prostate-specific membrane antigen (PSMA) and uses thereof
US12065503B2 (en) 2018-10-10 2024-08-20 Astellas Pharma Inc. Pharmaceutical composition containing tagged site-antihuman antibody fab fragment complex
US11945869B2 (en) 2018-10-11 2024-04-02 Inhibrx, Inc. PD-1 single domain antibodies and therapeutic compositions thereof
WO2020076977A2 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Dll3 single domain antibodies and therapeutic compositions thereof
WO2020077257A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Pd-1 single domain antibodies and therapeutic compositions thereof
US11208485B2 (en) 2018-10-11 2021-12-28 Inhibrx, Inc. PD-1 single domain antibodies and therapeutic compositions thereof
WO2020076992A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. 5t4 single domain antibodies and therapeutic compositions thereof
WO2020076970A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. B7h3 single domain antibodies and therapeutic compositions thereof
WO2020113141A2 (en) 2018-11-30 2020-06-04 Alpine Immune Sciences, Inc. Cd86 variant immunomodulatory proteins and uses thereof
EP4427810A2 (en) 2018-11-30 2024-09-11 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020117257A1 (en) 2018-12-06 2020-06-11 Genentech, Inc. Combination therapy of diffuse large b-cell lymphoma comprising an anti-cd79b immunoconjugates, an alkylating agent and an anti-cd20 antibody
WO2020205623A1 (en) 2019-03-29 2020-10-08 Rakuten Medical, Inc. Methods for photoimmunotherapy and related biomarkers
WO2020232169A1 (en) 2019-05-14 2020-11-19 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat follicular lymphoma
WO2020257681A1 (en) 2019-06-21 2020-12-24 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind psma and cd3 in combination with 4-1bb co-stimulation
WO2020257604A1 (en) 2019-06-21 2020-12-24 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind muc16 and cd3 in combination with 4-1bb co-stimulation
US12091460B2 (en) 2019-06-21 2024-09-17 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind MUC16 and CD3 in combination with 4-1BB co-stimulation
US11896682B2 (en) 2019-09-16 2024-02-13 Regeneron Pharmaceuticals, Inc. Radiolabeled MET binding proteins for immuno-PET imaging and methods of use thereof
WO2021055350A1 (en) 2019-09-16 2021-03-25 Regeneron Pharmaceuticals, Inc. Radiolabeled met binding proteins for immuno-pet imaging
WO2021076196A1 (en) 2019-10-18 2021-04-22 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2021113776A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
WO2021113780A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to gprc5d-targeted binding domains and related compositions and methods
WO2021155071A1 (en) 2020-01-29 2021-08-05 Inhibrx, Inc. Cd28 single domain antibodies and multivalent and multispecific constructs thereof
WO2021217051A1 (en) 2020-04-24 2021-10-28 Genentech, Inc. Methods of using anti-cd79b immunoconjugates
WO2022029660A1 (en) 2020-08-05 2022-02-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
WO2022147463A2 (en) 2020-12-31 2022-07-07 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
WO2022241446A1 (en) 2021-05-12 2022-11-17 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023019092A1 (en) 2021-08-07 2023-02-16 Genentech, Inc. Methods of using anti-cd79b immunoconjugates to treat diffuse large b-cell lymphoma
WO2023034750A1 (en) 2021-08-30 2023-03-09 Genentech, Inc. Anti-polyubiquitin multispecific antibodies
WO2023172883A1 (en) 2022-03-07 2023-09-14 Alpine Immune Sciences, Inc. Immunomodulatory proteins of variant cd80 polypeptides, cell therapies thereof and related methods and uses
WO2024151515A2 (en) 2023-01-09 2024-07-18 Odyssey Therapeutics, Inc. Anti-tnfr2 antigen-binding proteins and uses thereof
WO2024173876A1 (en) 2023-02-17 2024-08-22 Regeneron Pharmaceuticals, Inc. Radiolabeled anti-lag3 antibodies for immuno-pet imaging
WO2024192065A1 (en) 2023-03-14 2024-09-19 Odyssey Therapeutics, Inc. Anti-cd25 antigen-binding proteins and uses thereof

Also Published As

Publication number Publication date
RU2562862C2 (en) 2015-09-10
JP2013510093A (en) 2013-03-21
CA2780216A1 (en) 2011-05-12
EP2496270A1 (en) 2012-09-12
CN102596260B (en) 2017-04-05
KR20120102625A (en) 2012-09-18
CN102596260A (en) 2012-07-18
BR112012007774A2 (en) 2016-11-22
US20100111856A1 (en) 2010-05-06
KR20170136652A (en) 2017-12-11
US20150017094A1 (en) 2015-01-15
MX2012005211A (en) 2012-06-13
EP2496270B1 (en) 2018-01-31
MX340674B (en) 2016-07-20
JP5850843B2 (en) 2016-02-03
RU2012123007A (en) 2013-12-10

Similar Documents

Publication Publication Date Title
EP2496270B1 (en) Zirconium-radiolabeled, cysteine engineered antibody conjugates
US11873330B2 (en) Cysteine engineered antibodies and conjugates
AU2005286607B2 (en) Cysteine engineered antibodies and conjugates
MX2007003404A (en) Cysteine engineered antibodies and conjugates

Legal Events

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

Ref document number: 201080050134.4

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10776064

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010776064

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2012537224

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 3939/CHENP/2012

Country of ref document: IN

Ref document number: MX/A/2012/005211

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2780216

Country of ref document: CA

Ref document number: 20127011607

Country of ref document: KR

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2012123007

Country of ref document: RU

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012007774

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012007774

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20120404