WO2017201449A1 - Protac antibody conjugates and methods of use - Google Patents

Protac antibody conjugates and methods of use Download PDF

Info

Publication number
WO2017201449A1
WO2017201449A1 PCT/US2017/033611 US2017033611W WO2017201449A1 WO 2017201449 A1 WO2017201449 A1 WO 2017201449A1 US 2017033611 W US2017033611 W US 2017033611W WO 2017201449 A1 WO2017201449 A1 WO 2017201449A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
alkyl
seq
amino acid
alkylene
Prior art date
Application number
PCT/US2017/033611
Other languages
French (fr)
Inventor
Thomas Pillow
Jack SADOWSKY
Leanna STABEN
Steven Staben
Binqing Wei
Ingrid WERTZ
Pragya ADHIKARI
Nicole Blaquiere
Peter Dragovich
Wayne Fairbrother
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 PL17728325T priority Critical patent/PL3458101T3/en
Priority to EP17728325.6A priority patent/EP3458101B1/en
Priority to JP2018560504A priority patent/JP2019522633A/en
Priority to CN201780030893.6A priority patent/CN109152843A/en
Priority to ES17728325T priority patent/ES2858151T3/en
Publication of WO2017201449A1 publication Critical patent/WO2017201449A1/en
Priority to US16/194,897 priority patent/US11666581B2/en
Priority to JP2022107984A priority patent/JP2022126886A/en
Priority to US18/310,913 priority patent/US20230330102A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/93Ligases (6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y203/00Acyltransferases (2.3)
    • C12Y203/02Aminoacyltransferases (2.3.2)
    • C12Y203/02007Aspartyltransferase (2.3.2.7)

Definitions

  • BACKGROUND Cell maintenance and normal function requires controlled degradation of cellular proteins.
  • degradation of regulatory proteins triggers events in the cell cycle, such as DNA replication, chromosome segregation, etc. Accordingly, such degradation of proteins has implications for the cell’s proliferation, differentiation, and death.
  • inhibitors of proteins can block or reduce protein activity in a cell
  • protein degradation in a cell can also reduce activity or remove altogether the target protein.
  • Utilizing a cell’s protein degradation pathway can, therefore, provide a means for reducing or removing protein activity.
  • One of the cell’s major degradation pathways is known as the ubiquitin-proteasome system.
  • a protein is marked for degradation by the proteasome by ubiquitinating the protein.
  • the ubiqitinization of the protein is accomplished by an E3 ubiquitin ligase that binds to a protein and adds ubiquitin molecules to the protein.
  • the E3 ubiquitin ligase is part of a pathway that includes E1 and E2 ubiquitin ligases, which make ubiquitin available to the E3 ubiquitin ligase to add to the protein.
  • PROTACs bring together an E3 ubiquitin ligase with a protein that is to be targeted for degradation.
  • the PROTAC is comprised of a group that binds to an E3 ubiquitin ligase and a group that binds to the protein one wishes to degrade. These groups are typically connected with a linker. This molecular construct can bring the E3 ubiquitin ligase in proximity with the protein so that it is ubiquitinated and marked for degradation.
  • Targeted delivery using antibody-PROTAC conjugates can enhance delivery of PROTACs to particular cells using the specificity of an antibody and can also enhance the pharmacokinetics of delivery of PROTACs to cells relative to other modes of administration of PROTACs, such as infusion.
  • PROTAC-antibody conjugate having the formula:
  • D is a PROTAC having the structure E3LB ⁇ L2 ⁇ PB; wherein, E3LB is an E3 ligase binding group covalently bound to L2; L2 is a linker covalently bound to E3LB and PB; PB is a protein binding group covalently bound to L2; Ab is an antibody covalently bound to L1; L1 is a linker, covalently bound to Ab and to D; and p has a value from about 1 to about 8.
  • compositions comprising a PAC, and one or more pharmaceutically acceptable excipients.
  • Another aspect of the subject matter described herein is the use of a PAC in methods of treating conditions and diseases by administering to a subject a pharmaceutical composition comprising a PAC.
  • FIGURES Figure 1 shows the detection of ER- ⁇ by Western blot for PROTAC (without Ab), compound P1, and PROTAC-antibody conjugates (PACs) PAC1, and PAC2.
  • Figure 2 depicts the quantitation of ER ⁇ as determined by fluorescence intensity for Endox-XIAP PACs treated for 3 days in an engineered HER2-MCF7 line.
  • Media 10% CS- FBS in phenol red free-RPMI.
  • PACs antibody-proteolysis targeting chimera conjugates
  • the subject matter described herein utilizes antibody targeting to direct a PROTAC to a target cell or tissue. As described herein, connecting an antibody to a PROTAC to form a PAC has been shown to deliver the PROTAC to a target cell or tissue.
  • a cell that expresses an antigen can be targeted by an antigen specific PAC, whereby the PROTAC portion of the PAC is delivered intracellularly to the target cell.
  • PACs that comprise an antibody directed to an antigen that is not found on the cell do not result in significant intracellualr delivery of the PROTAC to the cell.
  • the subject matter described herein is directed to PROTAC-antibody conjugate (PAC) compositions that result in the ubiquitination of a target protein and subsequent degradation of the protein.
  • the compositions comprise an antibody covalently linked to a linker (L1), which is covalently linked at any available point of attachment to a PROTAC, in which the PROTAC comprises an E3 ubiquitin ligase binding (E3LB) moiety, wherein the E3LB moiety recognizes a E3 ubiquitin ligase protein and a protein binding moiety (PB) that recognizes a target protein.
  • E3LB E3 ubiquitin ligase binding
  • PB protein binding moiety
  • PROTAC refers to proteolysis-targeting chimera molecules having generally three components, an E3 ubiquitin ligase binding group (E3LB), a linker L2, and a protein binding group (PB).
  • E3LB E3 ubiquitin ligase binding group
  • PB protein binding group
  • residue refers to a component that is covalently bound or linked to another component.
  • a“residue of a PROTAC” refers to a PROTAC that is covalently linked to one or more groups such as a Linker L2, which itself can be optionally further linked to an antibody.
  • covalently bound or“covalently linked” refers to a chemical bond formed by sharing of one or more pairs of electrons.
  • peptidomimetic or PM as used herein means a non-peptide chemical moiety.
  • Peptides are short chains of amino acid monomers linked by peptide (amide) bonds, the covalent chemical bonds formed when the carboxyl group of one amino acid reacts with the amino group of another.
  • the shortest peptides are dipeptides, consisting of 2 amino acids joined by a single peptide bond, followed by tripeptides, tetrapeptides, etc.
  • a peptidomimetic chemical moiety includes non-amino acid chemical moieties.
  • a peptidomimetic chemical moiety may also include one or more amino acid that are separated by one or more non- amino acid chemical units.
  • a peptidomimetic chemical moiety does not contain in any portion of its chemical structure two or more adjacent amino acids that are linked by peptide bonds.
  • 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 (complementary determining regions) on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody.
  • An antibody includes a full-length immunoglobulin molecule or an immunologically active portion of a full-length immunoglobulin molecule, i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease.
  • the immunoglobulin disclosed herein can be of any type (e.g., IgG, IgE, IgM, IgD, and IgA), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule.
  • the immunoglobulins can be derived from any species. In one aspect, however, the immunoglobulin is of human, murine, or rabbit origin.
  • antibody fragment(s) as used herein comprises a portion of a full length antibody, generally the antigen binding or variable region thereof.
  • antibody fragments include Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies;
  • 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.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • 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 subject matter described herein 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.
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
  • The“class” of an antibody refers to the type of constant domain or constant region possessed by its heavy chain.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the term“intact antibody” as used herein is one comprising a VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1, CH2 and CH3.
  • the constant domains may be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variant thereof.
  • the intact antibody may have one or more“effector functions” which refer to those biological activities attributable to the Fc constant region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell- mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.
  • Fc region as used hererin means a C-terminal region of an
  • immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
  • variable domain residues other than hypervariable region (HVR) residues The FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2- H2(L2)-FR3-H3(L3)-FR4.
  • full length antibody “intact antibody,” and“whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • A“human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody- encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • A“humanized” antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • A“humanized form” of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization.
  • an“isolated antibody” is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC).
  • electrophoretic e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • An“isolated nucleic acid” refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an antibody refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • A“naked antibody” refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel.
  • the naked antibody may be present in a pharmaceutical formulation.
  • “Native antibodies” refer to naturally occurring immunoglobulin molecules with varying structures.
  • native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3). Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain.
  • VH variable heavy domain
  • VL variable region
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office,
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • 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., IgG1, IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • 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).
  • human consensus framework refers to a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NIH Publication 91-3242, Bethesda MD (1991), vols.1-3.
  • the subgroup is subgroup kappa I as in Kabat et al., supra.
  • the subgroup is subgroup III as in Kabat et al., supra.
  • An“acceptor human framework” for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework“derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • variable region or“variable domain” as used herein refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J. Immunol.150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
  • hypervariable region refers to each of the regions of an antibody variable domain that are hypervariable in sequence and/or form structurally defined loops (“hypervariable loops”).
  • native four-chain antibodies comprise six HVRs; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3).
  • HVRs generally comprise amino acid residues from the hypervariable loops and/or from the “complementarity determining regions” (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition.
  • CDRs complementarity determining regions
  • Exemplary hypervariable loops occur at amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3).
  • Exemplary CDRs CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3 occur at amino acid residues 24-34 of L1, 50-56 of L2, 89-97 of L3, 31-35B of H1, 50-65 of H2, and 95-102 of H3.
  • CDRs generally comprise the amino acid residues that form the hypervariable loops.
  • CDRs also comprise“specificity determining residues,” or“SDRs,” which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated-CDRs, or a- CDRs.
  • Exemplary a-CDRs (a-CDR-L1, a-CDR-L2, a-CDR-L3, a-CDR-H1, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31-34 of L1, 50-55 of L2, 89-96 of L3, 31-35B of H1, 50-58 of H2, and 95-102 of H3.
  • HVR residues and other residues in the variable domain are numbered herein according to Kabat et al., supra.
  • “Effector functions” refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: C1q binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
  • epitope refers to the particular site on an antigen molecule to which an antibody binds.
  • The“epitope 4D5” or“4D5 epitope” or“4D5” is the region in the extracellular domain of HER2 to which the antibody 4D5 (ATCC CRL 10463) and trastuzumab bind. This epitope is close to the transmembrane domain of HER2, and within domain IV of HER2.
  • a routine cross-blocking assay such as that described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988), can be performed.
  • epitope mapping can be performed to assess whether the antibody binds to the 4D5 epitope of HER2 (e.g. any one or more residues in the region from about residue 550 to about residue 610, inclusive, of HER2 (SEQ ID NO: 39).
  • The“epitope 2C4” or“2C4 epitope” is the region in the extracellular domain of HER2 to which the antibody 2C4 binds.
  • a routine cross-blocking assay such as that described in Antibodies, A
  • Epitope 2C4 comprises residues from domain II in the extracellular domain of HER2.
  • the 2C4 antibody and pertuzumab bind to the extracellular domain of HER2 at the junction of domains I, II and III (Franklin et al. Cancer Cell 5:317-328 (2004)).
  • Bind refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
  • binding affinity refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • an antibody as described herein has dissociation constant (Kd) of ⁇ 1 ⁇ M, ⁇ 100 nM, ⁇ 10 nM, ⁇ 5 nm, ⁇ 4 nM, ⁇ 3 nM, ⁇ 2 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g., 10 -8 M or less, e.g. from 10 -8 M to 10 -13 M, e.g., from 10 -9 M to 10 -13 M).
  • Kd dissociation constant
  • An“affinity matured” antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
  • HVRs hypervariable regions
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self- replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as “expression vectors.”
  • 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.
  • amino acid as used herein means glycine, alanine, valine, leucine, isoleucine, phenylalanine, proline, serine, threonine, tyrosine, cysteine, methionine, lysine, arginine, histidine, tryptophan, aspartic acid, glutamic acid, asparagine, glutamine or citrulline.
  • Linker means a chemical moiety comprising a chain of atoms that covalently attaches a PROTAC moiety to an antibody, or a component of a PROTAC to another component of the PROTAC.
  • a linker is a divalent radical, specified as L1 or L2.
  • A“patient” or“individual” or“subject” is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • the patient, individual, or subject is a human.
  • the patient may be a“cancer patient,” i.e. one who is suffering or at risk for suffering from one or more symptoms of cancer.
  • A“patient population” refers to a group of cancer patients. Such populations can be used to demonstrate statistically significant efficacy and/or safety of a drug.
  • A“relapsed” patient is one who has signs or symptoms of cancer after remission.
  • the patient has relapsed after adjuvant or neoadjuvant therapy.
  • a cancer or biological sample which“displays HER expression, amplification, or activation” is one which, in a diagnostic test, expresses (including overexpresses) a HER receptor, has amplified HER gene, and/or otherwise demonstrates activation or
  • Neoadjuvant therapy or“preoperative therapy” herein refers to therapy given prior to surgery.
  • the goal of neoadjuvant therapy is to provide immediate systemic treatment, potentially eradicating micrometastases that would otherwise proliferate if the standard sequence of surgery followed by systemic therapy were followed.
  • Neoadjuvant therapy may also help to reduce tumor size thereby allowing complete resection of initially unresectable tumors or preserving portions of the organ and its functions.
  • neoadjuvant therapy permits an in vivo assessment of drug efficacy, which may guide the choice of subsequent treatments.
  • adjuvant therapy herein refers to therapy given after definitive surgery, where no evidence of residual disease can be detected, so as to reduce the risk of disease recurrence.
  • the goal of adjuvant therapy is to prevent recurrence of the cancer, and therefore to reduce the chance of cancer-related death.
  • adjuvant therapy herein specifically excludes
  • Definitive surgery includes, for example, procedures, surgical or otherwise, that result in removal or resection of the tumor, including those that result in the removal or resection of all grossly visible tumor.
  • Definitive surgery includes, for example, complete or curative resection or complete gross resection of the tumor.
  • Definitive surgery includes procedures that occur in one or more stages, and includes, for example, multi-stage surgical procedures where one or more surgical or other procedures are performed prior to resection of the tumor.
  • Definitive surgery includes procedures to remove or resect the tumor including involved organs, parts of organs and tissues, as well as surrounding organs, such as lymph nodes, parts of organs, or tissues. Removal may be incomplete such that tumor cells might remain even though undetected.
  • “Survival” refers to the patient remaining alive, and includes disease free survival (DFS), progression free survival (PFS) and overall survival (OS). Survival can be estimated by the Kaplan-Meier method, and any differences in survival are computed using the stratified log-rank test.
  • DFS disease free survival
  • PFS progression free survival
  • OS overall survival
  • PFS progression-Free Survival
  • DFS Disease free survival
  • Disease free survival refers to the patient remaining alive, without return of the cancer, for a defined period of time such as about 1 year, about 2 years, about 3 years, about 4 years, about 5 years, about 10 years, etc., from initiation of treatment or from initial diagnosis.
  • DFS is analyzed according to the intent-to-treat principle, i.e., patients are evaluated on the basis of their assigned therapy.
  • the events used in the analysis of DFS can include local, regional and distant recurrence of cancer, occurrence of secondary cancer, and death from any cause in patients without a prior event (e.g, breast cancer recurrence or second primary cancer).
  • “Overall survival” refers to the patient remaining alive for a defined period of time, such as about 1 year, about 2 years, about 3 years, about 4 years, about 5 years, about 10 years, etc., from initiation of treatment or from initial diagnosis.
  • extending survival is meant increasing DFS and/or OS in a treated patient relative to an untreated patient, or relative to a control treatment protocol. Survival is monitored for at least about six months, or at least about 1 year, or at least about 2 years, or at least about 3 years, or at least about 4 years, or at least about 5 years, or at least about 10 years, etc., following the initiation of treatment or following the initial diagnosis.
  • “monotherapy” is meant a therapeutic regimen that includes only a single therapeutic agent for the treatment of the cancer or tumor during the course of the treatment period.
  • maintenance therapy is meant a therapeutic regimen that is given to reduce the likelihood of disease recurrence or progression. Maintenance therapy can be provided for any length of time, including extended time periods up to the life-span of the subject.
  • Maintenance therapy can be provided after initial therapy or in conjunction with initial or additional therapies. Dosages used for maintenance therapy can vary and can include diminished dosages as compared to dosages used for other types of therapy.
  • the terms“host cell,”“host cell line,” and“host cell culture” are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include“transformants” and“transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • cancer and“cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation.
  • A“tumor” comprises one or more cancerous cells. Examples of cancer are provided elsewhere herein.
  • A“HER2-positive” cancer comprises cancer cells which have higher than normal levels of HER2.
  • Examples of HER2-positive cancer include HER2-positive breast cancer and HER2-positive gastric cancer.
  • HER2-positive cancer has an
  • IHC immunohistochemistry
  • ISH in situ hybridization
  • EBC early stage breast cancer
  • early breast cancer or“early breast cancer” is used herein to refer to breast cancer that has not spread beyond the breast or the axillary lymph nodes. This includes ductal carcinoma in situ and stage I, stage IIA, stage IIB, and stage IIIA breast cancers.
  • a Stage 0 cancer is an in situ lesion
  • a Stage I cancer is small localized tumor
  • a Stage II and III cancer is a local advanced tumor which exhibits involvement of the local lymph nodes
  • a Stage IV cancer represents metastatic cancer.
  • the specific stages for each type of tumor are known to the skilled clinician.
  • metastatic breast cancer means the state of breast cancer where the cancer cells are transmitted from the original site to one or more sites elsewhere in the body, by the blood vessels or lymphatics, to form one or more secondary tumors in one or more organs besides the breast.
  • An“advanced” cancer is one which has spread outside the site or organ of origin, either by local invasion or metastasis. Accordingly, the term“advanced” cancer includes both locally advanced and metastatic disease.
  • A“recurrent” cancer is one which has regrown, either at the initial site or at a distant site, after a response to initial therapy, such as surgery.
  • A“locally recurrent” cancer is cancer that returns after treatment in the same place as a previously treated cancer.
  • An“operable” or“resectable” cancer is cancer which is confined to the primary organ and suitable for surgery (resection).
  • A“non-resectable” or “unresectable” cancer is not able to be removed (resected) by surgery.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.
  • Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal
  • chemotherapeutic agent refers to a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa;
  • ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine;
  • acetogenins especially bullatacin and bullatacinone
  • dronabinol, MARINOL® beta-lapachone
  • lapachol colchicines
  • betulinic acid a camptothecin (including the synthetic analogue topotecan (HYCAMTIN®), CPT-11
  • spongistatin nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard;
  • nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard;
  • nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gamma1I and calicheamicin omegaI1 (see, e.g., Nicolaou et al., Angew. Chem Intl. Ed.
  • antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gamma1I and calicheamicin omegaI1 (see, e.g., Nicolaou et al., Angew. Chem Intl. Ed.
  • CAELYX® deoxydoxorubicin
  • epirubicin epirubicin
  • esorubicin idarubicin
  • marcellomycin mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin
  • anti-metabolites such as methotrexate, gemcitabine (GEMZAR®), tegafur (UFTORAL®), capecitabine (XELODA®), an epothilone, and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine;
  • amsacrine bestrabucil
  • bisantrene edatraxate
  • defofamine demecolcine
  • diaziquone diaziquone
  • elfornithine elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; 2- ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 22’2’trichlorotriethylamine; trichothecenes (especially T 2 toxin verracurin A roridin A and anguidine); urethan; vindesine (ELDISINE
  • ELOXATIN® etoposide
  • carboplatin etoposide
  • vincas which prevent tubulin polymerization from forming microtubules, including vinblastine (VELBAN®), vincristine (ONCOVIN®), vindesine (ELDISINE®, FILDESIN®), and vinorelbine (NAVELBINE®); etoposide (VP-16);
  • retinoids such as retinoic acid, including bexarotene (TARGRETIN®); bisphosphonates such as clodronate (for example, BONEFOS® or OSTAC®), etidronate (DIDROCAL®), NE-58095, zoledronic acid/zoledronate (ZOMETA®), alendronate (FOSAMAX®), pamidronate (AREDIA®), tiludronate (SKELID®), or risedronate (ACTONEL®); troxacitabine (a 1,3- dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those that inhibit expression of genes in signaling pathways implicated in
  • Chemotherapeutic agents as defined herein include“anti-hormonal agents” or “endocrine therapeutics” which act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer They may be hormones themselves including, but not limited to: anti-estrogens with mixed agonist/antagonist profile, including, tamoxifen (NOLVADEX®), 4-hydroxytamoxifen, toremifene (FARESTON®), idoxifene, droloxifene, raloxifene (EVISTA®), trioxifene, keoxifene, and selective estrogen receptor modulators (SERMs) such as SERM3; pure anti-estrogens without agonist properties, such as fulvestrant (FASLODEX®), and EM800 (such agents may block estrogen receptor (ER) dimerization, inhibit DNA binding, increase ER turnover, and/or suppress ER levels);
  • anti-estrogens with mixed agonist/antagonist profile including, but not limited to: anti
  • aromatase inhibitors including steroidal aromatase inhibitors such as formestane and exemestane (AROMASIN®), and nonsteroidal aromatase inhibitors such as anastrazole (ARIMIDEX®), letrozole (FEMARA®) and aminoglutethimide, and other aromatase inhibitors include vorozole (RIVISOR®), megestrol acetate (MEGASE®), fadrozole, and 4(5)-imidazoles; lutenizing hormone-releaseing hormone agonists, including leuprolide (LUPRON® and ELIGARD®), goserelin, buserelin, and tripterelin; sex steroids, including progestines such as megestrol acetate and medroxyprogesterone acetate, estrogens such as diethylstilbestrol and premarin, and androgens/retinoids such as fluoxymesterone, all transretionic acid and fenretinide; on
  • immunosuppressive agent refers to substances that act to suppress or mask the immune system of the mammal being treated herein. This would include substances that suppress cytokine production, down-regulate or suppress self-antigen expression, or mask the MHC antigens. Examples of such agents include 2-amino-6-aryl-5-substituted pyrimidines (see U.S. Pat.
  • non-steroidal anti-inflammatory drugs NSAIDs
  • ganciclovir tacrolimus, glucocorticoids such as cortisol or aldosterone
  • anti-inflammatory agents such as a cyclooxygenase inhibitor, a 5- lipoxygenase inhibitor, or a leukotriene receptor antagonist
  • purine antagonists such as azathioprine or mycophenolate mofetil (MMF)
  • alkylating agents such as cyclophosphamide; bromocryptine; danazol; dapsone; glutaraldehyde (which masks the MHC antigens, as described in U.S. Pat.
  • anti-idiotypic antibodies for MHC antigens and MHC fragments include cyclosporin A; steroids such as corticosteroids or glucocorticosteroids or glucocorticoid analogs, e.g., prednisone, methylprednisolone, including SOLU-MEDROL ® methylprednisolone sodium succinate, and dexamethasone; dihydrofolate reductase inhibitors such as methotrexate (oral or subcutaneous); anti-malarial agents such as chloroquine and hydroxychloroquine; sulfasalazine; leflunomide; cytokine or cytokine receptor antibodies including anti-interferon-alpha, -beta, or -gamma antibodies, anti-tumor necrosis factor(TNF)-alpha antibodies (infliximab (REMICADE®) or adalimumab), anti-TNF-al
  • steroids such as cortic
  • streptokinase transforming growth factor-beta (TGF-beta); streptodornase; RNA or DNA from the host; FK506; RS-61443; , chlorambucil; deoxyspergualin; rapamycin; T-cell receptor (Cohen et al., U.S. Pat.
  • T-cell receptor fragments Offner et al., Science, 251: 430-432 (1991); WO 90/11294; Ianeway, Nature, 341: 482 (1989); and WO 91/01133
  • BAFF antagonists such as BAFF antibodies and BR3 antibodies and zTNF4 antagonists (for review, see Mackay and Mackay, Trends Immunol., 23:113-5 (2002) and see also definition below)
  • biologic agents that interfere with T cell helper signals such as anti- CD40 receptor or anti-CD40 ligand (CD154), including blocking antibodies to CD40-CD40 ligand (e.g., Durie et al., Science, 261: 1328-30 (1993); Mohan et al., J.
  • T10B9 T10B9
  • Some preferred immunosuppressive agents herein include cyclophosphamide, chlorambucil, azathioprine, leflunomide, MMF, or methotrexate.
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • antibodies of the subject matter described herein are used to delay development of a disease or to slow the progression of a disease.
  • a drug that is administered“concurrently” with one or more other drugs is
  • the concurrently administered drugs are each administered on day-1 of a 3-week cycle.
  • an “effective amount” of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • an effective amount of the drug for treating cancer may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • the effective amount may extend progression free survival (e.g.
  • the term“therapeutically effective amount” means any amount which, as compared to a corresponding subject who has not received such amount, results in treatment of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • therapeutically effective amounts of a PAC, as well as salts thereof may be administered as the raw chemical.
  • the active ingredient may be presented as a pharmaceutical composition.
  • the term“optionally” means that the subsequently described event(s) may or may not occur, and includes both event(s) that occur and event(s) that do not occur.
  • the phrase“optionally substituted”, “substituted” or variations thereof denote an optional substitution, including multiple degrees of substitution, with one or more substituent group, for example, one, two or three.
  • the phrase should not be interpreted as duplicative of the substitutions herein described and depicted.
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • A“pharmaceutically acceptable excipient” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • pharmaceutically acceptable excipient includes, but is not limited to, a buffer, carrier, stabilizer, or preservative.
  • 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,
  • 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.
  • salts which are not pharmaceutically acceptable, may be useful in the preparation of compounds of described herein and these should be considered to form a further aspect of the subject matter.
  • These salts such as oxalic or trifluoroacetate, while not in themselves pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining the compounds described herein and their pharmaceutically acceptable salts.
  • conjugate refers to two or more conjugates. Each conjugate can be the same or different from any other conjugate in the plurality.
  • A“small molecule” or“small molecular compound” generally refers to an organic molecule that is less than about 5 kilodaltons (Kd) in size. In some embodiments, the small molecule is less than about 4 Kd, 3 Kd, about 2 Kd, or about 1 Kd. In some embodiments, the small molecule is less than about 800 daltons (D), about 600 D, about 500 D, about 400 D, about 300 D, about 200 D, or about 100 D. In some embodiments, a small molecule is less than about 2000 g/mol, less than about 1500 g/mol, less than about 1000 g/mol, less than about 800 g/mol, or less than about 500 g/mol. In some embodiments, small molecules are non- polymeric.
  • Small molecules are not proteins, polypeptides, oligopeptides, peptides, polynucleotides, oligonucleotides, polysaccharides, glycoproteins, proteoglycans, etc.
  • a derivative of a small molecule refers to a molecule that shares the same structural core as the original small molecule, but which can be prepared by a series of chemical reactions from the original small molecule.
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical of any length from one to twelve carbon atoms (C 1 ⁇ C 12 ), wherein the alkyl radical may be optionally substituted independently with one or more substituents described below.
  • an alkyl radical is one to eight carbon atoms (C 1 ⁇ C 8 ), or one to six carbon atoms (C 1 ⁇ C 6 ).
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1-propyl (n-Pr, n-propyl, - CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, -CH(CH 3 ) 2 ), 1-butyl (n-Bu, n-butyl, - CH 2 CH 2 CH 2 CH 3 ), 2-methyl-1-propyl (i-Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, - CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH 3 ) 3 ), 1-pentyl (n-pentyl, - CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH(CH)
  • alkylene refers to a saturated linear or branched-chain divalent hydrocarbon radical of any length from one to twelve carbon atoms (C 1 ⁇ C 12 ), wherein the alkylene radical may be optionally substituted independently with one or more substituents described below.
  • an alkylene radical is one to eight carbon atoms (C 1 ⁇ C 8 ), or one to six carbon atoms (C 1 ⁇ C 6 ).
  • alkylene groups include, but are not limited to, methylene (-CH 2 -), ethylene ( ⁇ CH 2 CH 2 ⁇ ), propylene ( ⁇ CH 2 CH 2 CH 2 ⁇ ), and the like.
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical of any length from two to eight carbon atoms (C 2 ⁇ C 8 ) with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, ethynyl (-C ⁇ CH), propynyl (propargyl, -CH 2 C ⁇ CH), and the like.
  • alkynylene refers to a linear or branched divalent hydrocarbon radical of any length from two to eight carbon atoms (C 2 ⁇ C 8 ) with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynylene radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, ethynylene (-C ⁇ C-), propynylene (propargylene, -CH 2 C ⁇ C-), and the like.
  • “carbocycle”,“carbocyclyl”,“carbocyclic ring” and“cycloalkyl” refer to a monovalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms (C 3 ⁇ C 12 ) as a monocyclic ring or 7 to 12 carbon atoms as a bicyclic ring.
  • Bicyclic carbocycles having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, and bicyclic carbocycles having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane. Spiro moieties are also included within the scope of this definition.
  • Examples of monocyclic carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3- enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
  • Carbocyclyl groups are optionally substituted independently with one or more substituents described herein.
  • Aryl means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms (C 6 ⁇ C 20 ) derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Some aryl groups are represented in the exemplary structures as“Ar”. Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic ring.
  • Typical aryl groups include, but are not limited to, radicals derived from benzene (phenyl), substituted benzenes, naphthalene, anthracene, biphenyl, indenyl, indanyl, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthyl, and the like.
  • Aryl groups are optionally substituted independently with one or more substituents described herein.
  • Arylene means a divalent aromatic hydrocarbon radical of 6-20 carbon atoms (C 6 ⁇ C 20 ) derived by the removal of two hydrogen atom from a two carbon atoms of a parent aromatic ring system. Some arylene groups are represented in the exemplary structures as “Ar”. Arylene includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic ring.
  • Typical arylene groups include, but are not limited to, radicals derived from benzene (phenylene), substituted benzenes, naphthalene, anthracene, biphenylene, indenylene, indanylene, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthyl, and the like.
  • Arylene groups are optionally substituted with one or more substituents described herein.
  • heterocycle “heterocyclyl” and“heterocyclic ring” are used
  • a heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 4 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 6 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system. Heterocycles are described in Paquette, Leo A.;
  • Heterocyclyl also includes radicals where heterocycle radicals are fused with a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring.
  • heterocyclic rings include, but are not limited to, morpholin-4-yl, piperidin-1-yl, piperazinyl, piperazin-4-yl-2-one, piperazin-4-yl-3-one, pyrrolidin-1-yl, thiomorpholin-4-yl, S-dioxothiomorpholin-4-yl, azocan-1-yl, azetidin-1-yl, octahydropyrido[1,2-a]pyrazin-2-yl, [1,4]diazepan-1-yl, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, a
  • heteroaryl refers to a monovalent aromatic radical of 5-, 6-, or 7- membered rings, and includes fused ring systems (at least one of which is aromatic) of 5-20 atoms, containing one or more heteroatoms independently selected from nitrogen, oxygen, and sulfur.
  • heteroaryl groups are pyridinyl (including, for example, 2- hydroxypyridinyl), imidazolyl, imidazopyridinyl, 1-methyl-1H-benzo[d]imidazole,
  • [1,2,4]triazolo[1,5-a]pyridine pyrimidinyl (including, for example, 4-hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxadiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, thiadiazolyl, furazanyl,
  • the heterocycle or heteroaryl groups may be carbon (carbon-linked), or nitrogen (nitrogen-linked) bonded where such is possible.
  • carbon bonded heterocycles or heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6,
  • nitrogen bonded heterocycles or heteroaryls are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3- pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2- pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, 1H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ - carboline.
  • stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • Enantiomers refer to two stereoisomers of a compound which are non- superimposable mirror images of one another. 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).
  • d and l 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.
  • the terms “racemic mixture” and“racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • Wild-type (“WT");
  • Cysteine engineered mutant antibody (“thio”); light chain (“LC”); heavy chain (“HC”); 6- maleimidocaproyl (“MC”); maleimidopropanoyl (“MP”); valine-citrulline (“val-cit” or“vc”), alanine-phenylalanine (“ala-phe”), p-aminobenzyl (“PAB”), and p-aminobenzyloxycarbonyl (“PABC”);
  • the PROTAC-Antibody Conjugate (PAC) molecules described herein comprise an antibody conjugated via a linker (L1) to a PROTAC, wherein the PROTAC comprises a ubiquitin E3 ligase binding groug (“E3LB”), a linker (“L2”) and a protein binding group (“PB”).
  • E3LB ubiquitin E3 ligase binding groug
  • L2 linker
  • PB protein binding group
  • D is PROTAC having the structure E3LB ⁇ L2 ⁇ PB; wherein, E3LB is an E3 ligase binding group covalently bound to L2; L2 is a linker covalently bound to E3LB and PB; PB is a protein binding group covalently bound to L2; Ab is an antibody covalently bound to L1; L1 is a linker, covalently bound to Ab and to D; and p has a value from about 1 to about 50.
  • the variable p reflects that an antibody can be connected to one or more L1-D groups. In one embodiment, p is from about 1 to 8. In another embodiment, p is about 2.
  • antibodies e.g., a monoclonal antibodies (mABs) are used to deliver a PROTAC to target cells, e.g., cells that express the specific protein that is targeted by the antibody.
  • the antibody portion of a PAC can target a cell that expresses an antigen whereby the antigen specific PAC is delivered intracellularly to the target cell, typically through endocytosis While PACs that comprise an antibody directed to an antigen that is not found on the cell surface may result in less specific intracellular delivery of the PROTAC portion into the cell, the PAC may still undergo pinocytosis.
  • the PACs and method of their use described herein advantageously utilize antibody recognition of the cellular surface and/or endocytosis of the PAC to deliver the PROTAC portion inside cells.
  • an antibody provided herein is a human antibody.
  • Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol.5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol.20:450-459 (2008).
  • Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol., 133: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp.51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al., J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al., Proc. Natl. Acad. Sci. USA, 103:3557-3562 (2006).
  • Additional methods include those described, for example, in U.S. Patent No.7,189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas).
  • Human hybridoma technology Trioma technology
  • Vollmers and Brandlein, Histology and Histopathology, 20(3):927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3):185-91 (2005).
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • Antibodies for use in a PAC may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in
  • Phage typically display antibody fragments, either as single- chain Fv (scFv) fragments or as Fab fragments.
  • Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas.
  • the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al., EMBO J, 12: 725-734 (1993).
  • naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol., 227: 381-388 (1992).
  • Patent publications describing human antibody phage libraries include, for example: US Patent No.5,750,373, and US Patent Publication Nos.2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598,
  • Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • an antibody provided herein is a chimeric antibody.
  • Certain chimeric antibodies are described, e.g., in U.S. Patent No.4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984)).
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a“class switched” antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • HVRs e.g., CDRs, (or portions thereof) are derived from a non-human antibody
  • FRs or portions thereof
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the HVR residues are derived
  • Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the“best-fit” method (see, e.g., Sims et al. J. Immunol.151:2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol., 151:2623 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front.
  • framework regions selected using the“best-fit” method see, e.g., Sims et al. J. Immunol.151:2296 (1993)
  • framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions see, e.g.,
  • an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • the term“multispecific antibody” as used herein refers to an antibody comprising an antigen-binding domain that has polyepitopic specificity (i.e., is capable of binding to two, or more, different epitopes on one molecule or is capable of binding to epitopes on two, or more, different molecules).
  • multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different antigen binding sites (such as a bispecific antibody).
  • the first antigen-binding domain and the second antigen- binding domain of the multispecific antibody may bind the two epitopes within one and the same molecule (intramolecular binding).
  • the first antigen-binding domain and the second antigen-binding domain of the multispecific antibody may bind to two different epitopes on the same protein molecule.
  • the two different epitopes that a multispecific antibody binds are epitopes that are not normally bound at the same time by one monospecific antibody, such as e.g. a conventional antibody or one immunoglobulin single variable domain.
  • the first antigen-binding domain and the second antigen-binding domain of the multispecific antibody may bind epitopes located within two distinct molecules (intermolecular binding).
  • the first antigen- binding domain of the multispecific antibody may bind to one epitope on one protein molecule
  • the second antigen-binding domain of the multispecific antibody may bind to another epitope on a different protein molecule, thereby cross-linking the two molecules.
  • the antigen-binding domain of a multispecific antibody comprises two VH/VL units, wherein a first VH/VL unit binds to a first epitope and a second VH/VL unit binds to a second epitope, wherein each VH/VL unit comprises a heavy chain variable domain (VH) and a light chain variable domain (VL).
  • Such multispecific antibodies include, but are not limited to, full length antibodies, antibodies having two or more VL and VH domains, and antibody fragments (such as Fab, Fv, dsFv, scFv, diabodies, bispecific diabodies and triabodies, antibody fragments that have been linked covalently or non-covalently).
  • a VH/VL unit that further comprises at least a portion of a heavy chain variable region and/or at least a portion of a light chain variable region may also be referred to as an“arm” or“hemimer” or“half antibody.”
  • a hemimer comprises a sufficient portion of a heavy chain variable region to allow
  • a hemimer comprises a knob mutation or a hole mutation, for example, to allow
  • heterodimerization with a second hemimer or half antibody that comprises a complementary hole mutation or knob mutation.
  • Knob mutations and hole mutations are discussed further below.
  • a multispecific antibody provided herein may be a bispecific antibody.
  • the term“bispecific antibody” as used herein refers to a multispecific antibody comprising an antigen-binding domain that is capable of binding to two different epitopes on one molecule or is capable of binding to epitopes on two different molecules.
  • a bispecific antibody may also be referred to herein as having“dual specificity” or as being“dual specific.”
  • Exemplary bispecific antibodies may bind both protein and any other antigen.
  • one of the binding specificities is for protein and the other is for CD3. See, e.g., U.S. Patent No.5,821,337.
  • bispecific antibodies may bind to two different epitopes of the same protein molecule.
  • bispecific antibodies may bind to two different epitopes on two different protein molecules. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express protein.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983), WO 93/08829, and Traunecker et al., EMBO J.10: 3655 (1991)), and“knob-in-hole” engineering (see, e.g., U.S. Patent No.5,731,168, WO2009/089004, US2009/0182127, US2011/0287009, Marvin and Zhu, Acta Pharmacol. Sin. (2005) 26(6):649-658, and Kontermann (2005) Acta Pharmacol. Sin., 26:1-9).
  • knock-into-hole or“KnH” technology as used herein refers to the technology directing the pairing of two polypeptides together in vitro or in vivo by introducing a protuberance (knob) into one polypeptide and a cavity (hole) into the other polypeptide at an interface in which they interact.
  • KnHs have been introduced in the Fc:Fc binding interfaces, CL:CH1 interfaces or VH/VL interfaces of antibodies (see, e.g., US 2011/0287009, US2007/0178552, WO 96/027011, WO 98/050431, Zhu et al., 1997, Protein Science 6:781-788, and WO2012/106587).
  • KnHs drive the pairing of two different heavy chains together during the manufacture of multispecific antibodies.
  • multispecific antibodies having KnH in their Fc regions can further comprise single variable domains linked to each Fc region, or further comprise different heavy chain variable domains that pair with similar or different light chain variable domains.
  • KnH technology can be also be used to pair two different receptor extracellular domains together or any other polypeptide sequences that comprises different target recognition sequences (e.g., including affibodies, peptibodies and other Fc fusions).
  • knock mutation refers to a mutation that introduces a protuberance (knob) into a polypeptide at an interface in which the polypeptide interacts with another polypeptide.
  • the other polypeptide has a hole mutation.
  • hole mutation refers to a mutation that introduces a cavity (hole) into a polypeptide at an interface in which the polypeptide interacts with another polypeptide.
  • the other polypeptide has a knob mutation.
  • A“protuberance” refers to at least one amino acid side chain which projects from the interface of a first polypeptide and is therefore positionable in a compensatory cavity in the adjacent interface (i.e. the interface of a second polypeptide) so as to stabilize the
  • the protuberance may exist in the original interface or may be introduced synthetically (e.g., by altering nucleic acid encoding the interface).
  • nucleic acid encoding the interface of the first polypeptide is altered to encode the
  • nucleic acid encoding at least one“original” amino acid residue in the interface of the first polypeptide is replaced with nucleic acid encoding at least one“import” amino acid residue which has a larger side chain volume than the original amino acid residue. It will be appreciated that there can be more than one original and corresponding import residue.
  • the side chain volumes of the various amino residues are shown, for example, in Table 1 of US2011/0287009. A mutation to introduce a
  • protuberance may be referred to as a“knob mutation.”
  • import residues for the formation of a protuberance are naturally occurring amino acid residues selected from arginine (R), phenylalanine (F), tyrosine (Y) and tryptophan (W).
  • an import residue is tryptophan or tyrosine.
  • the original residue for the formation of the protuberance has a small side chain volume, such as alanine, asparagine, aspartic acid, glycine, serine, threonine or valine.
  • A“cavity” refers to at least one amino acid side chain which is recessed from the interface of a second polypeptide and therefore accommodates a corresponding protuberance on the adjacent interface of a first polypeptide.
  • the cavity may exist in the original interface or may be introduced synthetically (e.g. by altering nucleic acid encoding the interface).
  • nucleic acid encoding the interface of the second polypeptide is altered to encode the cavity.
  • the nucleic acid encoding at least one“original” amino acid residue in the interface of the second polypeptide is replaced with DNA encoding at least one“import” amino acid residue which has a smaller side chain volume than the original amino acid residue. It will be appreciated that there can be more than one original and corresponding import residue.
  • import residues for the formation of a cavity are naturally occurring amino acid residues selected from alanine (A), serine (S), threonine (T) and valine (V).
  • an import residue is serine, alanine or threonine.
  • the original residue for the formation of the cavity has a large side chain volume, such as tyrosine, arginine, phenylalanine or tryptophan.
  • a mutation to introduce a“cavity” may be referred to as a“hole mutation.”
  • the protuberance is“positionable” in the cavity which means that the spatial location of the protuberance and cavity on the interface of a first polypeptide and second polypeptide respectively and the sizes of the protuberance and cavity are such that the protuberance can be located in the cavity without significantly perturbing the normal association of the first and second polypeptides at the interface. Since protuberances such as Tyr, Phe and Trp do not typically extend perpendicularly from the axis of the interface and have preferred
  • the alignment of a protuberance with a corresponding cavity may, in some instances, rely on modeling the protuberance/cavity pair based upon a three-dimensional structure such as that obtained by X-ray crystallography or nuclear magnetic resonance (NMR). This can be achieved using widely accepted techniques in the art.
  • a knob mutation in an IgG1 constant region is T366W (EU numbering).
  • a hole mutation in an IgG1 constant region comprises one or more mutations selected from T366S, L368A and Y407V (EU numbering).
  • a hole mutation in an IgG1 constant region comprises T366S, L368A and Y407V (EU numbering).
  • a knob mutation in an IgG4 constant region is T366W (EU numbering).
  • a hole mutation in an IgG4 constant region comprises one or more mutations selected from T366S, L368A, and Y407V (EU numbering).
  • a hole mutation in an IgG4 constant region comprises T366S, L368A, and Y407V (EU numbering).
  • Multispecific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No.4,676,980, and Brennan et al., Science, 229: 81 (1985)); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny et al., J. Immunol., 148(5):1547-1553 (1992)); using "diabody” technology for making bispecific antibody fragments (see, e.g., Hollinger et al., Proc. Natl. Acad. Sci.
  • Engineered antibodies with three or more functional antigen binding sites including “Octopus antibodies” or“dual-variable domain immunoglobulins” (DVDs) are also included herein (see, e.g., US 2006/0025576A1, and Wu et al. Nature Biotechnology (2007)).
  • the antibody or fragment herein also includes a“Dual Acting FAb” or“DAF” comprising an antigen binding site that binds to a target protein as well as another, different antigen (see, US 2008/0069820, for example).
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab’, Fab’-SH, F(ab’) 2 , Fv, and scFv fragments, and other fragments described below.
  • Fab fragment antigen
  • Fab fragment antigen binding protein
  • Fab fragment antigen binding protein
  • Fab fragment antigen binding protein
  • Fab fragment antigen binding protein
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al., Nat. Med.
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No.6,248,516 B1).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • recombinant host cells e.g. E. coli or phage
  • amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
  • antibody variants having one or more amino acid having one or more amino acid
  • Sites of interest for substitutional mutagenesis include the HVRs and FRs.
  • Conservative substitutions are shown in Table 1 under the heading of“preferred substitutions.” More substantial changes are provided in Table 1 under the heading of “exemplary substitutions,” and as further described below in reference to amino acid side chain classes.
  • Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Amino acids may be grouped according to common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody.
  • An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
  • Alterations may be made in HVRs, e.g., to improve antibody affinity.
  • Such alterations may be made in HVR“hotspots,” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol.207:179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity.
  • HVR“hotspots i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol.207:179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity.
  • Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in
  • affinity maturation diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis).
  • a secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity.
  • Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
  • substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen.
  • conservative alterations e.g., conservative substitutions as provided herein
  • Such alterations may be outside of HVR“hotspots” or SDRs.
  • each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
  • a useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called“alanine scanning mutagenesis” as described by
  • a residue or group of target residues e.g., charged residues such as arg, asp, his, lys, and glu
  • a neutral or negatively charged amino acid e.g., alanine or polyalanine
  • substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions.
  • a crystal structure of an antigen-antibody complex is used to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution.
  • Variants may be screened to determine whether they contain the desired properties.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for antibody directed enzyme prodrug therapy (ADEPT)) or a polypeptide which increases the serum half-life of the antibody.
  • ADEPT antibody directed enzyme prodrug therapy
  • cysteine engineered antibodies e.g.,“THIOMABTM antibody”
  • one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as L1-PROTAC groups, to create a PAC, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; A140 (EU numbering) of the heavy chain; L174 (EU numbering) of the heavy chain; Y373 (EU numbering) of the heavy chain; K149 (Kabat numbering) of the light chain; A118 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
  • the antibodies described herein comprise the HC-A140C (EU numbering) cysteine substitution.
  • the antibodies described herein comprise the LC-K149C (Kabat numbering) cysteine substitution.
  • the antibodies described herein comprise the HC-A118C (EU numbering) cysteine substitution.
  • Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No.7,521,541.
  • the antibody comprises one of the following heavy chain cysteine substitutions:
  • the antibody comprises one of the following light chain cysteine substitutions:
  • a nonlimiting exemplary hu7C2.v2.2.LA light chain (LC) K149C THIOMABTM antibody has the heavy chain and light chain amino acid sequences of SEQ ID NOs: 26 and 30, respectively.
  • a nonlimiting exemplary hu7C2.v2.2.LA heavy chain (HC) A118C THIOMABTM antibody has the heavy chain and light chain amino acid sequences of SEQ ID NOs: 31 and 25, respectively.
  • PACs 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 TM antibody. 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 TM antibody 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 for use in a PAC are in the ranges of 0.6 to 1.0; 0.7 to 1.0; or 0.8 to 1.0.
  • 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.
  • Cysteine amino acids may be engineered at reactive sites in an antibody and which do not form intrachain or intermolecular disulfide linkages (Junutula, et al., 2008b Nature Biotech., 26(8):925-932; Dornan et al (2009) Blood 114(13):2721-2729; US 7521541; US 7723485; WO2009/052249, Shen et al (2012) Nature Biotech., 30(2):184-191; Junutula et al (2008) Jour of Immun. Methods 332:41-52).
  • the engineered cysteine thiols may react with linker reagents or the Linker L1-PROTAC intermediates described herein, which have thiol- reactive, electrophilic groups such as maleimides, activated disulfides (such as a 4- nitropyridyl disulfide), or alpha-halo amides to form a PAC with cysteine engineered antibodies (THIOMAB TM antibodies) and the PROTAC residue.
  • the location of the PROTAC moiety can thus be designed, controlled, and known.
  • PROTAC/antibody ratio (“PAR”) can be controlled since the engineered cysteine thiol groups typically react with thiol-reactive linker reagents or linker L1-PROTAC intermediates in high yield.
  • Engineering an antibody to introduce a cysteine amino acid by substitution at a single site on the heavy or light chain gives two new cysteines on the symmetrical antibody.
  • a PAR of about 2 can be achieved and near homogeneity of the conjugation product.
  • cysteine engineered antibodies 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, signaling 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, signaling proteins, cell survival regulatory factors, cell proliferation regulatory factors, molecules associated with (for e.g., known or suspected to contribute
  • 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).
  • Cysteine engineered antibodies are prepared for conjugation with linker L1 intermediates by reduction and reoxidation of intrachain disulfide groups.
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997).
  • oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the“stem” of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody may be made in order to create antibody variants with certain improved properties.
  • antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region.
  • the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5% to 65% or from 20% to 40%.
  • the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd).
  • Examples of publications related to“defucosylated” or“fucose- deficient” antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140; US
  • Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by
  • Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean- Mairet et al.); US Patent No.6,602,684 (Umana et al.); and US 2005/0123546 (Umana et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function.
  • one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant.
  • the Fc region variant may comprise a human Fc region sequence (e.g., a human IgG1, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.
  • the subject matter described herein is directed to an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half-life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks Fc ⁇ R binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • NK cells express Fc(RIII only, whereas monocytes express Fc(RI, Fc(RII and Fc(RIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol.
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry
  • 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. Proc. Nat’l Acad. Sci. USA 95:652-656 (1998).
  • C1q binding assays may also be carried out to confirm that the antibody is unable to bind C1q and hence lacks CDC activity.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996); Cragg, M.S. et al., Blood 101:1045-1052 (2003); and Cragg, M.S. and M.J. Glennie, Blood 103:2738-2743 (2004)).
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al., Int’l. Immunol.18(12):1759- 1769 (2006)).
  • one or more amino acid modifications may be introduced into the Fc portion of the antibody provided herein in order to increase IgG binding to the neonatal Fc receptor.
  • the antibody comprises the following three mutations according to EU numbering: M252Y, S254T, and T256E (the“YTE mutation”) (US Patent No.8,697,650; see also Dall’Acqua et al., Journal of Biological Chemistry 281(33):23514-23524 (2006).
  • the YTE mutation does not affect the ability of the antibody to bind to its cognate antigen.
  • the YTE mutation increases the antibody’s serum half-life compared to the native (i.e., non-YTE mutant) antibody.
  • the YTE mutation increases the serum half-life of the antibody by 3-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 2-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 4-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by at least 5-fold compared to the native (i.e., non-YTE mutant) antibody.
  • the YTE mutation increases the serum half-life of the antibody by at least 10-fold compared to the native (i.e., non-YTE mutant) antibody. See, e.g., US Patent No.8,697,650; see also Dall’Acqua et al., Journal of Biological Chemistry 281(33):23514-23524 (2006).
  • the YTE mutant provides a means to modulate antibody-dependent cell-mediated cytotoxicity (ADCC) activity of the antibody.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the YTEO mutant provides a means to modulate ADCC activity of a humanized IgG antibody directed against a human antigen. See, e.g., US Patent No.
  • the YTE mutant allows the simultaneous modulation of serum half-life, tissue distribution, and antibody activity (e.g., the ADCC activity of an IgG antibody). See, e.g., US Patent No.8,697,650; see also Dall’Acqua et al., Journal of
  • Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 according to EU numbering (U S Patent No 6737056)
  • Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327 according to EU numbering, including the so-called“DANA” Fc mutant with substitution of residues 265 and 297 to alanine according to EU numbering (i.e., D265A and N297A according to EU numbering) (US Patent No.7,332,581).
  • the Fc mutant comprises the following two amino acid substitutions: D265A and N297A.
  • the Fc mutant consists of the following two amino acid substitutions: D265A and N297A.
  • the proline at position329 (EU numbering) (P329) of a wild- type human Fc region is substituted with glycine or arginine or an amino acid residue large enough to destroy the proline sandwich within the Fc/Fc ⁇ receptor interface, that is formed between the P329 of the Fc and tryptophane residues W87 and W110 of FcgRIII
  • At least one further amino acid substitution in the Fc variant is S228P, E233P, L234A, L235A, L235E, N297A, N297D, or P331S and still in another embodiment said at least one further amino acid substitution is L234A and L235A of the human IgG1 Fc region or S228P and L235E of the human IgG4 Fc region, all according to EU numbering (U.S. Patent No.
  • a polypeptide comprises the Fc variant of a wild-type human IgG Fc region wherein the polypeptide has P329 of the human IgG Fc region substituted with glycine and wherein the Fc variant comprises at least two further amino acid substitutions at L234A and L235A of the human IgG1 Fc region or S228P and L235E of the human IgG4 Fc region, and wherein the residues are numbered according to the EU numbering (U.S. Patent No.8,969,526).
  • the polypeptide comprising the P329G, L234A and L235A (EU numbering) substitutions exhibit a reduced affinity to the human Fc ⁇ RIIIA and Fc ⁇ RIIA, for down-modulation of ADCC to at least 20% of the ADCC induced by the polypeptide comprising the wild-type human IgG Fc region, and/or for down-modulation of ADCP (U.S. Patent No.8,969,526).
  • the polypeptide comprising an Fc variant of a wild-type human Fc polypeptide comprises a triple mutation: an amino acid substitution at position Pro329, a L234A and a L235A mutation according to EU numbering (P329 / LALA) (U.S. Patent No.8,969,526).
  • the polypeptide comprises the following amino acid substitutions: P329G, L234A, and L235A according to EU numbering.
  • Certain antibody variants with improved or diminished binding to FcRs are described. (See, e.g., U.S. Patent No.6,737,056; WO 2004/056312, and Shields et al., J. Biol. Chem. 9(2): 6591-6604 (2001).)
  • an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering).
  • alterations are made in the Fc region that result in altered (i.e., either improved or diminished) C1q binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No.6,194,551, WO 99/51642, and Idusogie et al. J. Immunol.164: 4178-4184 (2000).
  • CDC Complement Dependent Cytotoxicity
  • Antibodies with increased half lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus are described in US2005/0014934A1 (Hinton et al.). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn.
  • Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No.7,371,826) according to EU numbering. See also Duncan & Winter, Nature 322:738-40 (1988); U.S. Patent No.
  • an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3- dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propylene glycol homopolymers, polypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol),
  • PEG poly
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer is attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
  • Antibodies including but not limited to cysteine engineered antibodies, which may be useful in the PACs described herein in the treatment of cancer include, but are not limited to, antibodies against cell surface receptors and tumor-associated antigens (TAA).
  • TAA tumor-associated antigens
  • Certain tumor-associated antigens are known in the art, and can be prepared for use in generating antibodies using methods and information which are well known in the art.
  • TAA tumor-associated antigens
  • tumor-associated polypeptides are more abundantly expressed on the surface of the cancer cells as compared to on the surface of the non-cancerous cells.
  • the identification of such tumor-associated cell surface antigen polypeptides has given rise to the ability to more specifically target cancer cells for destruction via antibody-based therapies.
  • TAA tumor-associated antigens
  • examples of tumor-associated antigens TAA include, but are not limited to, those 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 listed below 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, and/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.
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No.4,816,567.
  • isolated nucleic acid encoding an antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NS0, Sp20 cell).
  • a method of making an antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be 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 the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2003), pp.245-254, describing expression of antibody fragments in E. coli.
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been“humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat.
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos.5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES TM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol.36:59 (1977); baby hamster kidney cells (BHK); mouse sertoli cells (TM4 cells as described, e.g., in Mather, Biol.
  • monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al., Annals N.Y. Acad. Sci.383:44-68 (1982); MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR- CHO cells (Urlaub et al., Proc. Natl. Acad. Sci.
  • the antibody binds to one or more tumor-associated antigens or cell-surface receptors selected from (1)-(53):
  • 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 (2) E16 (LAT1, SLC7A5, Genbank accession no. NM_003486)
  • 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+
  • 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);
  • AAK74120.3; AF361486_1 MPF (MPF, MSLN, SMR, megakaryocyte potentiating factor, mesothelin,
  • Napi2b Napi3b, NAPI-3B, NPTIIb, SLC34A2, solute carrier family 34
  • 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.
  • WO2003003984 (Claim 1); WO200206339 (Claim 1; Page 50); WO200188133 (Claim 1; Page 41-43, 48-58); WO2003054152 (Claim 20); WO2003101400 (Claim 11);
  • 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);
  • 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);
  • WO200216413 (Claim 1; Page 94-95, 105); WO200222808 (Claim 2; Fig 1); US5854399 (Example 2; Col 17-18); US5792616 (Fig 2);
  • MIM 187395; NP_003203.1; NM_003212_1 (14) 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);
  • WO2003055439 (Claim 29; Fig 1A-B); WO2003025228 (Claim 37; Fig 5C);
  • WO200222636 (Example 13; Page 95-107); WO200212341 (Claim 68; Fig 7);
  • NCA NCA (CEACAM6, Genbank accession no. M18728);
  • WO2004031238 WO2003042661 (Claim 12); WO200278524 (Example 2); WO200286443 (Claim 27; Page 427); WO200260317 (Claim 2);
  • WO200222153 (Page 45-47); US2002042366 (Page 20-21); WO200146261 (Page
  • 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.
  • 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. AJ29
  • WO2003003906 (Claim 10; Page 288); WO200140309 (Example 1; Fig 17);
  • 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);
  • 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);
  • MIM 606269; NP_443177.1; NM_052945_1; AF132600 (27) CD22 (B-cell receptor CD22-B isoform, BL-CAM, Lyb-8, Lyb8, SIGLEC-2,
  • CD79a (CD79A, CD79 ⁇ , 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: 19q13.2, 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: 11q23.3, Genbank accession No. NP_001707.1)
  • HLA-DOB Beta subunit of MHC class II molecule (Ia 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)
  • 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: 9p13.3, 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: 5q12, Genbank accession No.
  • FcRH1 Fc receptor-like protein 1, a putative receptor for the immunoglobulin Fc domain that contains C2 type Ig-like and ITAM domains, may have a role in B-lymphocyte differentiation
  • WO2003089624 (claim 7); (35) FCRH5 (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: 1q21, Genbank accession No.
  • TENB2 tomoregulin, TPEF, HPP1, 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
  • TMEFF1 transmembrane protein with EGF-like and two follistatin-like domains 1; Tomoregulin-1
  • H7365 C9orf2; C9ORF2; U19878; X83961; NM_080655; NM_003692; Harms, P.W. (2003) Genes Dev.17 (21), 2624-2629; Gery, S.
  • GDNF-Ra1 GDNF family receptor alpha 1; GFRA1; GDNFR; GDNFRA; RETL1; TRNR1; RET1L; GDNFR-alpha1; GFR-ALPHA-1
  • CD33 a member of the sialic acid binding, immunoglobulin-like lectin family, is a 67- kDa glycosylated transmembrane protein. CD33is expressed on most myeloid and monocytic leukemia cells in addition to committed myelomonocytic and erythroid progenitor cells. It is not seen on the earliest pluripotent stem cells, mature granulocytes, lymphoid cells, or nonhematopoietic cells (Sabbath et al., (1985) J. Clin. Invest.75:756-56; Andrews et al., (1986) Blood 68:1030-5).
  • CD33 contains two tyrosine residues on its cytoplasmic tail, each of which is followed by hydrophobic residues similar to the immunoreceptor tyrosine-based inhibitory motif (ITIM) seen in many inhibitory receptors.
  • ITIM immunoreceptor tyrosine-based inhibitory motif
  • CLL-1 CLEC12A, MICL, and DCAL2
  • CTL/CTLD C-type lectin/C-type lectin-like domain
  • CLL-1 has been shown to be a type II transmembrane receptor comprising a single C-type lectin-like domain (which is not predicted to bind either calcium or sugar), a stalk region, a
  • transmembrane domain and a short cytoplasmic tail containing an ITIM motif.
  • the antibody of the PAC may be an antibody that is directed to a protein that is found on numerous cells or tissue types.
  • examples of such antibodies include gD and EpCAM.
  • a PAC can be used to deliver a PROTAC to many cells or tissues rather thanspecific cell types or tissue types as when using a using a targeted antibody.
  • a PAC may comprise an antibody, e.g., an antibody selected from:
  • a PAC can comprise anti-Ly6E antibodies.
  • Lymphocyte antigen 6 complex locus E (Ly6E), also known as retinoic acid induced gene E (RIG-E) and stem cell antigen 2 (SCA-2). It is a GPI linked, 131 amino acid length, ⁇ 8.4kDa protein of unknown function with no known binding partners. It was initially identified as a transcript expressed in immature thymocyte, thymic medullary epithelial cells in mice (Mao, et al. (1996) Proc. Natl. Acad. Sci. U.S.A.93:5910-5914).
  • the subject matter described herein provides a PAC comprising an anti-Ly6E antibody described in PCT Publication No. WO 2013/177055.
  • the subject matter described herein provides a PAC comprising an anti-Ly6E antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
  • HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12
  • HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13
  • HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14
  • HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9
  • the subject matter described herein provides a PAC comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14.
  • the subject matter described herein provides a PAC comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
  • a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 12, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 14; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
  • a PAC comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
  • an anti-Ly6E antibody of a PAC is humanized.
  • an anti-Ly6E antibody comprises HVRs as in any of the above
  • a human acceptor framework e.g. a human
  • immunoglobulin framework or a human consensus framework.
  • an anti-Ly6E antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 8.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:8 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to Ly6E.
  • the anti-Ly6E antibody comprises the VH sequence of SEQ ID NO: 8, including post- translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14.
  • an anti-Ly6E antibody of a PAC comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 7.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:7 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to Ly6E
  • substitutions e.g., conservative substitutions
  • insertions, or deletions relative to the reference sequence but an anti-Ly6E antibody comprising that sequence retains the ability to bind to Ly6E
  • a total of 1 to 10 amino acids have been substituted inserted and/or deleted in SEQ ID NO: 7.
  • a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 7.
  • the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
  • the anti-Ly6E antibody comprises the VL sequence of SEQ ID NO: 7, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
  • a PAC comprising an anti-Ly6E antibody
  • the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • a PAC comprises the VH and VL sequences in SEQ ID NO: 8 and SEQ ID NO: 7, respectively, including post-translational modifications of those sequences.
  • PACs comprising antibodies that bind to the same epitope as an anti-Ly6E antibody provided herein.
  • PACs comprising antibodies that bind to the same epitope as an anti-Ly6E antibody provided herein.
  • a PAC comprising an antibody that binds to the same epitope as an anti-Ly6E antibody comprising a VH sequence of SEQ ID NO: 8 and a VL sequence of SEQ ID NO: 7, respectively.
  • an anti-Ly6E antibody of a PAC is a monoclonal antibody, including a human antibody.
  • an anti-Ly6E antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
  • a PAC comprises an anti-Ly6E antibody comprising a heavy chain and a light chain comprising the amino acid sequences of SEQ ID NO: 16 and 15, respectively. Table 4. Ly6E AntibodySequences.
  • PACs comprise anti-HER2 antibodies.
  • an anti-HER2 antibody of a PAC comprises a humanized anti-HER2 antibody, e.g., huMAb4D5-1, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8, as described in Table 3 of US 5821337.
  • Those antibodies contain human framework regions with the complementarity-determining regions of a murine antibody (4D5) that binds to HER2.
  • the humanized antibody huMAb4D5-8 is also referred to as trastuzumab, commercially available under the tradename HERCEPTIN ⁇ .
  • an anti-HER2 antibody of a PAC comprises a humanized anti-HER2 antibody, e.g., humanized 2C4, as described in US7862817.
  • An exemplary humanized 2C4 antibody is pertuzumab, commercially available under the tradename PERJETA ⁇ .
  • an anti-HER2 antibody of a PAC comprises a humanized 7C2 anti-HER2 antibody.
  • a humanized 7C2 antibody is an anti-HER2 antibody.
  • PACs comprising an anti-HER2 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • PACs comprising an anti-HER2 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 68; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 22, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 24 or 29; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 24; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR- L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • an anti-HER2 antibody of a PAC is humanized.
  • an anti-HER2 antibody of a PAC comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
  • immunoglobulin framework or a human consensus framework.
  • an anti-HER2 antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 18.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 18 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti HER2 antibody comprising that sequence retains the ability to bind to HER2
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 18.
  • a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 18.
  • the anti- HER2 antibody comprises the VH sequence of SEQ ID NO: 18, including post- translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24.
  • an anti-HER2 antibody of a PAC comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 17.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 17 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-HER2 antibody comprising that sequence retains the ability to bind to HER2.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 17. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 17. In certain embodiments,
  • the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
  • the anti-HER2 antibody comprises the VL sequence of SEQ ID NO: 17, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
  • a PAC comprising an anti-HER2 antibody
  • the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • a PAC comprising an antibody comprising an antibody, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 18 and SEQ ID NO: 17, respectively, including post-translational modifications of those sequences.
  • a PAC comprising an antibody is provided, wherein the antibody comprises the humanized 7C2.v2.2.LA (hu7C2) K149C kappa light chain sequence of SEQ ID NO: 30
  • a PAC comprising an antibody
  • the antibody comprises the Hu7C2 A118C IgG1 heavy chain sequence of SEQ ID NO: 31
  • PACs comprising antibodies that bind to the same epitope as an anti-HER2 antibody provided herein.
  • PACs comprising antibodies that bind to the same epitope as an anti-HER2 antibody provided herein.
  • a PAC comprising an antibody that binds to the same epitope as an anti-HER2 antibody comprising a VH sequence of SEQ ID NO: 18 and a VL sequence of SEQ ID NO: 17, respectively.
  • an anti-HER2 antibody of a PAC is a monoclonal antibody, including a human antibody.
  • an anti-HER2 antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment.
  • a PAC comprises an antibody that is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
  • PACs comprise anti-MUC16 antibodies.
  • PACs comprising an anti-MUC16 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
  • a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 35, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 37; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
  • PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
  • an anti-MUC16 antibody of a PAC is humanized.
  • an anti-MUC16 antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
  • immunoglobulin framework or a human consensus framework.
  • an anti-MUC16 antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 39.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 39 contains substitutions (e g conservative substitutions) insertions or deletions relative to the reference sequence, but an anti-MUC16 antibody comprising that sequence retains the ability to bind to MUC16.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain
  • the anti-MUC16 antibody comprises the VH sequence of SEQ ID NO: 39, including post-translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37.
  • an anti-MUC16 antibody of a PAC comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 38.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:38 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-MUC16 antibody comprising that sequence retains the ability to bind to MUC16.
  • the anti-MUC16 antibody comprises the VL sequence of SEQ ID NO: 38, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
  • a PAC comprising an anti-MUC16 antibody
  • the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • a PAC comprising antibodies that bind to the same epitope as an anti-MUC16 antibody provided herein.
  • a PAC is provided comprising an antibody that binds to the same epitope as an anti-MUC16 antibody comprising a VH sequence of SEQ ID NO: 39 and a VL sequence of SEQ ID NO: 38, respectively.
  • an anti-MUC16 antibody of a PAC is a monoclonal antibody, including a human antibody.
  • an anti-MUC16 antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
  • PACs comprise anti STEAP 1 antibodies
  • PACs comprising an anti-STEAP-1 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
  • a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 40, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 42; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
  • PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
  • an anti-STEAP-1 antibody of a PAC is humanized.
  • an anti-STEAP-1 antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
  • immunoglobulin framework or a human consensus framework.
  • an anti-STEAP-1 antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 46.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 46 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-STEAP-1 antibody comprising that sequence retains the ability to bind to STEAP-1.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain
  • the anti-STEAP-1 antibody comprises the VH sequence of SEQ ID NO: 46, including post-translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42.
  • an anti-STEAP-1 antibody of an a PAC comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 47.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 47 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-STEAP-1 antibody comprising that sequence retains the ability to bind to STEAP-1.
  • the anti-STEAP-1 antibody comprises the VL sequence of SEQ ID NO: 47, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
  • a PAC comprising an anti-STEAP-1 antibody
  • the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • a PAC comprises the VH and VL sequences in SEQ ID NO: 46 and SEQ ID NO: 47, respectively, including post- translational modifications of those sequences.
  • PACs comprising antibodies that bind to the same epitope as an anti-STEAP-1 antibody provided herein.
  • a PAC comprising an antibody that binds to the same epitope as an anti-STEAP-1 antibody comprising a VH sequence of SEQ ID NO: 46 and a VL sequence of SEQ ID NO: 47, respectively.
  • an anti-STEAP-1 antibody of a PAC is a monoclonal antibody, including a human antibody.
  • an anti-STEAP-1 antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
  • a PAC comprises anti-NaPi2b antibodies.
  • PACs comprising an anti-NaPi2b antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
  • a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 48, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 50; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
  • PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
  • an anti-NaPi2b antibody of a PAC is humanized.
  • an anti-NaPi2b antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
  • immunoglobulin framework or a human consensus framework.
  • an anti-NaPi2b antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 54.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 54 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-NaPi2b antibody comprising that sequence retains the ability to bind to NaPi2b.
  • the anti-NaPi2b antibody comprises the VH sequence of SEQ ID NO: 54, including post- translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50.
  • an anti-NaPi2b antibody of a PAC comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 55
  • VL sequence having at least 90% 91% 92% 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 55 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-NaPi2b antibody comprising that sequence retains the ability to bind to anti-NaPi2b.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 55.
  • the anti-NaPi2b antibody comprises the VL sequence of SEQ ID NO: 55, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
  • a PAC comprising an anti-NaPi2b antibody
  • the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • a PAC comprises the VH and VL sequences in SEQ ID NO: 54 and SEQ ID NO: 55, respectively, including post- translational modifications of those sequences.
  • PACs comprising antibodies that bind to the same epitope as an anti-NaPi2b antibody provided herein.
  • PACs comprising antibodies that bind to the same epitope as an anti-NaPi2b antibody provided herein.
  • a PAC comprising an antibody that binds to the same epitope as an anti-NaPi2b antibody comprising a VH sequence of SEQ ID NO: 54 and a VL sequence of SEQ ID NO: 55, respectively.
  • an anti-NaPi2b antibody of a PAC is a monoclonal antibody, including a human antibody.
  • an anti-NaPi2b antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
  • PACs comprise anti-CD79b antibodies.
  • PACs comprising an anti-CD79b antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60.
  • the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60.
  • PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
  • the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
  • a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 58, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 60; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
  • PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; (c) HVR H3 comprising the amino acid sequence of SEQ ID NO: 60; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
  • an anti-CD79b antibody of a PAC is humanized.
  • an anti-CD79b antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
  • immunoglobulin framework or a human consensus framework.
  • an anti-CD79b antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 56.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 56 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-CD79b antibody comprising that sequence retains the ability to bind to CD79b.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 56. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 56. In certain embodiments,
  • the anti-CD79b antibody comprises the VH sequence of SEQ ID NO: 8, including post-translational modifications of that sequence.
  • the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60.
  • an anti-CD79b antibody of a PAC comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 57.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 57 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to CD79b.
  • the anti-CD79b antibody comprises the VL sequence of SEQ ID NO: 57, including post-translational modifications of that sequence.
  • the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
  • PACs comprising an anti-CD79b antibody
  • the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • a PAC comprises the VH and VL sequences in SEQ ID NO: 56 and SEQ ID NO: 57, respectively, including post- translational modifications of those sequences.
  • PACs comprising antibodies that bind to the same epitope as an anti-CD79b antibody provided herein.
  • antibodies that bind to the same epitope as an anti-CD79b antibody provided herein.
  • a PAC comprising an antibody that binds to the same epitope as an anti-CD79b antibody comprising a VH sequence of SEQ ID NO: 56 and a VL sequence of SEQ ID NO: 57, respectively.
  • an anti-CD79b antibody of a PAC is a monoclonal antibody, including a human antibody.
  • an anti-CD79b antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’) 2 fragment.
  • the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
  • a PAC can comprise anti-CD22 antibodies, which comprise three light chain hypervariable regions (HVR-L1, HVR-L2 and HVR-L3) and three heavy chain hypervariable regions (HVR-H1, HVR-H2 and HVR-H3).
  • the anti-CD22 antibody of a PAC comprises three light chain hypervariable regions and three heavy chain hypervariable regions (SEQ ID NO: 66-71), the sequences of which are shown below.
  • the anti-CD22 antibody of a PAC comprises the variable light chain sequence of SEQ ID NO: 72 and the variable heavy chain sequence of SEQ ID NO: 73.
  • the anti-CD22 antibody of PACs of the present invention comprises the light chain sequence of SEQ ID NO: 74 and the heavy chain sequence of SEQ ID NO: 75:
  • a PAC can comprise anti-CD33 antibodies, which comprise three light chain hypervariable regions and three heavy chain hypervariable regions, the sequences (SEQ ID NO:76-81) of which are shown below.
  • the anti- CD33 antibody of a PAC comprises the variable light chain sequence of SEQ ID NO: 82 and the variable heavy chain sequce of SEQ ID NO: 83. Table 11.
  • the anti-CD33 antibody of a PAC comprises the light chain sequence of SEQ ID NO: 84 and the heavy chain sequence of SEQ ID NO: 85. In one embodiment, the anti-CD33 antibody of a PAC comprises three light chain hypervariable regions and three heavy chain hypervariable regions, the sequences (Seq ID NO: 84-89) of which are shown below. In one embodiment, the anti-CD33 antibody of a PAC comprises the variable light chain sequence of SEQ ID NO: 90 and the variable heavy chain sequce of SEQ ID NO: 91. In one embodiment, the anti-CD33 antibody of PAC comprises the variable light chain sequence of SEQ ID NO: 92 and the variable heavy chain sequce of SEQ ID NO: 93.
  • the anti-CD33 antibody of the present invention comprises the variable light chain sequence of SEQ ID NO: 94 and the variable heavy chain sequce of SEQ ID NO: 95. In one embodiment, the anti-CD33 antibody of the present invention comprises the variable light chain sequence of SEQ ID NO: 96 and the variable heavy chain sequce of SEQ ID NO: 97.
  • an antibody provided herein has a dissociation constant (Kd) of ⁇ 1 ⁇ M, ⁇ 100 nM, ⁇ 50 nM, ⁇ 10 nM, ⁇ 5 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM, and optionally is ⁇ 10 -13 M. (e.g.10 -8 M or less, e.g. from 10 -8 M to 10 -13 M, e.g., from 10 -9 M to 10 -13 M).
  • Kd dissociation constant
  • Kd is measured by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay.
  • Solution binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of ( 125 I)-labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et al., J. Mol. Biol.293:865-881(1999)).
  • MICROTITER ® multi-well plates (Thermo Scientific) are coated overnight with 5 ⁇ g/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and
  • Fab of interest e.g., consistent with assessment of the anti-VEGF antibody, Fab-12, in Presta et al., Cancer Res.57:4593-4599 (1997).
  • the Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached.
  • Kd is measured using surface plasmon resonance assays using a BIACORE ® -2000 or a BIACORE ® -3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at ⁇ 10 response units (RU).
  • carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N- ethyl-N’- (3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N- hydroxysuccinimide (NHS) according to the supplier’s instructions.
  • Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 ⁇ g/ml ( ⁇ 0.2 ⁇ M) before injection at a flow rate of 5 ⁇ l/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups.
  • a“linker” is a bifunctional or multifunctional moiety that can be used to link one or more PROTAC moieties (D) to an antibody (Ab) to form a PAC.
  • PACs can be prepared using a L1 having reactive functionalities for covalently attaching to the PROTAC and to the antibody. For example, in some
  • a cysteine thiol of an antibody can form a bond with a reactive functional group of a linker or a linker L1-PROTAC group to make a PAC.
  • the chemical structure of the linker can have significant impact on both the efficacy and the safety of a PAC (Ducry & Stump, Bioconjugate Chem, 2010, 21, 5-13). Choosing the right linker influences proper drug delivery to the intended cellular compartment of target cells.
  • Linkers can be generally divided into two categories: cleavable (such as peptide, hydrzone, or disulfide) or non-cleavable (such as thioether).
  • Peptide linkers such as Valine- Citrulline (Val-Cit), that can be hydrolyzed by lysosomal enzymes (such as Cathepsin B) have been used to connect the drug with the antibody (US 6,214,345). They have been particularly useful, due in part to their relative stability in systemic circulation and the ability to efficiently release the drug in tumor.
  • non-peptide linkers which act like peptides and can be effectively cleaved by lysosomal proteases.
  • the greater diversity of non-peptide structures may yield novel, beneficial properties that are not afforded by the peptide linkers.
  • linker L1 Provided herein are different types of non-peptide linkers for linker L1 that can be cleaved by lysosomal enzymes.
  • a. Peptidomimetic Linkers Provided herein are different types of non-peptide, peptidomimetic linkers for PAC that are cleavable by lysosomal enzymes.
  • the amide bond in the middle of a dipeptide e.g. Val-Cit
  • an amide mimic e.g., an amide mimic
  • entire amino acid e.g., valine amino acid in Val-Cit dipeptide
  • L1 is a peptidomimetic linker, it is represented by the following formula
  • Str is a stretcher unit covalently attached to Ab
  • Sp is a bond or spacer unit covalently attached to a PROTAC moiety
  • PM is a non-peptide chemical moiety selected from the group consisting of:
  • W is—NH-heterocycloalkyl- or heterocycloalkyl
  • Y is heteroaryl, aryl, -C(O)C 1 -C 6 alkylene, C 1 -C 6 alkylene-NH 2 , C 1 -C 6 alkylene-NH-CH 3 , C 1 - C 6 alkylene-N-(CH 3 ) 2 , C 1 -C 6 alkenyl or C 1 -C 6 alkylenyl;
  • each R 1 is independently C 1 -C 10 alkyl, C 1 -C 10 alkenyl, (C 1 -C 10 alkyl)NHC(NH)NH 2 or (C 1 - C 10 alkyl)NHC(O)NH 2 ;
  • R 3 and R 2 are each independently H, C 1 -C 10 alkyl, C 1 -C 10 alkenyl, arylalkyl or heteroarylalkyl, or R 3 and R 2 together may form a C 3 -C 7 cycloalkyl;
  • R 4 and R 5 are each independently C 1 -C 10 alkyl, C 1 -C 10 alkenyl, arylalkyl, heteroarylalkyl, (C 1 - C 10 alkyl)OCH 2 -, or R 4 andR 5 may form a C 3 -C 7 cycloalkyl ring.
  • L1 may be connected to the PROTAC through any of the E3LB, L2, or PB groups.
  • Y is heteroaryl; R 4 and R 5 together form a cyclobutyl ring.
  • Y is a moiety selected from the group consisting of: .
  • Str is a chemical moiety represented by the following formula:
  • R 6 is selected from the group consisting of C 1 -C 10 alkylene, C 1 -C 10 alkenyl, C 3 - C 8 cycloalkyl, (C 1 -C 8 alkylene)O-, and C 1 -C 10 alkylene ⁇ C(O)N(R a ) ⁇ C 2 -C 6 alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C 3 -C 8 cycloalkyl, C 4 - C 7 heterocycloalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each R a is independently H or C 1 -C 6 alkyl; Sp is—A
  • Str has the formula:
  • R 7 is selected from C 1 -C 10 alkylene, C 1 -C 10 alkenyl, (C 1 -C 10 alkylene)O-, N(R c ) ⁇ (C 2 -C 6 alkylene) ⁇ N(R c ) and N(R c ) ⁇ (C 2 -C 6 alkylene); where each R c is independently H or C 1 -C 6 alkyl; Sp is—Ar—R b —, wherein Ar is aryl or heteroaryl, R b is (C 1 - C 10 alkylene)O- or Sp -C 1 -C 6 alkylene-C(O)NH-.
  • L1 is a non-peptide chemical moiety represented by the following formula
  • R 1 is C 1 -C 6 alkyl, C 1 -C 6 alkenyl, (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 ;
  • R 3 and R 2 are each independently H or C 1 -C 10 alkyl.
  • L1 is a non-peptide chemical moiety represented by the following formula
  • R 1 is C 1 -C 6 alkyl, (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 ;
  • R 4 and R 5 together form a C 3 -C 7 cycloalkyl ring.
  • L1 is a non-peptide chemical moiety represented by the following formula
  • R 1 is C 1 -C 6 alkyl, (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 and W is as defined above.
  • the linker may be a peptidomimetic linker such as those described in WO2015/095227, WO2015/095124 or WO2015/095223.
  • a Linker L1 forms a disulfide bond with the antibody.
  • the linker has the structure:
  • R 1 and R 2 are independently selected from H and C 1 -C 6 alkyl, or R 1 and R 2 form a 3, 4, 5, or 6-membered cycloalkyl or heterocyclyl group.
  • the linker is covalently bound to an antibody and a PROTAC as follows:
  • the carbonyl group of the linker is connected to an amine group in the PROTAC. It is also noted that the sulfur atom connected to Ab is a sulfur group from a cysteine in the antibody.
  • a linker L1 has a functionality that is capable of reacting with a free cysteine present on an antibody to form a covalent bond.
  • Nonlimiting examples of such reactive functionalities include maleimide, haloacetamides, ⁇ -haloacetyl, activated esters such as succinimide esters, 4-nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates, and isothiocyanates.
  • succinimide esters 4-nitrophenyl esters
  • pentafluorophenyl esters pentafluorophenyl esters, tetrafluorophenyl esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates, and isothiocyanates.
  • a linker has a functionality that is capable of reacting with an electrophilic group present on an antibody.
  • electrophilic groups include, but are not limited to, aldehyde and ketone carbonyl groups.
  • a heteroatom of the reactive functionality of the linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit.
  • reactive functionalities include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide.
  • a linker may comprise one or more linker components.
  • exemplary linker components include 6-maleimidocaproyl (“MC”), maleimidopropanoyl (“MP”), valine-citrulline (“val- cit” or“vc”), alanine-phenylalanine (“ala-phe”), p-aminobenzyloxycarbonyl (a“PAB”), N- Succinimidyl 4-(2-pyridylthio) pentanoate (“SPP”), and 4-(N-maleimidomethyl)
  • MCC cyclohexane-1 carboxylate
  • a linker may be a“cleavable linker,” facilitating release of a PROTAC.
  • Nonlimiting exemplary cleavable linkers include acid-labile linkers (e.g., comprising hydrazone), protease-sensitive (e.g., peptidase-sensitive) linkers, photolabile linkers, or disulfide- containing linkers (Chari et al., Cancer Research 52:127-131 (1992); US 5208020).
  • a linker has the following Formula:
  • A is a“stretcher unit”, and a is an integer from 0 to 1; W is an“amino acid unit”, and w is an integer from 0 to 12; Y is a“spacer unit”, and y is 0, 1, or 2.
  • Exemplary embodiments of such linkers are described in U.S. Patent No.7,498,298.
  • a linker component comprises a“stretcher unit” that links an antibody to another linker component or to a PROTAC moiety.
  • stretcher units are shown below (wherein the wavy line indicates sites of covalent attachment to an antibody, PROTAC, or additional linker components):
  • PROTAC (“D”) Useful PROTACs have the general formula described above. Particular PROTACs are described in US 7,208,157, WO2013/106643, WO2013/106646, and WO2015/160845. PROTACs include those having the following components.
  • E3 Ubiquitin Ligases Binding Groups (E3LB)
  • E3 ubiquitin ligases (of which over 600 are known in humans) confer substrate specificity for ubiquitination. There are known ligands which bind to these ligases. As described herein, an E3 ubiquitin ligase binding group is a peptide or small molecule that can bind an E3 ubiquitin ligase.
  • E3 ubiquitin ligases include: von Hippel-Lindau (VHL); cereblon, XIAP, E3A; MDM2; Anaphase-promoting complex (APC); UBR5 (EDD1); SOCS/ BC-box/ eloBC/ CUL5/ RING; LNXp80; CBX4; CBLL1; HACE1; HECTD1; HECTD2; HECTD3; HECW1; HECW2; HERC1; HERC2; HERC3; HERC4; HUWE1; ITCH; NEDD4; NEDD4L; PPIL2; PRPF19; PIAS1; PIAS2; PIAS3; PIAS4; RANBP2; RNF4; RBX1; SMURF1; SMURF2; STUB1; TOPORS; TRIP12; UBE3A; UBE3B; UBE3C; UBE4A; UBE4B; UBOX5
  • a particular E3 ubiquitin ligase is von Hippel-Lindau (VHL) tumor suppressor, the substrate recognition subunit of the E3 ligase complex VCB, which also consists of elongins B and C, Cul2 and Rbxl.
  • the primary substrate of VHL is Hypoxia Inducible Factor l ⁇ (HIF- l ⁇ ), a transcription factor that upregulates genes such as the pro-angiogenic growth factor VEGF and the red blood cell inducing cytokine erythropoietin in response to low oxygen levels.
  • HIF- l ⁇ Hypoxia Inducible Factor l ⁇
  • Compounds that bind VHL may be hydroxyproline compounds such as those disclosed in WO2013/106643, and other compounds described in US2016/0045607, WO2014187777, US20140356322, and US 9,249,153.
  • MDM2 Another particular E3 ubiquitin ligase is MDM2.
  • small molecular binding compounds for MDM2 include the“nutlin” compounds, e.g., nutlin 3a and nutlin 3, having the structure:
  • MDM2 binding compounds are those described in WO2012/121361;
  • XIAP X-linked inhibitor of apoptosis
  • XIAP is a protein that stops apoptotic cell death. Deregulation of XIAP has been associated with cancer, neurodegenerative disorders and autoimmunity. In the development of lung cancer, the overexpression of XIAP inhibits caspases. In developing prostate cancer, XIAP is one of four IAPs overexpressed in the prostatic epithelium. Mutations in the XIAP gene can result in a severe and rare type of inflammatory bowel disease. Defects in the XIAP gene can also result in an extremely rare condition called X-linked lymphoproliferative disease.
  • Degradation of XIAP can enhance apoptosis by preventing XIAP from binding to caspases. This allows normal caspase activity to proceed.
  • small molecular binding compounds for XIAP include compounds disclosed in US 9,096,544; WO 2015187998; WO 2015071393; US 9,278,978; US 9,249,151; US 20160024055; US 20150307499; US 20140135270; US 20150284427; US 20150259359; US 20150266879; US 20150246882; US 20150252072; US 20150225449; US 8,883,771, J. Med. Chem., 2015, 58(16) 6574-6588 and Small-molecule Pan-IAP Antagonists: A Patent Review (2010) Expert Opin Ther Pat; 20: 251-67 (Flygare & Fairbrother). Specific compounds include all the tetrahydro-benzodiazinone compounds of the following formula:
  • XIAP binding moiety can bind to the BIR2 or BIR3 domain of XIAP or both.
  • Cereblon is a protein that forms an E3 ubiquitin ligase complex with damaged DNA binding protein 1 (DDB1), Cullin-4A (CUL4A), and regulator of cullins 1 (ROC1). This complex ubiquitinates a number of other proteins. Cereblon ubquitination of target proteins results in increased levels of fibroblast growth factor 8 (FGF8) and fibroblast growth factor 10 (FGF10). FGF8 in turn regulates a number of developmental processes, such as limb and auditory vesicle formation. In the absence of cereblon, DDB1 forms a complex with DDB2 that functions as a DNA damage- binding protein.
  • DDB1 forms a complex with DDB2 that functions as a DNA damage- binding protein.
  • Thalidomide, lenalidomide, pomalidomide and analogs thereof are known to bind to cereblon.
  • the crystal structure of cereblon with thalidomide and derivative compounds are described in US2015/0374678.
  • Other small molecule compounds that bind to cereblon are also known, e.g., the compounds disclosed as an in US2016/0058872 and US2015/0291562.
  • phthalimide conjugation with binders, such as antagonists, of BET bromodomains can provide PROTACs with highly-selective cereblon-dependent BET protein degradation. Winter et al., Science, June 19, 2015, p.1376.
  • Such PROTACs can be conjugated to an antibody as described herein to form a PAC.
  • the PB component is a group which binds to a target protein intended to be degraded.
  • protein includes oligopeptides and polypeptide sequences of sufficient length that they can bind to a PB group. Any protein in a eukaryotic system or a microbial system, including a virus, bacteria or fungus, as otherwise described herein, are targets for ubiquitination mediated by the compounds described herein.
  • PB groups include, for example, any moiety which binds to a protein specifically (binds to a target protein) and includes the following non-limiting examples of small molecule target protein moieties: Hsp90 inhibitors, kinase inhibitors, MDM2 inhibitors, compounds targeting Human BET Bromodomain- containing proteins, HDAC inhibitors, human lysine methyltransferase inhibitors, angiogenesis inhibitors, immunosuppressive compounds, and compounds targeting the aryl hydrocarbon receptor (AHR), among numerous others.
  • the compositions described below exemplify some of the members of these nine types of small molecule target protein binding moieties.
  • Such small molecule target protein binding moieties also include pharmaceutically acceptable salts, enantiomers, solvates and polymorphs of these compositions, as well as other small molecules that may target a protein of interest.
  • target proteins may include, for example, structural proteins, receptors, enzymes, cell surface proteins, proteins pertinent to the integrated function of a cell, including proteins involved in catalytic activity, aromatase activity, motor activity, helicase activity, metabolic processes (anabolism and catrabolism), antioxidant activity, proteolysis, biosynthesis, proteins with kinase activity, oxidoreductase activity, transferase activity, hydrolase activity, lyase activity, isomerase activity, ligase activity, enzyme regulator activity, signal transducer activity, structural molecule activity, binding activity (protein, lipid carbohydrate), receptor activity, cell motility, membrane fusion, cell communication, regulation of biological processes, development, cell differentiation, response to stimulus, behavioral proteins, cell adhesion proteins, proteins involved in cell death, proteins involved in transport (including protein transporter activity, nuclear transport, ion transporter activity, channel transporter activity, carrier activity, permease activity, secretion activity, electron transporter activity, pathogenesis, chaperone regulator activity, nucleic acid
  • Proteins of interest can include proteins from eurkaryotes and prokaryotes including humans as targets for drug therapy, other animals, including domesticated animals, microbials for the determination of targets for antibiotics and other antimicrobials and plants, and even viruses, among numerous others.
  • the PB component of a PAC is any peptide or small molecule that bind protein targets such as FoxOl, HDAC, DP-1, E2F, ABL, AMPK, BRK, BRSK I, BRSK2, BTK, CAMKK1, CAMKK alpha, CAMKK beta, Rb, Suv39HI, SCF, p19INK4D, GSK-3, pi 8 INK4, myc, cyclin E, CDK2, CDK9, CDG4/6, Cycline D, pl6 INK4A, cdc25A, BMI1, SCF, Akt, CHKl/2, C 1 delta, CK1 gamma, C 2, CLK2, CSK, DDR2, DYRK1A/2/3, EF2K, EPH-A2/A4/B1/B2/B3/B4, EIF2A 3, Smad2, Smad3, Smad4, Smad7, p53, p21 Cipl, PAX, Fyn, CAS, C
  • PB groups are small molecule compounds such as those disclosed in
  • HSP90 Heat Shock Protein 90
  • Kinase and Phosphatase inhibitors MDM2 inhibitors
  • HDAC inhibitors Human Lysine Methyltransferase Inhibitors
  • Angiogenesis inhibitors Immunosuppressive compounds, as well as compounds that target: Human BET Bromodomain-containing proteins, the aryl hydrocarbon receptor (AHR), REF receptor kinase, FKBP, Androgen Receptor (AR), Estrogen receptor (ER), Thyroid Hormone
  • AHR aryl hydrocarbon receptor
  • REF receptor kinase FKBP
  • AR Androgen Receptor
  • Estrogen receptor ER
  • Thyroid Hormone Thyroid Hormone
  • the E3LB and PB groups of PROTACs as described herein can be connected with linker (L2).
  • the linker group L2 is a group comprising one or more covalently connected structural units of A (e.g., -A 1 ... A q -), wherein A 1 is a group coupled to at least one of a E3LB, a PB, or a combination thereof.
  • a 1 links a E3LB, a PB, or a combination thereof directly to another E3LB, PB, or combination thereof.
  • a 1 links a EL3B, a PB, or a combination thereof indirectly to another E3LB, PB, or combination thereof through A q .
  • a 1 to A q are, each independently, a bond, CR La R Lb , O, S, SO, SO 2 , NR Lc , SO 2 NR Lc , SONR Lc , CONR Lc , NR Lc CONR Ld , NR Lc SO 2 NR Ld , CO, CR La ⁇ CR Lb , C ⁇ C, SiR La R Lb , P(O)R La , P(O)OR La , NR Lc C( ⁇ NCN)NR Ld , NR Lc C( ⁇ NCN),
  • NR Lc C( ⁇ CNO 2 )NR Ld C 3-11 cycloalkyl optionally substituted with 0-6 R La and/or R Lb groups, C 3-11 heterocyclyl optionally substituted with 0-6 R La and/or R Lb groups, aryl optionally substituted with 0-6 R La and/or R Lb groups, heteroaryl optionally substituted with 0-6 R La and/or R Lb groups, where R La or R Lb , each independently, can be linked to other A groups to form cycloalkyl and/or heterocyclyl moeity which can be further substituted with 0-4 R Le groups; wherein R La , R Lb , R Lc , R Ld and R Le are, each independently, H, halo, C 1-8 alkyl, OC 1- 8 alkyl, SC 1-8 alkyl, NHC 1-8 alkyl, N(C 1-8 alkyl) 2 , C 3-11 cycloalkyl, aryl, heteroaryl,
  • q is an integer greater than or equal to 0. In certain embodiments, q is an integer greater than or equal to 1.
  • a q is a group which is connected to an E3LB moiety, and A 1 and A q are connected via structural units of A (number of such structural units of A: q-2).
  • a q is a group which is connected to A 1 and to an E3LB moiety.
  • the structure of the linker group L2 is -A 1 -, and A 1 is a group which is connected to an E3LB moiety and a PB moiety.
  • q is an integer from 1 to 100, 1 to 90, 1 to 80, 1 to 70, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, or 1 to 10.
  • the linker (L2) is selected from the group consisting of):
  • the linker group is an optionally substituted
  • (poly)ethyleneglycol having between 1 and about 100 ethylene glycol units, between about 1 and about 50 ethylene glycol units, between 1 and about 25 ethylene glycol units, between about 1 and 10 ethylene glycol units, between 1 and about 8 ethylene glycol units and 1 and 6 ethylene glycol units, between 2 and 4 ethylene glycol units, or optionally substituted alkyl groups interdispersed with optionally substituted, O, N, S, P or Si atoms.
  • the linker is substituted with an aryl, phenyl, benzyl, alkyl, alkylene, or heterocycle group.
  • the linker may be asymmetric or symmetrical.
  • the linker group may be any suitable moiety as described herein.
  • the linker is a substituted or unsubstituted polyethylene glycol group ranging in size from about 1 to about 12 ethylene glycol units, between 1 and about 10 ethylene glycol units, about 2 about 6 ethylene glycol units, between about 2 and 5 ethylene glycol units, between about 2 and 4 ethylene glycol units.
  • the E3LB group and PB group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker.
  • the linker is independently covalently bonded to the E3LB group and the PB group preferably through an amide, ester, thioester, keto group, carbamate (urethane), carbon or ether, each of which groups may be inserted anywhere on the E3LB group and PB group to provide maximum binding of the E3LB group on the ubiquitin ligase and the PB group on the target protein to be degraded.
  • the target protein for degradation may be the ubiquitin ligase itself.
  • the linker may be linked to an optionally substituted alkyl, alkylene, alkene or alkyne group, an aryl group or a heterocyclic group on the E3LB and/or PB groups. It is noted that an E3LB group or a PB group may need to be derivatized to make a chemical functional group that is reactive with a chemical functional group on the linker. Alternatively, the linker may need to be derivatized to include a chemical functional group that can react with a functional group found on E3LB and/or PB.
  • L2 can also be represented by the formula:
  • Z is a group which links E3LB to X; and X is a group linking Z to group PB.
  • Z is absent (a bond), -(CH 2 )i-O, -(CH 2 )i-S, -(CH 2 )i-N-R, a (CH 2 ) i - X 1 Y 1 group wherein X 1 Y 1 forms an amide group, or a urethane group, ester or thioester group, or a
  • each R is H, or a C 1 -C 3 alkyl, an alkanol group or a heterocycle (including a water soluble heterocycle, preferably, a morpholino , piperidine or piperazine group to promote water solubility of the linker group); each Y is independently a bond, O, S or N-R; and each i is independently 0 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
  • X is a
  • each V is independently a bond (absent)
  • j is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
  • k is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5; preferably k is 1, 2, 3, 4, or 5;
  • m' is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
  • n is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, l to 8, 1 to 6, 1 , 2, 3, 4 or 5;
  • X 1 is O, S or N-R, preferably O;
  • CON is a connector group (which may be a bond) which connects Z to X, when present in the linker group.
  • CON is a bond (absent), a heterocycle including a water soluble heterocycle such as a piperazinyl or other group or a group,
  • X 2 is O, S, NR 4 , S(O), S(O) 2 , -S(O) 2 O, -OS(O) 2 , or OS(O) 2 O;
  • X 3 is O, S, CHR 4 , NR 4 ;
  • R is H or a C 1 -C 3 alkyl group optionally substituted with one or two hydroxyl groups, or a pharmaceutically acceptable salt, enantiomer or stereoisomer thereof.
  • the linker group is a (poly)ethyleneglycol having between 1 and about 100 ethylene glycol units, between about 1 and about 50 ethylene glycol units, between 1 and about 25 ethylene glycol units, between about 1 and 10 ethylene glycol units, between 1 and about 8 ethylene glycol units and 1 and 6 ethylene glycol units, between 2 and 4 ethylene glycol units.
  • CON is N-phenyl
  • the E3LB group and PB group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker, in preferred aspects, the linker is independently covalently bonded to the E3LB group and the PB group through an amide, ester, thioester, keto group, carbamate (urethane) or ether, each of which groups may be inserted anywhere on the E3LB group and PB group to allow binding of the E3LB group to the ubiquitin ligase and the PB group to the target protein to be degraded.
  • the linker can be designed and connected to E3LB and PB to minimize, eliminate, or neutralize any impact its presence might have on the binding of E3LB and PB to their respective binding partners.
  • the targeted protein for degradation may be an ubiquitin ligase.
  • a PAC can comprise a single antibody where the single antibody can have more than one PROTAC, each PROTAC covalently linked to the antibody through a linker L1.
  • The“PROTAC loading” is the average number of PROTAC moieties per antibody.
  • PROTAC loading may range from 1 to 8 PROTAC (D) per antibody (Ab). That is, in the PAC formula, Ab ⁇ (L1 ⁇ D) p , p has a value from about 1 to about 50, from about 1 to about 8, from about 1 to about 5, from about 1 to about 4, or from about 1 to about 3.
  • Each PROTAC covalently linked to the antibody through linker L1 can be the same or different PROTAC and can have a linker of the same type or different type as any other L1 covalently linked to the antibody.
  • Ab is a cysteine engineered antibody and p is about 2.
  • ELISA assay for detection of PACs does not determine where the PROTAC moieties are attached to the antibody, such as the heavy chain or light chain fragments, or the particular amino acid residues.
  • separation, purification, and characterization of homogeneous PACs where p is a certain value from PACs with other PROTAC loadings may be achieved by means such as reverse phase HPLC or
  • p may be limited by the number of attachment sites on the antibody.
  • an antibody may have only one or several cysteine thiol groups, or may have only one or several sufficiently reactive thiol groups through which a linker may be attached.
  • Another reactive site on an Ab to connect L1-Ds are the amine functional group of lysine residues.
  • Values of p include values from about 1 to about 50, from about 1 to about 8, from about 1 to about 5, from about 1 about 4, from about 1 to about 3, and where p is equal to 2.
  • the subject matter described herein is directed to any the PACs, wherein p is about 1, 2, 3, 4, 5, 6, 7, or 8.
  • an antibody may contain, for example, many lysine residues that do not react with the linker L1-PROTAC group (L1-D) or linker reagent. Only the most reactive lysine groups may react with an amine-reactive linker reagent. Also, only the most reactive cysteine thiol groups may react with a thiol-reactive linker reagent or linker L1-PROTAC group. Generally, antibodies do not contain many, if any, free and reactive cysteine thiol groups which may be linked to a PROTAC moiety.
  • cysteine thiol residues in the antibodies of the compounds exist as disulfide bridges and must be reduced with a reducing agent such as dithiothreitol (DTT) or TCEP, under partial or total reducing conditions.
  • a reducing agent such as dithiothreitol (DTT) or TCEP
  • DTT dithiothreitol
  • TCEP TCEP
  • the PROTAC loading (PROTAC/antibody ratio,“PAR”) of a PAR may be controlled in several different manners, including: (i) limiting the molar excess of linker L1-PROTAC group or linker reagent relative to antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) partial or limiting reductive conditions for cysteine thiol modification.
  • PROTACs described herein can be covalently linked to a linker L1 to prepare L1- PROTAC groups. These compounds have the following general formula:
  • D is a PROTAC having the structure E3LB ⁇ L2 ⁇ PB; wherein, E3LB is an E3 ligase binding group covalently bound to L2; L2 is a linker covalently bound to E3LB and PB; PB is a protein binding group covalently bound to L2; and L1 is a linker, covalently bound to D.
  • E3LB is an E3 ligase binding group covalently bound to L2
  • L2 is a linker covalently bound to E3LB and PB
  • PB is a protein binding group covalently bound to L2;
  • L1 is a linker, covalently bound to D.
  • L1 is as described elsewhere herein, including a peptidomimetic linker.
  • the L1-PROTAC has the following formula:
  • Str is a stretcher unit
  • Sp is a bond or a spacer unit covalently attached to D, i.e., a PROTAC moiety
  • R 1 is C 1 -C 10 alkyl, (C 1 -C 10 alkyl)NHC(NH)NH 2 or (C 1 -C 10 alkyl)NHC(O)NH 2 ;
  • R 4 and R 5 are each independently C 1 -C 10 alkyl, arylalkyl, heteroarylalkyl, (C 1 -C 10 alkyl )OCH 2 -, or R 4 and R 5 may form a C 3 -C 7 cycloalkyl ring;
  • D is a PROTAC moiety.
  • An L1-PROTAC compound can be represented by the following formula:
  • R 6 is C 1 -C 10 alkylene
  • R 4 and R 5 together form a C 3 -C 7 cycloalkyl ring
  • D is a PROTAC moeity.
  • An L1-PROTAC compound can be represented by the following formula:
  • R 1 , R 4 and R 5 are as described elsewhere herein, and D is a PROTAC moiety.
  • Str is a stretcher unit
  • Sp is an optional spacer unit covalently attached to D, i.e., a PROTAC moiety
  • Y is heteroaryl, aryl, -C(O)C 1 -C 6 alkylene, C 1 -C 6 alkylene-NH 2 , C 1 -C 6 alkylene-NH-CH 3 , C 1 - C 6 alkylene-N-(CH 3 ) 2 , C 1 -C 6 alkenyl or C 1 -C 6 alkylenyl;
  • R 1 is C 1 -C 10 alkyl, (C 1 -C 10 alkyl)NHC(NH)NH 2 or (C 1 -C 10 alkyl)NHC(O)NH 2 ;
  • R 3 and R 2 are each independently H, C 1 -C 10 alkyl, arylalkyl or heteroarylalkyl, or R 3 and R 2 together may form a C 3 -C 7 cycloalkyl;
  • D is a PROTAC moiety.
  • An L1-PROTAC compound can be represented by the following formula:
  • R 6 is C 1 -C 10 alkylene, and R 1 , R 2 and R 3 are as described elsewhere herein, and D is a PROTAC moiety
  • An L1-PROTAC compound can be represented by the following formula:
  • R 1 , R 2 and R 3 are as described elsewhere herein, and D is a PROTAC moiety.
  • Str can have the following formula:
  • R 6 is selected from the group consisting of C 1 -C 10 alkylene, C 3 -C 8 cycloalkyl, O-(C 1 - C 8 alkylene), and C 1 -C 10 alkylene ⁇ C(O)N(R a ) ⁇ C 2 -C 6 alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo,
  • each R a is independently H or C 1 -C 6 alkyl
  • Sp is—Ar—R b —, wherein Ar is aryl or heteroaryl, R b is (C 1 -C 10 alkylene)O-.
  • R 6 is C 1 -C 10 alkylene
  • Sp is—Ar—R b —
  • Ar is aryl
  • R b is (C 1 -C 6 alkylene)O-
  • R 6 is–(CH 2 ) q is 1-10;
  • Str can have the following formula:
  • R 7 is selected from C 1 -C 10 alkylene, C 1 -C 10 alkylene ⁇ O, N(R c ) ⁇ (C 2 -C 6 alkylene) ⁇ N(R c ) and N(R c ) ⁇ (C 2 - C 6 alkylene); where each R c is independently H or C 1 -C 6 alkyl;
  • Sp is—Ar—R b —, wherein Ar is aryl or heteroaryl, R b is (C 1 -C 10 alkylene)O-; or wherein R 6 is C 1 -C 10 alkylene, Sp is—Ar—R b —, wherein Ar is aryl R b is (C 1 -C 6 alkylene)O-.
  • L1-PROTAC can have the following formulae, wherein in each instance, D is a PROTAC moiety:
  • PB is as described elsewhere herein, or is selected from the group consisting of Heat Shock Protein 90 (HSP90) inhibitors, Kinase and Phosphatase inhibitors, MDM2 inhibitors, HDAC inhibitors, Human Lysine Methyltransferase Inhibitors, Angiogenesis inhibitors, Immunosuppressive compounds, as well as compounds that target: Human BET Bromodomain-containing proteins, the aryl hydrocarbon receptor (AHR), REF receptor kinase, FKBP, Andrpgen Receptor (AR), Estrogen receptor (ER), Thyroid Hormone Receptor, HIV Protease, HIV Integrase, HCV Protease, Acyl-protein Thioesterase-1 and -2 (APT1 and APT2).
  • HSP90 Heat Shock Protein 90
  • AHR aryl hydrocarbon receptor
  • REF receptor kinase FKBP
  • AR Andrpgen Receptor
  • Estrogen receptor ER
  • Thyroid Hormone Receptor
  • E3LB is as described elsewhere herein, including a group that binds XIAP, VHL, cereblon and MDM2.
  • the subject matter described herein is also directed to methods of preparing a PAC from a L1-PROTAC compound, the method comprising contacting an antibody, or variants, mutations, splice variants, indels and fusions thereof, with a L1-PROTAC under conditions where the antibody is covalently bound to any available point of attachment on a L1- PROTAC, wherein a PAC is prepared.
  • the subject matter described herein is also directed to methods of preparing a PAC from an Ab-L1 portion, i.e., an antibody, or variants, mutations, splice variants, indels and fusions thereof, covalently attached to a L1, the methods comprising contacting a PROTAC with an Ab-L1 under conditions where the PROTAC is covalently bound to any available point of attachment on the Ab-L1, wherein a PAC is prepared.
  • the methods can further comprise routine isolation and purification of the PACs.
  • a PAC and a L1-PROTAC compound as described herein, these can exist in solid or liquid form. In the solid state, it may exist in crystalline or noncrystalline form, or as a mixture thereof.
  • pharmaceutically acceptable solvates may be formed for crystalline or non-crystalline compounds.
  • solvent molecules are incorporated into the crystalline lattice during crystallization.
  • Solvates may involve non-aqueous solvents such as, but not limited to, ethanol, isopropanol, DMSO, acetic acid, ethanolamine, or ethyl acetate, or they may involve water as the solvent that is incorporated into the crystalline lattice.
  • Hydrates wherein water is the solvent incorporated into the crystalline lattice are typically referred to as "hydrates.” Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The subject matter described herein includes all such solvates. The skilled artisan will further appreciate that certain compounds and PACs described herein that exist in crystalline form, including the various solvates thereof, may exhibit polymorphism (i.e. the capacity to occur in different crystalline structures). These different crystalline forms are typically known as “polymorphs.” The subject matter disclosed herein includes all such polymorphs. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state.
  • Polymorphs may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification. The skilled artisan will appreciate that different polymorphs may be produced, for example, by changing or adjusting the reaction conditions or reagents, used in making the compound. For example, changes in temperature, pressure, or solvent may result in polymorphs. In addition, one polymorph may spontaneously convert to another polymorph under certain conditions.
  • Compounds and PACs described herein or a salt thereof may exist in stereoisomeric forms (e.g., it contains one or more asymmetric carbon atoms).
  • the individual stereoisomers (enantiomers and diastereomers) and mixtures of these are included within the scope of the subject matter disclosed herein.
  • a compound or salt of Formula (I) may exist in tautomeric forms other than that shown in the formula and these are also included within the scope of the subject matter disclosed herein. It is to be understood that the subject matter disclosed herein includes all combinations and subsets of the particular groups described herein.
  • the subject matter disclosed herein also includes isotopically-labelled forms of the compounds described herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds described herein and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulphur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 Cl, 123 I and 125 I.
  • Isotopically-labelled compounds are disclosed herein, for example those into which radioactive isotopes such as 3 H, 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are commonly used for their ease of preparation and detectability.
  • 11 C and 18 F isotopes are useful in PET (positron emission tomography), and 125 I isotopes are useful in SPECT (single photon emission computerized tomography), all useful in brain imaging.
  • substitution with heavier isotopes such as deuterium, i.e., 2 H can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances.
  • Isotopically labelled compounds of formula I can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples below, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
  • D is a PROTAC having the structure E3LB ⁇ L2 ⁇ PB;
  • E3LB is an E3 ligase binding group covalently bound to L2;
  • L2 is a linker covalently bound to E3LB and PB;
  • PB is a protein binding group covalently bound to L2;
  • Ab is an antibody covalently bound to L1;
  • L1 is a linker, covalently bound to Ab and to D;
  • p has a value from about 1 to about 8.
  • E3LB is a group that binds an E3 ligase
  • E3 ligase is listed in Tables 13-27, e.g., Table 13, Table 14, Table 15, Table 16, Table 17, Table 18, Table 19, Table 20, Table 21, Table 22, Table 23, Table 24, Table 25, Table 26, or Table 27.
  • E3LB is a group that binds an E3 ligase, wherein the E3 ligase is selected from the group consisting of von Hippel-Lindau (VHL); cereblon; XIAP; E3A; MDM2; Anaphase-promoting complex (APC); UBR5
  • ETD1 ELD1
  • SOCS/ BC-box/ eloBC/ CUL5/ RING LNXp80
  • CBX4 CBLL1
  • HACE1 HACE1
  • RANBP2 RANBP2; RNF4; RBX1; SMURF1; SMURF2; STUB1; TOPORS; TRIP12; UBE3A;
  • HOIP/RNF31 cIAP-1/HIAP-2; cIAP-2/HIAP-1; cIAP (pan); ITCH/AIP4; KAP1; MARCH8; MDM2/HDM2; Mind Bomb 1/MIB1; Mind Bomb 2/MIB2; MuRF1/TRIM63; NDFIP1; NEDD4; NleL; Parkin; RNF2; RNF4; RNF8; RNF168; RNF43; SART1; Skp2; SMURF2; TRAF-1; TRAF-2; TRAF-3; TRAF-4; TRAF-5; TRAF-6; TRIM5; TRIM21; TRIM32;
  • E3LB is a group that binds an E3 ligase selected from the group consisting of XIAP, VHL, cereblon and MDM2.
  • E3LB is selected from the group consisting of a compound that binds VHL, a hydroxyproline compound that binds VHL, a compound that binds MDM2, a compound that binds cereblon, a tetrahhydro- benzodiazepinone, a nutlin, and small molecular binding compounds as described herein. 6.
  • E3LB is a XIAP inhibitor that is a tetrahydro-benzodiazepinone having the formula:
  • R1, R2, R3, R4 and R5 are as described in WO/2015/071393, and including all compounds therein.
  • PB is a group that binds FoxOl, HDAC, DP-1, E2F, ABL, AMPK, BRK, BRSK I, BRSK2, BTK, CAMKK1, CAMKK alpha, CAMKK beta, Rb, Suv39HI, SCF, p19INK4D, GSK-3, pi 8 INK4, myc, cyclin E, CDK2, CDK9, CDG4/6, Cycline D, pl6 INK4A, cdc25A, BMI1, SCF, Akt, CHKl/2, C 1 delta, CK1 gamma, C 2, CLK2, CSK, DDR2, DYRK1A/2/3, EF2K, EPH-A2/A4/B1/B2/B3/B4, EIF2A 3, Smad2, Smad3, Smad4, Smad7, p53, p21 Cipl, PAX, Fyn, CAS, C3G, SOS, Tal
  • PB is selected from the group consisting of Heat Shock Protein 90 (HSP90) inhibitors, Kinase and Phosphatase inhibitors, MDM2 inhibitors, HDAC inhibitors, Human Lysine Methyltransferase Inhibitors,
  • Angiogenesis inhibitors IL-1 angiogenesis inhibitors, Immunosuppressive compounds, and compounds that target:
  • PB is a compound that targets Estrogen Receptor alpha (ER ⁇ ).
  • Ly6G6D LGR5; RET; LY6K; GPR19; GPR54; ASPHD1; Tyrosinase; TMEM118;
  • GPR172A MUC16 and CD33.
  • TrpM4 CRIPTO; CD21; CD79b; FcRH2; B7-H4; HER2; CD22; CD79a; CD72; LY64; Ly6E; MUC16; and CD33.
  • Str is a stretcher unit covalently attached to Ab
  • Sp is a bond or spacer unit covalently attached to a PROTAC moiety
  • PM is a non-peptide chemical moiety selected from the group consisting of:
  • W is–NH-heterocycloalkyl- or heterocycloalkyl
  • Y is heteroaryl, aryl, -C(O)C 1 -C 6 alkylene, C 1 -C 6 alkylene-NH 2 , C 1 -C 6 alkylene-NH-CH 3 , C 1 - C 6 alkylene-N-(CH 3 ) 2 , C 1 -C 6 alkenyl or C 1 -C 6 alkylenyl; each R 1 is independently C 1 -C 10 alkyl, C 1 -C 10 alkenyl, (C 1 -C 10 alkyl)NHC(NH)NH 2 or (C 1 - C 10 alkyl)NHC(O)NH 2 ; R 3 and R 2 are each independently H, C 1 -C 10 alkyl, C 1 -C 10 alkenyl, arylalkyl or heteroarylalkyl, or R 3 and R 2 together may form a C 3 -C 7 cyclo
  • Str is a chemical moiety represented by the following formula:
  • R 6 is selected from the group consisting of C 1 -C 10 alkylene, C 1 -C 10 alkenyl, C 3 - C 8 cycloalkyl, (C 1 -C 8 alkylene)O-, and C 1 -C 10 alkylene ⁇ C(O)N(R a ) ⁇ C 2 -C 6 alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C 3 -C 8 cycloalkyl, C 4 - C 7 heterocycloalkyl, heteroarylalkyl, aryl arylalkyl, heteroarylalkyl and heteroaryl each R a is independently H or C 1 -C 6 alkylene
  • R 7 is selected from C 1 -C 10 alkylene, C 1 -C 10 alkenyl, (C 1 -C 10 alkylene)O-, N(R c ) ⁇ (C 2 - C 6 alkylene) ⁇ N(R c ) and N(R c ) ⁇ (C 2 -C 6 alkylene); where each R c is independently H or C 1 -C 6 alkyl; Sp is -C 1 -C 6 alkylene-C(O)NH- or—Ar—R b —, wherein Ar is aryl or heteroaryl, R b is (C 1 - C 10 alkylene)O-.
  • R 1 is C 1 -C 6 alkyl, C 1 -C 6 alkenyl, (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 ;
  • R 3 and R 2 are each independently H, C 1 -C 10 alkyl ; and Str and Sp are as defined herein.
  • R 1 is C 1 -C 6 alkyl, (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 ; Str and Sp are as defined herein; and R 4 and R 5 together form a C 3 -C 7 cycloalkyl ring. 25. The conjugate of any above embodiment, wherein L1 has the following formula
  • Str and Sp are as defined herein; and R 1 is C 1 -C 6 alkyl, (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 . 26.
  • R 6 is selected from the group consisting of C 1 -C 10 alkylene, and C 1 - C 10 alkylene ⁇ C(O)N(R a ) ⁇ C 2 -C 6 alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C 3 -C 8 cycloalkyl, C 4 -C 7 heterocycloalkyl, heteroarylalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each R a is independently H or C 1 -C 6 alkyl; Ab and Sp are as defined herein; and p is 1, 2, 3 or 4. 27.
  • Str is a chemical moiety represented by the following formula:
  • R 6 is selected from the group consisting of C 1 -C 10 alkylene, and C 1 - C 10 alkylene ⁇ C(O)N(R a ) ⁇ C 2 -C 6 alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C 3 -C 8 cycloalkyl, C 4 -C 7 heterocycloalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each R a is independently H or C 1 -C 6 alkyl; R 1 , R 4 , R 5 , Ab, D and Sp are as defined herein; and p is 1,
  • Str is a chemical moiety represented by the following formula: , R 6 is C 1 -C 6 alkylene;
  • Sp is -C 1 -C 6 alkylene-C(O)NH- or—Ar—R b —, where Ar is aryl, R b is (C 1 -C 3 alkylene)O-. 33.
  • R 1 is C 1 -C 6 alkyl-NH 2 , (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 ; and p is 1, 2 ,3 or 4.
  • Ab and D are as defined herein; p is 1, 2, 3 or 4; R 1 is C 1 -C 6 alkyl-NH 2 , (C 1 -C 6 alkyl)NHC(NH)NH 2 or (C 1 -C 6 alkyl)NHC(O)NH 2 ; R 4 and R 5 are each independently C 1 -C 6 alkyl, wherein the alkyl are unsubstituted, or R 4 and R 5 together with the carbon to which each is attached can form a C 3 -C 7 cycloalkyl ring, such as a cyclobutyl. 35.
  • L1 has the following formula:
  • R 1 and R 2 are independently selected from H and C 1 -C 6 alkyl, or R 1 and R 2 form a 3, 4, 5, or 6-membered cycloalkyl or heterocyclyl group.
  • a method of treating a disease in a human in need thereof comprising administering to the human an effective amount of a conjugate of any above embodiment.
  • a pharmaceutical composition comprising a conjugate of any above embodiment and a pharmaceutically acceptable excipient.
  • a hyperproliferative disorder including, benign or malignant solid tumors and hematological disorders, disorders involving neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic, immunologic, and autoimmune conditions.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, and head and neck cancer.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer
  • autoimmune disease is selected from the group consisting of rheumatologic disorders (such as, for example, rheumatoid arthritis, Sjögren's syndrome, scleroderma, lupus such as systemic lupus erythematosus (SLE) and lupus nephritis, polymyositis/dermatomyositis, cryoglobulinemia, anti-phospholipid antibody syndrome, and psoriatic arthritis), osteoarthritis, autoimmune gastrointestinal and liver disorders (such as, for example, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), autoimmune gastritis and pernicious anemia, autoimmune hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, and celiac disease), vasculitis (such as, for example, ANCA-associated vasculitis, including Churg-Strauss
  • rheumatologic disorders such
  • autoimmune disease is selected from the group consisiting of rheumatoid arthritis, ulcerative colitis, ANCA-associated vasculitis, lupus, multiple sclerosis, Sjögren's syndrome, Graves’ disease, IDDM, pernicious anemia, thyroiditis, and glomerulonephritis.
  • a method of preparing a PAC comprising contacting a Linker L1- PROTAC with an antibody.
  • PACs and compounds described herein can be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein, and those for other heterocycles described in:
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz or CBZ) and 9-fluorenylmethyleneoxycarbonyl (Fmoc).
  • BOC t-butoxycarbonyl
  • CBz or CBZ benzyloxycarbonyl
  • Fmoc 9-fluorenylmethyleneoxycarbonyl
  • a PAC may be prepared by connecting a PROTAC with a L1 linker reagent according to the procedures of WO 2013/055987; WO 2015/023355; WO
  • a PAC may be prepared by first connecting an antibody or variant, mutation, splice variant, indel and fusion thereof, including a cysteine engineered antibody, described herein with a L1 linker reagent, and conjugating it with any PROTAC.
  • the following synthetic routes describe exemplary methods of preparing PACs and components thereof. Other synthetic routes for preparing PACs and components thereof are disclosed elsewhere herein.
  • Schemes 1-4 depict synthesis routes to exemplary linkers L1 for disulfide attachment to antibody Ab.
  • the Ab is connected to L1 through a disulfide bond and the PROTAC is connected to L1 through any available attachment on the
  • cysteine engineered antibodies for conjugation by reduction and reoxidation, they can be prepared generally as follows. Light chain amino acids are numbered according to Kabat (Kabat et al., Sequences of proteins of immunological interest, (1991) 5th Ed., US Dept of Health and Human Service, National Institutes of Health, Bethesda, MD). Heavy chain amino acids are numbered according to the EU numbering system (Edelman et al (1969) Proc. Natl. Acad. of Sci.63(1):78-85), except where noted as the Kabat system. Single letter amino acid abbreviations are used.
  • THIOMABTM antibodies Full length, cysteine engineered monoclonal antibodies (THIOMABTM antibodies) expressed in CHO cells bear cysteine adducts (cystines) or are glutathionylated on the engineered cysteines due to cell culture conditions.
  • cysteine adducts cysteine adducts
  • cysteine glutathionylated on the engineered cysteines due to cell culture conditions.
  • THIOMABTM antibodies purified from CHO cells cannot be conjugated to Cys-reactive linker L1-PROTAC intermediates.
  • Cysteine engineered antibodies may be made reactive for conjugation with linker-PROTAC intermediates described herein, 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) followed by re- formation of the inter-chain disulfide bonds (re-oxidation) with a mild oxidant such as dehydroascorbic acid.
  • 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) followed by re- formation of the inter
  • THIOMABTM antibodies expressed in CHO cells (Gomez et al (2010) Biotechnology and Bioeng.105(4):748-760; Gomez et al (2010) Biotechnol. Prog.26:1438-1445) were reduced, for example, with about a 50 fold excess of DTT overnight in 50 mM Tris, pH 8.0 with 2 mM EDTA at room temperature, which removes Cys and glutathione adducts as well as reduces interchain disulfide bonds in the antibody. Removal of the adducts was monitored by reverse-phase LCMS using a PLRP-S column. The reduced THIOMABTM antibody was diluted and acidified by addition to at least four volumes of 10 mM sodium succinate, pH 5 buffer.
  • the antibody was diluted and acidified by adding to at least four volumes of 10 mM succinate, pH 5 and titration with 10% acetic acid until pH was approximately five.
  • the pH-lowered and diluted THIOMABTM antibody was subsequently loaded onto a HiTrap S cation exchange column, washed with several column volumes of 10 mM sodium acetate, pH 5 and eluted with 50 mM Tris, pH 8.0, 150 mM sodium chloride. Disulfide bonds were reestablished between cysteine residues present in the parent Mab by carrying out reoxidation.
  • the eluted reduced THIOMABTM antibody described above is treated with 15X dehydroascorbic acid (DHAA) for about 3 hours or, alternatively, with 200 nM to 2 mM aqueous copper sulfate (CuSO 4 ) at room temperature overnight.
  • DHAA dehydroascorbic acid
  • CuSO 4 aqueous copper sulfate
  • Other oxidants, i.e. oxidizing agents, and oxidizing conditions, which are known in the art may be used.
  • Ambient air oxidation may also be effective. This mild, partial reoxidation step forms intrachain disulfides efficiently with high fidelity. Reoxidation was monitored by reverse- phase LCMS using a PLRP-S column.
  • the reoxidized THIOMABTM antibody was diluted with succinate buffer as described above to reach pH approximately 5 and purification on an S column was carried out as described above with the exception that elution was performed with a gradient of 10 mM succinate, pH 5, 300 mM sodium chloride (buffer B) in 10 mM succinate, pH 5 (buffer A).
  • Buffer B 300 mM sodium chloride
  • EDTA was added to a final concentration of 2 mM and concentrated, if necessary, to reach a final concentration of more than 5 mg/mL.
  • the resulting THIOMABTM antibody, ready for conjugation, was stored at -20 o C or -80 o C in aliquots. Liquid chromatography/Mass Spectrometric Analysis was performed on a 6200 series TOF or QTOF Agilent LC/MS. Samples were
  • antibodies or conjugates were partially digested with LysC (0.25 ⁇ g per 50 ⁇ g (microgram) antibody or conjugate) for 15 minutes at 37 °C to give a Fab and Fc fragment for analysis by LCMS. Peaks in the deconvoluted LCMS spectra were assigned and quantitated. PROTAC-to-antibody ratios (PAR) were calculated by calculating the ratio of intensities of the peak or peaks corresponding to PROTAC-conjugated antibody relative to all peaks observed.
  • LysC 0.25 ⁇ g per 50 ⁇ g (microgram) antibody or conjugate
  • TIOMABTM antibody in 10 mM succinate, pH 5, 150 mM NaCl, 2 mM EDTA, is pH- adjusted to pH 7.5-8.5 with 1M Tris.
  • An excess, from about 3 molar to 20 equivalents of a linker-PROTAC intermediate with a thiol-reactive group e.g., maleimide or 4-nitropyridy disulfide
  • a linker-PROTAC intermediate with a thiol-reactive group e.g., maleimide or 4-nitropyridy disulfide
  • the reaction is incubated at room temperature or 37 C and monitored until completion (1 to about 24 hours), as determined by LC-MS analysis of the reaction mixture.
  • the conjugate is purified by one or any combination of several methods, the goal being to remove remaining unreacted linker- PROTAC intermediate and aggregated protein (if present at significant levels).
  • the conjugate may be diluted with 10 mM histidine-acetate, pH 5.5 until final pH is approximately 5.5 and purified by S cation exchange chromatography using either HiTrap S columns connected to an Akta purification system (GE Healthcare) or S maxi spin columns (Pierce).
  • the conjugate may be purified by gel filtration chromatography using an S200 column connected to an Akta purification system or Zeba spin columns.
  • dialysis may be used.
  • the THIOMAB TM antibody PROTAC conjugates were formulated into 20 mM His/acetate, pH 5, with 240 mM sucrose using either gel filtration or dialysis.
  • the purified conjugate is concentrated by centrifugal ultrafiltration and filtered through a 0.2- ⁇ m filter under sterile conditions and frozen for storage.
  • the PACs were characterized by BCA assay to determine protein concentration, analytical SEC (size- exclusion chromatography) for aggregation analysis and LC-MS after treatment with Lysine C endopeptidase (LysC) to calculate PAR.
  • Size exclusion chromatography is performed on conjugates using a Shodex KW802.5 column in 0.2M potassium phosphate pH 6.2 with 0.25 mM potassium chloride and 15% IPA at a flow rate of 0.75 ml/min. Aggregation state of the conjugate was determined by integration of eluted peak area absorbance at 280 nm.
  • LC-MS analysis may be performed on PAC using an Agilent QTOF 6520 ESI instrument.
  • the PAR is treated with 1:500 w/w Endoproteinase Lys C (Promega) in Tris, pH 7.5, for 30 min at 37°C.
  • the resulting cleavage fragments are loaded onto a 1000 ⁇ (Angstrom), 8 ⁇ m (micron) PLRP-S (highly cross-linked polystyrene) column heated to 80 °C and eluted with a gradient of 30% B to 40% B in 5 minutes.
  • Mobile phase A was H 2 O with 0.05% TFA and mobile phase B was acetonitrile with 0.04% TFA.
  • the flow rate was 0.5ml/min.
  • compositions of therapeutic PROTAC-antibody-conjugates can be prepared for parenteral administration, e.g., bolus, intravenous, intratumor injection with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form.
  • a PAC having the desired degree of purity is optionally mixed with one or more pharmaceutically acceptable excipients (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation for reconstitution or an aqueous solution.
  • PACs can be formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition. According to this aspect, there is provided a pharmaceutical composition comprising a PAC in association with one or more pharmaceutically acceptable excipients.
  • a typical formulation is prepared by mixing PACs with excipients, such as carriers and/or diluents.
  • excipients such as carriers and/or diluents.
  • Suitable carriers, diluents and other excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like.
  • the particular carrier, diluent or other excipient used will depend upon the means and purpose for which the PAC is being applied.
  • Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
  • GRAS solvents recognized by persons skilled in the art as safe
  • safe solvents are non-toxic aqueous solvents such as water and other non- toxic solvents that are soluble or miscible in water.
  • Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof.
  • 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,
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the PAC or aid in the manufacturing of the pharmaceutical product.
  • the formulations may be prepared using conventional dissolution and mixing procedures.
  • Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed.
  • physiologically acceptable carriers i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed.
  • the pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8.
  • Formulation in an acetate buffer at pH 5 is a suitable embodiment.
  • the PAC formulations can be sterile.
  • formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished by filtration through sterile filtration membranes.
  • the PAC ordinarily can be stored as a solid composition, a lyophilized formulation or as an aqueous solution.
  • compositions comprising a PAC can be formulated, dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • The“therapeutically effective amount” of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the coagulation factor mediated disorder. Such amount is preferably below the amount that is toxic to the host or renders the host significantly more susceptible to bleeding.
  • the PAC can be formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.
  • the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form. Suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
  • the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • the pharmaceutical compositions may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension.
  • a sterile injectable preparation such as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such 1,3-butanediol.
  • the sterile injectable preparation may also be prepared as a lyophilized powder.
  • the acceptable vehicles and solvents that may be employed are water, Ringer’s solution and isotonic sodium chloride solution.
  • sterile fixed oils may conventionally be employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may likewise be used in the preparation of injectables.
  • a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight).
  • the pharmaceutical composition can be prepared to provide easily measurable amounts for administration.
  • an aqueous solution intended for intravenous infusion may contain from about 3 to 500 ⁇ g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
  • 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.
  • the formulations may be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use.
  • sterile liquid carrier for example water
  • Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described.
  • 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.
  • 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 or by any other desired route.
  • PROTAC-antibody conjugates may be used to treat various diseases or disorders.
  • exemplary hyperproliferative disorders include benign or malignant solid tumors and hematological disorders such as leukemia and lymphoid malignancies.
  • Others include neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic and immunologic, including autoimmune, disorders.
  • the disease or disorder to be treated is a hyperproliferative disease such as cancer.
  • cancer to be treated herein include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer,
  • Autoimmune diseases for which the PAC may be used in treatment include rheumatologic disorders (such as, for example, rheumatoid arthritis, Sjögren's syndrome, scleroderma, lupus such as systemic lupus erythematosus (SLE) and lupus nephritis, polymyositis/dermatomyositis, cryoglobulinemia, anti-phospholipid antibody syndrome, and psoriatic arthritis), osteoarthritis, autoimmune gastrointestinal and liver disorders (such as, for example, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), autoimmune gastritis and pernicious anemia, autoimmune hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, and celiac disease), vasculitis (such as, for example, ANCA- associated vasculitis, including Churg-Strauss vasculitis,
  • More preferred such diseases include, for example, rheumatoid arthritis, ulcerative colitis, ANCA-associated vasculitis, lupus, multiple sclerosis, Sjögren's syndrome, Graves’ disease, IDDM, pernicious anemia, thyroiditis, and
  • a PAC comprising an anti-NaPi2b antibody, such as those described above, is used in a method of treating solid tumor, e.g., ovarian.
  • a PAC an anti-CD33 antibody such as those described herein, is used in a method of treating hematological malignancies such as non-Hodgkin's lymphoma (NHL), diffuse large hematopoietic lymphoma, follicular lymphoma, mantle cell lymphoma, chronic lymphocytic leukemia, multiple myeloma, acute myeloid leukemia (AML), and myeloid cell leukemia (MCL), and including B-cell related cancers and proliferative disorders.
  • NHL non-Hodgkin's lymphoma
  • diffuse large hematopoietic lymphoma diffuse large hematopoietic lymphoma
  • follicular lymphoma mantle cell lymphoma
  • a PAC comprising an anti-MUC16 antibody, such as those described herein, is used in a method of treating ovarian, breast and pancreatic cancers.
  • the cancer may be associated with the expression or activity of a MUC16/CA125/O772P polypeptide. See: WO 2007/001851; US 7989595; US 8449883; US 7723485; Chen et al (2007) Cancer Res.67(10): 4924-4932; Junutula, et al., (2008) Nature Biotech., 26(8):925- 932.
  • a PAC comprising an anti-HER2 antibody, such as those described above, is used in a method of treating cancer, e.g., breast or gastric cancer, more specifically HER2+ breast or gastric cancer, wherein the method comprises administering such PAC to a patient in need of such treatment.
  • the PAC comprises the anti-HER2 antibody trastuzumab or pertuzumab.
  • a PAC may be administered by any route appropriate to the condition to be treated.
  • the PAC will typically be administered parenterally, i.e. infusion, subcutaneous,
  • a PAC can be used either alone or in combination with other agents in a therapy.
  • a PAC may be co-administered with at least one additional therapeutic agent.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the PAC can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • a PAC can also be used in combination with radiation therapy.
  • a PAC can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
  • the appropriate dosage of a PAC (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of PAC, the severity and course of the disease, whether the PAC is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the PAC, and the discretion of the attending physician.
  • the PAC is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g.0.1mg/kg-10mg/kg) of a PAC can be an initial candidate dosage for
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of a PAC would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g.
  • kits containing materials useful for the treatment of the diseases and disorders described above.
  • the kit comprises a container comprising a PAC.
  • the kit may further comprise a label or package insert, on or associated with the container.
  • the term“package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • Suitable containers include, for example, bottles, vials, syringes, blister pack, etc.
  • a “vial” is a container suitable for holding a liquid or lyophilized preparation.
  • the vial is a single-use vial, e.g. a 20-cc single-use vial with a stopper.
  • the container may be formed from a variety of materials such as glass or plastic.
  • the container may hold a PAC or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a PAC.
  • the label or package insert indicates that the composition is used for treating the condition of choice, such as cancer.
  • the label or package insert may indicate that the patient to be treated is one having a disorder such as a hyperproliferative disorder, neurodegeneration, cardiac hypertrophy, pain, migraine or a neurotraumatic disease or event.
  • the label or package inserts indicates that the composition comprising a PAC can be used to treat a disorder resulting from abnormal cell growth.
  • the label or package insert may also indicate that the composition can be used to treat other disorders.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer’s solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as phosphate-buffered saline, Ringer’s solution and dextrose solution.
  • the kit may further comprise directions for the administration of the PAC and, if present, the second pharmaceutical formulation.
  • the kit may further comprise directions for the simultaneous, sequential or separate administration of the first and second pharmaceutical compositions to a patient in need thereof.
  • kits are suitable for the delivery of solid oral forms of a PAC, such as tablets or capsules.
  • a kit preferably includes a number of unit dosages.
  • Such kits can include a card having the dosages oriented in the order of their intended use.
  • An example of such a kit is a“blister pack”.
  • Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms.
  • a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered.
  • a kit may comprise (a) a first container with a PAC contained therein; and optionally (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a second compound with anti-hyperproliferative activity.
  • the kit may further comprise a third container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer’s solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet; however, the separate compositions may also be contained within a single, undivided container.
  • the kit comprises directions for the administration of the separate components.
  • the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.

Abstract

The subject matter described herein is directed to antibody-PROTAC conjugates (PACs), to pharmaceutical compositions containing them, and to their use in treating diseases and conditions where targeted protein degradation is beneficial.

Description

PROTAC ANTIBODY CONJUGATES AND METHODS OF USE CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of priority to U.S. provisional Application No. 62/339,257 filed 20 May 2016, the contents of which application is hereby incorporated by reference in its entirety. REFERENCE TO A SEQUENCE LISTING SUBMITTED AS A TEXT FILE VIA EFS-WEB The official copy of the sequence listing is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file named SEQLIST.TXT, created on May 20, 2016, and having a size of 77 kilobytes and is filed concurrently with the specification. The sequence listing contained in this ASCII formatted document is part of the specification and is herein incorporated by reference in its entirety. SEQ ID NOs.1-6 are intentionally omitted. FIELD OF THE INVENTION The subject matter described herein relates generally to antibody-(proteolysis- targeting chimera) (PROTAC) conjugate molecules that are useful for facilitating
intracellular degradation of target proteins. BACKGROUND Cell maintenance and normal function requires controlled degradation of cellular proteins. For example, degradation of regulatory proteins triggers events in the cell cycle, such as DNA replication, chromosome segregation, etc. Accordingly, such degradation of proteins has implications for the cell’s proliferation, differentiation, and death.
While inhibitors of proteins can block or reduce protein activity in a cell, protein degradation in a cell can also reduce activity or remove altogether the target protein.
Utilizing a cell’s protein degradation pathway can, therefore, provide a means for reducing or removing protein activity. One of the cell’s major degradation pathways is known as the ubiquitin-proteasome system. In this system, a protein is marked for degradation by the proteasome by ubiquitinating the protein. The ubiqitinization of the protein is accomplished by an E3 ubiquitin ligase that binds to a protein and adds ubiquitin molecules to the protein. The E3 ubiquitin ligase is part of a pathway that includes E1 and E2 ubiquitin ligases, which make ubiquitin available to the E3 ubiquitin ligase to add to the protein.
To harness this degradation pathway, PROTACs have been developed. PROTACs bring together an E3 ubiquitin ligase with a protein that is to be targeted for degradation. To facilitate a protein for degradation by the proteasome, the PROTAC is comprised of a group that binds to an E3 ubiquitin ligase and a group that binds to the protein one wishes to degrade. These groups are typically connected with a linker. This molecular construct can bring the E3 ubiquitin ligase in proximity with the protein so that it is ubiquitinated and marked for degradation.
There is an ongoing need in the art for enhanced and targeted delivery of PROTACs to cells that contain the protein target. Targeted delivery using antibody-PROTAC conjugates can enhance delivery of PROTACs to particular cells using the specificity of an antibody and can also enhance the pharmacokinetics of delivery of PROTACs to cells relative to other modes of administration of PROTACs, such as infusion.
SUMMARY OF THE INVENTION
In one aspect, the subject matter described herein is directed to a PROTAC-antibody conjugate (PAC) having the formula:
Ab―(L1―D)p,
wherein, D is a PROTAC having the structure E3LB―L2―PB; wherein, E3LB is an E3 ligase binding group covalently bound to L2; L2 is a linker covalently bound to E3LB and PB; PB is a protein binding group covalently bound to L2; Ab is an antibody covalently bound to L1; L1 is a linker, covalently bound to Ab and to D; and p has a value from about 1 to about 8.
Another aspect of the subject matter described herein is a pharmaceutical composition comprising a PAC, and one or more pharmaceutically acceptable excipients.
Another aspect of the subject matter described herein is the use of a PAC in methods of treating conditions and diseases by administering to a subject a pharmaceutical composition comprising a PAC.
Another aspect of the subject matter described herein is a method of making a PAC. Another aspect of the subject matter described herein is an article of manufacture comprising a pharmaceutical composition comprising a PAC, a container, and a package insert or label indicating that the pharmaceutical composition can be used to treat a disease or condition. BRIEF DESCRIPTION OF THE FIGURES Figure 1 shows the detection of ER-α by Western blot for PROTAC (without Ab), compound P1, and PROTAC-antibody conjugates (PACs) PAC1, and PAC2.
Figure 2 depicts the quantitation of ERα as determined by fluorescence intensity for Endox-XIAP PACs treated for 3 days in an engineered HER2-MCF7 line. Media: 10% CS- FBS in phenol red free-RPMI. DETAILED DESCRIPTION Disclosed herein, are antibody-proteolysis targeting chimera conjugates, referred to herein as PROTAC-Antibody conjugates (PACs), that are useful in targeted protein degradation, and the treatment of related diseases and disorders. The subject matter described herein utilizes antibody targeting to direct a PROTAC to a target cell or tissue. As described herein, connecting an antibody to a PROTAC to form a PAC has been shown to deliver the PROTAC to a target cell or tissue. As shown herein, e.g. in Examples 1 and 2, a cell that expresses an antigen can be targeted by an antigen specific PAC, whereby the PROTAC portion of the PAC is delivered intracellularly to the target cell. Also as shown herein, PACs that comprise an antibody directed to an antigen that is not found on the cell do not result in significant intracellualr delivery of the PROTAC to the cell.
Accordingly, the subject matter described herein is directed to PROTAC-antibody conjugate (PAC) compositions that result in the ubiquitination of a target protein and subsequent degradation of the protein. The compositions comprise an antibody covalently linked to a linker (L1), which is covalently linked at any available point of attachment to a PROTAC, in which the PROTAC comprises an E3 ubiquitin ligase binding (E3LB) moiety, wherein the E3LB moiety recognizes a E3 ubiquitin ligase protein and a protein binding moiety (PB) that recognizes a target protein. The subject matter described herein is useful for regulating protein activity, and treating diseases and conditions related to protein activity. The presently disclosed subject matter will now be described more fully hereinafter. However, many modifications and other embodiments of the presently disclosed subject matter set forth herein will come to mind to one skilled in the art to which the presently disclosed subject matter pertains having the benefit of the teachings presented in the foregoing descriptions. Therefore, it is to be understood that the presently disclosed subject matter is not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. In other words, the subject matter described herein covers all alternatives, modifications, and equivalents. In the event that one or more of the incorporated literature, patents, and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in this field. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.
I. Definitions
The term“PROTAC” refers to proteolysis-targeting chimera molecules having generally three components, an E3 ubiquitin ligase binding group (E3LB), a linker L2, and a protein binding group (PB).
The terms“residue,”“moiety” or“group” refers to a component that is covalently bound or linked to another component. For example a“residue of a PROTAC” refers to a PROTAC that is covalently linked to one or more groups such as a Linker L2, which itself can be optionally further linked to an antibody.
The term“covalently bound” or“covalently linked” refers to a chemical bond formed by sharing of one or more pairs of electrons.
The term“peptidomimetic” or PM as used herein means a non-peptide chemical moiety. Peptides are short chains of amino acid monomers linked by peptide (amide) bonds, the covalent chemical bonds formed when the carboxyl group of one amino acid reacts with the amino group of another. The shortest peptides are dipeptides, consisting of 2 amino acids joined by a single peptide bond, followed by tripeptides, tetrapeptides, etc. A peptidomimetic chemical moiety includes non-amino acid chemical moieties. A peptidomimetic chemical moiety may also include one or more amino acid that are separated by one or more non- amino acid chemical units. A peptidomimetic chemical moiety does not contain in any portion of its chemical structure two or more adjacent amino acids that are linked by peptide bonds.
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 (complementary determining regions) on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody. An antibody includes a full-length immunoglobulin molecule or an immunologically active portion of a full-length immunoglobulin molecule, i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease. The immunoglobulin disclosed herein can be of any type (e.g., IgG, IgE, IgM, IgD, and IgA), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule. The immunoglobulins can be derived from any species. In one aspect, however, the immunoglobulin is of human, murine, or rabbit origin.
The term "antibody fragment(s)" as used herein comprises 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 subject matter described herein 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.
The term“chimeric” antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
The“class” of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called α, δ, ε, γ, and µ, respectively.
The term“intact antibody” as used herein is one comprising a VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1, CH2 and CH3. The constant domains may be native sequence constant domains (e.g., human native sequence constant domains) or amino acid sequence variant thereof. The intact antibody may have one or more“effector functions” which refer to those biological activities attributable to the Fc constant region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell- mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.
The term“Fc region” as used hererin means a C-terminal region of an
immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. In one embodiment, a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
The term“framework” or“FR” as used herein refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FR1-H1(L1)-FR2- H2(L2)-FR3-H3(L3)-FR4.
The terms“full length antibody,”“intact antibody,” and“whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
A“human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody- encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
A“humanized” antibody refers to a chimeric antibody comprising amino acid residues from non-human HVRs and amino acid residues from human FRs. In certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody. A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A“humanized form” of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization.
An“isolated antibody” is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC). For review of methods for assessment of antibody purity, see, e.g., Flatman et al., J. Chromatogr. B 848:79-87 (2007).
An“isolated nucleic acid” refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
“Isolated nucleic acid encoding an antibody” refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
A“naked antibody” refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel. The naked antibody may be present in a pharmaceutical formulation.
“Native antibodies” refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3). Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain.
“Percent (%) amino acid sequence identity” with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office,
Washington D.C., 20559, where it is registered under U.S. Copyright Registration No.
TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program’s alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program.
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., IgG1, 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).
The term“human consensus framework” as used herein refers to a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences. Generally, the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences. Generally, the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NIH Publication 91-3242, Bethesda MD (1991), vols.1-3. In one embodiment, for the VL, the subgroup is subgroup kappa I as in Kabat et al., supra. In one embodiment, for the VH, the subgroup is subgroup III as in Kabat et al., supra.
An“acceptor human framework” for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below. An acceptor human framework“derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
The term“variable region” or“variable domain” as used herein refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). (See, e.g., Kindt et al. Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007).) A single VH or VL domain may be sufficient to confer antigen-binding specificity. Furthermore, antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J. Immunol.150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
The term“hypervariable region” or“HVR,” as used herein, refers to each of the regions of an antibody variable domain that are hypervariable in sequence and/or form structurally defined loops (“hypervariable loops”). Generally, native four-chain antibodies comprise six HVRs; three in the VH (H1, H2, H3), and three in the VL (L1, L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the “complementarity determining regions” (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. Exemplary hypervariable loops occur at amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3). (Chothia and Lesk, J. Mol. Biol.196:901-917 (1987).) Exemplary CDRs (CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3) occur at amino acid residues 24-34 of L1, 50-56 of L2, 89-97 of L3, 31-35B of H1, 50-65 of H2, and 95-102 of H3. (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991).) With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. CDRs also comprise“specificity determining residues,” or“SDRs,” which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated-CDRs, or a- CDRs. Exemplary a-CDRs (a-CDR-L1, a-CDR-L2, a-CDR-L3, a-CDR-H1, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31-34 of L1, 50-55 of L2, 89-96 of L3, 31-35B of H1, 50-58 of H2, and 95-102 of H3. (See Almagro and Fransson, Front. Biosci.13:1619- 1633 (2008).) Unless otherwise indicated, HVR residues and other residues in the variable domain (e.g., FR residues) are numbered herein according to Kabat et al., supra.
“Effector functions” refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: C1q binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
The term“epitope” refers to the particular site on an antigen molecule to which an antibody binds.
The“epitope 4D5” or“4D5 epitope” or“4D5” is the region in the extracellular domain of HER2 to which the antibody 4D5 (ATCC CRL 10463) and trastuzumab bind. This epitope is close to the transmembrane domain of HER2, and within domain IV of HER2. To screen for antibodies which bind to the 4D5 epitope, a routine cross-blocking assay such as that described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988), can be performed. Alternatively, epitope mapping can be performed to assess whether the antibody binds to the 4D5 epitope of HER2 (e.g. any one or more residues in the region from about residue 550 to about residue 610, inclusive, of HER2 (SEQ ID NO: 39).
The“epitope 2C4” or“2C4 epitope” is the region in the extracellular domain of HER2 to which the antibody 2C4 binds. In order to screen for antibodies which bind to the 2C4 epitope, a routine cross-blocking assay such as that described in Antibodies, A
Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988), can be performed. Alternatively, epitope mapping can be performed to assess whether the antibody binds to the 2C4 epitope of HER2. Epitope 2C4 comprises residues from domain II in the extracellular domain of HER2. The 2C4 antibody and pertuzumab bind to the extracellular domain of HER2 at the junction of domains I, II and III (Franklin et al. Cancer Cell 5:317-328 (2004)).
“Affinity” refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein,“binding affinity” refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following. In certain embodiments, an antibody as described herein has dissociation constant (Kd) of≤ 1µM,≤ 100 nM,≤ 10 nM, ≤ 5 nm,≤ 4 nM,≤ 3 nM,≤ 2 nM,≤ 1 nM,≤ 0.1 nM,≤ 0.01 nM, or≤ 0.001 nM (e.g., 10-8 M or less, e.g. from 10-8 M to 10-13 M, e.g., from 10-9 M to 10-13 M).
An“affinity matured” antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
The term“vector” as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a self- replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as “expression vectors.”
The term“free cysteine amino acid” as used herein 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“amino acid” as used herein means glycine, alanine, valine, leucine, isoleucine, phenylalanine, proline, serine, threonine, tyrosine, cysteine, methionine, lysine, arginine, histidine, tryptophan, aspartic acid, glutamic acid, asparagine, glutamine or citrulline.
The term“Linker”,“Linker Unit”, or“link” as used herein means a chemical moiety comprising a chain of atoms that covalently attaches a PROTAC moiety to an antibody, or a component of a PROTAC to another component of the PROTAC. In various embodiments, a linker is a divalent radical, specified as L1 or L2.
A“patient” or“individual” or“subject” is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the patient, individual, or subject is a human. In some embodiments, the patient may be a“cancer patient,” i.e. one who is suffering or at risk for suffering from one or more symptoms of cancer. A“patient population” refers to a group of cancer patients. Such populations can be used to demonstrate statistically significant efficacy and/or safety of a drug.
A“relapsed” patient is one who has signs or symptoms of cancer after remission. Optionally, the patient has relapsed after adjuvant or neoadjuvant therapy.
A cancer or biological sample which“displays HER expression, amplification, or activation” is one which, in a diagnostic test, expresses (including overexpresses) a HER receptor, has amplified HER gene, and/or otherwise demonstrates activation or
phosphorylation of a HER receptor.
“Neoadjuvant therapy” or“preoperative therapy” herein refers to therapy given prior to surgery. The goal of neoadjuvant therapy is to provide immediate systemic treatment, potentially eradicating micrometastases that would otherwise proliferate if the standard sequence of surgery followed by systemic therapy were followed. Neoadjuvant therapy may also help to reduce tumor size thereby allowing complete resection of initially unresectable tumors or preserving portions of the organ and its functions. Furthermore, neoadjuvant therapy permits an in vivo assessment of drug efficacy, which may guide the choice of subsequent treatments.
“Adjuvant therapy” herein refers to therapy given after definitive surgery, where no evidence of residual disease can be detected, so as to reduce the risk of disease recurrence. The goal of adjuvant therapy is to prevent recurrence of the cancer, and therefore to reduce the chance of cancer-related death. Adjuvant therapy herein specifically excludes
neoadjuvant therapy.
“Definitive surgery” is used as that term is used within the medical community.
Definitive surgery includes, for example, procedures, surgical or otherwise, that result in removal or resection of the tumor, including those that result in the removal or resection of all grossly visible tumor. Definitive surgery includes, for example, complete or curative resection or complete gross resection of the tumor. Definitive surgery includes procedures that occur in one or more stages, and includes, for example, multi-stage surgical procedures where one or more surgical or other procedures are performed prior to resection of the tumor. Definitive surgery includes procedures to remove or resect the tumor including involved organs, parts of organs and tissues, as well as surrounding organs, such as lymph nodes, parts of organs, or tissues. Removal may be incomplete such that tumor cells might remain even though undetected. “Survival” refers to the patient remaining alive, and includes disease free survival (DFS), progression free survival (PFS) and overall survival (OS). Survival can be estimated by the Kaplan-Meier method, and any differences in survival are computed using the stratified log-rank test.
“Progression-Free Survival” (PFS) is the time from the first day of treatment to documented disease progression (including isolated CNS progression) or death from any cause on study, whichever occurs first.
“Disease free survival (DFS)” refers to the patient remaining alive, without return of the cancer, for a defined period of time such as about 1 year, about 2 years, about 3 years, about 4 years, about 5 years, about 10 years, etc., from initiation of treatment or from initial diagnosis. In one aspect of the subject matter described herein, DFS is analyzed according to the intent-to-treat principle, i.e., patients are evaluated on the basis of their assigned therapy. The events used in the analysis of DFS can include local, regional and distant recurrence of cancer, occurrence of secondary cancer, and death from any cause in patients without a prior event (e.g, breast cancer recurrence or second primary cancer).
“Overall survival” refers to the patient remaining alive for a defined period of time, such as about 1 year, about 2 years, about 3 years, about 4 years, about 5 years, about 10 years, etc., from initiation of treatment or from initial diagnosis.
By“extending survival” is meant increasing DFS and/or OS in a treated patient relative to an untreated patient, or relative to a control treatment protocol. Survival is monitored for at least about six months, or at least about 1 year, or at least about 2 years, or at least about 3 years, or at least about 4 years, or at least about 5 years, or at least about 10 years, etc., following the initiation of treatment or following the initial diagnosis.
By“monotherapy” is meant a therapeutic regimen that includes only a single therapeutic agent for the treatment of the cancer or tumor during the course of the treatment period.
By“maintenance therapy” is meant a therapeutic regimen that is given to reduce the likelihood of disease recurrence or progression. Maintenance therapy can be provided for any length of time, including extended time periods up to the life-span of the subject.
Maintenance therapy can be provided after initial therapy or in conjunction with initial or additional therapies. Dosages used for maintenance therapy can vary and can include diminished dosages as compared to dosages used for other types of therapy. The terms“host cell,”“host cell line,” and“host cell culture” are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include“transformants” and“transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
The terms“cancer” and“cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth/proliferation. A“tumor” comprises one or more cancerous cells. Examples of cancer are provided elsewhere herein.
A“HER2-positive” cancer comprises cancer cells which have higher than normal levels of HER2. Examples of HER2-positive cancer include HER2-positive breast cancer and HER2-positive gastric cancer. Optionally, HER2-positive cancer has an
immunohistochemistry (IHC) score of 2+ or 3+ and/or an in situ hybridization (ISH) amplification ratio≥2.0. The term“HER2-positive cell” refers to a cell that expresses HER2 on its surface.
The term“early stage breast cancer (EBC)” or“early breast cancer” is used herein to refer to breast cancer that has not spread beyond the breast or the axillary lymph nodes. This includes ductal carcinoma in situ and stage I, stage IIA, stage IIB, and stage IIIA breast cancers.
Reference to a tumor or cancer as a“Stage 0,”“Stage I,”“Stage II,”“Stage III,” or “Stage IV”, and various sub-stages within this classification, indicates classification of the tumor or cancer using the Overall Stage Grouping or Roman Numeral Staging methods known in the art. Although the actual stage of the cancer is dependent on the type of cancer, in general, a Stage 0 cancer is an in situ lesion, a Stage I cancer is small localized tumor, a Stage II and III cancer is a local advanced tumor which exhibits involvement of the local lymph nodes, and a Stage IV cancer represents metastatic cancer. The specific stages for each type of tumor are known to the skilled clinician.
The term“metastatic breast cancer” means the state of breast cancer where the cancer cells are transmitted from the original site to one or more sites elsewhere in the body, by the blood vessels or lymphatics, to form one or more secondary tumors in one or more organs besides the breast.
An“advanced” cancer is one which has spread outside the site or organ of origin, either by local invasion or metastasis. Accordingly, the term“advanced” cancer includes both locally advanced and metastatic disease. A“recurrent” cancer is one which has regrown, either at the initial site or at a distant site, after a response to initial therapy, such as surgery. A“locally recurrent” cancer is cancer that returns after treatment in the same place as a previously treated cancer. An“operable” or“resectable” cancer is cancer which is confined to the primary organ and suitable for surgery (resection). A“non-resectable” or “unresectable” cancer is not able to be removed (resected) by surgery.
The term“cytotoxic agent” as used herein refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At211, I131, I125, Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below.
A "chemotherapeutic agent" refers to a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa;
ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine;
acetogenins (especially bullatacin and bullatacinone); delta-9-tetrahydrocannabinol
(dronabinol, MARINOL®); beta-lapachone; lapachol; colchicines; betulinic acid; a camptothecin (including the synthetic analogue topotecan (HYCAMTIN®), CPT-11
(irinotecan, CAMPTOSAR®), acetylcamptothecin, scopolectin, and 9-aminocamptothecin); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); podophyllotoxin; podophyllinic acid; teniposide; cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin;
spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard;
nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gamma1I and calicheamicin omegaI1 (see, e.g., Nicolaou et al., Angew. Chem Intl. Ed. Engl., 33: 183-186 (1994)); CDP323, an oral alpha-4 integrin inhibitor; dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including ADRIAMYCIN®, morpholino-doxorubicin, cyanomorpholino- doxorubicin, 2-pyrrolino-doxorubicin, doxorubicin HCl liposome injection (DOXIL®), liposomal doxorubicin TLC D-99 (MYOCET®), peglylated liposomal doxorubicin
(CAELYX®), and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate, gemcitabine (GEMZAR®), tegafur (UFTORAL®), capecitabine (XELODA®), an epothilone, and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti- adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil;
amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone;
elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; 2- ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 22’2’trichlorotriethylamine; trichothecenes (especially T 2 toxin verracurin A roridin A and anguidine); urethan; vindesine (ELDISINE®, FILDESIN®); dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); thiotepa; taxoid, e.g., paclitaxel (TAXOL®), albumin-engineered nanoparticle formulation of paclitaxel (ABRAXANETM), and docetaxel (TAXOTERE®); chloranbucil; 6-thioguanine; mercaptopurine; methotrexate; platinum agents such as cisplatin, oxaliplatin (e.g.,
ELOXATIN®), and carboplatin; vincas, which prevent tubulin polymerization from forming microtubules, including vinblastine (VELBAN®), vincristine (ONCOVIN®), vindesine (ELDISINE®, FILDESIN®), and vinorelbine (NAVELBINE®); etoposide (VP-16);
ifosfamide; mitoxantrone; leucovorin; novantrone; edatrexate; daunomycin; aminopterin; ibandronate; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid, including bexarotene (TARGRETIN®); bisphosphonates such as clodronate (for example, BONEFOS® or OSTAC®), etidronate (DIDROCAL®), NE-58095, zoledronic acid/zoledronate (ZOMETA®), alendronate (FOSAMAX®), pamidronate (AREDIA®), tiludronate (SKELID®), or risedronate (ACTONEL®); troxacitabine (a 1,3- dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those that inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Raf, H-Ras, and epidermal growth factor receptor (EGF- R); vaccines such as THERATOPE® vaccine and gene therapy vaccines, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and VAXID® vaccine; topoisomerase 1 inhibitor (e.g., LURTOTECAN®); rmRH (e.g., ABARELIX®); BAY439006 (sorafenib; Bayer); SU-11248 (sunitinib, SUTENT®, Pfizer); perifosine, COX-2 inhibitor (e.g., celecoxib or etoricoxib), proteosome inhibitor (e.g., PS341); bortezomib (VELCADE®); CCI-779; tipifarnib (R11577); orafenib, ABT510; Bcl-2 inhibitor such as oblimersen sodium (GENASENSE®, an antisence oligonucleotide); pixantrone; EGFR inhibitors (see definition below); tyrosine kinase inhibitors; serine-threonine kinase inhibitors such as rapamycin (sirolimus, RAPAMUNE®); farnesyltransferase inhibitors such as lonafarnib (SCH 6636, SARASARTM); and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone; and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATINTM) combined with 5-FU and leucovorin.
Chemotherapeutic agents as defined herein include“anti-hormonal agents” or “endocrine therapeutics” which act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer They may be hormones themselves including, but not limited to: anti-estrogens with mixed agonist/antagonist profile, including, tamoxifen (NOLVADEX®), 4-hydroxytamoxifen, toremifene (FARESTON®), idoxifene, droloxifene, raloxifene (EVISTA®), trioxifene, keoxifene, and selective estrogen receptor modulators (SERMs) such as SERM3; pure anti-estrogens without agonist properties, such as fulvestrant (FASLODEX®), and EM800 (such agents may block estrogen receptor (ER) dimerization, inhibit DNA binding, increase ER turnover, and/or suppress ER levels);
aromatase inhibitors, including steroidal aromatase inhibitors such as formestane and exemestane (AROMASIN®), and nonsteroidal aromatase inhibitors such as anastrazole (ARIMIDEX®), letrozole (FEMARA®) and aminoglutethimide, and other aromatase inhibitors include vorozole (RIVISOR®), megestrol acetate (MEGASE®), fadrozole, and 4(5)-imidazoles; lutenizing hormone-releaseing hormone agonists, including leuprolide (LUPRON® and ELIGARD®), goserelin, buserelin, and tripterelin; sex steroids, including progestines such as megestrol acetate and medroxyprogesterone acetate, estrogens such as diethylstilbestrol and premarin, and androgens/retinoids such as fluoxymesterone, all transretionic acid and fenretinide; onapristone; anti-progesterones; estrogen receptor down- regulators (ERDs); anti-androgens such as flutamide, nilutamide and bicalutamide; and pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above.
The term "immunosuppressive agent" as used herein for adjunct therapy refers to substances that act to suppress or mask the immune system of the mammal being treated herein. This would include substances that suppress cytokine production, down-regulate or suppress self-antigen expression, or mask the MHC antigens. Examples of such agents include 2-amino-6-aryl-5-substituted pyrimidines (see U.S. Pat. No.4,665,077); non-steroidal anti-inflammatory drugs (NSAIDs); ganciclovir, tacrolimus, glucocorticoids such as cortisol or aldosterone, anti-inflammatory agents such as a cyclooxygenase inhibitor, a 5- lipoxygenase inhibitor, or a leukotriene receptor antagonist; purine antagonists such as azathioprine or mycophenolate mofetil (MMF); alkylating agents such as cyclophosphamide; bromocryptine; danazol; dapsone; glutaraldehyde (which masks the MHC antigens, as described in U.S. Pat. No.4,120,649); anti-idiotypic antibodies for MHC antigens and MHC fragments; cyclosporin A; steroids such as corticosteroids or glucocorticosteroids or glucocorticoid analogs, e.g., prednisone, methylprednisolone, including SOLU-MEDROL® methylprednisolone sodium succinate, and dexamethasone; dihydrofolate reductase inhibitors such as methotrexate (oral or subcutaneous); anti-malarial agents such as chloroquine and hydroxychloroquine; sulfasalazine; leflunomide; cytokine or cytokine receptor antibodies including anti-interferon-alpha, -beta, or -gamma antibodies, anti-tumor necrosis factor(TNF)-alpha antibodies (infliximab (REMICADE®) or adalimumab), anti-TNF-alpha immunoadhesin (etanercept), anti-TNF-beta antibodies, anti-interleukin-2 (IL-2) antibodies and anti-IL-2 receptor antibodies, and anti-interleukin-6 (IL-6) receptor antibodies and antagonists (such as ACTEMRATM (tocilizumab)); anti-LFA-1 antibodies, including anti- CD11a and anti-CD18 antibodies; anti-L3T4 antibodies; heterologous anti-lymphocyte globulin; pan-T antibodies, preferably anti-CD3 or anti-CD4/CD4a antibodies; soluble peptide containing a LFA-3 binding domain (WO 90/08187 published 7/26/90);
streptokinase; transforming growth factor-beta (TGF-beta); streptodornase; RNA or DNA from the host; FK506; RS-61443; , chlorambucil; deoxyspergualin; rapamycin; T-cell receptor (Cohen et al., U.S. Pat. No.5,114,721); T-cell receptor fragments (Offner et al., Science, 251: 430-432 (1991); WO 90/11294; Ianeway, Nature, 341: 482 (1989); and WO 91/01133); BAFF antagonists such as BAFF antibodies and BR3 antibodies and zTNF4 antagonists (for review, see Mackay and Mackay, Trends Immunol., 23:113-5 (2002) and see also definition below); biologic agents that interfere with T cell helper signals, such as anti- CD40 receptor or anti-CD40 ligand (CD154), including blocking antibodies to CD40-CD40 ligand (e.g., Durie et al., Science, 261: 1328-30 (1993); Mohan et al., J. Immunol., 154: 1470- 80 (1995)) and CTLA4-Ig (Finck et al., Science, 265: 1225-7 (1994)); and T-cell receptor antibodies (EP 340,109) such as T10B9. Some preferred immunosuppressive agents herein include cyclophosphamide, chlorambucil, azathioprine, leflunomide, MMF, or methotrexate.
As used herein,“treatment” (and grammatical variations thereof such as“treat” or “treating”) refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, antibodies of the subject matter described herein are used to delay development of a disease or to slow the progression of a disease.
A drug that is administered“concurrently” with one or more other drugs is
administered during the same treatment cycle, on the same day of treatment as the one or more other drugs, and, optionally, at the same time as the one or more other drugs. For instance, for cancer therapies given every 3 weeks, the concurrently administered drugs are each administered on day-1 of a 3-week cycle.
An“effective amount” of an agent, e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. For example, an effective amount of the drug for treating cancer may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. The effective amount may extend progression free survival (e.g. as measured by Response Evaluation Criteria for Solid Tumors, RECIST, or CA-125 changes), result in an objective response (including a partial response, PR, or complete response, CR), increase overall survival time, and/or improve one or more symptoms of cancer (e.g. as assessed by FOSI).
As used herein, the term“therapeutically effective amount” means any amount which, as compared to a corresponding subject who has not received such amount, results in treatment of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function. For use in therapy, therapeutically effective amounts of a PAC, as well as salts thereof, may be administered as the raw chemical. Additionally, the active ingredient may be presented as a pharmaceutical composition.
As used herein, unless defined otherwise in a claim, the term“optionally” means that the subsequently described event(s) may or may not occur, and includes both event(s) that occur and event(s) that do not occur.
As used herein, unless defined otherwise, the phrase“optionally substituted”, “substituted” or variations thereof denote an optional substitution, including multiple degrees of substitution, with one or more substituent group, for example, one, two or three. The phrase should not be interpreted as duplicative of the substitutions herein described and depicted.
The term“pharmaceutical formulation” refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
A“pharmaceutically acceptable excipient” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A
pharmaceutically acceptable excipient includes, but is not limited to, a buffer, carrier, stabilizer, or preservative.
The phrase“pharmaceutically acceptable salt,” as used herein, refers to
pharmaceutically acceptable organic or inorganic salts of a molecule. Exemplary salts include, but are not limited, to sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate,
methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1,1’-methylene-bis -(2-hydroxy-3- 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.
Other salts, which are not pharmaceutically acceptable, may be useful in the preparation of compounds of described herein and these should be considered to form a further aspect of the subject matter. These salts, such as oxalic or trifluoroacetate, while not in themselves pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining the compounds described herein and their pharmaceutically acceptable salts.
As used herein, the term“plurality” refers to two or more conjugates. Each conjugate can be the same or different from any other conjugate in the plurality.
A“small molecule” or“small molecular compound” generally refers to an organic molecule that is less than about 5 kilodaltons (Kd) in size. In some embodiments, the small molecule is less than about 4 Kd, 3 Kd, about 2 Kd, or about 1 Kd. In some embodiments, the small molecule is less than about 800 daltons (D), about 600 D, about 500 D, about 400 D, about 300 D, about 200 D, or about 100 D. In some embodiments, a small molecule is less than about 2000 g/mol, less than about 1500 g/mol, less than about 1000 g/mol, less than about 800 g/mol, or less than about 500 g/mol. In some embodiments, small molecules are non- polymeric. Small molecules are not proteins, polypeptides, oligopeptides, peptides, polynucleotides, oligonucleotides, polysaccharides, glycoproteins, proteoglycans, etc. A derivative of a small molecule refers to a molecule that shares the same structural core as the original small molecule, but which can be prepared by a series of chemical reactions from the original small molecule.
The term“alkyl” as used herein refers to a saturated linear or branched-chain monovalent hydrocarbon radical of any length from one to twelve carbon atoms (C1−C12), wherein the alkyl radical may be optionally substituted independently with one or more substituents described below. In another embodiment, an alkyl radical is one to eight carbon atoms (C1−C8), or one to six carbon atoms (C1−C6). Examples of alkyl groups include, but are not limited to, methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1-propyl (n-Pr, n-propyl, - CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH(CH3)2), 1-butyl (n-Bu, n-butyl, - CH2CH2CH2CH3), 2-methyl-1-propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, - CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n-pentyl, - CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2- methyl-2-butyl (-C(CH3)2CH2CH3), 3-methyl-2-butyl (-CH(CH3)CH(CH3)2), 3-methyl-1- butyl (-CH2CH2CH(CH3)2), 2-methyl-1-butyl (-CH2CH(CH3)CH2CH3), 1-hexyl (- CH2CH2CH2CH2CH2CH3), 2-hexyl (-CH(CH3)CH2CH2CH2CH3), 3-hexyl (- CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (-C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (-CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (-CH(CH3)CH2CH(CH3)2), 3-methyl-3- pentyl (-C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (-CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2- butyl (-C(CH3)2CH(CH3)2), 3,3-dimethyl-2-butyl (-CH(CH3)C(CH3)3, 1-heptyl, 1-octyl, and the like.
The term“alkylene” as used herein refers to a saturated linear or branched-chain divalent hydrocarbon radical of any length from one to twelve carbon atoms (C1−C12), wherein the alkylene radical may be optionally substituted independently with one or more substituents described below. In another embodiment, an alkylene radical is one to eight carbon atoms (C1−C8), or one to six carbon atoms (C1−C6). Examples of alkylene groups include, but are not limited to, methylene (-CH2-), ethylene (−CH2CH2−), propylene (− CH2CH2CH2−), and the like.
The term“alkenyl” refers to linear or branched-chain monovalent hydrocarbon radical of any length from two to eight carbon atoms (C2−C8) with at least one site of unsaturation, i.e., a carbon-carbon, sp2 double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having“cis” and“trans” orientations, or alternatively,“E” and“Z” orientations. Examples include, but are not limited to, ethylenyl or vinyl (−CH=CH2), allyl (−
CH2CH=CH2), and the like.
The term“alkenylene” refers to linear or branched-chain divalent hydrocarbon radical of any length from two to eight carbon atoms (C2−C8) with at least one site of unsaturation, i.e., a carbon-carbon, sp2 double bond, wherein the alkenylene radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having“cis” and“trans” orientations, or alternatively,“E” and“Z” orientations. Examples include, but are not limited to, ethylenylene or vinylene (−CH=CH−), allyl (− CH2CH=CH−), and the like.
The term“alkynyl” refers to a linear or branched monovalent hydrocarbon radical of any length from two to eight carbon atoms (C2−C8) with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, ethynyl (-C≡CH), propynyl (propargyl, -CH2C≡CH), and the like.
The term“alkynylene” refers to a linear or branched divalent hydrocarbon radical of any length from two to eight carbon atoms (C2−C8) with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynylene radical may be optionally substituted independently with one or more substituents described herein. Examples include, but are not limited to, ethynylene (-C≡C-), propynylene (propargylene, -CH2C≡C-), and the like.
The terms“carbocycle”,“carbocyclyl”,“carbocyclic ring” and“cycloalkyl” refer to a monovalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms (C3−C12) as a monocyclic ring or 7 to 12 carbon atoms as a bicyclic ring. Bicyclic carbocycles having 7 to 12 atoms can be arranged, for example, as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, and bicyclic carbocycles having 9 or 10 ring atoms can be arranged as a bicyclo [5,6] or [6,6] system, or as bridged systems such as bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane. Spiro moieties are also included within the scope of this definition. Examples of monocyclic carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3- enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like. Carbocyclyl groups are optionally substituted independently with one or more substituents described herein.
“Aryl” means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms (C6− C20) derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Some aryl groups are represented in the exemplary structures as“Ar”. Aryl includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic ring. Typical aryl groups include, but are not limited to, radicals derived from benzene (phenyl), substituted benzenes, naphthalene, anthracene, biphenyl, indenyl, indanyl, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthyl, and the like. Aryl groups are optionally substituted independently with one or more substituents described herein.
“Arylene” means a divalent aromatic hydrocarbon radical of 6-20 carbon atoms (C6− C20) derived by the removal of two hydrogen atom from a two carbon atoms of a parent aromatic ring system. Some arylene groups are represented in the exemplary structures as “Ar”. Arylene includes bicyclic radicals comprising an aromatic ring fused to a saturated, partially unsaturated ring, or aromatic carbocyclic ring. Typical arylene groups include, but are not limited to, radicals derived from benzene (phenylene), substituted benzenes, naphthalene, anthracene, biphenylene, indenylene, indanylene, 1,2-dihydronaphthalene, 1,2,3,4-tetrahydronaphthyl, and the like. Arylene groups are optionally substituted with one or more substituents described herein.
The terms“heterocycle,”“heterocyclyl” and“heterocyclic ring” are used
interchangeably herein and refer to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) carbocyclic radical of 3 to about 20 ring atoms in which at least one ring atom is a heteroatom selected from nitrogen, oxygen, phosphorus and sulfur, the remaining ring atoms being C, where one or more ring atoms is optionally substituted independently with one or more substituents described below. A heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 4 heteroatoms selected from N, O, P, and S) or a bicycle having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 6 heteroatoms selected from N, O, P, and S), for example: a bicyclo [4,5], [5,5], [5,6], or [6,6] system. Heterocycles are described in Paquette, Leo A.;
“Principles of Modern Heterocyclic Chemistry” (W.A. Benjamin, New York, 1968), particularly Chapters 1, 3, 4, 6, 7, and 9;“The Chemistry of Heterocyclic Compounds, A series of Monographs” (John Wiley & Sons, New York, 1950 to present), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566.“Heterocyclyl” also includes radicals where heterocycle radicals are fused with a saturated, partially unsaturated ring, or aromatic carbocyclic or heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, morpholin-4-yl, piperidin-1-yl, piperazinyl, piperazin-4-yl-2-one, piperazin-4-yl-3-one, pyrrolidin-1-yl, thiomorpholin-4-yl, S-dioxothiomorpholin-4-yl, azocan-1-yl, azetidin-1-yl, octahydropyrido[1,2-a]pyrazin-2-yl, [1,4]diazepan-1-yl, pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H- pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl,
dihydropyranyl, dihydrothienyl, dihydrofuranyl, pyrazolidinylimidazolinyl, imidazolidinyl, 3- azabicyco[3.1.0]hexanyl, 3-azabicyclo[4.1.0]heptanyl, azabicyclo[2.2.2]hexanyl, 3H-indolyl quinolizinyl and N-pyridyl ureas. Spiro moieties are also included within the scope of this definition. Examples of a heterocyclic group wherein 2 ring atoms are substituted with oxo (=O) moieties are pyrimidinonyl and 1,1-dioxo-thiomorpholinyl. The heterocycle groups herein are optionally substituted independently with one or more substituents described herein.
The term“heteroaryl” refers to a monovalent aromatic radical of 5-, 6-, or 7- membered rings, and includes fused ring systems (at least one of which is aromatic) of 5-20 atoms, containing one or more heteroatoms independently selected from nitrogen, oxygen, and sulfur. Examples of heteroaryl groups are pyridinyl (including, for example, 2- hydroxypyridinyl), imidazolyl, imidazopyridinyl, 1-methyl-1H-benzo[d]imidazole,
[1,2,4]triazolo[1,5-a]pyridine, pyrimidinyl (including, for example, 4-hydroxypyrimidinyl), pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxadiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl. Heteroaryl groups are optionally substituted
independently with one or more substituents described herein.
The heterocycle or heteroaryl groups may be carbon (carbon-linked), or nitrogen (nitrogen-linked) bonded where such is possible. By way of example and not limitation, carbon bonded heterocycles or heteroaryls are bonded at position 2, 3, 4, 5, or 6 of a pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2, 3, or 4 of an azetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline.
By way of example and not limitation, nitrogen bonded heterocycles or heteroaryls are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3- pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2- pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, 1H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β- carboline.
The term“chiral” refers to molecules which have the property of non- superimposability of the mirror image partner, while the term“achiral” refers to molecules which are superimposable on their mirror image partner.
The term“stereoisomers” refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
“Diastereomer” refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
“Enantiomers” refer to two stereoisomers of a compound which are non- superimposable mirror images of one another. 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 l 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.
Other terms, definitions and abbreviations herein include: Wild-type ("WT");
Cysteine engineered mutant antibody ("thio"); light chain ("LC"); heavy chain ("HC"); 6- maleimidocaproyl (“MC”); maleimidopropanoyl (“MP”); valine-citrulline (“val-cit” or“vc”), alanine-phenylalanine (“ala-phe”), p-aminobenzyl (“PAB”), and p-aminobenzyloxycarbonyl (“PABC”); A118C (EU numbering) = A121C (Sequential numbering) = A114C (Kabat numbering) of heavy chain K149C (Kabat numbering) of light chain. Still additional definitions and abbreviations are provided elsehwere herein.
II. PROTAC-Antibody Conjugate (PAC)
The PROTAC-Antibody Conjugate (PAC) molecules described herein comprise an antibody conjugated via a linker (L1) to a PROTAC, wherein the PROTAC comprises a ubiquitin E3 ligase binding groug (“E3LB”), a linker (“L2”) and a protein binding group (“PB”). The general formula of a PAC is:
Ab―(L1―D)p,
wherein, D is PROTAC having the structure E3LB―L2―PB; wherein, E3LB is an E3 ligase binding group covalently bound to L2; L2 is a linker covalently bound to E3LB and PB; PB is a protein binding group covalently bound to L2; Ab is an antibody covalently bound to L1; L1 is a linker, covalently bound to Ab and to D; and p has a value from about 1 to about 50. The variable p reflects that an antibody can be connected to one or more L1-D groups. In one embodiment, p is from about 1 to 8. In another embodiment, p is about 2.
The following sections describe the components that comprise the PAC. To obtain a PAC having potent efficacy and a desirable therapeutic index, the following components are provided.
1. Antibody (Ab)
As described herein, antibodies, e.g., a monoclonal antibodies (mABs) are used to deliver a PROTAC to target cells, e.g., cells that express the specific protein that is targeted by the antibody. The antibody portion of a PAC can target a cell that expresses an antigen whereby the antigen specific PAC is delivered intracellularly to the target cell, typically through endocytosis While PACs that comprise an antibody directed to an antigen that is not found on the cell surface may result in less specific intracellular delivery of the PROTAC portion into the cell, the PAC may still undergo pinocytosis. The PACs and method of their use described herein advantageously utilize antibody recognition of the cellular surface and/or endocytosis of the PAC to deliver the PROTAC portion inside cells.
a. Human Antibodies
In certain embodiments, an antibody provided herein is a human antibody. Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol.5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol.20:450-459 (2008).
Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous
immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal’s chromosomes. In such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated. For review of methods for obtaining human antibodies from transgenic animals, see Lonberg, Nat. Biotech.23:1117-1125 (2005). See also, e.g., U.S. Patent Nos.6,075,181 and 6,150,584 describing XENOMOUSETM technology; U.S. Patent No.5,770,429 describing HUMAB® technology; U.S. Patent No.7,041,870 describing K-M MOUSE® technology, and U.S. Patent Application Publication No. US 2007/0061900, describing VELOCIMOUSE® technology). Human variable regions from intact antibodies generated by such animals may be further modified, e.g., by combining with a different human constant region.
Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol., 133: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp.51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al., J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al., Proc. Natl. Acad. Sci. USA, 103:3557-3562 (2006). Additional methods include those described, for example, in U.S. Patent No.7,189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas). Human hybridoma technology (Trioma technology) is also described in Vollmers and Brandlein, Histology and Histopathology, 20(3):927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3):185-91 (2005).
Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
b. Library-Derived Antibodies
Antibodies for use in a PAC may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in
Hoogenboom et al. in Methods in Molecular Biology 178:1-37 (O’Brien et al., ed., Human Press, Totowa, NJ, 2001) and further described, e.g., in the McCafferty et al., Nature 348:552-554; Clackson et al., Nature 352: 624-628 (1991); Marks et al., J. Mol. Biol.222: 581-597 (1992); Marks and Bradbury, in Methods in Molecular Biology 248:161-175 (Lo, ed., Human Press, Totowa, NJ, 2003); Sidhu et al., J. Mol. Biol.338(2): 299-310 (2004); Lee et al., J. Mol. Biol.340(5): 1073-1093 (2004); Fellouse, Proc. Natl. Acad. Sci. USA 101(34): 12467-12472 (2004); and Lee et al., J. Immunol. Methods 284(1-2): 119-132(2004). In certain phage display methods, repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al., Ann. Rev. Immunol., 12: 433-455 (1994). Phage typically display antibody fragments, either as single- chain Fv (scFv) fragments or as Fab fragments. Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas. Alternatively, the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al., EMBO J, 12: 725-734 (1993). Finally, naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol., 227: 381-388 (1992). Patent publications describing human antibody phage libraries include, for example: US Patent No.5,750,373, and US Patent Publication Nos.2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598,
2007/0237764, 2007/0292936, and 2009/0002360.
Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
c. Chimeric and Humanized Antibodies
In certain embodiments, an antibody provided herein is a chimeric antibody. Certain chimeric antibodies are described, e.g., in U.S. Patent No.4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 81:6851-6855 (1984)). In one example, a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region. In a further example, a chimeric antibody is a“class switched” antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
In certain embodiments, a chimeric antibody is a humanized antibody. Typically, a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody. Generally, a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences. A humanized antibody optionally will also comprise at least a portion of a human constant region. In some embodiments, some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
Humanized antibodies and methods of making them are reviewed, e.g., in Almagro and Fransson, Front. Biosci.13:1619-1633 (2008), and are further described, e.g., in
Riechmann et al., Nature 332:323-329 (1988); Queen et al., Proc. Nat’l Acad. Sci. USA 86:10029-10033 (1989); US Patent Nos.5, 821,337, 7,527,791, 6,982,321, and 7,087,409; Kashmiri et al., Methods 36:25-34 (2005) (describing SDR (a-CDR) grafting); Padlan, Mol. Immunol.28:489-498 (1991) (describing“resurfacing”); Dall’Acqua et al., Methods 36:43-60 (2005) (describing“FR shuffling”); and Osbourn et al., Methods 36:61-68 (2005) and Klimka et al., Br. J. Cancer, 83:252-260 (2000) (describing the“guided selection” approach to FR shuffling).
Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the“best-fit” method (see, e.g., Sims et al. J. Immunol.151:2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol., 151:2623 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front. Biosci.13:1619-1633 (2008)); and framework regions derived from screening FR libraries (see, e.g., Baca et al., J. Biol. Chem.272:10678-10684 (1997) and Rosok et al., J. Biol. Chem.271:22611-22618 (1996)).
d. Multispecific Antibodies
In certain embodiments, an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody. The term“multispecific antibody” as used herein refers to an antibody comprising an antigen-binding domain that has polyepitopic specificity (i.e., is capable of binding to two, or more, different epitopes on one molecule or is capable of binding to epitopes on two, or more, different molecules).
In some embodiments, multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different antigen binding sites (such as a bispecific antibody). In some embodiments, the first antigen-binding domain and the second antigen- binding domain of the multispecific antibody may bind the two epitopes within one and the same molecule (intramolecular binding). For example, the first antigen-binding domain and the second antigen-binding domain of the multispecific antibody may bind to two different epitopes on the same protein molecule. In certain embodiments, the two different epitopes that a multispecific antibody binds are epitopes that are not normally bound at the same time by one monospecific antibody, such as e.g. a conventional antibody or one immunoglobulin single variable domain. In some embodiments, the first antigen-binding domain and the second antigen-binding domain of the multispecific antibody may bind epitopes located within two distinct molecules (intermolecular binding). For example, the first antigen- binding domain of the multispecific antibody may bind to one epitope on one protein molecule, whereas the second antigen-binding domain of the multispecific antibody may bind to another epitope on a different protein molecule, thereby cross-linking the two molecules.
In some embodiments, the antigen-binding domain of a multispecific antibody (such as a bispecific antibody) comprises two VH/VL units, wherein a first VH/VL unit binds to a first epitope and a second VH/VL unit binds to a second epitope, wherein each VH/VL unit comprises a heavy chain variable domain (VH) and a light chain variable domain (VL). Such multispecific antibodies include, but are not limited to, full length antibodies, antibodies having two or more VL and VH domains, and antibody fragments (such as Fab, Fv, dsFv, scFv, diabodies, bispecific diabodies and triabodies, antibody fragments that have been linked covalently or non-covalently). A VH/VL unit that further comprises at least a portion of a heavy chain variable region and/or at least a portion of a light chain variable region may also be referred to as an“arm” or“hemimer” or“half antibody.” In some embodiments, a hemimer comprises a sufficient portion of a heavy chain variable region to allow
intramolecular disulfide bonds to be formed with a second hemimer. In some embodiments, a hemimer comprises a knob mutation or a hole mutation, for example, to allow
heterodimerization with a second hemimer or half antibody that comprises a complementary hole mutation or knob mutation. Knob mutations and hole mutations are discussed further below.
In certain embodiments, a multispecific antibody provided herein may be a bispecific antibody. The term“bispecific antibody” as used herein refers to a multispecific antibody comprising an antigen-binding domain that is capable of binding to two different epitopes on one molecule or is capable of binding to epitopes on two different molecules. A bispecific antibody may also be referred to herein as having“dual specificity” or as being“dual specific.” Exemplary bispecific antibodies may bind both protein and any other antigen. In certain embodiments, one of the binding specificities is for protein and the other is for CD3. See, e.g., U.S. Patent No.5,821,337. In certain embodiments, bispecific antibodies may bind to two different epitopes of the same protein molecule. In certain embodiments, bispecific antibodies may bind to two different epitopes on two different protein molecules. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express protein.
Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983), WO 93/08829, and Traunecker et al., EMBO J.10: 3655 (1991)), and“knob-in-hole” engineering (see, e.g., U.S. Patent No.5,731,168, WO2009/089004, US2009/0182127, US2011/0287009, Marvin and Zhu, Acta Pharmacol. Sin. (2005) 26(6):649-658, and Kontermann (2005) Acta Pharmacol. Sin., 26:1-9). The term“knob-into-hole” or“KnH” technology as used herein refers to the technology directing the pairing of two polypeptides together in vitro or in vivo by introducing a protuberance (knob) into one polypeptide and a cavity (hole) into the other polypeptide at an interface in which they interact. For example, KnHs have been introduced in the Fc:Fc binding interfaces, CL:CH1 interfaces or VH/VL interfaces of antibodies (see, e.g., US 2011/0287009, US2007/0178552, WO 96/027011, WO 98/050431, Zhu et al., 1997, Protein Science 6:781-788, and WO2012/106587). In some embodiments, KnHs drive the pairing of two different heavy chains together during the manufacture of multispecific antibodies. For example, multispecific antibodies having KnH in their Fc regions can further comprise single variable domains linked to each Fc region, or further comprise different heavy chain variable domains that pair with similar or different light chain variable domains. KnH technology can be also be used to pair two different receptor extracellular domains together or any other polypeptide sequences that comprises different target recognition sequences (e.g., including affibodies, peptibodies and other Fc fusions).
The term“knob mutation” as used herein refers to a mutation that introduces a protuberance (knob) into a polypeptide at an interface in which the polypeptide interacts with another polypeptide. In some embodiments, the other polypeptide has a hole mutation. The term“hole mutation” as used herein refers to a mutation that introduces a cavity (hole) into a polypeptide at an interface in which the polypeptide interacts with another polypeptide. In some embodiments, the other polypeptide has a knob mutation.
A“protuberance” refers to at least one amino acid side chain which projects from the interface of a first polypeptide and is therefore positionable in a compensatory cavity in the adjacent interface (i.e. the interface of a second polypeptide) so as to stabilize the
heteromultimer, and thereby favor heteromultimer formation over homomultimer formation, for example. The protuberance may exist in the original interface or may be introduced synthetically (e.g., by altering nucleic acid encoding the interface). In some embodiments, nucleic acid encoding the interface of the first polypeptide is altered to encode the
protuberance. To achieve this, the nucleic acid encoding at least one“original” amino acid residue in the interface of the first polypeptide is replaced with nucleic acid encoding at least one“import” amino acid residue which has a larger side chain volume than the original amino acid residue. It will be appreciated that there can be more than one original and corresponding import residue. The side chain volumes of the various amino residues are shown, for example, in Table 1 of US2011/0287009. A mutation to introduce a
“protuberance” may be referred to as a“knob mutation.”
In some embodiments, import residues for the formation of a protuberance are naturally occurring amino acid residues selected from arginine (R), phenylalanine (F), tyrosine (Y) and tryptophan (W). In some embodiments, an import residue is tryptophan or tyrosine. In some embodiment, the original residue for the formation of the protuberance has a small side chain volume, such as alanine, asparagine, aspartic acid, glycine, serine, threonine or valine.
A“cavity” refers to at least one amino acid side chain which is recessed from the interface of a second polypeptide and therefore accommodates a corresponding protuberance on the adjacent interface of a first polypeptide. The cavity may exist in the original interface or may be introduced synthetically (e.g. by altering nucleic acid encoding the interface). In some embodiments, nucleic acid encoding the interface of the second polypeptide is altered to encode the cavity. To achieve this, the nucleic acid encoding at least one“original” amino acid residue in the interface of the second polypeptide is replaced with DNA encoding at least one“import” amino acid residue which has a smaller side chain volume than the original amino acid residue. It will be appreciated that there can be more than one original and corresponding import residue. In some embodiments, import residues for the formation of a cavity are naturally occurring amino acid residues selected from alanine (A), serine (S), threonine (T) and valine (V). In some embodiments, an import residue is serine, alanine or threonine. In some embodiments, the original residue for the formation of the cavity has a large side chain volume, such as tyrosine, arginine, phenylalanine or tryptophan. A mutation to introduce a“cavity” may be referred to as a“hole mutation.”
The protuberance is“positionable” in the cavity which means that the spatial location of the protuberance and cavity on the interface of a first polypeptide and second polypeptide respectively and the sizes of the protuberance and cavity are such that the protuberance can be located in the cavity without significantly perturbing the normal association of the first and second polypeptides at the interface. Since protuberances such as Tyr, Phe and Trp do not typically extend perpendicularly from the axis of the interface and have preferred
conformations, the alignment of a protuberance with a corresponding cavity may, in some instances, rely on modeling the protuberance/cavity pair based upon a three-dimensional structure such as that obtained by X-ray crystallography or nuclear magnetic resonance (NMR). This can be achieved using widely accepted techniques in the art.
In some embodiments, a knob mutation in an IgG1 constant region is T366W (EU numbering). In some embodiments, a hole mutation in an IgG1 constant region comprises one or more mutations selected from T366S, L368A and Y407V (EU numbering). In some embodiments, a hole mutation in an IgG1 constant region comprises T366S, L368A and Y407V (EU numbering).
In some embodiments, a knob mutation in an IgG4 constant region is T366W (EU numbering). In some embodiments, a hole mutation in an IgG4 constant region comprises one or more mutations selected from T366S, L368A, and Y407V (EU numbering). In some embodiments, a hole mutation in an IgG4 constant region comprises T366S, L368A, and Y407V (EU numbering).
Multispecific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No.4,676,980, and Brennan et al., Science, 229: 81 (1985)); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny et al., J. Immunol., 148(5):1547-1553 (1992)); using "diabody" technology for making bispecific antibody fragments (see, e.g., Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993)); and using single-chain Fv (sFv) dimers (see,e.g. Gruber et al., J. Immunol., 152:5368 (1994)); and preparing trispecific antibodies as described, e.g., in Tutt et al. J. Immunol.147: 60 (1991).
Engineered antibodies with three or more functional antigen binding sites, including “Octopus antibodies” or“dual-variable domain immunoglobulins” (DVDs) are also included herein (see, e.g., US 2006/0025576A1, and Wu et al. Nature Biotechnology (2007)).). The antibody or fragment herein also includes a“Dual Acting FAb” or“DAF” comprising an antigen binding site that binds to a target protein as well as another, different antigen (see, US 2008/0069820, for example).
e. Antibody Fragments
In certain embodiments, an antibody provided herein is an antibody fragment.
Antibody fragments include, but are not limited to, Fab, Fab’, Fab’-SH, F(ab’)2, Fv, and scFv fragments, and other fragments described below. For a review of certain antibody fragments, see Hudson et al. Nat. Med.9:129-134 (2003). For a review of scFv fragments, see, e.g., Pluckthün, in The Pharmacology of Monoclonal Antibodies, vol.113, Rosenburg and Moore eds., (Springer-Verlag, New York), pp.269-315 (1994); see also WO 93/16185; and U.S. Patent Nos.5,571,894 and 5,587,458. For discussion of Fab and F(ab')2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see U.S. Patent No.5,869,046.
Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al., Nat. Med.
9:129-134 (2003); and Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat. Med.9:129-134 (2003).
Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No.6,248,516 B1).
Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
f. Antibody Variants In certain embodiments, amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody. Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
i. Substitution, Insertion, and Deletion Variants
In certain embodiments, antibody variants having one or more amino acid
substitutions are provided. Sites of interest for substitutional mutagenesis include the HVRs and FRs. Conservative substitutions are shown in Table 1 under the heading of“preferred substitutions.” More substantial changes are provided in Table 1 under the heading of “exemplary substitutions,” and as further described below in reference to amino acid side chain classes. Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
Table 1. Amino acid substitutions.
Figure imgf000040_0001
Figure imgf000041_0001
Amino acids may be grouped according to common side-chain properties:
(1) hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile;
(2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gln;
(3) acidic: Asp, Glu;
(4) basic: His, Lys, Arg;
(5) residues that influence chain orientation: Gly, Pro;
(6) aromatic: Trp, Tyr, Phe.
Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
One type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody). Generally, the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody. An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display-based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
Alterations (e.g., substitutions) may be made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR“hotspots,” i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol.207:179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity. Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in
Hoogenboom et al. in Methods in Molecular Biology 178:1-37 (O’Brien et al., ed., Human Press, Totowa, NJ, (2001).) In some embodiments of affinity maturation, diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis). A secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity. Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
In certain embodiments, substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen. For example, conservative alterations (e.g., conservative substitutions as provided herein) that do not substantially reduce binding affinity may be made in HVRs. Such alterations may be outside of HVR“hotspots” or SDRs. In certain embodiments of the variant VH and VL sequences provided above, each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
A useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called“alanine scanning mutagenesis” as described by
Cunningham and Wells (1989) Science, 244:1081-1085. In this method, a residue or group of target residues (e.g., charged residues such as arg, asp, his, lys, and glu) are identified and replaced by a neutral or negatively charged amino acid (e.g., alanine or polyalanine) to determine whether the interaction of the antibody with antigen is affected. Further
substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions. Alternatively, or additionally, a crystal structure of an antigen-antibody complex is used to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution. Variants may be screened to determine whether they contain the desired properties.
Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue. Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for antibody directed enzyme prodrug therapy (ADEPT)) or a polypeptide which increases the serum half-life of the antibody.
ii. Cysteine engineered antibody variants
In certain embodiments, it may be desirable to create cysteine engineered antibodies, e.g.,“THIOMAB™ antibody,” in which one or more residues of an antibody are substituted with cysteine residues. In particular embodiments, the substituted residues occur at accessible sites of the antibody. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as L1-PROTAC groups, to create a PAC, as described further herein. In certain embodiments, any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; A140 (EU numbering) of the heavy chain; L174 (EU numbering) of the heavy chain; Y373 (EU numbering) of the heavy chain; K149 (Kabat numbering) of the light chain; A118 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region. In specific embodiments, the antibodies described herein comprise the HC-A140C (EU numbering) cysteine substitution. In specific embodiments, the antibodies described herein comprise the LC-K149C (Kabat numbering) cysteine substitution. In specific embodiments, the antibodies described herein comprise the HC-A118C (EU numbering) cysteine substitution.
Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No.7,521,541.
In certain embodiments, the antibody comprises one of the following heavy chain cysteine substitutions:
Table 2. HC Cysteine Substitutions.
Figure imgf000044_0002
In certain embodiments, the antibody comprises one of the following light chain cysteine substitutions:
Table 3. LC Cysteine Substitutions.
Figure imgf000044_0001
A nonlimiting exemplary hu7C2.v2.2.LA light chain (LC) K149C THIOMAB™ antibody has the heavy chain and light chain amino acid sequences of SEQ ID NOs: 26 and 30, respectively. A nonlimiting exemplary hu7C2.v2.2.LA heavy chain (HC) A118C THIOMAB™ antibody has the heavy chain and light chain amino acid sequences of SEQ ID NOs: 31 and 25, respectively.
PACs 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 THIOMABTM antibody 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 THIOMABTM antibody 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 for use in a PAC are in the ranges of 0.6 to 1.0; 0.7 to 1.0; or 0.8 to 1.0.
To prepare a cysteine engineered antibody by 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.
Cysteine amino acids may be engineered at reactive sites in an antibody and which do not form intrachain or intermolecular disulfide linkages (Junutula, et al., 2008b Nature Biotech., 26(8):925-932; Dornan et al (2009) Blood 114(13):2721-2729; US 7521541; US 7723485; WO2009/052249, Shen et al (2012) Nature Biotech., 30(2):184-191; Junutula et al (2008) Jour of Immun. Methods 332:41-52). The engineered cysteine thiols may react with linker reagents or the Linker L1-PROTAC intermediates described herein, which have thiol- reactive, electrophilic groups such as maleimides, activated disulfides (such as a 4- nitropyridyl disulfide), or alpha-halo amides to form a PAC with cysteine engineered antibodies (THIOMABTM antibodies) and the PROTAC residue. The location of the PROTAC moiety can thus be designed, controlled, and known. PROTAC/antibody ratio (“PAR”) can be controlled since the engineered cysteine thiol groups typically react with thiol-reactive linker reagents or linker L1-PROTAC intermediates in high yield. Engineering an antibody to introduce a cysteine amino acid by substitution at a single site on the heavy or light chain gives two new cysteines on the symmetrical antibody. A PAR of about 2 can be achieved and near homogeneity of the conjugation product.
Cysteine engineered antibodies 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, signaling 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).
Cysteine engineered antibodies are prepared for conjugation with linker L1 intermediates by reduction and reoxidation of intrachain disulfide groups.
iii. Glycosylation Variants
In certain embodiments, an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
Where the antibody comprises an Fc region, the carbohydrate attached thereto may be altered. Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997). The
oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the“stem” of the biantennary oligosaccharide structure. In some embodiments, modifications of the oligosaccharide in an antibody may be made in order to create antibody variants with certain improved properties. In one embodiment, antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. For example, the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5% to 65% or from 20% to 40%. The amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example. Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ± 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd). Examples of publications related to“defucosylated” or“fucose- deficient” antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140; US
2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; WO 2005/035586; WO 2005/035778; WO2005/053742; WO2002/031140; Okazaki et al. J. Mol. Biol.336:1239-1249 (2004); Yamane-Ohnuki et al. Biotech. Bioeng.87: 614 (2004). Examples of cell lines capable of producing defucosylated antibodies include Lec13 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys.249:533-545 (1986); US Pat Appl No US 2003/0157108 A1, Presta, L; and WO 2004/056312 A1, Adams et al., especially at Example 11), and knockout cell lines, such as alpha-1,6- fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki et al.
Biotech. Bioeng.87: 614 (2004); Kanda, Y. et al., Biotechnol. Bioeng., 94(4):680-688 (2006); and WO2003/085107).
Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by
GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean- Mairet et al.); US Patent No.6,602,684 (Umana et al.); and US 2005/0123546 (Umana et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087 (Patel et al.); WO 1998/58964 (Raju S ); and WO 1999/22764 (Raju S ) iv. Fc region variants
In certain embodiments, one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant. The Fc region variant may comprise a human Fc region sequence (e.g., a human IgG1, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g. a substitution) at one or more amino acid positions.
In certain embodiments, the subject matter described herein is directed to an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half-life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious. In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcγR binding (hence likely lacking ADCC activity), but retains FcRn binding ability. The primary cells for mediating ADCC, NK cells, express Fc(RIII only, whereas monocytes express Fc(RI, Fc(RII and Fc(RIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol.
9:457-492 (1991). Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No.5,500,362 (see, e.g. Hellstrom, I. et al. Proc. Nat’l Acad. Sci. USA 83:7059-7063 (1986)) and Hellstrom, I et al., Proc. Nat’l Acad. Sci. USA 82:1499-1502 (1985); 5,821,337 (see Bruggemann, M. et al., J. Exp. Med.
166:1351-1361 (1987)). Alternatively, non-radioactive assays methods may be employed (see, for example, ACTI™ non-radioactive cytotoxicity assay for flow cytometry
(CellTechnology, Inc. Mountain View, CA; and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, WI). 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. Proc. Nat’l Acad. Sci. USA 95:652-656 (1998). C1q binding assays may also be carried out to confirm that the antibody is unable to bind C1q and hence lacks CDC activity. See, e.g., C1q and C3c binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996); Cragg, M.S. et al., Blood 101:1045-1052 (2003); and Cragg, M.S. and M.J. Glennie, Blood 103:2738-2743 (2004)). FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al., Int’l. Immunol.18(12):1759- 1769 (2006)).
In some embodiments, one or more amino acid modifications may be introduced into the Fc portion of the antibody provided herein in order to increase IgG binding to the neonatal Fc receptor. In certain embodiments, the antibody comprises the following three mutations according to EU numbering: M252Y, S254T, and T256E (the“YTE mutation”) (US Patent No.8,697,650; see also Dall’Acqua et al., Journal of Biological Chemistry 281(33):23514-23524 (2006). In certain embodiments, the YTE mutation does not affect the ability of the antibody to bind to its cognate antigen. In certain embodiments, the YTE mutation increases the antibody’s serum half-life compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 3-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 2-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by 4-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by at least 5-fold compared to the native (i.e., non-YTE mutant) antibody. In some embodiments, the YTE mutation increases the serum half-life of the antibody by at least 10-fold compared to the native (i.e., non-YTE mutant) antibody. See, e.g., US Patent No.8,697,650; see also Dall’Acqua et al., Journal of Biological Chemistry 281(33):23514-23524 (2006).
In certain embodiments, the YTE mutant provides a means to modulate antibody- dependent cell-mediated cytotoxicity (ADCC) activity of the antibody. In certain
embodiments, the YTEO mutant provides a means to modulate ADCC activity of a humanized IgG antibody directed against a human antigen. See, e.g., US Patent No.
8,697,650; see also Dall’Acqua et al., Journal of Biological Chemistry 281(33):23514-23524 (2006).
In certain embodiments, the YTE mutant allows the simultaneous modulation of serum half-life, tissue distribution, and antibody activity (e.g., the ADCC activity of an IgG antibody). See, e.g., US Patent No.8,697,650; see also Dall’Acqua et al., Journal of
Biological Chemistry 281(33):23514-23524 (2006).
Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 according to EU numbering (U S Patent No 6737056) Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327 according to EU numbering, including the so-called“DANA” Fc mutant with substitution of residues 265 and 297 to alanine according to EU numbering (i.e., D265A and N297A according to EU numbering) (US Patent No.7,332,581). In certain embodiments the Fc mutant comprises the following two amino acid substitutions: D265A and N297A. In certain embodiments the Fc mutant consists of the following two amino acid substitutions: D265A and N297A.
In certain embodiments, the proline at position329 (EU numbering) (P329) of a wild- type human Fc region is substituted with glycine or arginine or an amino acid residue large enough to destroy the proline sandwich within the Fc/Fcγ receptor interface, that is formed between the P329 of the Fc and tryptophane residues W87 and W110 of FcgRIII
(Sondermann et al.: Nature 406, 267-273 (20 July 2000)). In a further embodiment, at least one further amino acid substitution in the Fc variant is S228P, E233P, L234A, L235A, L235E, N297A, N297D, or P331S and still in another embodiment said at least one further amino acid substitution is L234A and L235A of the human IgG1 Fc region or S228P and L235E of the human IgG4 Fc region, all according to EU numbering (U.S. Patent No.
8,969,526).
In certain embodiments, a polypeptide comprises the Fc variant of a wild-type human IgG Fc region wherein the polypeptide has P329 of the human IgG Fc region substituted with glycine and wherein the Fc variant comprises at least two further amino acid substitutions at L234A and L235A of the human IgG1 Fc region or S228P and L235E of the human IgG4 Fc region, and wherein the residues are numbered according to the EU numbering (U.S. Patent No.8,969,526). In certain embodiments, the polypeptide comprising the P329G, L234A and L235A (EU numbering) substitutions exhibit a reduced affinity to the human FcγRIIIA and FcγRIIA, for down-modulation of ADCC to at least 20% of the ADCC induced by the polypeptide comprising the wild-type human IgG Fc region, and/or for down-modulation of ADCP (U.S. Patent No.8,969,526).
In a specific embodiment the polypeptide comprising an Fc variant of a wild-type human Fc polypeptide comprises a triple mutation: an amino acid substitution at position Pro329, a L234A and a L235A mutation according to EU numbering (P329 / LALA) (U.S. Patent No.8,969,526). In specific embodiments, the polypeptide comprises the following amino acid substitutions: P329G, L234A, and L235A according to EU numbering. Certain antibody variants with improved or diminished binding to FcRs are described. (See, e.g., U.S. Patent No.6,737,056; WO 2004/056312, and Shields et al., J. Biol. Chem. 9(2): 6591-6604 (2001).)
In certain embodiments, an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering).
In some embodiments, alterations are made in the Fc region that result in altered (i.e., either improved or diminished) C1q binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No.6,194,551, WO 99/51642, and Idusogie et al. J. Immunol.164: 4178-4184 (2000).
Antibodies with increased half lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol.117:587 (1976) and Kim et al., J. Immunol.24:249 (1994)), are described in US2005/0014934A1 (Hinton et al.). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn. Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No.7,371,826) according to EU numbering. See also Duncan & Winter, Nature 322:738-40 (1988); U.S. Patent No.
5,648,260; U.S. Patent No.5,624,821; and WO 94/29351 concerning other examples of Fc region variants.
g. Antibody Derivatives
In certain embodiments, an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available. The moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers. Non-limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3- dioxolane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propylene glycol homopolymers, polypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof.
Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water. The polymer may be of any molecular weight, and may be branched or unbranched. The number of polymers attached to the antibody may vary, and if more than one polymer is attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
In another embodiment, conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided. In one embodiment, the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)). The radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
h. Tumor-Associated Antigens
Antibodies, including but not limited to cysteine engineered antibodies, which may be useful in the PACs described herein in the treatment of cancer include, but are not limited to, antibodies against cell surface receptors and tumor-associated antigens (TAA). Certain tumor-associated antigens are known in the art, and can be 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 more specifically target cancer cells for destruction via antibody-based therapies.
Examples of tumor-associated antigens TAA include, but are not limited to, those 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 listed below 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, and/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. i. Recombinant Methods and Compositions
Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No.4,816,567. In one embodiment, isolated nucleic acid encoding an antibody described herein is provided. Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody). In a further embodiment, one or more vectors (e.g., expression vectors) comprising such nucleic acid are provided. In a further embodiment, a host cell comprising such nucleic acid is provided. In one such embodiment, a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody. In one embodiment, the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NS0, Sp20 cell). In one embodiment, a method of making an antibody is provided, wherein the method comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
For recombinant production of an antibody, nucleic acid encoding an antibody, e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acid may be 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 the antibody).
Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein. For example, antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed. For expression of antibody fragments and polypeptides in bacteria, see, e.g., U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. (See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2003), pp.245-254, describing expression of antibody fragments in E. coli.) After expression, the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been“humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat.
Biotech.22:1409-1414 (2004), and Li et al., Nat. Biotech.24:210-215 (2006).
Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos.5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants).
Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol.36:59 (1977); baby hamster kidney cells (BHK); mouse sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod.23:243-251 (1980); monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al., Annals N.Y. Acad. Sci.383:44-68 (1982); MRC 5 cells; and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR- CHO cells (Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); and myeloma cell lines such as Y0, NS0 and Sp2/0. For a review of certain mammalian host cell lines suitable for antibody production, see, e.g., Yazaki and Wu, Methods in Molecular Biology, Vol.248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ), pp.255-268 (2003).
Referring now to antibody affinity, in embodiments, the antibody binds to one or more tumor-associated antigens or cell-surface receptors selected from (1)-(53):
(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); US 798959. Cross-references: GI:34501467;
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) Napi2b (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: GI:37182378; AAQ88991.1; AY358628_1 (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, s1-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 111 198 206; 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 1a; 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: GI:22655488; AAN04080.1; AF455138_1 (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, CR1, CRGF, CRIPTO, TDGF1, 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, CD79β, 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, Muller 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, SPAP1A (SH2 domain containing phosphatase anchor protein 1a), 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_110391.2; NM_030764_1 (17) HER2 (ErbB2, Genbank accession no. M11730)
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 1I); 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; M11767; AAA35808.1. EMBL; M11761;
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);
WO9946284 (Fig 9);
Cross-references: MIM:179780; AAH17023.1; BC017023_1 (20) IL20Rα (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); WO9837193 (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 1b); WO200032752 (Claim 18; Fig 1);
WO9851805 (Claim 17; Page 97); WO9851824 (Claim 10; Page 94); WO9840403
(Claim 2; Fig 1B);
Accession: O43653; 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: GI:30102449; 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; AF132600 (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, CD79α, 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: 19q13.2, Genbank accession No. NP_001774.10)
WO2003088808, US20030228319; WO2003062401 (claim 9); US2002150573 (claim 4, pages 13-14); WO9958658 (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: 11q23.3, 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); WO9928468 (claim 1, page 38); US5440021 (Example 2, col 49-52); WO9428931 (pages 56-58);
WO9217497 (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 (Ia 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; WO9958658 (claim 13, Fig 15); US6153408 (Col 35-38); US5976551 (col 168-170);
US6011146 (col 145-146); Kasahara et al (1989) Immunogenetics 30(1):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: 17p13.3, 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: 9p13.3, 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: 5q12, 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; WO9744452 (claim 8 pages 57 61); WO200012130 (pages 24 26); (34) FcRH1 (Fc receptor-like protein 1, a putative receptor for the immunoglobulin Fc domain that contains C2 type Ig-like and ITAM domains, may have a role in B-lymphocyte differentiation); 429 aa, pI: 5.28, MW: 46925 TM: 1 [P] Gene Chromosome: 1q21-1q22, 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) FCRH5 (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: 1q21, 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, HPP1, 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; gp100) 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; C9ORF2; 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-Ra1 (GDNF family receptor alpha 1; GFRA1; GDNFR; GDNFRA; RETL1; TRNR1; RET1L; GDNFR-alpha1; 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-1);
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; WO 2013/17705; (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 (1):278-287; (43) LGR5 (leucine-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-ELE1); 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_115940.2; 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) ASPHD1 (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; OCA1A; 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 (11):6759-6764; Takeda, S. et al (2002) FEBS Lett.520 (1-3):97-101. (52) CD33, a member of the sialic acid binding, immunoglobulin-like lectin family, is a 67- kDa glycosylated transmembrane protein. CD33is expressed on most myeloid and monocytic leukemia cells in addition to committed myelomonocytic and erythroid progenitor cells. It is not seen on the earliest pluripotent stem cells, mature granulocytes, lymphoid cells, or nonhematopoietic cells (Sabbath et al., (1985) J. Clin. Invest.75:756-56; Andrews et al., (1986) Blood 68:1030-5). CD33 contains two tyrosine residues on its cytoplasmic tail, each of which is followed by hydrophobic residues similar to the immunoreceptor tyrosine-based inhibitory motif (ITIM) seen in many inhibitory receptors. (53) CLL-1 (CLEC12A, MICL, and DCAL2), encodes a member of the C-type lectin/C-type lectin-like domain (CTL/CTLD) superfamily. Members of this family share a common protein fold and have diverse functions, such as cell adhesion, cell-cell signalling, glycoprotein turnover, and roles in inflammation and immune response. The protein encoded by this gene is a negative regulator of granulocyte and monocyte function. Several alternatively spliced transcript variants of this gene have been described, but the full-length nature of some of these variants has not been determined. This gene is closely linked to other CTL/CTLD superfamily members in the natural killer gene complex region on chromosome 12p13 (Drickamer K (1999) Curr. Opin. Struct. Biol.9 (5):585–90; van Rhenen A, et al., (2007) Blood 110 (7):2659–66; Chen CH, et al. (2006) Blood 107 (4):1459–67; Marshall AS, et al. (2006) Eur. J. Immunol.36 (8):2159–69; Bakker AB, et al (2005) Cancer Res.64 (22):8443–50; Marshall AS, et al (2004) J. Biol. Chem.279 (15):14792–802). CLL-1 has been shown to be a type II transmembrane receptor comprising a single C-type lectin-like domain (which is not predicted to bind either calcium or sugar), a stalk region, a
transmembrane domain and a short cytoplasmic tail containing an ITIM motif.
In an aspect, the antibody of the PAC may be an antibody that is directed to a protein that is found on numerous cells or tissue types. Examples of such antibodies include gD and EpCAM. In other words, a PAC can be used to deliver a PROTAC to many cells or tissues rather thanspecific cell types or tissue types as when using a using a targeted antibody.
As described herein, a PAC may comprise an antibody, e.g., an antibody selected from:
Anti-Ly6E Antibodies
In certain embodiments, a PAC can comprise anti-Ly6E antibodies. Lymphocyte antigen 6 complex, locus E (Ly6E), also known as retinoic acid induced gene E (RIG-E) and stem cell antigen 2 (SCA-2). It is a GPI linked, 131 amino acid length, ~8.4kDa protein of unknown function with no known binding partners. It was initially identified as a transcript expressed in immature thymocyte, thymic medullary epithelial cells in mice (Mao, et al. (1996) Proc. Natl. Acad. Sci. U.S.A.93:5910-5914). In some embodiments, the subject matter described herein provides a PAC comprising an anti-Ly6E antibody described in PCT Publication No. WO 2013/177055.
In some embodiments, the subject matter described herein provides a PAC comprising an anti-Ly6E antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
In one aspect, the subject matter described herein provides a PAC comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14. In a further embodiment, the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14.
In another aspect, the subject matter described herein provides a PAC comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11. In one embodiment, the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
In another aspect, a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 12, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 14; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11. In another aspect, the subject matter described herein provides a PAC comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
In any of the above embodiments, an anti-Ly6E antibody of a PAC is humanized. In one embodiment, an anti-Ly6E antibody comprises HVRs as in any of the above
embodiments, and further comprises a human acceptor framework, e.g. a human
immunoglobulin framework or a human consensus framework.
In another aspect, an anti-Ly6E antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 8. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:8 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to Ly6E. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 8. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 8. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-Ly6E antibody comprises the VH sequence of SEQ ID NO: 8, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 12, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 13, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 14.
In another aspect, an anti-Ly6E antibody of a PAC is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 7. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:7 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to Ly6E In certain embodiments a total of 1 to 10 amino acids have been substituted inserted and/or deleted in SEQ ID NO: 7. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 7. In certain
embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-Ly6E antibody comprises the VL sequence of SEQ ID NO: 7, including post-translational modifications of that sequence. In a particular
embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 9; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 10; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 11.
In another aspect, a PAC comprising an anti-Ly6E antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
In one embodiment, a PAC is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 8 and SEQ ID NO: 7, respectively, including post-translational modifications of those sequences.
In a further aspect, provided herein are PACs comprising antibodies that bind to the same epitope as an anti-Ly6E antibody provided herein. For example, in certain
embodiments, a PAC is provided comprising an antibody that binds to the same epitope as an anti-Ly6E antibody comprising a VH sequence of SEQ ID NO: 8 and a VL sequence of SEQ ID NO: 7, respectively.
In a further aspect, an anti-Ly6E antibody of a PAC according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-Ly6E antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’)2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein. In some embodiments, a PAC comprises an anti-Ly6E antibody comprising a heavy chain and a light chain comprising the amino acid sequences of SEQ ID NO: 16 and 15, respectively. Table 4. Ly6E AntibodySequences.
Figure imgf000072_0001
Figure imgf000073_0001
Anti-HER2 Antibodies
In certain embodiments, PACs comprise anti-HER2 antibodies. In one embodiment, an anti-HER2 antibody of a PAC comprises a humanized anti-HER2 antibody, e.g., huMAb4D5-1, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5-8, as described in Table 3 of US 5821337. Those antibodies contain human framework regions with the complementarity-determining regions of a murine antibody (4D5) that binds to HER2. The humanized antibody huMAb4D5-8 is also referred to as trastuzumab, commercially available under the tradename HERCEPTIN ^. In another embodiment, an anti-HER2 antibody of a PAC comprises a humanized anti-HER2 antibody, e.g., humanized 2C4, as described in US7862817. An exemplary humanized 2C4 antibody is pertuzumab, commercially available under the tradename PERJETA ^.
In another embodiment, an anti-HER2 antibody of a PAC comprises a humanized 7C2 anti-HER2 antibody. A humanized 7C2 antibody is an anti-HER2 antibody.
In some embodiments, described herein are PACs comprising an anti-HER2 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In some embodiments, described herein are PACs comprising an anti-HER2 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
In one aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29. In one aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24. In a further embodiment, the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 68; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29. In a further embodiment, the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24.
In another aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In one embodiment, the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
In another aspect, a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 22, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 24 or 29; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In another aspect, a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 24; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
In another aspect, described herein are PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, 27, or 28; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24 or 29; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21. In another aspect, described herein are PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (e) HVR- L2 comprising the amino acid sequence of SEQ ID NO: 20; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
In any of the above embodiments, an anti-HER2 antibody of a PAC is humanized. In one embodiment, an anti-HER2 antibody of a PAC comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
immunoglobulin framework or a human consensus framework.
In another aspect, an anti-HER2 antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 18. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 18 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti HER2 antibody comprising that sequence retains the ability to bind to HER2 In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 18. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 18. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti- HER2 antibody comprises the VH sequence of SEQ ID NO: 18, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 22, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 23, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 24.
In another aspect, an anti-HER2 antibody of a PAC is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 17. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 17 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-HER2 antibody comprising that sequence retains the ability to bind to HER2. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 17. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 17. In certain
embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-HER2 antibody comprises the VL sequence of SEQ ID NO: 17, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 19; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 20; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 21.
In another aspect, a PAC comprising an anti-HER2 antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
In one embodiment, a PAC comprising an antibody is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 18 and SEQ ID NO: 17, respectively, including post-translational modifications of those sequences. In one embodiment, a PAC comprising an antibody is provided, wherein the antibody comprises the humanized 7C2.v2.2.LA (hu7C2) K149C kappa light chain sequence of SEQ ID NO: 30
In one embodiment, a PAC comprising an antibody is provided, wherein the antibody comprises the Hu7C2 A118C IgG1 heavy chain sequence of SEQ ID NO: 31
In a further aspect, provided herein are PACs comprising antibodies that bind to the same epitope as an anti-HER2 antibody provided herein. For example, in certain
embodiments, a PAC is provided, comprising an antibody that binds to the same epitope as an anti-HER2 antibody comprising a VH sequence of SEQ ID NO: 18 and a VL sequence of SEQ ID NO: 17, respectively.
In a further aspect, an anti-HER2 antibody of a PAC according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-HER2 antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’)2 fragment. In another embodiment, a PAC comprises an antibody that is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
Table 5. Humanized 7C2 anti-HER2 antibody sequences.
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Anti-MUC16 Antibodies
In certain embodiments, PACs comprise anti-MUC16 antibodies.
In some embodiments, described herein are PACs comprising an anti-MUC16 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
In one aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37. In a further embodiment, the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37.
In another aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34. In one embodiment, the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
In another aspect, a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 35, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 37; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
In another aspect, described herein are PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
In any of the above embodiments, an anti-MUC16 antibody of a PAC is humanized. In one embodiment, an anti-MUC16 antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
immunoglobulin framework or a human consensus framework.
In another aspect, an anti-MUC16 antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 39. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 39 contains substitutions (e g conservative substitutions) insertions or deletions relative to the reference sequence, but an anti-MUC16 antibody comprising that sequence retains the ability to bind to MUC16. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 39. In certain
embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-MUC16 antibody comprises the VH sequence of SEQ ID NO: 39, including post-translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 35, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 36, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 37.
In another aspect, an anti-MUC16 antibody of a PAC is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 38. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO:38 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-MUC16 antibody comprising that sequence retains the ability to bind to MUC16. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 38. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 38. In certain embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-MUC16 antibody comprises the VL sequence of SEQ ID NO: 38, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 32; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 33; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 34.
In another aspect, a PAC comprising an anti-MUC16 antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
In one embodiment, a PAC is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 39 and SEQ ID NO: 38, respectively, including post- translational modifications of those sequences In a further aspect, provided herein are PACs comprising antibodies that bind to the same epitope as an anti-MUC16 antibody provided herein. For example, in certain embodiments, a PAC is provided comprising an antibody that binds to the same epitope as an anti-MUC16 antibody comprising a VH sequence of SEQ ID NO: 39 and a VL sequence of SEQ ID NO: 38, respectively.
In a further aspect, an anti-MUC16 antibody of a PAC according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-MUC16 antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’)2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
Table 6. MUC16 Antibody Sequences.
Figure imgf000082_0001
Anti-STEAP-1 Antibodies
In certain embodiments PACs comprise anti STEAP 1 antibodies In some embodiments, described herein are PACs comprising an anti-STEAP-1 antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
In one aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42. In a further embodiment, the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42.
In another aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45. In one embodiment, the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
In another aspect, a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 40, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 42; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
In another aspect, described herein are PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
In any of the above embodiments, an anti-STEAP-1 antibody of a PAC is humanized. In one embodiment, an anti-STEAP-1 antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
immunoglobulin framework or a human consensus framework.
In another aspect, an anti-STEAP-1 antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 46. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 46 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-STEAP-1 antibody comprising that sequence retains the ability to bind to STEAP-1. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 46. In certain
embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-STEAP-1 antibody comprises the VH sequence of SEQ ID NO: 46, including post-translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 40, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 41, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 42.
In another aspect, an anti-STEAP-1 antibody of an a PAC is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 47. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 47 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-STEAP-1 antibody comprising that sequence retains the ability to bind to STEAP-1. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 47 In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 47. In certain embodiments the substitutions insertions or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-STEAP-1 antibody comprises the VL sequence of SEQ ID NO: 47, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 43; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 44; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 45.
In another aspect, a PAC comprising an anti-STEAP-1 antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
In one embodiment, a PAC is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 46 and SEQ ID NO: 47, respectively, including post- translational modifications of those sequences.
In a further aspect, provided herein are PACs comprising antibodies that bind to the same epitope as an anti-STEAP-1 antibody provided herein. For example, in certain embodiments, a PAC is provided comprising an antibody that binds to the same epitope as an anti-STEAP-1 antibody comprising a VH sequence of SEQ ID NO: 46 and a VL sequence of SEQ ID NO: 47, respectively.
In a further aspect, an anti-STEAP-1 antibody of a PAC according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-STEAP-1 antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’)2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein. Table 7. STEAP Antibody Sequences.
Figure imgf000085_0001
Figure imgf000086_0001
Anti-NaPi2b Antibodies
In certain embodiments, a PAC comprises anti-NaPi2b antibodies.
In some embodiments, described herein are PACs comprising an anti-NaPi2b antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52 and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
In one aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50. In a further embodiment, the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50.
In another aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53. In one embodiment, the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
In another aspect, a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 48, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 50; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
In another aspect, described herein are PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48 (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
In any of the above embodiments, an anti-NaPi2b antibody of a PAC is humanized. In one embodiment, an anti-NaPi2b antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
immunoglobulin framework or a human consensus framework.
In another aspect, an anti-NaPi2b antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 54. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 54 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-NaPi2b antibody comprising that sequence retains the ability to bind to NaPi2b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 54. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 54. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-NaPi2b antibody comprises the VH sequence of SEQ ID NO: 54, including post- translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 48, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 49, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 50.
In another aspect, an anti-NaPi2b antibody of a PAC is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 55 In certain embodiments a VL sequence having at least 90% 91% 92% 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 55 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-NaPi2b antibody comprising that sequence retains the ability to bind to anti-NaPi2b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 55. In certain
embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 55. In certain embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-NaPi2b antibody comprises the VL sequence of SEQ ID NO: 55, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 51; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 52; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 53.
In another aspect, a PAC comprising an anti-NaPi2b antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
In one embodiment, a PAC is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 54 and SEQ ID NO: 55, respectively, including post- translational modifications of those sequences.
In a further aspect, provided herein are PACs comprising antibodies that bind to the same epitope as an anti-NaPi2b antibody provided herein. For example, in certain
embodiments, a PAC is provided comprising an antibody that binds to the same epitope as an anti-NaPi2b antibody comprising a VH sequence of SEQ ID NO: 54 and a VL sequence of SEQ ID NO: 55, respectively.
In a further aspect, an anti-NaPi2b antibody of a PAC according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-NaPi2b antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’)2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
Figure imgf000089_0001
Anti-CD79b Antibodies
In certain embodiments, PACs comprise anti-CD79b antibodies.
In some embodiments, described herein are PACs comprising an anti-CD79b antibody comprising at least one, two, three, four, five, or six HVRs selected from (a) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
In one aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VH HVR sequences selected from (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60. In a further embodiment, the antibody comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60.
In another aspect, described herein are PACs comprising an antibody that comprises at least one, at least two, or all three VL HVR sequences selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63. In one embodiment, the antibody comprises (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
In another aspect, a PAC comprises an antibody comprising (a) a VH domain comprising at least one, at least two, or all three VH HVR sequences selected from (i) HVR- H1 comprising the amino acid sequence of SEQ ID NO: 58, (ii) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59, and (iii) HVR-H3 comprising an amino acid sequence selected from SEQ ID NO: 60; and (b) a VL domain comprising at least one, at least two, or all three VL HVR sequences selected from (i) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61, (ii) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62, and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
In another aspect, described herein are PACs comprising an antibody that comprises (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58; (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59; (c) HVR H3 comprising the amino acid sequence of SEQ ID NO: 60; (d) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (e) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (f) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
In any of the above embodiments, an anti-CD79b antibody of a PAC is humanized. In one embodiment, an anti-CD79b antibody comprises HVRs as in any of the above embodiments, and further comprises a human acceptor framework, e.g. a human
immunoglobulin framework or a human consensus framework.
In another aspect, an anti-CD79b antibody of a PAC comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 56. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 56 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-CD79b antibody comprising that sequence retains the ability to bind to CD79b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 56. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 56. In certain embodiments,
substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-CD79b antibody comprises the VH sequence of SEQ ID NO: 8, including post-translational modifications of that sequence. In a particular embodiment, the VH comprises one, two or three HVRs selected from: (a) HVR-H1 comprising the amino acid sequence of SEQ ID NO: 58, (b) HVR-H2 comprising the amino acid sequence of SEQ ID NO: 59, and (c) HVR-H3 comprising the amino acid sequence of SEQ ID NO: 60.
In another aspect, an anti-CD79b antibody of a PAC is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 57. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence of SEQ ID NO: 57 contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-Ly6E antibody comprising that sequence retains the ability to bind to CD79b. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 57. In certain embodiments, a total of 1 to 5 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 57. In certain embodiments the substitutions insertions or deletions occur in regions outside the HVRs (i.e., in the FRs). Optionally, the anti-CD79b antibody comprises the VL sequence of SEQ ID NO: 57, including post-translational modifications of that sequence. In a particular embodiment, the VL comprises one, two or three HVRs selected from (a) HVR-L1 comprising the amino acid sequence of SEQ ID NO: 61; (b) HVR-L2 comprising the amino acid sequence of SEQ ID NO: 62; and (c) HVR-L3 comprising the amino acid sequence of SEQ ID NO: 63.
In another aspect, described herein are PACs comprising an anti-CD79b antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
In one embodiment, a PAC is provided, wherein the antibody comprises the VH and VL sequences in SEQ ID NO: 56 and SEQ ID NO: 57, respectively, including post- translational modifications of those sequences.
In a further aspect, provided herein are PACs comprising antibodies that bind to the same epitope as an anti-CD79b antibody provided herein. For example, in certain
embodiments, a PAC is provided comprising an antibody that binds to the same epitope as an anti-CD79b antibody comprising a VH sequence of SEQ ID NO: 56 and a VL sequence of SEQ ID NO: 57, respectively.
In a further aspect, an anti-CD79b antibody of a PAC according to any of the above embodiments is a monoclonal antibody, including a human antibody. In one embodiment, an anti-CD79b antibody of a PAC is an antibody fragment, e.g., a Fv, Fab, Fab’, scFv, diabody, or F(ab’)2 fragment. In another embodiment, the antibody is a substantially full length antibody, e.g., an IgG1 antibody, IgG2a antibody or other antibody class or isotype as defined herein.
Table 9. CD79b Antibody Sequences.
Figure imgf000092_0001
Figure imgf000093_0001
Anti-CD22 Antibodies
In certain embodiments, a PAC can comprise anti-CD22 antibodies, which comprise three light chain hypervariable regions (HVR-L1, HVR-L2 and HVR-L3) and three heavy chain hypervariable regions (HVR-H1, HVR-H2 and HVR-H3). In one embodiment, the anti-CD22 antibody of a PAC comprises three light chain hypervariable regions and three heavy chain hypervariable regions (SEQ ID NO: 66-71), the sequences of which are shown below. In one embodiment, the anti-CD22 antibody of a PAC comprises the variable light chain sequence of SEQ ID NO: 72 and the variable heavy chain sequence of SEQ ID NO: 73. In one embodiment, the anti-CD22 antibody of PACs of the present invention comprises the light chain sequence of SEQ ID NO: 74 and the heavy chain sequence of SEQ ID NO: 75:
Figure imgf000093_0002
Figure imgf000094_0001
Anti-CD33 Antibodies
In certain embodiments, a PAC can comprise anti-CD33 antibodies, which comprise three light chain hypervariable regions and three heavy chain hypervariable regions, the sequences (SEQ ID NO:76-81) of which are shown below. In one embodiment, the anti- CD33 antibody of a PAC comprises the variable light chain sequence of SEQ ID NO: 82 and the variable heavy chain sequce of SEQ ID NO: 83. Table 11.
Figure imgf000095_0001
In one embodiment, the anti-CD33 antibody of a PAC comprises the light chain sequence of SEQ ID NO: 84 and the heavy chain sequence of SEQ ID NO: 85. In one embodiment, the anti-CD33 antibody of a PAC comprises three light chain hypervariable regions and three heavy chain hypervariable regions, the sequences (Seq ID NO: 84-89) of which are shown below. In one embodiment, the anti-CD33 antibody of a PAC comprises the variable light chain sequence of SEQ ID NO: 90 and the variable heavy chain sequce of SEQ ID NO: 91. In one embodiment, the anti-CD33 antibody of PAC comprises the variable light chain sequence of SEQ ID NO: 92 and the variable heavy chain sequce of SEQ ID NO: 93. In one embodiment, the anti-CD33 antibody of the present invention comprises the variable light chain sequence of SEQ ID NO: 94 and the variable heavy chain sequce of SEQ ID NO: 95. In one embodiment, the anti-CD33 antibody of the present invention comprises the variable light chain sequence of SEQ ID NO: 96 and the variable heavy chain sequce of SEQ ID NO: 97.
Figure imgf000096_0001
Figure imgf000097_0001
Antibody Affinity
In certain embodiments, an antibody provided herein has a dissociation constant (Kd) of ≤ 1µM,≤ 100 nM,≤ 50 nM,≤ 10 nM,≤ 5 nM,≤ 1 nM,≤ 0.1 nM,≤ 0.01 nM, or≤ 0.001 nM, and optionally is≥ 10-13 M. (e.g.10-8 M or less, e.g. from 10-8 M to 10-13 M, e.g., from 10-9 M to 10-13 M).
In one embodiment, Kd is measured by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay. Solution binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of (125I)-labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et al., J. Mol. Biol.293:865-881(1999)). To establish conditions for the assay,
MICROTITER® multi-well plates (Thermo Scientific) are coated overnight with 5 µg/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and
subsequently blocked with 2% (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23°C). In a non-adsorbent plate (Nunc #269620), 100 pM or 26 pM [125I]-antigen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of the anti-VEGF antibody, Fab-12, in Presta et al., Cancer Res.57:4593-4599 (1997)). The Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour). The solution is then removed and the plate washed eight times with 0.1% polysorbate 20 (TWEEN- 20®) in PBS. When the plates have dried, 150 µl/well of scintillant (MICROSCINT-20 TM;
Packard) is added, and the plates are counted on a TOPCOUNT TM gamma counter (Packard) for ten minutes. Concentrations of each Fab that give less than or equal to 20% of maximal binding are chosen for use in competitive binding assays.
According to another embodiment, Kd is measured using surface plasmon resonance assays using a BIACORE®-2000 or a BIACORE ®-3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at ~10 response units (RU). Briefly,
carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N- ethyl-N’- (3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N- hydroxysuccinimide (NHS) according to the supplier’s instructions. Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 µg/ml (~0.2 µM) before injection at a flow rate of 5 µl/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% polysorbate 20 (TWEEN-20TM) surfactant (PBST) at 25°C at a flow rate of approximately 25 µl/min. Association rates (kon) and dissociation rates (koff) are calculated using a simple one-to-one Langmuir binding model (BIACORE ® Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (Kd) is calculated as the ratio koff/kon. See, e.g., Chen et al., J. Mol. Biol.293:865-881 (1999). If the on-rate exceeds 106 M-1 s-1 by the surface plasmon resonance assay above, then the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity (excitation = 295 nm; emission = 340 nm, 16 nm band-pass) at 25oC of a 20 nM anti-antigen antibody (Fab form) in PBS, pH 7.2, in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophotometer (Aviv Instruments) or a 8000-series SLM-AMINCO TM spectrophotometer
(ThermoSpectronic) with a stirred cuvette.
2. Linkers (L1)
As described herein, a“linker” (L1) is a bifunctional or multifunctional moiety that can be used to link one or more PROTAC moieties (D) to an antibody (Ab) to form a PAC. In some embodiments, PACs can be prepared using a L1 having reactive functionalities for covalently attaching to the PROTAC and to the antibody. For example, in some
embodiments, a cysteine thiol of an antibody (Ab) can form a bond with a reactive functional group of a linker or a linker L1-PROTAC group to make a PAC. Particularly, the chemical structure of the linker can have significant impact on both the efficacy and the safety of a PAC (Ducry & Stump, Bioconjugate Chem, 2010, 21, 5-13). Choosing the right linker influences proper drug delivery to the intended cellular compartment of target cells.
Linkers can be generally divided into two categories: cleavable (such as peptide, hydrzone, or disulfide) or non-cleavable (such as thioether). Peptide linkers, such as Valine- Citrulline (Val-Cit), that can be hydrolyzed by lysosomal enzymes (such as Cathepsin B) have been used to connect the drug with the antibody (US 6,214,345). They have been particularly useful, due in part to their relative stability in systemic circulation and the ability to efficiently release the drug in tumor. However, the chemical space represented by natural peptides is limited; therefore, it is desirable to have a variety of non-peptide linkers which act like peptides and can be effectively cleaved by lysosomal proteases. The greater diversity of non-peptide structures may yield novel, beneficial properties that are not afforded by the peptide linkers. Provided herein are different types of non-peptide linkers for linker L1 that can be cleaved by lysosomal enzymes.
a. Peptidomimetic Linkers Provided herein are different types of non-peptide, peptidomimetic linkers for PAC that are cleavable by lysosomal enzymes. For example, the amide bond in the middle of a dipeptide (e.g. Val-Cit) was replaced with an amide mimic; and/or entire amino acid (e.g., valine amino acid in Val-Cit dipeptide) was replaced with a non-amino acid moiety (e.g., cycloalkyl dicarbonyl structures (for example, ring size = 4 or 5)). When L1 is a peptidomimetic linker, it is represented by the following formula
—Str—(PM)—Sp—,
wherein:
Str is a stretcher unit covalently attached to Ab;
Sp is a bond or spacer unit covalently attached to a PROTAC moiety; and
PM is a non-peptide chemical moiety selected from the group consisting of:
Figure imgf000100_0001
Figure imgf000100_0002
Figure imgf000100_0003
W is–NH-heterocycloalkyl- or heterocycloalkyl;
Y is heteroaryl, aryl, -C(O)C1-C6alkylene, C1-C6alkylene-NH2, C1-C6alkylene-NH-CH3, C1- C6alkylene-N-(CH3)2, C1-C6alkenyl or C1-C6alkylenyl;
each R1 is independently C1-C10alkyl, C1-C10alkenyl, (C1-C10alkyl)NHC(NH)NH2 or (C1- C10alkyl)NHC(O)NH2;
R3 and R2 are each independently H, C1-C10alkyl, C1-C10alkenyl, arylalkyl or heteroarylalkyl, or R3 and R2 together may form a C3-C7cycloalkyl; and
R4 and R5 are each independently C1-C10alkyl, C1-C10alkenyl, arylalkyl, heteroarylalkyl, (C1- C10alkyl)OCH2-, or R4 andR5 may form a C3-C7cycloalkyl ring.
It is noted that L1 may be connected to the PROTAC through any of the E3LB, L2, or PB groups.
In embodiments, Y is heteroaryl; R4 and R5 together form a cyclobutyl ring.
In embodiments, Y is a moiety selected from the group consisting of:
Figure imgf000100_0004
. In embodiments, Str is a chemical moiety represented by the following formula:
Figure imgf000101_0001
wherein R6 is selected from the group consisting of C1-C10alkylene, C1-C10alkenyl, C3- C8cycloalkyl, (C1-C8alkylene)O-, and C1-C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4- C7heterocycloalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each Ra is independently H or C1-C6alkyl; Sp is—Ar—Rb—, wherein Ar is aryl or heteroaryl, Rb is (C1-C10alkylene)O-.
In embodiments, Str has the formula:
Figure imgf000101_0002
wherein R7 is selected from C1-C10alkylene, C1-C10alkenyl, (C1-C10alkylene)O-, N(Rc)−(C2-C6 alkylene)−N(Rc) and N(Rc)−(C2-C6alkylene); where each Rc is independently H or C1-C6 alkyl; Sp is—Ar—Rb—, wherein Ar is aryl or heteroaryl, Rb is (C1- C10alkylene)O- or Sp -C1-C6alkylene-C(O)NH-.
In embodiments, L1 is a non-peptide chemical moiety represented by the following formula
Figure imgf000101_0003
R1 is C1-C6alkyl, C1-C6alkenyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2; R3 and R2 are each independently H or C1-C10alkyl. In embodiments, L1 is a non-peptide chemical moiety represented by the following formula
Figure imgf000102_0001
R1 is C1-C6 alkyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2;
R4 and R5 together form a C3-C7cycloalkyl ring.
In embodiments, L1 is a non-peptide chemical moiety represented by the following formula
Figure imgf000102_0002
R1 is C1-C6alkyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2 and W is as defined above.
In some embodiments, the linker may be a peptidomimetic linker such as those described in WO2015/095227, WO2015/095124 or WO2015/095223.
b. Non-peptidomimetic Linkers
In an aspect, a Linker L1 forms a disulfide bond with the antibody. In an aspect, the linker has the structure:
Figure imgf000102_0003
,
wherein, R1 and R2 are independently selected from H and C1-C6 alkyl, or R1 and R2 form a 3, 4, 5, or 6-membered cycloalkyl or heterocyclyl group. The linker is covalently bound to an antibody and a PROTAC as follows:
Figure imgf000103_0001
. In one aspect the carbonyl group of the linker is connected to an amine group in the PROTAC. It is also noted that the sulfur atom connected to Ab is a sulfur group from a cysteine in the antibody. In another aspect, a linker L1 has a functionality that is capable of reacting with a free cysteine present on an antibody to form a covalent bond. Nonlimiting exemples of such reactive functionalities include maleimide, haloacetamides, α-haloacetyl, activated esters such as succinimide esters, 4-nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates, and isothiocyanates. See, e.g., the conjugation method at page 766 of Klussman, et al (2004), Bioconjugate Chemistry 15(4):765-773, and the Examples herein.
In some embodiments, a linker has a functionality that is capable of reacting with an electrophilic group present on an antibody. Examples of such electrophilic groups include, but are not limited to, aldehyde and ketone carbonyl groups. In some embodiments, a heteroatom of the reactive functionality of the linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit. Nonlimiting examples of such reactive functionalities include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide.
A linker may comprise one or more linker components. Exemplary linker components include 6-maleimidocaproyl (“MC”), maleimidopropanoyl (“MP”), valine-citrulline (“val- cit” or“vc”), alanine-phenylalanine (“ala-phe”), p-aminobenzyloxycarbonyl (a“PAB”), N- Succinimidyl 4-(2-pyridylthio) pentanoate (“SPP”), and 4-(N-maleimidomethyl)
cyclohexane-1 carboxylate (“MCC”). Various linker components are known in the art, some of which are described below.
A linker may be a“cleavable linker,” facilitating release of a PROTAC. Nonlimiting exemplary cleavable linkers include acid-labile linkers (e.g., comprising hydrazone), protease-sensitive (e.g., peptidase-sensitive) linkers, photolabile linkers, or disulfide- containing linkers (Chari et al., Cancer Research 52:127-131 (1992); US 5208020).
In certain embodiments, a linker has the following Formula:
Figure imgf000104_0002
wherein A is a“stretcher unit”, and a is an integer from 0 to 1; W is an“amino acid unit”, and w is an integer from 0 to 12; Y is a“spacer unit”, and y is 0, 1, or 2. Exemplary embodiments of such linkers are described in U.S. Patent No.7,498,298.
In some embodiments, a linker component comprises a“stretcher unit” that links an antibody to another linker component or to a PROTAC moiety. Nonlimiting exemplary stretcher units are shown below (wherein the wavy line indicates sites of covalent attachment to an antibody, PROTAC, or additional linker components):
Figure imgf000104_0001
3. PROTAC (“D”) Useful PROTACs have the general formula described above. Particular PROTACs are described in US 7,208,157, WO2013/106643, WO2013/106646, and WO2015/160845. PROTACs include those having the following components.
a. E3 Ubiquitin Ligases Binding Groups (E3LB)
E3 ubiquitin ligases (of which over 600 are known in humans) confer substrate specificity for ubiquitination. There are known ligands which bind to these ligases. As described herein, an E3 ubiquitin ligase binding group is a peptide or small molecule that can bind an E3 ubiquitin ligase.
Specific E3 ubiquitin ligases include: von Hippel-Lindau (VHL); cereblon, XIAP, E3A; MDM2; Anaphase-promoting complex (APC); UBR5 (EDD1); SOCS/ BC-box/ eloBC/ CUL5/ RING; LNXp80; CBX4; CBLL1; HACE1; HECTD1; HECTD2; HECTD3; HECW1; HECW2; HERC1; HERC2; HERC3; HERC4; HUWE1; ITCH; NEDD4; NEDD4L; PPIL2; PRPF19; PIAS1; PIAS2; PIAS3; PIAS4; RANBP2; RNF4; RBX1; SMURF1; SMURF2; STUB1; TOPORS; TRIP12; UBE3A; UBE3B; UBE3C; UBE4A; UBE4B; UBOX5; UBR5; WWP1; WWP2; Parkin; A20/TNFAIP3; AMFR/gp78; ARA54; beta-TrCP1/BTRC; BRCA1; CBL; CHIP/STUB1; E6; E6AP/UBE3A; F-box protein 15/FBXO15; FBXW7/Cdc4;
GRAIL/RNF128; HOIP/RNF31; cIAP-1/HIAP-2; cIAP-2/HIAP-1; cIAP (pan); ITCH/AIP4; KAP1; MARCH8;; Mind Bomb 1/MIB1; Mind Bomb 2/MIB2; MuRF1/TRIM63; NDFIP1; NEDD4; NleL; Parkin; RNF2; RNF4; RNF8; RNF168; RNF43; SART1; Skp2; SMURF2; TRAF-1; TRAF-2; TRAF-3; TRAF-4; TRAF-5; TRAF-6; TRIM5; TRIM21; TRIM32;
UBR5; and ZNRF3.
The following Tables 13-27 list certain E3 ligases.
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0002
A particular E3 ubiquitin ligase is von Hippel-Lindau (VHL) tumor suppressor, the substrate recognition subunit of the E3 ligase complex VCB, which also consists of elongins B and C, Cul2 and Rbxl. The primary substrate of VHL is Hypoxia Inducible Factor lα (HIF- lα), a transcription factor that upregulates genes such as the pro-angiogenic growth factor VEGF and the red blood cell inducing cytokine erythropoietin in response to low oxygen levels. Compounds that bind VHL may be hydroxyproline compounds such as those disclosed in WO2013/106643, and other compounds described in US2016/0045607, WO2014187777, US20140356322, and US 9,249,153.
Another particular E3 ubiquitin ligase is MDM2. Examples of small molecular binding compounds for MDM2 include the“nutlin” compounds, e.g., nutlin 3a and nutlin 3, having the structure:
Figure imgf000121_0001
.
Also included as MDM2 binding compounds are those described in WO2012/121361;
WO2014/038606; WO2010/082612; WO2014/044401; WO2009/151069; WO2008/072655;
Figure imgf000122_0001
Chem. Lett, 2013, 4, 466-469; J. Med. Chem., 2015, 58, 1038-1052; Bioorg. Med. Chem. Lett. 25 (2015) 3621-3625; Bioorg. Med. Chem. Lett.16 (2006) 3310-3314. Further specific examples of small molecular binding compounds for MDM2 contemplated for use with a PAC include RG7112, RG7388, MI 773/SAR 405838, AMG 232, DS-3032b, RO6839921, RO5045337, RO5503781, Idasanutlin, CGM-097, MK-8242
Another particular E3 ubiquitin ligase is X-linked inhibitor of apoptosis (XIAP). XIAP is a protein that stops apoptotic cell death. Deregulation of XIAP has been associated with cancer, neurodegenerative disorders and autoimmunity. In the development of lung cancer, the overexpression of XIAP inhibits caspases. In developing prostate cancer, XIAP is one of four IAPs overexpressed in the prostatic epithelium. Mutations in the XIAP gene can result in a severe and rare type of inflammatory bowel disease. Defects in the XIAP gene can also result in an extremely rare condition called X-linked lymphoproliferative disease.
Degradation of XIAP can enhance apoptosis by preventing XIAP from binding to caspases. This allows normal caspase activity to proceed.
Examples of small molecular binding compounds for XIAP include compounds disclosed in US 9,096,544; WO 2015187998; WO 2015071393; US 9,278,978; US 9,249,151; US 20160024055; US 20150307499; US 20140135270; US 20150284427; US 20150259359; US 20150266879; US 20150246882; US 20150252072; US 20150225449; US 8,883,771, J. Med. Chem., 2015, 58(16) 6574-6588 and Small-molecule Pan-IAP Antagonists: A Patent Review (2010) Expert Opin Ther Pat; 20: 251-67 (Flygare & Fairbrother). Specific compounds include all the tetrahydro-benzodiazinone compounds of the following formula:
Figure imgf000123_0001
,
as disclosed in WO 2015/071393. Other small molecular binding compounds for XIAP include AEG35156, Embelin, TWX006 and TWX024. When an XIAP bindig moeity is used as part of a PROTAC, the XIAP binding moiety can bind to the BIR2 or BIR3 domain of XIAP or both.
Another particular E3 ubiquitin ligase is cereblon. Cereblon is a protein that forms an E3 ubiquitin ligase complex with damaged DNA binding protein 1 (DDB1), Cullin-4A (CUL4A), and regulator of cullins 1 (ROC1). This complex ubiquitinates a number of other proteins. Cereblon ubquitination of target proteins results in increased levels of fibroblast growth factor 8 (FGF8) and fibroblast growth factor 10 (FGF10). FGF8 in turn regulates a number of developmental processes, such as limb and auditory vesicle formation. In the absence of cereblon, DDB1 forms a complex with DDB2 that functions as a DNA damage- binding protein. Thalidomide, lenalidomide, pomalidomide and analogs thereof are known to bind to cereblon. The crystal structure of cereblon with thalidomide and derivative compounds are described in US2015/0374678. Other small molecule compounds that bind to cereblon are also known, e.g., the compounds disclosed as an in US2016/0058872 and US2015/0291562. Further, phthalimide conjugation with binders, such as antagonists, of BET bromodomains can provide PROTACs with highly-selective cereblon-dependent BET protein degradation. Winter et al., Science, June 19, 2015, p.1376. Such PROTACs can be conjugated to an antibody as described herein to form a PAC.
b. Protein Binding Group (PB)
The PB component is a group which binds to a target protein intended to be degraded. The term "protein" includes oligopeptides and polypeptide sequences of sufficient length that they can bind to a PB group. Any protein in a eukaryotic system or a microbial system, including a virus, bacteria or fungus, as otherwise described herein, are targets for ubiquitination mediated by the compounds described herein.
PB groups include, for example, any moiety which binds to a protein specifically (binds to a target protein) and includes the following non-limiting examples of small molecule target protein moieties: Hsp90 inhibitors, kinase inhibitors, MDM2 inhibitors, compounds targeting Human BET Bromodomain- containing proteins, HDAC inhibitors, human lysine methyltransferase inhibitors, angiogenesis inhibitors, immunosuppressive compounds, and compounds targeting the aryl hydrocarbon receptor (AHR), among numerous others. The compositions described below exemplify some of the members of these nine types of small molecule target protein binding moieties. Such small molecule target protein binding moieties also include pharmaceutically acceptable salts, enantiomers, solvates and polymorphs of these compositions, as well as other small molecules that may target a protein of interest.
In general, target proteins may include, for example, structural proteins, receptors, enzymes, cell surface proteins, proteins pertinent to the integrated function of a cell, including proteins involved in catalytic activity, aromatase activity, motor activity, helicase activity, metabolic processes (anabolism and catrabolism), antioxidant activity, proteolysis, biosynthesis, proteins with kinase activity, oxidoreductase activity, transferase activity, hydrolase activity, lyase activity, isomerase activity, ligase activity, enzyme regulator activity, signal transducer activity, structural molecule activity, binding activity (protein, lipid carbohydrate), receptor activity, cell motility, membrane fusion, cell communication, regulation of biological processes, development, cell differentiation, response to stimulus, behavioral proteins, cell adhesion proteins, proteins involved in cell death, proteins involved in transport (including protein transporter activity, nuclear transport, ion transporter activity, channel transporter activity, carrier activity, permease activity, secretion activity, electron transporter activity, pathogenesis, chaperone regulator activity, nucleic acid binding activity, transcription regulator activity, extracellular organization and biogenesis activity, translation regulator activity. Proteins of interest can include proteins from eurkaryotes and prokaryotes including humans as targets for drug therapy, other animals, including domesticated animals, microbials for the determination of targets for antibiotics and other antimicrobials and plants, and even viruses, among numerous others.
Accordingly, the PB component of a PAC is any peptide or small molecule that bind protein targets such as FoxOl, HDAC, DP-1, E2F, ABL, AMPK, BRK, BRSK I, BRSK2, BTK, CAMKK1, CAMKK alpha, CAMKK beta, Rb, Suv39HI, SCF, p19INK4D, GSK-3, pi 8 INK4, myc, cyclin E, CDK2, CDK9, CDG4/6, Cycline D, pl6 INK4A, cdc25A, BMI1, SCF, Akt, CHKl/2, C 1 delta, CK1 gamma, C 2, CLK2, CSK, DDR2, DYRK1A/2/3, EF2K, EPH-A2/A4/B1/B2/B3/B4, EIF2A 3, Smad2, Smad3, Smad4, Smad7, p53, p21 Cipl, PAX, Fyn, CAS, C3G, SOS, Tal, Raptor, RACK-1, CRK, Rapl, Rac, KRas, NRas, HRas, GRB2, FAK, PI3K, spred, Spry, mTOR, MPK, LKBl, PAK 1/2/4/5/6, PDGFRA, PYK2, Src, SRPK1, PLC, PKC, PKA, PKB alpha/beta, PKC alpha/gamma/zeta, PKD, PLKl, PRAK, PRK2, WAVE-2, TSC2, DAPKl, BAD, IMP, C-TAK1, TAKl, TAOl, TBK1, TESK1, TGFBR1, TIE2, TLK1, TrkA, TSSK1, TTBK1/2, TTK, Tpl2/cotl, MEK1, MEK2, PLDL Erkl, Erk2, Erk5, Erk8, p90RSK, PEA-15, SRF, p27 KIP1, TIF la, HMGN1, ER81, MKP-3, c-Fos, FGF-R1, GCK, GSK3 beta, HER4, HIPK1/2/3/, IGF-1R, cdc25, UBF, LAMTOR2, Statl, StaO,CREB, JAK, Src, PTEN, NF-kappaB, HECTH9, Bax, HSP70, HSP90, Apaf-1, Cyto c, BCL-2, Bcl-xL, Smac, XIAP, Caspase-9, Caspase-3, Caspase-6, Caspase-7, CDC37, TAB, IKK, TRADD, TRAF2, R1P1, FLIP, TAKl, JNKl/2/3, Lck, A-Raf, B-Raf, C-Raf, MOS, MLKl/3, MN l/2, MSKl, MST2/3/4, MPSK1, MEKKl , ME K4, MEL , ASK1, MINK1 , MKK 1 /2/3/4/6/7, NE 2a/6/7, NUAK1, OSR1, SAP , STK33, Syk, Lyn, PDK1, PHK, PIM 1/2/3, Ataxin- 1, mTORCl, MDM2, p21 Wafl , Cyclin Dl, Lamln A, Tpl2, Myc, catenin, Wnt, IKK-beta, IKK- gamma, IKK-alpha, IKK-epsilon, ELK, p65RelA, IRAKI, IRA 2, IRAK4, IRR, FADD, TRAF6, TRAF3, MKK3, MKK6, ROCK2, RSK1/2, SGK 1, SmMLCK, SIK2/3, ULK1/2, VEGFR1, WNK l , YES1, ZAP70, MAP4K3, MAP4K5, MAPKlb, MAPKAP-K2 K3, p38 alpha/beta/delta/gamma MAPK, Aurora A, Aurora B, Aurora C, MCAK, Clip, MAPKAPK, FAK, MARK 1 /2/3/4, Mucl , SHC, CXCR4, Gap-1, Myc, beta-catenin/TCF, Cbl, BRM, Mcl-1, BRD2, BRD3, BRD4, AR, RAS, ErbB3, EGFR, IRE1, HPK1, RIPK2, and ERα, including all variants, mutations, splice variants, indels and fusions of these target proteins listed.
Specific PB groups are small molecule compounds such as those disclosed in
US2014/0356322 and US2016/0045607. Compounds disclosed therein can be classified as Heat Shock Protein 90 (HSP90) inhibitors, Kinase and Phosphatase inhibitors, MDM2 inhibitors, HDAC inhibitors, Human Lysine Methyltransferase Inhibitors, Angiogenesis inhibitors, Immunosuppressive compounds, as well as compounds that target: Human BET Bromodomain-containing proteins, the aryl hydrocarbon receptor (AHR), REF receptor kinase, FKBP, Androgen Receptor (AR), Estrogen receptor (ER), Thyroid Hormone
Receptor, HIV Protease, HIV Integrase, HCV Protease, Acyl-protein Thioesterase-1 and -2 (APT1 and APT2).
c. Linker L2
The E3LB and PB groups of PROTACs as described herein can be connected with linker (L2). In certain embodiments, the linker group L2 is a group comprising one or more covalently connected structural units of A (e.g., -A1... Aq-), wherein A1 is a group coupled to at least one of a E3LB, a PB, or a combination thereof. In certain embodiments, A1 links a E3LB, a PB, or a combination thereof directly to another E3LB, PB, or combination thereof. In other embodiments, A1 links a EL3B, a PB, or a combination thereof indirectly to another E3LB, PB, or combination thereof through Aq.
In certain embodiments, A1 to Aq are, each independently, a bond, CRLaRLb, O, S, SO, SO2, NRLc, SO2NRLc, SONRLc, CONRLc, NRLcCONRLd, NRLcSO2NRLd, CO, CRLa═CRLb, C≡C, SiRLaRLb, P(O)RLa, P(O)ORLa, NRLcC(═NCN)NRLd, NRLcC(═NCN),
NRLcC(═CNO2)NRLd, C3-11cycloalkyl optionally substituted with 0-6 RLa and/or RLb groups, C3-11heterocyclyl optionally substituted with 0-6 RLa and/or RLb groups, aryl optionally substituted with 0-6 RLa and/or RLb groups, heteroaryl optionally substituted with 0-6 RLa and/or RLb groups, where RLa or RLb, each independently, can be linked to other A groups to form cycloalkyl and/or heterocyclyl moeity which can be further substituted with 0-4 RLe groups; wherein RLa, RLb, RLc, RLd and RLe are, each independently, H, halo, C1-8alkyl, OC1- 8alkyl, SC1-8alkyl, NHC1-8alkyl, N(C1-8alkyl)2, C3-11cycloalkyl, aryl, heteroaryl, C3- 11heterocyclyl, OC1-8cycloalkyl, SC1-8cycloalkyl, NHC1-8cycloalkyl, N(C1-8cycloalkyl)2, N(C1-8cycloalkyl)(C1-8alkyl), OH, NH2, SH, SO2C1-8alkyl, P(O)(OC1-8alkyl)(C1-8alkyl), P(O)(OC1-8alkyl)2, CC—C1-8alkyl, CCH, CH═CH(C1-8alkyl), C(C1-8alkyl)═CH(C1-8alkyl), C(C1-8alkyl)═C(C1-8alkyl)2, Si(OH)3, Si(C1-8alkyl)3, Si(OH)(C1-8alkyl)2, COC1-8alkyl, CO2H, halogen, CN, CF3, CHF2, CH2F, NO2, SF5, SO2NHC1-8alkyl, SO2N(C1-8alkyl)2, SONHC1- 8alkyl, SON(C1-8alkyl)2, CONHC1-8alkyl, CON(C1-8alkyl)2, N(C1-8alkyl)CONH(C1-8alkyl), N(C1-8alkyl)CON(C1-8alkyl)2, NHCONH(C1-8alkyl), NHCON(C1-8alkyl)2, NHCONH2, N(C1- 8alkyl)SO2NH(C1-8alkyl), N(C1-8alkyl) SO2N(C1-8alkyl)2, NH SO2NH(C1-8alkyl), NH
SO2N(C1-8alkyl)2, NH SO2NH2.
In certain embodiments, q is an integer greater than or equal to 0. In certain embodiments, q is an integer greater than or equal to 1.
In certain embodiments, e.g., where q is greater than 2, Aq is a group which is connected to an E3LB moiety, and A1 and Aq are connected via structural units of A (number of such structural units of A: q-2).
In certain embodiments, e.g., where q is 2, Aq is a group which is connected to A1 and to an E3LB moiety.
In certain embodiments, e.g., where q is 1, the structure of the linker group L2 is -A1-, and A1 is a group which is connected to an E3LB moiety and a PB moiety.
In additional embodiments, q is an integer from 1 to 100, 1 to 90, 1 to 80, 1 to 70, 1 to 60, 1 to 50, 1 to 40, 1 to 30, 1 to 20, or 1 to 10.
In certain embodiments, the linker (L2) is selected from the group consisting of):
Figure imgf000128_0001
Figure imgf000129_0001
In additional embodiments, the linker group is an optionally substituted
(poly)ethyleneglycol having between 1 and about 100 ethylene glycol units, between about 1 and about 50 ethylene glycol units, between 1 and about 25 ethylene glycol units, between about 1 and 10 ethylene glycol units, between 1 and about 8 ethylene glycol units and 1 and 6 ethylene glycol units, between 2 and 4 ethylene glycol units, or optionally substituted alkyl groups interdispersed with optionally substituted, O, N, S, P or Si atoms. In certain embodiments, the linker is substituted with an aryl, phenyl, benzyl, alkyl, alkylene, or heterocycle group. In certain embodiments, the linker may be asymmetric or symmetrical.
In any of the embodiments of the compounds described herein, the linker group may be any suitable moiety as described herein. In one embodiment, the linker is a substituted or unsubstituted polyethylene glycol group ranging in size from about 1 to about 12 ethylene glycol units, between 1 and about 10 ethylene glycol units, about 2 about 6 ethylene glycol units, between about 2 and 5 ethylene glycol units, between about 2 and 4 ethylene glycol units. Although the E3LB group and PB group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker. The linker is independently covalently bonded to the E3LB group and the PB group preferably through an amide, ester, thioester, keto group, carbamate (urethane), carbon or ether, each of which groups may be inserted anywhere on the E3LB group and PB group to provide maximum binding of the E3LB group on the ubiquitin ligase and the PB group on the target protein to be degraded. In certain aspects where the PB group is an E3LB group, the target protein for degradation may be the ubiquitin ligase itself. In certain aspects, the linker may be linked to an optionally substituted alkyl, alkylene, alkene or alkyne group, an aryl group or a heterocyclic group on the E3LB and/or PB groups. It is noted that an E3LB group or a PB group may need to be derivatized to make a chemical functional group that is reactive with a chemical functional group on the linker. Alternatively, the linker may need to be derivatized to include a chemical functional group that can react with a functional group found on E3LB and/or PB.
L2 can also be represented by the formula:
Figure imgf000130_0001
,
Where Z is a group which links E3LB to X; and X is a group linking Z to group PB. In embodiments, Z is absent (a bond), -(CH2)i-O, -(CH2)i-S, -(CH2)i-N-R, a (CH2)i- X1Y1 group wherein X1Y1 forms an amide group, or a urethane group, ester or thioester group, or a
Figure imgf000130_0002
where, each R is H, or a C1-C3 alkyl, an alkanol group or a heterocycle (including a water soluble heterocycle, preferably, a morpholino , piperidine or piperazine group to promote water solubility of the linker group); each Y is independently a bond, O, S or N-R; and each i is independently 0 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
In embodiments, X is a
Figure imgf000131_0001
,
where each V is independently a bond (absent),
Figure imgf000131_0002
Figure imgf000131_0003
j is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
k is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5; preferably k is 1, 2, 3, 4, or 5;
m' is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, 1 to 8, 1 to 6, 1, 2, 3, 4 or 5;
n is 1 to 100, 1 to 75, 1 to 60, 1 to 55, 1 to 50, 1 to 45, 1 to 40, 2 to 35, 3 to 30, 1 to 15, 1 to 10, l to 8, 1 to 6, 1 , 2, 3, 4 or 5;
X1 is O, S or N-R, preferably O;
Y is the same as above;
and CON is a connector group (which may be a bond) which connects Z to X, when present in the linker group.
In embodiments, CON is a bond (absent), a heterocycle including a water soluble heterocycle such as a piperazinyl or other group or a group,
Figure imgf000132_0001
,
where X2 is O, S, NR4, S(O), S(O)2, -S(O)2O, -OS(O)2, or OS(O)2O;
X3 is O, S, CHR4, NR4; and
R is H or a C1-C3 alkyl group optionally substituted with one or two hydroxyl groups, or a pharmaceutically acceptable salt, enantiomer or stereoisomer thereof.
In alternative preferred aspects, the linker group is a (poly)ethyleneglycol having between 1 and about 100 ethylene glycol units, between about 1 and about 50 ethylene glycol units, between 1 and about 25 ethylene glycol units, between about 1 and 10 ethylene glycol units, between 1 and about 8 ethylene glycol units and 1 and 6 ethylene glycol units, between 2 and 4 ethylene glycol units.
In embodiments, CON is
Figure imgf000132_0002
,
or an amide group.
Although the E3LB group and PB group may be covalently linked to the linker group through any group which is appropriate and stable to the chemistry of the linker, in preferred aspects, the linker is independently covalently bonded to the E3LB group and the PB group through an amide, ester, thioester, keto group, carbamate (urethane) or ether, each of which groups may be inserted anywhere on the E3LB group and PB group to allow binding of the E3LB group to the ubiquitin ligase and the PB group to the target protein to be degraded. In other words, as shown herein, the linker can be designed and connected to E3LB and PB to minimize, eliminate, or neutralize any impact its presence might have on the binding of E3LB and PB to their respective binding partners. In certain aspects, the targeted protein for degradation may be an ubiquitin ligase.
Additional linkers L2 are disclosed in US Application Publication Nos.
2016/0058872; 2016/0045607; 2014/0356322; and 2015/0291562, and WO2014/063061.
Refering now to a PAC, a PAC can comprise a single antibody where the single antibody can have more than one PROTAC, each PROTAC covalently linked to the antibody through a linker L1. The“PROTAC loading” is the average number of PROTAC moieties per antibody. PROTAC loading may range from 1 to 8 PROTAC (D) per antibody (Ab). That is, in the PAC formula, Ab―(L1―D)p, p has a value from about 1 to about 50, from about 1 to about 8, from about 1 to about 5, from about 1 to about 4, or from about 1 to about 3. Each PROTAC covalently linked to the antibody through linker L1 can be the same or different PROTAC and can have a linker of the same type or different type as any other L1 covalently linked to the antibody. In one embodiment, Ab is a cysteine engineered antibody and p is about 2.
The average number of PROTACs per antibody in preparations of PACs from conjugation reactions may be characterized by conventional means such as mass
spectrometry, ELISA assay, electrophoresis, and HPLC. The quantitative distribution of PACs in terms of p may also be determined. By ELISA, the averaged value of p in a particular preparation of PAC may be determined (Hamblett et al (2004) Clin. Cancer Res. 10:7063-7070; Sanderson et al (2005) Clin. Cancer Res.11:843-852). However, the distribution of the value of p is not discernible by the antibody-antigen binding and detection limitation of ELISA. Also, ELISA assay for detection of PACs does not determine where the PROTAC moieties are attached to the antibody, such as the heavy chain or light chain fragments, or the particular amino acid residues. In some instances, separation, purification, and characterization of homogeneous PACs where p is a certain value from PACs with other PROTAC loadings may be achieved by means such as reverse phase HPLC or
electrophoresis.
For some PACs, p may be limited by the number of attachment sites on the antibody. For example, an antibody may have only one or several cysteine thiol groups, or may have only one or several sufficiently reactive thiol groups through which a linker may be attached. Another reactive site on an Ab to connect L1-Ds are the amine functional group of lysine residues. Values of p include values from about 1 to about 50, from about 1 to about 8, from about 1 to about 5, from about 1 about 4, from about 1 to about 3, and where p is equal to 2. In some embodiments, the subject matter described herein is directed to any the PACs, wherein p is about 1, 2, 3, 4, 5, 6, 7, or 8.
Generally, fewer than the theoretical maximum of PROTAC moieties is conjugated to an antibody during a conjugation reaction. An antibody may contain, for example, many lysine residues that do not react with the linker L1-PROTAC group (L1-D) or linker reagent. Only the most reactive lysine groups may react with an amine-reactive linker reagent. Also, only the most reactive cysteine thiol groups may react with a thiol-reactive linker reagent or linker L1-PROTAC group. Generally, antibodies do not contain many, if any, free and reactive cysteine thiol groups which may be linked to a PROTAC moiety. Most cysteine thiol residues in the antibodies of the compounds exist as disulfide bridges and must be reduced with a reducing agent such as dithiothreitol (DTT) or TCEP, under partial or total reducing conditions. However, the PROTAC loading (PROTAC/antibody ratio,“PAR”) of a PAR may be controlled in several different manners, including: (i) limiting the molar excess of linker L1-PROTAC group or linker reagent relative to antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) partial or limiting reductive conditions for cysteine thiol modification.
III. L1-PROTAC Compounds
The PROTACs described herein can be covalently linked to a linker L1 to prepare L1- PROTAC groups. These compounds have the following general formula:
(L1―D),
wherein, D is a PROTAC having the structure E3LB―L2―PB; wherein, E3LB is an E3 ligase binding group covalently bound to L2; L2 is a linker covalently bound to E3LB and PB; PB is a protein binding group covalently bound to L2; and L1 is a linker, covalently bound to D. Useful groups for each of these components is as described above.
In particular embodiments, L1 is as described elsewhere herein, including a peptidomimetic linker. In these embodiments, the L1-PROTAC has the following formula:
Figure imgf000135_0001
wherein
Str is a stretcher unit;
Sp is a bond or a spacer unit covalently attached to D, i.e., a PROTAC moiety;
R1 is C1-C10alkyl, (C1-C10alkyl)NHC(NH)NH2 or (C1-C10alkyl)NHC(O)NH2;
R4 and R5 are each independently C1-C10alkyl, arylalkyl, heteroarylalkyl, (C1-C10alkyl )OCH2-, or R4 and R5 may form a C3-C7cycloalkyl ring;
D is a PROTAC moiety.
An L1-PROTAC compound can be represented by the following formula:
Figure imgf000135_0002
, wherein R6 is C1-C10alkylene; R4 and R5 together form a C3-C7cycloalkyl ring, and D is a PROTAC moeity.
An L1-PROTAC compound can be represented by the following formula:
Figure imgf000135_0003
, wherein R1, R4 and R5 are as described elsewhere herein, and D is a PROTAC moiety.
An L1-PROTAC com ound can be re resented b the following formula:
Figure imgf000135_0004
wherein
Str is a stretcher unit;
Sp is an optional spacer unit covalently attached to D, i.e., a PROTAC moiety;
Y is heteroaryl, aryl, -C(O)C1-C6alkylene, C1-C6alkylene-NH2, C1-C6alkylene-NH-CH3, C1- C6alkylene-N-(CH3)2, C1-C6alkenyl or C1-C6alkylenyl; R1 is C1-C10alkyl, (C1-C10alkyl)NHC(NH)NH2 or (C1-C10alkyl)NHC(O)NH2;
R3 and R2 are each independently H, C1-C10alkyl, arylalkyl or heteroarylalkyl, or R3 and R2 together may form a C3-C7cycloalkyl; and
D is a PROTAC moiety.
An L1-PROTAC compound can be represented by the following formula:
Figure imgf000136_0001
wherein, R6 is C1-C10alkylene, and R1, R2 and R3 are as described elsewhere herein, and D is a PROTAC moiety
An L1-PROTAC compound can be represented by the following formula:
Figure imgf000136_0002
wherein R1, R2 and R3 are as described elsewhere herein, and D is a PROTAC moiety.
In any of the above L1-PROTAC compounds, Str can have the following formula:
Figure imgf000136_0003
wherein R6 is selected from the group consisting of C1-C10alkylene, C3-C8cycloalkyl, O-(C1- C8alkylene), and C1-C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo,
trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4-C7heterocycloalkyl aryl, arylalkyl, heteroarylalkyl and heteroaryl; each Ra is independently H or C1-C6alkyl; Sp is—Ar—Rb—, wherein Ar is aryl or heteroaryl, Rb is (C1-C10alkylene)O-. In certain L1-PROTAC compounds, R6 is C1-C10alkylene, Sp is—Ar—Rb—, wherein Ar is aryl Rb is (C1-C6alkylene)O-; or R6 is–(CH2)q is 1-10;
In any of the above L1-PROTAC compounds, Str can have the following formula:
Figure imgf000137_0001
,
wherein, indicates a moiety capable of conjugating to an antibody, R7 is selected from C1-C10alkylene, C1-C10alkylene−O, N(Rc)−(C2-C6 alkylene)−N(Rc) and N(Rc)−(C2- C6alkylene); where each Rc is independently H or C1-C6 alkyl;
Sp is—Ar—Rb—, wherein Ar is aryl or heteroaryl, Rb is (C1-C10 alkylene)O-; or wherein R6 is C1-C10 alkylene, Sp is—Ar—Rb—, wherein Ar is aryl Rb is (C1-C6 alkylene)O-.
An L1-PROTAC can have the following formulae, wherein in each instance, D is a PROTAC moiety:
Figure imgf000137_0002
Referring now to the PB group of the PROTAC, in particular embodiments, PB is as described elsewhere herein, or is selected from the group consisting of Heat Shock Protein 90 (HSP90) inhibitors, Kinase and Phosphatase inhibitors, MDM2 inhibitors, HDAC inhibitors, Human Lysine Methyltransferase Inhibitors, Angiogenesis inhibitors, Immunosuppressive compounds, as well as compounds that target: Human BET Bromodomain-containing proteins, the aryl hydrocarbon receptor (AHR), REF receptor kinase, FKBP, Andrpgen Receptor (AR), Estrogen receptor (ER), Thyroid Hormone Receptor, HIV Protease, HIV Integrase, HCV Protease, Acyl-protein Thioesterase-1 and -2 (APT1 and APT2).
In particular embodiments, E3LB is as described elsewhere herein, including a group that binds XIAP, VHL, cereblon and MDM2.
The subject matter described herein is also directed to methods of preparing a PAC from a L1-PROTAC compound, the method comprising contacting an antibody, or variants, mutations, splice variants, indels and fusions thereof, with a L1-PROTAC under conditions where the antibody is covalently bound to any available point of attachment on a L1- PROTAC, wherein a PAC is prepared. The subject matter described herein is also directed to methods of preparing a PAC from an Ab-L1 portion, i.e., an antibody, or variants, mutations, splice variants, indels and fusions thereof, covalently attached to a L1, the methods comprising contacting a PROTAC with an Ab-L1 under conditions where the PROTAC is covalently bound to any available point of attachment on the Ab-L1, wherein a PAC is prepared. The methods can further comprise routine isolation and purification of the PACs.
Referring now to a PAC and a L1-PROTAC compound, as described herein, these can exist in solid or liquid form. In the solid state, it may exist in crystalline or noncrystalline form, or as a mixture thereof. The skilled artisan will appreciate that pharmaceutically acceptable solvates may be formed for crystalline or non-crystalline compounds. In crystalline solvates, solvent molecules are incorporated into the crystalline lattice during crystallization. Solvates may involve non-aqueous solvents such as, but not limited to, ethanol, isopropanol, DMSO, acetic acid, ethanolamine, or ethyl acetate, or they may involve water as the solvent that is incorporated into the crystalline lattice. Solvates wherein water is the solvent incorporated into the crystalline lattice are typically referred to as "hydrates." Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water. The subject matter described herein includes all such solvates. The skilled artisan will further appreciate that certain compounds and PACs described herein that exist in crystalline form, including the various solvates thereof, may exhibit polymorphism (i.e. the capacity to occur in different crystalline structures). These different crystalline forms are typically known as "polymorphs." The subject matter disclosed herein includes all such polymorphs. Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification. The skilled artisan will appreciate that different polymorphs may be produced, for example, by changing or adjusting the reaction conditions or reagents, used in making the compound. For example, changes in temperature, pressure, or solvent may result in polymorphs. In addition, one polymorph may spontaneously convert to another polymorph under certain conditions.
Compounds and PACs described herein or a salt thereof may exist in stereoisomeric forms (e.g., it contains one or more asymmetric carbon atoms). The individual stereoisomers (enantiomers and diastereomers) and mixtures of these are included within the scope of the subject matter disclosed herein. Likewise, it is understood that a compound or salt of Formula (I) may exist in tautomeric forms other than that shown in the formula and these are also included within the scope of the subject matter disclosed herein. It is to be understood that the subject matter disclosed herein includes all combinations and subsets of the particular groups described herein. The scope of the subject matter disclosed herein includes mixtures of stereoisomers as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures. It is to be understood that the subject matter disclosed herein includes all combinations and subsets of the particular groups defined hereinabove.
The subject matter disclosed herein also includes isotopically-labelled forms of the compounds described herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds described herein and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulphur, fluorine, iodine, and chlorine, such as 2H, 3H, 11C, 13C, 14C, 15N, 17O, 18O, 31P, 32P, 35S, 18F, 36Cl, 123I and 125I. Compounds and PACs as disclosed herein and pharmaceutically acceptable salts thereof that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of the subject matter disclosed herein. Isotopically-labelled compounds are disclosed herein, for example those into which radioactive isotopes such as 3H, 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are commonly used for their ease of preparation and detectability.11C and 18F isotopes are useful in PET (positron emission tomography), and 125I isotopes are useful in SPECT (single photon emission computerized tomography), all useful in brain imaging. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labelled compounds of formula I can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples below, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
The subject matter described herein includes the following embodiments:
1. A conjugate having the chemical structure
Ab―(L1―D)p,
wherein,
D is a PROTAC having the structure E3LB―L2―PB;
E3LB is an E3 ligase binding group covalently bound to L2;
L2 is a linker covalently bound to E3LB and PB;
PB is a protein binding group covalently bound to L2;
Ab is an antibody covalently bound to L1;
L1 is a linker, covalently bound to Ab and to D;
p has a value from about 1 to about 8.
2. The conjugate of embodiment 1, wherein E3LB is a group that binds an E3 ligase, wherein the E3 ligase is listed in Tables 13-27, e.g., Table 13, Table 14, Table 15, Table 16, Table 17, Table 18, Table 19, Table 20, Table 21, Table 22, Table 23, Table 24, Table 25, Table 26, or Table 27.
3. The conjugate of any above embodiment, wherein E3LB is a group that binds an E3 ligase, wherein the E3 ligase is selected from the group consisting of von Hippel-Lindau (VHL); cereblon; XIAP; E3A; MDM2; Anaphase-promoting complex (APC); UBR5
(EDD1); SOCS/ BC-box/ eloBC/ CUL5/ RING; LNXp80; CBX4; CBLL1; HACE1;
HECTD1; HECTD2; HECTD3; HECW1; HECW2; HERC1; HERC2; HERC3; HERC4; HUWE1; ITCH; NEDD4; NEDD4L; PPIL2; PRPF19; PIAS1; PIAS2; PIAS3; PIAS4;
RANBP2; RNF4; RBX1; SMURF1; SMURF2; STUB1; TOPORS; TRIP12; UBE3A;
UBE3B; UBE3C; UBE4A; UBE4B; UBOX5; UBR5; WWP1; WWP2; Parkin;
A20/TNFAIP3; AMFR/gp78; ARA54; beta-TrCP1/BTRC; BRCA1; CBL; CHIP/STUB1; E6; E6AP/UBE3A; F-box protein 15/FBXO15; FBXW7/Cdc4; GRAIL/RNF128;
HOIP/RNF31; cIAP-1/HIAP-2; cIAP-2/HIAP-1; cIAP (pan); ITCH/AIP4; KAP1; MARCH8; MDM2/HDM2; Mind Bomb 1/MIB1; Mind Bomb 2/MIB2; MuRF1/TRIM63; NDFIP1; NEDD4; NleL; Parkin; RNF2; RNF4; RNF8; RNF168; RNF43; SART1; Skp2; SMURF2; TRAF-1; TRAF-2; TRAF-3; TRAF-4; TRAF-5; TRAF-6; TRIM5; TRIM21; TRIM32;
UBR5; and ZNRF3.
4. The conjugate of any above embodiment, wherein E3LB is a group that binds an E3 ligase selected from the group consisting of XIAP, VHL, cereblon and MDM2.
5. The conjugate of any above embodiment, wherein E3LB is selected from the group consisting of a compound that binds VHL, a hydroxyproline compound that binds VHL, a compound that binds MDM2, a compound that binds cereblon, a tetrahhydro- benzodiazepinone, a nutlin, and small molecular binding compounds as described herein. 6. The conjugate of any above embodiment, wherein E3LB is a XIAP inhibitor that is a tetrahydro-benzodiazepinone having the formula:
Figure imgf000141_0001
,
wherein, R1, R2, R3, R4 and R5 are as described in WO/2015/071393, and including all compounds therein.
7. The conjugate of any above embodiment, wherein PB is a group that binds FoxOl, HDAC, DP-1, E2F, ABL, AMPK, BRK, BRSK I, BRSK2, BTK, CAMKK1, CAMKK alpha, CAMKK beta, Rb, Suv39HI, SCF, p19INK4D, GSK-3, pi 8 INK4, myc, cyclin E, CDK2, CDK9, CDG4/6, Cycline D, pl6 INK4A, cdc25A, BMI1, SCF, Akt, CHKl/2, C 1 delta, CK1 gamma, C 2, CLK2, CSK, DDR2, DYRK1A/2/3, EF2K, EPH-A2/A4/B1/B2/B3/B4, EIF2A 3, Smad2, Smad3, Smad4, Smad7, p53, p21 Cipl, PAX, Fyn, CAS, C3G, SOS, Tal, Raptor, RACK-1, CRK, Rapl, Rac, KRas, NRas, HRas, GRB2, FAK, PI3K, spred, Spry, mTOR, MPK, LKBl, PAK 1/2/4/5/6, PDGFRA, PYK2, Src, SRPK1, PLC, PKC, PKA, PKB alpha/beta, PKC alpha/gamma/zeta, PKD, PLKl, PRAK, PRK2, RIPK2, WAVE-2, TSC2, DAPKl, BAD, IMP, C-TAK1, TAKl, TAOl, TBK1, TESK1, TGFBR1, TIE2, TLK1, TrkA, TSSK1, TTBK1/2, TTK, Tpl2/cotl, MEK1, MEK2, PLDL Erkl, Erk2, Erk5, Erk8, p90RSK, PEA- 15, SRF, p27 KIP1, TIF la, HMGN1, ER81, MKP-3, c-Fos, FGF-R1, GCK, GSK3 beta, HER4, HIPK1/2/3/, IGF-1R, cdc25, UBF, LAMTOR2, Statl, StaO,CREB, JAK, Src, PTEN, NF- kappaB, HECTH9, Bax, HSP70, HSP90, Apaf-1, Cyto c, BCL-2, Bcl-xL, Smac, XIAP, Caspase-9, Caspase-3, Caspase-6, Caspase-7, CDC37, TAB, IKK, TRADD, TRAF2, R1P1, FLIP, TAKl, JNKl/2/3, Lck, A-Raf, B-Raf, C-Raf, MOS, MLKl/3, MN l/2, MSKl, MST2/3/4, MPSK1, MEKKl , ME K4, MEL , ASK1, MINK1 , MKK 1 /2/3/4/6/7, NE 2a/6/7, NUAK1, OSR1, SAP , STK33, Syk, Lyn, PDK1, PHK, PIM 1/2/3, Ataxin- 1, mTORCl, MDM2, p21 Wafl , Cyclin Dl, Lamln A, Tpl2, Myc, catenin, Wnt, IKK-beta, IKK- gamma, IKK-alpha, IKK-epsilon, ELK, p65RelA, IRAKI, IRA 2, IRAK4, IRR, FADD, TRAF6, TRAF3, MKK3, MKK6, ROCK2, RSK1/2, SGK 1, SmMLCK, SIK2/3, ULK1/2, VEGFR1, WNK l , YES1, ZAP70, MAP4K3, MAP4K5, MAPKlb, MAPKAP-K2 K3, p38 alpha/beta/delta/gamma MAPK, Aurora A, Aurora B, Aurora C, MCAK, Clip, MAPKAPK, FAK, MARK 1 /2/3/4, Mucl , SHC, CXCR4, Gap-1, Myc, beta-catenin/TCF, Cbl, BRM, Mcl1, BRD2, BRD3, BRD4, AR, RAS, ErbB3, EGFR, IRE1, HPK1, RIPK2, and Era, including all variants, mutations, splice variants, indels and fusions thereof.
8. The conjugate of any above embodiment, wherein PB is selected from the group consisting of Heat Shock Protein 90 (HSP90) inhibitors, Kinase and Phosphatase inhibitors, MDM2 inhibitors, HDAC inhibitors, Human Lysine Methyltransferase Inhibitors,
Angiogenesis inhibitors, Immunosuppressive compounds, and compounds that target:
Human BET Bromodomain-containing proteins, the aryl hydrocarbon receptor (AHR), REF receptor kinase, FKBP, Androgen Receptor (AR), Estrogen receptor (ER), Thyroid Hormone Receptor, HIV Protease, HIV Integrase, HCV Protease, and Acyl-protein Thioesterase-1 and -2 (APT1 and APT2). 9. The conjugate of any above embodiment, wherein PB is a compound that targets Estrogen Receptor alpha (ERα).
10. The conjugate of any above embodiment, wherein the Ab is selected from Tables 4- 12, e.g., Table 4, Table 5, Table 6, Table 7, Table 8, Table 9, Table 10, Table 11, or Table 12. 11. The conjugate of any above embodiment, wherein the Ab is a cysteine engineered antibody or variants thereof.
12. The conjugate of any above embodiment, wherein Ab binds to one or more of polypeptides selected from the group consisting of DLL3; EDAR; CLL1; BMPR1B; E16; STEAP1; 0772P; MPF; NaPi2b; Sema 5b; PSCA hlg; ETBR; MSG783; STEAP2; TrpM4; CRIPTO; CD21; CD79b; FcRH2; B7-H4; HER2; NCA; MDP; IL20Rα; Brevican; EphB2R; ASLG659; PSCA; GEDA; BAFF-R; CD22; CD79a; CXCR5; HLA-DOB; P2X5; CD72; LY64; FcRH1; IRTA2; TENB2; PMEL17; TMEFF1; GDNF-Ra1; Ly6E; TMEM46;
Ly6G6D; LGR5; RET; LY6K; GPR19; GPR54; ASPHD1; Tyrosinase; TMEM118;
GPR172A; MUC16 and CD33.
13. The conjugate of any above embodiment, wherein Ab binds to one or more of polypeptides selected from the group consisting of CLL1; STEAP1; NaPi2b; STEAP2;
TrpM4; CRIPTO; CD21; CD79b; FcRH2; B7-H4; HER2; CD22; CD79a; CD72; LY64; Ly6E; MUC16; and CD33.
14. The conjugate of any above embodiment, wherein Ab is an antibody that binds to one or more polypeptides selected from the group consisting of B7-H4, Her2, CLL1, CD33, CD22 and NaPi2b.
15. The conjugate of any above embodiment, wherein the antibody binds to HER2 or B7- H4.
16. The conjugate of any above embodiment, wherein the antibody binds to Her2.
17. The conjugate of any above embodiment, wherein L1 is a peptidomimetic linker. 18. The conjugate of any above embodiment, wherein L1 is a peptidomimetic linker represented by the following formula
—Str—(PM)—Sp— wherein, Str is a stretcher unit covalently attached to Ab; Sp is a bond or spacer unit covalently attached to a PROTAC moiety; PM is a non-peptide chemical moiety selected from the group consisting of:
Figure imgf000144_0001
Figure imgf000144_0002
W is–NH-heterocycloalkyl- or heterocycloalkyl; Y is heteroaryl, aryl, -C(O)C1-C6alkylene, C1-C6alkylene-NH2, C1-C6alkylene-NH-CH3, C1- C6alkylene-N-(CH3)2, C1-C6alkenyl or C1-C6alkylenyl; each R1 is independently C1-C10alkyl, C1-C10alkenyl, (C1-C10alkyl)NHC(NH)NH2 or (C1- C10alkyl)NHC(O)NH2; R3 and R2 are each independently H, C1-C10alkyl, C1-C10alkenyl, arylalkyl or heteroarylalkyl, or R3 and R2 together may form a C3-C7cycloalkyl; and R4 and R5 are each independently C1-C10alkyl, C1-C10alkenyl, arylalkyl, heteroarylalkyl, (C1- C10alkyl )OCH2-, or R4 and R5 together may form a C3-C7cycloalkyl ring. 19. The conjugate of any above embodiment, wherein Y is heteroaryl; R4 and R5 together form a cyclobutyl ring.
20. The conjugate of any above embodiment, wherein Y is a moiety selected from the group consisting of
Figure imgf000145_0001
. 21. The conjugate of any above embodiment, wherein
Str is a chemical moiety represented by the following formula:
Figure imgf000145_0002
wherein R6 is selected from the group consisting of C1-C10alkylene, C1-C10alkenyl, C3- C8cycloalkyl, (C1-C8alkylene)O-, and C1-C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4- C7heterocycloalkyl, heteroarylalkyl, aryl arylalkyl, heteroarylalkyl and heteroaryl each Ra is independently H or C1-C6alkyl; Sp is -C1-C6alkylene-C(O)NH- or—Ar—Rb—, wherein Ar is aryl or heteroaryl, Rb is (C1- C10alkylene)O-. 22. The conjugate of any above embodiment, wherein Str has the formula:
Figure imgf000145_0003
wherein R7 is selected from C1-C10alkylene, C1-C10alkenyl, (C1-C10alkylene)O-, N(Rc)−(C2- C6 alkylene)−N(Rc) and N(Rc)−(C2-C6alkylene); where each Rc is independently H or C1-C6 alkyl; Sp is -C1-C6alkylene-C(O)NH- or—Ar—Rb—, wherein Ar is aryl or heteroaryl, Rb is (C1- C10alkylene)O-. 23. The conjugate of any above embodiment, wherein L1 has the following formula
Figure imgf000146_0001
R1 is C1-C6alkyl, C1-C6alkenyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2; R3 and R2 are each independently H, C1-C10alkyl ; and Str and Sp are as defined herein. 24. The conjugate of any above embodiment, wherein L1 has the following formula
Figure imgf000146_0002
R1 is C1-C6alkyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2; Str and Sp are as defined herein; and R4 and R5 together form a C3-C7cycloalkyl ring. 25. The conjugate of any above embodiment, wherein L1 has the following formula
Figure imgf000146_0003
Str and Sp are as defined herein; and R1 is C1-C6alkyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2. 26. The conjugate of any above embodiment, having the formula:
Figure imgf000147_0001
wherein Str is a chemical moiety represented by the following formula:
Figure imgf000147_0002
R6 is selected from the group consisting of C1-C10alkylene, and C1- C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4-C7heterocycloalkyl, heteroarylalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each Ra is independently H or C1-C6alkyl; Ab and Sp are as defined herein; and p is 1, 2, 3 or 4. 27. The conjugate of any above embodiment, having the formula:
Figure imgf000147_0003
wherein Str is a chemical moiety represented by the following formula:
Figure imgf000148_0001
R6 is selected from the group consisting of C1-C10alkylene, and C1- C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4-C7heterocycloalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each Ra is independently H or C1-C6alkyl; R1, R4, R5, Ab, D and Sp are as defined herein; and p is 1, 2, 3 or 4. 28. The conjugate of any above embodiment, wherein Y is heteroaryl, aryl or alkenyl; R6 is C1-C10alkylene. 29. The conjugate of any above embodiment, wherein Y is
Figure imgf000148_0002
.
30. The conjugate of any above embodiment, wherein Y is
Figure imgf000148_0003
.
31. The conjugate of any above embodiment, wherein Y is
Figure imgf000148_0004
. 32. The conjugate of any above embodiment, wherein
Str is a chemical moiety represented by the following formula:
Figure imgf000149_0001
, R6 is C1-C6alkylene;
Sp is -C1-C6alkylene-C(O)NH- or—Ar—Rb—, where Ar is aryl, Rb is (C1-C3alkylene)O-. 33. The conjugate of any above embodiment, having the formula:
Figure imgf000149_0002
wherein
Ab, D, R2 and R3 are as defined herein;
R1 is C1-C6alkyl-NH2, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2 ;and p is 1, 2 ,3 or 4.
34. The conjugate of any above embodiment, having the formula:
Figure imgf000149_0003
wherein
Ab and D are as defined herein; p is 1, 2, 3 or 4; R1 is C1-C6alkyl-NH2, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2; R4 and R5 are each independently C1-C6alkyl, wherein the alkyl are unsubstituted, or R4 and R5 together with the carbon to which each is attached can form a C3-C7cycloalkyl ring, such as a cyclobutyl. 35. The conjugate of any above embodiment, wherein L1 has the following formula:
Figure imgf000150_0001
,
wherein, R1 and R2 are independently selected from H and C1-C6 alkyl, or R1 and R2 form a 3, 4, 5, or 6-membered cycloalkyl or heterocyclyl group.
36. The conjugate of any above embodiment selected from the group consisting of PAC1, PAC2, PAC3, PAC4, and PAC5.
37. The conjugate of any above embodiment, wherein the ratio of PROTAC per Antibody (“PAR”) is from about 1.5 to about 3.
38. The conjugate of any above embodiment, wherein the ratio of PROTAC per Antibody (“PAR”) is about 2.
39. A method of treating a disease in a human in need thereof, comprising administering to the human an effective amount of a conjugate of any above embodiment.
40. A pharmaceutical composition comprising a conjugate of any above embodiment and a pharmaceutically acceptable excipient.
41. A method of treating a disease in a human in need thereof, comprising administering to said human an effective amount of a pharmaceutical composition of embodiment 40.
42. A method of treating a disease with a conjugate of any above embodiment, wherein the disease is a hyperproliferative disorder, including, benign or malignant solid tumors and hematological disorders, disorders involving neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic, immunologic, and autoimmune conditions.
43. A method of treating a disease with a conjugate of any above embodiment, wherein the disease is cancer.
44. A method of treating a disease with a conjugate of any above embodiment, wherein the cancer is selected from the group consisting of a carcinoma, lymphoma, blastoma, sarcoma, leukemia, lymphoid malignancies, squamous cell cancer (e.g. epithelial squamous cell cancer), lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, and head and neck cancer.
45. The method of embodiment 43, wherein the cancer is a HER2-positive cancer.
46. The method of embodiment 45, wherein the HER2-positive cancer is breast cancer or gastric cancer.
47. A method of treating a disease with a conjugate of any above embodiment, wherein the disease is an autoimmune disease.
48. The method of embodiment 47, wherein said autoimmune disease is selected from the group consisting of rheumatologic disorders (such as, for example, rheumatoid arthritis, Sjögren's syndrome, scleroderma, lupus such as systemic lupus erythematosus (SLE) and lupus nephritis, polymyositis/dermatomyositis, cryoglobulinemia, anti-phospholipid antibody syndrome, and psoriatic arthritis), osteoarthritis, autoimmune gastrointestinal and liver disorders (such as, for example, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), autoimmune gastritis and pernicious anemia, autoimmune hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, and celiac disease), vasculitis (such as, for example, ANCA-associated vasculitis, including Churg-Strauss vasculitis, Wegener's granulomatosis, and polyarteriitis), autoimmune neurological disorders (such as, for example, multiple sclerosis, opsoclonus myoclonus syndrome, myasthenia gravis, neuromyelitis optica, Parkinson’s disease, Alzheimer’s disease, and autoimmune polyneuropathies), renal disorders (such as, for example, glomerulonephritis, Goodpasture’s syndrome, and Berger’s disease), autoimmune dermatologic disorders (such as, for example, psoriasis, urticaria, hives, pemphigus vulgaris, bullous pemphigoid, and cutaneous lupus erythematosus), hematologic disorders (such as, for example, thrombocytopenic purpura, thrombotic thrombocytopenic purpura, post-transfusion purpura, and autoimmune hemolytic anemia), atherosclerosis, uveitis, autoimmune hearing diseases (such as, for example, inner ear disease and hearing loss), Behcet's disease, Raynaud's syndrome, organ transplant, and autoimmune endocrine disorders (such as, for example, diabetic-related autoimmune diseases such as insulin- dependent diabetes mellitus (IDDM), Addison’s disease, and autoimmune thyroid disease (e.g., Graves’ disease and thyroiditis)).
49. The method of embodiment 47, wherein said autoimmune disease is selected from the group consisiting of rheumatoid arthritis, ulcerative colitis, ANCA-associated vasculitis, lupus, multiple sclerosis, Sjögren's syndrome, Graves’ disease, IDDM, pernicious anemia, thyroiditis, and glomerulonephritis.
50. Use of a conjugate of any above embodiment in the manufacture of a medicament for the treatment of cancer in a mammal.
51. A method of preparing a PAC, said method comprising contacting a Linker L1- PROTAC with an antibody.
IV. Synthesis Routes
PACs and compounds described herein can be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein, and those for other heterocycles described in:
Comprehensive Heterocyclic Chemistry II, Editors Katritzky and Rees, Elsevier, 1997, e.g. Volume 3; Liebigs Annalen der Chemie, (9):1910-16, (1985); Helvetica Chimica Acta, 41:1052-60, (1958); Arzneimittel-Forschung, 40(12):1328-31, (1990). Starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, WI) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v.1-23, Wiley, N.Y. (1967-2006 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer-Verlag, Berlin, including supplements (also available via the Beilstein online database). Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the PACs and groups as described herein and necessary reagents and intermediates are known in the art and include, for example, those described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and P. G .M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley and Sons (1999); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995) and subsequent editions thereof. In preparing PACs and compounds, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz or CBZ) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
The General Procedures and Examples provide exemplary methods for preparing PACs and compounds described herein. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the PACs and compounds. Although specific starting materials and reagents are depicted and discussed in the Schemes, General
Procedures, and Examples, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the exemplary compounds prepared by the described methods can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
Generally, a PAC may be prepared by connecting a PROTAC with a L1 linker reagent according to the procedures of WO 2013/055987; WO 2015/023355; WO
2010/009124; WO 2015/095227, and conjugating it with any of the antibodies or variants, mutations, splice variants, indels and fusions thereof, including cysteine engineered antibodies, described herein. Alternatively, a PAC may be prepared by first connecting an antibody or variant, mutation, splice variant, indel and fusion thereof, including a cysteine engineered antibody, described herein with a L1 linker reagent, and conjugating it with any PROTAC. The following synthetic routes describe exemplary methods of preparing PACs and components thereof. Other synthetic routes for preparing PACs and components thereof are disclosed elsewhere herein.
1. Linker L1
With respect to Linker L1, Schemes 1-4 depict synthesis routes to exemplary linkers L1 for disulfide attachment to antibody Ab. The Ab is connected to L1 through a disulfide bond and the PROTAC is connected to L1 through any available attachment on the
PROTAC.
Figure imgf000154_0001
Scheme 1
Referring to Scheme 1, 1,2-Di(pyridin-2-yl)disulfane and 2-mercaptoethanol were reacted in pyridine and methanol at room temperature to give 2-(pyridin-2- yldisulfanyl)ethanol. Acylation with 4-nitrophenyl carbonochloridate in triethylamine and acetonitrile gave 4-nitrophenyl 2-(pyridin-2-yldisulfanyl)ethyl carbonate 9.
Figure imgf000154_0002
Scheme 2
Referring to Scheme 2, to a mixture of 1,2-bis(5-nitropyridin-2-yl)disulfane 10 (1.0 g, 3.22 mmol) in anhydrous DMF/MeOH (25 mL/25 mL) was added HOAc (0.1 mL), followed by 2-aminoethanethiol hydrochloride 11 (183 mg, 1.61 mmol). After the reaction mixture was stirred at r.t. overnight, it was concentrated under vacuum to remove the solvent, and the residue was washed with DCM (30 mL × 4) to afford 2-((5-nitropyridin-2- yl)disulfanyl)ethanamine hydrochloride 12 as pale yellow solid (300 mg, 69.6 %). 1H NMR (400 MHz, DMSO-d6) δ 9.28 (d, J = 2.4 Hz, 1H), 8.56 (dd, J = 8.8, 2.4 Hz, 1H), 8.24 (s, 4H), 8.03 (d, J = 8.8 Hz, 1H), 3.15 - 3.13 (m, 2H), 3.08 - 3.06 (m, 2H).
Figure imgf000155_0001
Scheme 3
Referring to Scheme 3, a solution of 1,2-bis(5-nitropyridin-2-yl)disulfane 10 (9.6 g, 30.97 mmol) and 2-mercaptoethanol (1.21 g, 15.49 mmol) in anhydrous DCM/CH3OH (250 mL/250 mL) was stirred at r.t. under N2 for 24 h. After the mixture was concentrated under vacuum, and the residue was diluted with DCM (300 mL). MnO2 (10 g) was added and the mixture was stirred at r.t. for another 0.5 h. The mixture was purified by column
chromatography on silica gel (DCM/MeOH = 100/1 to 100/1) to afford 2-((5-nitropyridin-2- yl)disulfanyl)ethanol 13 (2.2 g, 61.1 %) as brown oil. 1H NMR (400 MHz, CDCl3) δ 9.33 (d, J = 2.8 Hz, 1H), 8.38 - 8.35 (dd, J = 9.2, 2.8 Hz, 1H), 7.67 (d, J = 9.2 Hz, 1H), 4.10 (t, J = 7.2 Hz, 1H), 3.81 - 3.76 (q, 2H), 3.01 (t, J = 5.2 Hz, 2H).
To a solution of 13 (500 mg, 2.15 mmol) in anhydrous DMF (10 mL) was added DIEA (834 mg, 6.45 mmol), followed by PNP carbonate (bis(4-nitrophenyl) carbonate, 1.31g, 4.31 mmol). The reaction solution was stirred at r.t for 4 h and the mixture was purified by prep-HPLC (FA) to afford 4-nitrophenyl 2-((5-nitropyridin-2-yl)disulfanyl)ethyl carbonate 14 (270 mg, 33.1 %) as light brown oil. 1H NMR (400 MHz, CDCl3) δ 9.30 (d, J = 2.4 Hz, 1H), 8.43 - 8.40 (dd, J = 8.8, 2.4 Hz, 1H), 8.30 - 8.28 (m, 2H), 7.87 (d, J = 8.8 Hz, 1H), 7.39 - 7.37 (m, 2H), 4.56 (t, J = 6.4 Hz, 2H), 3.21 (t, J = 6.4 Hz, 2H).
Figure imgf000156_0001
Scheme 4
Referring to Scheme 4, sulfuryl chloride (2.35 mL of a 1.0M solution in DCM, 2.35 mmol) was added drop-wise to a stirred suspension of 5-nitropyridine-2-thiol (334 mg, 2.14 mmol) in dry DCM (7.5 mL) at 0°C (ice/acetone) under an argon atmosphere. The reaction mixture turned from a yellow suspension to a yellow solution and was allowed to warm to room temperature then stirred for 2 hours after which time the solvent was removed by evaporation in vacuo to provide a yellow solid. The solid was re-dissolved in DCM (15 mL) and treated drop-wise with a solution of (R)-2-mercaptopropan-1-ol (213 mg, 2.31 mmol) in dry DCM (7.5 mL) at 0°C under an argon atmosphere. The reaction mixture was allowed to warm to room temperature and stirred for 20 hours at which point analysis by LC/MS revealed substantial product formation at retention time 1.41 minutes (ES+) m/z 247 ([M+ H]+., ~100% relative intensity). The precipitate was removed by filtration and the filtrate evaporated in vacuo to give an orange solid which was treated with H2O (20 mL) and basified with ammonium hydroxide solution. The mixture was extracted with DCM (3 x 25 mL) and the combined extracts washed with H2O (20 mL), brine (20 mL), dried (MgSO4), filtered and evaporated in vacuo to give the crude product. Purification by flash chromatography
(gradient elution in 1% increments: 100% DCM to 98:2 v/v DCM/MeOH) gave (R)-2-((5- nitropyridin-2-yl)disulfanyl)propan-1-ol 15 as an oil (111 mg, 21% yield).
To a solution of triphosgene, Cl3COCOOCCl3, Sigma Aldrich, CAS Reg. No.32315- 10-9 (241 mg, 0.812 mmol) in DCM (10 mL) was added a solution of (R)-2-((5-nitropyridin- 2-yl)disulfanyl)propan-1-ol 15 (500 mg, 2.03 mmol) and pyridine (153 mg, 1.93 mmol) in DCM (10 mL) dropwise at 20 oC. After the reaction mixture was stirred at 20oC for 30 min, it was concentrated and (R)-2-((5-nitropyridin-2-yl)disulfanyl)propyl carbonochloridate 16 can be used directly without further purification to covalently link through the carbonochloridate group any available group on the PROTAC. 2. Cysteine Engineered Antibodies
With regard to cysteine engineered antibodies for conjugation by reduction and reoxidation, they can be prepared generally as follows. Light chain amino acids are numbered according to Kabat (Kabat et al., Sequences of proteins of immunological interest, (1991) 5th Ed., US Dept of Health and Human Service, National Institutes of Health, Bethesda, MD). Heavy chain amino acids are numbered according to the EU numbering system (Edelman et al (1969) Proc. Natl. Acad. of Sci.63(1):78-85), except where noted as the Kabat system. Single letter amino acid abbreviations are used.
Full length, cysteine engineered monoclonal antibodies (THIOMAB™ antibodies) expressed in CHO cells bear cysteine adducts (cystines) or are glutathionylated on the engineered cysteines due to cell culture conditions. As is, THIOMAB™ antibodies purified from CHO cells cannot be conjugated to Cys-reactive linker L1-PROTAC intermediates. Cysteine engineered antibodies may be made reactive for conjugation with linker-PROTAC intermediates described herein, 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) followed by re- formation of the inter-chain disulfide bonds (re-oxidation) with a mild oxidant such as dehydroascorbic acid. Full length, cysteine engineered monoclonal antibodies
(THIOMAB™ antibodies) expressed in CHO cells (Gomez et al (2010) Biotechnology and Bioeng.105(4):748-760; Gomez et al (2010) Biotechnol. Prog.26:1438-1445) were reduced, for example, with about a 50 fold excess of DTT overnight in 50 mM Tris, pH 8.0 with 2 mM EDTA at room temperature, which removes Cys and glutathione adducts as well as reduces interchain disulfide bonds in the antibody. Removal of the adducts was monitored by reverse-phase LCMS using a PLRP-S column. The reduced THIOMAB™ antibody was diluted and acidified by addition to at least four volumes of 10 mM sodium succinate, pH 5 buffer.
Alternatively, the antibody was diluted and acidified by adding to at least four volumes of 10 mM succinate, pH 5 and titration with 10% acetic acid until pH was approximately five. The pH-lowered and diluted THIOMAB™ antibody was subsequently loaded onto a HiTrap S cation exchange column, washed with several column volumes of 10 mM sodium acetate, pH 5 and eluted with 50 mM Tris, pH 8.0, 150 mM sodium chloride. Disulfide bonds were reestablished between cysteine residues present in the parent Mab by carrying out reoxidation. The eluted reduced THIOMAB™ antibody described above is treated with 15X dehydroascorbic acid (DHAA) for about 3 hours or, alternatively, with 200 nM to 2 mM aqueous copper sulfate (CuSO4) 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 may also be effective. This mild, partial reoxidation step forms intrachain disulfides efficiently with high fidelity. Reoxidation was monitored by reverse- phase LCMS using a PLRP-S column. The reoxidized THIOMAB™ antibody was diluted with succinate buffer as described above to reach pH approximately 5 and purification on an S column was carried out as described above with the exception that elution was performed with a gradient of 10 mM succinate, pH 5, 300 mM sodium chloride (buffer B) in 10 mM succinate, pH 5 (buffer A). To the eluted THIOMABTM antibody , EDTA was added to a final concentration of 2 mM and concentrated, if necessary, to reach a final concentration of more than 5 mg/mL. The resulting THIOMAB™ antibody, ready for conjugation, was stored at -20 oC or -80 oC in aliquots. Liquid chromatography/Mass Spectrometric Analysis was performed on a 6200 series TOF or QTOF Agilent LC/MS. Samples were
chromatographed on a PRLP-S®, 1000 A, microbore column (50mm × 2.1mm, Polymer Laboratories, Shropshire, UK) heated to 80 °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 eluent was directly ionized using the electrospray source. Data were collected and deconvoluted by the MassHunter software (Agilent). Prior to LC/MS analysis, antibodies or conjugates (50 micrograms) were treated with PNGase F (2 units/ml; PROzyme, San Leandro, CA) for 2 hours at 37 °C to remove N-linked carbohydrates.
Alternatively, antibodies or conjugates were partially digested with LysC (0.25 µg per 50 µg (microgram) antibody or conjugate) for 15 minutes at 37 °C to give a Fab and Fc fragment for analysis by LCMS. Peaks in the deconvoluted LCMS spectra were assigned and quantitated. PROTAC-to-antibody ratios (PAR) were calculated by calculating the ratio of intensities of the peak or peaks corresponding to PROTAC-conjugated antibody relative to all peaks observed.
3. Conjugation of Linker L1-PROTAC group to antibodies
In one method of conjugating Linker L1-PROTAC compounds to antibodies, after the reduction and reoxidation procedures above, the cysteine-engineered antibody
(THIOMAB™ antibody), in 10 mM succinate, pH 5, 150 mM NaCl, 2 mM EDTA, is pH- adjusted to pH 7.5-8.5 with 1M Tris. An excess, from about 3 molar to 20 equivalents of a linker-PROTAC intermediate with a thiol-reactive group (e.g., maleimide or 4-nitropyridy disulfide), is dissolved in DMF, DMA or propylene glycol and added to the reduced, reoxidized, and pH-adjusted antibody. The reaction is incubated at room temperature or 37 C and monitored until completion (1 to about 24 hours), as determined by LC-MS analysis of the reaction mixture. When the reaction is complete, the conjugate is purified by one or any combination of several methods, the goal being to remove remaining unreacted linker- PROTAC intermediate and aggregated protein (if present at significant levels). For example, the conjugate may be diluted with 10 mM histidine-acetate, pH 5.5 until final pH is approximately 5.5 and purified by S cation exchange chromatography using either HiTrap S columns connected to an Akta purification system (GE Healthcare) or S maxi spin columns (Pierce). Alternatively, the conjugate may be purified by gel filtration chromatography using an S200 column connected to an Akta purification system or Zeba spin columns. Alternatively, dialysis may be used. The THIOMABTM antibody PROTAC conjugates were formulated into 20 mM His/acetate, pH 5, with 240 mM sucrose using either gel filtration or dialysis. The purified conjugate is concentrated by centrifugal ultrafiltration and filtered through a 0.2-µm filter under sterile conditions and frozen for storage. The PACs were characterized by BCA assay to determine protein concentration, analytical SEC (size- exclusion chromatography) for aggregation analysis and LC-MS after treatment with Lysine C endopeptidase (LysC) to calculate PAR.
Size exclusion chromatography is performed on conjugates using a Shodex KW802.5 column in 0.2M potassium phosphate pH 6.2 with 0.25 mM potassium chloride and 15% IPA at a flow rate of 0.75 ml/min. Aggregation state of the conjugate was determined by integration of eluted peak area absorbance at 280 nm.
LC-MS analysis may be performed on PAC using an Agilent QTOF 6520 ESI instrument. As an example, the PAR is treated with 1:500 w/w Endoproteinase Lys C (Promega) in Tris, pH 7.5, for 30 min at 37°C. The resulting cleavage fragments are loaded onto a 1000Å (Angstrom), 8 µm (micron) PLRP-S (highly cross-linked polystyrene) column heated to 80 °C and eluted with a gradient of 30% B to 40% B in 5 minutes. Mobile phase A was H2O with 0.05% TFA and mobile phase B was acetonitrile with 0.04% TFA. The flow rate was 0.5ml/min. Protein elution was monitored by UV absorbance detection at 280nm prior to electrospray ionization and MS analysis. Chromatographic resolution of the unconjugated Fc fragment, residual unconjugated Fab and drugged Fab was usually achieved. The obtained m/z spectra were deconvoluted using Mass Hunter™ software (Agilent Technologies) to calculate the mass of the antibody fragments. V. Formulations
Pharmaceutical formulations of therapeutic PROTAC-antibody-conjugates (PACs) as described herein can be prepared for parenteral administration, e.g., bolus, intravenous, intratumor injection with a pharmaceutically acceptable parenteral vehicle and in a unit dosage injectable form. A PAC having the desired degree of purity is optionally mixed with one or more pharmaceutically acceptable excipients (Remington's Pharmaceutical Sciences (1980) 16th edition, Osol, A. Ed.), in the form of a lyophilized formulation for reconstitution or an aqueous solution.
PACs can be formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition. According to this aspect, there is provided a pharmaceutical composition comprising a PAC in association with one or more pharmaceutically acceptable excipients.
A typical formulation is prepared by mixing PACs with excipients, such as carriers and/or diluents. Suitable carriers, diluents and other excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like. The particular carrier, diluent or other excipient used will depend upon the means and purpose for which the PAC is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
In general, safe solvents are non-toxic aqueous solvents such as water and other non- toxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof. 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).
The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the PAC or aid in the manufacturing of the pharmaceutical product. The formulations may be prepared using conventional dissolution and mixing procedures.
Formulation may be conducted by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed. The pH of the formulation depends mainly on the particular use and the concentration of compound, but may range from about 3 to about 8. Formulation in an acetate buffer at pH 5 is a suitable embodiment.
The PAC formulations can be sterile. In particular, formulations to be used for in vivo administration must be sterile. Such sterilization is readily accomplished by filtration through sterile filtration membranes.
The PAC ordinarily can be stored as a solid composition, a lyophilized formulation or as an aqueous solution.
The pharmaceutical compositions comprising a PAC can be formulated, dosed and administered in a fashion, i.e., amounts, concentrations, schedules, course, vehicles and route of administration, consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The“therapeutically effective amount” of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the coagulation factor mediated disorder. Such amount is preferably below the amount that is toxic to the host or renders the host significantly more susceptible to bleeding. The PAC can be formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen. The pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug. Generally, an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form. Suitable containers are well known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like. The container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package. In addition, the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
The pharmaceutical compositions 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 1,3-butanediol. The sterile injectable preparation may also be 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 PAC 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, a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weight:weight). The pharmaceutical composition can be prepared to provide easily measurable amounts for 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.
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 subject matter 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 or by any other desired route.
VI. Indications and Methods of Treatment
It is contemplated that the PROTAC-antibody conjugates (PAC) disclosed herein may be used to treat various diseases or disorders. Exemplary hyperproliferative disorders include benign or malignant solid tumors and hematological disorders such as leukemia and lymphoid malignancies. Others include neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic and immunologic, including autoimmune, disorders.
Generally, the disease or disorder to be treated is a hyperproliferative disease such as cancer. Examples of cancer to be treated herein include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g. epithelial squamous cell cancer), lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
Autoimmune diseases for which the PAC may be used in treatment include rheumatologic disorders (such as, for example, rheumatoid arthritis, Sjögren's syndrome, scleroderma, lupus such as systemic lupus erythematosus (SLE) and lupus nephritis, polymyositis/dermatomyositis, cryoglobulinemia, anti-phospholipid antibody syndrome, and psoriatic arthritis), osteoarthritis, autoimmune gastrointestinal and liver disorders (such as, for example, inflammatory bowel diseases (e.g., ulcerative colitis and Crohn's disease), autoimmune gastritis and pernicious anemia, autoimmune hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, and celiac disease), vasculitis (such as, for example, ANCA- associated vasculitis, including Churg-Strauss vasculitis, Wegener's granulomatosis, and polyarteriitis), autoimmune neurological disorders (such as, for example, multiple sclerosis, opsoclonus myoclonus syndrome, myasthenia gravis, neuromyelitis optica, Parkinson’s disease, Alzheimer’s disease, and autoimmune polyneuropathies), renal disorders (such as, for example, glomerulonephritis, Goodpasture’s syndrome, and Berger’s disease), autoimmune dermatologic disorders (such as, for example, psoriasis, urticaria, hives, pemphigus vulgaris, bullous pemphigoid, and cutaneous lupus erythematosus), hematologic disorders (such as, for example, thrombocytopenic purpura, thrombotic thrombocytopenic purpura, post-transfusion purpura, and autoimmune hemolytic anemia), atherosclerosis, uveitis, autoimmune hearing diseases (such as, for example, inner ear disease and hearing loss), Behcet's disease, Raynaud's syndrome, organ transplant, and autoimmune endocrine disorders (such as, for example, diabetic-related autoimmune diseases such as insulin- dependent diabetes mellitus (IDDM), Addison’s disease, and autoimmune thyroid disease (e.g., Graves’ disease and thyroiditis)). More preferred such diseases include, for example, rheumatoid arthritis, ulcerative colitis, ANCA-associated vasculitis, lupus, multiple sclerosis, Sjögren's syndrome, Graves’ disease, IDDM, pernicious anemia, thyroiditis, and
glomerulonephritis.
In certain embodiments, a PAC comprising an anti-NaPi2b antibody, such as those described above, is used in a method of treating solid tumor, e.g., ovarian. In another embodiment, a PAC an anti-CD33 antibody, such as those described herein, is used in a method of treating hematological malignancies such as non-Hodgkin's lymphoma (NHL), diffuse large hematopoietic lymphoma, follicular lymphoma, mantle cell lymphoma, chronic lymphocytic leukemia, multiple myeloma, acute myeloid leukemia (AML), and myeloid cell leukemia (MCL), and including B-cell related cancers and proliferative disorders. See: US 8226945; Li et al (2013) Mol. Cancer. Ther.12(7):1255- 1265; Polson et al (2010) Leukemia 24:1566-1573; Polson et al (2011) Expert Opin. Investig. Drugs 20(1):75-85.
In another embodiment, a PAC comprising an anti-MUC16 antibody, such as those described herein, is used in a method of treating ovarian, breast and pancreatic cancers. The cancer may be associated with the expression or activity of a MUC16/CA125/O772P polypeptide. See: WO 2007/001851; US 7989595; US 8449883; US 7723485; Chen et al (2007) Cancer Res.67(10): 4924-4932; Junutula, et al., (2008) Nature Biotech., 26(8):925- 932.
In certain embodiments, a PAC comprising an anti-HER2 antibody, such as those described above, is used in a method of treating cancer, e.g., breast or gastric cancer, more specifically HER2+ breast or gastric cancer, wherein the method comprises administering such PAC to a patient in need of such treatment. In one such embodiment, the PAC comprises the anti-HER2 antibody trastuzumab or pertuzumab.
A PAC may be administered by any route appropriate to the condition to be treated. The PAC will typically be administered parenterally, i.e. infusion, subcutaneous,
intramuscular, intravenous, intradermal, intrathecal and epidural.
A PAC can be used either alone or in combination with other agents in a therapy. For instance, a PAC may be co-administered with at least one additional therapeutic agent. Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the PAC can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. A PAC can also be used in combination with radiation therapy.
A PAC (and any additional therapeutic agent) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.
For the prevention or treatment of disease, the appropriate dosage of a PAC (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of PAC, the severity and course of the disease, whether the PAC is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the PAC, and the discretion of the attending physician. The PAC is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 µg/kg to 15 mg/kg (e.g.0.1mg/kg-10mg/kg) of a PAC can be an initial candidate dosage for
administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 µg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of a PAC would be in the range from about 0.05 mg/kg to about 10 mg/kg. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses). An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
VII. Articles of Manufacture
In another aspect, described herein are articles of manufacture, for example, a“kit”, containing materials useful for the treatment of the diseases and disorders described above is provided. The kit comprises a container comprising a PAC. The kit may further comprise a label or package insert, on or associated with the container. The term“package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products. Suitable containers include, for example, bottles, vials, syringes, blister pack, etc. A “vial” is a container suitable for holding a liquid or lyophilized preparation. In one embodiment, the vial is a single-use vial, e.g. a 20-cc single-use vial with a stopper. The container may be formed from a variety of materials such as glass or plastic. The container may hold a PAC or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
At least one active agent in the composition is a PAC. The label or package insert indicates that the composition is used for treating the condition of choice, such as cancer. In addition, the label or package insert may indicate that the patient to be treated is one having a disorder such as a hyperproliferative disorder, neurodegeneration, cardiac hypertrophy, pain, migraine or a neurotraumatic disease or event. In one embodiment, the label or package inserts indicates that the composition comprising a PAC can be used to treat a disorder resulting from abnormal cell growth. The label or package insert may also indicate that the composition can be used to treat other disorders. Alternatively, or additionally, the article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer’s solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
The kit may further comprise directions for the administration of the PAC and, if present, the second pharmaceutical formulation. For example, if the kit comprises a first composition comprising a PAC, and a second pharmaceutical formulation, the kit may further comprise directions for the simultaneous, sequential or separate administration of the first and second pharmaceutical compositions to a patient in need thereof.
In another embodiment, the kits are suitable for the delivery of solid oral forms of a PAC, such as tablets or capsules. Such a kit preferably includes a number of unit dosages. Such kits can include a card having the dosages oriented in the order of their intended use. An example of such a kit is a“blister pack”. Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms. If desired, a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered. According to one embodiment, a kit may comprise (a) a first container with a PAC contained therein; and optionally (b) a second container with a second pharmaceutical formulation contained therein, wherein the second pharmaceutical formulation comprises a second compound with anti-hyperproliferative activity. Alternatively, or additionally, the kit may further comprise a third container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer’s solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
In certain other embodiments wherein the kit comprises a PAC and a second therapeutic agent, the kit may comprise a container for containing the separate compositions such as a divided bottle or a divided foil packet; however, the separate compositions may also be contained within a single, undivided container. Typically, the kit comprises directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
The following examples are offered by way of illustration and not by way of limitation.
EXAMPLES Example 1
Syntheses of a PAC
A. Chemical Synthesis of a PROTAC:
i. Attachment of a Linker (L2) to an E3 Ligase Binding Group (E3LB)
Figure imgf000168_0001
Methyl 4-[[(2S,3S)-3-[(2S)-2-[[(tert-butoxy)carbonyl](methyl)amino]propanamido]-8- cyano-5-[(2-methoxynaphthalen-1-yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5- benzodiazepin-1-yl]carbonyl]benzoate
To a solution of tert-butyl N-[(1S)-1-[[(3S,4S)-7-cyano-1-[(2-methoxynaphthalen-1- yl)methyl]-4-methyl-2-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-3-yl]carbamoyl]ethyl]- N-methylcarbamate (3.00 g, 5.25 mmol) in 1,2-dichloroethane (50 mL) was added triethylamine (2.6 g, 25.7 mmol) and methyl 4-(carbonochloridoyl)benzoate (3.10 g, 15.61 mmol) under nitrogen. The resulting solution was stirred for 5 h at 80 oC and allowed to cool to room temperature. Water (100 mL) was added. The resulting solution was extracted with 3x100 mL of dichloromethane and the organic layers combined and dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by flash
chromatography on silica gel eluting with ethyl acetate/petroleum ether (1:1). This resulted in 3.10 g (81%) of methyl 4-[[(2S,3S)-3-[(2S)-2-[[(tert- butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5-[(2-methoxynaphthalen-1- yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1-yl]carbonyl]benzoate as a brown solid. MS (ESI): [M+H]+ = 734.4.
Figure imgf000169_0001
4-[[(2S,3S)-3-[(2S)-2-[[(Tert-butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5- [(2-methoxynaphthalen-1-yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5- benzodiazepin-1-yl]carbonyl]benzoic acid
Aqueous LiOH solution (30 mL, 1 M) was added to a solution of methyl 4-[[(2S,3S)-3-[(2S)- 2-[[(tert-butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5-[(2-methoxynaphthalen-1- yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1-yl]carbonyl]benzoate (3.10 g, 4.22 mmol) in tetrahydrofuran (30 mL) at room temperature. The resulting solution was stirred for 5 h at room temperature. Ethyl ether (20 mL) was added. Phases were separated The aqueous phase was acidified with 1 N HCl solution until pH about 7 The resulting mixture was extracted with 2x80 mL of ethyl acetate and the organic layers were combined and dried over anhydrous sodium sulfate and concentrated under vacuum. This resulted in 2.5 g of 4-[[(2S,3S)-3-[(2S)-2-[[(tert- butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5-[(2-methoxynaphthalen-1- yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1-yl]carbonyl]benzoic acid as a brown solid. MS (ESI): [M+H]+ = 720.5.
Figure imgf000170_0001
Methyl 2-(2-(2-aminoethoxy)ethoxy)acetate hydrochloride
To a solution of 2-[2-(2-aminoethoxy)ethoxy]acetic acid hydrochloride (500 mg, 2.505 mmol) in 2,2-dimethoxypropane (5 mL, 40.327 mmol) was added dropwise concentrated HCl (0.2 mL) at room temperature. The reaction mixture was stirred for 15 h at 25 oC and concentrated under vacuum. The residue was used directly without further purification.
Figure imgf000170_0002
Methyl 2-(2-[2-[(4-[[(2S,3S)-3-[(2S)-2-[[(tert- butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5-[(2-methoxynaphthalen-1- yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1- yl]carbonyl]phenyl)formamido]ethoxy]ethoxy)acetate
To a solution of [(2S,3S)-3-[(2S)-2-[[(tert-butoxy)carbonyl](methyl)amino]propanamido]-8- cyano-5-[(2-methoxynaphthalen-1-yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5- benzodiazepin-1-yl]carbonyl]benzoic acid (500 mg, 0.695 mmol) in N,N-dimethylformamide (6 mL) was added crude methyl 2-[2-(2-aminoethoxy)ethoxy]acetate HCl salt from the previous step (500 mg), HATU (528 mg, 1.389 mmol) and DIPEA (897 mg, 6.94 mmol) under nitrogen at room temperature. The resulting solution was stirred for 1 hour at 25oC, and quenched with water. The resulting solution was extracted with dichloromethane and the organic layers combined The organic phases were washed with brine and dried over anhydrous sodium sulfate and concentrated under vacuum. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (20:1). This resulted in 550 mg (90%) of methyl 2-(2-[2-[(4-[[(2S,3S)-3-[(2S)-2-[[(tert- butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5-[(2-methoxynaphthalen-1- yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1- yl]carbonyl]phenyl)formamido]ethoxy]ethoxy)acetate as a yellow solid. MS (ESI): [M+H]+ = 879.5.
Figure imgf000171_0001
2-(2-[2-[(4-[[(2S,3S)-3-[(2S)-2-[[(Tert-butoxy)carbonyl](methyl)amino]propanami do]-8-cyano-5-[(2-methoxynaphthalen-1-yl)methyl]-2-methyl-4-oxo-2,3,4,5- tetrahydro-1H-1,5-benzodiazepin-1- yl]carbonyl]phenyl)formamido]ethoxy]ethoxy)acetic acid
To a solution of methyl 2-(2-[2-[(4-[[(2S,3S)-3-[(2S)-2-[[(tert- butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5-[(2-methoxynaphthalen-1- yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1- yl]carbonyl]phenyl)formamido]ethoxy]ethoxy)acetate (500 mg, 0.569 mmol) in
tetrahydrofuran (8 mL) was added a solution of lithium hydroxide monohydrate (95 mg, 2.26 mmol) in water (1 mL) at room temperature. The mixture was stirred for 1 hour at 25 oC. The mixture was diluted with water and acidified with 1 N citric acid to pH about 4, extracted with ethyl acetate (2x). The organic phases were combined and washed with brine, dried over sodium sulfate, and concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography on C18 silica gel, mobile phase: 5 mM aqueous NH4HCO3 and CH3CN (0-95%) to afford 370 mg (75%) of 2-(2-[2-[(4-[[(2S,3S)-3-[(2S)-2-[[(tert- butoxy)carbonyl](methyl)amino]propanamido]-8-cyano-5-[(2-methoxynaphthalen-1- yl)methyl]-2-methyl-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1- yl]carbonyl]phenyl)formamido]ethoxy]ethoxy)acetic acid as a white solid. MS (ESI):
[M+H]+= 865.5.1H NMR (400 MHz, DMSO-d6): δ (ppm)
8.70 (d, J = 8.6 Hz, 1H), 8.46 (t, J = 5.5 Hz, 1H), 8.21 (d, J = 8.7 Hz, 1H), 8.10 (d, J = 8.6 Hz, 1H), 7.95 (d, J = 9.1 Hz, 1H), 7.89 (dd, J = 8.6, 1.9 Hz, 1H), 7.69 (d, J = 8.1
Hz, 1H), 7.50 (d, J = 9.2 Hz, 1H), 7.30 (m, 1H), 7.18 (d, J = 1.9 Hz, 1H), 7.13 (t, J =
7.5 Hz, 1H), 7.00 (d, J = 8.1 Hz, 2H), 6.13 (d, J = 15.1 Hz, 1H), 5.78 (d, J = 8.0 Hz,
2H), 5.51 (d, J = 15.0 Hz, 1H), 5.04 (brs, 1H), 4.61-4.52 (m, 1H), 4.22 (dd, J = 11.9, 8.5 Hz, 1H), 3.99 (s, 2H), 3.96 (s, 3H), 3.68– 3.44 (m, 7H), 3.37(s, 1H), 2.76 (s, 3H), 1.40-1.31 (m, 12H), 1.12 (d, J = 6.1 Hz, 3H).
ii. Attachment of a PB to an E3LB via a Linker (L2)
Figure imgf000172_0001
4-((2S,3S)-8-Cyano-5-((2-methoxynaphthalen-1-yl)methyl)-2-methyl-3-((S)-2- (methylamino)propanamido)-4-oxo-2,3,4,5-tetrahydro-1H-benzo[b][1,4]diazepine-1- carbonyl)-N-(2-(2-(2-((2-(4-(1-(4-hydroxyphenyl)-2-phenylbut-1-en-1- yl)phenoxy)ethyl)(methyl)amino)-2-oxoethoxy)ethoxy)ethyl)benzamide (“compound P1”)
To a solution of 2-[2-[2-[[4-[(3S,4S)-3-[[(2S)-2-[tert- butoxycarbonyl(methyl)amino]propanoyl] amino]-7-cyano-1-[(2-methoxy-1- naphthyl)methyl]-4-methyl-2-oxo-3,4-dihydro-1,5-benzodiazepine-5- carbonyl]benzoyl]amino]ethoxy]ethoxy]acetic acid (74 mg, 0.0855 mmol) in 2- methyltetrahydrofuran (0.855 mL) was added HATU (1.1 equiv., 36.5 mg, 0.0941 mmol) and N,N-diisopropylethylamine (3.0 equiv., 0.045 mL, 0.257 mmol). The mixture was stirred at room temperature for 30 minutes, then a solution of 4-[1-[4-[2- (methylamino)ethoxy]phenyl]-2-phenyl-but-1-enyl]phenol (1.05 equiv., 33.5 mg, 0.0898 mmol) in 2-methyltetrahydrofuran (60, 0.5 mL, 400 mg, 5 mmol) was added, followed by 0.2 mL DMF. The mixture was stirred at room temperature for 22 h. Water was added and the solution was extracted 3 times with iPrOAc. The organic layers were combined then dried with sodium sulfate and concentrated in vacuo.
The crude material was dissolved in dichloromethane (0.85 mL) and trifluoroacetic acid (0.26 mL) was added dropwise. The reaction was stirred at room temperature until no gas evolution was observed. After 1 h, the solution was concentrated in vacuo and purified by reverse- phase HPLC to obtain 45 mg (45 % yield over 2 steps) of the desired product.
M+H = 560.9, 1120.7; δ 1H NMR (400 MHz, DMSO-d6) δ 9.39, 9.14 (overlapping s, 1H), 8.85– 8.70 (m, 1H), 8.44– 8.36 (m, 1H), 8.20 (d, J = 8.8 Hz, 1H), 8.10 (d, J = 8.6 Hz, 1H), 7.97– 7.82 (m, 2H), 7.70– 7.64 (m, 1H), 7.50– 7.44 (m, 1H), 7.34– 7.27 (m, 1H), 7.21– 7.04 (m, 9H), 7.03– 6.87 (m, 4H), 6.78– 6.67 (m, 2H), 6.62– 6.54 (m, 2H), 6.44– 6.33 (m, 1H), 6.12 (d, J = 15.1 Hz, 1H), 5.78 (d, J = 8.0 Hz, 2H), 5.52 (d, J = 15.0 Hz, 1H), 4.99 - 4.89 (m, 1H), 4.33– 4.04 (m, 4H), 4.00– 3.88 (m, 1H), 3.94 (s, 3H), 3.68– 3.61 (m, 1H), 3.60– 3.42 (m, 5H), 3.38– 3.32 (m, 1H), 3.03– 2.78 (m, 3H), 2.45– 2.35 (m, 2H), 2.32 (s, 3H), 1.23 (d, J = 6.4 Hz, 3H), 1.11 (d, J = 6.1 Hz, 3H), 0.89– 0.76 (m, 4H).
B. Preparation of L1-PROTAC
iii. Attachment of Linker L1 to PROTAC
Figure imgf000173_0001
Figure imgf000173_0002
N-[(1S)-1-[[(1S)-4-(Carbamoylamino)-1-[(4-[4-[(1Z)-1-(4-[2-[2-(2-[2-[(4-[[(2S,3S)-8- cyano-5-[(2-methoxynaphthalen-1-yl)methyl]-2-methyl-3-[(2S)-2- (methylamino)propanamido]-4-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1- yl]carbonyl]phenyl)formamido]ethoxy]ethoxy)-N-methylacetamido]ethoxy]phenyl)-2- phenylbut-1-en-1-yl]phenoxymethyl]phenyl)carbamoyl]butyl]carbamoyl]-2- methylpropyl]-6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanamide (“compound LP2”) To a solution of (2S)-N-[(3S,4S)-7-cyano-5-[(4-[[2-(2-[[(2-[4-[(1Z)-1-(4-hydroxyphenyl)-2- phenylbut-1-en-1-yl]phenoxy]ethyl)(methyl)carbamoyl]methoxy]
ethoxy)ethyl]carbamoyl]phenyl)carbonyl]-1-[(2-methoxynaphthalen-1-yl)methyl]-4-methyl- 2-oxo-2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-3-yl]-2-(methylamino)propanamide
(compound P1, 48 mg, 0.043 mmol) and N-[(1S)-1-{[(1S)-4-(carbamoylamino)-1-{[4- (chloromethyl)phenyl]carbamoyl}butyl]carbamoyl} -2-methylpropyl]-6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanamide (90 mg, 0.152 mmol) in DMF (0.9 mL) at 0 °C was added K2CO3 (60 mg, 0.43 mmol). The reaction mixture was stirred for 4 h at 0 oC, and diluted with precooled DMF (0.9 mL). The solid was filtered off. The filtrate was purified by Preparative HPLC with the following conditions: Column, SunFire Prep C18 OBD Column, 19*150mm 5um, 10 nm; mobile phase, water (0.1%TFA) and CH3CN (5% CH3CN up to 48% in 10 min); Detector, UV 254/220 nm to afford 22 mg (31%) of N-[(1S)-1-[[(1S)-4- (carbamoylamino)-1-[(4-[4-[(1Z)-1-(4-[2-[2-(2-[2-[(4-[[(2S,3S)-8-cyano-5-[(2- methoxynaphthalen-1-yl)methyl]-2-methyl-3-[(2S)-2-(methylamino)propanamido]-4-oxo- 2,3,4,5-tetrahydro-1H-1,5-benzodiazepin-1-yl]carbonyl]phenyl)formamido]ethoxy]ethoxy)- N-methylacetamido]ethoxy]phenyl)-2-phenylbut-1-en-1- yl]phenoxymethyl]phenyl)carbamoyl]butyl]carbamoyl]-2-methylpropyl]-6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanamide as a white solid. MS (ESI): [M+H]+ = 1675.1; 1H-NMR (400 MHz, DMSO-d6): δ (ppm) 10.15 (s, 1H), 9.95-9.72 (m, 1H), 9.52-9.41 (m, 1H), 9.31- 9.14 (m, 1H), 8.43 (brs, 1H), 8.20-8.18 (m, 1H), 8.17-8.07 (m, 2H), 7.93 (m, 2H), 7.70 (d, J = 8 Hz, 1H), 7.70-7.65 (m, 3H), 7.48-7.42 (m, 2H), 7.41-7.36 (m, 1H), 7.33-7.28 (m, 1H), 7.23 (m, 1H), 7.18-7.07 (m, 7H), 7.02-7.01 (m, 1H), 6.99 (s, 3H), 6.96-6.92 (m, 2H), 6.76-6.67 (m, 2H), 6.60-6.58 (m, 2H), 6.40 (d, J = 8.4 Hz, 1H), 6.13-6.08 (m, 1H), 5.99 (s, 1H), 5.80 (d, J = 7.6 Hz, 2H), 5.55 (d, J = 15.2 Hz, 1H), 5.44 (brs, 1H), 5.02-4.93 (m, 1H), 7.43-4.36 (m, 3H), 4.26-4.02(m, 6H), 3.95-3.88 (m, 4H), 3.66-3.63 (m, 3H), 3.57-3.53 (m, 4H), 3.39-3.36 (m, 4H), 3.05-2.81 (m, 5H), 2.68 (s, 2H), 2.42-2.39 (m, 3H), 2.22-2.07 (m, 2H), 2.03-1.91 (m, 1H), 1.71 (brs, 2H), 1.62 (brs, 3H), 1.50-1.36 (m, 6H), 1.21-1.17 (m, 5H), 0.87-0.82 (m, 10H).
C. Preparation of PAC iv. Attachment of Antibody (Ab) to PROTAC via Linker L1 Conjugation of a PROTAC to HER2 and B7H4 antibodies to yield PROTAC- antibody conjugates (PACs) was accomplished as follows.
A cysteine-engineered antibody (THIOMAB™ antibody), in 10 mM succinate, pH 5, 150 mM NaCl, 2 mM EDTA, is pH-adjusted to pH 7.5-8.5 with 1M Tris. Between three and ten equivalents of a linker L1-PROTAC with a thiol-reactive group is dissolved in DMF or DMA and added to the reduced, reoxidized, and pH-adjusted antibody. The reaction is incubated at room temperature or 37°C and monitored until completion (1 to about 24 hours), as determined by LC-MS analysis of the reaction mixture. When the reaction is complete, the conjugate is purified by one or any combination of several methods, the goal being to remove remaining unreacted linker L1-PROTAC intermediate and aggregated protein (if present at significant levels). For example, the conjugate may be diluted with 10 mM histidine-acetate, pH 5.5 until final pH is approximately 5.5 and purified by S cation exchange chromatography using either HiTrap S columns connected to an Akta purification system (GE Healthcare) or S maxi spin columns (Pierce). Alternatively, the conjugate may be purified by gel filtration chromatography using an S200 column connected to an Akta purification system or Zeba spin columns. Alternatively, dialysis may be used.
The THIOMABTM antibody PROTAC conjugates were formulated into 20 mM His/acetate, pH 5, with 240 mM sucrose using either gel filtration or dialysis. The purified conjugate is concentrated by centrifugal ultrafiltration and filtered through a 0.2-µm filter under sterile conditions and frozen for storage.
Example 2
Characterization of PACs
The PACs were characterized to determine protein concentration (e.g., by BCA assay), aggregation level (by analytical SEC), PAR (e.g., by LC-MS).
Size exclusion chromatography is performed on conjugates using a Shodex KW802.5 column in 0.2M potassium phosphate pH 6.2 with 0.25 mM potassium chloride and 15% IPA at a flow rate of 0.75 ml/min. Aggregation state of the conjugate was determined by integration of eluted peak area absorbance at 280 nm.
LC-MS analysis is performed on conjugates using an Agilent TOF 6530 ESI instrument. As an example, the PAC is treated with 1:500 w/w Endoproteinase Lys C
(Promega) in Tris, pH 7.5, for 30 min at 37°C. The resulting cleavage fragments are loaded onto a 1000Å (Angstrom) 8 µm (micron) PLRP S (highly cross linked polystyrene) column heated to 80 °C and eluted with a gradient of 30% B to 40% B in 10 minutes. Mobile phase A was H2O with 0.05% TFA and mobile phase B was acetonitrile with 0.04% TFA. The flow rate was 0.5ml/min. Protein elution was monitored by UV absorbance detection at 280nm prior to electrospray ionization and MS analysis. Chromatographic resolution of the unconjugated Fc fragment, residual unconjugated Fab and PROTAC-conjugated Fab was usually achieved. The obtained m/z spectra were deconvoluted using Mass Hunter™ software (Agilent Technologies) to calculate the mass of the antibody fragments.
Table 28. Characterization of several PACs.
Figure imgf000176_0001
MC-VC-PAB refers to (6-maleimidocaproyl)-(valine-citrulline)-(p-aminobenzyl).
Thio means THIOMABTM antibody.
LC K149C means that the K at position 149 has been changed to C in the light chain Hu means human
Anti HER2 and Anti B7-H4 mean antibodies that bind to HER2 and B7-H4 respectively.
Example 3
Detection of Effects of a HER-2 containing PAC on Estrogen Receptor alpha Density The following experimental describes the detection of estrogen receptor alpha (ER-α) in MCF7-neo/HER2 cell lines by Western blot after treatment with PROTACs and PROTAC- antibody conjugates.
MCF7 cells engineered to express the HER2/NEU receptor were seeded in a 12 well plate with a cell density of 40x104 cells per well. To deplete estradiol, cells were grown in phenol red free RPMI media containing 10% (v/v) charcoal stripped serum (Gemini Bio- products), 4 mM L-glutamine, 100U each of penicillin and streptomycin and nonessential amino acids (Gibco, Life Technologies). After 3 days, cells were trypsinized and re-seeded at half the density.
The following day unconjugated PROTAC or PROTAC conjugated to antibody (PAC) was added to the medium at 10 µg/ml (=134 nM PROTAC) and 1 µg/ml (=13.4 nM PROTAC). After 3 days, cells were washed once with PBS and lysed in 100µL urea lysis buffer (6M Urea, 20mM Tris pH7.5, 12.5 mM NaCl, 2.5 mM MgCl2, 0.1% Triton X-100, protease inhibitor cocktail (Roche)). Total protein concentrations were determined by BCA (ThermoFisher). For each sample, 10ug of total cellular protein was separated on 4-12% Bis- Tris gel and transferred to Invitrolon PVDF membrane (Thermofisher). Membranes were blocked in PBS-0.1 % Tween-20 containing 10% non fat dry milk and probed with primary antibodies against Estrogen Receptor α (Santa Cruz, SC-8002) and GAPDH (Santa Cruz, SC25778 HRP) at 1:1000 dilution followed by secondary antibodies against mouse IgG (GE Healthcare) at 1:5000 dilution.
Protein bands were visualized using chemiluminescence (Perkin Elmer). The results are shown in Figure 1.
Example 4
Quantitation of Effects of a HER-2 containing PAC on Estrogen Receptor alpha Density The following experimental describes the quantitation of ERα in MCF7-neo/HER2 cells after treatment with PROTACs and PROTAC-antibody conjugates.
Cell plating neoMCF7/HER2 cells (CL130220) were thawed at 37 oC then transferred to growth media (RPMI, 10% Fetal Bovine Serum F2442 (FBS)) by spinning cells down twice at 1200 RCF for three minutes and removing and replacing supernatant with growth media. Cells were then transferred to a 150 cm2 flask (Ref: 431465) and grown until confluent. Once confluent, cells were washed once in PBS, PBS aspirated, and 10 ml of 10X 0.5% Trypsin (Cat# 15400-054) was added to the flask to cover cells. Excess trypsin was immediately aspirated and flask was transferred to 37 oC incubator for 5 min. After incubation 20 ml growth media (phenol red free-RPMI (Cat# 11835-030) 10% charcoal stripped-FBS (SKU: F6765)) was added to flask and cell density was determined using a Vicell instrument.
Growth media was added to make cellular density of 10,000 cells/0.05 ml.50 ul of cells were then transferred to each well of two Greiner 384 well plates (Ref: 781946) that were stored overnight in a 37 oC incubator before compound treatment the following day.
Compound treatment PACs were thawed at RT and were each diluted to 60 ug/ml in 37 C growth media, followed by a 20-point 2X serial dilution across a 384 well plate (Ref:
781091).10 ul of each sample from the serial dilution was transferred to the wells of the cell plates. The highest working concentration of the PACs was 10 ug/ml. Cell plate columns 1, 2, 23 and 24 were left untreated for data normalization while Columns 3-22 contained the PAC dilutions. After compound treatment cell plates were stored in a 37 C incubator for 72 h. Staining After the 72 h compound treatment, cells were fixed for 30 min with 15 ul of 16% paraformaldehyde (Cat# 15710-S) in each well. Contents were aspirated, 50 ul of Permeabilization / Block buffer (PBS, BSA 0.5% (w/v), Triton X1000.1% (v/v)), was added to each well. After 10 min the Permeabilization / Block buffer was aspirated and washed twice with PBS. Wells were then aspirated and treated with 25 ul 1/1000 of mAb Anti-ESR1 (Clone F10) (Santa Cruz sc-8002) in Permeabilization/Block Buffer and incubated at RT for 2 h before being washed 4X in PBS. Contents of the wells were aspirated again and treated with 25 ul 1/1000 Alexafluor 488 conjugated anti-mouse (LifeTechnologies #A21202) and 1/1000 Hoechst 33258 (Cat# H3569) and incubated for 3 h. Plates were then washed 3X in PBS and sealed.
Data Collection Cellomics Arrayscan was used to collect the cell count and ERα fluorescence intensity (MeanCircAvgIntenCh2), which is proportional to the amount of ERα present in the nucleus. Data was then normalized to non-treated cell controls, and plotted in GraphPad Prism. GraphPad Prism calculated the EC50s using the“lag(inhibitor) vs. response –Variable Slope” function.
Table 29 reports the IC50 data from the quantitation experiment.
Table 29.
Figure imgf000178_0001
Figure imgf000179_0001
The results are shown in Figure 2. Treatment of HER2 expressing cells with HER2 antibody containing PAC Anti-HER2(Endox-XIAP) resulted in a marked decreased Estrogen Receptor-alpha (ERα) levels. Treatment of the HER2 expressing cells with the B7-H4 antibody containing PAC Anti-B7-H4(Endox-XIAP) did not result in a substantial decrease in ERα levels.
Efforts have been made to ensure accuracy with respect to numbers used (e.g.
amounts, temperature, etc.) but some experimental errors and deviations should be accounted for.
One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practicing the subject matter described herein. The present disclosure is in no way limited to just 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 subject matter 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.
Throughout this specification and the claims, the words“comprise,”“comprises,” and “comprising” are used in a non-exclusive sense, except where the context requires otherwise. It is understood that embodiments described herein include“consisting of” and/or“consisting essentially of” embodiments.
As used herein, the term“about,” when referring to a value is meant to encompass variations of, in some embodiments ± 50%, in some embodiments ± 20%, in some embodiments ± 10%, in some embodiments ± 5%, in some embodiments ± 1%, in some embodiments ± 0.5%, and in some embodiments ± 0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods or employ the disclosed compositions. Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit, unless the context clearly dictates otherwise, between the upper and lower limit of the range and any other stated or intervening value in that stated range, is encompassed. The upper and lower limits of these small ranges which may independently be included in the smaller rangers is also encompassed, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included.
Many modifications and other embodiments set forth herein will come to mind to one skilled in the art to which this subject matter pertains having the benefit of the teachings presented in the foregoing descriptions and the associated drawings. Therefore, it is to be understood that the subject matter is not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.

Claims

What is Claimed is: 1. A conjugate having the chemical structure
Ab―(L1―D)p,
wherein,
D is a PROTAC having the structure E3LB―L2―PB;
E3LB is an E3 ligase binding group covalently bound to L2;
L2 is a linker covalently bound to E3LB and PB;
PB is a protein binding group covalently bound to L2;
Ab is an antibody covalently bound to L1;
L1 is a linker, covalently bound to Ab and D; and
p has a value from about 1 to about 8.
2. The conjugate of claim 1, wherein E3LB is a group that binds an E3 ligase, wherein said E3 ligase is listed in Tables 13-27.
3. The conjugate of claim 1, wherein E3LB is a group that binds an E3 ligase, wherein said E3 ligase is selected from the group consisting of von Hippel-Lindau (VHL); cereblon; XIAP; E3A; MDM2; Anaphase-promoting complex (APC); UBR5 (EDD1); SOCS/ BC-box/ eloBC/ CUL5/ RING; LNXp80; CBX4; CBLL1; HACE1; HECTD1; HECTD2; HECTD3; HECW1; HECW2; HERC1; HERC2; HERC3; HERC4; HUWE1; ITCH; NEDD4; NEDD4L; PPIL2; PRPF19; PIAS1; PIAS2; PIAS3; PIAS4; RANBP2; RNF4; RBX1;
SMURF1; SMURF2; STUB1; TOPORS; TRIP12; UBE3A; UBE3B; UBE3C; UBE4A;
UBE4B; UBOX5; UBR5; WWP1; WWP2; Parkin; A20/TNFAIP3; AMFR/gp78; ARA54; beta-TrCP1/BTRC; BRCA1; CBL; CHIP/STUB1; E6; E6AP/UBE3A; F-box protein
15/FBXO15; FBXW7/Cdc4; GRAIL/RNF128; HOIP/RNF31; cIAP-1/HIAP-2; cIAP- 2/HIAP-1; cIAP (pan); ITCH/AIP4; KAP1; MARCH8;; Mind Bomb 1/MIB1; Mind Bomb 2/MIB2; MuRF1/TRIM63; NDFIP1; NEDD4; NleL; Parkin; RNF2; RNF4; RNF8; RNF168; RNF43; SART1; Skp2; SMURF2; TRAF-1; TRAF-2; TRAF-3; TRAF-4; TRAF-5; TRAF-6; TRIM5; TRIM21; TRIM32; UBR5; and ZNRF3.
4. The conjugate of claim 1, wherein E3LB is a group that binds an E3 ligase selected from the group consisting of XIAP, VHL, cereblon and MDM2.
5. The conjugate of claim 1, wherein E3LB is selected from the group consisting of a compound that binds VHL, a hydroxyproline compound that binds VHL, a compound that binds MDM2, a compound that binds cereblon, a tetrahhydro-benzodiazepinone a nutlin, thalidomide, lenalidomide, and pomalidomide.
6. The conjugate of claim 1, wherein E3LB is a XIAP inhibitor that is a tetrahydro-benzodiazepinone having the formula:
Figure imgf000182_0001
,
wherein, R1, R2, R3, R4 and R5 are as described in WO/2015/071393.
7. The conjugate of claim 1, wherein PB is a group that binds FoxOl, HDAC, DP-1, E2F, ABL, AMPK, BRK, BRSK I, BRSK2, BTK, CAMKK1, CAMKK alpha, CAMKK beta, Rb, Suv39HI, SCF, p19INK4D, GSK-3, pi 8 INK4, myc, cyclin E, CDK2, CDK9, CDG4/6, Cycline D, pl6 INK4A, cdc25A, BMI1, SCF, Akt, CHKl/2, C 1 delta, CK1 gamma, C 2, CLK2, CSK, DDR2, DYRK1A/2/3, EF2K, EPH-A2/A4/B1/B2/B3/B4, EIF2A 3, Smad2, Smad3, Smad4, Smad7, p53, p21 Cipl, PAX, Fyn, CAS, C3G, SOS, Tal, Raptor, RACK-1, CRK, Rapl, Rac, KRas, NRas, HRas, GRB2, FAK, PI3K, spred, Spry, mTOR, MPK, LKBl, PAK 1/2/4/5/6, PDGFRA, PYK2, Src, SRPK1, PLC, PKC, PKA, PKB alpha/beta, PKC alpha/gamma/zeta, PKD, PLKl, PRAK, PRK2, RIPK2, WAVE-2, TSC2, DAPKl, BAD, IMP, C-TAK1, TAKl, TAOl, TBK1, TESK1, TGFBR1, TIE2, TLK1, TrkA, TSSK1, TTBK1/2, TTK, Tpl2/cotl, MEK1, MEK2, PLDL Erkl, Erk2, Erk5, Erk8, p90RSK, PEA- 15, SRF, p27 KIP1, TIF la, HMGN1, ER81, MKP-3, c-Fos, FGF-R1, GCK, GSK3 beta, HER4, HIPK1/2/3/, IGF-1R, cdc25, UBF, LAMTOR2, Statl, StaO, CREB, JAK, Src, PTEN, NF- kappaB, HECTH9, Bax, HSP70, HSP90, Apaf-1, Cyto c, BCL-2, Bcl-xL, Smac, XIAP, Caspase-9, Caspase-3, Caspase-6, Caspase-7, CDC37, TAB, IKK, TRADD, TRAF2, R1P1, FLIP, TAKl, JNKl/2/3, Lck, A-Raf, B-Raf, C-Raf, MOS, MLKl/3, MN l/2, MSKl, MST2/3/4, MPSK1, MEKKl , ME K4, MEL , ASK1, MINK1 , MKK 1 /2/3/4/6/7, NE 2a/6/7, NUAK1, OSR1, SAP , STK33, Syk, Lyn, PDK1, PHK, PIM 1/2/3, Ataxin- 1, mTORCl, MDM2, p21 Wafl, Cyclin Dl, Lamln A, Tpl2, Myc, catenin, Wnt, IKK-beta, IKK- gamma, IKK-alpha, IKK-epsilon, ELK, p65RelA, IRAKI, IRA 2, IRAK4, IRR, FADD, TRAF6, TRAF3, MKK3, MKK6, ROCK2, RSK1/2, SGK 1, SmMLCK, SIK2/3, ULK1/2, VEGFR1, WNK l , YES1, ZAP70, MAP4K3, MAP4K5, MAPKlb, MAPKAP-K2 K3, p38 alpha/beta/delta/gamma MAPK, Aurora A, Aurora B, Aurora C, MCAK, Clip, MAPKAPK, FAK, MARK 1 /2/3/4, Mucl , SHC, CXCR4, Gap-1, Myc, beta-catenin/TCF, Cbl, BRM, Mcl1, BRD2, BRD3, BRD4, AR, RAS, ErbB3, EGFR, IRE1, HPK1, RIPK2, and ERα, including all variants, mutations, splice variants, indels and fusions thereof.
8. The conjugate of claim 1, wherein PB is selected from the group consisting of Heat Shock Protein 90 (HSP90) inhibitors, Kinase and Phosphatase inhibitors, MDM2 inhibitors, HDAC inhibitors, Human Lysine Methyltransferase Inhibitors, Angiogenesis inhibitors, Immunosuppressive compounds, and compounds that target: Human BET Bromodomain-containing proteins, the aryl hydrocarbon receptor (AHR), REF receptor kinase, FKBP, Androgen Receptor (AR), Estrogen receptor (ER), Thyroid Hormone Receptor, HIV Protease, HIV Integrase, HCV Protease, and Acyl-protein Thioesterase-1 and -2 (APT1 and APT2).
9. The conjugate of claim 1, wherein PB is a group that targets Estrogen Receptor alpha (ERa).
10. The conjugate of claim 1, wherein said Ab is a cysteine engineered antibody or variant thereof.
11. The conjugate of claim 1, wherein Ab binds to one or more of polypeptides selected from the group consisting of DLL3, EDAR, CLL1; BMPR1B; E16; STEAP1;
0772P; MPF; NaPi2b; Sema 5b; PSCA hlg; ETBR; MSG783; STEAP2; TrpM4; CRIPTO; CD21; CD79b; FcRH2; B7-H4; HER2; NCA; MDP; IL20Rα; Brevican; EphB2R;
ASLG659; PSCA; GEDA; BAFF-R; CD22; CD79a; CXCR5; HLA-DOB; P2X5; CD72; LY64; FcRH1; IRTA2; TENB2; PMEL17; TMEFF1; GDNF-Ra1; Ly6E; TMEM46;
Ly6G6D; LGR5; RET; LY6K; GPR19; GPR54; ASPHD1; Tyrosinase; TMEM118;
GPR172A; MUC16 and CD33.
12. The conjugate of claim 10, wherein Ab binds to one or more of polypeptides selected from the group consisting of CLL1 STEAP1 NaPi2b STEAP2 TrpM4 CRIPTO CD21, CD79b, FcRH2, B7-H4, HER2, CD22, CD79a, CD72, LY64, Ly6E, MUC16, and CD33.
13. The conjugate of claim 12, wherein Ab is an antibody that binds to one or more polypeptides selected from the group consisting of B7-H4, HER2, CLL1, CD33, CD22 and NaPi2b.
14. The conjugate of claim 12, wherein the antibody binds to HER2 or B7-H4.
15. The conjugate of claim 14, wherein the antibody binds to HER2.
16. The conjugate of claim 1, wherein L1 is a peptidomimetic linker.
17. The conjugate of claim 16, wherein L1 is a peptidomimetic linker represented by the following formula:
—Str—(PM)—Sp— wherein, Str is a stretcher unit covalently attached to Ab; Ab is an antibody; Sp is a bond or spacer unit covalently attached to a PROTAC moiety; PM is a non-peptide chemical moiety selected from the group consisting of:
Figure imgf000185_0001
Figure imgf000185_0002
Figure imgf000185_0003
W is–NH-heterocycloalkyl- or heterocycloalkyl; Y is heteroaryl, aryl, -C(O)C1-C6alkylene, C1-C6alkylene-NH2, C1-C6alkylene-NH-CH3, C1- C6alkylene-N-(CH3)2, C1-C6alkenyl or C1-C6alkylenyl; each R1 is independently C1-C10alkyl, C1-C10alkenyl, (C1-C6alkyl)NHC(NH)NH2, (C1- C6alkyl)NHC(O)NH2, (C1-C10alkyl)NHC(NH)NH2 or (C1-C10alkyl)NHC(O)NH2; R3 and R2 are each independently H, C1-C10alkyl, C1-C10alkenyl, arylalkyl or heteroarylalkyl, or R3 and R2 together may form a C3-C7cycloalkyl; and R4 and R5 are each independently C1-C10alkyl, C1-C10alkenyl, arylalkyl, heteroarylalkyl, (C1- C10alkyl )OCH2-, or R4 and R5 together may form a C3-C7cycloalkyl ring.
18. The conjugate of claim 17, wherein Y is heteroaryl; R4 and R5 together form a cyclobutyl ring.
19. The conjugate of claim 17, wherein Y is a moiety selected from the group consisting of
Figure imgf000186_0001
.
20. The conjugate of claim 17, wherein
Str is a chemical moiety represented by the following formula:
Figure imgf000186_0002
wherein R6 is selected from the group consisting of C1-C10alkylene, C1-C10alkenyl, C3- C8cycloalkyl, (C1-C8alkylene)O-, and C1-C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4- C7heterocycloalkyl, heteroarylalkyl, aryl arylalkyl, heteroarylalkyl and heteroaryl each Ra is independently H or C1-C6alkyl; and Sp is -C1-C6alkylene-C(O)NH- or—Ar—Rb—, wherein Ar is aryl or heteroaryl, and Rb is (C1-C10alkylene)O-.
21. The conjugate of claim 17, wherein Str has the formula:
wherein R7 is selected from C1-C10alkylene, C1-C10alkenyl, (C1-C10alkylene)O-, N(Rc)−(C2- C6 alkylene)−N(Rc) and N(Rc)−(C2-C6alkylene); where each Rc is independently H or C1-C6 alkyl; and Sp is -C1-C6alkylene-C(O)NH- or—Ar—Rb—, wherein Ar is aryl or heteroaryl, and Rb is (C1-C10alkylene)O-.
22. The conjugate of claim 17, wherein L1 has the following formula
Figure imgf000187_0001
R1 is C1-C6alkyl, C1-C6alkenyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2; R3 and R2 are each independently H, C1-C10alkyl.
23. The conjugate of claim 17, wherein L1 has the following formula:
Figure imgf000187_0002
R1 is C1-C6alkyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2;
R4 and R5 together form a C3-C7cycloalkyl ring.
24. The conjugate of claim 17, wherein L1 has the following formula:
Figure imgf000187_0003
R1 is C1-C6alkyl, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2.
25. The conjugate compound of claim 1, having the formula:
Figure imgf000188_0001
wherein Sp is a bond or spacer unit covalently attached to PROTAC moiety D; Y is heteroaryl, aryl, -C(O)C1-C6alkylene, C1-C6alkylene-NH2, C1-C6alkylene-NH-CH3, C1- C6alkylene-N-(CH3)2, C1-C6alkenyl or C1-C6alkylenyl; R1 is independently C1-C10alkyl, C1-C10alkenyl, (C1-C6alkyl)NHC(NH)NH2, (C1- C6alkyl)NHC(O)NH2, (C1-C10alkyl)NHC(NH)NH2 or (C1-C10alkyl)NHC(O)NH2; R3 and R2 are each independently H, C1-C10alkyl, C1-C10alkenyl, arylalkyl or heteroarylalkyl, or R3 and R2 together may form a C3-C7cycloalkyl; and Str is a chemical moiety represented by the following formula:
Figure imgf000188_0002
R6 is selected from the group consisting of C1-C10alkylene, and C1- C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4-C7heterocycloalkyl, heteroarylalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each Ra is independently H or C1-C6alkyl; p is 1, 2, 3 or 4.
26. The conjugate of claim 1, having the formula:
Figure imgf000189_0001
wherein Sp is a bond or spacer unit covalently attached to PROTAC moiety D; R4 and R5 are each independently C1-C10alkyl, C1-C10alkenyl, arylalkyl, heteroarylalkyl, (C1- C10alkyl )OCH2-, or R4 and R5 together may form a C3-C7cycloalkyl ring R1 is independently C1-C10alkyl, C1-C10alkenyl, (C1-C6alkyl)NHC(NH)NH2, (C1- C6alkyl)NHC(O)NH2, (C1-C10alkyl)NHC(NH)NH2 or (C1-C10alkyl)NHC(O)NH2; Str is a chemical moiety represented by the following formula:
Figure imgf000189_0002
R6 is selected from the group consisting of C1-C10alkylene, and C1- C10alkylene−C(O)N(Ra)−C2-C6alkylene, where each alkylene may be substituted by one to five substituents selected from the group consisting of halo, trifluoromethyl, difluoromethyl, amino, alkylamino, cyano, sulfonyl, sulfonamide, sulfoxide, hydroxy, alkoxy, ester, carboxylic acid, alkylthio, C3-C8cycloalkyl, C4-C7heterocycloalkyl, aryl, arylalkyl, heteroarylalkyl and heteroaryl each Ra is independently H or C1-C6alkyl; p is 1, 2, 3 or 4.
27. The conjugate of claim 25, wherein Y is heteroaryl, aryl or alkenyl; R6 is C1- C10alkylene.
28. The conjugate of claim 25, wherein Y is
Figure imgf000190_0001
.
29. The conjugate of claim 2 h r in Y is
Figure imgf000190_0002
.
30. The conjugate of claim 25, wherein Y is
Figure imgf000190_0003
.
31. The conjugate of claim 25, wherein
Str is a chemical moiety represented by the following formula:
Figure imgf000190_0004
,
R6 is C1-C6alkylene;
Sp is -C1-C6alkylene-C(O)NH- or—Ar—Rb—, where Ar is aryl, Rb is (C1-C3alkylene)O-.
32. The conjugate of claim 1, having the formula:
Figure imgf000191_0001
wherein Y is heteroaryl, aryl, -C(O)C1-C6alkylene, C1-C6alkylene-NH2, C1-C6alkylene-NH-CH3, C1- C6alkylene-N-(CH3)2, C1-C6alkenyl or C1-C6alkylenyl; R1 is C1-C6alkyl-NH2, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2; R3 and R2 are each independently H, C1-C10alkyl, C1-C10alkenyl, arylalkyl or heteroarylalkyl, or R3 and R2 together may form a C3-C7cycloalkyl; and p is 1, 2 ,3 or 4.
33. The conjugate of claim 1, having the formula:
Figure imgf000191_0002
wherein p is 1, 2, 3 or 4; R1 is C1-C6alkyl-NH2, (C1-C6alkyl)NHC(NH)NH2 or (C1-C6alkyl)NHC(O)NH2; R4 and R5 are each independently C1-C6alkyl, wherein said alkyl are unsubstituted, or R4 andR5 may form a C3-C7cycloalkyl ring.
34. The conjugate of claim 1, wherein L1 has the following formula:
Figure imgf000192_0001
,
wherein, R1 and R2 are independently selected from H and C1-C6 alkyl, or R1 and R2 form a 3, 4, 5, or 6-membered cycloalkyl or heterocyclyl group.
35. The conjugate of claim 1 selected from the group consisting of PAC1, PAC2, PAC3, PAC4 and PAC5.
36. The conjugate of claim 1, wherein p is from about 1.0 to about 3.
37. The conjugate of claim 1, wherein p is about 2.
38. A pharmaceutical composition comprising a conjugate of claim 1 and one or more pharmaceutically acceptable excipients.
39. A method of treating a disease in a human in need thereof, comprising administering to said human an effective amount of a conjugate of claim 1 or a composition of claim 38.
40. The method of claim 39, wherein said disease is cancer.
41. The method of claim 40, wherein said cancer is selected from the group consisting of a carcinoma, lymphoma, blastoma, sarcoma, leukemia, lymphoid malignancies, squamous cell cancer, lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, and head and neck cancer.
42. The method of claim 41, wherein the cancer is a HER2-positive cancer.
43. The method of claim 42, wherein the HER2-positive cancer is breast cancer or gastric cancer.
44. A method of claim 39, wherein said disease is an autoimmune disease.
45. The method of claim 44, wherein the autoimmune disease is selected from the group consisting of rheumatologic disorders, osteoarthritis, autoimmune gastrointestinal and liver disorders, vasculitis, autoimmune neurological disorders, renal disorders, autoimmune dermatologic disorders, hematologic disorders, atherosclerosis, uveitis, autoimmune hearing diseases, Behcet's disease, Raynaud's syndrome, organ transplant, autoimmune endocrine disorders, Addison’s disease, and autoimmune thyroid disease.
46. The method of claim 45, wherein the autoimmune disease is selected from the group consisiting of rheumatoid arthritis, ulcerative colitis, ANCA-associated vasculitis, lupus, multiple sclerosis, Sjögren's syndrome, Graves’ disease, IDDM, pernicious anemia, thyroiditis, and glomerulonephritis.
PCT/US2017/033611 2016-05-20 2017-05-19 Protac antibody conjugates and methods of use WO2017201449A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
PL17728325T PL3458101T3 (en) 2016-05-20 2017-05-19 Protac antibody conjugates and methods of use
EP17728325.6A EP3458101B1 (en) 2016-05-20 2017-05-19 Protac antibody conjugates and methods of use
JP2018560504A JP2019522633A (en) 2016-05-20 2017-05-19 PROTAC antibody conjugates and methods of use
CN201780030893.6A CN109152843A (en) 2016-05-20 2017-05-19 PROTAC antibody conjugates and its application method
ES17728325T ES2858151T3 (en) 2016-05-20 2017-05-19 PROTAC-Antibody Conjugates and Procedures for Use
US16/194,897 US11666581B2 (en) 2016-05-20 2018-11-19 PROTAC antibody conjugates and methods of use
JP2022107984A JP2022126886A (en) 2016-05-20 2022-07-04 Protac-antibody conjugates and methods of use
US18/310,913 US20230330102A1 (en) 2016-05-20 2023-05-02 Protac antibody conjugates and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662339257P 2016-05-20 2016-05-20
US62/339,257 2016-05-20

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/194,897 Continuation US11666581B2 (en) 2016-05-20 2018-11-19 PROTAC antibody conjugates and methods of use

Publications (1)

Publication Number Publication Date
WO2017201449A1 true WO2017201449A1 (en) 2017-11-23

Family

ID=59014750

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/033611 WO2017201449A1 (en) 2016-05-20 2017-05-19 Protac antibody conjugates and methods of use

Country Status (7)

Country Link
US (2) US11666581B2 (en)
EP (1) EP3458101B1 (en)
JP (2) JP2019522633A (en)
CN (1) CN109152843A (en)
ES (1) ES2858151T3 (en)
PL (1) PL3458101T3 (en)
WO (1) WO2017201449A1 (en)

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108047312A (en) * 2017-12-05 2018-05-18 北京大学深圳研究生院 A kind of stable polypeptide targeting proteins chimer molecules and its preparation method and application
CN108690137A (en) * 2018-05-31 2018-10-23 山东省医学科学院基础医学研究所 M8Sac71 antibody and the preparation method and application thereof
WO2018227023A1 (en) * 2017-06-07 2018-12-13 Silverback Therapeutics, Inc. Antibody construct conjugates
WO2019133531A1 (en) * 2017-12-26 2019-07-04 Kymera Therapeutics, Inc. Irak degraders and uses thereof
WO2019140003A1 (en) * 2018-01-10 2019-07-18 Development Center For Biotechnology Antibody protac conjugates
US10450310B2 (en) 2016-04-22 2019-10-22 Dana-Farber Cancer Institute, Inc. Bifunctional molecules for degradation of EGFR and methods of use
WO2019207538A1 (en) 2018-04-26 2019-10-31 Aurigene Discovery Technologies Limited Pyridazine derivatives as smarca2/4 degraders
CN110407770A (en) * 2018-04-27 2019-11-05 复旦大学 3- substitution -1,5- benzo-aza * class compound and its medicinal usage
WO2020027225A1 (en) 2018-07-31 2020-02-06 ファイメクス株式会社 Heterocyclic compound
WO2020073930A1 (en) * 2018-10-09 2020-04-16 嘉兴优博生物技术有限公司 Targeted protease degradation platform
WO2020086858A1 (en) * 2018-10-24 2020-04-30 Genentech, Inc. Conjugated chemical inducers of degradation and methods of use
US10646575B2 (en) 2016-05-10 2020-05-12 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
US10660968B2 (en) 2016-05-10 2020-05-26 C4 Therapeutics, Inc. Spirocyclic degronimers for target protein degradation
WO2020191377A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Extracellular vesicle conjugates and uses thereof
WO2020191369A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
WO2020207396A1 (en) * 2019-04-09 2020-10-15 Ranok Therapeutics (Hangzhou) Co. Ltd. Methods and compositions for targeted protein degradation
US10849982B2 (en) 2016-05-10 2020-12-01 C4 Therapeutics, Inc. C3-carbon linked glutarimide degronimers for target protein degradation
CN112029866A (en) * 2020-09-21 2020-12-04 河南尚泰科诺生物科技有限公司 Application of WWP1 in pancreatic cancer
WO2021016521A1 (en) * 2019-07-25 2021-01-28 Beth Israel Deaconess Medical Center, Inc. Photo induced control of protein destruction
WO2021020585A1 (en) 2019-07-31 2021-02-04 ファイメクス株式会社 Heterocyclic compound
JP2021506814A (en) * 2017-12-15 2021-02-22 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Degradation of target proteins mediated by stabilizing peptides
US11117889B1 (en) 2018-11-30 2021-09-14 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
EP3878472A1 (en) 2020-03-12 2021-09-15 Julius-Maximilians-Universitaet Wuerzburg Proteolysis targeting chimera (protac) for degradation of aurora a-kinase
WO2021237100A1 (en) 2020-05-21 2021-11-25 Codiak Biosciences, Inc. Methods of targeting extracellular vesicles to lung
WO2022020288A1 (en) * 2020-07-21 2022-01-27 Genentech, Inc. Antibody-conjugated chemical inducers of degradation of brm and methods thereof
US11254672B2 (en) 2017-09-04 2022-02-22 C4 Therapeutics, Inc. Dihydrobenzimidazolones for medical treatment
WO2022066928A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
KR20220042136A (en) 2019-07-31 2022-04-04 피맥스 가부시키가이샤 heterocyclic compound
US11292792B2 (en) 2018-07-06 2022-04-05 Kymera Therapeutics, Inc. Tricyclic CRBN ligands and uses thereof
US11358948B2 (en) 2017-09-22 2022-06-14 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
WO2022148459A1 (en) * 2021-01-11 2022-07-14 和径医药科技(上海)有限公司 Class of novel smad3 protein degraders and application thereof
EP3867395A4 (en) * 2018-10-16 2022-07-20 Technology Innovation Momentum Fund (Israel) Limited Partnership Systems and method for screening small molecules of interest
US11401256B2 (en) 2017-09-04 2022-08-02 C4 Therapeutics, Inc. Dihydroquinolinones for medical treatment
US11459335B2 (en) 2017-06-20 2022-10-04 C4 Therapeutics, Inc. N/O-linked Degrons and Degronimers for protein degradation
US11485743B2 (en) 2018-01-12 2022-11-01 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11485750B1 (en) 2019-04-05 2022-11-01 Kymera Therapeutics, Inc. STAT degraders and uses thereof
US11512080B2 (en) 2018-01-12 2022-11-29 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11524949B2 (en) 2017-11-16 2022-12-13 C4 Therapeutics, Inc. Degraders and Degrons for targeted protein degradation
WO2023275394A1 (en) 2021-07-02 2023-01-05 Merck Patent Gmbh Anti-protac antibodies and complexes
US11584748B2 (en) 2018-04-16 2023-02-21 C4 Therapeutics, Inc. Spirocyclic compounds
US11591332B2 (en) 2019-12-17 2023-02-28 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11597768B2 (en) 2017-06-26 2023-03-07 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
WO2023056069A1 (en) * 2021-09-30 2023-04-06 Angiex, Inc. Degrader-antibody conjugates and methods of using same
US11623932B2 (en) 2017-09-22 2023-04-11 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11623929B2 (en) 2018-06-04 2023-04-11 C4 Therapeutics, Inc. Spirocyclic compounds
US11666581B2 (en) 2016-05-20 2023-06-06 Genentech, Inc. PROTAC antibody conjugates and methods of use
WO2023104941A1 (en) 2021-12-08 2023-06-15 European Molecular Biology Laboratory Hydrophilic tetrazine-functionalized payloads for preparation of targeting conjugates
US11679109B2 (en) 2019-12-23 2023-06-20 Kymera Therapeutics, Inc. SMARCA degraders and uses thereof
US11685750B2 (en) 2020-06-03 2023-06-27 Kymera Therapeutics, Inc. Crystalline forms of IRAK degraders
US11701357B2 (en) 2016-08-19 2023-07-18 Beigene Switzerland Gmbh Treatment of B cell cancers using a combination comprising Btk inhibitors
US11707457B2 (en) 2019-12-17 2023-07-25 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
WO2023147328A1 (en) 2022-01-26 2023-08-03 Genentech, Inc. Antibody-conjugated chemical inducers of degradation with hydolysable maleimide linkers and methods thereof
US11753397B2 (en) 2018-03-26 2023-09-12 C4 Therapeutics, Inc. Cereblon binders for the degradation of ikaros
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
US11802131B2 (en) 2017-09-04 2023-10-31 C4 Therapeutics, Inc. Glutarimides for medical treatment
US11865114B2 (en) 2019-09-12 2024-01-09 Aurigene Oncology Limited Method for treating diseases using SMARCA2/4 degraders
US11932846B2 (en) 2017-08-08 2024-03-19 Technology Innovation Momentum Fund (Israel) Limited Partnership Chloramphenicol resistant split protein and uses thereof
US11932624B2 (en) 2020-03-19 2024-03-19 Kymera Therapeutics, Inc. MDM2 degraders and uses thereof

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2022536602A (en) * 2019-05-14 2022-08-18 ジェネンテック, インコーポレイテッド Methods of using anti-CD79B immunoconjugates to treat follicular lymphoma
CN112010858B (en) * 2019-05-31 2023-07-18 四川海思科制药有限公司 BTK inhibitor, preparation method and pharmaceutical application thereof
WO2020257730A1 (en) 2019-06-19 2020-12-24 Flagship Pioneering Innovations Vi, Llc Compositions comprising circular polyribonucleotides for protein modulation and uses thereof
EP4010081A4 (en) * 2019-08-08 2023-07-26 Institute For Cancer Research d/b/a The Research Institute of Fox Chase Cancer Center Combination therapy for treatment of cancer
CN112552293A (en) * 2019-09-25 2021-03-26 珠海宇繁生物科技有限责任公司 PROTAC small molecular compound and application thereof
CN111298106B (en) * 2019-11-25 2022-04-08 清华大学 Conjugate and application thereof in degradation of beta-catenin protein
CN110885332B (en) * 2019-12-06 2022-03-18 中国人民解放军第二军医大学 PDE delta protein degradation targeting chimera and preparation method and application thereof
EP4096715A1 (en) 2020-01-29 2022-12-07 Flagship Pioneering Innovations VI, LLC Compositions comprising linear polyribonucleotides for protein modulation and uses thereof
WO2021219077A1 (en) * 2020-04-29 2021-11-04 泰比棣医药科技(石家庄)有限公司 Proteolysis targeting compound having tissue targeting capability and use thereof
CN111437391B (en) * 2020-05-29 2022-04-08 南通大学 Application of knock-down circHECTD1 in preparation of drug for treating glioma
TW202228775A (en) 2020-10-14 2022-08-01 美商維里迪恩醫療股份有限公司 Compositions and methods for treatment of thyroid eye disease
CN112375150A (en) * 2020-11-25 2021-02-19 天津科技大学 Application of binary polypeptide compound
TW202304524A (en) 2021-04-10 2023-02-01 美商普方生物製藥美國公司 Folr1 binding agents, conjugates thereof and methods of using the same
EP4326768A1 (en) 2021-04-23 2024-02-28 Profoundbio Us Co. Anti-cd70 antibodies, conjugates thereof and methods of using the same
KR20220167240A (en) * 2021-06-11 2022-12-20 주식회사 나이벡 Bio PROTAC Protein having Intracellular Delivery Function and Pharmaceutical Composition Comprising the Same
TW202320857A (en) 2021-07-06 2023-06-01 美商普方生物製藥美國公司 Linkers, drug linkers and conjugates thereof and methods of using the same
CN114917359B (en) * 2022-05-15 2023-03-21 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) PROTAC composition aiming at cell cycle multi-space-time distribution anti-cancer targets
WO2023229535A1 (en) 2022-05-27 2023-11-30 Engine Biosciences Pte. Ltd. Bet polypeptide therapeutic agents for generating synthetic lethality in tumors

Citations (465)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US798959A (en) 1904-12-19 1905-09-05 George W Goss Corn-husker.
US4120649A (en) 1975-04-10 1978-10-17 Israel Schechter Transplants
US4665077A (en) 1979-03-19 1987-05-12 The Upjohn Company Method for treating rejection of organ or skin grafts with 6-aryl pyrimidine compounds
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0340109A2 (en) 1988-04-28 1989-11-02 The Board Of Trustees Of The Leland Stanford Junior University Anti-T-cell receptor determinants as autoimmune disease treatment
WO1990008187A1 (en) 1989-01-19 1990-07-26 Dana Farber Cancer Institute Soluble two domain cd2 protein
WO1990011294A1 (en) 1989-03-21 1990-10-04 The Immune Response Corporation Vaccination and methods against diseases resulting from pathogenic responses by specific t cell populations
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1991001133A1 (en) 1989-07-19 1991-02-07 Arthur Allen Vandenbark T cell receptor peptides as therapeutics for autoimmune and malignant disease
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
WO1992007574A1 (en) 1990-10-25 1992-05-14 Tanox Biosystems, Inc. Glycoproteins associated with membrane-bound immunoglobulins as antibody targets on b cells
US5114721A (en) 1988-03-15 1992-05-19 Yeda Research And Development Co. Ltd. Preparation of t-cell and t-cell membrane for use in prevention and treatment of autoimmune diseases
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 & CO. LTD. A human endothelin receptor
JPH053790A (en) 1990-04-19 1993-01-14 Fujisawa Pharmaceut Co Ltd Dehydropeptidase-i
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1994010312A1 (en) 1992-10-23 1994-05-11 Chugai Seiyaku Kabushiki Kaisha Gene coding for megakaryocyte potentiator
WO1994028931A1 (en) 1993-06-11 1994-12-22 Genentech, Inc. Methods for treating inflammatory disorders
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5440021A (en) 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
WO1996002642A1 (en) 1994-07-20 1996-02-01 University Of Dundee INTERRUPTION OF BINDING OF MDM2 AND p53 PROTEIN AND THERAPEUTIC APPLICATION THEREOF
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
WO1996027011A1 (en) 1995-03-01 1996-09-06 Genentech, Inc. A method for making heteromultimeric polypeptides
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
WO1997007198A2 (en) 1995-08-11 1997-02-27 Genetics Institute, Inc. Dna sequences and secreted proteins encoded thereby
WO1997009343A2 (en) 1995-09-04 1997-03-13 Rhone-Poulenc Rorer S.A. Antagonists of the oncogenic activity of the protein mdm2, and use thereof in the treatment of cancers
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
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
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
WO1997044452A1 (en) 1996-05-17 1997-11-27 Schering Corporation Human b-cell antigens, related reagents
WO1998001467A2 (en) 1996-07-05 1998-01-15 Novartis Ag Inhibitors of the interaction between p53 and mdm2
US5750373A (en) 1990-12-03 1998-05-12 Genentech, Inc. Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5773223A (en) 1993-09-02 1998-06-30 Chiron Corporation Endothelin B1, (ETB1) receptor polypeptide and its encoding nucleic acid methods, and uses thereof
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
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)
WO1998050431A2 (en) 1997-05-02 1998-11-12 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components
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
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
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
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
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
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
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
WO1999055858A2 (en) 1998-04-28 1999-11-04 Metagen Gesellschaft Für Genomforschung Mbh Human nucleic acid sequences obtained from pancreas tumor tissue
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
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
WO2000015657A1 (en) 1998-09-12 2000-03-23 Astrazeneca Ab Piperizine-4-phenyl derivatives as inhibitors of the interaction between mdm2 and 53
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
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
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
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
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
WO2001000244A2 (en) 1999-06-25 2001-01-04 Genentech, Inc. METHODS OF TREATMENT USING ANTI-ErbB ANTIBODY-MAYTANSINOID CONJUGATES
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
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
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
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
WO2001040309A2 (en) 1999-10-29 2001-06-07 Genentech, Inc. Anti-prostate stem cell antigen (psca) antibody compositions and methods of use
WO2001040269A2 (en) 1999-11-30 2001-06-07 Corixa Corporation Compositions and methods for 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
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
WO2001046232A2 (en) 1999-12-23 2001-06-28 Zymogenetics, Inc. Soluble interleukin-20 receptor
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
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
WO2002002634A2 (en) 2000-06-30 2002-01-10 Incyte Genomics, Inc. Human extracellular matrix and cell adhesion polypeptides
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
WO2002010382A2 (en) 2000-07-28 2002-02-07 Ulrich Wissenbach Trp8, trp9 and trp10, markers for cancer
WO2002010187A1 (en) 2000-07-27 2002-02-07 Mayo Foundation For Medical Education And Research B7-h3 and b7-h4, novel immunoregulatory molecules
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
US20020034749A1 (en) 1997-11-18 2002-03-21 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
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
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
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
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
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
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
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
WO2002072596A1 (en) 2001-03-09 2002-09-19 Incyte Genomics, Inc. Steap-related protein
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
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
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
WO2002099122A1 (en) 2001-06-05 2002-12-12 Exelixis, Inc. 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
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
WO2003003906A2 (en) 2001-07-03 2003-01-16 Eos Biotechnology, Inc. Diagnostic and screening methods for bladder 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
WO2003004529A2 (en) 2001-07-02 2003-01-16 Licentia Ltd. Ephrin-tie receptor materials and methods
WO2003003984A2 (en) 2001-07-05 2003-01-16 Curagen Corporation Novel proteins and nucleic acids encoding same
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
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
WO2003014294A2 (en) 2001-08-03 2003-02-20 Genentech, Inc. Tacis and br3 polypeptides and uses thereof
WO2003016494A2 (en) 2001-08-16 2003-02-27 Vitivity, Inc. Diagnosis and treatment of vascular disease
WO2003016475A2 (en) 2001-08-14 2003-02-27 The General Hospital Corporation Nucleic acid and amino acid sequences involved in pain
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
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
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
US20030060612A1 (en) 1997-10-28 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
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
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
WO2003029262A2 (en) 2001-08-29 2003-04-10 Vanderbilt University The human mob-5 (il-24) receptors and uses thereof
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
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
US20030096961A1 (en) 2001-06-01 2003-05-22 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
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
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
WO2003051359A1 (en) 2001-12-18 2003-06-26 F.Hoffmann-La Roche Ag Cis-2,4,5- triphenyl-imidazolines and their use in the treatment of tumors
US20030119131A1 (en) 2000-01-20 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
US20030119130A1 (en) 1999-08-17 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
US20030119128A1 (en) 1999-07-20 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
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
WO2003051360A1 (en) 2001-12-18 2003-06-26 F. Hoffmann-La Roche Ag Cis-imidazolines as mdm2 inhibitors
US20030124140A1 (en) 1998-12-17 2003-07-03 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2003054152A2 (en) 2001-12-10 2003-07-03 Nuvelo, Inc. Novel nucleic acids and polypeptides
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
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
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
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
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US20030157089A1 (en) 1997-02-25 2003-08-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20030165504A1 (en) 1999-09-24 2003-09-04 Retter Marc W. Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2003072035A2 (en) 2002-02-22 2003-09-04 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO2003072036A2 (en) 2002-02-21 2003-09-04 Duke University Treatment methods using anti-cd22 antibodies
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
WO2003081210A2 (en) 2002-03-21 2003-10-02 Sunesis Pharmaceuticals, Inc. Identification of kinase inhibitors
US20030185830A1 (en) 1997-02-25 2003-10-02 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
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
WO2003083041A2 (en) 2002-03-22 2003-10-09 Biogen, Inc. Cripto-specific antibodies
WO2003083047A2 (en) 2002-03-01 2003-10-09 Exelixis, Inc. MP53s AS MODIFIERS OF THE p53 PATHWAY AND METHODS OF USE
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
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
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
WO2003087306A2 (en) 2002-04-05 2003-10-23 Agensys, Inc. Nucleic acid and corresponding protein entitled 98p4b6 useful in treatment and detection of cancer
WO2003087768A2 (en) 2002-04-12 2003-10-23 Mitokor Targets for therapeutic intervention identified in the mitochondrial proteome
WO2003089904A2 (en) 2002-04-17 2003-10-30 Baylor College Of Medicine Aib1 as a prognostic marker and predictor of resistance to encocrine therapy
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
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
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
WO2003097803A2 (en) 2002-05-15 2003-11-27 Avalon Pharmaceuticals Cancer-linked gene as target for chemotherapy
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
WO2003104275A2 (en) 2002-06-06 2003-12-18 Oncotherapy Science, Inc. Genes and polypeptides relating to human colon cancers
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
US20030232056A1 (en) 1999-09-10 2003-12-18 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2003104399A2 (en) 2002-06-07 2003-12-18 Avalon Pharmaceuticals, Inc Cancer-linked gene as target for chemotherapy
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
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
WO2004000221A2 (en) 2002-06-20 2003-12-31 The Regents Of The University Of California Compositions and methods for modulating lymphocyte activity
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
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
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
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
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2004042346A2 (en) 2002-04-24 2004-05-21 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
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
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
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
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
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
WO2004047749A2 (en) 2002-11-21 2004-06-10 University Of Utah Research Foundation Purinergic modulation of smell
WO2004053079A2 (en) 2002-12-06 2004-06-24 Diadexus, Inc. Compositions, splice variants and methods relating to ovarian specific genes and proteins
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
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
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
WO2004063709A2 (en) 2003-01-08 2004-07-29 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
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
WO2004080460A1 (en) 2003-03-13 2004-09-23 F. Hoffmann-La Roche Ag SUBSTITUTED PIPERIDINES AS NOVEL MDM2-p53 INHIBITORS
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
WO2005002575A1 (en) 2003-06-17 2005-01-13 F. Hoffmann-La Roche Ag Cis-imidazolines as mdm2 inhibitors
WO2005003097A1 (en) 2003-06-17 2005-01-13 F. Hoffmann-La Roche Ag Cis-2,4,5-triaryl-imidazolines
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US20050048572A1 (en) 2002-10-31 2005-03-03 Genentech, Inc. Methods and compositions for increasing antibody production
US20050079574A1 (en) 2003-01-16 2005-04-14 Genentech, Inc. Synthetic antibody phage libraries
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
US20050119455A1 (en) 2002-06-03 2005-06-02 Genentech, Inc. Synthetic antibody phage libraries
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
WO2005110996A1 (en) 2004-05-18 2005-11-24 F.Hoffmann-La Roche Ag Novel cis-imidazolines
US20050266000A1 (en) 2004-04-09 2005-12-01 Genentech, Inc. Variable domain library and uses
WO2005123691A1 (en) 2004-06-17 2005-12-29 F-Hoffmann-La Roche Ag Novel cis-imidazolines
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006032631A1 (en) 2004-09-22 2006-03-30 Janssen Pharmaceutica N.V. Inhibitors of the interaction between mdm2 and p53
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
WO2006091646A2 (en) 2005-02-22 2006-08-31 The Regents Of The University Of Michigan Small molecule inhibitors of mdm2 and uses thereof
WO2006097261A1 (en) 2005-03-16 2006-09-21 F.Hoffmann-La Roche Ag Cis-2,4,5-triaryl-imidazolines and their use as anti-cancer medicaments
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2006136606A2 (en) 2005-06-24 2006-12-28 F. Hoffmann-La Roche Ag Oxindole derivatives
WO2007001851A2 (en) 2005-06-20 2007-01-04 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7189826B2 (en) 1997-11-24 2007-03-13 Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
US20070061900A1 (en) 2000-10-31 2007-03-15 Murphy Andrew J Methods of modifying eukaryotic cells
US7208157B2 (en) 2000-09-08 2007-04-24 California Institute Of Technology Proteolysis targeting chimeric pharmaceutical
US20070117126A1 (en) 1999-12-15 2007-05-24 Genentech, Inc. Shotgun scanning
WO2007063013A1 (en) 2005-12-01 2007-06-07 F. Hoffmann-La Roche Ag 2,4,5-triphenyl imidazoline derivatives as inhibitors of the interaction between p53 and mdm2 proteins for use as anticancer agents
US20070160598A1 (en) 2005-11-07 2007-07-12 Dennis Mark S Binding polypeptides with diversified and consensus vh/vl hypervariable sequences
WO2007082805A1 (en) 2006-01-18 2007-07-26 F. Hoffmann-La Roche Ag Cis-4, 5-biaryl-2-heterocyclic-imidazolines as mdm2 inhibitors
WO2007104714A1 (en) 2006-03-13 2007-09-20 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2007104664A1 (en) 2006-03-13 2007-09-20 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2007107545A1 (en) 2006-03-22 2007-09-27 Janssen Pharmaceutica N.V. Cyclic-alkylaminederivatives as inhibitors of the interaction between mdm2 and p53
WO2007107543A1 (en) 2006-03-22 2007-09-27 Janssen Pharmaceutica N.V. Inhibitors of the interaction between mdm2 and p53
US20070237764A1 (en) 2005-12-02 2007-10-11 Genentech, Inc. Binding polypeptides with restricted diversity sequences
US20070292936A1 (en) 2006-05-09 2007-12-20 Genentech, Inc. Binding polypeptides with optimized scaffolds
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008036168A2 (en) 2006-08-30 2008-03-27 The Regents Of The University Of Michigan New small molecule inhibitors of mdm2 and the uses thereof
WO2008034736A2 (en) 2006-09-21 2008-03-27 F. Hoffmann-La Roche Ag Oxindole derivatives as anticancer agents
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
WO2008055812A1 (en) 2006-11-09 2008-05-15 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2008072655A1 (en) 2006-12-14 2008-06-19 Daiichi Sankyo Company, Limited Imidazothiazole derivatives
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2008106507A2 (en) 2007-02-27 2008-09-04 University Of South Florida Mdm2/mdmx inhibitor peptide
WO2008125487A1 (en) 2007-04-12 2008-10-23 F. Hoffmann-La Roche Ag Diphenyl-dihydro-imidazopyridinones
WO2008130614A2 (en) 2007-04-20 2008-10-30 University Of Pittsburg-Of The Commonwealth System Of Higher Education Selective and dual-action p53/mdm2/mdm4 antagonists
WO2008141917A1 (en) 2007-05-17 2008-11-27 F. Hoffmann-La Roche Ag 3,3-spiroindolinone derivatives
WO2008141975A1 (en) 2007-05-23 2008-11-27 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
US20090002360A1 (en) 2007-05-25 2009-01-01 Innolux Display Corp. Liquid crystal display device and method for driving same
WO2009019274A1 (en) 2007-08-06 2009-02-12 Janssen Pharmaceutica Nv Substituted phenylenediamines as inhibitors of the interaction between mdm2 and p53
US7498298B2 (en) 2003-11-06 2009-03-03 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2009037343A1 (en) 2007-09-21 2009-03-26 Janssen Pharmaceutica Nv Inhibitors of the interaction between mdm2 and p53
WO2009037308A1 (en) 2007-09-21 2009-03-26 Janssen Pharmaceutica Nv Inhibitors of the interaction between mdm2 and p53
WO2009047161A1 (en) 2007-10-09 2009-04-16 F. Hoffmann-La Roche Ag Chiral cis-imidazolines
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2009052249A1 (en) 2007-10-19 2009-04-23 Genentech, Inc. Cysteine engineered anti-tenb2 antibodies and antibody drug conjugates
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
WO2009077357A1 (en) 2007-12-14 2009-06-25 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2009080488A1 (en) 2007-12-19 2009-07-02 F. Hoffmann-La Roche Ag Spiroindolinone derivatives as anticancer agents
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
US20090182127A1 (en) 2006-06-22 2009-07-16 Novo Nordisk A/S Production of Bispecific Antibodies
WO2009115425A1 (en) 2008-03-19 2009-09-24 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2009151069A1 (en) 2008-06-12 2009-12-17 第一三共株式会社 Imidazothiazole derivative having 4,7-diazaspiro[2.5]octane ring structure
WO2010009124A2 (en) 2008-07-15 2010-01-21 Genentech, Inc. Anthracycline derivative conjugates, process for their preparation and their use as antitumor compounds
WO2010028862A1 (en) 2008-09-15 2010-03-18 Priaxon Ag Pyrrolidin-2-ones as hdm2 ligands
US7723485B2 (en) 2007-05-08 2010-05-25 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
WO2010058819A1 (en) 2008-11-19 2010-05-27 学校法人 慶應義塾 Peptide capable of inhibiting interaction between human tumor protein mdm2 and human tumor suppression protein p53, and use thereof
WO2010082612A1 (en) 2009-01-16 2010-07-22 第一三共株式会社 Imidazothiazole derivative having proline ring structure
WO2010084097A1 (en) 2009-01-26 2010-07-29 F. Hoffmann-La Roche Ag Spiroindolinone derivative prodrugs
WO2010091979A1 (en) 2009-02-10 2010-08-19 F. Hoffmann-La Roche Ag Spiroindolinone pyridine derivative
WO2010094622A1 (en) 2009-02-17 2010-08-26 F. Hoffmann-La Roche Ag 3, 3' -spiroindolinone derivatives as anticancer agents
WO2010121995A1 (en) 2009-04-23 2010-10-28 F. Hoffmann-La Roche Ag 3,3'-spiroindolinone derivatives and their use for cancer
US7862817B2 (en) 1999-06-25 2011-01-04 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
WO2011005219A1 (en) 2009-07-07 2011-01-13 Agency For Science, Technology And Research Novel mdm2 binding peptides and uses thereof
WO2011023677A1 (en) 2009-08-26 2011-03-03 Novartis Ag Tetra-substituted heteroaryl compounds and their use as mdm2 and/or mdm4 modulators
WO2011045257A1 (en) 2009-10-14 2011-04-21 F. Hoffmann-La Roche Ag Novel n-substituted-pyrrolidines as inhibitors of mdm2-p-53 interactions
WO2011060049A2 (en) 2009-11-12 2011-05-19 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists
WO2011061139A1 (en) 2009-11-17 2011-05-26 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
WO2011067185A1 (en) 2009-12-02 2011-06-09 F. Hoffmann-La Roche Ag Spiroindolinone pyrrolidines
WO2011076786A1 (en) 2009-12-22 2011-06-30 Novartis Ag Substituted isoquinolinones and quinazolinones
WO2011098398A1 (en) 2010-02-09 2011-08-18 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
WO2011101297A1 (en) 2010-02-17 2011-08-25 F. Hoffmann-La Roche Ag Substituted spiroindolinones
WO2011106650A2 (en) 2010-02-27 2011-09-01 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Novel p53-mdm2/p53-mdm4 antagonists to treat proliferative disease
WO2011134925A1 (en) 2010-04-28 2011-11-03 F. Hoffmann-La Roche Ag Spiroindolinone pyrrolidines
US20110287009A1 (en) 2010-04-23 2011-11-24 Genentech, Inc. Production of Heteromultimeric Proteins
WO2011153509A1 (en) 2010-06-04 2011-12-08 Amgen Inc. Piperidinone derivatives as mdm2 inhibitors for the treatment of cancer
WO2012007409A1 (en) 2010-07-12 2012-01-19 F. Hoffmann-La Roche Ag N-substituted pyrrolidines
WO2012022707A1 (en) 2010-08-18 2012-02-23 F. Hoffmann-La Roche Ag Substituted heteroaryl spiropyrrolidine mdm2 antagonists
WO2012033525A2 (en) 2010-09-08 2012-03-15 University Of Pittsburgh - Of The Commonwealth System Of Higher Education P53-mdm2 antagonists
WO2012034954A1 (en) 2010-09-15 2012-03-22 F. Hoffmann-La Roche Ag Substituted hexahydropyrrolo [1, 2 - c] imidazolones useful as mdm2 inhibitors
WO2012038307A1 (en) 2010-09-20 2012-03-29 F. Hoffmann-La Roche Ag Spiro substituted pyrrolo[1,2-c]imidazole derivatives useful as mdm2 inhibitors
WO2012047587A2 (en) 2010-09-27 2012-04-12 Serrata, Llc Mdm2 inhibitors for treatment of ocular conditions
WO2012065022A2 (en) 2010-11-12 2012-05-18 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists
WO2012066095A1 (en) 2010-11-19 2012-05-24 Novartis Ag Crystalline form of an inhibitor of mdm2/4 and p53 interaction
WO2012076513A1 (en) 2010-12-09 2012-06-14 F. Hoffmann-La Roche Ag 3-cyano-1-hydroxymethyl-2-phenylpyrrolidine derivatives as inhibitors of mdm2-p53 interactions useful for the treatment of cancer
US8226945B2 (en) 2006-05-30 2012-07-24 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
WO2012106587A1 (en) 2011-02-04 2012-08-09 Genentech, Inc. Fc VARIANTS AND METHODS FOR THEIR PRODUCTION
WO2012116989A1 (en) 2011-03-03 2012-09-07 F. Hoffmann-La Roche Ag Substituted heteroaryl 2', 3', 7', 7a' - tetrahydrospiro-[pyrrole-3, 6' - pyrrolo [1, 2-c] imidazole] - 1', 2 (1h, 5'h) -diones as anticancer agents
WO2012121361A1 (en) 2011-03-10 2012-09-13 第一三共株式会社 Dispiropyrrolidine derivative
WO2012155066A2 (en) 2011-05-11 2012-11-15 Shaomeng Wang Spiro-oxindole mdm2 antagonists
WO2012176123A1 (en) 2011-06-22 2012-12-27 Novartis Ag 3 - imidazolyl- indoles for the treatment of proliferative diseases
WO2012175487A1 (en) 2011-06-20 2012-12-27 Novartis Ag Cyclohexyl isoquinolinone compounds
WO2012175520A1 (en) 2011-06-20 2012-12-27 Novartis Ag Hydroxy substituted isoquinolinone derivatives
WO2013017705A1 (en) 2011-08-03 2013-02-07 Salvador Moreno Rufino Baltasar Panel system for construction with backlighting based on light-emitting diodes
WO2013049250A1 (en) 2011-09-27 2013-04-04 Amgen Inc. Heterocyclic compounds as mdm2 inhibitors for the treatment of cancer
WO2013055987A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2013080141A1 (en) 2011-11-29 2013-06-06 Novartis Ag Pyrazolopyrrolidine compounds
WO2013096150A1 (en) 2011-12-21 2013-06-27 Merck Sharp & Dohme Corp. Substituted piperidines as hdm2 inhibitors
WO2013106643A2 (en) 2012-01-12 2013-07-18 Yale University Compounds & methods for the enhanced degradation of targeted proteins & other polypeptides by an e3 ubiquitin ligase
WO2013106646A2 (en) 2012-01-12 2013-07-18 Yale University Compounds and methods for the inhibition of vcb e3 ubiquitin ligase
WO2013105037A1 (en) 2012-01-09 2013-07-18 Universidade Do Porto Inhibitors of p53-mdm2 interaction
WO2013111105A1 (en) 2012-01-26 2013-08-01 Novartis Ag Imidazopyrrolidinone compounds
WO2013135648A1 (en) 2012-03-15 2013-09-19 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
WO2013177055A2 (en) 2012-05-21 2013-11-28 Genentech, Inc. ANTI-Ly6E ANTIBODIES AND IMMUNOCONJUGATES AND METHODS OF USE
WO2013175417A1 (en) 2012-05-24 2013-11-28 Novartis Ag Pyrrolopyrrolidinone compounds
WO2013178570A1 (en) 2012-05-30 2013-12-05 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
WO2014038606A1 (en) 2012-09-06 2014-03-13 第一三共株式会社 Crystal of dispiropyrrolidine derivative
WO2014044622A1 (en) * 2012-09-19 2014-03-27 F. Hoffmann-La Roche Ag 2-oxo-2,3,4,5-tetrahydro-1 h-benzo[b]diazepines and their use in the treatment of cancer
WO2014044401A2 (en) 2012-09-24 2014-03-27 Glanzstoff Bohemia S.R.O. Yarn having flame-retardant effect and textile fabric formed therefrom
US8697650B2 (en) 2010-02-16 2014-04-15 Medimmune, Llc HSA-related compositions and methods of use
WO2014063061A1 (en) 2012-10-19 2014-04-24 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US20140135270A1 (en) 2012-11-09 2014-05-15 Ensemble Therapeutics Corp. Macrocyclic compounds for inhibition of inhibitors of apoptosis
WO2014082889A1 (en) 2012-11-28 2014-06-05 F. Hoffmann-La Roche Ag Novel imidazolines as dual inhibitors of mdm2 and mdmx
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
WO2014100071A2 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted pyrrolopyrimidines as hdm2 inhibitors
WO2014115080A1 (en) 2013-01-22 2014-07-31 Novartis Ag Pyrazolo[3,4-d]pyrimidinone compounds as inhibitors of the p53/mdm2 interaction
WO2014115077A1 (en) 2013-01-22 2014-07-31 Novartis Ag Substituted purinone compounds
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
WO2014123882A1 (en) 2013-02-07 2014-08-14 Merck Sharp & Dohme Corp. 2,6,7 substituted purines as hdm2 inhibitors
WO2014130470A1 (en) 2013-02-19 2014-08-28 Amgen Inc. Cis-morpholinone and other compounds as mdm2 inhibitors for the treatment of cancer
WO2014134207A2 (en) 2013-03-01 2014-09-04 Google Inc. Auxiliary content suggestions relating to user generated content
WO2014151863A1 (en) 2013-03-14 2014-09-25 Amgen Inc. Heteroaryl acid morpholinone compounds as mdm2 inhibitors for the treatment of cancer
WO2014165506A1 (en) * 2013-04-01 2014-10-09 Immunomedics, Inc. Anti-mucin antibodies for early detection and treament of pancreatic cancer
US8883771B2 (en) 2012-08-23 2014-11-11 The Regents Of The University Of Michigan Bivalent inhibitors of IAP proteins and therapeutic methods using the same
WO2014187777A1 (en) 2013-05-21 2014-11-27 Mediapharma S.R.L. Novel inhibitors of pvhl-elongin c binding
WO2014191896A1 (en) 2013-05-27 2014-12-04 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
WO2014198266A1 (en) 2013-06-12 2014-12-18 Schaeffler Technologies Gmbh & Co. Kg Bicycle drive and bicycle comprising a bicycle drive
WO2014200937A1 (en) 2013-06-10 2014-12-18 Amgen Inc. Processes of making and crystalline forms of a mdm2 inhibitor
WO2015004610A1 (en) 2013-07-11 2015-01-15 Adamed Sp. Z O.O. 1,5-dihydropyrrol-2-one derivatives as inhibitors of p53-mdm2/mdm4 protein-protein interaction
WO2015023355A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
US8969526B2 (en) 2011-03-29 2015-03-03 Roche Glycart Ag Antibody Fc variants
WO2015070224A2 (en) 2013-11-11 2015-05-14 Amgen Inc. Combination therapy including an mdm2 inhibitor and one or more additional pharmaceutically active agents for the treatment of cancers
WO2015071393A1 (en) 2013-11-18 2015-05-21 F. Hoffmann-La Roche Ag Tetrahydro-benzodiazepinones
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095223A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095124A1 (en) 2013-12-16 2015-06-25 Genentech Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US9096544B2 (en) 2009-12-23 2015-08-04 Wayne State University Therapeutic compounds
US20150246882A1 (en) 2012-10-11 2015-09-03 Hoffmann-La Roche Inc. Indolines
US20150252072A1 (en) 2012-10-11 2015-09-10 Hoffmann-La Roche Inc. Azaindolines
US20150259359A1 (en) 2012-10-19 2015-09-17 Astex Therapeutics Limited Bicyclic heterocycle compounds and their uses in therapy
US20150266879A1 (en) 2012-10-11 2015-09-24 Hoffmann-La Roche Inc. Azaindolines
US20150284427A1 (en) 2012-11-09 2015-10-08 Bristol-Myers Squibb Company Macrocyclic compounds for inhibition of inhibitors of apoptosis
WO2015155332A1 (en) 2014-04-11 2015-10-15 Boehringer Ingelheim International Gmbh Spiro[3h-indole-3,2'-pyrrolidin]-2(1h)-one derivatives and their use as mdm2-p53 inhibitors
US20150291562A1 (en) 2014-04-14 2015-10-15 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
WO2015161032A1 (en) 2014-04-17 2015-10-22 The Regents Of The University Of Michigan Mdm2 inhibitors and therapeutic methods using the same
WO2015158648A1 (en) 2014-04-15 2015-10-22 F. Hoffmann-La Roche Ag Solid forms of a pharmaceutically active compound
US20150307499A1 (en) 2012-11-30 2015-10-29 Sanford-Burnham Medical Research Institute Inhibitor of apoptosis protein (iap) antagonists
WO2015187998A2 (en) 2014-06-04 2015-12-10 Sanford-Burnham Medical Research Institute Use of inhibitor of apoptosis protein (iap) antagonists in hiv therapy
WO2015189799A1 (en) 2014-06-12 2015-12-17 Adamed Sp. Z O.O. Compounds comprising 1,1',2,5'-tetrahydrospiro[indole-3,2'-pyrrole]-2,5'-dione system as inhibitors p53-mdm2 protein-protein interaction
US20150374678A1 (en) 2014-06-27 2015-12-31 Celgene Corporation Compositions and methods for inducing conformational changes in cereblon and other e3 ubiquitin ligases
WO2016001376A1 (en) 2014-07-03 2016-01-07 Boehringer Ingelheim International Gmbh New spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors
US20160024055A1 (en) 2012-12-11 2016-01-28 Hoffmann-La Roche Inc. Dimeric compounds
US9249153B2 (en) 2011-03-18 2016-02-02 Pusan National University Industry-University Cooperation Foundation Pharmaceutical composition for treating aging-associated diseases, containing progerin expression inhibitor as active ingredient, and screening method of said progerin expression inhibitor
US9249151B2 (en) 2013-08-23 2016-02-02 Boehringer Ingelheim International Gmbh Bis-amido pyridines
US20160045607A1 (en) 2014-08-11 2016-02-18 Yale University Estrogen-related receptor alpha based protac compounds and associated methods of use
WO2016026937A1 (en) 2014-08-21 2016-02-25 Boehringer Ingelheim International Gmbh New spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors
WO2016028391A2 (en) 2014-08-18 2016-02-25 Hudson Biopharma Inc. Spiropyrrolidines as mdm2 inhibitors
US20160058872A1 (en) 2014-04-14 2016-03-03 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
US9278978B2 (en) 2013-08-23 2016-03-08 Boehringer Ingelheim International Gmbh 6-Alkynyl Pyridine

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011130499A1 (en) * 2010-04-15 2011-10-20 Genentech, Inc. Anti-polyubiquitin antibodies and methods of use
WO2015081282A1 (en) * 2013-11-27 2015-06-04 Redwood Bioscience, Inc. Hydrazinyl-pyrrolo compounds and methods for producing a conjugate
JP6730261B2 (en) * 2014-09-17 2020-07-29 ジェネンテック, インコーポレイテッド Immune complex containing anti-HER2 antibody
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use

Patent Citations (492)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US798959A (en) 1904-12-19 1905-09-05 George W Goss Corn-husker.
US4120649A (en) 1975-04-10 1978-10-17 Israel Schechter Transplants
US4665077A (en) 1979-03-19 1987-05-12 The Upjohn Company Method for treating rejection of organ or skin grafts with 6-aryl pyrimidine compounds
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
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
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5114721A (en) 1988-03-15 1992-05-19 Yeda Research And Development Co. Ltd. Preparation of t-cell and t-cell membrane for use in prevention and treatment of autoimmune diseases
EP0340109A2 (en) 1988-04-28 1989-11-02 The Board Of Trustees Of The Leland Stanford Junior University Anti-T-cell receptor determinants as autoimmune disease treatment
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
WO1990008187A1 (en) 1989-01-19 1990-07-26 Dana Farber Cancer Institute Soluble two domain cd2 protein
WO1990011294A1 (en) 1989-03-21 1990-10-04 The Immune Response Corporation Vaccination and methods against diseases resulting from pathogenic responses by specific t cell populations
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1991001133A1 (en) 1989-07-19 1991-02-07 Arthur Allen Vandenbark T cell receptor peptides as therapeutics for autoimmune and malignant disease
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
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US6417429B1 (en) 1989-10-27 2002-07-09 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
JPH053790A (en) 1990-04-19 1993-01-14 Fujisawa Pharmaceut Co Ltd Dehydropeptidase-i
US5792616A (en) 1990-05-29 1998-08-11 The United States Of America Antibodies to human cripto protein
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1992007574A1 (en) 1990-10-25 1992-05-14 Tanox Biosystems, Inc. Glycoproteins associated with membrane-bound immunoglobulins as antibody targets on b cells
US5750373A (en) 1990-12-03 1998-05-12 Genentech, Inc. Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
WO1992017497A1 (en) 1991-03-29 1992-10-15 Genentech, Inc. Human pf4a receptors and their use
US5440021A (en) 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
EP0522868A1 (en) 1991-07-12 1993-01-13 SHIONOGI SEIYAKU KABUSHIKI KAISHA trading under the name of SHIONOGI & 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
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
US6153408A (en) 1991-11-15 2000-11-28 Institut Pasteur And Institut National De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
US6011146A (en) 1991-11-15 2000-01-04 Institut Pasteur Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
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
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1994010312A1 (en) 1992-10-23 1994-05-11 Chugai Seiyaku Kabushiki Kaisha Gene coding for megakaryocyte potentiator
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
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
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5773223A (en) 1993-09-02 1998-06-30 Chiron Corporation Endothelin B1, (ETB1) receptor polypeptide and its encoding nucleic acid methods, and uses thereof
WO1996002642A1 (en) 1994-07-20 1996-02-01 University Of Dundee INTERRUPTION OF BINDING OF MDM2 AND p53 PROTEIN AND THERAPEUTIC APPLICATION THEREOF
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
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
WO1996027011A1 (en) 1995-03-01 1996-09-06 Genentech, Inc. A method for making heteromultimeric polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
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
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
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
WO1997009343A2 (en) 1995-09-04 1997-03-13 Rhone-Poulenc Rorer S.A. Antagonists of the oncogenic activity of the protein mdm2, and use thereof in the treatment of cancers
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
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
WO1997044452A1 (en) 1996-05-17 1997-11-27 Schering Corporation Human b-cell antigens, related reagents
WO1998001467A2 (en) 1996-07-05 1998-01-15 Novartis Ag Inhibitors of the interaction between p53 and mdm2
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
US6555339B1 (en) 1997-04-14 2003-04-29 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human protein-coupled receptors
US20030105292A1 (en) 1997-04-14 2003-06-05 Liaw Chen W. Non-endogenous, constitutively activated human G protein-coupled receptors
EP0875569A1 (en) 1997-04-28 1998-11-04 Smithkline Beecham Corporation A human sodium dependent phosphate transporter (IPT-1)
WO1998050431A2 (en) 1997-05-02 1998-11-12 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components
US20070178552A1 (en) 1997-05-02 2007-08-02 Genentech, Inc. Method for Making Multispecific Antibodies Having Heteromultimeric and Common Components
WO1998051824A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting disease of the urinary tract
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
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US20030060612A1 (en) 1997-10-28 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
US20020034749A1 (en) 1997-11-18 2002-03-21 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
US7189826B2 (en) 1997-11-24 2007-03-13 Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
WO1999028468A1 (en) 1997-12-02 1999-06-10 The Regents Of The University Of California Modulating b lymphocyte chemokine/receptor interactions
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
WO1999046284A2 (en) 1998-03-13 1999-09-16 The Burnham Institute Molecules that home to various selected organs or tissues
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO1999055858A2 (en) 1998-04-28 1999-11-04 Metagen Gesellschaft Für Genomforschung Mbh Human nucleic acid sequences obtained from pancreas tumor tissue
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
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
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
WO2000015657A1 (en) 1998-09-12 2000-03-23 Astrazeneca Ab Piperizine-4-phenyl derivatives as inhibitors of the interaction between mdm2 and 53
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
US20030124140A1 (en) 1998-12-17 2003-07-03 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
WO2000036107A2 (en) 1998-12-17 2000-06-22 Corixa Corporation Compositions and methods for therapy and diagnosis of ovarian cancer
US20030065143A1 (en) 1998-12-30 2003-04-03 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2000040614A2 (en) 1998-12-30 2000-07-13 Beth Israel Deaconess Medical Center, Inc. Characterization of the soc/crac calcium channel protein family
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US7332581B2 (en) 1999-01-15 2008-02-19 Genentech, Inc. Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
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
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US20030119122A1 (en) 1999-05-11 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2000075655A1 (en) 1999-06-03 2000-12-14 Takeda Chemical Industries, Ltd. Screening method with the use of cd100
US7862817B2 (en) 1999-06-25 2011-01-04 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
US20030206918A1 (en) 1999-09-10 2003-11-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030129192A1 (en) 1999-09-10 2003-07-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
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
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
WO2001040309A2 (en) 1999-10-29 2001-06-07 Genentech, Inc. Anti-prostate stem cell antigen (psca) antibody compositions and methods of use
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
US20070117126A1 (en) 1999-12-15 2007-05-24 Genentech, Inc. Shotgun scanning
US20020042366A1 (en) 1999-12-23 2002-04-11 Penny Thompson Method for treating inflammation
WO2001046232A2 (en) 1999-12-23 2001-06-28 Zymogenetics, Inc. Soluble interleukin-20 receptor
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
US20040005320A1 (en) 1999-12-23 2004-01-08 Penny Thompson Method for treating inflammation
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
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
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
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
WO2002002634A2 (en) 2000-06-30 2002-01-10 Incyte Genomics, Inc. Human extracellular matrix and cell adhesion polypeptides
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
US7208157B2 (en) 2000-09-08 2007-04-24 California Institute Of Technology Proteolysis targeting chimeric pharmaceutical
WO2002022660A2 (en) 2000-09-11 2002-03-21 Hyseq, Inc. Novel nucleic acids and polypeptides
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
US20030186373A1 (en) 2000-09-15 2003-10-02 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
US20040005598A1 (en) 2000-09-26 2004-01-08 Genentech, Inc. PUMPCn compositions and uses thereof
WO2002026822A2 (en) 2000-09-26 2002-04-04 Genentech, Inc. Pumpcn compositions and uses thereof
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
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
US20070061900A1 (en) 2000-10-31 2007-03-15 Murphy Andrew J Methods of modifying eukaryotic cells
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
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
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
WO2002081646A2 (en) 2001-04-06 2002-10-17 Mannkind Corporation Epitope sequences
WO2003008537A2 (en) 2001-04-06 2003-01-30 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
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
WO2003000842A2 (en) 2001-06-04 2003-01-03 Curagen Corporation Novel proteins and nucleic acids encoding 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
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
US20030091580A1 (en) 2001-06-18 2003-05-15 Mitcham Jennifer L. Compositions and methods for the therapy and diagnosis of ovarian 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
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
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
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
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
WO2003025148A2 (en) 2001-09-19 2003-03-27 Nuvelo, Inc. Novel nucleic acids and polypeptides
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
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
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
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
WO2003051359A1 (en) 2001-12-18 2003-06-26 F.Hoffmann-La Roche Ag Cis-2,4,5- triphenyl-imidazolines and their use in the treatment of tumors
WO2003051360A1 (en) 2001-12-18 2003-06-26 F. Hoffmann-La Roche Ag Cis-imidazolines as mdm2 inhibitors
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
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
WO2003083041A2 (en) 2002-03-22 2003-10-09 Biogen, Inc. Cripto-specific antibodies
EP1347046A1 (en) 2002-03-22 2003-09-24 Research Association for Biotechnology Full-length cDNA sequences
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
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2003087768A2 (en) 2002-04-12 2003-10-23 Mitokor Targets for therapeutic intervention identified in the mitochondrial proteome
US20040101874A1 (en) 2002-04-12 2004-05-27 Mitokor Inc. Targets for therapeutic intervention identified in the mitochondrial proteome
WO2003088808A2 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20030228319A1 (en) 2002-04-16 2003-12-11 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)
US20050119455A1 (en) 2002-06-03 2005-06-02 Genentech, Inc. Synthetic antibody phage libraries
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
WO2004001004A2 (en) 2002-06-21 2003-12-31 Johns Hopkins University School Of Medicine Membrane associated tumor endothelium markers
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
EP1394274A2 (en) 2002-08-06 2004-03-03 Genox Research, Inc. Methods of testing for bronchial asthma or chronic obstructive pulmonary disease
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
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
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
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
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
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
US20050079574A1 (en) 2003-01-16 2005-04-14 Genentech, Inc. Synthetic antibody phage libraries
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
WO2004080460A1 (en) 2003-03-13 2004-09-23 F. Hoffmann-La Roche Ag SUBSTITUTED PIPERIDINES AS NOVEL MDM2-p53 INHIBITORS
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
WO2005002575A1 (en) 2003-06-17 2005-01-13 F. Hoffmann-La Roche Ag Cis-imidazolines as mdm2 inhibitors
WO2005003097A1 (en) 2003-06-17 2005-01-13 F. Hoffmann-La Roche Ag Cis-2,4,5-triaryl-imidazolines
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US7498298B2 (en) 2003-11-06 2009-03-03 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
US20050266000A1 (en) 2004-04-09 2005-12-01 Genentech, Inc. Variable domain library and uses
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
WO2005110996A1 (en) 2004-05-18 2005-11-24 F.Hoffmann-La Roche Ag Novel cis-imidazolines
WO2005123691A1 (en) 2004-06-17 2005-12-29 F-Hoffmann-La Roche Ag Novel cis-imidazolines
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006032631A1 (en) 2004-09-22 2006-03-30 Janssen Pharmaceutica N.V. Inhibitors of the interaction between mdm2 and p53
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2006091646A2 (en) 2005-02-22 2006-08-31 The Regents Of The University Of Michigan Small molecule inhibitors of mdm2 and uses thereof
WO2006097261A1 (en) 2005-03-16 2006-09-21 F.Hoffmann-La Roche Ag Cis-2,4,5-triaryl-imidazolines and their use as anti-cancer medicaments
US7989595B2 (en) 2005-06-20 2011-08-02 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8449883B2 (en) 2005-06-20 2013-05-28 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2007001851A2 (en) 2005-06-20 2007-01-04 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2006136606A2 (en) 2005-06-24 2006-12-28 F. Hoffmann-La Roche Ag Oxindole derivatives
US20070160598A1 (en) 2005-11-07 2007-07-12 Dennis Mark S Binding polypeptides with diversified and consensus vh/vl hypervariable sequences
WO2007063013A1 (en) 2005-12-01 2007-06-07 F. Hoffmann-La Roche Ag 2,4,5-triphenyl imidazoline derivatives as inhibitors of the interaction between p53 and mdm2 proteins for use as anticancer agents
US20070129416A1 (en) 2005-12-01 2007-06-07 Qingjie Ding 4,4,5,5, Tetrasubstituted imidazolines
US20070237764A1 (en) 2005-12-02 2007-10-11 Genentech, Inc. Binding polypeptides with restricted diversity sequences
WO2007082805A1 (en) 2006-01-18 2007-07-26 F. Hoffmann-La Roche Ag Cis-4, 5-biaryl-2-heterocyclic-imidazolines as mdm2 inhibitors
WO2007104714A1 (en) 2006-03-13 2007-09-20 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2007104664A1 (en) 2006-03-13 2007-09-20 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2007107543A1 (en) 2006-03-22 2007-09-27 Janssen Pharmaceutica N.V. Inhibitors of the interaction between mdm2 and p53
WO2007107545A1 (en) 2006-03-22 2007-09-27 Janssen Pharmaceutica N.V. Cyclic-alkylaminederivatives as inhibitors of the interaction between mdm2 and p53
US20070292936A1 (en) 2006-05-09 2007-12-20 Genentech, Inc. Binding polypeptides with optimized scaffolds
US8226945B2 (en) 2006-05-30 2012-07-24 Genentech, Inc. Antibodies and immunoconjugates and uses therefor
US20090182127A1 (en) 2006-06-22 2009-07-16 Novo Nordisk A/S Production of Bispecific Antibodies
WO2008036168A2 (en) 2006-08-30 2008-03-27 The Regents Of The University Of Michigan New small molecule inhibitors of mdm2 and the uses thereof
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008034736A2 (en) 2006-09-21 2008-03-27 F. Hoffmann-La Roche Ag Oxindole derivatives as anticancer agents
WO2008055812A1 (en) 2006-11-09 2008-05-15 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2008072655A1 (en) 2006-12-14 2008-06-19 Daiichi Sankyo Company, Limited Imidazothiazole derivatives
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2008106507A2 (en) 2007-02-27 2008-09-04 University Of South Florida Mdm2/mdmx inhibitor peptide
WO2008125487A1 (en) 2007-04-12 2008-10-23 F. Hoffmann-La Roche Ag Diphenyl-dihydro-imidazopyridinones
WO2008130614A2 (en) 2007-04-20 2008-10-30 University Of Pittsburg-Of The Commonwealth System Of Higher Education Selective and dual-action p53/mdm2/mdm4 antagonists
US7723485B2 (en) 2007-05-08 2010-05-25 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
WO2008141917A1 (en) 2007-05-17 2008-11-27 F. Hoffmann-La Roche Ag 3,3-spiroindolinone derivatives
WO2008141975A1 (en) 2007-05-23 2008-11-27 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
US20090002360A1 (en) 2007-05-25 2009-01-01 Innolux Display Corp. Liquid crystal display device and method for driving same
WO2009019274A1 (en) 2007-08-06 2009-02-12 Janssen Pharmaceutica Nv Substituted phenylenediamines as inhibitors of the interaction between mdm2 and p53
WO2009037343A1 (en) 2007-09-21 2009-03-26 Janssen Pharmaceutica Nv Inhibitors of the interaction between mdm2 and p53
WO2009037308A1 (en) 2007-09-21 2009-03-26 Janssen Pharmaceutica Nv Inhibitors of the interaction between mdm2 and p53
WO2009047161A1 (en) 2007-10-09 2009-04-16 F. Hoffmann-La Roche Ag Chiral cis-imidazolines
WO2009052249A1 (en) 2007-10-19 2009-04-23 Genentech, Inc. Cysteine engineered anti-tenb2 antibodies and antibody drug conjugates
WO2009077357A1 (en) 2007-12-14 2009-06-25 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2009080488A1 (en) 2007-12-19 2009-07-02 F. Hoffmann-La Roche Ag Spiroindolinone derivatives as anticancer agents
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
WO2009115425A1 (en) 2008-03-19 2009-09-24 F. Hoffmann-La Roche Ag Spiroindolinone derivatives
WO2009151069A1 (en) 2008-06-12 2009-12-17 第一三共株式会社 Imidazothiazole derivative having 4,7-diazaspiro[2.5]octane ring structure
WO2010009124A2 (en) 2008-07-15 2010-01-21 Genentech, Inc. Anthracycline derivative conjugates, process for their preparation and their use as antitumor compounds
WO2010028862A1 (en) 2008-09-15 2010-03-18 Priaxon Ag Pyrrolidin-2-ones as hdm2 ligands
WO2010058819A1 (en) 2008-11-19 2010-05-27 学校法人 慶應義塾 Peptide capable of inhibiting interaction between human tumor protein mdm2 and human tumor suppression protein p53, and use thereof
WO2010082612A1 (en) 2009-01-16 2010-07-22 第一三共株式会社 Imidazothiazole derivative having proline ring structure
WO2010084097A1 (en) 2009-01-26 2010-07-29 F. Hoffmann-La Roche Ag Spiroindolinone derivative prodrugs
WO2010091979A1 (en) 2009-02-10 2010-08-19 F. Hoffmann-La Roche Ag Spiroindolinone pyridine derivative
WO2010094622A1 (en) 2009-02-17 2010-08-26 F. Hoffmann-La Roche Ag 3, 3' -spiroindolinone derivatives as anticancer agents
WO2010121995A1 (en) 2009-04-23 2010-10-28 F. Hoffmann-La Roche Ag 3,3'-spiroindolinone derivatives and their use for cancer
WO2011005219A1 (en) 2009-07-07 2011-01-13 Agency For Science, Technology And Research Novel mdm2 binding peptides and uses thereof
WO2011023677A1 (en) 2009-08-26 2011-03-03 Novartis Ag Tetra-substituted heteroaryl compounds and their use as mdm2 and/or mdm4 modulators
WO2011045257A1 (en) 2009-10-14 2011-04-21 F. Hoffmann-La Roche Ag Novel n-substituted-pyrrolidines as inhibitors of mdm2-p-53 interactions
WO2011060049A2 (en) 2009-11-12 2011-05-19 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists
WO2011061139A1 (en) 2009-11-17 2011-05-26 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
WO2011067185A1 (en) 2009-12-02 2011-06-09 F. Hoffmann-La Roche Ag Spiroindolinone pyrrolidines
WO2011076786A1 (en) 2009-12-22 2011-06-30 Novartis Ag Substituted isoquinolinones and quinazolinones
US9096544B2 (en) 2009-12-23 2015-08-04 Wayne State University Therapeutic compounds
WO2011098398A1 (en) 2010-02-09 2011-08-18 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
US8697650B2 (en) 2010-02-16 2014-04-15 Medimmune, Llc HSA-related compositions and methods of use
WO2011101297A1 (en) 2010-02-17 2011-08-25 F. Hoffmann-La Roche Ag Substituted spiroindolinones
WO2011106650A2 (en) 2010-02-27 2011-09-01 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Novel p53-mdm2/p53-mdm4 antagonists to treat proliferative disease
US20110287009A1 (en) 2010-04-23 2011-11-24 Genentech, Inc. Production of Heteromultimeric Proteins
WO2011134925A1 (en) 2010-04-28 2011-11-03 F. Hoffmann-La Roche Ag Spiroindolinone pyrrolidines
WO2011153509A1 (en) 2010-06-04 2011-12-08 Amgen Inc. Piperidinone derivatives as mdm2 inhibitors for the treatment of cancer
WO2012007409A1 (en) 2010-07-12 2012-01-19 F. Hoffmann-La Roche Ag N-substituted pyrrolidines
WO2012022707A1 (en) 2010-08-18 2012-02-23 F. Hoffmann-La Roche Ag Substituted heteroaryl spiropyrrolidine mdm2 antagonists
WO2012033525A2 (en) 2010-09-08 2012-03-15 University Of Pittsburgh - Of The Commonwealth System Of Higher Education P53-mdm2 antagonists
WO2012034954A1 (en) 2010-09-15 2012-03-22 F. Hoffmann-La Roche Ag Substituted hexahydropyrrolo [1, 2 - c] imidazolones useful as mdm2 inhibitors
WO2012038307A1 (en) 2010-09-20 2012-03-29 F. Hoffmann-La Roche Ag Spiro substituted pyrrolo[1,2-c]imidazole derivatives useful as mdm2 inhibitors
WO2012047587A2 (en) 2010-09-27 2012-04-12 Serrata, Llc Mdm2 inhibitors for treatment of ocular conditions
WO2012065022A2 (en) 2010-11-12 2012-05-18 The Regents Of The University Of Michigan Spiro-oxindole mdm2 antagonists
WO2012066095A1 (en) 2010-11-19 2012-05-24 Novartis Ag Crystalline form of an inhibitor of mdm2/4 and p53 interaction
WO2012076513A1 (en) 2010-12-09 2012-06-14 F. Hoffmann-La Roche Ag 3-cyano-1-hydroxymethyl-2-phenylpyrrolidine derivatives as inhibitors of mdm2-p53 interactions useful for the treatment of cancer
WO2012106587A1 (en) 2011-02-04 2012-08-09 Genentech, Inc. Fc VARIANTS AND METHODS FOR THEIR PRODUCTION
WO2012116989A1 (en) 2011-03-03 2012-09-07 F. Hoffmann-La Roche Ag Substituted heteroaryl 2', 3', 7', 7a' - tetrahydrospiro-[pyrrole-3, 6' - pyrrolo [1, 2-c] imidazole] - 1', 2 (1h, 5'h) -diones as anticancer agents
WO2012121361A1 (en) 2011-03-10 2012-09-13 第一三共株式会社 Dispiropyrrolidine derivative
US9249153B2 (en) 2011-03-18 2016-02-02 Pusan National University Industry-University Cooperation Foundation Pharmaceutical composition for treating aging-associated diseases, containing progerin expression inhibitor as active ingredient, and screening method of said progerin expression inhibitor
US8969526B2 (en) 2011-03-29 2015-03-03 Roche Glycart Ag Antibody Fc variants
WO2012155066A2 (en) 2011-05-11 2012-11-15 Shaomeng Wang Spiro-oxindole mdm2 antagonists
WO2012175520A1 (en) 2011-06-20 2012-12-27 Novartis Ag Hydroxy substituted isoquinolinone derivatives
WO2012175487A1 (en) 2011-06-20 2012-12-27 Novartis Ag Cyclohexyl isoquinolinone compounds
WO2012176123A1 (en) 2011-06-22 2012-12-27 Novartis Ag 3 - imidazolyl- indoles for the treatment of proliferative diseases
WO2013017705A1 (en) 2011-08-03 2013-02-07 Salvador Moreno Rufino Baltasar Panel system for construction with backlighting based on light-emitting diodes
WO2013049250A1 (en) 2011-09-27 2013-04-04 Amgen Inc. Heterocyclic compounds as mdm2 inhibitors for the treatment of cancer
WO2013055987A1 (en) 2011-10-14 2013-04-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2013080141A1 (en) 2011-11-29 2013-06-06 Novartis Ag Pyrazolopyrrolidine compounds
WO2013096150A1 (en) 2011-12-21 2013-06-27 Merck Sharp & Dohme Corp. Substituted piperidines as hdm2 inhibitors
WO2013105037A1 (en) 2012-01-09 2013-07-18 Universidade Do Porto Inhibitors of p53-mdm2 interaction
WO2013106646A2 (en) 2012-01-12 2013-07-18 Yale University Compounds and methods for the inhibition of vcb e3 ubiquitin ligase
US20140356322A1 (en) 2012-01-12 2014-12-04 Yale University Compounds & Methods for the Enhanced Degradation of Targeted Proteins & Other Polypeptides by an E3 Ubiquitin Ligase
WO2013106643A2 (en) 2012-01-12 2013-07-18 Yale University Compounds & methods for the enhanced degradation of targeted proteins & other polypeptides by an e3 ubiquitin ligase
WO2013111105A1 (en) 2012-01-26 2013-08-01 Novartis Ag Imidazopyrrolidinone compounds
WO2013135648A1 (en) 2012-03-15 2013-09-19 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
WO2013177055A2 (en) 2012-05-21 2013-11-28 Genentech, Inc. ANTI-Ly6E ANTIBODIES AND IMMUNOCONJUGATES AND METHODS OF USE
WO2013175417A1 (en) 2012-05-24 2013-11-28 Novartis Ag Pyrrolopyrrolidinone compounds
WO2013178570A1 (en) 2012-05-30 2013-12-05 F. Hoffmann-La Roche Ag Substituted pyrrolidine-2-carboxamides
US8883771B2 (en) 2012-08-23 2014-11-11 The Regents Of The University Of Michigan Bivalent inhibitors of IAP proteins and therapeutic methods using the same
WO2014038606A1 (en) 2012-09-06 2014-03-13 第一三共株式会社 Crystal of dispiropyrrolidine derivative
WO2014044622A1 (en) * 2012-09-19 2014-03-27 F. Hoffmann-La Roche Ag 2-oxo-2,3,4,5-tetrahydro-1 h-benzo[b]diazepines and their use in the treatment of cancer
US20150225449A1 (en) 2012-09-19 2015-08-13 Hoffmann-La Roche Inc. 2-oxo-2,3,4,5-tetrahydro-1 h-benzo[b]diazepines and their use in the treatment of cancer
WO2014044401A2 (en) 2012-09-24 2014-03-27 Glanzstoff Bohemia S.R.O. Yarn having flame-retardant effect and textile fabric formed therefrom
US20150252072A1 (en) 2012-10-11 2015-09-10 Hoffmann-La Roche Inc. Azaindolines
US20150246882A1 (en) 2012-10-11 2015-09-03 Hoffmann-La Roche Inc. Indolines
US20150266879A1 (en) 2012-10-11 2015-09-24 Hoffmann-La Roche Inc. Azaindolines
US20150259359A1 (en) 2012-10-19 2015-09-17 Astex Therapeutics Limited Bicyclic heterocycle compounds and their uses in therapy
WO2014063061A1 (en) 2012-10-19 2014-04-24 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US20140135270A1 (en) 2012-11-09 2014-05-15 Ensemble Therapeutics Corp. Macrocyclic compounds for inhibition of inhibitors of apoptosis
US20150284427A1 (en) 2012-11-09 2015-10-08 Bristol-Myers Squibb Company Macrocyclic compounds for inhibition of inhibitors of apoptosis
WO2014082889A1 (en) 2012-11-28 2014-06-05 F. Hoffmann-La Roche Ag Novel imidazolines as dual inhibitors of mdm2 and mdmx
US20150307499A1 (en) 2012-11-30 2015-10-29 Sanford-Burnham Medical Research Institute Inhibitor of apoptosis protein (iap) antagonists
US20160024055A1 (en) 2012-12-11 2016-01-28 Hoffmann-La Roche Inc. Dimeric compounds
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
WO2014100071A2 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted pyrrolopyrimidines as hdm2 inhibitors
WO2014115080A1 (en) 2013-01-22 2014-07-31 Novartis Ag Pyrazolo[3,4-d]pyrimidinone compounds as inhibitors of the p53/mdm2 interaction
WO2014115077A1 (en) 2013-01-22 2014-07-31 Novartis Ag Substituted purinone compounds
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
WO2014123882A1 (en) 2013-02-07 2014-08-14 Merck Sharp & Dohme Corp. 2,6,7 substituted purines as hdm2 inhibitors
WO2014130470A1 (en) 2013-02-19 2014-08-28 Amgen Inc. Cis-morpholinone and other compounds as mdm2 inhibitors for the treatment of cancer
WO2014134207A2 (en) 2013-03-01 2014-09-04 Google Inc. Auxiliary content suggestions relating to user generated content
WO2014151863A1 (en) 2013-03-14 2014-09-25 Amgen Inc. Heteroaryl acid morpholinone compounds as mdm2 inhibitors for the treatment of cancer
WO2014165506A1 (en) * 2013-04-01 2014-10-09 Immunomedics, Inc. Anti-mucin antibodies for early detection and treament of pancreatic cancer
WO2014187777A1 (en) 2013-05-21 2014-11-27 Mediapharma S.R.L. Novel inhibitors of pvhl-elongin c binding
WO2014191896A1 (en) 2013-05-27 2014-12-04 Novartis Ag Pyrazolopyrrolidine derivatives and their use in the treatment of disease
WO2014200937A1 (en) 2013-06-10 2014-12-18 Amgen Inc. Processes of making and crystalline forms of a mdm2 inhibitor
WO2014198266A1 (en) 2013-06-12 2014-12-18 Schaeffler Technologies Gmbh & Co. Kg Bicycle drive and bicycle comprising a bicycle drive
WO2015004610A1 (en) 2013-07-11 2015-01-15 Adamed Sp. Z O.O. 1,5-dihydropyrrol-2-one derivatives as inhibitors of p53-mdm2/mdm4 protein-protein interaction
WO2015023355A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
US9249151B2 (en) 2013-08-23 2016-02-02 Boehringer Ingelheim International Gmbh Bis-amido pyridines
US9278978B2 (en) 2013-08-23 2016-03-08 Boehringer Ingelheim International Gmbh 6-Alkynyl Pyridine
WO2015070224A2 (en) 2013-11-11 2015-05-14 Amgen Inc. Combination therapy including an mdm2 inhibitor and one or more additional pharmaceutically active agents for the treatment of cancers
WO2015071393A1 (en) 2013-11-18 2015-05-21 F. Hoffmann-La Roche Ag Tetrahydro-benzodiazepinones
WO2015095124A1 (en) 2013-12-16 2015-06-25 Genentech Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095223A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015155332A1 (en) 2014-04-11 2015-10-15 Boehringer Ingelheim International Gmbh Spiro[3h-indole-3,2'-pyrrolidin]-2(1h)-one derivatives and their use as mdm2-p53 inhibitors
WO2015160845A2 (en) 2014-04-14 2015-10-22 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
US20150291562A1 (en) 2014-04-14 2015-10-15 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
US20160058872A1 (en) 2014-04-14 2016-03-03 Arvinas, Inc. Imide-based modulators of proteolysis and associated methods of use
WO2015158648A1 (en) 2014-04-15 2015-10-22 F. Hoffmann-La Roche Ag Solid forms of a pharmaceutically active compound
WO2015161032A1 (en) 2014-04-17 2015-10-22 The Regents Of The University Of Michigan Mdm2 inhibitors and therapeutic methods using the same
WO2015187998A2 (en) 2014-06-04 2015-12-10 Sanford-Burnham Medical Research Institute Use of inhibitor of apoptosis protein (iap) antagonists in hiv therapy
WO2015189799A1 (en) 2014-06-12 2015-12-17 Adamed Sp. Z O.O. Compounds comprising 1,1',2,5'-tetrahydrospiro[indole-3,2'-pyrrole]-2,5'-dione system as inhibitors p53-mdm2 protein-protein interaction
US20150374678A1 (en) 2014-06-27 2015-12-31 Celgene Corporation Compositions and methods for inducing conformational changes in cereblon and other e3 ubiquitin ligases
WO2016001376A1 (en) 2014-07-03 2016-01-07 Boehringer Ingelheim International Gmbh New spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors
US20160045607A1 (en) 2014-08-11 2016-02-18 Yale University Estrogen-related receptor alpha based protac compounds and associated methods of use
WO2016028391A2 (en) 2014-08-18 2016-02-25 Hudson Biopharma Inc. Spiropyrrolidines as mdm2 inhibitors
WO2016026937A1 (en) 2014-08-21 2016-02-25 Boehringer Ingelheim International Gmbh New spiro[3h-indole-3,2´-pyrrolidin]-2(1h)-one compounds and derivatives as mdm2-p53 inhibitors

Non-Patent Citations (302)

* Cited by examiner, † Cited by third party
Title
"Liebigs Annalen der Chemie", 1985, pages: 1910 - 16
"Small-molecule Pan-IAP Antagonists: A Patent Review", EXPERT OPIN THER PAT, vol. 20, 2010, pages 251 - 67
"The Chemistry of Heterocyclic Compounds, A series of Monographs", 1950, JOHN WILEY & SONS, NEW YORK, article "Volumes 13, 14, 16, 19, and 28;"
ALMAGRO; FRANSSON, FRONT. BIOSCI., vol. 13, 2008, pages 1619 - 1633
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
ANDREWS ET AL., BLOOD, vol. 68, 1986, pages 1030 - 5
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
ARZNEIMITTEL-FORSCHUNG, vol. 40, no. 12, 1990, pages 1328 - 31
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, NEW YORK, N.Y.
BACA ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 10678 - 10684
BAKKER AB ET AL., CANCER RES., vol. 64, no. 22, 2005, pages 8443 - 50
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
BECK ET AL., J. MOL. BIOL., vol. 228, 1992, pages 433 - 441
BECK ET AL., J. MOL. BIOL., vol. 255, 1996, pages 1 - 13
BEILSTEIN: "Beilsteins Handbuch der organischen Chemie, 4. Aufl.,", SPRINGER-VERLAG
BIOCHEM. BIOPHYS. RES. COMMUN., vol. 255, no. 2, 1999, pages 283 - 288
BIOCHEM. BIOPHYS. RES. COMMUN., vol. 275, no. 3, 2000, pages 783 - 788
BIOORG. MED. CHEM. LETT., vol. 16, 2006, pages 3310 - 3314
BIOORG. MED. CHEM. LETT., vol. 25, 2015, pages 3621 - 3625
BISHOP, D.T. ET AL., NAT. GENET., vol. 41, no. 8, 2009, pages 920 - 925
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
BOERNER ET AL., J. IMMUNOL., vol. 147, 1991, pages 86
BOURGEOIS C. ET AL., J. CLIN. ENDOCRINOL. METAB., vol. 82, 1997, pages 3116 - 3123
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81
BRODEUR ET AL.: "Monoclonal Antibody Production Techniques and Applications", 1987, MARCEL DEKKER, INC., NEW YORK, pages: 51 - 63
BRUGGEMANN, M. ET AL., J. EXP. MED., vol. 166, 1987, pages 1351 - 1361
CANCER RES., vol. 61, no. 15, 2001, pages 5857 - 5860
CARTER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 4285
CELL, vol. 109, no. 3, 2002, pages 397 - 407
CHAN,J.; WATT, V.M., ONCOGENE, vol. 6, no. 6, 1991, pages 1057 - 1061
CHARI ET AL., CANCER RESEARCH, vol. 52, 1992, pages 127 - 131
CHARLTON: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, TOTOWA, NJ, pages: 245 - 254
CHEN CH ET AL., BLOOD, vol. 107, no. 4, 2006, pages 1459 - 67
CHEN ET AL., CANCER RES., vol. 67, no. 10, 2007, pages 4924 - 4932
CHEN ET AL., J. MOL. BIOL., vol. 293, 1999, pages 865 - 881
CHO H.-S. ET AL., NATURE, vol. 421, 2003, pages 756 - 760
CHOTHIA; LESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHOWDHURY, METHODS MOL. BIOL., vol. 207, 2008, pages 179 - 196
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
CLARKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLYNES ET AL., PROC. NAT'LACAD. SCI. USA, vol. 95, 1998, pages 652 - 656
COUSSENS L. ET AL., SCIENCE, vol. 230, no. 4730, 1985, pages 1132 - 1139
CRAGG, M.S. ET AL., BLOOD, vol. 101, 2003, pages 1045 - 1052
CRAGG, M.S.; M.J. GLENNIE, BLOOD, vol. 103, 2004, pages 2738 - 2743
CUNNINGHAM; WELLS, SCIENCE, vol. 244, 1989, pages 1081 - 1085
DALL'ACQUA ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 281, no. 33, 2006, pages 23514 - 23524
DALL'ACQUA ET AL., METHODS, vol. 36, 2005, pages 43 - 60
DAVIS ET AL., PROC. NATL. ACAD. SCI USA, vol. 98, no. 17, 2001, pages 9772 - 9777
DE NOOIJ-VAN DALEN, A.G. ET AL., INT. J. CANCER, vol. 103, no. 6, 2003, pages 768 - 774
DOBNER ET AL., EUR. J. IMMUNOL., vol. 22, 1992, pages 2795 - 2799
DORNAN ET AL., BLOOD, vol. 114, no. 13, 2009, pages 2721 - 2729
DRICKAMER K, CURR. OPIN. STRUCT. BIOL., vol. 9, no. 5, 1999, pages 585 - 90
DUCRY; STUMP, BIOCONJUGATE CHEM, vol. 21, 2010, pages 5 - 13
DUMOUTIER L. ET AL., J. IMMUNOL., vol. 167, 2001, pages 3545 - 3549
DUNCAN; WINTER, NATURE, vol. 322, 1988, pages 738 - 40
DURIE E, SCIENCE, vol. 261, 1993, pages 1328 - 30
ED HARLOW AND DAVID LANE: "Antibodies, A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
EDELMAN ET AL., PROC. NATL. ACAD. OF SCI., vol. 63, no. 1, 1969, pages 78 - 85
EHSANI A. ET AL., GENOMICS, vol. 15, 1993, pages 426 - 429
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC., NEW YORK
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
FEILD, J.A. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 258, no. 3, 1999, pages 578 - 582
FELLOUSE, PROC. NATL. ACAD. SCI. USA, vol. 101, no. 34, 2004, pages 12467 - 12472
FINCK ET AL., SCIENCE, vol. 265, 1994, pages 1225 - 7
FLATMAN ET AL., J. CHROMATOGR. B, vol. 848, 2007, pages 79 - 87
FRANKLIN ET AL., CANCER CELL, vol. 5, 2004, pages 317 - 328
FUCHS S. ET AL., MOL. MED., vol. 7, 2001, pages 115 - 124
FUJISAKU ET AL., J. BIOL. CHEM., vol. 264, no. 4, 1989, pages 2118 - 2125
FURUSHIMA, K. ET AL., DEV. BIOL., vol. 306, no. 2, 2007, pages 480 - 492
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
GENOME RES., vol. 13, no. 10, 2003, pages 2265 - 2270
GENOMICS, vol. 62, no. 2, 1999, pages 281 - 284
GERHARD, D.S. ET AL., GENOME RES., vol. 14, no. 10B, 2004, pages 2121 - 2127
GERNGROSS, NAT. BIOTECH., vol. 22, 2004, pages 1409 - 1414
GERY, S. ET AL., ONCOGENE, vol. 22, no. 18, 2003, pages 2723 - 2727
GETZ ET AL., ANAL. BIOCHEM., vol. 273, 1999, pages 73 - 80
GLYNNE-JONES ET AL., INT J CANCER., vol. 94, no. 2, 15 October 2001 (2001-10-15), pages 178 - 84
GOMEZ ET AL., BIOTECHNOL. PROG., vol. 26, 2010, pages 1438 - 1445
GOMEZ ET AL., BIOTECHNOLOGY AND BIOENG, vol. 105, no. 4, 2010, pages 748 - 760
GRAHAM ET AL., J. GEN VIROL., vol. 36, 1977, pages 59
GRIFFITHS ET AL., EMBO J, vol. 12, 1993, pages 725 - 734
GRUBER ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368
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 ET AL., CLIN. CANCER RES., vol. 10, 2004, pages 7063 - 7070
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
HELLSTROM, I ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 82, 1985, pages 1499 - 1502
HELLSTROM, I. ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 83, 1986, pages 7059 - 7063
HELVETICA CHIMICA ACTA, vol. 41, 1958, pages 1052 - 60
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
HOLLINGER ET AL., PROC. NATL. ACAD SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOOGENBOOM ET AL.: "Methods in Molecular Biology", vol. 178, 2001, HUMAN PRESS, TOTOWA, NJ, pages: 1 - 37
HOOGENBOOM; WINTER, J. MOL. BIOL., vol. 227, 1992, pages 381 - 388
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
HUDSON ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
IANEWAY, NATURE, vol. 341, 1989, pages 482
IDUSOGIE ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
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
J. AM. CHEM. SOC., vol. 82, 1960, pages 5566
J. BIOL. CHEM., vol. 270, no. 37, 1995, pages 21984 - 21990
J. BIOL. CHEM., vol. 276, no. 29, 2001, pages 27371 - 27375
J. BIOL. CHEM., vol. 277, no. 22, 2002, pages 19665 - 19672
J. BIOL. CHEM., vol. 278, no. 33, 2003, pages 30813 - 30820
J. MED. CHEM., vol. 58, 2015, pages 1038 - 1052
J. MED. CHEM., vol. 58, no. 16, 2015, pages 6574 - 6588
JANEWAY, C.; TRAVERS, P.; WALPORT, M.; SHLOMCHIK: "Immuno Biology, 5th ed.,", 2001, GARLAND PUBLISHING, NEW YORK
JANEWAY, C.; TRAVERS, P.; WALPORT, M.; SHLOMCHIK: "Immunobiology, 5th ed.,", 2001, GARLAND PUBLISHING, NEW YORK
JONSSON ET AL., IMMUNOGENETICS, vol. 29, no. 6, 1989, pages 411 - 413
JUNUTULA ET AL., JOUR OF IMMUN. METHODS, vol. 332, 2008, pages 41 - 52
JUNUTULA ET AL., NATURE BIOTECH., vol. 26, no. 8, 2008, pages 925 - 932
KABAT ET AL.: "Sequences of Proteins of Immunological Interest, 5th ed.,", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH, BETHESDA, MD
KABAT ET AL.: "Sequences of Proteins of Immunological Interest, Fifth edition", vol. 1-3, 1991, NIH PUBLICATION 91-3242, BETHESDA MD
KABAT ET AL.: "Sequences ofproteins of immunological interest 5th Ed.,", 1991, NATIONAL INSTITUTES OF HEALTH, BETHESDA, MD
KAM ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 11600 - 11605
KANDA, Y. ET AL., BIOTECHNOL. BIOENG., vol. 94, no. 4, 2006, pages 680 - 688
KASAHARA ET AL., IMMUNOGENETICS, vol. 30, no. 1, 1989, pages 66 - 68
KASHMIRI ET AL., METHODS, vol. 36, 2005, pages 25 - 34
KATRITZKY AND REES,: "Comprehensive Heterocyclic Chemistry II", vol. 3, 1997, ELSEVIER
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
KLIMKA ET AL., BR. J. CANCER, vol. 83, 2000, pages 252 - 260
KLUSSMAN ET AL., BIOCONJUGATE CHEMISTRY, vol. 15, no. 4, 2004, pages 765 - 773
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
KONTERMANN, ACTA PHARMACOL. SIN., vol. 26, 2005, pages 1 - 9
KOSTELNY ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1547 - 1553
KOZBOR, J. IMMUNOL., vol. 133, 1984, pages 3001
KUHNS J.J. ET AL., J. BIOL. CHEM., vol. 274, 1999, pages 36422 - 36427
KUMMER, M.P. ET AL., J. BIOL. CHEM., vol. 284, no. 4, 2009, pages 2296 - 2306
L. PAQUETTE,: "Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
LAB. INVEST., vol. 82, no. 11, 2002, pages 1573 - 1582
LARHAMMAR ET AL., J. BIOL. CHEM., vol. 260, no. 26, 1985, pages 14111 - 14119
LE ET AL., FEBS LETT., vol. 418, no. 1-2, 1997, pages 195 - 199
LEE ET AL., J. IMMUNOL. METHODS, vol. 284, no. 1-2, 2004, pages 119 - 132
LEE ET AL., J. MOL. BIOL., vol. 340, no. 5, 2004, pages 1073 - 1093
LI ET AL., MOL. CANCER. THER., vol. 12, no. 7, 2013, pages 1255 - 1265
LI ET AL., NAT. BIOTECH., vol. 24, 2006, pages 210 - 215
LI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 3557 - 3562
LIANG ET AL., CANCER RES., vol. 60, 2000, pages 4907 - 12
LONBERG, CURR. OPIN. IMMUNOL., vol. 20, 2008, pages 450 - 459
LONBERG, NAT. BIOTECH., vol. 23, 2005, pages 1117 - 1125
LOUIS F. FIESER; MARY FIESER: "Reagents for Organic Synthesis", vol. 1-23, 1967, WILEY, N.Y.
LUND ET AL., EUR. J. BIOCHEM., vol. 267, 2000, pages 7246 - 7256
MACKAY; MACKAY, TRENDS IMMUNOL., vol. 23, 2002, pages 113 - 5
MALLYA, M. ET AL., GENOMICS, vol. 80, no. 1, 2002, pages 113 - 123
MAO ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 93, 1996, pages 5910 - 5914
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
MARKS ET AL., J. MOL. BIOL., vol. 222, 1992, pages 581 - 597
MARKS; BRADBURY: "Methods in Molecular Biology", vol. 248, 2003, HUMAN PRESS, TOTOWA, NJ, pages: 161 - 175
MARSHALL AS ET AL., EUR. J. IMMUNOL., vol. 36, no. 8, 2006, pages 2159 - 69
MARSHALL AS ET AL., J. BIOL. CHEM., vol. 279, no. 15, 2004, pages 14792 - 802
MARVIN; ZHU, ACTA PHARMACOL. SIN., vol. 26, no. 6, 2005, pages 649 - 658
MATHER ET AL.: "Annals N.Y. Acad. Sci.", vol. 383, 1982, pages: 44 - 68
MATHER, BIOL. REPROD., vol. 23, 1980, pages 243 - 251
MCCAFFERTY ET AL., NATURE, vol. 348, pages 552 - 554
MCGLINCHEY, R.P. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 106, no. 33, 2009, pages 13731 - 13736
MED. CHEM. LETT, vol. 4, 2013, pages 466 - 469
MILLER ET AL., JOUR. OF IMMUNOLOGY, vol. 170, 2003, pages 4854 - 4861
MILSTEIN; CUELLO, NATURE, vol. 305, 1983, pages 537
MIURA ET AL., BLOOD, vol. 92, 1998, pages 2815 - 2822
MIURA ET AL., GENOMICS, vol. 38, no. 3, 1996, pages 299 - 304
MOHAN ET AL., J. IMMUNOL., vol. 154, 1995, pages 1470 - 80
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
NAGASE T. ET AL., DNA RES., vol. 7, no. 2, 2000, pages 143 - 150
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
NARUSE ET AL., 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
NI, XIANDAI MIANYIXUE, vol. 26, no. 4, 2006, pages 265 - 268
NICOLAOU ET AL., ANGEW. CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
O'BRIEN ET AL.,: "Methods in Molecular Biology", vol. 178, 2001, HUMAN PRESS, TOTOWA, NJ, pages: 1 - 37
O'DOWD, B.F. ET AL., FEBS LETT., vol. 394, no. 3, 1996, pages 325 - 329
OFFNER ET AL., SCIENCE, vol. 251, 1991, pages 430 - 432
OGAWA Y. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 178, 1991, pages 248 - 255
OKAMOTO Y. ET AL., BIOL. CHEM., vol. 272, 1997, pages 21589 - 21596
OKAZAKI ET AL., J. MOL. BIOL., vol. 336, 2004, pages 1239 - 1249
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
OSBOURN ET AL., METHODS, vol. 36, 2005, pages 61 - 68
OSOL, A.: "Remington's Pharmaceutical Sciences 16th edition,", 1980
PADLAN, MOL. IMMUNOL., vol. 28, 1991, pages 489 - 498
PAQUETTE, LEO A.: "Principles of Modern Heterocyclic Chemistry", 1968, W.A. BENJAMIN, NEW YORK, article "Chapters 1, 3, 4, 6, 7, and 9;"
PARRISH-NOVAK J. ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 47517 - 47523
PETKOVA, S.B. ET AL., INT'L. IMMUNOL., vol. 18, no. 12, 2006, pages 1759 - 1769
PINGAULT V. ET AL., HUM. GENET., vol. 111, 2002, pages 198 - 206
PIYA SUJAN ET AL: "BRD4 Proteolysis Targeting Chimera (PROTAC) Leads to Sustained Degradation of BRD4 with Broad Activity Against Acute Leukemias and Overcomes Stroma Mediated Resistance By Modulating Surface Expression of CXCR4", vol. 126, no. 23, 1 December 2015 (2015-12-01), pages 1 (675) - 6, XP008185697, ISSN: 0006-4971, Retrieved from the Internet <URL:http://www.bloodjournal.org/content/126/23/675> *
PLETNEV S. ET AL., BIOCHEMISTRY, vol. 42, 2003, pages 12617 - 12624
PLUCKTHUN: "The Pharmacology of Monoclonal Antibodies", vol. 113, 1994, SPRINGER-VERLAG, NEW YORK, pages: 269 - 315
POISON ET AL., EXPERT OPIN. INVESTIG. DRUGS, vol. 20, no. 1, 2011, pages 75 - 85
POISON ET AL., LEUKEMIA, vol. 24, 2010, pages 1566 - 1573
PORTOLANO ET AL., J. IMMUNOL., vol. 150, 1993, pages 880 - 887
PRESTA ET AL., CANCER RES., vol. 57, 1997, pages 4593 - 4599
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623
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
QUEEN ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 86, 1989, pages 10029 - 10033
R. LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS
RAVETCH; KINET, ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
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., NATURE, vol. 332, 1988, pages 323 - 329
RIPKA ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
ROSOK ET AL., J. BIOL. CHEM., vol. 271, 1996, pages 22611 - 22618
ROSS ET AL., CANCER RES., vol. 62, 2002, pages 2546 - 2553
ROUX ET AL., J. IMMUNOL., vol. 161, 1998, pages 4083 - 4090
S. P. PARKER,: "McGraw-Hill Dictionary of Chemical Terms", 1984, MCGRAW-HILL BOOK COMPANY, NEW YORK
SABBATH ET AL., J. CLIN. INVEST., vol. 75, 1985, pages 756 - 56
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. ET AL., AM. J. EPIDEMIOL., vol. 170, no. 5, 2009, pages 537 - 545
SAMBROOK ET AL.: "Molecular Cloning, A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SANDERSON ET AL., CLIN. CANCER RES., vol. 11, 2005, pages 843 - 852
SCHERER, S.E. ET AL., NATURE, vol. 440, no. 7082, 2006, pages 346 - 351
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
SHEIKH F. ET AL., J. IMMUNOL., vol. 172, 2004, pages 2006 - 2010
SHEN ET AL., NATURE BIOTECH., vol. 30, no. 2, 2012, pages 184 - 191
SHIELDS ET AL., J. BIOL. CHEM., vol. 9, no. 2, 2001, pages 6591 - 6604
SIDHU ET AL., J. MOL. BIOL., vol. 338, no. 2, 2004, pages 299 - 310
SIMS ET AL., J. IMMUNOL., vol. 151, 1993, pages 2296
SINGLETON ET AL.: "Dictionary of Microbiology and Molecular Biology, 2nd ed.,", 1994, J. WILEY & SONS, NEW YORK, NY
SINHA S.K. ET AL., J. IMMUNOL., vol. 150, 1993, pages 5311 - 5320
SONDERMANN ET AL., NATURE, vol. 406, 20 July 2000 (2000-07-20), pages 267 - 273
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
SVENSSON P.J. ET AL., HUM. GENET., vol. 103, 1998, pages 145 - 148
SWIERCZ J.M. ET AL., J. CELL BIOL., vol. 165, 2004, pages 869 - 880
T. W. GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS, NEW YORK
T. W. GREENE; P. G .M. WUTS: "Protective Groups in Organic Synthesis 3rd Ed.,", 1999, JOHN WILEY AND SONS
TAKEDA, S. ET AL., FEBS LETT., vol. 520, no. 1-3, 2002, pages 97 - 101
TAWARAGI Y. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 150, 1988, pages 89 - 96
TEN DIJKE,P. ET AL., SCIENCE, vol. 264, no. 5155, 1994, pages 101 - 104
THOMPSON, J.S. ET AL., SCIENCE, vol. 293, no. 5537, 2001, pages 2108 - 2111
TONNELLE ET AL., EMBO J., vol. 4, no. 11, 1985, pages 2839 - 2847
TOUCHMAN ET AL., GENOME RES., vol. 10, 2000, pages 165 - 173
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655
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
TUTT ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
UCHIDA ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 266, 1999, pages 593 - 602
URLAUB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216
VAN DIJK; VAN DE WINKEL, CURR. OPIN. PHARMACOL., vol. 5, 2001, pages 368 - 74
VAN RHENEN A ET AL., BLOOD, vol. 110, no. 7, 2007, pages 2659 - 66
VERHEIJ J.B. ET AL., AM. J. MED. GENET., vol. 108, 2002, pages 223 - 225
VOLLMERS; BRANDLEIN, HISTOLOGY ANDHISTOPATHOLOGY, vol. 20, no. 3, 2005, pages 927 - 937
VOLLMERS; BRANDLEIN, METHODS AND FINDINGS IN EXPERIMENTAL AND CLINICAL PHARMACOLOGY, vol. 27, no. 3, 2005, pages 185 - 91
VON HOEGEN ET AL., J. IMMUNOL., vol. 144, no. 12, 1990, pages 4870 - 4877
WEIS J.J. ET AL., 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
WILSON ET AL., J. EXP. MED., vol. 173, 1991, pages 137 - 146
WINTER ET AL., ANN. REV. IMMUNOL., vol. 12, 1994, pages 433 - 455
WINTER ET AL., SCIENCE, 19 June 2015 (2015-06-19), pages 1376
WRIGHT ET AL., TIBTECH, vol. 15, 1997, pages 26 - 32
WU ET AL., NATURE BIOTECHNOLOGY, 2007
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 T. ET AL., NATURE, vol. 319, 1986, pages 230 - 234
YAMAMOTO, Y. ET AL., HEPATOLOGY, vol. 37, no. 3, 2003, pages 528 - 533
YAMANE-OHNUKI ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614
YAZAKI; WU: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, TOTOWA, NJ, pages: 255 - 268
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
ZHU ET AL., PROTEIN SCIENCE, vol. 6, 1997, pages 781 - 788

Cited By (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11161842B2 (en) 2016-04-22 2021-11-02 Dana-Farber Cancer Institute, Inc. Bifunctional molecules for degradation of EGFR and methods of use
US10450310B2 (en) 2016-04-22 2019-10-22 Dana-Farber Cancer Institute, Inc. Bifunctional molecules for degradation of EGFR and methods of use
US11185592B2 (en) 2016-05-10 2021-11-30 C4 Therapeutics, Inc. Spirocyclic degronimers for target protein degradation
US10646575B2 (en) 2016-05-10 2020-05-12 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
US10849982B2 (en) 2016-05-10 2020-12-01 C4 Therapeutics, Inc. C3-carbon linked glutarimide degronimers for target protein degradation
US10905768B1 (en) 2016-05-10 2021-02-02 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
US10660968B2 (en) 2016-05-10 2020-05-26 C4 Therapeutics, Inc. Spirocyclic degronimers for target protein degradation
US11666581B2 (en) 2016-05-20 2023-06-06 Genentech, Inc. PROTAC antibody conjugates and methods of use
US11701357B2 (en) 2016-08-19 2023-07-18 Beigene Switzerland Gmbh Treatment of B cell cancers using a combination comprising Btk inhibitors
WO2018227023A1 (en) * 2017-06-07 2018-12-13 Silverback Therapeutics, Inc. Antibody construct conjugates
US11459335B2 (en) 2017-06-20 2022-10-04 C4 Therapeutics, Inc. N/O-linked Degrons and Degronimers for protein degradation
US11597768B2 (en) 2017-06-26 2023-03-07 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
US11932846B2 (en) 2017-08-08 2024-03-19 Technology Innovation Momentum Fund (Israel) Limited Partnership Chloramphenicol resistant split protein and uses thereof
US11401256B2 (en) 2017-09-04 2022-08-02 C4 Therapeutics, Inc. Dihydroquinolinones for medical treatment
US11802131B2 (en) 2017-09-04 2023-10-31 C4 Therapeutics, Inc. Glutarimides for medical treatment
US11254672B2 (en) 2017-09-04 2022-02-22 C4 Therapeutics, Inc. Dihydrobenzimidazolones for medical treatment
US11787802B2 (en) 2017-09-04 2023-10-17 C4 Therapeutics, Inc. Dihydrobenzimidazolones for medical treatment
US11358948B2 (en) 2017-09-22 2022-06-14 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11623932B2 (en) 2017-09-22 2023-04-11 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11524949B2 (en) 2017-11-16 2022-12-13 C4 Therapeutics, Inc. Degraders and Degrons for targeted protein degradation
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
CN108047312B (en) * 2017-12-05 2021-03-23 北京大学深圳研究生院 Stable polypeptide protein targeted chimera molecule and preparation method and application thereof
CN108047312A (en) * 2017-12-05 2018-05-18 北京大学深圳研究生院 A kind of stable polypeptide targeting proteins chimer molecules and its preparation method and application
JP2021506814A (en) * 2017-12-15 2021-02-22 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Degradation of target proteins mediated by stabilizing peptides
JP2021508703A (en) * 2017-12-26 2021-03-11 カイメラ セラピューティクス, インコーポレイテッド IRAK Degradants and Their Use
US11318205B1 (en) 2017-12-26 2022-05-03 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US10874743B2 (en) 2017-12-26 2020-12-29 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11723980B2 (en) 2017-12-26 2023-08-15 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
WO2019133531A1 (en) * 2017-12-26 2019-07-04 Kymera Therapeutics, Inc. Irak degraders and uses thereof
WO2019140003A1 (en) * 2018-01-10 2019-07-18 Development Center For Biotechnology Antibody protac conjugates
EP3737422A4 (en) * 2018-01-10 2021-10-06 Development Center for Biotechnology Antibody protac conjugates
JP2021510375A (en) * 2018-01-10 2021-04-22 ディベロップメント センター フォー バイオテクノロジーDevelopment Center For Biotechnology Antibody PROTAC conjugate
CN112135637A (en) * 2018-01-10 2020-12-25 财团法人生物技术开发中心 Antibody PROTAC conjugates
US11485743B2 (en) 2018-01-12 2022-11-01 Kymera Therapeutics, Inc. Protein degraders and uses thereof
US11512080B2 (en) 2018-01-12 2022-11-29 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11932635B2 (en) 2018-01-12 2024-03-19 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
US11753397B2 (en) 2018-03-26 2023-09-12 C4 Therapeutics, Inc. Cereblon binders for the degradation of ikaros
US11584748B2 (en) 2018-04-16 2023-02-21 C4 Therapeutics, Inc. Spirocyclic compounds
JP2021520372A (en) * 2018-04-26 2021-08-19 オーリジーン ディスカバリー テクノロジーズ リミテッドAurigene Discovery Technologies Limited Pyridazine derivative as SMARCA 2/4 degrading agent
JP7387630B2 (en) 2018-04-26 2023-11-28 オーリジーン オンコロジー リミテッド Pyridazine derivatives as SMARCA2/4 decomposers
WO2019207538A1 (en) 2018-04-26 2019-10-31 Aurigene Discovery Technologies Limited Pyridazine derivatives as smarca2/4 degraders
CN112771038A (en) * 2018-04-26 2021-05-07 奥里吉恩发现科技有限公司 Pyridazine derivatives as SMARCA2/4 degradants
CN110407770B (en) * 2018-04-27 2022-12-30 复旦大学 3-substituted-1, 5-benzazepine compound and pharmaceutical use thereof
CN110407770A (en) * 2018-04-27 2019-11-05 复旦大学 3- substitution -1,5- benzo-aza * class compound and its medicinal usage
CN108690137A (en) * 2018-05-31 2018-10-23 山东省医学科学院基础医学研究所 M8Sac71 antibody and the preparation method and application thereof
US11623929B2 (en) 2018-06-04 2023-04-11 C4 Therapeutics, Inc. Spirocyclic compounds
US11292792B2 (en) 2018-07-06 2022-04-05 Kymera Therapeutics, Inc. Tricyclic CRBN ligands and uses thereof
US11897882B2 (en) 2018-07-06 2024-02-13 Kymera Therapeutics, Inc. Tricyclic crbn ligands and uses thereof
WO2020027225A1 (en) 2018-07-31 2020-02-06 ファイメクス株式会社 Heterocyclic compound
US11639354B2 (en) 2018-07-31 2023-05-02 Fimecs, Inc. Heterocyclic compound
JP2022513360A (en) * 2018-10-09 2022-02-07 ユーブラス バイオセラピューティクス インコーポレーテッド Target Protease Degradation Platform (TED)
WO2020073930A1 (en) * 2018-10-09 2020-04-16 嘉兴优博生物技术有限公司 Targeted protease degradation platform
CN111018857B (en) * 2018-10-09 2023-06-02 嘉兴优博生物技术有限公司 Targeted protease degradation platform (TED)
CN111018857A (en) * 2018-10-09 2020-04-17 嘉兴优博生物技术有限公司 Targeted protease degradation platform (TED)
EP3865152A4 (en) * 2018-10-09 2022-11-16 Eubulus Biotherapeutics Inc. Targeted protease degradation platform
CN112955182A (en) * 2018-10-09 2021-06-11 嘉兴优博生物技术有限公司 Targeted protease degradation platform (TED)
EP3867395A4 (en) * 2018-10-16 2022-07-20 Technology Innovation Momentum Fund (Israel) Limited Partnership Systems and method for screening small molecules of interest
JP2022505450A (en) * 2018-10-24 2022-01-14 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Conjugated chemical decomposition inducers and usage
WO2020086858A1 (en) * 2018-10-24 2020-04-30 Genentech, Inc. Conjugated chemical inducers of degradation and methods of use
CN113056287A (en) * 2018-10-24 2021-06-29 豪夫迈·罗氏有限公司 Conjugated chemical degradation inducers and methods of use
US11807636B2 (en) 2018-11-30 2023-11-07 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11117889B1 (en) 2018-11-30 2021-09-14 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11352350B2 (en) 2018-11-30 2022-06-07 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
WO2020191377A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Extracellular vesicle conjugates and uses thereof
WO2020191369A1 (en) 2019-03-21 2020-09-24 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
US11746120B2 (en) 2019-04-05 2023-09-05 Kymera Therapeutics, Inc. Stat degraders and uses thereof
US11485750B1 (en) 2019-04-05 2022-11-01 Kymera Therapeutics, Inc. STAT degraders and uses thereof
WO2020207396A1 (en) * 2019-04-09 2020-10-15 Ranok Therapeutics (Hangzhou) Co. Ltd. Methods and compositions for targeted protein degradation
WO2021016521A1 (en) * 2019-07-25 2021-01-28 Beth Israel Deaconess Medical Center, Inc. Photo induced control of protein destruction
KR20220042136A (en) 2019-07-31 2022-04-04 피맥스 가부시키가이샤 heterocyclic compound
WO2021020585A1 (en) 2019-07-31 2021-02-04 ファイメクス株式会社 Heterocyclic compound
US11865114B2 (en) 2019-09-12 2024-01-09 Aurigene Oncology Limited Method for treating diseases using SMARCA2/4 degraders
US11779578B2 (en) 2019-12-17 2023-10-10 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11591332B2 (en) 2019-12-17 2023-02-28 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11707457B2 (en) 2019-12-17 2023-07-25 Kymera Therapeutics, Inc. IRAK degraders and uses thereof
US11679109B2 (en) 2019-12-23 2023-06-20 Kymera Therapeutics, Inc. SMARCA degraders and uses thereof
WO2021180787A1 (en) 2020-03-12 2021-09-16 Julius-Maximilians-Universitaet Wuerzburg Proteolysis targeting chimera (protac) for degradation of aurora a-kinase
EP3878472A1 (en) 2020-03-12 2021-09-15 Julius-Maximilians-Universitaet Wuerzburg Proteolysis targeting chimera (protac) for degradation of aurora a-kinase
US11932624B2 (en) 2020-03-19 2024-03-19 Kymera Therapeutics, Inc. MDM2 degraders and uses thereof
WO2021237100A1 (en) 2020-05-21 2021-11-25 Codiak Biosciences, Inc. Methods of targeting extracellular vesicles to lung
US11685750B2 (en) 2020-06-03 2023-06-27 Kymera Therapeutics, Inc. Crystalline forms of IRAK degraders
WO2022020288A1 (en) * 2020-07-21 2022-01-27 Genentech, Inc. Antibody-conjugated chemical inducers of degradation of brm and methods thereof
CN112029866A (en) * 2020-09-21 2020-12-04 河南尚泰科诺生物科技有限公司 Application of WWP1 in pancreatic cancer
CN112029866B (en) * 2020-09-21 2024-02-02 河南尚泰科诺生物科技有限公司 Application of WWP1 in pancreatic cancer
WO2022066928A2 (en) 2020-09-23 2022-03-31 Codiak Biosciences, Inc. Process for preparing extracellular vesicles
WO2022148459A1 (en) * 2021-01-11 2022-07-14 和径医药科技(上海)有限公司 Class of novel smad3 protein degraders and application thereof
WO2023275394A1 (en) 2021-07-02 2023-01-05 Merck Patent Gmbh Anti-protac antibodies and complexes
WO2023056069A1 (en) * 2021-09-30 2023-04-06 Angiex, Inc. Degrader-antibody conjugates and methods of using same
WO2023104941A1 (en) 2021-12-08 2023-06-15 European Molecular Biology Laboratory Hydrophilic tetrazine-functionalized payloads for preparation of targeting conjugates
WO2023147328A1 (en) 2022-01-26 2023-08-03 Genentech, Inc. Antibody-conjugated chemical inducers of degradation with hydolysable maleimide linkers and methods thereof

Also Published As

Publication number Publication date
US20230330102A1 (en) 2023-10-19
EP3458101B1 (en) 2020-12-30
US11666581B2 (en) 2023-06-06
PL3458101T3 (en) 2021-05-31
US20190175612A1 (en) 2019-06-13
JP2019522633A (en) 2019-08-15
CN109152843A (en) 2019-01-04
JP2022126886A (en) 2022-08-30
ES2858151T3 (en) 2021-09-29
EP3458101A1 (en) 2019-03-27

Similar Documents

Publication Publication Date Title
US20230330102A1 (en) Protac antibody conjugates and methods of use
US10639373B2 (en) Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US20230067037A1 (en) Conjugated chemical inducers of degradation and methods of use
EP3522933B1 (en) Methods for preparing antibody drug conjugates
EP3847166A1 (en) Polycyclic amides as cytotoxic agents
WO2020157491A1 (en) G-a crosslinking cytotoxic agents
EP4188927A1 (en) Cytotoxic agents

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2018560504

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17728325

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017728325

Country of ref document: EP

Effective date: 20181220