WO2020159504A1 - Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer - Google Patents

Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer Download PDF

Info

Publication number
WO2020159504A1
WO2020159504A1 PCT/US2019/015900 US2019015900W WO2020159504A1 WO 2020159504 A1 WO2020159504 A1 WO 2020159504A1 US 2019015900 W US2019015900 W US 2019015900W WO 2020159504 A1 WO2020159504 A1 WO 2020159504A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
fragment
amino acid
acid sequence
Prior art date
Application number
PCT/US2019/015900
Other languages
English (en)
Inventor
Ehsun Sarafraz-Yazdi
Original Assignee
Nomocan Pharmaceuticals Llc
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 Nomocan Pharmaceuticals Llc filed Critical Nomocan Pharmaceuticals Llc
Priority to PCT/US2019/015900 priority Critical patent/WO2020159504A1/fr
Priority to AU2019427766A priority patent/AU2019427766A1/en
Priority to EP19705076.8A priority patent/EP3917968A1/fr
Priority to US17/426,984 priority patent/US20220098328A1/en
Priority to CA3127776A priority patent/CA3127776A1/fr
Publication of WO2020159504A1 publication Critical patent/WO2020159504A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]

Definitions

  • the present invention relates, inter alia, to certain anti-M(H)DM2/4 antibodies (including chimeric and humanized antibodies) or antigen- binding fragments thereof, pharmaceutical compositions comprising anti-M(H)DM2/4 antibodies or antigen-binding fragments thereof, antibody-drug conjugates comprising anti-M(H)DM2/4 antibodies or antigen-binding fragments thereof bound to a cytotoxic drug, and the use of such antibodies, fragments, compositions and conjugates for treating cancer and/or for preventing metastases. 4. BACKGROUND
  • the MDM2 (MDM2 is a mouse homologue of HDM2) protein is composed of 489 amino acids and contains a p53 binding domain, two nuclear localization signals (amino acids 176-182 and 464-471), and zinc-finger motifs (amino acids 297-326 and 436-477) (see UniProt website, at UniProt Accession No. P23804).
  • Its human homologue, HDM2 is composed of 491 amino acids and contains a p53 binding domain, two nuclear localization signals (amino acids 179-185 and 466-473) and zinc-finger motifs (amino acids 299-328 and 438-479) (see UniProt website, at UniProt Accession No. Q00987).
  • Mouse protein MDM4 (also identified as MDMX) is a homologue of the MDM2 protein (see UniProt website, at UniProt Accession No. O35618), and both MDM2 and MDM4 are major negative regulators of p53 (Wade et al., 2013, Nat Rev. Cancer 13:83-96; Marine et al., 2004, Cell Cycle 3:900-904; Momand et al., 2011, Gene 486:23- 30).
  • HDM4 (also identified as HDMX) is a human homologue of MDM4 (see UniProt website, at UniProt Accession No. O15151).
  • M(H)DM2 and M(H)DM4 proteins The most conserved domain within all M(H)DM2 and M(H)DM4 proteins is the RING domain which is responsible for ubiquitination of its target proteins, including p53 protein, and heterodimerization between M(H)DM2 and M(H)DM4. M(H)DM4 is required for M(H)DM2-mediated polyubiquitination of p53.
  • a distinctive feature of M(H)DM2 and M(H)DM4 are their very complex expression pattern.
  • M(H)DM2 Multiple-sized transcripts and protein products of M(H)DM2 have been identified in cancer cells by a number of groups (Olson et al., 1993, Oncogene 8:2353-2360; Bartel et al., 2002, Cancer Cell 2:9-15 (“Bartel 2002”); Sigalas et al., 1996, Nat. Med.2:912-917 (“Sigalas 1996”); Iwakuma & Lozano, 2003, Mol. Cancer Res.1:993-1000 (“Iwakuma & Lozano 2003”)). Many types of human cancers overexpress MDM2 protein and a common characteristic among these cancers is an associated increase in mdm2 splice variants.
  • M(H)DM2 variants have been shown to be expressed in a variety of tumors such as human ovarian, bladder, breast and astrocytic neoplasms, glioblastomas, leukemia and pediatric Rhabdomyosarcoma tumors (reviewed by Iwakuma & Lozano 2003; Rosso et al., 2014, Subcell Biochem.85:247-61 (“Rosso 2014”)). Most interestingly they have been found to be more frequent in tumors of advanced stage (Bartel 2002).
  • the multiple-sized M(H)DM2 transcripts that have been shown to be splice variants forms of the M(H)DM2 mRNA have been reported to be expressed more frequently in tumor cells than in normal cells (Bartel et al., 2004, Mol. Cancer Res.2:29 (“Bartel 2004”)). It has been proposed that a mRNA surveillance system exists in untransformed cells, which degrades spliced transcripts and protects the cells from errors of transcription, mRNA processing, or mRNA transport whereas in transformed cells this system may not be functioning correctly (Bartel 2004).
  • MDM2-A and MDM2-B are common to several tumor types (Sigalas 1996). Others have only been found in specific tumors, for example MDM2-FB25 and MDM2-FB26 in pediatric rhabdomyosarcoma.
  • M(H)DM2 compartmentalization of the full-length M(H)DM2 was due to binding and sequestration by an alternative-spliced M(H)DM2 product (HDM2ALT1).
  • HDM2ALT1 alternative-spliced M(H)DM2 product
  • MDM2-D150-230 localized to the cytoplasm of U20S cells (Schuster et al., 2007, Mol. Cancer Res.5:403-412 (“Schuster 2007”)).
  • Both of these M(H)DM2 protein variants lacked part of the NH 2 -terminal region that contains a nuclear localization signal (NLS), suggesting that loss of this signal prevented the nuclear entry of these two proteins (Schuster 2007).
  • NLS nuclear localization signal
  • M(H)DM4 protein variants have also been characterized, including splicing variant MDMX-S (Lenos and Jochemsen, 2011, J. Biomed Biotechnol.,
  • HDM2 was found to be expressed in the plasma membrane of cancer cells (Sarafraz- Yazdi et al., 2010, PNAS 107:1918-1923 (“Sarafraz-Yazdi 2010”). Further, anti- cancer peptides, PNC-27 and PNC-28, which bind to HDM2 expressed in the cancer cell membranes and kill cancer cells by inducing necrosis, have been developed (Sarafraz-Yazdi 2010; Davitt et al., 2014, Annals Clin. Lab. Sci.44:241248) (the amino acid sequences of PNC-27 and PNC-28 are provided in Table I of U.S. Patent Application Publication No.2012/0177566). PNC-27 has been reported to bind within amino acids 25-109 of HDM2 (Do et al., 2003, Oncogene
  • U.S. Patent Application Publication No.2012/0177566 discloses methods of selectively necrosing cells by administering to the cells a compound (such as PNC-27 and PNC-28), including an HDM-2 targeting component and a cytotoxic component attached to the HDM-2 targeting component, wherein said compound comprises a membrane-active form.
  • a compound such as PNC-27 and PNC-28
  • cytotoxic component attached to the HDM-2 targeting component
  • PNC-27 and PNC-28 peptides which comprise a membrane resident peptide (“MRP”) and a p53 sequence
  • MRP membrane resident peptide
  • p53 sequence component the cargo (i.e., the p53 sequence component) is attached to the MRP component so as to form a cytotoxic structure (Kanovsky et al, 2001, PNAS 98:12438- 12443 (“Kanovsky 2001”); Bowne et al., 2008, Ann Surg Oncol.15:3588-3600 “Bowne 2008”)).
  • the MRP component or the HDM-2 targeting component i.e.
  • U.S. Patent No.9,765,117 discloses HDM2 targeting peptides and fusion peptides comprising an HDM2 targeting peptide and a transmembrane penetrating sequence, such as MRP; it is disclosed that MRP is required for induction of cell necrosis (see col.4, lines 27-28).
  • U.S. Patent No.9,765,117 indicates that expression of the p53 HDM2 targeting sequence in the absence of the MRP in cancer cells causes p53-dependent apoptosis and not tumor necrosis (see col.4, lines 29-32).
  • CPPs cell-penetrating peptides
  • MRPs membrane delivery of the peptides, and molecules attached to the peptides, to plasma membrane lipid bilayers, including those of normal healthy cells.
  • TAT transcriptional activator
  • Penetratin peptides enable cellular membrane delivery of the peptides, and molecules attached to the peptides, to plasma membrane lipid bilayers, including those of normal healthy cells.
  • TAT trans-activating transcriptional activator
  • Penetratin peptides enable cellular membrane delivery of the peptides, and molecules attached to the peptides, to plasma membrane lipid bilayers, including those of normal healthy cells.
  • TAT trans-activating transcriptional activator
  • Penetratin peptides enable cellular membrane delivery of the peptides, and molecules attached to the peptides, to plasma membrane lipid bilayers, including those of normal healthy cells.
  • TAT trans-activating transcriptional activator
  • Penetratin peptides enable cellular
  • CPPs such as MRPs, Membrane Transduction Domain of Antennapedia, TAT and Penetratin peptides
  • cargos such as other peptides, DNA, RNA, small molecules, antibodies or fragments thereof
  • PNC-27 and PNC-28 peptides are examples of Penetratin-/MRP-cargo conjugates that exhibit a cytotoxic function that is dependent on the attachment and linkage of their cargo to the MRP, which is required for the formation of their membrane active structure, and hence, cytotoxic function (Kanovsky 2001; Rosal 2005; Bowne 2008).
  • described herein is an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4.
  • antibodies or fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4, wherein said antibodies or fragments inhibit tumor growth in vivo (or inhibit tumor cell proliferation in vivo).
  • the antibodies or fragments described herein are not bound to a cell-penetrating peptide (e.g., a membrane resident peptide).
  • the antibodies or fragments described herein are not bound to a cytotoxic component (i.e., not bound to a cytotoxic agent).
  • antibodies or fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of
  • MCNTNMSVPTDGAVT SEQ ID NO:1
  • TTSQIPASEQE SEQ ID NO:2
  • CPVCRQPIQMIVLTYFP SEQ ID NO:3
  • M(H)DM2/4 e.g., HDM2
  • the antibody or fragment specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or
  • the antibody or fragment binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide is MCNTNMSVPTDGAVT (SEQ ID NO:1). In one embodiment, the antibody or fragment binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide is TTSQIPASEQE (SEQ ID NO:2). In one embodiment, the antibody or fragment binds to an extracellularly accessible epitope of
  • described herein is an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1.
  • described herein is an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:2. In one embodiment, described herein is an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:3.
  • a humanized antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising: (i) a heavy chain variable region (VH) comprising VH complementarity determining region (“CDR”) 1, VH CDR 2, and VH CDR3; said VH CDR 1, VH CDR 2 and VH CDR 3 being the CDRs of a VH that has an amino acid sequence selected from the group consisting of SEQ ID NO:36, SEQ ID NO:38, and SED ID NO:40, or (ii) a light chain variable region (VL) comprising VL CDR 1, VL CDR 2 and VL CDR 3 being the CDRs of a VL that has an amino acid sequence selected from the group consisting of SEQ ID NO:37, SEQ ID NO:39, and SEQ ID NO:41.
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • VH CDR 2 and VH CDR 3 being the CDRs of a VH that has an amino acid
  • the humanized antibody or a fragment that specifically binds to HDM2 comprises a VH wherein VH CDR 1, VH CDR 2 and VH CDR 3 are of a VH having the amino acid sequence of SEQ ID NO:36. In one embodiment, the humanized antibody or a fragment that specifically binds to HDM2 comprises a VH wherein VH CDR 1, VH CDR 2 and VH CDR 3 are of a VH having the amino acid sequence of SEQ ID NO:38. In one embodiment, the humanized antibody or a fragment that specifically binds to HDM2 comprises a VH wherein VH CDR 1, VH CDR 2 and VH CDR 3 are of a VH having the amino acid sequence of SEQ ID NO:40.
  • the humanized antibody or a fragment that specifically binds to HDM2 comprises a VL wherein VL CDR 1, VL CDR 2 and VL CDR 3 are of a VL having the amino acid sequence of SEQ ID NO:37 (and, optionally, a VH wherein VH CDR 1, VH CDR 2 and VH CDR 3 are of a VH having the amino acid sequence of SEQ ID NO:36).
  • the humanized antibody or a fragment that specifically binds to HDM2 comprises a VL wherein VL CDR 1, VL CDR 2 and VL CDR 3 are of a VL having the amino acid sequence of SEQ ID NO:39 (and, optionally, a VH wherein VH CDR 1, VH CDR 2 and VH CDR 3 are of a VH having the amino acid sequence of SEQ ID NO:38).
  • the humanized antibody or a fragment that specifically binds to HDM2 comprises a VL wherein VL CDR 1, VL CDR 2 and VL CDR 3 are of a VL having the amino acid sequence of SEQ ID NO:41 (and, optionally, the a VH wherein VH CDR 1, VH CDR 2 and VH CDR 3 are of a VH having the amino acid sequence of SEQ ID NO:40).
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • VH CDR 1 has the amino acid sequence GFTFTHY (SEQ ID NO:18)
  • VH CDR 2 has the amino acid sequence RNKAKGYT (SEQ ID NO:19)
  • VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • VH CDR 1 has the amino acid sequence GFTFTHYYMS (SEQ ID NO:42), the VH CDR 2 has the amino acid sequence FIRNKAKGYTAE (SEQ ID NO:45), and the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • the VH CDR 1 has the amino acid sequence HYYMS (SEQ ID NO:43)
  • the VH CDR 2 has the amino acid sequence FIRNKAKGYTAEYSASVKG (SEQ ID NO:46)
  • the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • the VH CDR 1 has the amino acid sequence THYYMS (SEQ ID NO:44), the VH CDR 2 has the amino acid sequence WLGFIRNKAKGYTAE (SEQ ID NO:47), and the VH CDR 3 has the amino acid sequence ARDIGD (SEQ ID NO:48); or
  • the VH CDR 1 has the amino acid sequence FTFTHYY (SEQ ID NO:144), the VH CDR 2 has the amino acid sequence IRNKAKGYTA (SEQ ID NO:145), and the VH CDR 3 has the amino acid sequence ARDIGDN (SEQ ID NO:146).
  • an antibody or a fragment thereof that specifically binds to M(H)DM2/4 (e.g., HDM2), said antibody or fragment comprising a heavy chain variable region (VH) comprising VH complementarity determining region (“CDR”) 1, VH CDR 2, and VH CDR 3, wherein:
  • VH CDR 1 has the amino acid sequence GDTLSGS (SEQ ID NO:24)
  • VH CDR 2 has the amino acid sequence HLNRGT (SEQ ID NO:25)
  • VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • VH CDR 1 has the amino acid sequence GDTLSGSWMH (SEQ ID NO:52)
  • VH CDR 2 has the amino acid sequence EIHLNRGTTN (SEQ ID NO:55)
  • VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • the VH CDR 1 has the amino acid sequence GSWMH (SEQ ID NO:53), the VH CDR 2 has the amino acid sequence EIHLNRGTTNYNEKFKG (SEQ ID NO:56), and the VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • the VH CDR 1 has the amino acid sequence SGSWMH (SEQ ID NO:54), the VH CDR 2 has the amino acid sequence WIGEIHLNRGTTN (SEQ ID NO:57), and the VH CDR 3 has the amino acid sequence ARSPGFA (SEQ ID NO:58); or
  • the VH CDR 1 has the amino acid sequence GDTLSGSW (SEQ ID NO:148)
  • the VH CDR 2 has the amino acid sequence IHLNRGTT (SEQ ID NO:143)
  • the VH CDR 3 has the amino acid sequence ARSPGFA (SEQ ID NO:58).
  • an antibody or a fragment thereof that specifically binds to M(H)DM2/4 e.g., HDM2
  • said antibody or fragment comprising a heavy chain variable region (VH) comprising VH complementarity determining region (“CDR”) 1, VH CDR 2, and VH CDR 3, wherein:
  • the VH CDR 1 has the amino acid sequence GYTFTSY (SEQ ID NO:30)
  • the VH CDR 2 has the amino acid sequence NPRNGG (SEQ ID NO:31)
  • the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32);
  • the VH CDR 1 has the amino acid sequence GYTFTSYYMY (SEQ ID NO:62), the VH CDR 2 has the amino acid sequence GINPRNGGTN (SEQ ID NO:65), and the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32); (iii) the VH CDR 1 has the amino acid sequence SYYMY (SEQ ID NO:63), the VH CDR 2 has the amino acid sequence GINPRNGGTNFNEKFKN (SEQ ID NO:66), and the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32); or
  • the VH CDR 1 has the amino acid sequence TSYYMY (SEQ ID NO:64), the VH CDR 2 has the amino acid sequence WIGGINPRNGGTN (SEQ ID NO:67), and the VH CDR 3 has the amino acid sequence TRSGYYAMD (SEQ ID NO:68).
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • VH CDR 1 has the amino acid sequence GFTFTHY (SEQ ID NO:18)
  • VH CDR 2 has the amino acid sequence RNKAKGYT (SEQ ID NO:19)
  • VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • VH CDR 1 has the amino acid sequence GFTFTHYYMS (SEQ ID NO:42), the VH CDR 2 has the amino acid sequence FIRNKAKGYTAE (SEQ ID NO:45), and the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • the VH CDR 1 has the amino acid sequence HYYMS (SEQ ID NO:43)
  • the VH CDR 2 has the amino acid sequence FIRNKAKGYTAEYSASVKG (SEQ ID NO:46)
  • the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • the VH CDR 1 has the amino acid sequence THYYMS (SEQ ID NO:44), the VH CDR 2 has the amino acid sequence WLGFIRNKAKGYTAE (SEQ ID NO:47), and the VH CDR 3 has the amino acid sequence ARDIGD (SEQ ID NO:48); or
  • the VH CDR 1 has the amino acid sequence FTFTHYY (SEQ ID NO:144)
  • the VH CDR 2 has the amino acid sequence IRNKAKGYTA (SEQ ID NO:145)
  • the VH CDR 3 has the amino acid sequence ARDIGDN (SEQ ID NO:146).
  • a humanized antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising a VH, wherein the VH comprises VH CDR1, VH CDR2, and VH CDR 3, wherein:
  • VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • VH CDR 3 has the amino acid sequence ARDIGD (SEQ ID NO:48); or
  • VH CDR 3 has the amino acid sequence ARDIGDN (SEQ ID NO:146).
  • VL light chain variable region
  • CDR VL complementarity determining region
  • VL CDR 1 has the amino acid sequence RSSKNLLHSNGITYLY (SEQ ID NO:21)
  • the VL CDR 2 has the amino acid sequence RVSNLAS (SEQ ID NO:22)
  • the VL CDR 3 has the amino acid sequence AQLLELPYT (SEQ ID NO:23);
  • VL CDR 1 has the amino acid sequence LHSNGITYLYWY (SEQ ID NO:49)
  • VL CDR 2 has the amino acid sequence LLISRVSNLA (SEQ ID NO:50)
  • VL CDR 3 has the amino acid sequence AQLLELPY (SEQ ID NO:51);
  • the VL CDR 1 has the amino acid sequence KNLLHSNGITY (SEQ ID NO:147), the VL CDR 2 has the amino acid sequence RVS, and the VL CDR 3 has the amino acid sequence AQLLELPYT (SEQ ID NO:23).
  • a humanized antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising a light chain variable region (VL) comprising VL complementarity determining region (“CDR”) 1, VL CDR 2, and VL CDR 3, wherein:
  • VL CDR 1 has the amino acid sequence RSSKNLLHSNGITYLY (SEQ ID NO:21)
  • the VL CDR 2 has the amino acid sequence RVSNRAS (SEQ ID NO:236)
  • the VL CDR 3 has the amino acid sequence AQLLELPYT (SEQ ID NO:23);
  • VL CDR 1 has the amino acid sequence LHSNGITYLYWY (SEQ ID NO:49)
  • VL CDR 2 has the amino acid sequence LLISRVSNRAS (SEQ ID NO:237)
  • VL CDR 3 has the amino acid sequence AQLLELPY (SEQ ID NO:51).
  • an antibody or a fragment comprising:
  • VL light chain variable region
  • CDR VL complementarity determining region
  • VL CDR 1 has the amino acid sequence RSSKNLLHSNGITYLY (SEQ ID NO:21)
  • the VL CDR 2 has the amino acid sequence RVSNRAS (SEQ ID NO:236)
  • the VL CDR 3 has the amino acid sequence AQLLELPYT (SEQ ID NO:23);
  • VL CDR 1 has the amino acid sequence LHSNGITYLYWY (SEQ ID NO:49)
  • VL CDR 2 has the amino acid sequence LLISRVSNRAS (SEQ ID NO:237)
  • VL CDR 3 has the amino acid sequence AQLLELPY (SEQ ID NO:51); and
  • VH heavy chain variable region comprising VH complementarity determining region (“CDR”) 1, VH CDR 2, and VH CDR 3, wherein:
  • VH CDR 1 has the amino acid sequence GFTFTHY (SEQ ID NO:18)
  • VH CDR 2 has the amino acid sequence RNKAKGYT (SEQ ID NO:19)
  • VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • VH CDR 1 has the amino acid sequence GFTFTHYYMS (SEQ ID NO:42), the VH CDR 2 has the amino acid sequence FIRNKAKGYTAE (SEQ ID NO:45), and the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • the VH CDR 1 has the amino acid sequence HYYMS (SEQ ID NO:43)
  • the VH CDR 2 has the amino acid sequence FIRNKAKGYTAEYSASVKG (SEQ ID NO:46)
  • the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • the VH CDR 1 has the amino acid sequence THYYMS (SEQ ID NO:44), the VH CDR 2 has the amino acid sequence WLGFIRNKAKGYTAE (SEQ ID NO:47), and the VH CDR 3 has the amino acid sequence ARDIGD (SEQ ID NO:48); or
  • the VH CDR 1 has the amino acid sequence FTFTHYY (SEQ ID NO:144)
  • the VH CDR 2 has the amino acid sequence IRNKAKGYTA (SEQ ID NO:145)
  • the VH CDR 3 has the amino acid sequence ARDIGDN (SEQ ID NO:146).
  • a humanized antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising:
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • VH CDR 1 has the amino acid sequence GFTFTHY (SEQ ID NO:18)
  • VH CDR 2 has the amino acid sequence RNKAKGYT (SEQ ID NO:19)
  • VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • VH CDR 1 has the amino acid sequence GFTFTHYYMS (SEQ ID NO:42), the VH CDR 2 has the amino acid sequence FIRNKAKGYTAE (SEQ ID NO:45), and the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20);
  • the VH CDR 1 has the amino acid sequence HYYMS (SEQ ID NO:43), the VH CDR 2 has the amino acid sequence FIRNKAKGYTAEYSASVKG (SEQ ID NO:46), and the VH CDR 3 has the amino acid sequence DIGDN (SEQ ID NO:20); (iv) the VH CDR 1 has the amino acid sequence THYYMS (SEQ ID NO:44), the VH CDR 2 has the amino acid sequence WLGFIRNKAKGYTAE (SEQ ID NO:47), and the VH CDR 3 has the amino acid sequence ARDIGD (SEQ ID NO:48); or
  • the VH CDR 1 has the amino acid sequence FTFTHYY (SEQ ID NO:144), the VH CDR 2 has the amino acid sequence IRNKAKGYTA (SEQ ID NO:145), and the VH CDR 3 has the amino acid sequence ARDIGDN (SEQ ID NO:146); and
  • VL light chain variable region
  • CDR VL complementarity determining region
  • VL CDR 1 has the amino acid sequence RSSKNLLHSNGITYLY (SEQ ID NO:21)
  • the VL CDR 2 has the amino acid sequence RVSNLAS (SEQ ID NO:22)
  • the VL CDR 3 has the amino acid sequence AQLLELPYT (SEQ ID NO:23);
  • VL CDR 1 has the amino acid sequence LHSNGITYLYWY (SEQ ID NO:49)
  • VL CDR 2 has the amino acid sequence LLISRVSNLA (SEQ ID NO:50)
  • VL CDR 3 has the amino acid sequence AQLLELPY (SEQ ID NO:51);
  • the VL CDR 1 has the amino acid sequence KNLLHSNGITY (SEQ ID NO:147), the VL CDR 2 has the amino acid sequence RVS, and the VL CDR 3 has the amino acid sequence AQLLELPYT (SEQ ID NO:23).
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • VH CDR 1 has the amino acid sequence GDTLSGS (SEQ ID NO:24)
  • VH CDR 2 has the amino acid sequence HLNRGT (SEQ ID NO:25)
  • VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • VH CDR 1 has the amino acid sequence GDTLSGSWMH (SEQ ID NO:52)
  • VH CDR 2 has the amino acid sequence EIHLNRGTTN (SEQ ID NO:55)
  • VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • the VH CDR 1 has the amino acid sequence GSWMH (SEQ ID NO:53), the VH CDR 2 has the amino acid sequence EIHLNRGTTNYNEKFKG (SEQ ID NO:56), and the VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • the VH CDR 1 has the amino acid sequence SGSWMH (SEQ ID NO:54), the VH CDR 2 has the amino acid sequence WIGEIHLNRGTTN (SEQ ID NO:57), and the VH CDR 3 has the amino acid sequence ARSPGFA (SEQ ID NO:58); or
  • the VH CDR 1 has the amino acid sequence GDTLSGSW (SEQ ID NO:148)
  • the VH CDR 2 has the amino acid sequence IHLNRGTT (SEQ ID NO:143)
  • the VH CDR 3 has the amino acid sequence ARSPGFA (SEQ ID NO:58).
  • an antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising a VH, wherein the VH comprises VH CDR1, VH CDR2, and VH CDR 3, wherein:
  • VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26); or
  • VH CDR 3 has the amino acid sequence ARSPGFA (SEQ ID NO:58).
  • VL light chain variable region
  • CDR VL complementarity determining region
  • VL CDR 1 has the amino acid sequence RSSKSLLHSNGNSYLY (SEQ ID NO:27)
  • the VL CDR 2 has the amino acid sequence RMSNLAS (SEQ ID NO:28)
  • the VL CDR 3 has the amino acid sequence MQHLEYPFT (SEQ ID NO:29);
  • the VL CDR 1 has the amino acid sequence LHSNGNSYLYWF (SEQ ID NO:59), the VL CDR 2 has the amino acid sequence LLIYRMSNLA (SEQ ID NO:60), and the VL CDR 3 has the amino acid sequence MQHLEYPF (SEQ ID NO:61); or
  • the VL CDR 1 has the amino acid sequence KSLLHSNGNSY (SEQ ID NO:141), the VL CDR 2 has the amino acid sequence RMS, and the VL CDR 3 has the amino acid sequence MQHLEYPFT (SEQ ID NO:29).
  • an antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising:
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • the VH CDR 1 has the amino acid sequence GDTLSGS (SEQ ID NO:24), the VH CDR 2 has the amino acid sequence HLNRGT (SEQ ID NO:25), and the VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • the VH CDR 1 has the amino acid sequence GDTLSGSWMH (SEQ ID NO:52), the VH CDR 2 has the amino acid sequence EIHLNRGTTN (SEQ ID NO:55), and the VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • the VH CDR 1 has the amino acid sequence GSWMH (SEQ ID NO:53), the VH CDR 2 has the amino acid sequence EIHLNRGTTNYNEKFKG (SEQ ID NO:56), and the VH CDR 3 has the amino acid sequence SPGFAY (SEQ ID NO:26);
  • the VH CDR 1 has the amino acid sequence SGSWMH (SEQ ID NO:54), the VH CDR 2 has the amino acid sequence WIGEIHLNRGTTN (SEQ ID NO:57), and the VH CDR 3 has the amino acid sequence ARSPGFA (SEQ ID NO:58); or
  • the VH CDR 1 has the amino acid sequence GDTLSGSW (SEQ ID NO:148), the VH CDR 2 has the amino acid sequence IHLNRGTT (SEQ ID NO:143), and the VH CDR 3 has the amino acid sequence ARSPGFA (SEQ ID NO:58); and
  • VL light chain variable region
  • CDR VL complementarity determining region
  • VL CDR 1 has the amino acid sequence RSSKSLLHSNGNSYLY (SEQ ID NO:27)
  • the VL CDR 2 has the amino acid sequence RMSNLAS (SEQ ID NO:28)
  • the VL CDR 3 has the amino acid sequence MQHLEYPFT (SEQ ID NO:29);
  • the VL CDR 1 has the amino acid sequence LHSNGNSYLYWF (SEQ ID NO:59), the VL CDR 2 has the amino acid sequence LLIYRMSNLA (SEQ ID NO:60), and the VL CDR 3 has the amino acid sequence MQHLEYPF (SEQ ID NO:61); or
  • the VL CDR 1 has the amino acid sequence KSLLHSNGNSY (SEQ ID NO:141), the VL CDR 2 has the amino acid sequence RMS, and the VL CDR 3 has the amino acid sequence MQHLEYPFT (SEQ ID NO:29).
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • the VH CDR 1 has the amino acid sequence GYTFTSY (SEQ ID NO:30), the VH CDR 2 has the amino acid sequence NPRNGG (SEQ ID NO:31), and the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32);
  • the VH CDR 1 has the amino acid sequence GYTFTSYYMY (SEQ ID NO:62), the VH CDR 2 has the amino acid sequence GINPRNGGTN (SEQ ID NO:65), and the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32);
  • the VH CDR 1 has the amino acid sequence SYYMY (SEQ ID NO:63)
  • the VH CDR 2 has the amino acid sequence GINPRNGGTNFNEKFKN (SEQ ID NO:66)
  • the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32); or
  • the VH CDR 1 has the amino acid sequence TSYYMY (SEQ ID NO:64)
  • the VH CDR 2 has the amino acid sequence WIGGINPRNGGTN (SEQ ID NO:67)
  • the VH CDR 3 has the amino acid sequence TRSGYYAMD (SEQ ID NO:68).
  • an antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising a VH, wherein the VH comprises VH CDR1, VH CDR2, and VH CDR 3, wherein:
  • VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32); or
  • VH CDR 3 has the amino acid sequence TRSGYYAMD (SEQ ID NO:68).
  • an antibody or fragment further comprises a light chain variable region (VL) comprising VL complementarity determining region (“CDR”) 1, VL CDR 2, and VL CDR 3, wherein:
  • VL CDR 1 has the amino acid sequence RASQDISNFLN (SEQ ID NO:33)
  • VL CDR 2 has the amino acid sequence YTSRLHS (SEQ ID NO:34)
  • VL CDR 3 has the amino acid sequence QQGNTLPRT (SEQ ID NO:35);
  • the VL CDR 1 has the amino acid sequence SNFLNWY (SEQ ID NO:69)
  • the VL CDR 2 has the amino acid sequence LLIYYTSRLH (SEQ ID NO:70)
  • the VL CDR 3 has the amino acid sequence QQGNTLPR (SEQ ID NO:71).
  • an antibody or a fragment thereof that specifically binds to HDM2 said antibody or fragment comprising:
  • VH heavy chain variable region
  • CDR VH complementarity determining region
  • the VH CDR 1 has the amino acid sequence GYTFTSY (SEQ ID NO:30), the VH CDR 2 has the amino acid sequence NPRNGG (SEQ ID NO:31), and the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32);
  • the VH CDR 1 has the amino acid sequence GYTFTSYYMY (SEQ ID NO:62), the VH CDR 2 has the amino acid sequence GINPRNGGTN (SEQ ID NO:65), and the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32);
  • the VH CDR 1 has the amino acid sequence SYYMY (SEQ ID NO:63)
  • the VH CDR 2 has the amino acid sequence GINPRNGGTNFNEKFKN (SEQ ID NO:66)
  • the VH CDR 3 has the amino acid sequence SGYYAMDY (SEQ ID NO:32); or
  • the VH CDR 1 has the amino acid sequence TSYYMY (SEQ ID NO:64), the VH CDR 2 has the amino acid sequence WIGGINPRNGGTN (SEQ ID NO:67), and the VH CDR 3 has the amino acid sequence TRSGYYAMD (SEQ ID NO:68); and
  • VL light chain variable region
  • CDR VL complementarity determining region
  • VL CDR 1 has the amino acid sequence RASQDISNFLN (SEQ ID NO:33)
  • VL CDR 2 has the amino acid sequence YTSRLHS (SEQ ID NO:34)
  • VL CDR 3 has the amino acid sequence QQGNTLPRT (SEQ ID NO:35);
  • the VL CDR 1 has the amino acid sequence SNFLNWY (SEQ ID NO:69)
  • the VL CDR 2 has the amino acid sequence LLIYYTSRLH (SEQ ID NO:70)
  • the VL CDR 3 has the amino acid sequence QQGNTLPR (SEQ ID NO:71).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VH having the amino acid sequence of SEQ ID NO:36, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:36.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VL having the amino acid sequence of SEQ ID NO:37, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:37 (and, optionally, comprising a VH having the amino acid sequence of SEQ ID NO:36, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:36).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VH having the amino acid sequence of SEQ ID NO:38, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:38.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VL having the amino acid sequence of SEQ ID NO:39, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:39 (and, optionally, comprising a VH having the amino acid sequence of SEQ ID NO:38, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:38).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VH having the amino acid sequence of SEQ ID NO:40, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:40.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VL having the amino acid sequence of SEQ ID NO:41, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:41 (and, optionally, comprising a VH having the amino acid sequence of SEQ ID NO:40, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:40).
  • VH heavy chain variable region
  • VL light chain variable region
  • a humanized antibody or a fragment thereof that specifically binds to HDM2 or MDM2 said antibody or fragment comprising: (a) a heavy chain variable region (VH) comprising a VH having an amino acid sequence selected from the group consisting of SEQ ID NO:287, SEQ ID NO:291, SEQ ID NO:295, SEQ ID NO:299, SEQ ID NO:305, and SEQ ID NO:309, and
  • VL light chain variable region
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VH having the amino acid sequence of SEQ ID NO:287, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:287.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VH having the amino acid sequence of SEQ ID NO:291, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:291.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VH having the amino acid sequence of SEQ ID NO:295, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:295.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VH having the amino acid sequence of SEQ ID NO:299, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:299.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VH having the amino acid sequence of SEQ ID NO:305, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:305.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VH having the amino acid sequence of SEQ ID NO:309, or a VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:309.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:289, or a VL having at least 95% sequence identity to the amino acid sequence of SEQ ID NO:289.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:293, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:293.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:297, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:297.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:301, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:301.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:303, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:303.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:307, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:307.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:311, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:311.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:299, and (ii) a VL having the amino acid sequence of SEQ ID NO:297.
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising: (a) a heavy chain variable region (VH) comprising a VH having an amino acid sequence selected from the group consisting of SEQ ID NO:287, SEQ ID NO:291, SEQ ID NO:295, and SEQ ID NO:299, and (ii) a light chain variable region (VL) comprising a VL having an amino acid sequence selected from the group consisting of SEQ ID NO:289, SEQ ID NO:293, SEQ ID NO:297, SEQ ID NO:301, and SEQ ID NO:303.
  • VH heavy chain variable region
  • VL light chain variable region
  • a humanized antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising: (a) a heavy chain variable region (VH) comprising a VH having an amino acid sequence selected from the group consisting of SEQ ID NO:299, SEQ ID NO:305, and SEQ ID NO:309, and (ii) a light chain variable region (VL) comprising a VL having an amino acid sequence selected from the group consisting of SEQ ID NO:297, SEQ ID NO:307, and SEQ ID NO:311.
  • VH heavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VH having the amino acid sequence of SEQ ID NO: 1
  • VH having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:283.
  • an antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a VL having the amino acid sequence of SEQ ID NO:285, or a VL having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:285.
  • an antibody or fragment thereof that specifically binds to HDM2 or MDM2 said antibody comprising:
  • an antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a heavy chain having the amino acid sequence of SEQ ID NO:312, or a heavy chain having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO:312.
  • an antibody or fragment thereof that specifically binds to HDM2 or MDM2 comprising a light chain having the amino acid sequence of SEQ ID NO:313, or a light chain having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the amino acid sequence of SEQ ID NO: 313.
  • an antibody or fragment thereof that specifically binds to HDM2 or MDM2, which inhibits tumor cell proliferation in vivo.
  • the anti-M(H)DM2/4 antibody described herein is a monoclonal antibody.
  • the anti-M(H)DM2/4 antibody described herein is a human, humanized, or a chimeric antibody (e.g., a human, humanized or chimeric monoclonal antibody).
  • the anti-M(H)DM2/4 antibody described herein is a human antibody.
  • the anti-M(H)DM2/4 antibody described herein is a humanized antibody.
  • the anti-M(H)DM2/4 antibody described herein is a chimeric antibody.
  • the anti-M(H)DM2/4 antibody described herein is a purified antibody.
  • the anti-M(H)DM2/4 antibody described herein is an immunoglobulin (e.g., IgG or IgM).
  • the immunoglobulin is an IgG.
  • the immunoglobulin is an IgM.
  • the immunoglobulin is of IgG1 isotype.
  • the immunoglobulin is of IgG3 isotype.
  • the immunoglobulin is of IgG2 isotype.
  • the immunoglobulin is of IgG4 isotype.
  • the anti- M(H)DM2/4 antibody described herein comprises an Fc region, wherein the Fc region is a human IgG Fc region or a human IgM Fc region.
  • the anti- M(H)DM2/4 antibody described herein comprises an Fc region, which is a human IgG1 Fc region, a human IgG2 Fc region, or a human IgG3 Fc region, or a human IgG4 Fc region.
  • the anti- M(H)DM2/4 antibody described herein comprises a human IgG1 Fc region.
  • the anti-M(H)DM2/4 antibody described herein comprises a human IgG3 Fc region.
  • the anti- M(H)DM2/4 antibody described herein comprises a human IgG4 Fc region. In one embodiment, the anti- M(H)DM2/4 antibody described herein comprises a human IgG2 Fc region. In one embodiment, the anti- M(H)DM2/4 antibody described herein comprises a human IgM Fc region. In one embodiment, the anti-M(H)DM2/4 antibody described herein comprises a human IgE Fc region.
  • the anti-M(H)DM2/4 antibody or fragment described herein is an antigen-binding fragment of an anti-M(H)DM2/4 antibody.
  • the antibody or fragment described herein is an Fv fragment, a Fab fragment, a Fab ⁇ fragment, a F(ab ⁇ ) 2 fragment, a single chain antibody molecule, or a single chain Fv (scFv).
  • the antibody or fragment described herein is an Fv fragment.
  • the antibody or fragment described herein is a Fab fragment.
  • the antibody or fragment described herein is a Fab ⁇ fragment.
  • the antibody or fragment described herein is a F(ab ⁇ ) 2 fragment.
  • the antibody or fragment described herein is a single chain antibody molecule.
  • the antibody or fragment described herein is a single chain Fv (scFv).
  • the anti-M(H)DM2/4 antibody or antigen-binding fragment described herein mediates complement-dependent cytotoxicity (CDC) or antibody-dependent cell-mediated cytoxicity (ADCC). In one embodiment, the anti- M(H)DM2/4 antibody or antigen-binding fragment mediates complement-dependent cytotoxicity (CDC). [0069] In certain embodiments, the anti-M(H)DM2/4 antibody described herein is a bispecific antibody that also specifically binds to a cell surface antigen of an effector cell (e.g., a T cell, a B lymphocyte, a neutrophil, a macrophage, a natural killer cell, or a dendritic cell).
  • an effector cell e.g., a T cell, a B lymphocyte, a neutrophil, a macrophage, a natural killer cell, or a dendritic cell.
  • the antibody or fragment described herein specifically binds to an extracellularly accessible epitope of M(H)DM2 (e.g., HDM2) and does not bind to M(H)DM4 (e.g., HDM4).
  • the antibody or fragment described herein specifically binds to extracellularly accessible epitopes of both M(H)DM2 (e.g., HDM2) and M(H)DM4 (e.g., HDM4).
  • the antibody or fragment described herein specifically binds to an extracellularly accessible epitope of HDM2, and optionally, may also bind to an
  • the anti-HDM2 antibody or fragment described herein specifically binds HDM2 within amino acids of SEQ ID NO: 1 (which are amino acids 1 to 15 of HDM2 (SEQ ID NO:4)).
  • the anti-HDM2 antibody described herein specifically binds HDM2 within amino acids of SEQ ID NO: 2 (which are amino acids 15 to 25 of HDM2 (SEQ ID NO:4)).
  • the anti-HDM2 antibody described herein specifically binds HDM2 within amino acids of SEQ ID NO: 3 (which are amino acids 475-491 of HDM2 (SEQ ID NO:4)).
  • the anti- HDM2 antibody described herein specifically binds within amino acids 19 to 50 of SEQ ID NO: 4. In another specific embodiment, the anti-HDM2 antibody described herein specifically binds within amino acids 154 to 167 of SEQ ID NO: 4. In yet another specific embodiment, the anti- HDM2 antibody described herein specifically binds within amino acids 1 to 60 of SEQ ID NO: 4. In yet another specific embodiment, the anti- HDM2 antibody described herein specifically binds within amino acids 1 to 100 of SEQ ID NO: 4. In another specific embodiment, the anti- HDM2 antibody described herein specifically binds within amino acids 100 to 110 of SEQ ID NO: 4.
  • the anti-HDM2 antibody described herein specifically binds within amino acids 50 to 60 of SEQ ID NO: 4. In yet another specific embodiment, the anti-HDM2 antibody described herein specifically binds within amino acids 1 to 109 of SEQ ID NO: 4. In another specific embodiment, the anti-HDM2 antibody described herein specifically binds within amino acids 26 to 60 of SEQ ID NO: 4. In one specific embodiment, the anti- HDM2 antibody described herein specifically binds within the terminal 60 amino acids at the C- terminus of the HDM2 on the plasma membrane of the cancer cells. In another specific embodiment, the anti-HDM2 antibody described herein specifically binds within the terminal 100 amino acids at the C-terminus of the HDM2 on the plasma membrane of the cancer cells.
  • the anti- M(H)DM2/4 antibody described herein does not bind within amino acids 101 to 200 of SEQ ID NO:4. In one embodiment, the anti- M(H)DM2/4 antibody described herein does not bind to the epitope of HDM2 or MDM2 to which“MDM2 monoclonal antibody (M01), clone 1A7” (Abnova, Cat. No. H00004193-M01) binds. In one embodiment, the anti- M(H)DM2/4 antibody described herein does not compete with“MDM2 monoclonal antibody (M01), clone 1A7” (Abnova, Cat. No. H00004193-M01) for binding to HDM2.
  • the anti- M(H)DM2/4 antibody described herein binds within amino acids 101 to 200 of SEQ ID NO:4. In one embodiment, the anti- M(H)DM2/4 antibody described herein binds to the epitope of HDM2 or MDM2 to which“MDM2 monoclonal antibody (M01), clone 1A7” (Abnova, Cat. No. H00004193-M01) binds. In one embodiment, the anti- M(H)DM2/4 antibody described herein competes with“MDM2 monoclonal antibody (M01), clone 1A7” (Abnova, Cat. No. H00004193-M01) for binding to HDM2.
  • the anti- M(H)DM2/4 antibody described herein does not bind within amino acids 153 to 222 of SEQ ID NO:4. In one embodiment, the anti- M(H)DM2/4 antibody described herein does not bind within amino acids 26 to 169 of SEQ ID NO:4. In a specific embodiment, the anti- M(H)DM2/4 antibody described herein does not bind within amino acids 26 to 222 of SEQ ID NO:4.
  • the M(H)DM2/4 exposed on the surface of cancer cells being targeted by the antibodies or fragments described herein is an M(H)DM2/4 variant that lacks one or more nuclear localization signal domains.
  • the HDM2 exposed on the surface of cancer cells being targeted by the antibodies or fragments described herein is an HDM2 variant that lacks the sequence of amino acids 179 to 185 of SEQ ID NO: 4 and/or the sequence of amino acids 464 to 471 of SEQ ID NO: 4.
  • the HDM2 exposed on the surface of cancer cells being targeted by the antibodies or fragments described herein is an HDM2 variant that lacks the sequence of amino acids 181 to 185 of SEQ ID NO: 4.
  • antibodies or fragments thereof that compete for binding to M(H)DM2/4 with an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment thereof described herein.
  • anti-M(H)DM2/4 e.g., anti-HDM2
  • such antibodies or fragments that compete for binding are monoclonal antibodies or fragments thereof.
  • antibodies or fragments thereof that compete for binding to M(H)DM2/4 with a mouse anti-HDM2 immunoglobulin (preferably IgG) antibody selected from the group consisting of: (i) an antibody comprising a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO:36, and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO:37; (ii) an antibody comprising a VH having the amino acid sequence of SEQ ID NO:38, and a VL having the amino acid sequence of SEQ ID NO:39; and (iii) an antibody comprising a VH having the amino acid sequence of SEQ ID NO:40, and a VL having the amino acid sequence of SEQ ID NO:41.
  • a mouse anti-HDM2 immunoglobulin (preferably IgG) antibody selected from the group consisting of: (i) an antibody comprising a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO:36, and a light chain variable region (
  • antibodies or fragments thereof that: (i) compete for binding to a peptide of sequence SEQ ID NO:1 with a mouse anti-HDM2 IgG1 antibody comprising a heavy chain variable region (VH) having the amino acid sequence of SEQ ID NO:36, and a light chain variable region (VL) having the amino acid sequence of SEQ ID NO:37; or (ii) compete for binding to a peptide of SEQ ID NO:2 with a mouse anti-HDM2 IgG3 antibody comprising a VH having the amino acid sequence of SEQ ID NO:38, and a VL having the amino acid sequence of SEQ ID NO:39; or (iii) compete for binding to a peptide of SEQ ID NO:3 with a mouse IgM antibody comprising a VH having the amino acid sequence of SEQ ID NO:40, and a VL having the amino acid sequence of SEQ ID NO:41.
  • antibody-drug conjugates comprising any antibody or fragment described herein (e.g., an antibody-drug conjugate in which an anti-M(H)DM2/4 antibody or fragment thereof described herein is covalently bound to a cytotoxic drug).
  • compositions comprising a therapeutically effective amount of any antibody or fragment described herein.
  • compositions comprising a therapeutically effective amount of any chimeric or humanized anti-M(H)DM2/4 antibody or fragment described herein.
  • kits for treating cancer in a subject in need thereof comprising administering to the subject: (i) any anti-M(H)DM2/4 antibody described herein; (ii) an antibody or a fragment thereof that specifically binds to an
  • a cell-penetrating peptide e.g., a membrane resident peptide
  • an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug , (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • provided herein are methods for treating cancer in a subject in need thereof, said method comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • a method for treating cancer in a subject in need thereof comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell-penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug , (iii) any
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate described herein.
  • a method for treating cancer in a subject in need thereof comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of
  • MCNTNMSVPTDGAVT SEQ ID NO:1
  • TTSQIPASEQE SEQ ID NO:2
  • CPVCRQPIQMIVLTYFP SEQ ID NO:3
  • a method for treating cancer in a subject in need thereof comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • provided herein is a method for treating cancer in a subject in need thereof, said method comprising administering to the subject any chimeric or humanized anti-M(H)DM2/4 antibody or fragment described herein.
  • methods for inhibiting tumor growth in a subject in need thereof comprising administering to the subject: (i) any anti-M(H)DM2/4 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein said antibody or fragment is not bound to a cell- penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody- drug conjugate described herein.
  • a cell- penetrating peptide e.
  • provided herein are methods for inhibiting tumor growth in a subject in need thereof, said method comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • a method for inhibiting tumor growth in a subject in need thereof comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell- penetrating peptide(e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetraitng peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • any anti-HDM2 antibody or fragment described herein comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 bound to a cytotoxic drug, wherein said antibody or fragment is not bound to a cell- penetrating peptide.
  • a method for inhibiting tumor growth in a subject in need thereof comprising administering to the subject an anti- M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3).
  • provided herein is a method for inhibiting tumor growth in a subject in need thereof, said method comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • a method for inhibiting tumor growth in a subject in need thereof said method comprising administering to the subject any chimeric or humanized anti- M(H)DM2/4 antibody or fragment described herein.
  • methods for inhibiting tumor progression in a subject in need thereof said method comprising
  • any anti-M(H)DM2/4 antibody or fragment described herein comprising an extracellularly accessible epitope of M(H)DM2/4, wherein said antibody or fragment is not bound to a cell- penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody- drug conjugate described herein.
  • a cell- penetrating peptide e.g., a membrane resident peptide
  • an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug,
  • any pharmaceutical composition described herein or (iv) any antibody- drug conjugate described herein.
  • provided herein are methods for inhibiting tumor progression in a subject in need thereof, said method comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • a method for inhibiting tumor progression in a subject in need thereof comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell- penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • a cell- penetrating peptide e.g., a membrane resident peptide
  • an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 bound to a cytotoxic drug, wherein said antibody or fragment is not bound to a cell- penetrating peptide.
  • a method for inhibiting tumor progression in a subject in need thereof comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3).
  • provided herein is a method for inhibiting tumor progression in a subject in need thereof, said method comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • a method for preventing cancer recurrence or relapse in a subject in need thereof comprising administering to the subject: (i) any anti-M(H)DM2/4 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an
  • a cell- penetrating peptide e.g., a membrane resident peptide
  • an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • a method for preventing cancer recurrence or relapse in a subject in need thereof comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 e.g., HDM2
  • a method for preventing cancer recurrence or relapse in a subject in need thereof comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell- penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell- penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • any anti-HDM2 antibody or fragment described herein comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HD
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 bound to a cytotoxic drug, wherein said antibody or fragment is not bound to a cell-penetrating peptide.
  • a method for preventing cancer recurrence or relapse in a subject in need thereof comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or
  • CPVCRQPIQMIVLTYFP SEQ ID NO:3
  • a method for preventing cancer recurrence or relapse in a subject in need thereof comprising administering to the subject an anti- M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • provided herein is a method for preventing cancer recurrence or relapses in a subject in need thereof, said method comprising administering to the subject any chimeric or humanized anti-M(H)DM2/4 antibody or fragment described herein.
  • a method for increasing survival in a subject having a cancer comprising administering to the subject: (i) any anti-M(H)DM2/4 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an
  • a cell- penetrating peptide e.g., a membrane resident peptide
  • an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • a method for increasing survival in a subject having a cancer comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 e.g., HDM2
  • a method for increasing survival in a subject a cancer comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell- penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell- penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • any anti-HDM2 antibody or fragment described herein comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 bound to a cytotoxic drug, wherein said antibody or fragment is not bound to a cell-penetrating peptide.
  • a method for increasing survival in a subject having a cancer comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3).
  • a method for increasing survival in a subject having a cancer comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • provided herein is a method for increasing survival in a subject having a cancer (e.g., relative to a subject not treated with anti-M(H)DM2/4 antibody or fragment thereof described herein), said method comprising administering to the subject any chimeric or humanized anti-M(H)DM2/4 antibody or fragment described herein.
  • kits for preventing metastasis in a subject are provided herein.
  • any anti- M(H)DM2/4 antibody or fragment described herein comprising administering to the subject: (i) any anti- M(H)DM2/4 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein said antibody or fragment is not bound to a cell-penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug, (iii) any
  • a pharmaceutical composition described herein or (iv) any antibody-drug conjugate described herein.
  • methods for preventing metastasis in a subject in need thereof comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2)
  • methods for preventing metastasis in a subject having a cancer comprising administering to the subject: (i) any anti-HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell- penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell- penetrating peptide) bound to a cytotoxic drug, (iii) any pharmaceutical composition described herein, or (iv) any antibody-drug conjugate described herein.
  • a cell- penetrating peptide e.g., a membrane resident peptide
  • an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell- penetrating peptide) bound to a
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 bound to a cytotoxic drug, wherein said antibody or fragment is not bound to a cell-penetrating peptide.
  • a method for preventing metastasis in a subject having a cancer comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3).
  • a method for preventing metastasis in a subject having a cancer comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • a method for preventing metastasis in a subject having a cancer comprising administering to the subject any chimeric or humanized anti-M(H)DM2/4 antibody or fragment described herein.
  • methods for inhibiting metastasis e.g., reducing the number, size or invasiveness of metastases
  • a subject having a metastatic cancer comprising administering to the subject: (i) any anti-M(H)DM2/4 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed, wherein said antibody or fragment is not bound to a cell-penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic
  • provided herein are methods for inhibiting metastasis in a subject in need thereof, said method comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibody or fragment is not bound to a cytotoxic component.
  • a method for inhibiting metastasis comprising administering to the subject: (i) any anti- HDM2 antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell-penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is not bound to a cell-penetrating peptide) bound to a cytotoxic drug, (iii) any combination of the antibody or fragment described herein; (ii) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2, wherein said antibody or fragment is not bound to a cell-penetrating peptide (e.g., a membrane resident peptide), or an antibody-drug conjugate comprising the antibody or fragment (i.e., said antibody or fragment that is
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an antibody-drug conjugate described herein.
  • a method for inhibiting metastasis e.g., reducing the number, size or invasiveness of metastases
  • a method for inhibiting metastasis comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3).
  • a method for inhibiting metastasis comprising administering to the subject an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 contained within a peptide of SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • a method for inhibiting metastasis comprising administering to the subject any chimeric or humanized anti-M(H)DM2/4 antibody or fragment described herein.
  • kits for treating cancer in a subject who has experienced an accelerated rate of cancer growth in response to administration to the subject of an inhibitor of one or more inhibitory checkpoint molecules comprising
  • an antibody-drug conjugate comprising any anti-M(H)DM2/4 antibody or fragment described herein, bound to a cytotoxic drug, or
  • a pharmaceutical composition comprising a therapeutically effective amount of any anti- M(H)DM2/4 antibody or fragment described herein or an antibody-drug conjugate comprising any anti-M(H)DM2/4 antibody or fragment described herein, bound to a cytotoxic drug.
  • kits for treating cancer in a subject who has experienced an accelerated rate of cancer growth in response to administration to the subject of an inhibitor of one or more inhibitory checkpoint molecules wherein the one or more inhibitory checkpoint molecules are selected from the group consisting of: CTLA-4, PD-1, PD-L1, and PD- L2.
  • provided herein are methods of treating cancer in a subject who has experienced an accelerated rate of cancer growth in response to administration to the subject of an inhibitor of one or more inhibitory checkpoint molecules, wherein the one or more inhibitory checkpoint molecules is PD-1.
  • kits for treating cancer in a subject who has experienced an accelerated rate of cancer growth in response to administration to the subject of an inhibitor of one or more inhibitory checkpoint molecules wherein said method comprises administering to the subject any anti-M(H)DM2/4 antibody or fragment described herein.
  • kits for treating cancer in a subject who has experienced an accelerated rate of cancer growth in response to administration to the subject of an inhibitor of one or more inhibitory checkpoint molecules comprising administering to the subject any anti-M(H)DM2/4 antibody or fragment described herein, wherein the anti-M(H)DM2/4 antibody or fragment is not bound to a cell-penetrating peptide.
  • kits for treating cancer in a subject who has experienced an accelerated rate of cancer growth in response to administration to the subject of an inhibitor of one or more inhibitory checkpoint molecules comprising administering to the subject any pharmaceutical composition comprising a therapeutically effective amount of any anti-M(H)DM2/4 antibody or fragment described herein or an antibody- drug conjugate comprising any anti-M(H)DM2/4 antibody or fragment described herein, bound to a cytotoxic drug, wherein the antibody or fragment is not bound to a cell-penetrating peptide.
  • any method described herein comprising administering an anti-M(H)DM2/4 antibody or fragment which competes for binding to M(H)DM2/4 with a humanized antibody or a fragment that specifically binds to HDM2 or MDM2, said humanized antibody or fragment comprising:
  • VH heavy chain variable region
  • VL light chain variable region
  • the method comprises administering an anti-M(H)DM2/4 antibody or fragment which competes for binding to M(H)DM2/4 with a humanized antibody or a fragment that specifically binds to HDM2 or MDM2, said humanized antibody or fragment comprising:
  • VH heavy chain variable region
  • VL light chain variable region
  • any method described herein comprising administering an anti-M(H)DM2/4 antibody or fragment which competes for binding to M(H)DM2/4 with an antibody or a fragment that specifically binds to HDM2 or MDM2, said antibody or fragment comprising:
  • VH heavy chain variable region
  • VL light chain variable region
  • any method described herein comprising administering an anti-M(H)DM2/4 antibody or fragment which competes for binding to M(H)DM2/4 with a chimeric antibody that specifically binds to HDM2 or MDM2, said chimeric antibody comprising:
  • provided herein is any method described herein, wherein the anti- M(H)DM2/4 antibody or fragment is purified.
  • a subject e.g., a human having a cancer
  • said method comprising: (a) identifying a subject having a cancer wherein an antibody or a fragment thereof (e.g., a labeled antibody or fragment) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) binds to the surface of an intact cell of the cancer; and (b) administering to the subject (i) any M(H)DM2/4 (e.g., anti- HDM2) antibody or fragment described herein or an antibody-drug conjugate comprising said antibody or fragment (e.g., an anti-M(H)DM2/4 antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide, wherein the sequence of the peptide consists of SEQ ID NO:1, SEQ ID NO:2,
  • the antibody or fragment thereof in step (b) can be the same or different from the antibody or fragment thereof in step (a).
  • methods that further comprise, before step (b), a step of determining whether the antibody or fragment binds to the surface of the intact cell of the cancer e.g., using FACS or cell-based ELISA analysis) (using any anti-M(H)DM2/4 antibody described herein).
  • methods that further comprise, before the determining step, the step of obtaining intact cells of the cancer e.g., by biopsy of the cancerous tumor in the subject, or by obtaining a blood sample with circulating cancer cells from the subject).
  • the method comprises administering to the subject an antibody-drug conjugate comprising the antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 bound to a cytotoxic drug, wherein said antibody or fragment is not bound to a cell-penetrating peptide.
  • a subject e.g., a human
  • methods for selecting a subject having a cancer for treatment comprising: (a) selecting a subject having a cancer for treatment by: (i) obtaining an intact cancer cell from the subject (e.g., by biopsy of the cancerous tumor in the subject, or by obtaining a blood sample with circulating cancer cells from the subject), and (ii) determining whether an antibody or a fragment thereof (e.g., a labeled antibody or fragment) that specifically binds to M(H)DM2/4 (e.g., an antibody or fragment that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, such as any anti-M(H)DM2/4 antibody or fragment described herein) binds to the surface of the intact cancer cell obtained from the subject (e.g., using FACS or cell-based ELISA analysis), and (b) if the binding is detected in step (a), administering to the subject said antibody or fragment,
  • an antibody or a fragment thereof e.
  • a subject for treatment and treating cancer in the subject comprising: (a) selecting a subject having a cancer for treatment by: (i) obtaining an intact cancer cell from the subject (e.g., by biopsy of the cancerous tumor in the subject, or by obtaining a blood sample with circulating cancer cells from the subject), and (ii) determining whether an antibody or a fragment thereof that specifically binds to HDM2 (e.g., an antibody or fragment that specifically binds to an extracellularly accessible epitope of HDM2, such as any anti-M(H)DM2/4 antibody or fragment described herein) binds to the surface of the intact cancer cell obtained from the subject (e.g., using FACS or cell-based ELISA analysis), and (b) if the binding is detected in step (a), administering to the subject said antibody or fragment, wherein said antibody or fragment is not bound to a cell-penetrating peptide.
  • HDM2 e.g., an antibody or fragment that specifically binds to an extracellularly accessible epitop
  • kits for treating cancer in a subject who has experienced an accelerated rate of cancer growth in response to administration to the subject of an inhibitor of one or more inhibitory checkpoint molecules comprising:
  • the method further comprises before step (b), a step of determining whether any antibody or fragment thereof described herein that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 binds to the surface of intact cells of the cancer.
  • step (a) further comprises selecting a subject who has a gene amplification of
  • step (a) further comprises selecting a subject who has an increased protein expression of M(H)DM2/4 in the cells of the cancer relative to the level of protein expression of M(H)DM2/4 in normal cells. In certain embodiments of the methods, step (a) further comprises selecting a subject who has a cancer wherein there is an increased binding of an antibody or a fragment thereof that specifically binds to an
  • extracellularly accessible epitope of M(H)DM2/4 to the surface of intact cells of the cancer relative to the binding of the antibody or fragment thereof to the surface of intact normal cells.
  • kits for treating cancer in a subject who is a hyper-progressor in response to administration of an inhibitor of one or more inhibitory checkpoint molecules comprising: (a) identifying a subject, wherein the subject has (i) a cancer wherein an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 binds to the surface of intact cells of the cancer, and (ii) a gene amplification of M(H)DM2/4, or an increased protein expression of M(H)DM2/4 in the cells of the cancer relative to the level of protein expression of M(H)DM2/4 in normal cells; and (b) administering to the subject: (i) an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, or an antibody-drug conjugate comprising the antibody or fragment bound to a cytotoxic drug, wherein said antibody or fragment is not bound to a cell-penetrating
  • the method further comprises before step (b) a step of determining whether the antibody or fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 binds to the surface of intact cells of the cancer.
  • step (a) further comprises selecting a subject who has a gene amplification of M(H)DM2/4.
  • step (a) further comprises selecting a subject who has an increased binding of an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 to the surface of intact cells of the cancer relative to its binding to the surface of intact normal cells.
  • the cancer treated in accordance with the methods described herein is a type of cancer that is known to metastasize.
  • the cancer treated in accordance with the methods described herein is an advanced stage cancer.
  • the cancer treated in accordance with the methods described herein is an early stage cancer.
  • the cancer treated in accordance with the methods described herein is a metastatic cancer.
  • the cancer being treated can be a solid cancer or a non- solid cancer (e.g., leukemia or lymphoma).
  • the cancer treated in accordance with the methods described herein is a cervical cancer, an endometrial cancer, an ovarian cancer, a pancreatic cancer, a melanoma (e.g., a uveal melanoma), a breast cancer, a triple negative breast cancer, a colorectal cancer (e.g. a colon cancer), a bladder cancer, an astrocytic neoplasm, a glioblastoma, a pediatric Rhabdomyosarcoma, or a lung cancer (e.g., a non-small cell lung carcinoma).
  • a melanoma e.g., a uveal melanoma
  • breast cancer e.g., a triple negative breast cancer
  • a colorectal cancer e.g. a colon cancer
  • a bladder cancer e.g. a bladder cancer
  • an astrocytic neoplasm e.g., a glioblastom
  • the cancer treated in accordance with the methods described herein is a melanoma, a pancreatic cancer, a breast cancer, or an ovarian cancer.
  • the cancer treated in accordance with the methods described herein is a lung cancer.
  • the cancer treated in accordance with the methods described herein is a colorectal cancer.
  • the cancer treated in accordance with the methods described herein is a colon cancer.
  • the cancer treated in accordance with the methods described herein is a melanoma.
  • the cancer treated in accordance with the methods described herein is a pancreatic cancer.
  • the cancer treated in accordance with the methods described herein is a breast cancer.
  • the cancer treated in accordance with the methods described herein is an ovarian cancer.
  • the subject treated in accordance with the methods described herein can be a human or a non-human animal (such as a mammal). In a preferred embodiment, the subject is a human.
  • the anti-M(H)DM2/4 antibodies or fragments described herein are administered intravenously, intraperitoneally, intramuscularly, subcutaneously, or intratumorally. In other embodiments, the anti-M(H)DM2/4 antibodies or fragments described herein are administered orally.
  • the subject being treated in accordance with the methods described herein is further administered an additional anti-cancer therapy that is different from said antibody or fragment or antibody-drug conjugate (e.g., vaccine, targeted therapy, chemotherapy, radiotherapy, surgery, or immunotherapy).
  • the additional therapy is a vaccine. In one embodiment, the additional therapy is a targeted therapy.
  • the additional therapy is a chemotherapy (e.g., gemcitabine, paclitaxel, nab- paclitaxel, or a combination of gemcitabine and nab-paclitaxel).
  • the additional therapy is an immunotherapy.
  • the additional therapy is a radiotherapy.
  • the additional therapy is a surgery (e.g., to remove part or all of the cancerous tumor being treated).
  • the additional therapy is an inhibitor of the function of one or more checkpoint inhibitory molecules (e.g., an inhibitor, such as an inhibitory antibody to, one or more of: CTLA-4, PD-1, PD-L1, PD-L2, TIM-3, OX40, and LAG-3).
  • the additional therapy is not a cell cycle inhibitor.
  • the subject treated using the methods described herein is not administered a cell cycle inhibitor during the course of treatment with said antibody or fragment.
  • the anti-M(H)DM2/4 antibodies or fragments described herein are administered alone, without any additional anti-cancer therapy (e.g., the subject treated using the methods described herein is not administered an additional anti-cancer therapy during the course of treatment with said antibody or fragment).
  • the subject being treated in accordance with the methods described herein is further administered an additional cancer therapy that is different from an anti-M(H)DM2/4 antibody or fragment or antibody-drug conjugate, wherein the additional cancer therapy is an inhibitor of one or more of: EGFR, KRAS, STK11, ALK, BRAF, ERBB2, RET, ROS1, B2M, HLA, POLE, IGF-1, ERK/MAPK, PI3K/AKT, TGF-B, DNMT3A, IFN ⁇ , JAK1/JAK2/JAK3, CD274, PTEN, ART, AND CDK.
  • the additional cancer therapy is an inhibitor of one or more of: EGFR, KRAS, STK11, ALK, BRAF, ERBB2, RET, ROS1, B2M, HLA, POLE, IGF-1, ERK/MAPK, PI3K/AKT, TGF-B, DNMT3A, IFN ⁇ , JAK1/JAK2/JAK3, CD274, P
  • the subject being treated in accordance with the methods described herein is further administered an additional cancer therapy that is different from an anti-M(H)DM2/4 antibody or fragment or antibody-drug conjugate, wherein the additional cancer therapy is a peptide inhibitor of p53-M(H)DM2/4 interaction.or a small molecule inhibitor of p53-M(H)DM2/4 interaction.
  • the subject being treated in accordance with the methods described herein is further administered chemotherapy, wherein the chemotherapy is cisplatin.
  • the subject being treated in accordance with the methods described herein is further administered chemotherapy, wherein the chemotherapy is 5-FU.
  • the subject being treated in accordance with the methods described herein is further administered chemotherapy, wherein the chemotherapy is paclitaxel.
  • the subject being treated in accordance with the methods described herein is further administered chemotherapy, wherein the chemotherapy is paclitaxel formulated as albumin-bound particles (e.g., ABRAXANE ® ).
  • the subject being treated in accordance with the methods described herein is further administered chemotherapy, wherein the chemotherapy is gemcitabine (e.g., where the cancer being treated is a pancreatic cancer).
  • the subject being treated in accordance with the methods described herein is further administered chemotherapy, wherein the chemotherapy is nab- paclitaxel (e.g., where the cancer being treated is a pancreatic cancer).
  • the cancer is a pancreatic cancer
  • the subject being treated in accordance with the methods described herein is further administered chemotherapy, wherein the chemotherapy is a combination of gemcitabine and nab-paclitaxel.
  • the gemcitabine and/or nab-paclitaxel are administered in doses that are lower than doses used when gemcitabine and/or nab-paclitaxel are administered not in combination with an anti-cancer antibody (such as an anti- M(H)DM2/4 antibody or fragment described herein).
  • an anti-cancer antibody such as an anti- M(H)DM2/4 antibody or fragment described herein.
  • gemcitabine is administered in a dose that is less than 1,500 mg/m 2
  • nab-paclitaxel is administered in a dose that is less than 300 mg/m 2 .
  • gemcitabine is administered in a dose that is equal to or less than 1,000 mg/m 2 and/or the nab-paclitaxel is administered in a dose that is equal to or less than 125 mg/m 2 . In one embodiment, wherein the subject is human, gemcitabine is administered in a dose that is equal to or less than 500 mg/m 2 and/or the nab-paclitaxel is administered in a dose that is equal to or less than 62.5 mg/m 2 . In certain embodiments, the combination of gemcitabine and nab-paclitaxel is administered with a frequency of every 2 weeks or less.
  • the subject being treated in accordance with the methods described herein is resistant to other cancer therapies (e.g., vaccine, targeted therapy, chemotherapy, radiotherapy, surgery, or immunotherapy).
  • the subject being treated in accordance with the methods described herein is resistant to chemotherapy.
  • the subject being treated in accordance with the methods described herein has a chemotherapy-resistant ovarian cancer.
  • the subject being treated in accordance with the methods described herein is resistant to one or more inhibitor of an inhibitory immune checkpoint molecule.
  • the subject being treated in accordance with the methods described herein is resistant to radiotherapy.
  • the anti-M(H)DM2/4 antibody or fragment used in the methods described herein specifically binds within amino acids 19 to 50 of SEQ ID NO:4. In another specific embodiment, the anti-M(H)DM2/4 antibody used in the methods described herein specifically binds within amino acids 154 to 167 of SEQ ID NO:4. In yet another specific embodiment, the anti-M(H)DM2/4 antibody used in the methods described herein specifically binds within amino acids 1 to 60 of SEQ ID NO:4. In yet another specific embodiment, the anti- M(H)DM2/4 antibody used in the methods described herein specifically binds within amino acids 1 to 100 of SEQ ID NO:4.
  • the anti-M(H)DM2/4 antibody used in the methods described herein specifically binds within amino acids 1 to 109 of SEQ ID NO:4. In another specific embodiment, the anti-M(H)DM2/4 antibody used in the methods described herein specifically binds within amino acids 26 to 60 of SEQ ID NO: 4. In one specific embodiment, the anti-M(H)DM2/4 antibody used in the methods described herein specifically binds within the terminal 60 amino acids at the C-terminus of the HDM2 on the plasma membrane of cancer cells. In another specific embodiment, the anti-M(H)DM2/4 antibody used in the methods described herein specifically binds within the terminal 100 amino acids at the C-terminus of the HDM2 on the plasma membrane of cancer cells. In one specific embodiment, the anti-M(H)DM2/4 antibody used in the methods described herein specifically binds within amino acids 101 to 200 of SEQ ID NO:4.
  • the anti-M(H)DM2/4 antibody or fragment used in the methods described herein competes for binding to M(H)DM2/4 with mouse anti-HDM2 antibody OP145 (which is described herein, see, e.g., Table 10).
  • the anti- M(H)DM2/4 antibody or fragment used in the methods described herein competes for binding to M(H)DM2/4 with mouse anti-HDM2 antibody 965 (SMP14) (which is described herein, see, e.g., Tables 3 and 10).
  • the anti- M(H)DM2/4 antibody or fragment used in the methods described herein competes for binding to M(H)DM2/4 with rabbit anti-HDM2 antibody sc-813 (N-20) (which is described herein, see, e.g., Table 10).
  • the anti- M(H)DM2/4 antibody or fragment used in the methods described herein competes for binding to M(H)DM2/4 with rabbit anti-HDM2 antibody sc-812 (C-18) (which is described herein, see, e.g., Table 10).
  • the anti- M(H)DM2/4 antibody or fragment used in the methods described herein competes for binding to M(H)DM2/4 with mouse anti-HDM2 antibody M01, clone 1A7 (which is described herein, see, e.g., Table 3).
  • kits for treating cancer or preventing metastases in a subject in need thereof comprising administering to the subject any anti-M(H)DM2/4 antibody described herein (such as an antibody that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 on the surface of cells of said cancer), wherein the antibody comprises a human IgG Fc region that mediates complement-dependent cytotoxicity (CDC) and/or antibody-dependent cell-mediated cytotoxicity (ADCC).
  • CDC complement-dependent cytotoxicity
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the extracellular region of HDM2 targeted by the anti-HDM2 antibodies or fragments used herein is within one of the following amino acid regions of HDM2: amino acids of SEQ ID NO: 1 (which are amino acids 1 to 15 of SEQ ID NO:4), amino acids of SEQ ID NO: 2 (which are amino acids 15 to 25 of SEQ ID NO:4), amino acids of SEQ ID NO: 3 (which are amino acids 475 to 491 of SEQ ID NO:4), amino acids 19 to 50 of SEQ ID NO: 4, amino acids 50 to 60 of SEQ ID NO: 4, amino acids 100 to 110 of SEQ ID NO: 4, amino acids 154 to 167 of SEQ ID NO: 4, amino acids 1 to 60 of SEQ ID NO: 4, or the terminal 60 amino acids at the C- terminus of the HDM2 on the plasma membrane of the cancer cells.
  • the cancer is a leukemia, a lung cancer, a colon cancer, a melanoma, a pancreatic cancer,
  • kits for diagnosing cancer in a subject comprising: (a) detecting whether an antibody or a fragment thereof (e.g., a labeled antibody or fragment) that specifically binds to M(H)DM2/4 (e.g., HDM2) binds to the surface of intact cells of the subject, wherein the antibody or fragment is any anti- M(H)DM2/4 antibody or fragment described herein (in a preferred example, wherein the antibody or fragment is any anti- M(H)DM2/4 antibody or fragment that specifically binds to a peptide of SEQ ID NO”1, SED ID NO:2, or SEQ ID NO:3); and (b) diagnosing the subject with cancer if binding is detected in step (a).
  • an antibody or a fragment thereof e.g., a labeled antibody or fragment
  • M(H)DM2/4 e.g., HDM2
  • the method of diagnosing is an ex vivo method. In one embodiment, the method of diagnosing further comprises, before step (a), obtaining intact cells from the subject. In one embodiment, the method of diagnosing comprises administering the antibody or fragment to the subject before the detecting in step (a), and wherein the detecting is performed by in vivo imaging of the subject.
  • kits for diagnosing a hyper-progressive disease in a subject who has cancer comprising: (a) determininig whether a gene amplification of M(H)DM2/4 is present in the cells of the cancer of the subject; (b) determining whether an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 binds to the surface of intact cells of the subject; and (c) diagnosing the subject with the hyper-progressive disease if the gene amplification is determined to be present in step (a) and binding is detected in step (b).
  • provided herein are methods of diagnosing a hyper-progressive disease in a subject who has cancer, said methods are ex vivo methods. In certrain aspects, provided herein are methods of diagnosing a hyper-progressive disease in a subject who has cancer, said methods further comprising obtaining intact cells from the subject before step (b). In certrain aspects, provided herein are methods of diagnosing a hyper-progressive disease in a subject who has cancer, said methods comprising administering the antibody or fragment to the subject before the detecting in step (b), and wherein the detecting is performed by in vivo imaging of the subject.
  • certrain aspects provided herein are methods of diagnosing a hyper-progressive disease in a subject who has cancer, wherein, in step (b), the antibody or fragment is labeled.
  • step (b) the antibody or fragment is labeled.
  • certrain aspects provided herein are methods of diagnosing a hyper-progressive disease in a subject who has cancer, wherein any anti-M(H)DM2/4 antibody or fragment thereof described herein is used.
  • the subject is a human.
  • vaccine compositions comprising: (i) an
  • MCNTNMSVPTDGAVT SEQ ID NO:1
  • TTSQIPASEQE SEQ ID NO:2
  • CPVCRQPIQMIVLTYFP SEQ ID NO:3
  • a polynucleotide encoding the peptide and (ii) a pharmaceutically acceptable carrier.
  • the peptide in the vaccine compositions is purified.
  • vaccine compositions further comprise an adjuvant.
  • provided herein are methods of vaccinating a subject at risk for developing cancer or a subject who has been dignosed with cancer by administering to the subject the vaccine composition described herein.
  • HDM2 refers to the human E3 ubiquitin-protein ligase of UniProt Accession Number Q00987 (SEQ ID NO:4) (i.e., full-length HDM2 protein) or a protein product of any splice variant of the full-length HDM2 protein known in the art or described herein.
  • the amino acid sequences of exemplary splice variants of the full-length HDM2 protein are shown as SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO: 15, SEQ ID NO: 16 and SEQ ID NO: 17.
  • MDM2 refers to the mouse E3 ubiquitin-protein ligase of UniProt Accession Number P23804 (SEQ ID NO:5) (i.e., full-length MDM2 protein) or a protein product of any splice variant of the full-length MDM2 protein known in the art or described herein.
  • M(H)DM2 refers to HDM2, MDM2, or an E3 ubiquitin- protein ligase from species other than human and mouse that is a homolog of HDM2 or MDM2.
  • HDM4 refers to the human protein of UniProt Accession Number O15151 (SEQ ID NO:6) (i.e., full-length HDM4 protein) or a protein product of any splice variant of the full-length HDM4 protein known in the art or described herein.
  • MDM4 refers to the mouse protein of UniProt Accession Number O35618 (i.e., full-length MDM4 protein) or a protein product of any splice variant of the full-length MDM4 protein known in the art or described herein.
  • the amino acid sequence of an exemplary splice variant of the full-length MDM4 protein is shown as SEQ ID NO:6.
  • Other splice variants of the full length MDM4 protein known in the art include, without limitation MDM4-S, MDM4-A, MDM4-G, MDM4-XALT1/XALT2 and MDM4-211.
  • M(H)DM4 refers to HDM4 (also called HDMX), MDM4 (also called MDMX), or a protein from a species other than human and mouse that is a homolog of HDM4 or MDM4.
  • the term“M(H)DM2/4” refers to HDM2, MDM2, HDM4, MDM4, or a protein from a species other than human and mouse that is a homolog of HDM2, MDM2, HDM4 or MDM4.
  • the term“about,” when used to modify a numeric value indicate that deviations of up to 10% above and below the numeric value remain within the intended meaning of the recited value.
  • the term“intact” with reference to a cell refers to a cell that is viable or fixed but not permeabilized.
  • the term“extracellularly accessible” with reference to an epitope of M(H)DM2/4 refers to an epitope of M(H)DM2/4 that, when the M(H)DM2/4 is expressed by an intact cell, the epitope is available for binding with an extracellular antibody (without a need for intracellular transport of the antibody).
  • An antibody or a fragment thereof can be determined to bind to an extracellularly accessible epitope of M(H)DM2/4, when the antibody, when extracellular, binds to M(H)DM2/4 expressed by an intact cell.
  • the M(H)DM2/4 expressed by an intact cell in the foregoing definition is the form of the M(H)DM2/4 retained on the plasma membrane (i.e., as a transmembrane protein).
  • Membrane-bound M(H)DM2/4 or“plasma membrane-bound M(H)DM2/4” refers to a transmembrane M(H)DM2/4 protein variant.
  • VL refers to the light chain variable region of an antibody.
  • VK refers to the kappa isotype of the light chain variable region of an antibody.
  • VH refers to the heavy chain variable region of an antibody.
  • hypo-progression refers to an accelerated rate of tumor or cancer growth therapy after administration of a therapy compared to prior to the administration of the therapy.
  • hyper-progressor refers to a subject who exhibits an accelerated rate of tumor or cancer growth after administration of a therapy compared to prior to the administration of the therapy.
  • the term“percent (%) amino acid sequence identity” or“percent 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. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are known in the art, for instance, using publicly available computer software such as BLASTp, BLAST-2, ALIGN (e.g., ALIGN-2) or Megalign (DNASTAR) software.
  • Figures 1A-D show that monoclonal antibodies NMC-103, NMC-204 and NMC-303 specifically bound to NMC-P1 (SEQ ID NO:1), NMC-P2 (SEQ ID NO:2) and NMC-P3 (SEQ ID NO:3) peptide antigens, respectively, in peptide-ELISA experiments.
  • B NMC-204 bound to NMC-P2 peptide while NMC-103 did not bind to NMC-P2.
  • C NMC-303 bound to NMC-P3 peptide while NMC-204 did not bind to NMC-P3.
  • Figure 2 shows that monoclonal antibodies NMC-103, NMC-204, NMC-303 bound to HDM2 recombinant protein.
  • Figures 3A-B show that monoclonal antibody NMC-103 bound to an extracellularly accessible epitope of HDM2 on intact human (A) and murine (B) cancer cells.
  • FIGs 4A-B show that monoclonal antibody NMC-204 bound to an extracellularly accessible epitope of HDM2 on intact human (A) and murine (B) cancer cells.
  • FIGs 5A-B show that monoclonal antibody NMC-303 bound to an extracellularly accessible epitope of HDM2 on intact human (A) and murine (B) cancer cells.
  • Figure 6 shows that monoclonal antibody NMC-204 bound to an extracellularly accessible epitope of HDM2 on intact human cancer cells but did not bind to intact normal human peripheral blood mononuclear cells.
  • Figures 7A-B depict the binding curves of the binding of monoclonal antibodies NMC-103 (A) and NMC-204 (B) to intact MIA PaCa-2 cells.
  • Figures 8A-C show that the binding of monoclonal antibody NMC-103 to its extracellularly accessible epitope of HDM2 on the plasma membrane of intact human pancreatic cancer MIA PaCa-2 cells was competed by the NMC-P1 peptide (A), the binding of monoclonal antibody NMC-204 to its extracellularly accessible epitope of HDM2 on the plasma membrane of intact human pancreatic cancer MIA PaCa-2 cells was competed by the NMC-P2 peptide (B), and the binding of monoclonal antibody NMC-303 to its extracellularly accessible epitope of HDM2 on the plasma membrane of intact human pancreatic cancer MIA PaCa-2 cells was competed by the NMC-P3 peptide (C).
  • FIG. 9 shows that the binding of monoclonal antibody NMC-103 to its
  • HDM2 extracellularly accessible epitope of HDM2 on the plasma membrane of intact human pancreatic cancer MIA PaCa-2 cells was competed by the full-length recombinant HDM2 protein.
  • Figure 10 shows the cell-ELISA binding results of monoclonal antibody NMC-103, monoclonal antibody NMC-204, an antibody against E-cadherin, and an antibody against Cytochrome-C to intact human pancreatic MiaPaCa-2 cells.
  • Figures 11A-C present flow cytometry data on % cells stained with monoclonal antibodies NMC-103 (A), NMC-204 (B), and anti-Na+/K+ ATPase a-1 (C), respectively.
  • Figures 12A-D show that monoclonal antibodies NMC-103 (A and D) and NMC-204 (B and D), but not an anti-Cytochrome-C antibody (C and D), inhibited cell proliferation of intact human pancreatic MIAPaCa-2 cells.
  • Figures 13A-C show that monoclonal antibody NMC-103 (B and C) in the presence of normal human serum induces complement-mediated cytotoxicity against human pancreatic MIAPaCa-2 cells as compared with cells treated with normal human serum in the absence of any antibody (A).
  • Figures 14A-B show the lack of binding of many commercially available monoclonal antibodies to either NMC-P1 (A) or NMC-P2 (B).
  • FIGs 15A-B show that an anti-HDM2 antibody termed“MDM2 monoclonal antibody (M01), clone 1A7” (Abnova, Cat. No. H00004193-M01) reacted with intact cancer cells (A), but an anti-HDM2 antibody termed“Anti-MDM2 (Ab-4) Mouse mAb (2A9C1.18)” (EMD Millipore, Cat. No. OP144) and an anti-HDM2 antibody termed“Anti-MDM2 (Ab-1) Mouse mAb (IF2)” (EMD Millipore, Cat. No. OP46) did not react with intact cancer cells (B).
  • Figure 16 depicts the effect of monoclonal antibody NMC-204 on tumor volume of the LL/2 syngeneic mouse model of lung cancer.
  • Figures 17A-B depict the effect of monoclonal antibody NMC-103 on tumor volume (A) and tumor cell proliferation (B) of the MC-38 syngeneic mouse model of colon cancer.
  • Figures 18A-B depict the effect of monoclonal antibody NMC-204 on tumor volume (A) and tumor cell proliferation (B) of the MC-38 syngeneic mouse model of colon cancer.
  • Figure 19 depicts the effect of monoclonal antibody NMC-103 alone (2 mg/kg), a combination of low dose Gemcitabine (25 mg/kg) and nab-Paclitaxel (5 mg/kg), a combination of low dose Gemcitabine (25 mg/kg), nab-Paclitaxel (5 mg/kg) and NMC-103 (2mg/kg), and isotype control mouse IgG1 (2 mg/kg), respectively, on tumor volume of the Panc-2 syngeneic mouse model of pancreatic cancer. Treatment started when tumors in mice reached
  • Figure 20 depicts the DNA sequence and protein sequence of the heavy chain variable region and the light chain variable region, respectively, of monoclonal antibody NMC- 103.
  • Figure 21 depicts the DNA sequence and protein sequence of the heavy chain variable region and the light chain variable region, respectively, of monoclonal antibody NMC- 204.
  • Figure 22 depicts the DNA sequence and protein sequence of the heavy chain variable region and the light chain variable region, respectively, of monoclonal antibody NMC- 303.
  • the leader sequence before the DNA and protein sequences of the heavy chain variable region and the light chain variable region is in bold (but not underlined).
  • Figure 23 shows the tumor size of mice treated with the anti-HDM2 antibody termed “MDM2 monoclonal antibody (M01), clone 1A7” (Abnova, Cat. No. H00004193-M01), the tumor size of mice treated with NMC-103, the tumor size of mice treated with NMC-204, and the tumor size of mice treated with isotype control.
  • MDM2 monoclonal antibody M01
  • clone 1A7 Abnova, Cat. No. H00004193-M01
  • FIGs 24A-F Anti-HDM2-specific antibodies stain the surface of cancer cells but not normal cells. Intact cells released either with either EDTA or Trypsin were blocked with 5% human serum albumin. Cells were then incubated with either polyclonal N-20 M(H)DM2- specific antibody (sc-813, N-20, rabbit IgG; from Santa Cruz;“N-20”) or monoclonal
  • M(H)DM2-specific OP145 antibody (OP145, mouse IgG1; from Calbiochem;“OP145”) for 90 min. on ice.
  • Another set of cells prepared under the same conditions were incubated with the same antibodies that were pre-incubated with their corresponding blocking peptides before incubation with cells. Following primary antibody incubation, cells were washed 3 times with ice-cold PBS followed by FITC-secondary antibody incubation for 60 min. Cells were then washed 3 times with PBS and were subjected to FACS analyzer.
  • Human melanoma cells 24A, 24B, and 24C
  • primary human ovarian cancer cells 24D and 24E
  • normal mouse splenocytes 24F.
  • Figure 24A area under curve #1 represents cells incubated with goat anti- rabbit secondary antibody only; area under curve #2 represents cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #3 represents cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody.
  • Figure 24B area under curve #1 represents cells incubated with goat anti-mouse secondary antibody only; area under curve #2 represents cells incubated with anti-HDM2 monoclonal antibody OP145 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #3 represents cells incubated with anti-HDM2 monoclonal antibody OP145 followed by goat anti-mouse secondary antibody.
  • Figure 24C area under curve #1 represents cells incubated with goat anti- rabbit secondary antibody only; area under curve #2 represents trypsin-released cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody; area under curve #3 represents EDTA-released cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #4 represents EDTA-released cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody.
  • Figures 24D & E area under curve #1 represents cells incubated with goat anti-rabbit secondary antibody only; area under curve #2 represents cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #3 represents cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti- rabbit secondary antibody.
  • Figure 24F area under curve #1 represents cells incubated with goat anti-rabbit secondary antibody only; area under curve #2 represents trypsin-released cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody; area under curve #3 represents EDTA-released cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #4 represents EDTA-released cells incubated with anti- HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody.
  • FIGS 25A-C Human pancreatic or ovarian cancer cells and normal human fibroblasts were treated with normal human serum (NHS) alone, NHS + anti-HDM2 OP145 monoclonal antibody (mouse IgG1, from Calbiochem,“OP145”) or control antibody (NHS + Cytochrome C). Extensive cell death as evident by Propidium Iodide (PI) staining was observed when cancer cells were treated with the OP145 antibody (see panels b and e) in the presence of NHS, whereas the same antibody had no effect on the viability of normal human fibroblasts (see panel g).
  • NHS normal human serum
  • OP145 normal human serum
  • PI Propidium Iodide
  • Control antibody to Cytochrome C shows no cytotoxicity (see panel c) beyond that observed in untreated cells (see panel a). Lack of cell death is manifested by no or little PI staining in panels a, c, d, f and g.
  • the cell death marker PI was visualized using Olympus FluoView FV1000 Confocal Laser Scanning Biological Microscope built on the Olympus IX81 Inverted Microscope.
  • M(H)DM2-specific antibodies are cytotoxic to pancreatic cancer MiaPaCa-2 cells in the presence of NHS. Quantitative representations of M(H)DM2-specific antibody-dependent complement cytotoxicity against human pancreatic cancer cells. Cells treated with anti- M(H)DM2 (C-18) antibody in the presence of NHS demonstrated cytotoxicity over 15-30 min. post-treatment, whereas anti-HDM2 OP46 shows no cytotoxic effect beyond that observed when cells were treated with control anti-Cytochrome C antibody or when cells were treated with NHS in the absence of anti-M(H)DM2 antibodies.
  • Figure 26 shows the tumor size of mice treated with anti-HDM2 antibody OP145 and the tumor size of mice treated with PBS control (in Panc02 syngeneic mouse model of pancreatic cancer).
  • the x axis shows days after tumor cell injection into the mice.
  • the y axis shows tumor volume in mm 3 .
  • the arrow shows the day on which the treatment was started.
  • Figure 28 depicts a Kaplan Meier survival analysis demonstrating survival benefit in mice that received NMC-103 alone or in combination with chemotherapy when compared to chemotherapy alone or control antibody under the experimental conditions described in Figure 27.
  • FIG 29 shows that mice previously treated with NMC-103 as described in Figure 27, become immune to tumor re-challenging after drug withdrawal.
  • mice that had previously received with a combination of G + nP mice in group C (mice that had been previously treated with NMC-103) and group D (mice that had been previously treated with a combination of NMC-103 + G + nP) were re-challenged by a second round of Panc-2 inoculation (subcutaneous injection of 2x10 6 cells/mouse), on the left dorsal flank. Tumor growth was monitored for 10 days at which point, a tumor of 90 mm 3 was measured in the mice from group B. No tumor was observed in mice from the two groups that had previously received NMC-103 antibody (Groups C and D).
  • Figure 30 depicts the effect of a single dose of monoclonal antibody NMC-103 (10 mg/kg) or isotype control mouse IgG1 (10 mg/kg), when added to a treatment regimen of a combination of low dose Gemcitabine (25 mg/kg) and nab-Paclitaxel (5 mg/kg), in the treatment of large size tumors (i.e. advanced cancers) on tumor volume of the Panc-2 syngeneic mouse model of pancreatic cancer. Mice were treated with pancreatic cancer standard of care
  • mice were then randomly divided in 2 groups that received a single dose of an isotype control mouse IgG1 (10 mg/kg) or NMC-103 (10 mg/kg). As shown in this figure, a single i.p. injection of NMC-103 reduced the tumor size by almost half 6 days post treatment (from 438 mm 3 to 233 mm 3 ).
  • Figure 31 depicts the effect of monoclonal antibody NMC-103 on tumor volume of the MC-38 syngeneic mouse model of colon cancer.
  • mice treated with NMC-103 at 10 mg/kg, 2 times per week for 2 weeks reached an average tumor size of 210 mm 3
  • mice in the group that weretreated with isotype control antibody at 10 mg/kg grew rapidly and reached 1168 mm 3 by day 12.
  • mice treated with NMC-103 at 0.4 mg/kg Figure 17
  • these data support the dose-dependent anti-tumor effect of NMC-103 antibody.
  • FIGs 32A and 32B depict the effect of a chimeric version of monoclonal antibody NMC-303.
  • Isotype class-switching was performed on a mouse NMC-303 to convert it from a mouse IgM to a chimeric IgG1.
  • the mouse Heavy and Light chain variable regions were cloned into a human Ig gamma-1 chain and human Ig kappa chain as constant region.
  • Figure 32A shows that by day 24 post tumor inoculation, mice treated with chimeric version of NMC-303 (10 mg/kg) reached an average tumor size of 726 mm3, while mice treated with control antibody (10 mg/kg) had an average tumor size of 1746 mm3. Furthermore, Figure 32B shows the individual mouse tumor sizes on day 24 post tumor inoculation.
  • Figure 33 shows the cell-based-ELISA results of the binding of humanized monoclonal NMC-H103 (VH4/VK3) and chimeric monoclonal (NMC-C303) antibodies to membrane-bound MDM2 on intact mouse Lewis Lung (LL/2) cancer cells.
  • An antibody to plasma membrane marker E-Cadherin also binds to intact un-permeabilized mouse LL/2 cancer cells, while antibodies to cytoplasmic proteins cytochrome C, cyclin D1 and Bcl-2 do not bind to intact un-permeabilized cells. All primary antibodies were used at 5 mg/mL.
  • Anti-mouse and anti-human secondary HRP conjugated antibodies were used at 1:2000 to 1:4000 range.
  • Figures 34A-B show the cell-based ELISA binding results demonstrating the binding of humanized NMC-H103 (VH4/VK3) and chimeric (NMC-C303) antibodies to a panel of intact, un-permeabilized human (34A) and murine (34B) cancer cells. No binding beyond background staining was shown using anti-human secondary antibody.
  • Figures 35A-B show the cell-based ELISA results demonstrating the binding of humanized monoclonal antibody NMC-H103 (VH4/VK3) to intact, un-permeabilized mouse LL/2 cancer cells, while pre-incubation of humanized monoclonal antibody NMC-H103 (VH4/VK3) (5 mg/mL) with NMC-P1, reduces the binding efficiency of humanized monoclonal antibody NMC-H103 (VH4/VK3) to its target. Pre-incubation of chimeric monoclonal antibody NMC-C303 with NMC-P1 has no effect on its binding to mouse LL/2 cancer cells.
  • Figure 35B shows flow cytometry results on live (7AAD negative) cells showing the binding of humanized monoclonal antibody NMC-H103 (VH4/VK3) (left panel; solid black curve) and chimeric monoclonal antibody NMC-C303 (right panel; solid black curve) to live OVCAR-3 (upper panel) and mouse LL/2 cancer cells (lower panel). Dashed line curves show the reduced binding of humanized monoclonal NMC-H103 (VH4/VK3) and chimeric monoclonal NMC-C303 antibodies to OVCAR-3 and mouse LL/2 cancer cells, when cells are pre-treated with trypsin.
  • Figures 36A-B show results from in vivo experiments utilizing NOD scid gamma mice (NSG: NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ) immune-deficient mice demonstrating that the anti-cancer activity of the humanized monoclonal NMC-H103 (VH4/VK3) and the chimeric monoclonal NMC-C303 antibodies is significantly more effective in presence of normal human peripheral blood monocytes (Hu-PBMCs) than in the absence of Hu-PBMCs.
  • NSG NOD.Cg-Prkdc scid Il2rg tm1Wjl /SzJ
  • Hu-PBMCs (7x10 6 Hu-PBMCs/mouse) were injected in tail vein of mice in group 1 (filled triangle) and group 3 (filled circle), whereas mice in group 2 (filled square) did not receive any Hu-PBMCs.
  • mice in group 1 received intraperitoneal (i.p.) injection of isotype control antibody (10 mg/kg), while mice in groups 2 and 3 received i.p.
  • Hu-PBMCs (7x10 6 Hu-PBMCs/mouse) were injected in tail vein of mice in group 1 (left bar) & 3 (right bar), whereas mice in group 2 (middle bar) did not receive any Hu-PBMCs.
  • Mice in group 1 received a single i.p.injection of isotype control antibody (10mg/kg), while mice in groups 2 and 3 received a single i.p. injection of humanized monoclonal antibody NMC-H103 (VH4/VK3) (10mg/kg).
  • Subcutaneous tumors were measured 4 days post antibody treatment.
  • Figures 37A-C show in vitro cell proliferation MTT assay results of M109 cells treated with anti-PD1 antibody (A), results of the in vivo effect of anti-PD1 antibody on the growth of M109 tumor in NSG mice (B), and flow cytometry results on freshly excised tumors from mice treated with isotype control or anti-PD1 antibodies.
  • Figure 37A shows MTT cell proliferation assay results of M109 cells treated with 75 mg/mL (middle bar) or 100 mg/mL (right bar) of anti-PD1 antibody for 48 hours. As control, M109 cell proliferation was also measured in presence of isotype control antibody at 100 mg/mL (left bar).
  • Figure 37B depicts the effect of anti-PD1 antibody on the growth of M109 tumor in mice.
  • Figures 38A-B show single cycle (A) and multiple cycle (B) sensorgrams data and fitted curves (1:1 binding model) for binding to NMC-P1 of chimeric monoclonal antibody NMC-C103 (VH0/VK0) and humanized monoclonal antibody NMC-H103 variants having VH/ and VK combinations identified in the figure.
  • the sequences of the respective VH and VK of humanized monoclonal antibody NMC-H103 variants are provided in Tables 15-27 and Section 11. 7.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2).
  • the extracellularly accessible epitope is contained within SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3.
  • Antibodies provided herein are described in Section 5.1, below.
  • antibody-drug conjugates comprising an antibody or fragment that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) described herein bound (e.g., covalently) to a cytotoxic drug.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), wherein said antibodies or fragments are not bound to a cytotoxic component.
  • compositions comprising an antibody or fragment that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) described herein.
  • such pharmaceutical compositions comprise a therapeutically effective amount of such antibody or fragment (i.e., an amount that can be used to treat a cancer in a subject, e.g. by achieving one or more anti-tumor effects described herein).
  • nucleic acids encoding the antibodies and antigen-binding fragments described herein.
  • vectors and cells comprising nucleic acids encoding such antibodies or antigen-binding fragments thereof.
  • Cells recombinantly producing the antibodies or antigen-binding fragments thereof described herein are also provided.
  • Chimeric antigen receptors are engineered receptors that provide both antigen binding and immune cell activation functions (Sadelain et al., 2013, Cancer Discovery 3:388- 398).
  • CARs comprising a single-chain variable fragment (scFv) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, such as a scFv comprising the VH and VL of an anti-M(H)DM2/4 antibody described herein, fused via a linker to a transmembrane domain (e.g., of CD3 zeta) fused to an intracellular T cell activation domain such as CD3 zeta intracellular domain, optionally further fused to a co-stimulatory domain (e.g., CD28 intracellular domain).
  • T cells expressing such CARs are also provided.
  • MCNTNMSVPTDGAVT SEQ ID NO:1
  • TTSQIPASEQE SEQ ID NO:2
  • the peptide can be, for example, synthetic or recombinant. In some embodiments, the peptide is purified. In some embodiments, the peptide islabeled with a detectable marker (e.g., a fluorescent marker or an isotope). In some
  • the peptide is tagged (e.g., with a GST, His, Strep, myc, FLAG, or HA tag).
  • a cysteine is added at one of the ends of the peptide(which may allow for linkage to a carrier protein).
  • the peptide islinked to a carrier protein (e.g., linked to Keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA), ovalbumin, thyroglobulin, tetanus toxoid, or diphtheria toxoid).
  • KLH Keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • ovalbumin ovalbumin
  • thyroglobulin thyroglobulin
  • tetanus toxoid or diphtheria toxoid
  • nucleic acids encoding a peptide described herein.
  • vectors and cells comprising a nucleic acid encoding a peptide described herein.
  • Cells recombinantly producing a peptide described herein are also provided.
  • uses of the peptides described herein as immunogens The peptides of SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3 described herein contain extracellularly accessible epitopes of MDM2 and HDM2.
  • an anti-M(H)DM2/4 antibody e.g., an antibody that specifically binds to M(H)DM2/4
  • an animal e.g., a mouse or a rabbit
  • an anti-M(H)DM2/4 antibody suitable for therapeutic use by contacting an anti-M(H)DM2/4 antibody with a peptide described herein under conditions suitable for binding between the antibody and the peptide, and detecting or measuring binding between the antibody and the peptide that occurs, and, if the binding between the antibody and the peptide is detected, using the antibody in the methods of treating cancer described herein.
  • provided herein are methods of identifying an anti-M(H)DM2/4 antibody suitable for diagnostic use by contacting an anti-M(H)DM2/4 antibody with a peptide described herein under conditions suitable for binding between the antibody and the peptide, and detecting or measuring binding between the antibody and the peptide that occurs, and, if the binding between the antibody and the peptide is detected, using the antibody in the methods of diagnosing cancer described herein.
  • an anti-M(H)DM2/4 antibody or fragment described herein in particular, an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (in particular, a region exposed on the plasma membrane surface of cancer cells).
  • the antibody or fragment thereof specifically binds to an extracellularly accessible epitope of HDM2 (in particular, a region exposed on the plasma membrane surface of cancer cells).
  • the antibody or fragment (e.g., for use in the methods described herein) does not bind or only minimally binds to the plasma membrane surface of normal cells of the tissue type from which the cancer in the subject originates.
  • the anti-M(H)DM2/4 antibody or fragment is any chimeric or humanized anti-M(H)DM2 antibody described herein.
  • an anti-M(H)DM2/4 antibody or fragment described herein in particular, an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 (in particular, a region exposed on the plasma membrane surface of cancer cells).
  • an antibody or fragment thereof specifically binds to an extracellularly accessible epitope of HDM2 (in particular, a region exposed on the plasma membrane surface of cancer cells).
  • the antibody or fragment (e.g., for use in the methods described herein) does not bind or only minimally binds to the plasma membrane surface of normal cells of the tissue type from which the cancer in the subject originates.
  • the anti-M(H)DM2/4 antibody or fragment is any chimeric or humanized anti-M(H)DM2 antibody described herein.
  • an antibody or a fragment thereof used herein specifically binds to an extracellularly accessible epitope of HDM2 and/or HDM4 (a region exposed on the plasma membrane surface of cancer cells).
  • an antibody or a fragment thereof used herein specifically binds to an extracellularly accessible epitope of HDM2 (a region exposed on the plasma membrane surface of cancer cells) (optionally, such antibody or fragment that does not bind to HDM4).
  • an antibody or a fragment thereof used herein specifically binds to an extracellularly accessible epitope of HDM4 (a region exposed on the plasma membrane surface of cancer cells) (optionally, such antibody or fragment that does not bind to HDM2).
  • an antibody or a fragment thereof used herein binds to an extracellularly accessible epitope of M(H)DM2/4 (a region exposed on the plasma membrane surface of cancer cells), where the M(H)DM2/4 is a homologue of HDM2 and/or HDM4 expressed in such animal.
  • an antibody or a fragment thereof used herein binds to an extracellularly accessible epitope of M(H)DM2 (a region exposed on the plasma membrane surface of cancer cells), where the M(H)DM2 is a homologue of HDM2 expressed in such animal (optionally, such antibody or fragment does not bind to M(H)DM4).
  • an antibody or a fragment thereof used herein binds to an extracellularly accessible of M(H)DM4 (a region exposed on the plasma membrane surface of cancer cells), where the M(H)DM4 is a homologue of HDM4 expressed in such animal (optionally, such antibody or fragment does not bind to M(H)DM2).
  • HDM2 and antibodies and antibody fragments thereto which shall be understood to be for use in treating a human; it will be clear to one skilled in the art that the description also should be deemed applicable to: (i) HDM4 and antibodies and antibody fragments thereto, and use thereof for treatment of humans (unless indicated otherwise explicitly or by context), and (ii) M(H)DM2/4 and antibodies and antibody fragments thereto, and use thereof for treatment of non-human animals, e.g., mammals (unless indicated otherwise explicitly or by context).
  • the patients or subjects being treated using the methods described herein are human.
  • the anti-HDM2 antibody or a fragment thereof used in accordance with the methods described herein mediates complement-dependent cytotoxicity (CDC), mediates antibody-dependent cell-mediated cytotoxicity (ADCC), and/or is bound to a cytotoxic drug or drugs (e.g., is an antibody-drug conjugate).
  • CDC complement-dependent cytotoxicity
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the invention provides for the use of antibodies that mediate complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC).
  • the anti-HDM2 antibody or a fragment thereof used in accordance with the methods described herein is not bound to a cell-penetrating peptide.
  • Cell penetrating peptides can insert into a cell plasma membrane and transport molecules to which they are attached into the cell.
  • Such cell-penetrating peptides include, without limitation, a membrane resident peptide (MRP), Membrane Transduction Domain of Antennapedia, trans- activating transcriptional activator (TAT), and a Penetratin peptide.
  • the anti-HDM2 antibody or a fragment thereof used in accordance with the methods described herein is not attached to a membrane resident peptide (MRP), Membrane Transduction Domain of Antennapedia, TAT, and/or a Penetratin peptide.
  • MRP membrane resident peptide
  • the anti-HDM2 antibody or a fragment thereof used in accordance with the methods described herein is not attached to any peptide sequence that can insert into the lipid bilayer of the plasma membrane of cells.
  • the anti-HDM2 antibody or a fragment thereof used in accordance with the methods described herein is not attached to an MRP.
  • the anti- HDM2 antibody or a fragment thereof used in accordance with the methods described herein is not attached to a Penetratin peptide.
  • HDM-2 targeting antibodies alone are selectively cytotoxic to cancer cells.
  • extracellularly accessible epitopes of HDM2 are appropriate therapeutic targets for anti-HDM2 antibodies, and that cancer cells expressing HDM2 on their surface can be successfully targeted and destroyed with antibodies to such extracellular regions of HDM2.
  • select HDM2-specific antibodies can bind to the extracellularly accessible sequences of HDM2 on the surface membrane of intact cancer cells, while exhibiting minimal binding to the surface membrane of normal human blood mononuclear cells.
  • HDM2- specific antibodies can inhibit the growth of cancer cells in vitro and in vivo, strongly suggesting that they can be used as therapeutic agents in vivo.
  • data presented in the examples show that such HDM2-specific antibodies can have a synergistic anti-tumor effect when combined with chemotherapeutic drugs.
  • data presented in the examples show that certain chimeric and humanized HDM2-specific antibodies described herein have anti-tumor effect.
  • the data presented in the examples show that subjects whose tumors undergo hyper-progression in response to treatment with an inhibitor of an inhibitory immune checkpoint molecule have an increased level of plasma-membrane-bound M(H)DM2 that could be targeted with HDM2-specific antibodies that bind to the extracellularly accessible epitopes of M(H)DM2 (e.g., certain chimeric and humanized HDM2-specific antibodies described herein).
  • HDM2-specific antibodies bind to the extracellularly accessible epitopes of M(H)DM2 (e.g., certain chimeric and humanized HDM2-specific antibodies described herein).
  • antibodies or antigen-binding fragments thereof that (immuno) specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) (a region exposed on the plasma membrane surface of cells).
  • M(H)DM2/4 e.g., HDM2
  • Specifically bind[s]/binding does not exclude cross-reactivity of the antibody or antigen-binding fragment; thus, for example, antibodies or antigen-binding fragments thereof that (immuno) specifically bind to an extracellularly accessible epitope of HDM2 exposed on the plasma membrane surface of cells may also specifically bind to (cross-react with) MDM2.
  • anti-M(H)DM2/4 antibodies and fragments thereof that (immuno) specifically bind to an extracellularly accessible epitope of M(H)DM2/4 and that have an anti-tumor effect (e.g., inhibit tumor growth in vivo).
  • an antibody or an antigen-binding fragment thereof specifically binds an epitope of M(H)DM2/4 that is extracellularly accessible on cancer cells but not on non-cancer cells (e.g., non-cancerous cells of the same organ type or tissue type as the cancer cells).
  • an antibody or an antigen-binding fragment thereof specifically binds an epitope of M(H)DM2/4, exposure or accessibility of which on the plasma membrane surface of cancer cells is increased relative to its exposure or accessibility on the plasma membrane surface of non-cancer cells (e.g., non-cancerous cells of the organ or tissues of the host).
  • antibodies or antigen-binding fragments thereof that (immuno) specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2), which are not bound to a cell-penetrating peptide (e.g., a membrane resident peptide).
  • antibodies or antigen-binding fragments thereof that (immuno) specifically bind to M(H)DM2/4 (e.g., HDM2), in particular to an extracellularly accessible epitope of M(H)DM2/4, wherein the antibody or fragment specifically binds to a peptide the sequence of which peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3).
  • MCNTNMSVPTDGAVT SEQ ID NO:1
  • TTSQIPASEQE SEQ ID NO:2
  • CPVCRQPIQMIVLTYFP SEQ ID NO:3
  • immuno specifically bind to M(H)DM2/4 (e.g., HDM2), in particular to an extracellularly accessible epitope of M(H)DM2/4
  • the antibody or fragment specifically binds to a peptide the sequence of which peptide consists of MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3); and wherein such antibodies or fragments have an anti-tumor effect in vivo, and/or wherein such antibodies or fragments are not bound to a cell-penetrating peptide.
  • anti- M(H)DM2/4 antibodies and fragments having heavy chain variable regions and/or light chain variable regions described herein see, e.g., having sequences of heavy chain variable regions and/or light chain variable regions of antibodies NMC-103, NMC-204 and NMC-303 provided herein, see, e.g., Section 8 and Figures 20-22.
  • anti-M(H)DM2/4 antibodies and fragments having one or more complementarity determining regions (CDRs) described herein see, e.g., CDRs provided in Tables 4-9 and Figures 20-22).
  • chimeric and humanized anti- M(H)DM2/4 antibodies and fragments having heavy chain variable regions and/or light chain variable regions described herein e.g., having sequences of heavy chain variable regions and/or light chain variable regions of antibodies NMC-C303, NMC-C103 and variants of NMC-H103 provided herein, see, e.g., Sections 9 and 11, and Tables 11-27.
  • anti-M(H)DM2/4 antibodies and fragments having one or more complementarity determining regions (CDRs) described herein see, e.g., CDRs provided in Tables 11-27
  • CDRs complementarity determining regions
  • chimeric and humanized anti-M(H)DM2/4 antibodies and fragments having one or more framework regions (FR) described herein see, e.g., heavy chain framework regions (HFR) and light chain framework regions (LFR) provided in Tables 11-27).
  • CDRs are defined in various ways in the art, including the Kabat, Chothia, AbM, Contact, and IMGT.
  • the CDRs of an antibody can be defined according to the Kabat system, which is based on sequence variability (see, e.g., Kabat EA & Wu TT (1971) Ann NY Acad Sci 190: 382-391; Kabat EA et al., (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No.91- 3242.
  • the VH CDR1 is present at amino acid positions 31 to 35 of the heavy chain; (ii) the VH CDR2 is present at amino acid positions 50 to 68 or 50 to 66 of the heavy chain; and (iii) the VH CDR3 is present at amino acid positions 101 to 105 or 99 to 104 or 99 to 106 of the heavy chain.
  • the VL CDR1 is present at amino acid positions 24 to 39 or 24 to 34 of the light chain;
  • the VH CDR2 is present at amino acid positions 55 to 61 or 50 to 56 of the light chain;
  • the VH CDR3 is present at amino acid positions 94 to 102 or 89 to 97 of the light chain.
  • the actual linear amino acid sequence of the antibody variable domain can contain fewer or additional amino acids due to a shortening or lengthening of a framework region (FR) and/or CDR and, as such, an amino acid’s Kabat number is not necessarily the same as its linear amino acid number.
  • the Kabat CDR positions may vary depending on the antibody, and may be determined according to methods known in the art.
  • the CDRs of the antibodies described herein are determined using the Kabat system.
  • the CDRs of an antibody can be defined according to the Chothia system, which is based on the location of immunoglobulin structural loop regions (see, e.g., Chothia C & Lesk AM, (1987), J Mol Biol 196: 901-917; Al-Lazikani B et al., (1997) J Mol Biol 273: 927-948; Chothia C et al., (1992) J Mol Biol 227: 799-817; Tramontano A et al., (1990) J Mol Biol 215(1): 175-82; and U.S. Patent No.7,709,226).
  • “Chothia CDRs,” and like terms are recognized in the art and refer to antibody CDR sequences as determined according to the method of Chothia and Lesk, 1987, J. Mol. Biol., 196:901-917, which will be referred to herein as the“Chothia CDRs” (see also, e.g., U.S. Patent No.7,709,226 and Martin, A.,“Protein Sequence and Structure Analysis of Antibody Variable Domains,” in Antibody Engineering, Kontermann and Dübel, eds., Chapter 31, pp.422-439, Springer-Verlag, Berlin (2001)).
  • the VH CDR1 is present at amino acid positions 26 to 32 of the heavy chain;
  • the VH CDR2 is present at amino acid positions 52 to 59 or 52 to 57 of the heavy chain; and
  • the VH CDR3 is present at amino acid positions 101 to 105 or 99 to 104 or 99 to 106 of the heavy chain.
  • the VL CDR1 is present at amino acid positions 24 to 39 or 24 to 34 of the light chain;
  • the VL CDR2 is present at amino acid positions 55 to 61 or 50 to 56 of the light chain;
  • the VL CDR3 is present at amino acid positions 94 to 102 or 89 to 97 of the light chain.
  • the Chothia CDR positions may vary depending on the antibody, and may be determined according to methods known in the art.
  • the CDRs of the antibodies described herein are determined using the Chothia system.
  • the CDRs of an antibody can be defined according to the AbM system, which is based on AbM hypervariable regions that represent a compromise between the Kabat CDRs and Chothia structural loops, and where CDRs are determined using Oxford Molecular's AbM antibody modeling software (Oxford Molecular Group, Inc.).
  • the VH CDR1 is present at amino acid positions 26 to 35 of the heavy chain;
  • the VH CDR2 is present at amino acid positions 50 to 61 or 50 to 59 of the heavy chain; and
  • the VH CDR3 is present at amino acid positions 101 to 105 or 99 to 104 or 99 to 106 of the heavy chain.
  • the VL CDR1 is present at amino acid positions 24 to 39 or 24 to 34 of the light chain;
  • the VH CDR2 is present at amino acid positions 55 to 61 or 50 to 56 of the light chain;
  • the VH CDR3 is present at amino acid positions 94 to 102 or 89 to 97 of the light chain.
  • the AbM CDR positions may vary depending on the antibody, and may be determined according to methods known in the art.
  • the CDRs of the antibodies described herein are determined using the AbM numbering system.
  • the CDRs of an antibody can be defined according to the IMGT system (see "IMGT®, the international ImMunoGeneTics information system® website imgt.org, founder and director: Marie-Paule Lefranc, adjoin, France; see, e.g., Lefranc, M.- P., 1999, The Immunologist, 7:132-136 and Lefranc, M.-P. et al., 1999, Nucleic Acids Res., 27:209-212, both of which are incorporated herein by reference in their entirety).
  • IMGT® the international ImMunoGeneTics information system® website imgt.org, founder and director: Marie-Paule Lefranc, adjoin, France; see, e.g., Lefranc, M.- P., 1999, The Immunologist, 7:132-136 and Lefranc, M.-P. et al., 1999, Nucleic Acids Res., 27:209-212, both of which are incorporated herein by reference in their
  • the VH CDR1 is present at amino acid positions 27 to 33 or 26 to 33 of the heavy chain;
  • the VH CDR2 is present at amino acid positions 51 to 60 or 51 to 58 of the heavy chain;
  • the VH CDR3 is present at amino acid positions 99 to 105 or 97 to 103 of the heavy chain.
  • the VL CDR1 is present at amino acid positions 27 to 37 of the light chain;
  • the VH CDR2 is present at amino acid positions 55 to 57 of the light chain; and
  • the VH CDR3 is present at amino acid positions 94 to 102 of the light chain.
  • the IMGT CDR positions may vary depending on the antibody, and may be determined according to methods known in the art. In a specific embodiment, the CDRs of the antibodies described herein are determined using the IMGT system.
  • the CDRs of an antibody can be defined according to the Contact system.
  • the Contact definition is based on an analysis of the available complex crystal structures (bioinf.org.uk/abs) (see MacCallum RM et al., (1996) J Mol Biol 5: 732-745; see also, e.g., Martin A.“Protein Sequence and Structure Analysis of Antibody Variable Domains,” in Antibody Engineering, Kontermann and Dübel, eds., Chapter 31, pp.422-439, Springer-Verlag, Berlin (2001)).
  • the VH CDR1 is present at amino acid positions 30 to 35 of the heavy chain;
  • the VH CDR2 is present at amino acid positions 47 to 61 or 47 to 59 of the heavy chain; and
  • the VH CDR3 is present at amino acid positions 99 to 104 or 97 to 103 or 97 to 105 of the heavy chain.
  • the VL CDR1 is present at amino acid positions 30 to 41 or 30 to 36 of the light chain;
  • the VH CDR2 is present at amino acid positions 51 to 60 or 46 to 55 of the light chain;
  • the VH CDR3 is present at amino acid positions 94 to 101 or 89 to 96 of the light chain.
  • the Contact CDR positions may vary depending on the antibody, and may be determined according to methods known in the art. In a specific embodiment, the CDRs of the antibodies described herein are determined using the Contact system.
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise CDRs of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303), which are defined according to any of the above-described systems.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs (preferably all three VH CDRs) of any anti-HDM2 antibody described herein (such as NMC- 103, NMC-204, or NMC-303).
  • VH heavy chain variable region
  • VHs contain VH CDRs surrounded by framework regions (the CDR and FR sequences appear in the following sequence in the VH: FR1-VH CDR 1-FR2-VH CDR 2-FR3-VH CDR 3-FR4), optionally the framework regions are human framework regions.
  • the framework regions are human framework regions.
  • antibodies or antigen- binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:36 (which is the VH of NMC-103).
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:38 (which is the VH of NMC- 204).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:40 (which is the VH of NMC-303).
  • VH heavy chain variable region
  • such antibody or fragment is a humanized antibody or fragment.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs (preferably all three VL CDRs) of any anti-HDM2 antibody described herein (such as NMC-103, NMC-204, or NMC-303).
  • VL light chain variable region having one, two or all three VL CDRs (preferably all three VL CDRs) of any anti-HDM2 antibody described herein (such as NMC-103, NMC-204, or NMC-303).
  • VLs contain VL CDRs surrounded by framework regions (the CDR and FR sequences appear in the following sequence in the VL: FR1-VL CDR 1-FR2-VL CDR 2-FR3-VL CDR 3-FR4); optionally the framework regions are human framework regions.
  • antibodies or antigen- binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:37 (which is the VL of NMC-103).
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:39 (which is the VL of NMC- 204).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:40 (which is the VL of NMC-303).
  • VL light chain variable region
  • such antibody or fragment is a humanized antibody or fragment.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs (preferably all three VH CDRs) of any anti-HDM2 antibody described herein (such as NMC- 103, NMC-204, or NMC-303) and comprise a light chain variable region (VL) having one, two or all three VL CDRs (preferably all three VL CDRs) of such anti-HDM2 antibody.
  • VH heavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 5 (providing VL CDRs of NMC-103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a haeavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 4; and comprises a light chain variable region (VL) having one, two or all threeVL CDRs identified in Table 5.
  • VH haeavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 7 (providing VL CDRs of NMC-204).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 6; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 7.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 8 (providing VH CDRs of NMC-303).
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 9 (providing VL CDRs of NMC-303).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 8; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 9.
  • VH heavy chain variable region
  • VL light chain variable region
  • the antibody or fragment thereof comprises the sequence (e.g., VH or VL sequence) and not the sequences having a certain percent identity (which is less than 100%) to the sequence.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VH of any antibody described herein, such as a VH of any antibody provided in Section 8 or Figures 20-22 (e.g., the VH of NMC-103, the VH of NMC-204, or the VH of NMC-303), or a VH having at least 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VL of any antibody described herein, such as a VL of any antibody provided in Section 8 or Figures 20-22 (e.g., the VL of NMC-103, the VL of NMC-204, or the VL of NMC-303), or a VL having at least 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VH and a VL of any antibody described herein, such as a VH and VL of any antibody provided in Section 8 or Figures 20-22 (e.g., the VH and VL of NMC-103, the VH and VL of NMC-204, or the VH and VL of NMC-303), or a VHand VL having at least 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:36, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:37, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:38, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:39, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:40, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:41, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more Kabat VL CDRs of a VL of any one of the antibodies described herein (any one of NMC-103, NMC-204, and NMC- 303) and/or one or more Kabat VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise Kabat VH CDR 3 of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three Kabat VL CDRs of a VL of any one of the antibodies described herein (any one of NMC- 103, NMC-204, and NMC-303) and/or three Kabat VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more Chothia VL CDRs of a VL of any one of the antibodies described herein (any one of NMC-103, NMC-204, and NMC- 303) and/or one or more Chothia VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • M(H)DM2/4 e.g., HDM2
  • Chothia VL CDRs of a VL of any one of the antibodies described herein any one of NMC-103, NMC-204, and NMC- 303
  • Chothia VH CDRs of a VH of any one of the antibodies described herein any one of antibodies NMC-103, NMC-204, and NMC-303.
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise Chothia VH CDR 3 of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three Chothia VL CDRs of a VL of any one of the antibodies described herein (any one of NMC-103, NMC-204, and NMC-303) and/or three Chothia VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more AbM VL CDRs of a VL of any one of the antibodies described herein (any one of NMC-103, NMC-204, and NMC- 303) and/or one or more AbM VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • M(H)DM2/4 e.g., HDM2
  • AbM VL CDRs of a VL of any one of the antibodies described herein any one of NMC-103, NMC-204, and NMC- 303
  • AbM VH CDRs of a VH of any one of the antibodies described herein any one of antibodies NMC-103, NMC-204, and NMC-303.
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise AbM VH CDR 3 of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three AbM VL CDRs of a VL of any one of the antibodies described herein (any one of NMC- 103, NMC-204, and NMC-303) and/or three AbM VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more Contact VL CDRs of a VL of any one of the antibodies described herein (any one of NMC-103, NMC-204, and NMC- 303) and/or one or more Contact VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • M(H)DM2/4 e.g., HDM2
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise Contact VH CDR 3 of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three Contact VL CDRs of a VL of any one of the antibodies described herein (any one of NMC-103, NMC-204, and NMC-303) and/or three Contact VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more IMGT VL CDRs of a VL of any one of the antibodies described herein (any one of NMC-103, NMC-204, and NMC- 303) and/or one or more IMGT VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • M(H)DM2/4 e.g., HDM2
  • IMGT VL CDRs of a VL of any one of the antibodies described herein any one of NMC-103, NMC-204, and NMC- 303
  • IMGT VH CDRs of a VH of any one of the antibodies described herein any one of antibodies NMC-103, NMC-204, and NMC-303.
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise IMGT VH CDR 3 of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three IMGT VL CDRs of a VL of any one of the antibodies described herein (any one of NMC- 103, NMC-204, and NMC-303) and/or three IMGT VH CDRs of a VH of any one of the antibodies described herein (any one of antibodies NMC-103, NMC-204, and NMC-303).
  • M(H)DM2/4 e.g., HDM2
  • IMGT VL CDRs of a VL of any one of the antibodies described herein any one of NMC- 103, NMC-204, and NMC-303
  • IMGT VH CDRs of a VH of any one of the antibodies described herein any one of antibodies NMC-103, NMC-204, and NMC-303.
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise CDRs of any one of the antibodies described herein (such as NMC-C303, NMC-C103 and variants of NMC-H103), which are defined according to any of the above-described systems.
  • antibodies e.g., chimeric or humanized antibodies
  • antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs (preferably all three VH CDRs) of any anti-HDM2 antibody described herein (such as NMC-C303, NMC-C103 and variants of NMC-H103).
  • VH heavy chain variable region
  • VHs contain VH CDRs surrounded by framework regions (the CDR and FR sequences appear in the following sequence in the VH: FR1-VH CDR 1-FR2-VH CDR 2-FR3- VH CDR 3-FR4), optionally the framework regions are humanized or human framework regions.
  • the framework regions are humanized or human framework regions.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:40 (which is the VH of NMC-C303).
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:283 (which is the VH of NMC-C103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:287 (which is the VH1 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:291 (which is the VH2 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:295 (which is the VH3 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:299 (which is the VH4 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:305 (which is the VH6 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs of a VH having the amino acid sequence of SEQ ID NO:309 (which is the VH7 variant of NMC-H103).
  • VH heavy chain variable region
  • chimeric or humanized antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs (preferably all fourVH FRs) of any anti-HDM2 antibody described herein (such as NMC-C303, NMC-C103 and variants of NMC-H103).
  • VH heavy chain variable region
  • VHs contain VH CDRs surrounded by framework regions (the CDR and FR sequences appear in the following sequence in the VH: FR1-VH CDR 1-FR2-VH CDR 2-FR3-VH CDR 3-FR4), optionally the framework regions are humanized or human framework regions.
  • the framework regions are humanized or human framework regions.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs of a VH having the amino acid sequence of SEQ ID NO:40 (which is the VH of NMC-C303).
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs of a VH having the amino acid sequence of SEQ ID NO:283 (which is the VH of NMC-C103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs of a VH having the amino acid sequence of SEQ ID NO:287 (which is the VH1 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs of a VH having the amino acid sequence of SEQ ID NO:291 (which is the VH2 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs of a VH having the amino acid sequence of SEQ ID NO:295 (which is the VH3 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs of a VH having the amino acid sequence of SEQ ID NO:299 (which is the VH4 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all fourVH FRs of a VH having the amino acid sequence of SEQ ID NO:305 (which is the VH6 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two, three or all four VH FRs of a VH having the amino acid sequence of SEQ ID NO:309 (which is the VH7 variant of NMC-H103).
  • VH heavy chain variable region
  • antibodies e.g., chimeric or humanized antibodies
  • antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs (preferably all three VL CDRs) of any anti-HDM2 antibody described herein (such as NMC-C303, NMC-C103 and variants of NMC-H103).
  • VL light chain variable region
  • VLs contain VL CDRs surrounded by framework regions (the CDR and FR sequences appear in the following sequence in the VL: FR1-VL CDR 1-FR2-VL CDR 2-FR3- VL CDR 3-FR4); optionally the framework regions are human or humanized framework regions.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:41 (which is the VL of NMC-C303).
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:285 (which is the VL of NMC-C103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:289 (which is the VK1 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:293 (which is the VK2 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:297 (which is the VK3 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:301 (which is the VK4 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:303 (which is the VK5 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:307 (which is the VK6 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two or all three VL CDRs of a VL having the amino acid sequence of SEQ ID NO:311 (which is the VK7 variant of NMC-H103).
  • VL light chain variable region
  • chimeric or humanized antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs (preferably all fourVL FRs) of any anti-HDM2 antibody described herein (such as NMC-C303, NMC-C103 and variants of NMC-H103).
  • VL light chain variable region
  • VLs contain VL CDRs surrounded by framework regions (the CDR and FR sequences appear in the following sequence in the VL: FR1-VL CDR 1-FR2-VL CDR 2-FR3-VL CDR 3-FR4), optionally the framework regions are humanized or human framework regions.
  • the framework regions are humanized or human framework regions.
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:41 (which is the VL of NMC-C303).
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:285 (which is the VL of NMC-C103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:289 (which is the VL1 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:293 (which is the VL2 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:297 (which is the VL3 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:301 (which is the VL4 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:303 (which is the VL5 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all fourVL FRs of a VL having the amino acid sequence of SEQ ID NO:307 (which is the VL6 variant of NMC-H103).
  • VL light chain variable region
  • antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a light chain variable region (VL) having one, two, three or all four VL FRs of a VL having the amino acid sequence of SEQ ID NO:311 (which is the VL7 variant of NMC-H103).
  • VL light chain variable region
  • chimeric or humanized antibodies or antigen-binding fragments thereof that specifically bind to an extracellularly accessible epitope of M(H)DM2/4 (e.g., HDM2) and comprise a heavy chain variable region (VH) having one, two or all three VH CDRs (preferably all three VH CDRs) of any chimeric or humanized anti-HDM2 antibody described herein (such as NMC-C103, NMC-C303, or variants of NMC-H103) and comprise a light chain variable region (VL) having one, two or all three VL CDRs (preferably all three VL CDRs) of such anti-HDM2 antibody.
  • VH heavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 12 (providing VL CDRs of NMC-C303).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a haeavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 11; and comprises a light chain variable region (VL) having one, two or all threeVL CDRs identified in Table 12.
  • VH haeavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 14 (providing VL CDRs of NMC-C103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 13; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 14.
  • VH heavy chain variable region
  • VL light chain variable region
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 13 (providing VH FRs of NMC-C103).
  • VL light chain variable region
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 13; and comprises a light chain variable region (VL) having one, two, three or all fourVL FRs identified in Table 14.
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 16 (providing VK1 variant CDRs of NMC-H103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 15; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 16.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 15 (providing VH1 variant FRs of NMC-H103).
  • VH heavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 18 (providing VK2 variant CDRs of NMC-H103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 17; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 18.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 17 (providing VH2 variant FRs of NMC-H103).
  • VH heavy chain variable region
  • NMC-H103 providing VH2 variant FRs of NMC-H103.
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 20 (providing VK3 variant CDRs of NMC-H103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 19; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 20.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 19 (providing VH3 variant FRs of NMC-H103).
  • VH heavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 22 (providing VK4 variant CDRs of NMC-H103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 21; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 22.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 21 (providing VH4 variant FRs of NMC-H103).
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 23 (providing VK5 variant CDRs of NMC-H103).
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two, three or all four VL FRs identified in Table 23 (providing VK5 variant CDRs of NMC-H103).
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 25 (providing VK6 variant CDRs of NMC-H103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 24; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 25.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 24 (providing VH6 variant FRs of NMC-H103).
  • VH heavy chain variable region
  • VL light chain variable region
  • VH heavy chain variable region
  • CDR-H3 VH CDR3
  • provided herein is an antibody or a fragment thereof that specifically binds to HDM2 and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 27 (providing VK7 variant CDRs of NMC-H103).
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two or all three VH CDRs identified in Table 26; and comprises a light chain variable region (VL) having one, two or all three VL CDRs identified in Table 27.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in Table 26 (providing VH7 variant FRs of NMC-H103).
  • VH heavy chain variable region
  • NMC-H103 light chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in any one of Tables 15, 17, 19, 21, 24, and 26; and comprises a light chain variable region (VL) having one, two, three or all four VL FRs identified in any one of Tables 16, 18, 20, 22, 23, 25, and 27.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, or all three or VH CDRs identified in any one of Tables 15, 17, 19, 21, 24, and 26; and comprises a light chain variable region (VL) having one, two, or all three VL CDRs identified in any one of Tables 16, 18, 20, 22, 23, 25, and 27.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in any one of Tables 15, 17, 19, and 21; and comprises a light chain variable region (VL) having one, two, three or all four VL FRs identified in any one of Tables 16, 18, 20, 22, and 23.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, or all three VH CDRs identified in any one of Tables 15, 17, 19, and 21; and comprises a light chain variable region (VL) having one, two, or all three VL CDRs identified in any one of Tables 16, 18, 20, 22, and 23.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, three or all four VH FRs identified in any one of Tables 21, 24, and 26; and comprises a light chain variable region (VL) having one, two, three or all four VL FRs identified in any one of Tables 20, 25, and 27.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or a fragment thereof that specifically binds to HDM2 and comprises a heavy chain variable region (VH) having one, two, or all three VH CDRs identified in any one of Tables 21, 24, and 26; and comprises a light chain variable region (VL) having one, two, or all three VL CDRs identified in any one of Tables 20, 25, and 27.
  • VH heavy chain variable region
  • VL light chain variable region
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VH of any antibody described herein, such as a VH of any antibody provided in Sections 9 and 11 (e.g., the VH of NMC-C103, the VH of NMC-C303, or the VH of any of NMC-H103 variants), or a VH having at least 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VL of any antibody described herein, such as a VL of any antibody provided in Sections 9 and 11 (e.g., the VL of NMC-C103, the VL of NMC-C303, or the VL of any of NMC-H103 variants), or a VL having at least 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising a VH and a VL of any antibody described herein, such as a VH and VL of any antibody provided in Sections 9 and 11 (e.g., the VH and VL of NMC- C103, the VH and VL of NMC-C303, or the VH and VL of any NMC-H103 variant, e.g., the VH and VL of any NMC-H103 variant tested for binding to NMC-P1 as described in Section 9.6, Example 18 and Figures 38A and 38B), or a VH and VL having at least 75%, 80%, 85%, 90%, 95%, 98%, or 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:40, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:41, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:40, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and (ii) a VL having the amino acid sequence of SEQ ID NO:41, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:283, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:285, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:283, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and (ii) a VL having the amino acid sequence of SEQ ID NO:285, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:287, 291, 295, 299, 305, or 309, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:289, 293, 297, 301, 303, 307, or 311, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:287, 291, 295, 299, 305, or 309, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and (ii) a VL having the amino acid sequence of SEQ ID NO:289, 293, 297, 301, 303, 307, or 311, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:287, 291, 295, or 299, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:289, 293, 297, 301, or 303, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO:287, 291, 295, or 299, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and (ii) a VL having the amino acid sequence of SEQ ID NO:289, 293, 297, 301, or 303, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO: 299, 305, or 309, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and/or (ii) a VL having the amino acid sequence of SEQ ID NO:297, 307, or 311, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • an antibody or fragment thereof that specifically binds to HDM2 comprising: (i) a VH having the amino acid sequence of SEQ ID NO: 299, 305, or 309, or a VH having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto; and (ii) a VL having the amino acid sequence of SEQ ID NO:297, 307, or 311, or a VL having at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% sequence identity thereto.
  • substitutions, insertions, or deletions in these sequences occur in regions outside the CDRs (i.e., in the FRs).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more Kabat VL CDRs and VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or one or more Kabat VH CDRs and VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three Kabat VL CDRs and four Kabat VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or three Kabat VH CDRs and four Kabat VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC- H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more Chothia VL CDRs and VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or one or more Chothia VH CDRs and VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three Chothia VL CDRs and four Chothia VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or three Chothia VH CDRs and four Chothia VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more AbM VL CDRs and VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or one or more AbM VH CDRs and VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three AbM VL CDRs and four AbM VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or three AbM VH CDRs and four AbM VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC- H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more Contact VL CDRs and VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or one or more Contact VH CDRs and VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three Contact VL CDRs and four Contact VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or three Contact VH CDRs and four Contact VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103).
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise one or more IMGT VL CDRs and VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or one or more IMGT VH CDRs and VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103).
  • M(H)DM2/4 e.g., HDM2
  • IMGT VL CDRs and VL FRs of a VL of any one of the chimeric or humanized antibodies described herein such as NMC-C103, NMC-C303, and variants of NMC-H103
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise three IMGT VL CDRs and four IMGT VL FRs of a VL of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC-H103), and/or three IMGT VH CDRs and four IMGT VH FRs of a VH of any one of the chimeric or humanized antibodies described herein (such as NMC-C103, NMC-C303, and variants of NMC- H103).
  • M(H)DM2/4 e.g., HDM2
  • 3 IMGT VL CDRs and four IMGT VL FRs of a VL of any one of the chimeric or humanized antibodies described herein such as NMC-C103, NMC-C303, and variants of NMC- H103.
  • antibodies or fragments thereof that specifically bind to M(H)DM2/4 (e.g., HDM2) and comprise combinations of Kabat CDRs, Chothia CDRs, AbM CDRs, IMGT CDRs, and Contact CDRs (or a combination of CDRs defined by any two, three, four or five of these CDR defining systems).
  • the position of one or more CDRs along the VH (e.g., CDR1, CDR2, or CDR3) and/or VL (e.g., CDR1, CDR2, or CDR3) region of an antibody described herein may vary by one, two, three, four, five, or six amino acid positions so long as immunospecific binding to M(H)DM2/4 (e.g., HDM2) is maintained (e.g., substantially maintained, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%).
  • M(H)DM2/4 e.g., HDM2
  • the position defining a CDR of any of antibody described herein may vary by shifting the N-terminal and/or C-terminal boundary of the CDR by one, two, three, four, five, or six amino acids, so long as immunospecific binding to M(H)DM2/4 (e.g., HDM2) is maintained (e.g., substantially maintained, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%).
  • M(H)DM2/4 e.g., HDM2
  • the length of one or more CDRs along the VH (e.g., CDR1, CDR2, or CDR3) and/or VL (e.g., CDR1, CDR2, or CDR3) region of an antibody described herein may vary (e.g., be shorter or longer) by one, two, three, four, five, or more amino acids, so long as immunospecific binding to M(H)DM2/4 (e.g., HDM2) is maintained (e.g., substantially maintained, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%).
  • M(H)DM2/4 e.g., HDM2
  • an anti-M(H)DM2/4 (e.g., HDM2) antibody described herein is a humanized immunoglobulin (e.g., an IgG) that comprises the 3 VH CDRs and the 3 VL CDRs (i.e., VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3) of any of the antibodies described herein (any one of murine antibodies NMC-103, NMC-204, and NMC-303), respectively, human or human-derived framework regions, and human or human- derived constant regions; antigen-binding fragments of such humanized antibodies are also provided by the present invention.
  • a humanized immunoglobulin e.g., an IgG
  • VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3 any one of murine antibodies NMC-103, NMC-204, and NMC-303
  • human or human-derived framework regions i.
  • a humanizedanti-M(H)DM2/4 (e.g., HDM2) antibody or antigen-binding fragment thereof comprises a VH with VH CDR1, VH CDR2, and VH CDR3 as described herein (e.g., those of MNC-103, NMC-204, or NMC-303), surrounded by VH framework regions that are human framework regions or derived from human framework regions.
  • an anti-M(H)DM2/4 (e.g., HDM2) antibody or antigen-binding fragment thereof comprises a VL with VL CDR1, VL CDR2, and VL CDR3 as described herein (e.g., those of MNC-103, NMC-204, or NMC-303), surrounded by VL framework regions that are human framework regions or derived from human framework regions.
  • VL CDR1, VL CDR2, and VL CDR3 as described herein (e.g., those of MNC-103, NMC-204, or NMC-303), surrounded by VL framework regions that are human framework regions or derived from human framework regions.
  • an anti-M(H)DM2/4 (e.g., HDM2) antibody or antigen-binding fragment thereof comprises (i) a VH with VH CDR1, VH CDR2, and VH CDR3 as described herein (e.g., those of MNC-103, NMC-204, or NMC-303), surrounded by VH framework regions that are human framework regions or derived from human framework regions; and (ii) a VL with VL CDR1, VL CDR2, and VL CDR3 as described herein (e.g., those of MNC-103, NMC-204, or NMC-303), surrounded by VL framework regions that are human framework regions or derived from human framework regions.
  • a VH with VH CDR1, VH CDR2, and VH CDR3 as described herein (e.g., those of MNC-103, NMC-204, or NMC-303), surrounded by VL framework regions that are human framework regions or derived from human framework regions.
  • Human framework regions that may be used include, without limitation: (i) framework regions selected using the "best-fit” method (see, e.g., Sims et al. J. Immunol.
  • 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)
  • framework regions derived from screening FR libraries see, e.g., Baca et al., J. Biol. Chem.
  • one or more of the CDRs of an anti-M(H)DM2/4 (e.g., HDM2) or antigen-binding fragment thereof described herein may be inserted within known framework regions.
  • the framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions.
  • described herein are polynucleotides comprising combinations of the framework regions and CDRs that encode an anti-M(H)DM2/4 (e.g., HDM2) or antigen-binding fragment thereof that specifically binds M(H)DM2/4 (e.g., HDM2).
  • One or more (e.g., one or two or three) amino acid substitutions may be made within the framework regions, preferably, one or more (e.g, one or two or three) amino acid substitutions may be made that improve binding of the antibody to M(H)DM2/4 (e.g., HDM2).
  • M(H)DM2/4 e.g., HDM2
  • the framework regions in the variable domains can be those of the native (e.g., murine) antibody).
  • Antibodies provided herein include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules (i.e., molecules that possess an antigen-binding site) that specifically bind to an extracellular region (epitope) of M(H)DM2/4 accessible on the plasma membrane surface of cancer cells (for example, an epitope that is expressed or exposed on the plasma membrane of cancer cells at greater levels than on non-cancer cells (e.g., when such cancer and non-cancer cells originated from the same tissue)).
  • immunoglobulin molecules i.e., molecules that possess an antigen-binding site
  • epitope that is expressed or exposed on the plasma membrane of cancer cells at greater levels than on non-cancer cells (e.g., when such cancer and non-cancer cells originated from the same tissue)
  • anti- M(H)DM2/4 antibodies described herein are monoclonal antibodies or fragments thereof.
  • the antibodies and fragments described herein are preferably human, humanized or chimeric.
  • a human antibody can be a human immunoglobulin, which may be isolated from a human immunoglobulin library or isolated from mice or other animals that express antibodies from human genes.
  • an antibody provided herein is human (or a fragment of a human antibody).
  • an antibody provided herein is humanized (or a fragment of a humanized antibody).
  • an antibody provided herein is chimeric (or a fragment of a chimeric antibody) (where a chimeric antibody is an antibody with a variable region of one species (e.g., murine) and a constant region of another species (e.g., human)).
  • a chimeric antibody is an antibody with a variable region of one species (e.g., murine) and a constant region of another species (e.g., human)).
  • an antibody provided herein is a human, humanized or chimeric monoclonal antibody (which is particularly suitable for treatment of human subjects).
  • an antibody provided herein is a synthetic antibody.
  • an antibody provided herein is a multi-specific antibody (e.g., a bi-specific antibody).
  • an antibody provided herein is a single chain antibody, e.g., a single chain Fv (scFv).
  • an antigen-binding fragment of an anti- M(H)DM2/4 antibody is provided herein wherein the fragment can be, without limitation, an Fv fragment, a Fab fragment, a F(ab ⁇ ) fragment, a F(ab ⁇ ) 2 fragment, or a disulfide-linked Fv (sdFv).
  • an antigen-binding fragment provided herein is an Fv fragment.
  • an antigen-binding fragment provided herein is a Fab fragment.
  • an antigen-binding fragment provided herein is a F(ab ⁇ ) 2 fragment. In one embodiment, an antigen- binding fragment provided herein is a F(ab ⁇ ) fragment.
  • an antibody provided herein is a multispecific antibody (such as a bi-specific antibody) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed on the plasma membrane surface of cancer cells and specifically binds to a second antigen, wherein such binding allows re-targeting of effector cells towards tumor cells (as an example of such engineered bi-specific antibodies directed to a different target see Chames et al., 2009, MAbs 1:539-547, describing an antibody termed catumaxomab, a T-cell targeting agent).
  • a multispecific antibody such as a bi-specific antibody
  • an antibody provided herein is a multispecific antibody (such as a bi- specific antibody) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed on the plasma membrane surface of cancer cells, and also binds to an antigen exposed on the plasma membrane surface of an effector cell.
  • Effector cells include but are not limited to T cells, natural killer cells, neutrophils, macrophages, dendritic cells and B lymphocytes.
  • an antibody provided herein is a multispecific antibody (e.g., a bi-specific antibody) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed on the plasma membrane surface of cancer cells, and also specifically binds to an antigen exposed on the surface of T cells (e.g., cytotoxic T cells).
  • an antibody provided herein is a multispecific antibody (e.g., a bi-specific antibody) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed on the plasma membrane surface of cancer cells, and also specifically binds to CD3.
  • an antibody provided herein is a multispecific antibody (e.g., a bi-specific antibody) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed on the plasma membrane surface of cancer cells, and also specifically binds to an antigen exposed on the surface of natural killer cells, neutrophils, macrophages, dendritic cells, and/or B-lymphocytes.
  • a multispecific antibody e.g., a bi-specific antibody
  • an antibody provided herein is a multispecific antibody (e.g., a bi-specific antibody) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed on the plasma membrane surface of cancer cells, and also specifically binds to an antigen exposed on the surface of neutrophils, macrophages, dendritic cells, and/or B-lymphocytes.
  • a multispecific antibody e.g., a bi-specific antibody
  • an antibody provided herein is a multispecific antibody (e.g., a bi-specific antibody) that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 exposed on the plasma membrane surface of cancer cells, and also specifically binds to an antigen exposed on the surface of natural killer cells, macrophage and/or dendritic cells.
  • a multispecific antibody e.g., a bi-specific antibody
  • the antibody used is a monoclonal antibody or an antigen-binding fragment thereof that is human, humanized or chimeric.
  • the antibody can be an antibody or fragment appropriate for use in the treated species (i.e., of that species).
  • the antibodies described herein can be from any animal species, such as mammals (e.g., mouse, donkey, sheep, rabbit, goat, guinea pig, camel, horse, dog, cat) or birds (e.g., chicken).
  • the antibody is an immunoglobulin
  • immunoglobulin molecules that can be used are of any type (e.g., IgG, IgE, IgM, IgD, IgA, IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, IgA2) or subclass of immunoglobulin molecule.
  • the antibody is an immunoglobulin, and, in particular, an IgG.
  • the antibody is an IgM.
  • the anti- M(H)DM2/4 antibodies or fragments described herein are antibodies or fragments that mediate complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytoxicity (ADCC), and/or cytotoxicity due to a cytotoxic drug bound to the antibody or fragment.
  • CDC complement-dependent cytotoxicity
  • ADCC antibody-dependent cell-mediated cytoxicity
  • cytotoxicity due to a cytotoxic drug bound to the antibody or fragment are antibodies or fragments that mediate complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytoxicity (ADCC), and/or cytotoxicity due to a cytotoxic drug bound to the antibody or fragment.
  • the anti- M(H)DM2/4 antibodies or fragments described herein are antibodies or fragments that are capable of inducing cytotoxicity against the cancer cells being targeted by such antibodies or fragments, where the cytotoxicity can be due to complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), or due to cytotoxicity of a drug bound to the antibody (where the antibody used is in a form of an antibody-drug conjugate).
  • CDC complement-dependent cytotoxicity
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • a drug bound to the antibody where the antibody used is in a form of an antibody-drug conjugate.
  • the anti- M(H)DM2/4 antibodies or fragments described herein are antibodies or fragments that mediate complement-dependent cytotoxicity (CDC).
  • CDC complement-dependent cytotoxicity
  • the Fc region of the antibody described herein is of a human IgG (e.g., IgG1, IgG2, IgG3, IgG4) type or a human IgM type.
  • the Fc region of the antibody described herein is of a mouse IgG (e.g., IgG1, IgG2a, IgG2b, IgG3) or mouse IgM type.
  • the Fc region of the antibody described herein is of a human IgG1 isotype.
  • the Fc region of the antibody described herein is of a human IgG3 isotype.
  • the Fc region of the antibody described herein is of a human IgG2 isotype. In one embodiment, the Fc region of the antibody described herein is bioengineered (e.g., mutated) to increase its CDC activity. In one embodiment, the antibody or fragment is a bispecific antibody or fragment that specifically binds to two distinct extracellular epitopes on M(H)DM2/4 (which may, e.g., lead to the amplification of complement activation, the increased deposition of fragments (C3b, iC3b, C3d, C3g, C4b) on the cancer cell surface membrane, and/or the increased cancer cell killing by the MAC).
  • M(H)DM2/4 which may, e.g., lead to the amplification of complement activation, the increased deposition of fragments (C3b, iC3b, C3d, C3g, C4b) on the cancer cell surface membrane, and/or the increased cancer cell killing by the MAC).
  • the antibody or fragment is a bispecific antibody or fragment that specifically binds to an extracellular epitope of M(H)DM2/4 and specifically binds to an extracellular epitope of a complement regulatory protein (CRP) (which may, e.g., prevent the degradation of the freshly deposited immunologically active fragments (C3b, iC3b, C3d, C3g, C4b) by CRPs, amplify the activation of the complement cascade, and/or amplify MAC induced cancer cell lysis).
  • CRP complement regulatory protein
  • Such bispecific anti-M(H)DM2/4 antibodies or fragments may increase CDC activity, increase ADCC activity, increase antibody-dependent cellular phagocytosis (ADCP) by neutrophils and macrophages, or increase CDC, ADCC and ADCP.
  • ADCP antibody-dependent cellular phagocytosis
  • the anti-M(H)DM2/4 antibodies or fragments described herein are antibodies or fragments that mediate antibody-dependent cell- mediated cytoxicity (ADCC) and/or antibody-dependent cellular phagocytosis (ADCP).
  • ADCC antibody-dependent cell- mediated cytoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • the Fc region of the antibody mediates its binding to an Fc receptor, FcR, on neutrophils, macrophages, natural killer cells, eosinophils and mast cells, which leads to ADCC and on neutrophils, macrophages and dendritic cells resulting in ADCP.
  • FcR Fc receptor
  • the Fc region of the antibody described herein is of a human IgG (e.g., IgG1, IgG2, IgG3) type or a human IgE type.
  • the Fc region of the antibody described herein is of a human IgG1 isotype.
  • the Fc region of the antibody described herein is bioengineered (e.g., via cross-linking, via di-sulfide bond formation, via oligosaccharide addition, or via mutation) to increase its ADCC and/or ADCP activity.
  • the Fc region of the antibody described herein is mutated to increase the lifespan of the intact antibody (e.g., in accordance with the methods described in Vaccaro et al., 2005, Nat Biotechnol.23:1283-8128, the disclosure of which is incorporated by reference herein).
  • amino acid substitutions in the Fc CH2 and CH3 domains can be employed to direct the efficacy towards ADCC/ADCP and away from CDC or to increase the efficiency of all three cytotoxic activities (i.e., ADCC, ADCP and CDC).
  • CH2 and/or CH3 domains of the Fc region of the antibody described herein are modified at their glycosylation sites to remove/reduce fucose residues in order to improve ADCC and/or ADCP function, e.g., in accordance with the methods described in, e.g., Satoh et al., 2006, Expert Opin Biol Ther.6:1161-1173 and/or Liu et al., 2015, Ca Immunol Res.3:173-183, the disclosures of which are incorporated by reference herein).
  • the anti-M(H)DM2/4 antibodies described herein having an IgG Fc region are bioengineered at their Fc region to change the N-glycan structure at their glycosylation site to the G0 glycan type terminating in GlcNAc (N-acetylglucosamine), and without fucose and sialic acid residues (which may result in, e.g., the activation of both the classic and alternate pathway of complement pathways and in increased binding to lectins including the mannose-binding lectins secreted during inflammatory responses).
  • GlcNAc N-acetylglucosamine
  • the Fc region of the antibody described herein is of a human IgG1 isotype and has alanine substitution at position 333 of its CH2 domain.
  • the Fc region of the antibody described herein is of a human IgG1 isotype and has a triple mutation S239D/I332E/A330L (which leads to a higher affinity for FcgRIIIa and a lower affinity for FcgRIIb resulting in enhanced ADCC) (such Fc modification can be made, e.g., in
  • the Fc region of the antibody described herein is of a human IgG1 isotype and has a triple mutation S239D/I332E/G236A (which leads to improved FcgRIIIa affinity and
  • the anti-M(H)DM2/4 antibodies or fragments described herein are antibodies or fragments that mediate both complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytoxicity (ADCC).
  • the anti- M(H)DM2/4 antibodies or fragments described herein are antibodies or fragments that mediate complement-dependent cytotoxicity (CDC), antibody- dependent cell-mediated cytoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP).
  • contemplated herein is an antibody or fragment that mediates only CDC or only ADCC activity.
  • the CDC and ADCC function of the antibodies described herein can be tested by any in vitro and/or in vivo cytotoxicity assays known in the art.
  • an antibody or fragment used herein comprises one or more amino acid mutations or substitutions in the Fc region that improve its CDC or ADCC activity (e.g., any mutations or substitutions described herein or known in the art (see, e.g., Idusogie et al., 2001. J Immunol.166(4):2571-5; Strohl, 2009, Curr Opin Biotechnol.20(6):685-91; Lazar et al., 2006, PNAS 103(11): 4005–4010, the disclosures of which are incorporated by reference herein)).
  • any mutations or substitutions described herein or known in the art see, e.g., Idusogie et al., 2001. J Immunol.166(4):2571-5; Strohl, 2009, Curr Opin Biotechnol.20(6):685-91; Lazar et al., 2006, PNAS 103(11): 4005–4010, the disclosures of which are incorporated by
  • the anti-M(H)DM2/4 antibodies or fragments are unconjugated, for example, are not conjugated to a cytotoxic drug).
  • the anti-M(H)DM2/4 antibodies or fragments described herein are not bound (e.g., not conjugated) to a drug (e.g., to a cytotoxic drug).
  • the anti-M(H)DM2/4 antibodies or fragments mediate CDC and/or ADCC.
  • an anti-M(H)DM2/4 antibody or fragment thereof specifically binds to an extracellularly accessible segment (i.e. epitope) within amino acids 1 to 15, 15 to 25 or 475 to 491 of HDM2 (SEQ ID NO:4).
  • an anti-HDM2 antibody or fragment thereof specifically binds to HDM2 within amino acids of SEQ ID NO:1 (which are amino acids 1 to 15 of HDM2 (SEQ ID NO:4)). Amino acids of SEQ ID NO:1 (which are amino acids 1 to 15 of SEQ ID NO:4) are in an extracellularly accessible epitope of HDM2.
  • an anti- HDM2 antibody or fragment thereof specifically binds to HDM2 within amino acids of SEQ ID NO:2 (which are amino acids 15 to 25 of HDM2 (SEQ ID NO:4)). Amino acids of SEQ ID NO:2 (which are amino acids 15 to 25 of SEQ ID NO:4) are in an extracellularly accessible epitope of HDM2.
  • an anti-HDM2 antibody or fragment thereof specifically binds to HDM2 within amino acids of SEQ ID NO:3 (which are amino acids 475 to 491 of HDM2 (SEQ ID NO:4)). Amino acids of SEQ ID NO:3 (which are amino acids 475 to 491 of SEQ ID NO:4) are in an extracellularly accessible epitope of HDM2.
  • anti-M(H)DM2/4 antibodies or fragments thereof specifically bind to an extracellularly accessible epitope of HDM2 within amino acids 50 to 60 of
  • anti-M(H)DM2/4 antibodies or fragments thereof specifically bind to an extracellularly accessible epitope of M(H)DM2/4 within amino acids 100 to 110 of M(H)DM2/4 (SEQ ID NO:4 or SEQ ID NO:6).
  • anti-M(H)DM2/4 antibodies or fragments thereof specifically bind to an extracellular epitope of M(H)DM2/4 within amino acids 1 to 126 of M(H)DM2/4 (SEQ ID NO:4 or SEQ ID NO:6).
  • anti-M(H)DM2/4 antibodies or fragments thereof specifically bind to an extracellularly accessible epitope of M(H)DM2/4 within amino acids 436 to 482 of M(H)DM2/4 (SEQ ID NO:4 or SEQ ID NO:6).
  • anti-M(H)DM2/4 antibodies or fragments thereof specifically bind to an extracellularly accessible epitope of M(H)DM2/4 within the terminal 100 amino acids at the C- terminus of the M(H)DM2/4 (e.g., M(H)DM2/4 protein variant (splice variant) known or expected to be expressed on the plasma membrane of cells of the cancer type being treated, or M(H)DM2/4 protein variant (splice variant) determined to be expressed on the plasma membrane of cancer cells of the subject being treated).
  • M(H)DM2/4 protein variant splice variant
  • the invention also provides anti-M(H)DM2/4 antibodies or fragments thereof that compete for binding to HDM2 with an antibody that specifically binds to HDM2 within the amino acid sequence of SEQ ID NO:1 (e.g., NMC-103 antibody described herein, or any antibody or fragment having the VH of NMC-103 (i.e., the VH of SEQ ID NO:36) and the VL of NMC-103 (i.e., the VL of SEQ ID NO:37), or any antibody or fragment having the VH and VL CDRs of NMC-103).
  • SEQ ID NO:1 e.g., NMC-103 antibody described herein, or any antibody or fragment having the VH of NMC-103 (i.e., the VH of SEQ ID NO:36) and the VL of NMC-103 (i.e., the VL of SEQ ID NO:37), or any antibody or fragment having the VH and VL CDRs of NMC-103.
  • the invention also provides anti-M(H)DM2/4 antibodies or fragments thereof that compete for binding to HDM2 with an antibody that specifically binds to HDM2 within the amino acid sequence of SEQ ID NO:2 (e.g., NMC-204 antibody described herein, or any antibody or fragment having the VH of NMC-204 (i.e., the VH of SEQ ID NO:38) and the VL of NMC-204 (i.e., the VL of SEQ ID NO:39), or any antibody or fragment having the VH and VL CDRs of NMC204).
  • an antibody that specifically binds to HDM2 within the amino acid sequence of SEQ ID NO:2 (e.g., NMC-204 antibody described herein, or any antibody or fragment having the VH of NMC-204 (i.e., the VH of SEQ ID NO:38) and the VL of NMC-204 (i.e., the VL of SEQ ID NO:39), or any antibody or fragment having the VH and VL CDRs
  • the invention also provides anti-M(H)DM2/4 antibodies or fragments thereof that compete for binding to HDM2 with an antibodythat specifically binds to HDM2 within the amino acid sequence of SEQ ID NO:3 (e.g., NMC-303 antibody described herein, or any antibody or fragment having the VH of NMC-303 (i.e., the VH of SEQ ID NO:40) and the VL of NMC-303 (i.e., the VL of SEQ ID NO:41), or any antibody or fragment having the VH and VL CDRs of NMC-303).
  • NMC-303 antibody described herein or any antibody or fragment having the VH of NMC-303 (i.e., the VH of SEQ ID NO:40) and the VL of NMC-303 (i.e., the VL of SEQ ID NO:41), or any antibody or fragment having the VH and VL CDRs of NMC-303).
  • any competition assay known in the art can be used to identify an antibody that competes with an antibody described herein for binding to M(H)DM2/4 (see Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY)).
  • immobilized M(H)DM2/4 e.g., immobilized on a microtiter plate or well
  • a solution comprising a first labeled antibody that binds to M(H)DM2/4 and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to M(H)DM2/4.
  • immobilized M(H)DM2/4 e.g., immobilized on a microtiter plate or well
  • M(H)DM2/4 can be incubated in a solution comprising the first labeled antibody but without the second unlabeled antibody. After incubation, excess unbound antibody is removed, and the amount of label associated with immobilized M(H)DM2/4 is measured. The substantial reduction of the amount of label in the test sample relative to the control sample indicates that the second antibody is competing with the first antibody for binding to M(H)DM2/4.
  • an antibody that competes with an antibody described herein e.g., antibodies having the VH and VL of NMC-103, NMC-204 or NMC-303 for binding to M(H)DM2/4 also binds to the same peptide derived from M(H)DM2/4 that is bound by such antibody (e.g., the peptide of SEQ ID NO:1, SEQ ID NO:2 or SEQ ID NO:3).
  • an antibody that competes with an antibody described herein e.g., antibodies having the VH and VL of NMC-103, NMC-204 or NMC-303 for binding to
  • M(H)DM2/4 also binds to the same epitope in M(H)DM2/4 that is bound by such antibody.
  • an anti-M(H)DM2/4 antibody or fragment thereof binds to the same epitope of M(H)DM2/4 as an antibody or fragment having a VH of SEQ ID NO:36 and a VL of SEQ ID NO:37.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein binds to the same epitope of M(H)DM2/4 as an antibody or fragment having a VH of SEQ ID NO:38 and a VL of SEQ ID NO:39.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein binds to the same epitope of M(H)DM2/4 as an antibody or fragment having a VH of SEQ ID NO:40 and a VL of SEQ ID NO:41.
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 19 to 50 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 19 to 108 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 154 to 167 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 1 to 60 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 1 to 100 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 1 to 108 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 26 to 60 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within the terminal 60 amino acids at the C-terminus of the HDM2 (e.g., HDM2 protein variant (splice variant) known or expected to be expressed on the plasma membrane of cells of the cancer type being treated, or HDM2 protein variant (splice variant) determined to be expressed on the plasma membrane of cancer cells of the subject being treated).
  • HDM2 protein variant splice variant known or expected to be expressed on the plasma membrane of cells of the cancer type being treated
  • HDM2 protein variant splice variant
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within the terminal 100 amino acids at the C-terminus of the HDM2 (e.g., HDM2 protein variant (splice variant) known or expected to be expressed on the plasma membrane of cells of the cancer type being treated, or HDM2 protein variant (splice variant) determined to be expressed on the plasma membrane of cancer cells of the subject being treated).
  • HDM2 protein variant splice variant known or expected to be expressed on the plasma membrane of cells of the cancer type being treated
  • HDM2 protein variant splice variant
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein specifically binds to an extracellularly accessible epitope within amino acids 101 to 200 of HDM2 (SEQ ID NO:4).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein competes for binding to HDM2 with antibody OP145 (monoclonal antibody commercially available from Calbiochem, Catalogue No. OP145-100UG; see Table 10, below, for further details regarding OP145).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein competes for binding to HDM2 with antibody 965 (SMP14) (monoclonal antibody commercially available from Santa Cruz,
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein competes for binding to HDM2 with antibody sc-813 (N-20) (polyclonal antibody commercially available from Santa Cruz, Catalogue No. Sc-813; see Table 10, below, for further details regarding sc-813 (N-20)).
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein competes for binding to HDM2 with antibody sc-812 (C-18) (polyclonal antibody commercially available from Santa Cruz, Catalogue No. Sc-812; see Table 10, below, for further details regarding sc-812 (C-18)).
  • C-18 polyclonal antibody commercially available from Santa Cruz, Catalogue No. Sc-812; see Table 10, below, for further details regarding sc-812 (C-18)
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein competes for binding to HDM2 with antibody M01, clone 1A7 (monoclonal antibody commercially available from Abnova, Catalogue No. H00004193-M01; see Table 3, below, for further details regarding M01, clone 1A7).
  • Any competition assay known in the art can be used to identify an antibody that competes with antibody OP145, SMP14, N-20, C-18, or M01, clone 1A7 for binding to
  • M(H)DM2/4 e.g., HDM2
  • an antibody that competes with antibody OP145, SMP14, N-20, C-18, or M01, clone 1A7 for binding to M(H)DM2/4 also binds to the same epitope that is bound by such antibodies.
  • an anti- M(H)DM2/4 antibody or fragment thereof for use in the methods described herein binds to the same epitope of HDM2 as antibody OP145 (see Table 10, below, for further details regarding OP145).
  • an anti- M(H)DM2/4 antibody or fragment thereof for use in the methods described herein binds to the same epitope of HDM2 as antibody 965 (SMP14) (see Tables 3 and 10, below, for further details regarding 965
  • an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein binds to one of the same epitope(s) of HDM2 as polyclonal antibody sc-813 (N-20) (see Table 10, below, for further details regarding 813 (N-20)). In one embodiment, an anti-M(H)DM2/4 antibody or fragment thereof for use in the methods described herein binds to one of the same epitope(s) of HDM2 as polyclonal antibody sc-812 (C-18) (see Table 10, below, for further details regarding sc-812 (C-18)).
  • an anti- M(H)DM2/4 antibody or fragment thereof for use in the methods described herein binds to the same epitope of HDM2 as antibody M01, clone 1A7 (i.e., monoclonal antibody commercially available from Abnova, Catalogue No. H00004193-M01; see Table 3, below, for further details regarding M01, clone 1A7).
  • the anti-M(H)DM2/4 antibody or fragment thereof described herein is purified.
  • an antibody or fragment 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) methods (see Flatman et al., J. Chromatogr. B 848:79-87 (2007) for review of methods for assessment of antibody purity).
  • electrophoretic e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • the anti-M(H)DM2/4 antibody or fragment described herein can be fused or conjugated (e.g., covalently or non-covalently linked) to a detectable label or substance.
  • a detectable label or substance e.g., covalently or non-covalently linked
  • detectable labels or substances include enzyme labels, radioisotopes (e.g., iodine, carbon, sulfur, tritium, indium, and technetium), luminescent labels, fluorescent labels, and biotin.
  • This methodology can be used to determine whether cells of a certain cancer (e.g., cells of cancer in a patient) express M(H)DM2/4, or a certain splice variant of M(H)DM2/4, on the plasma membrane, where the detection of M(H)DM2/4 using the antibody (or fragment) may indicate that the antibody (or fragment) (with or without the detectable label or substance) can be used in the diagnosis and treatment of the cancer or preventing metastases of the cancer.
  • the invention provides antibody-drug conjugates comprising an anti-M(H)DM2/4 antibody or fragment described herein bound (e.g., covalently bound) to a cytotoxic drug.
  • the antibody-drug conjugates are intended to mediate cytotoxicity by delivery of a cytotoxic drug to the cells of the cancer.
  • an anti-M(H)DM2/4 antibody or an antigen-binding fragment described herein can be bound or conjugated to one or more cytotoxic agents.
  • the cytotoxic agent can be any agent that inhibits or prevents a vital cellular function (e.g., cell division) and/or causes cell death or destruction.
  • chemotherapeutic agents e.g., any chemotherapeutic agent known in the art or described herein
  • toxins e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof
  • radioactive isotopes e.g., radioactive isotopes, growth inhibitory agents, and nucleolytic enzymes.
  • the antibody-drug conjugates and their methods of making are known in the art (see, e.g., Peters & Brown, 2015, Biosci. Rep.35, e00225, doi:10.1042/BSR20150089).
  • cytotoxic agents that can be conjugated to an anti-M(H)DM2/4 antibody or fragment described herein include, without limitation, anthracyclin, doxorubicin, methotreaxate, an anti-metabolite agent, an anti-folate agent, an auristatin (e.g., MMAE or MMAF), a maytansine, a calicheamicin, a duocarymucin, and a pyrrolobenzodiazepine (PBD) dimer.
  • anthracyclin doxorubicin
  • methotreaxate an anti-metabolite agent
  • an anti-folate agent an anti-folate agent
  • an auristatin e.g., MMAE or MMAF
  • a maytansine e.g., MMAE or MMAF
  • a calicheamicin e.g., a duocarymucin
  • PBD pyrrolobenzodiazepine
  • an M(H)DM2/4 antibody or fragment described herein is conjugated to one or more of the following drugs: a maytansinoid, an auristatin (such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF)), a dolastatin, a calicheamicin or derivative thereof, an anthracycline (such as daunomycin or doxorubicin), methotrexate, vindesine, a taxane (such as docetaxel), paclitaxel, larotaxel, tesetaxel, ortataxel, and a trichothecene.
  • a maytansinoid such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF)
  • a dolastatin such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF)
  • a calicheamicin or derivative thereof an anthracycline (such as daun
  • an M(H)DM2/4 antibody or fragment described herein is conjugated to a toxin or a fragment thereof (e.g., diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain, ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins, momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, or a tricothecene).
  • a toxin or a fragment thereof e.g., diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain, ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites for
  • an M(H)DM2/4 antibody or fragment described herein is conjugated to a radioactive isotope (e.g., At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 , or a radioactive isotope of Lu).
  • a radioactive isotope e.g., At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 , or a radioactive isotope of Lu.
  • an M(H)DM2/4 antibody or fragment thereof described herein is conjugated to nanoparticles or other targeting tools to promote concentrated delivery to and retention of the antibodies at the tumor site.
  • anti-M(H)DM2/4 antibodies or fragments described herein can be produced by any method known in the art.
  • monoclonal antibodies are also known in the art, and include the use of hybridoma, recombinant and phage display technologies, and the use of humanized mice.
  • monoclonal antibodies can be produced using hybridoma techniques as taught, for example, in Harlow E & Lane D, Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed.1988); Hammerling GJ et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563681 (Elsevier, N.Y., 1981), or in Kohler G & Milstein C (1975) Nature 256: 495.
  • human monoclonal antibodies can be produced using humanized mice as taught, for example, in Laffleur et al., 2012, Methods Mol. Biol.901:149-59. [00319] Methods for producing specific antibodies using hybridoma technology are routine and well known in the art. In particular, a mouse or another appropriate host animal can be immunized to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the target protein (i.e., extracellular region of M(H)DM2/4) used for immunization.
  • target protein i.e., extracellular region of M(H)DM2/4
  • Lymphocytes then are fused with myeloma cells to form a hybridoma cell (see Goding JW (Ed), Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986); see also Kozbor D (1984) J Immunol 133: 3001-5; Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp.51-63 (Marcel Dekker, Inc., New York, 1987)).
  • the hybridoma cells are then grown in a suitable culture medium, which can be assayed for production of monoclonal antibodies directed against M(H)DM2/4.
  • the binding specificity of monoclonal anti- M(H)DM2/4 antibodies produced by this method can be determined by methods known in the art, e.g., immunoprecipitation or an in vitro assay, such as
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the antibodies or fragments described herein can also be made using various phage display technologies known in the art (see Brinkman U et al., (1995) J Immunol Methods 182: 41-50; Ames RS et al., (1995) J Immunol Methods 184: 177-186; Kettleborough CA et al., (1994) Eur J Immunol 24: 952-958; Persic L et al., (1997) Gene 187: 9-18; and Burton DR & Barbas CF (1994) Advan Immunol 57: 191-280).
  • chimeric antibodies i.e., antibody with a variable region of one species (e.g., murine) and a constant region of another species (e.g., human)
  • a variable region of one species e.g., murine
  • a constant region of another species e.g., human
  • Fab and F(ab ⁇ ) 2 fragments can be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab ⁇ ) 2 fragments).
  • WO 93/17105 Tan P et al., (2002) J Immunol 169: 1119- 25; Caldas C et al., (2000) Protein Eng.13(5): 353-60; Morea V et al., (2000), Methods 20(3): 267-79; Baca M et al., (1997) J Biol Chem 272(16): 10678-84; Roguska MA et al., (1996) Protein Eng 9(10): 895904; Couto JR et al., (1995) Cancer Res.55 (23 Supp): 5973s-5977s; Couto JR et al., (1995) Cancer Res 55(8): 1717-22; Sandhu JS (1994) Gene 150(2): 409- 10 and Pedersen JT et al., (1994) J Mol Biol 235(3): 959-73).
  • a humanized antibody is made by CDR grafting.
  • Methods of making human antibodies are known in the art and include phage display methods using antibody libraries derived from human immunoglobulin sequences (see U.S. Patent Nos.4,444,887, 4,716,111, and 5,885,793; and International Publication Nos. WO
  • human antibodies can be produced using mouse-human hybridomas (see Shinmoto H et al., (2004) Cytotechnology 46: 19-23; Naganawa Y et al., (2005) Human Antibodies 14: 27-31).
  • scFv antibodies are also known in the art (see Ahmad et al., 2012, Clinical and Developmental Immunology, doi:10.1155/2012/980250; Wang et al., 2006, Anal. Chem.78, 997–1004; Pansri et al., 2009, BMC Biotechnology 9:6).
  • scFv antibodies can be constructed by fusing variable domains of heavy and light chains of immunoglobulins via short polypeptide linkers (using recombinant expression techniques), and scFv antibodies having desired antigen-binding properties can be selected by phage display technology.
  • a candidate antibody or fragment for use in the therapeutic and diagnostic methods provided herein is not yet known or has not yet been demonstrated to bind to a region of M(H)DM2/4 exposed on the surface of cancer cells, the antibody or fragment may optionally be tested by any of the following methods:
  • a method for identifying an anti- M(H)DM2/4 antibody or a fragment thereof for use in the methods described herein comprising: (a) contacting intact cancer cells (e.g., cancer cells expected, known, or determined to express M(H)DM2/4) with an anti- M(H)DM2/4 antibody or a fragment thereof; and (b) determining whether the antibody or fragment binds to the intact cancer cells, in particular on the extracellular surface of the cancer cells (e.g., relative to intact cancer cells not contacted by said antibody or fragment), wherein the binding of the anti-M(H)DM2/4 antibody or fragment to the intact cancer cells contacted with such antibody or fragment indicates that said antibody or fragment is suitable for use in the methods described herein.
  • the cancer cells are from the patient proposed to be treated with the antibody or fragment thereof.
  • a method for identifying an anti- M(H)DM2/4 antibody or a fragment thereof for use in the methods described herein comprising: (a) contacting intact cancer cells (e.g., cancer cells expected, known, or determined to express M(H)DM2/4) with an anti- M(H)DM2/4 antibody or a fragment thereof; and (b) determining whether the antibody or fragment binds to the intact cancer cells (in particular on the extracellular surface of the cancer cells) at an increased level relative to binding to intact normal cells (e.g., non-cancerous cells of the same tissue or organ type as the cancer cells), wherein increased binding of the anti- M(H)DM2/4 antibody or fragment to the intact cancer cells relative to normal cells indicates that said antibody or fragment is suitable for use in the methods described herein.
  • intact cancer cells e.g., cancer cells expected, known, or determined to express M(H)DM2/4
  • an anti- M(H)DM2/4 antibody or a fragment thereof comprising: (a) contacting intact cancer cells (e.g
  • a method for identifying an anti-M(H)DM2/4 antibody or a fragment thereof for use in the methods described herein comprising: (a) contacting intact cancer cells (e.g., cancer cells known, expected or determined to express M(H)DM2/4) with an anti-M(H)DM2/4 antibody or a fragment thereof; and (b) determining whether the contacting step results in an increased amount of cell death or destruction of the intact cancer cells (e.g., as determined by cell-death markers such as Propidium Iodide staining) relative to the amount of death or destruction of intact cancer cells not contacted by said antibody or fragment and/or relative to the amount of death or destruction of intact normal cells (e.g., non-cancerous cells of the same tissue or organ as the cancer cells) contacted by said antibody or fragment, wherein increased amount of cell death or destruction of the intact cancer cells contacted with the antibody or fragment indicates that said
  • a method for identifying an anti-M(H)DM2/4 antibody or a fragment thereof for use in the methods described herein comprising: (a) contacting intact cancer cells (e.g., cells known, expected or determined to express M(H)DM2/4) with an anti- M(H)DM2/4 antibody or fragment; and (b) determining whether the contacting step results in an increased level of complement-dependent cytotoxicity (CDC) or antibody-dependent cell- mediated cytotoxicity (ADCC) (as determined by one or more cytotoxicity assays) towards the intact cancer cells relative to the level of CDC or ADCC towards intact cancer cells not contacted by said antibody or fragment and/or relative to the level of CDC or ADCC towards intact normal cells (e.g., non- cancerous cells of the same tissue or organ as the cancer cells) contacted by said antibody or fragment, wherein increased level of CDC or ADCC towards the intact cancer cells indicates that said antibody
  • a method for identifying an anti-M(H)DM2/4 antibody or a fragment thereof for use in the methods described herein comprising: (a) contacting intact cancer cells (e.g., cells known, expected or determined to express M(H)DM2/4) with an anti-M(H)DM2/4 antibody or a fragment thereof; and (b) determining whether the contacting step results in increased inhibition of growth and proliferation of the intact cancer cells relative to the inhibition of growth and proliferation of intact cancer cells not contacted by said antibody or fragment and/or relative to the inhibition of growth and proliferation of intact normal cells (e.g., non-cancerous cells of the same tissue or organ as the cancer cells) contacted by said antibody or fragment, wherein increased inhibition of growth and proliferation of the intact cancer cells contacted with the antibody or fragment indicates that said antibody or fragment is suitable for use in the methods described herein.
  • intact cancer cells e.g., cells known, expected or determined to express M(H)DM2/4
  • an anti-M(H)DM2/4 antibody or a fragment thereof comprising:
  • compositions comprising an anti- M(H)DM2/4 antibody or an antigen-binding fragment thereof and a pharmaceutically acceptable carrier.
  • pharmaceutical compositions comprising an antibody-drug conjugate as described herein and a pharmaceutically acceptable carrier.
  • Appropriate pharmaceutically acceptable carriers including, but not limited to, excipients and stabilizers) are known in the art (see, e.g., Remington’s Pharmaceutical Sciences (1990) Mack Publishing Co., Easton, PA).
  • the anti-M(H)DM2/4 antibody or fragment, or antibody-drug conjugate, in the pharmaceutical compositions described herein can be purified.
  • Pharmaceutically acceptable carriers may include an isotonic agent, a buffer, a suspending agent, a dispersing agent, an emulsifying agent, a wetting agent, a sequestering or chelating agent, a pH buffering agent, a solubility enhancer, an antioxidant, an anesthetic, and/or an antimicrobial agent.
  • Suitable excipients include, without limitation, water, saline, glycerol, ethanol, starch, lactose, sucrose, gelatin, malt, propylene, silica gel, sodium stearate, base cream and dextrose.
  • suitable pharmaceutically acceptable carriers may include physiological saline or phosphate buffered saline (PBS), and solutions containing such agents as glucose, polyethylene glycol, polypropylene glycol or other agents.
  • PBS physiological saline or phosphate buffered saline
  • compositions provided herein comprise an anti-M(H)DM2/4 antibody or an antigen-binding fragment thereof, or antibody-drug conjugate, described herein, and optionally one or more other therapeutic (e.g., anti-cancer) agents, in a pharmaceutically acceptable carrier.
  • a pharmaceutical composition may be formulated for any route of administration to a subject.
  • Formulations for injections can be prepared as liquid solutions, suspensions, emulsions, or solid forms suitable for making into a solution or suspension prior to injection.
  • the anti-M(H)DM2/4 antibody or fragment thereof, or antibody-drug conjugate can be used or present in the pharmaceutical composition in a therapeutically effective amount.
  • the therapeutically effective amount of the antibody or conjugate can be determined by standard clinical techniques.
  • a pharmaceutical composition contemplated for use in the therapeutic methods described herein comprises an anti-M(H)DM2/4 antibody or an antigen- binding fragment thereof, or antibody-drug conjugate described herein, and does not comprise any additional anti-cancer agent or therapy.
  • a pharmaceutical composition contemplated for use in the therapeutic methods described herein comprises an anti- M(H)DM2/4 antibody or an antigen-binding fragment thereof, or antibody-drug conjugate described herein, and further comprises an additional anti-cancer agent or therapy (e.g., any one, two or more additional anti-cancer agents or therapies described herein).
  • a vaccine composition comprising: (i) an
  • MCNTNMSVPTDGAVT SEQ ID NO:1
  • TTSQIPASEQE SEQ ID NO:2
  • a vaccine composition comprising: (i) an immunogenic amount of a peptide, wherein the amino acid sequence of the peptide is MCNTNMSVPTDGAVT (SEQ ID NO:1), TTSQIPASEQE (SEQ ID NO:2), or CPVCRQPIQMIVLTYFP (SEQ ID NO:3); and (ii) a pharmaceutically acceptable carrier.
  • the peptide is purified.
  • the composition further comprises an adjuvant (such as an immune-stimulating adjuvant).
  • the pharmaceutically acceptable carrier is a liposome.
  • a vaccine composition can be produced using any method known in the art (see, e.g., Li et al., 2014, Vaccines 2:515-536).
  • the invention provides for treating cancer (e.g., inhibiting cancer proliferation, inhibiting cancer progression, and/or increasing survival) in a subject in need thereof comprising administering to the subject any anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein.
  • the invention provides a method of treating cancer in a subject in need thereof, said method comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4.
  • the invention provides a method of preventing metastasis in a subject in need thereof, said method comprising administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4.
  • the antibody or fragment is not bound to a cell-penetrating peptide.
  • the invention provides for treating cancer (e.g., inhibiting cancer proliferation, inhibiting cancer progression, and/or increasing survivial) in a subject in need thereof comprising administering to the subject an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2 exposed on the surface of cancer cells (e.g., where the cells of the type of cancer being treated are known or expected to have such extracellular region of HDM2 exposed on their plasma membrane surface), preferably wherein the antibody or fragment is not bound to a cell-penetrating peptide.
  • the method of treating cancer encompasses preventing metastasis of the cancer.
  • the method of treating cancer is a method for reducing tumor size (as measured, e.g., by tumor volume or diameter), inhibiting the growth of the tumor, reducing the growth of the tumor, or eradicating the tumor.
  • the invention provides for preventing metastases in a subject that has cancer comprising administering to the subject any anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein.
  • the invention provides for preventing metastases in a subject that has cancer comprising administering to the subject an antibody or a fragment thereof that specifically binds to an extracellular region of HDM2 exposed on the surface of cancer cells (e.g., where the cells of the type of cancer being treated are known or expected to have such extracellular region of HDM2 exposed on their plasma membrane surface).
  • the invention provides for reducing the number, size or invasiveness of metastases, or eradicating metastases, in a subject that has a metastatic cancer comprising administering to the subject any anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein.
  • anti-M(H)DM2/4 e.g., anti-HDM2
  • the invention provides for reducing the number, size or invasiveness of metastases, or eradicating metastases, in a subject that has a metastatic cancer comprising administering to the subject an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of HDM2 exposed on the surface of cancer cells (e.g., where the cells of the type of cancer being treated are known or expected to have such extracellularly accessible epitope of HDM2 exposed on their plasma membrane surface).
  • the invention provides for treating cancer (e.g., inhibiting cancer proliferation, inhibiting cancer progression, and/or increasing survival) in a subject who has experienced an accelerated rate of cancer growth in response to administration of an inhibitor of one or more inhibitory checkpoint molecules, comprising administering to the subject any anti- M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein.
  • cancer e.g., inhibiting cancer proliferation, inhibiting cancer progression, and/or increasing survival
  • anti- M(H)DM2/4 e.g., anti-HDM2
  • the invention provides for treating cancer (e.g., inhibiting cancer proliferation, inhibiting cancer progression, increasing survival) in a subject who has experienced hyper-progression in response to administration of an inhibitor of one or more inhibitory checkpoint molecules, comprising administering to the subject any anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein.
  • the invention provides for treating cancer (e.g., inhibiting cancer proliferation, inhibiting cancer progression, increasing survival) in a subject who is at risk for hyper-progression in response to administration of an inhibitor of one or more inhibitory checkpoint molecules, comprising administering to the subject any anti-M(H)DM2/4 (e.g., anti- HDM2) antibody or fragment described herein.
  • the methods described herein comprise administering to the subject: an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4.
  • the antibody or fragment is not bound to a cell- penetrating peptide.
  • the inhibitor of one or more inhibitory checkpoint molecules is an inhibitor, such as an inhibitory antibody to, of one or more of: CTLA-4, PD-1, PD-L1, PD-L2, TIM-3, OX40, and LAG-3.
  • the inhibitor of one or more inhibitory checkpoint molecules is an antibody that is nivolumab (Opdivo, Bristol-Myers Squibb), pembrolizumab (Keytruda, Merck), avelumab (Bavencio; EMD Serono, Pfizer), atezolizumab (Tecentriq, Genentech), durvalumab (Imfinzi, AstraZeneca), cemiplimab (Libtayo, Regeneron) or ipilimumab (Yervoy, Bristol-Myers Squibb).
  • the inhibitor of one or more inhibitory checkpoint molecules is an inhibitor of, such as an inhibitory antibody to, PD-1.
  • the methods described herein further comprise, before administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, identifying the subject as a hyper-progressor in response to administration of an inhibitor of one or more inhibitory checkpoint molecules (and, e.g., administering any anti-M(H)DM2/4 (e.g., anti- HDM2) antibody or fragment described herein if the subject is a hyper-progressor).
  • an anti-M(H)DM2/4 e.g., anti-HDM2
  • identifying the subject as a hyper-progressor in response to administration of an inhibitor of one or more inhibitory checkpoint molecules (and, e.g., administering any anti-M(H)DM2/4 (e.g., anti- HDM2) antibody or fragment described herein if the subject is a hyper-progressor).
  • the methods described herein further comprise, before administering an anti- M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, determining whether any anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein binds to the surface of intact cells of the cancer (e.g., intact cancer cells isolated from the subjects) (and, e.g., administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein if an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein binds to the surface of intact cells of the cancer).
  • the methods described herein further comprise, before administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, determining whether the subject has a gene amplification of
  • M(H)DM2/4 (and, e.g., administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein if the subject has a gene amplification of M(H)DM2/4).
  • an anti-M(H)DM2/4 e.g., anti-HDM2
  • the methods described herein further comprise, before administering an anti- M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, determining how many copies of the M(H)DM2/4 gene are present in the cancer cells of the subject, and, e.g., administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein if the cancer cells of the subject have 8 or more than 8 copies of M(H)DM2/4 gene.
  • an anti- M(H)DM2/4 e.g., anti-HDM2
  • an anti-M(H)DM2/4 e.g., anti-HDM2
  • the methods described herein further comprise, before administering an anti- M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, determining the degree of M(H)DM2/4 gene amplification in the cancer cells of the subject, and, e.g., administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein if the cancer cells of the subject have 50% or more than 50% amplification of the M(H)DM2/4 gene relative to the normal cells of the subject (or relative to the cancer cells of the subject prior to administration of an inhibitor of an immune checkpoint molecule).
  • the gene amplification of M(H)DM2/4 is gene amplification of HDM2.
  • the gene amplification of M(H)DM2/4 is gene amplification of HDM4. In one embodiment, the gene amplification of M(H)DM2/4 is gene amplification of HDM2 and HDM4.
  • the methods described herein further comprise, before administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, determining whether the subject has an increased protein expression of M(H)DM2/4 in the cells of the cancer relative to the level of protein expression of M(H)DM2/4 in normal cells (and, e.g., administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein if the subject has an increased protein expression of M(H)DM2/4 in the cells of the cancer relative to the level of protein expression of M(H)DM2/4 in normal cells).
  • the methods described herein further comprise, before administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, determining whether the subject has an increased protein expression of M(H)DM2/4 in the cells of the cancer relative to the level of protein expression of M(H)DM2/4 in cancer cells prior to administration of the inhibitor of a checkpoint molecule (and, e.g., administering an anti- M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein if the subject has an increased protein expression of M(H)DM2/4 in the cells of the cancer relative to the level of protein expression of M(H)DM2/4 in cancer cells prior to administration of the inhibitor of a checkpoint molecule).
  • an anti-M(H)DM2/4 e.g., anti-HDM22
  • the methods described herein further comprise, before administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein, determining whether the subject has an increased binding of an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 to the surface of intact cells of the cancer relative to its binding to the surface of intact normal cells (or relative to its binding to the surface of intact cells of the cancer prior to administration of an inhibitor of an immune checkpoint molecule).
  • an anti-M(H)DM2/4 e.g., anti-HDM22
  • the methods described herein comprise administering an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein if the subject has an increased binding of an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 to the surface of intact cells of the cancer relative to its binding to the surface of intact normal cells (or relative to its binding to the surface of intact cells of the cancer prior to administration of an inhibitor of an immune checkpoint molecule).
  • an anti-M(H)DM2/4 e.g., anti-HDM22
  • the cancer is a bladder cancer, a breast cancer (e.g., a triple negative breast cancer), a lung cancer, a colon cancer, a melanoma, a sarcoma (e.g., an endometrial stromal sarcoma), an ovarian cancer, a glioma, a head and neck cancer, an urothelial cancer, a pancreatic cancer, or a squamous cell carcinoma of the hypopharynx.
  • a breast cancer e.g., a triple negative breast cancer
  • a lung cancer e.g., a colon cancer
  • a melanoma e.g., a sarcoma (e.g., an endometrial stromal sarcoma)
  • an ovarian cancer e.g., a glioma, a head and neck cancer
  • an urothelial cancer e.g., a pancreatic cancer
  • an anti-M(H)DM2/4 e.g., anti-HDM2
  • another therapy such as immunotherapy (e.g., an inhibitor of an inhibitory checkpoint molecule), a chemotherapy, a small molecule inhibitor (such as an inhibitor of one or more of: EGFR, KRAS, STK11, ALK, BRAF, ERBB2, RET, ROS1, B2M, HLA, POLE, IGF-1, ERK/MAPK,
  • an anti-M(H)DM2/4 e.g., anti-HDM2 antibody or fragment described herein is administered as a monotherapy.
  • the invention provides for treatment of a cancer that is resistant to another cancer therapy or therapies (e.g., vaccine, targeted therapy (such as small molecule targeted therapy), chemotherapy, radiotherapy, or immunotherapy (such as treatment with another monoclonal antibody).
  • another cancer therapy or therapies e.g., vaccine, targeted therapy (such as small molecule targeted therapy), chemotherapy, radiotherapy, or immunotherapy (such as treatment with another monoclonal antibody).
  • the invention provides for treating a cancer resistant to chemotherapy (i.e., resistant to one or more chemotherapeutic drugs).
  • the invention provides for treating a cancer resistant to treatment with another monoclonal antibody or antibodies.
  • the invention provides for treating a cancer resistant to radiotherapy.
  • the invention provides for treating a cancer resistant to small molecule targeted therapy or therapies.
  • an anti-M(H)DM2/4 e.g., anti-HDM2
  • an anti-HDM2 antibody or antigen-binding fragment thereof in a patient who has cancer (e.g., has been diagnosed with cancer).
  • therapeutic use of an anti- HDM2 antibody (or a fragment thereof) in a patient who has cancer that is known to metastasize i.e., is a type of cancer that is commonly known to become metastatic cancer.
  • the patient being treated has metastatic cancer.
  • a patient with a cancer that has not metastasized is treated in accordance with a method described herein in order to prevent metastasis of the cancer.
  • the patient being treated has been previously treated with other cancer therapies (e.g. vaccine, targeted therapy,
  • the patient with a cancer that has metastasized is treated in accordance with a method described herein in order to reduce, slow down or stop metastases, or decrease the number or size of metastases of the cancer.
  • the methods described herein are suitable for treating cancers that are expected, known or determined to express anti-M(H)DM2/4 (e.g., HDM2) on the surface of their cells.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein can be one of the splice variants of HDM2 protein known in the art or described herein. Without intending to be bound by a mechanism of action, it is believed that HDM2 on the surface of cancer cells is usually a splice variant of the HDM2 protein that lacks at least one or all nuclear localization signals (e.g., the nuclear localization sequence at the N-terminal portion of HDM2, the nuclear localization signal at the C-terminal portion of HDM2, or both nuclear localization signals).
  • nuclear localization signals e.g., the nuclear localization sequence at the N-terminal portion of HDM2, the nuclear localization signal at the C-terminal portion of HDM2, or both nuclear localization signals.
  • HDM2 on the surface of cancer cells is a splice variant of the HDM2 protein that lacks at least one or all nuclear localization signals, and further lacks a nuclear export signal.
  • the HDM2 on the surface of cancer cells is HDM2 that lacks the nuclear localization signal at amino acids 179-185 of SEQ ID NO: 4 (i.e., lacks amino acids 179 to 185 of SEQ ID NO: 4).
  • the HDM2 on the surface of cancer cells is HDM2 that lacks the nuclear localization signal at amino acids 466-473 of SEQ ID NO: 4 (i.e., lacks amino acids 466 to 473 of SEQ ID NO: 4).
  • the HDM2 on the surface of cancer cells is HDM2 that lacks the nuclear localization signals at amino acids 179-185 and amino acids 466-473 of SEQ ID NO: 4 (i.e., lacks amino acids 179 to 185 and 466 to 473 of SEQ ID NO: 4).
  • the HDM2 on the surface of cancer cells is HDM2 that lacks the nuclear export signal, such as the nuclear export signal at amino acids 190-202 of SEQ ID NO: 4 (i.e., lacks amino acids 190 to 202 of SEQ ID NO: 4).
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein, such as one of the splice variants known in the art or described herein (see Table 1 and Table 2, and Section 8, for the list of HDM2 variants).
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-A (SEQ ID NO: 8), which lacks amino acids 28-222 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-A1 (SEQ ID NO: 9), which lacks amino acids 28-222 and 275-300 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-B (SEQ ID NO: 10), which lacks amino acids 28-300 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-C (SEQ ID NO: 11), which lacks amino acids 53-222 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-D (SEQ ID NO:12 ), which lacks amino acids 30-388 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-E (SEQ ID NO: 13), which lacks amino acids 76-102 and 103-491 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-F (SEQ ID NO: 14), which lacks amino acids 53-97 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-G (SEQ ID NO: 15), which lacks amino acids 115-169 of SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-11 (SEQ ID NO: 16), in which amino acid M has been substituted with M ® MVRSRQM in SEQ ID NO: 4.
  • the HDM2 on the surface of cancer cells targeted by the methods described herein is a splice variant of the HDM2 protein known as MDM2-KB2 (SEQ ID NO: 17), which lacks amino acids 157-248 of SEQ ID NO: 4.
  • the methods described herein target a splice variant of M(H)DM4 on the surface of cancer cells, for example, target one or more of the following splice variants: MDMX-S, MDM4-S, MDM4-A, MDM4-G, MDM4-XALT1/XALT2 and MDM4-211 (or a human equivalent of the listed splice variants).
  • the anti-M(H)DM2/4 e.g., anti-HDM2
  • anti-HDM2 anti-HDM2
  • This binding to HDM2 can be shown by, for example, the ability of the anti-HDM2 antibody or fragment to bind to an intact cancer cell of the tissue type of the tissue of origin of the cancer being treated (which can be but does not need to be from the subject being treated).
  • an anti-HDM2 antibody or a fragment thereof is tested and determined to be expected to bind to the intact cells of the cancer of a subject, before administering the antibody or fragment to the subject. This testing can be done, e.g., by showing binding of the anti-HDM2 antibody or fragment to the surface of intact cancer cells obtained by biopsy or to a cancer cell line of the appropriate tissue type.
  • the cancer cells of the prospective patient to be treated can be tested for expression of M(H)DM2/4 (e.g., HDM2) on their surface using techniques known in the art in order to determine whether the subject is an appropriate candidate for anti-HDM2 therapy described herein; however, such ordinarily would not be deemed necessary if the patient has a cancer of a tissue type that is known or expected to have an extracellularly accessible epitope of M(H)DM2/4.
  • M(H)DM2/4 e.g., HDM2
  • the cells of the cancer in the subject being treated have been tested and determined to have an extracellularly accessible epitope of HDM2 (targeted by the anti-HDM2 antibody or fragment thereof) exposed on their plasma membrane surface (e.g., determined to express a variant of HDM2 that is known to have this extracellular region exposed on the plasma membrane surface).
  • the cancer being treated using the methods described herein is a cancer that is known or determined to express a splice variant of HDM2 (for example, MDM2-A (SEQ ID NO: 8), MDM2-A1 (SEQ ID NO: 9), MDM2-B (SEQ ID NO: 10), MDM2-C (SEQ ID NO: 11), MDM2-D (SEQ ID NO:12 ), MDM2-E (SEQ ID NO: 13), MDM2-F (SEQ ID NO: 14), MDM2-G (SEQ ID NO: 15), MDM2-11 (SEQ ID NO: 16) or MDM-KB2 (SEQ ID NO: 17)), on the plasma membrane surface of its cells.
  • a splice variant of HDM2 for example, MDM2-A (SEQ ID NO: 8), MDM2-A1 (SEQ ID NO: 9), MDM2-B (SEQ ID NO: 10), MDM2-C (SEQ ID NO: 11), MDM2-D (SEQ ID NO:12 ), MDM2-E (S
  • an anti-M(H)DM2/4 e.g., anti- HDM2
  • an antigen-binding fragment thereof in accordance with the methods described herein can be carried out to achieve, or found to result in achieving, at least one, two, three, four or more of the following effects (e.g., in a subject with a cancerous tumor): (i) a decrease in tumor size (e.g., volume or diameter), (ii) a reduction in the growth of the tumor, (iii) inhibition of the progression of tumor growth, (iv) the regression of the tumor, (v) inhibition of a recurrence of the tumor, (vi) eradication of the tumor (e.g., primary tumor or metastatic tumor), (vii) prevention of metastasis of the tumor, (vii) reduction in the number, size or invasiveness of the metastases of the tumor, (viii) reduction or amelioration of the severity or duration of one or more symptoms of the tumor, (ix)
  • a decrease in tumor size e.g
  • the tumor size can be assessed using magnetic resonance imaging (MRI), dynamic contrast-enhanced MRI (DCE-MRI), X-ray, computed tomography (CT) scan, or positron emission tomography (PET) scan.
  • MRI magnetic resonance imaging
  • DCE-MRI dynamic contrast-enhanced MRI
  • CT computed tomography
  • PET positron emission tomography
  • the administration of an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or an antigen-binding fragment thereof in accordance with the methods described herein is effective to treat cancer in a subject (e.g., reduces tumor volume or diameter, reduces tumor growth, reduce tumor proliferation, eradicates the tumor, or improves one or more symptoms of cancer), when used alone or in combination with another therapy.
  • the administration of an anti-HDM2 antibody or an antigen-binding fragment thereof in accordance with the methods described herein is effective to prevent metastases in a subject that has cancer, when used alone or in combination with another therapy.
  • an anti-HDM2 antibody or an antigen-binding fragment thereof in accordance with the methods described herein is effective to treat a metastatic cancer (e.g., reduces the number, size or invasiveness of metastases, or eradicates metastases), when used alone or in combination with another therapy.
  • the administration of an anti-M(H)DM2/4 (e.g., anti- HDM2) antibody or an antigen-binding fragment thereof in accordance with the methods described herein is effective to treat cancer or prevent metastasis in a subject when used alone (i.e., without an additional therapy).
  • the administration of an anti-HDM2 antibody or an antigen-binding fragment thereof in accordance with the methods described herein is effective to treat cancer or prevent metastasis in a subject when used in combination with one or more of the additional therapies described herein.
  • the effectiveness of therapies described herein can be assessed by evaluating a parameter (e.g., tumor size) before and after administration of the therapies described herein to the subject being treated.
  • a parameter e.g., tumor size
  • the effectiveness of therapy can be assessed by evaluating a parameter (e.g., tumor size) before and after administration of the therapies described herein to an animal model (e.g., in an animal model, such as a mouse model, a rat model, or a hamster model, of the cancer being treated). Any assay known in the art can be used to evaluate the therapeutic effectiveness of the therapies described herein.
  • cancers that can be treated in accordance with the methods described herein include, but are not limited to, breast cancer, cervical cancer, ovarian cancer, endometrial cancer, uterine cancer, pancreatic cancer, skin cancer (e.g., melanoma), prostate cancer (e.g., hormone refractory, such as castration resistant, prostate cancer), lung cancer (e.g., small-cell lung cancer, or non-small cell lung cancer), colorectal cancer (e.g., colon cancer, or rectal cancer), gastrointestinal cancer, stomach cancer, small bowel cancer, appendix cancer, esophageal cancer, gastric cancer, renal cancer, bladder cancer, gallbladder cancer, kidney cancer (e.g., renal cell carcinoma, or Wilms tumor)), liver cancer (e.g., hepatic carcinoma, or hepatoma), central nervous system cancer (e.g., brain cancer), peripheral nervous system cancer, bronchial cancer, cancer of the oral cavity or pharynx (e.g., breast cancer, cervical
  • the cancer is cervical cancer, endometrial cancer, ovarian cancer, pancreatic cancer, melanoma, breast cancer, or colon cancer.
  • the cancer is a pancreatic cancer.
  • the cancer is a melanoma.
  • the cancer is a breast cancer.
  • the cancer is an ovarian cancer.
  • the cancer that can be treated in accordance with the methods described herein is resistant to another cancer therapy or therapies (e.g., vaccine, targeted therapy (such as small molecule targeted therapy), chemotherapy, radiotherapy, or immunotherapy (such as treatment with another monoclonal antibody)).
  • the cancer that can be treated in accordance with the methods described herein is resistant to chemotherapy.
  • the cancer that can be treated in accordance with the methods described herein is resistant to treatment with another monoclonal antibody or antibodies.
  • the cancer that can be treated in accordance with the methods described herein is resistant to radiation.
  • the cancer that can be treated in accordance with the methods described herein is resistant to small molecule targeted therapy.
  • the cancer treated in accordance with the methods described herein is a solid cancer.
  • the cancer treated in accordance with the methods described herein is a non-solid cancer (e.g., hematologic cancer).
  • the cancer treated in accordance with the invention is leukemia (e.g., acute leukemia (such as acute lymphocytic leukemia or acute myelocytic leukemia), chronic leukemia (such as chronic myelocytic leukemia or chronic lymphocytic leukemia), or hairy cell leukemia), lymphoma (e.g., Hodgkin's lymphoma, non-Hodgkin's lymphoma, B-cell lymphoma, Burkitt’s lymphoma, follicular lymphoma, lymphoblastic lymphoma, mantle cell lymphoma, or T-cell lymphoma).
  • acute leukemia such as acute lymphocytic leukemia or acute myelocytic leukemia
  • chronic leukemia such as chronic myelocytic leukemia or chronic lymphocytic leukemia
  • hairy cell leukemia e.g., lymphoma (e.g., Hodgkin's lymph
  • the cancer treated in accordance with the invention is carcinoma (e.g., adenocarcinoma, basal cell carcinoma, renal cell carcinoma, squamous cell carcinoma, osteocarcinoma, thyoma/thymic carcinoma, or choriocarcinoma), blastoma, sarcoma (e.g., soft tissue sarcoma, osteosarcoma, chondrosarcoma, rhabdomyosarcoma, or synovia sarcoma), lymphoma, leukemia, a germ cell tumor, myeloma (e.g., multiple myeloma), squamous cell cancer, mesothelioma, glioblastoma (e.g., glioblastoma multiforme), glioma, neuroblastoma, melanoma, astrocytoma, medulloblastoma, hepatoma
  • carcinoma e.g.,
  • craniopharyngioma ependymoma, pinealoma, hemangioblastoma, neuroma, oligodendroglioma, meningioma, or retinoblastoma.
  • the cancer treated in accordance with the methods described herein is a sarcoma or carcinoma, e.g., fibrosarcoma, myxosarcoma, liposarcoma,
  • chondrosarcoma chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma,
  • lymphangiosarcoma lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, endometrial stromal sarcoma, mast cell sarcoma, adult soft tissue sarcoma, uterine sarcoma, Kaposi sarcoma, merkel cell carcinoma, urothelial carcinoma, colon carcinoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary
  • adenocarcinomas cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, bile duct carcinoma, embryonal carcinoma, lung carcinoma (e.g., small cell lung carcinoma), bladder carcinoma, or epithelial carcinoma.
  • the cancer treated in accordance with the methods of the invention is metastatic.
  • the cancer treated in accordance with the methods described herein is a metastatic melanoma, a metastatic ovarian cancer, a metastatic cervical cancer, a metastatic endometrial cancer, a metastatic pancreatic cancer, a metastatic breast cancer, a metastatic colon cancer, or a metastatic brain cancer. 7.6.2 Methods of Administration
  • anti-M(H)DM2/4 e.g., anti-HDM2
  • antibodies or fragments described herein can be administered to a subject by any suitable means which include, but are not limited to, parenteral (e.g., intravenous, intraarterial, intramuscular, intraosseous, intracerebral, intracerebroventricular, intrathecal, subcutaneous), intraperitoneal, intratumoral, intrapulmonary, intradermal, transdermal, conjunctival, intraocular, intranasal, intratracheal, oral and local intralesional routes of administration.
  • parenteral e.g., intravenous, intraarterial, intramuscular, intraosseous, intracerebral, intracerebroventricular, intrathecal, subcutaneous
  • intraperitoneal intratumoral
  • intrapulmonary intradermal
  • transdermal transdermal
  • conjunctival intraocular
  • intranasal intratracheal
  • oral and local intralesional routes of administration e.g., parent
  • the anti-HDM2 antibodies or fragments described herein are administered intravenously, intraarterially, intramuscularly, intraperitoneally, intratumorally, or subcutaneously. In one embodiment, the anti-HDM2 antibodies or fragments described herein are administered intravenously. In one embodiment, the anti-HDM2 antibodies or fragments described herein are administered intraperitoneally. In one embodiment, the anti-HDM2 antibodies or fragments described herein are administered intramuscularly. In one embodiment, the anti-HDM2 antibodies or fragments described herein are administered subcutaneously. In one embodiment, the anti-HDM2 antibodies or fragments described herein are administered intratumorally (such as by an injection into the tumor of the cancer being treated). In particular embodiments, the anti-HDM2 antibodies or fragments described herein are administered intravenously, intraperitoneally, or intratumorally.
  • nano-particles coated with an anti-M(H)DM2/4 e.g., anti- HDM2
  • an anti-M(H)DM2/4 e.g., anti- HDM2
  • an anti-M(H)DM2/4 e.g., anti- HDM2
  • Arruebo et al. 2009, J. of Nanomater.2009:Article ID 439389, regarding nano-particle coating with antibodies.
  • provided herein are methods for treating cancer or preventing metastasis in a subject having a cancer comprising administering to the subject nano- particles coated with an an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or a fragment thereof.
  • an anti-M(H)DM2/4 e.g., anti-HDM2
  • Various dosing schedules of the anti-M(H)DM2/4 e.g., anti-HDM2
  • methods of administration include, without limitation, bolus administration, pulse infusions, and continuous infusions.
  • the therapeutic regimen for use in the methods described herein may include administration of anti-M(H)DM2/4 (e.g., anti-HDM2) antibodies or fragments thereof (and compositions comprising such antibodies) once every week, once every two weeks, once every three weeks, once every four weeks, once every six weeks, once every eight weeks or once every twelve weeks (e.g., such that the subject receives from at least two, at least three, at least four, at least five, at least six, at least eight, or at least ten doses of the antibody, or from two to twenty doses of the antibody).
  • anti-M(H)DM2/4 e.g., anti-HDM2
  • compositions comprising such antibodies once every week, once every two weeks, once every three weeks, once every four weeks, once every six weeks, once every eight weeks or once every twelve weeks (e.g., such that the subject receives from at least two, at least three, at least four, at least five, at least six, at least eight, or at least ten doses of the antibody, or from
  • anti-HDM2 antibodies or fragments thereof are administered daily, every other day, or two, three, or four times a week (e.g., for a period of time, such as one week, two weeks, three weeks, four weeks, six weeks, two months or three months).
  • the treatment regimens contemplated herein include regimens wherein the initial higher dose of the antibody may be followed by one or more lower doses, or wherein the initial lower dose of the antibody is followed by one or more higher doses.
  • An exemplary treatment course in which the anti-HDM2 antibody or fragment is administered) may last for one week, two weeks, three weeks, four weeks, six weeks, two months, three months, four months, five months, six months, one year, or over several years.
  • a withdrawal period in which the antibody is not administered for, e.g., a week, two weeks, three weeks, four weeks, six weeks, two months, three months, four months, six months or one year
  • a second treatment period where the antibody is administered, e.g., once a month, once in two weeks, once a week, twice a week or three times a week.
  • Such initial treatment and such second treatment periods can last, for example, two weeks, three weeks, four weeks, six weeks, two months, three months, four months, or six months (where the initial treatment period can be the same or different from the second treatment period).
  • This course of treatment (having the initial treatment period, a withdrawal period and a second treatment period) can be repeated twice, three times, four times, five times, six times, ten times or more than ten times.
  • two or more antibodies or fragments thereof with different binding specificities for M(H)DM2/4 are administered simultaneously or sequentially to the subject being treated.
  • anti-M(H)DM2/4 e.g., anti-HDM2
  • the appropriate dosage of anti-M(H)DM2/4 (e.g., anti-HDM2) antibodies or fragments for use in the methods described herein will depend on the type of antibody used, the type of cancer being treated, the severity of the cancer being treated, the route of administration, the target site, the condition of the patient (e.g., age, body weight, health), the responsiveness of the patient to the antibody, other medications used by the patient, and other factors to be considered at the discretion of the medical practitioner performing the treatment.
  • the dosage of an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein which is administered to the subject can be from about 1 ⁇ g/kg to 200 mg/kg of the patient’s body weight.
  • the dosage of an anti- M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein which is administered to the subject can be from about 1 ⁇ g/kg to 100 mg/kg of the patient’s body weight (e.g., from about 0.01 mg/kg to about 100 mg/kg, from about 0.05 mg/kg to about 100 mg/kg, or from about 0.5 mg/kg to about 100 mg/kg).
  • the dosage of an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein which is administered to the subject can be from about 1 mg/kg to 200 mg/kg of the patient’s body weight. In one embodiment, the dosage of an anti-HDM2 antibody or fragment described herein which is administered to the subject is from 0.025 mg/kg to about 5 mg/kg. In one embodiment, the dosage of an anti- HDM2 antibody or fragment described herein which is administered to the subject is from 0.05 mg/kg to about 2 mg/kg. In one embodiment, the dosage of an anti-HDM2 antibody or fragment described herein which is administered to the subject is from 5 mg/kg to about 30 mg/kg.
  • an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or fragment described herein which is administered to the subject can be from about 1 mg/kg to 200 mg/kg of the patient’s body weight. In one embodiment, the dosage of an anti-HDM2 antibody or fragment described herein which is administered to the
  • doses e.g., one or more doses of about 0.025 mg/kg, 0.05 mg/kg, 0.1 mg/kg, 0.2 mg/kg, 0.3 mg/kg, 0.4 mg/kg, 0.5 mg/kg, 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, 7.5 mg/kg, 10 mg/kg, 20 mg/kg, 30 mg/kg, 40 mg/kg, 50 mg/kg, 75 mg/kg, or 100 mg/kg of an anti-HDM2 antibody or fragment described herein can be administered to the subject being treated.
  • a dose (e.g., one or more doses) of about 0.1 mg/kg of an anti- HDM2 antibody or fragment described herein can be administered to the subject being treated (e.g., when the antibody is administered intratumorally). 7.7 Diagnostic, Companion Diagnostic and Prognostic Methods
  • kits for diagnosing cancer in a subject comprising: (a) detecting whether an antibody or a fragment thereof (e.g., a labeled antibody or fragment) that specifically binds to M(H)DM2/4 (e.g., HDM2) binds to the surface of an intact cell of the subject, wherein the antibody or fragment is any anti-M(H)DM2/4 antibody or fragment described herein (in particular, any antibody or fragment that specifically binds to SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3); and (b) diagnosing the subject with cancer if the binding is detected in step (a).
  • an antibody or a fragment thereof e.g., a labeled antibody or fragment
  • M(H)DM2/4 e.g., HDM2
  • the method of diagnosing further comprises, before the detecting in step (a), obtaining the intact cell from the subject, and then performing the detecting by, e.g., determining whether a labeled antibody or fragment binds to the intact cell from the subject using, e.g., FACS or cell-based ELISA analysis.
  • the method of diagnosing comprises administering the antibody or fragment to the subject before the detecting in step (a), and wherein the detecting is performed by in vivo imaging of the subject.
  • a patient is selected for treatment using an anti-M(H)DM2/4 (e.g., anti- HDM2) antibody or antibody fragment described herein based on the detection of binding of such antibody or fragment to the surface of intact cancer cells obtained from the patient.
  • an anti-M(H)DM2/4 e.g., anti- HDM2
  • antibody or antibody fragment described herein based on the detection of binding of such antibody or fragment to the surface of intact cancer cells obtained from the patient.
  • a method of selecting a patient having a cancer for treatment with an antibody or fragment that specifically binds to an extracellularly accessible epitope of HDM2 comprising: obtaining an intact cancer cell from the patient (e.g., by biopsy of the cancerous tumor in the patient, or by obtaining a blood sample with circulating cancer cells from the patient), and determining whether the antibody or fragment binds to the surface of the intact cancer cell of the patient (using any method known in the art or described herein, e.g., using cell-based ELISA or FACS analysis), wherein the detection of binding indicates that the patient can be treated with the antibody or fragment.
  • a method of selecting a patient having a cancer for treatment with an antibody or fragment that specifically binds to an extracellularly accessible epitope of HDM2 comprising: obtaining an intact cancer cell from the patient (e.g., by biopsy of the cancerous tumor in the patient, or by obtaining a blood sample with circulating cancer cells from the patient), determining whether the antibody or fragment binds to the surface of the intact cancer cell of the patient (using any method known in the art or described herein, e.g., using cell-based ELISA or FACS analysis), and, if the binding is detected, administering the antibody or fragment to the patient.
  • the antibody or fragment administered to the patient can be the same or different from the antibody or fragment used for selection of the patient to be treated.
  • a method of selecting a patient having an ovarian cancer for treatment with an antibody or fragment that specifically binds to an extracellularly accessible epitope of HDM2 comprising: obtaining an intact ovarian cancer cell from the patient, determining whether the antibody or fragment binds to the surface of the intact cancer cell of the patient, and, if the binding is detected, administering the antibody or fragment to the patient.
  • any antibody or a fragment thereof that specifically binds to SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3 is used for such patient selection and/or treatment.
  • any antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, which is not bound to a cell-penetrating peptide is used for such patient selection and/or treatment.
  • any antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4, which is not bound to a cell-penetrating peptide is used for such patient selection, and any antibody or a fragment thereof that specifically binds to SEQ ID NO:1, SEQ ID NO:2, or SEQ ID NO:3 is used for the treatment of the patient.
  • a hyper-progressive disease such as diagnosing a subject as exhibiting hyper-progression, or likely to exhibit hyper- progression, of a cancer in response to administration of an inhibitor of an inhibitory checkpoint molecule
  • subject e.g., a human who has a cancer
  • said method comprising: (a) optionally, determininig
  • the method of diagnosing further comprises, before step (b), obtaining (e.g., isolating) the intact cell from the subject, and then performing the determining in step (b) by, e.g., determining whether a labeled antibody or fragment binds to the intact cell from the subject using, e.g., FACS or cell-based ELISA analysis.
  • the method of diagnosing comprises administering the antibody or fragment to the subject before step (b), and wherein the determining in step (b) is performed by in vivo imaging of the subject.
  • the determining in step (a) is performed using PCR, Next Generation Sequencing or RNA sequencing.
  • the gene amplification is determined to be present in step (a) if the cancer cells of the subject have 8 or more than 8 copies of
  • the gene amplification is determined to be present in step (a) if the cancer cells of the subject have 50% or more than 50% amplification of the M(H)DM2/4 gene relative to the normal cells of the subject (or relative to the cells of the subject prior to administration of an inhibitor of an inhibitory checkpoint molecule).
  • the gene amplification of M(H)DM2/4 is gene amplification of HDM2. In one embodiment, the gene amplification of M(H)DM2/4 is gene amplification of HDM4. In one embodiment, the gene amplification of M(H)DM2/4 is gene amplification of HDM2 and HDM4.
  • Non-limiting exemplary samples that can be used for in diagnostic or patient selection methods using an anti-M(H)DM2/4 antibody (e.g., anti-HDM2 antibody) or fragment thereof described herein include: tissue biopsies, intact cells obtained from malignant tissues, and circulating cancer cells isolated from blood.
  • tissue sample can be obtained from a patient and immunohistochemistry can be performed to detect whether a labeled anti- M(H)DM2/4 antibody (e.g., anti-HDM2 antibody) or fragment thereof binds to the tissue sample.
  • intact cells can be isolated from a patient and FACS or cell-based ELISA analysis can be performed to detect whether a labeled anti- M(H)DM2/4 antibody (e.g., anti-HDM2 antibody) or fragment thereof binds to such cell.
  • a blood sample with circulating cancer cells can be obtained from a patient and FACS or cell-based ELISA analysis can be performed to detect whether a labeled anti- M(H)DM2/4 antibody (e.g., anti-HDM2 antibody) or fragment thereof binds to such cells.
  • the duration of treatment and/or dosage of an anti-M(H)DM2/4 (e.g., anti-HDM2) antibody or antibody fragment described herein to be used in the treatment of a patient is determined based on the detection of binding of such antibody or fragment to the surface of intact cancer cells obtained from the patient.
  • a method of determining whether to continue the treatment of a patient having a cancer with an antibody or fragment that specifically binds to an extracellularly accessible epitope of HDM2 comprising: administering the antibody or fragment to the patient for a first period of time (e.g., where the patient had been selected for treatment as described above), obtaining an intact cancer cell from the patient (e.g., by biopsy of the cancerous tumor in the patient, or by obtaining a blood sample with circulating cancer cells from the patient), and determining whether the antibody or fragment binds to the surface of the intact cancer cell of the patient (using any method known in the art or described herein, e.g., using cell-based ELISA or FACS analysis), and, if the binding is detected, continuing administering the antibody or fragment to the patient for a second period of time (but, e.g., if the binding is not detected, discontinuing the treatment).
  • a first period of time e.g., where the patient had been selected for treatment as described above
  • a method of determining whether to increase the dose of an antibody or fragment that specifically binds to an extracellularly accessible epitope of HDM2 for use in the treatment of a patient having a cancer comprising: administering a dose the antibody or fragment to the patient for a period of time (e.g., where the patient had been selected for treatment as described above), obtaining an intact cancer cell from the patient (e.g., by biopsy of the cancerous tumor in the patient, or by obtaining a blood sample with circulating cancer cells from the patient), and determining whether the antibody or fragment binds to the surface of the intact cancer cell of the patient (using any method known in the art or described herein, e.g., using cell-based ELISA or FACS analysis), and, if the binding is detected, administering a dose of the antibody or fragment to the patient for a second period of time, wherein the dose administered during the second period of time is higher than the dose administered during the first period of time (e.g., two times, or three
  • treatment with an anti-M(H)DM2/4 e.g., anti- HDM2
  • an anti-M(H)DM2/4 e.g., anti- HDM2
  • an antibody fragment described herein is monitored in a patient by determining the amount of HDM2 expressed on the surface of cancer cells obtained from the patient, before and after treatment, wherein a decrease in the amount is a positive prognosis.
  • the patients or subjects being treated in accordance with the methods described herein include, but are not limited to, humans and non-human vertebrates. In certain aspects,
  • the subject being treated is a mammal, e.g., a human, a dog, a cat, a monkey, a rabbit, a cow, a horse, a goat, a sheep, or a pig.
  • the subject being treated is a human.
  • the subject being treated in accordance with the methods described herein has been diagnosed with a cancer (e.g., using a biopsy or any another method known in the art).
  • the subject being treated has been diagnosed with an early stage cancer.
  • the subject being treated has been diagnosed with an advanced stage cancer.
  • the subject being treated has been diagnosed with a high-grade tumor.
  • the subject being treated has been diagnosed with a low-grade tumor.
  • the subject being treated has been diagnosed with a cancer that can metastasize.
  • the subject being treated has been diagnosed with a metastatic cancer.
  • the subject being treated in accordance with the methods described herein has been diagnosed with a cervical cancer, an endometrial cancer, an ovarian cancer, a pancreatic cancer, a melanoma, a breast cancer, a colorectal cancer (e.g., a colon cancer), a bladder cancer, an astrocytic neoplasm, a glioblastoma, a pediatric
  • the subject being treated in accordance with the methods described herein has been diagnosed with a melanoma, a pancreatic cancer, a breast cancer, or an ovarian cancer.
  • the subject being treated in accordance with the methods described herein has been diagnosed with a melanoma.
  • the subject being treated in accordance with the methods described herein has been diagnosed with a pancreatic cancer.
  • the subject being treated in accordance with the methods described herein has been diagnosed with a breast cancer.
  • the subject being treated in accordance with the methods described herein has been diagnosed with an ovarian cancer.
  • the subject being treated in accordance with the methods described herein has been diagnosed with a lung cancer.
  • the subject being treated is a hyper-progressor in response to treatment with an inhibitor of an inhibitory immune checkpoint molecule (e.g., the subject has been diagnosed with hyper-progression in response to treatment with an inhibitor of an inhibitory immune checkpoint molecule).
  • the subject being treated is at risk of being a hyper-progressor in response to treatment with an inhibitor of an inhibitory immune checkpoint molecule (e.g., the subject has a gene amplification of M(H)DM2/4, and/or the subject has an increased binding of an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 to the surface of intact cells of the cancer relative to its binding to the surface of intact normal cells or relative to its binding to the surface of intact cells of the cancer prior to administration of the inhibitor).
  • an inhibitor of an inhibitory immune checkpoint molecule e.g., the subject has a gene amplification of M(H)DM2/4, and/or the subject has an increased binding of an antibody or a fragment thereof that specifically binds to an extracellularly accessible epitope of M(H)DM2/4 to the surface of intact cells of the cancer relative to its binding to the surface of intact normal cells or relative to its binding to the surface of intact cells of the cancer prior to administration of the inhibitor.
  • the subject being treated has previously undergone one or more other cancer therapies (e.g., vaccine, targeted therapy (such as small molecule targeted therapy), chemotherapy, radiotherapy, or immunotherapy (such as treatment with another monoclonal antibody)), and the subject’s cancer has developed resistance to the one or more other cancer therapies.
  • the subject being treated is resistant to one or more other cancer therapies.
  • the subject being treated is resistant to radiotherapy. In one embodiment, the subject being treated is resistant to a small molecule targeted therapy. In one embodiment, the subject being treated is resistant to treatment with another monoclonal antibody.
  • the subject being treated has a type of a cancer that is known or expected to have M(H)DM2/4 (e.g., HDM2) on the surface of its cells.
  • M(H)DM2/4 e.g., HDM2
  • the subject being treated has a type of cancer, the cells of which express one or more of splice variants of HDM2 on their cell surface, for example (and without limitation), one or more of the following splice variants: MDM2-A (SEQ ID NO: 8), MDM2-A1 (SEQ ID NO: 9), MDM2-B (SEQ ID NO: 10), MDM2-C (SEQ ID NO: 11), MDM2-D (SEQ ID NO:12 ), MDM2-E (SEQ ID NO: 13), MDM2-F (SEQ ID NO: 14), MDM2-G (SEQ ID NO: 15), MDM2- 11 (SEQ ID NO: 16) or MDM-KB2 (SEQ ID NO: 17).
  • MDM2-A SEQ ID NO: 8
  • MDM2-A1 SEQ ID NO: 9
  • MDM2-B SEQ ID NO: 10
  • MDM2-C SEQ ID NO: 11
  • MDM2-D SEQ ID NO:12
  • MDM2-E SEQ ID NO: 13
  • the subject being treated has a cancer that has been tested and determined (using any assay known in the art) to carry M(H)DM2/4 (e.g., HDM2) on the plasma membrane of its cells.
  • M(H)DM2/4 e.g., HDM2
  • the subject being treated has a cancer, the cells of which have been tested and determined (by any method known in the art) to expose on their plasma membrane surface an extracellular region of HDM2 that can be targeted by an anti- HDM2 antibody or fragment (and such antibody can then be administered to the subject).
  • the subject being treated has a cancer, the cells of which have been tested and determined (by any method known in the art) to express one or more of splice variants of HDM2 on their cell surface, for example (and without limitation), one or more of the following splice variants: MDM2-A (SEQ ID NO: 8), MDM2-A1 (SEQ ID NO: 9), MDM2-B (SEQ ID NO: 10), MDM2-C (SEQ ID NO: 11), MDM2-D (SEQ ID NO:12 ), MDM2-E (SEQ ID NO: 13), MDM2-F (SEQ ID NO: 14), MDM2-G (SEQ ID NO: 15), MDM2-11 (SEQ ID NO: 16) or MDM-KB2 (SEQ ID NO: 17). 7.9 Combination therapies and kits
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with one or more anti-cancer therapies different from said antibody or fragment , e.g., a chemotherapy, a surgery, a radiation therapy, another antibody with an anti-cancer activity, a cytokine, a T cell therapy, a vaccine (e.g., a cellular vaccine), a small molecule with an anti-cancer activity, an anti-hormonal agent, or any other anti-cancer therapy known in the art.
  • a chemotherapy e.g., a surgery, a radiation therapy, another antibody with an anti-cancer activity, a cytokine, a T cell therapy, a vaccine (e.g., a cellular vaccine), a small molecule with an anti-cancer activity, an anti-hormonal agent, or any other anti-cancer therapy known in the art.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with chemotherapy.
  • chemotherapeutic agents include, without limitation, an alkylating agent, a nitrosourea agent, an antimetabolite, a topoisomerase inhibitor, an aromatase inhibitor, an antitumor antibiotic, an alkaloid derived from a plant, a hormone antagonist, a P-glycoprotein inhibitor, and a platimum complex derivative.
  • chemotherapeutic drugs that can be used in the methods described herein include, without limitation, taxol, paclitaxel, nab-paclitaxel, 5-fluorouracil (5-FU), gemcitabine, doxorubicin, daunorubicin, colchicin, mitoxantrone, tamoxifen, cyclophosphamide,
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is an anti- M(H)DM2/4 antibody or fragment thereof described herein.
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is
  • the cancer treated using a combination therapy described herein is a pancreatic cancer, a breast cancer, a lung cancer or an ovarian cancer.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with gemcitabine (e.g., for treatment of a non- small cell lung cancer, a pancreatic cancer or an ovarian cancer).
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with capecitabine.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with irinotecan.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with celecoxib.
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with paclitaxel (e.g., paclitaxel formulated as albumin-bound particles such as ABRAXANE®) (e.g., for treatment of a metasatic breast cancer).
  • paclitaxel e.g., paclitaxel formulated as albumin-bound particles such as ABRAXANE®
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with nab- paclitaxel.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with cisplatin.
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with 5-FU (e.g., for treatment of a colorectal cancer such as a colon cancer).
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with carboplatin.
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with gemcitabine and carboplatin (e.g., for treatment of an ovarian cancer).
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with paclitaxel (e.g., paclitaxel formulated as albumin- bound particles such as ABRAXANE®) and gemcitabine (e.g., for treatment of a breast cancer, or a pancreatic cancer such as adenocarcinoma of the pancreas or metastatic adenocarcinoma of the pancreas).
  • paclitaxel e.g., paclitaxel formulated as albumin- bound particles such as ABRAXANE®
  • gemcitabine e.g., for treatment of a breast cancer, or a pancreatic cancer such as adenocarcinoma of the pancreas or metastatic adenocarcinoma of the pancreas.
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with gemcitabine and cisplatin (e.g.
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with gemcitabine and 5-FU.
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with paclitaxel (e.g., paclitaxel formulated as albumin-bound particles such as ABRAXANE ® ) and carboplatin (e.g., for treatment of a non- small cell lung cancer).
  • paclitaxel e.g., paclitaxel formulated as albumin-bound particles such as ABRAXANE ®
  • carboplatin e.g., for treatment of a non- small cell lung cancer.
  • gemcitabine is administered in a dose of 1,500 mg/m 2 .
  • nab-paclitaxel is administered in a dose of 300 mg/m 2 . In certain embodiments, wherein the subject is human, gemcitabine is administered in a dose of 1,000 mg/m 2 . In certain embodiments, wherein the subject is human, nab-paclitaxel is administered in a dose of 125 mg/m 2 .
  • the gemcitabine and/or nab-paclitaxel are administered in doses that are lower than doses used when gemcitabine and/or nab-paclitaxel are administered not in combination with an anti-cancer antibody (such as an anti-M(H)DM2/4 antibody or fragment described herein).
  • an anti-cancer antibody such as an anti-M(H)DM2/4 antibody or fragment described herein.
  • gemcitabine is administered in a dose that is less than 1,500 mg/m 2
  • nab-paclitaxel is administered in a dose that is less than 300 mg/m 2 .
  • gemcitabine is administered in a dose that is less than 1,000 mg/m 2
  • nab-paclitaxel is administered in a dose that is less than 125 mg/m 2
  • gemcitabine is administered in a dose that is equal to or less than 500 mg/m 2 , 400 mg/m 2 , 300 mg/m 2 or 200 mg/m 2
  • the nab-paclitaxel is administered in a dose that is equal to or less than 62.5 mg/m 2 , 50 mg/m 2 , 40 mg/m 2 , 30 mg/m 2 , or 20 mg/m 2 .
  • gemcitabine is administered in a dose that is equal to or less than 900 mg/m 2 , 800 mg/m 2 , 700 mg/m 2 or 600 mg/m 2
  • the nab-paclitaxel is administered in a dose that is equal to or less than 110 mg/m 2 , 100 mg/m 2 , 90 mg/m 2 , 80 mg/m 2 , or 70 mg/m 2
  • gemcitabine and/or nab-paclitaxel are administered with a frequency of every 2 weeks or less (e.g., every 3 weeks, every 4 weeks, every 6 weeks, or every 8 weeks, or less).
  • gemcitabine is administered with a frequency of once a day, 4 times per week, 3 times per week, 2 times per week, or once per week.
  • nab-paclitaxel is administered with a frequency of once a day, 4 times per week, 3 times per week, 2 times per week, or once per week.
  • gemcitabine and nab- paclitaxel are administered with a frequency of once a day, 4 times per week, 3 times per week, 2 times per week, or once per week.
  • gemcitabine and/or nab-paclitazel is administered once a week.
  • the total duration of treatment with gemcitabine and/or nab-paclitaxel is, or is more than, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, or 12 weeks.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject with a cancer in combination with the chemotherapy drug(s) indicated for said cancer, which chemotherapy drug(s) can be optionally administered in the dosage and/or regime of administration indicated for said cancer.
  • chemotherapy drugs as well as their dosage and regime of administration indicated for various cancers are provided below.
  • Ovarian Cancer The following information is taken from Gemzar ® (gemcitabine for injection), Eli Lilly and Company, Highlights of Prescribing Information, revised March 2017, http://pi.lilly.com/us/gemzar.pdf (last accessed on July 27, 2017).
  • Gemcitabine (Gemzar ® ) in combination with carboplatin is indicated for the treatment of patients with advanced ovarian cancer that has relapsed at least 6 months after completion of platinum-based therapy.
  • the recommended dose of Gemzar ® is 1000 mg/m 2 as an intravenous infusion over 30 minutes on Days 1 and 8 of each 21-day cycle, in combination with carboplatin (AUC 4) intravenously after Gemzar ® administration on Day 1 of each 21-day cycle.
  • Non-Small Cell Lung Cancer The following information is taken from Gemzar ® (gemcitabine for injection), Eli Lilly and Company, Highlights of Prescribing Information, revised March 2017, http://pi.lilly.com/us/gemzar.pdf (last accessed on July 27, 2017).
  • Gemcitabine (Gemzar ® ) is indicated in combination with cisplatin for the first-line treatment of patients with inoperable, locally advanced (Stage IIIA or IIIB), or metastatic (Stage IV) non- small cell lung cancer. Every 4-week schedule: the recommended dose of Gemzar ® is 1000 mg/m 2 intravenously over 30 minutes on Days 1, 8, and 15 in combination with cisplatin therapy; cisplatin can be administered intravenously at 100 mg/m 2 on Day 1 after the infusion of Gemzar ® .
  • the recommended dose of Gemzar ® is 1250 mg/m 2 intravenously over 30 minutes on Days 1 and 8 in combination with cisplatin therapy; cisplatin can be administered intravenously at 100 mg/m 2 on Day 1 after the infusion of Gemzar ® .
  • Pancreatic Cancer The following information is taken from Gemzar ® (gemcitabine for injection), Eli Lilly and Company, Highlights of Prescribing Information, revised March 2017, http://pi.lilly.com/us/gemzar.pdf (last accessed on July 27, 2017).
  • Gemcitabine (Gemzar ® ) is indicated as first-line treatment for patients with locally advanced (nonresectable Stage II or Stage III) or metastatic (Stage IV) adenocarcinoma of the pancreas.
  • Gemzar ® is indicated for patients previously treated with 5-FU.
  • the recommended dose of Gemzar ® is 1000 mg/m 2 over 30 minutes intravenously.
  • the recommended treatment schedule is as follows: weeks 1-8 -- weekly dosing for the first 7 weeks followed by one week rest; after week 8 -- weekly dosing on Days 1, 8, and 15 of 28-day cycles.
  • Metastatic Breast Cancer The following information is taken from ABRAXANE ® (paclitaxel protein-bound particles for injectable suspension, albumin-bound), Celgene
  • ABRAXANE ® is indicated for the treatment of breast cancer after failure of combination chemotherapy for metastatic disease or relapse within 6 months of adjuvant chemotherapy. Prior therapy should have included an anthracycline unless clinically contraindicated. After failure of combination chemotherapy for metastatic breast cancer or relapse within 6 months of adjuvant chemotherapy, the recommended regimen for ABRAXANE® is 260 mg/m 2 administered intravenously over 30 minutes every 3 weeks.
  • Non-Small Cell Lung Cancer The following information is taken from
  • ABRAXANE ® paclitaxel protein-bound particles for injectable suspension, albumin-bound
  • Celgene Corporation Highlights of Prescribing Information, revised July 2015,
  • ABRAXANE ® is indicated for the first-line treatment of locally advanced or metastatic non- small cell lung cancer, in combination with carboplatin, in patients who are not candidates for curative surgery or radiation therapy.
  • the recommended dose of ABRAXANE ® is 100 mg/m 2 administered as an intravenous infusion over 30 minutes on Days 1, 8, and 15 of each 21-day cycle.
  • Carboplatin can be administered on Day 1 of each 21 day cycle immediately after ABRAXANE ® .
  • Adenocarcinoma of the Pancreas The following information is taken from
  • ABRAXANE ® paclitaxel protein-bound particles for injectable suspension, albumin-bound
  • Celgene Corporation Highlights of Prescribing Information, revised July 2015,
  • ABRAXANE ® is indicated for the first-line treatment of patients with metastatic
  • ABRAXANE ® adenocarcinoma of the pancreas, in combination with gemcitabine.
  • the recommended dose of ABRAXANE ® is 125 mg/m 2 administered as an intravenous infusion over 30-40 minutes on Days 1, 8 and 15 of each 28-day cycle.
  • Gemcitabine can be administered immediately after ABRAXANE ® on Days 1, 8 and 15 of each 28-day cycle.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with an immunomodulator (e.g., a cytokine, an antigen, or a checkpoint targeting agent).
  • an immunomodulator e.g., a cytokine, an antigen, or a checkpoint targeting agent.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with a checkpoint targeting agent such as, without limitation, an antagonist of PD-1, an antagonist of PD-L1, an antagonist of PD-L2, an antagonist of CTLA-4, an antagonist of TIM-3, an antagonist of GITR, an antagonist of OX40, an antagonist of LAG-3 (e.g., the antagonist of any of the above- mentioned checkpoint molecules can be an antibody, such as an inhibitory antibody to these molecules, an antibody fragment, or a small molecule).
  • a checkpoint targeting agent such as, without limitation, an antagonist of PD-1, an antagonist of PD-L1, an antagonist of PD-L2, an antagonist of CTLA-4, an antagonist of TIM-3, an antagonist of GITR, an antagonist of OX40, an antagonist of LAG-3 (e.g., the antagonist of any of the above- mentioned checkpoint molecules can be an antibody, such as an inhibitory antibody to these molecules, an antibody fragment, or a small molecule).
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with an inhibitor of PD-1, an inhibitor of PD-L1, or an inhibitor of CTLA-4 (where the inhibitor can be an antagonistic antibody, an antibody fragment, or a small molecule).
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with radiation therapy (e.g., x-rays, gamma- rays or another source of radiation).
  • radiation therapy e.g., x-rays, gamma- rays or another source of radiation.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with surgery (such as a surgery to remove part or all of the cancerous tumor being treated).
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with a Treg-inhibitory agent.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with a T-cell therapy.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with a tumor vaccine.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with an EGFR inhibitor (e.g., erlotinib).
  • an EGFR inhibitor e.g., erlotinib
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with an inhibitor of one or more of: EGFR, KRAS, STK11, ALK, BRAF, ERBB2, RET, ROS1, B2M, HLA, POLE, IGF-1,
  • ERK/MAPK PI3K/AKT
  • TGF-b TGF-b
  • DNMT3A IFN g
  • JAK1/JAK2/JAK3A IFN g
  • JAK1/JAK2/JAK3A IFN g
  • JAK1/JAK2/JAK3A IFN g
  • JAK1/JAK2/JAK3A IFN g
  • JAK1/JAK2/JAK3A IFN g
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is administered to a subject in combination with an inhibitor of p53-H(M)DM2 interaction (e.g., a small molecule inhibitor or a peptide inhibitor).
  • an inhibitor of p53-H(M)DM2 interaction e.g., a small molecule inhibitor or a peptide inhibitor.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is used to treat a subject that is not treated with a cell cycle inhibitor (i.e., the additional therapy is not an agent that inhibits cell cycle).
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein is used to treat a subject that is not concurrently (during the same treatment period) treated with a cell cycle inhibitor (i.e., the subject is not treated with an anti-M(H)DM2/4 antibody or fragment thereof and a cell cycle inhibitor during the same period of time, e.g., day or week).
  • an anti- M(H)DM2/4 antibody or fragment thereof described herein is used to treat a subject that has not been previously treated and is not concurrently treated with a cell cycle inhibitor.
  • an anti-M(H)DM2/4 antibody or fragment thereof described herein can be used before, during, or after the second therapy (e.g., a chemotherapy, a radiation therapy, a surgery, or any other therapy described herein or known in the art).
  • the second therapy e.g., a chemotherapy, a radiation therapy, a surgery, or any other therapy described herein or known in the art.
  • the subject being treated in accordance with the methods described herein has not received an anti-cancer therapy prior to the administration of an anti- M(H)DM2/4 antibody or fragment thereof.
  • an anti- M(H)DM2/4 antibody or fragment thereof is administered to a subject that has received an anti-cancer therapy prior to administration of the antibody or fragment.
  • anti- M(H)DM2/4 antibody or fragment thereof is administered to a subject recovering from or receiving an immunosuppressive therapy.
  • kits comprising an anti-M(H)DM2/4 antibody or a fragment thereof, and one or more additional anti-cancer agents.
  • kits comprising (i) an anti-M(H)DM2/4 antibody or a fragment thereof (e.g., in a therapeutically effective amount), and (ii) one or more of chemotherapeutic drugs, for example, gemcitabine, paclitaxel, or gemcitabine and nab-paclitaxel (e.g., in therapeutically effective amounts, such as any amounts described herein, which may be less than the therapeutically effective amount of the drug or drugs when the drug or drugs are used without the anti-M(H)DM2/4 antibody or fragment).
  • chemotherapeutic drugs for example, gemcitabine, paclitaxel, or gemcitabine and nab-paclitaxel
  • HDM2 a segment of HDM2
  • Three (3) different extracellularly accessible epitopes have been identified.
  • Specific segments of HDM2 that are extracellularly accessible include but are not limited to epitopes present in the NMC-P1, NMC-P2 and NMC-P3 peptide sequences (SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3, respectively). These extracellularly accessible sequences are appropriate therapeutic and diagnostic targets for anti-HDM2 antibodies.
  • cancer cells expressing HDM2 on their surface membrane can be targeted with antibodies to HDM2 for diagnostic and therapeutic (i.e., anti-tumor cytotoxic and inhibitory effect) purposes.
  • HDM2-specific antibodies bound to the extracellularly accessible sequences of M(H)DM2/4 on the surface membrane of intact cells of several rodent and human cancer cell lines as well as primary tumor cells from human patients.
  • the same HDM2-specific antibodies exhibited minimal binding to the surface membrane of normal human blood mononuclear cells. It was found that these HDM2-specific antibodies selectively bound to various cancer cells such as: intact human melanoma, uveal melanoma, pancreatic, breast, colon, lung and ovarian cancer cells in vitro.
  • HDM2-specific antibodies inhibited the growth of cancer cells in vitro and in vivo, strongly indicating that they can be used as therapeutic agents in vivo.
  • Data from in vivo studies and described herein showed that select HDM2-specific antibodies inhibited tumor growth and were cytotoxic against tumors in rodent tumor models.
  • select HDM2-specific antibodies were not only cytotoxic to tumor cells but also inhibited tumor growth in mouse models of pancreatic cancer, lung cancer and colon cancer.
  • Data herein further demonstrated that only select antibodies recognized extracellularly accessible epitopes of HDM2.
  • Anti-HDM2 antibodies that specifically bind to (i) NMC-P1, i.e., the peptide of SEQ ID NO:1 (“NMC-100s series of monoclonal antibodies)”, (ii) NMC-P2, i.e., the peptide of SEQ ID NO:2 (“NMC-200s series of monoclonal antibodies”), and (iii) NMC-P3, i.e., the peptide of SEQ ID NO:3 (“NMC-300s series of monoclonal antibodies”) were generated using the hybridoma approach.
  • NMC-P1, NMC-P2 and NMC-P3 peptides were conjugated to Keyhole limpet hemocyanin (KLH) using Sulfo-SMCC method (Thermo Scientific, Cat. No.22122). Briefly, Protein-NH2 was made in Conjugation Buffer (provided by manufacturer). Twenty-fold molar excess of crosslinker was added to the protein solution and the reaction mixture was incubated for 30 minutes at room temperature. Excess cross linker was then removed using desalting column equilibrated with Conjugation Buffer. Protein-SH and desalted Protein-NH2 were then combined, mixed and incubated for 30 minutes at room temperature.
  • Conjugation Buffer provided by manufacturer. Twenty-fold molar excess of crosslinker was added to the protein solution and the reaction mixture was incubated for 30 minutes at room temperature. Excess cross linker was then removed using desalting column equilibrated with Conjugation Buffer. Protein-SH and desalted Protein-NH2 were then combined,
  • mice BALB/c female
  • mice BALB/c female
  • Boosters were injected 5-7 times to provoke immune response.
  • Specific-antibody production was then evaluated by peptide-ELISA of the mice serum for antibody titration.
  • Spleens of mice with high antibody titer were then harvested from each mice and single cell suspension of splenocytes were prepared. Splenocytes were then fused with SP2/0 myeloma cells (1:5 ratio).
  • IMDM medium (10% FBS) was then added and kept in the incubator for 1 hour. Cells were then centrifuged and supernatant was removed. Cell pellet was then re-suspended in IMDM (20%
  • FBS FBS-containing HAT Fusion medium (hypoxanthine-aminopterin-thymidine medium). Cells were then plated in 96-well dishes and incubated for roughly 10 to 14 days. After 10-14 days when clones became visible, media supernatant from each well was tested by ELISA for its
  • NMC-P1, NMC-P2 or NMC-P3 binding to their corresponding specific immunogenic peptide
  • ELISA-positive wells were then selected for further clone selection, single-cell sub-cloning and monoclonal antibody purification.
  • Monoclonal antibody selection was done by peptide-ELISA using NMC-P1, NMC-P2 or NMC-P3 peptide antigen. To further select antibodies that react with the peptide antigen as well as with plasma membrane HDM2, binding assays were also.
  • Monoclonal antibody NMC-103 is an antibody that binds to NMC-P1 (SEQ ID NO:1) (it is one of the NMC-100s series of antibodies).
  • Monoclonal antibody NMC-204 is an antibody that binds to NMC-P2 (SEQ ID NO:2) (it is one of the NMC-200s series of antibodies).
  • Monoclonal antibody NMC-303 is an antibody that binds to NMC-P3 (SEQ ID NO:3) (it is one of the NMC-300s series of antibodies).
  • SEQ ID NO:3 is one of the NMC-300s series of antibodies.
  • the heavy chain/light chain frame work region sequences, complementarity determining region (CDR) sequences, and variable region sequences of these antibodies are listed in Section 8, below.
  • anti-HDM2 antibodies were used in the experiments described in Examples 1-9: (i) purified NMC-103 mouse monoclonal antibody (mAb) of the IgG1 isotype (NMC-103 mAbs produced by single-cell cloned hybridoma cells were purified on protein G/A columns), (ii) purified NMC-204 mouse mAb of the IgG3 isotype (NMC-204 mAbs produced by single-cell cloned hybridoma cells were purified on protein G/A columns); (iii) purified NMC- 303 mouse mAb of the IgM isotype (NMC-303 mAbs produced by single-cell cloned hybridoma cells were purified on protein G/A columns); (iv) an anti-HDM2 antibody termed“MDM2 monoclonal antibody (M01), clone 1A7” (Abnova, Cat.
  • MDM2 monoclonal antibody M01
  • Peptide-ELISA Methodology 5 mg/ml of NMC-P1, NMC-P2 or NMC-P3 peptide antigen was dried onto a 96-well ELISA plate overnight. Plates were then blocked with 5% BSA in 1x phosphate-buffered saline (PBS) (100 ml/well) for 2 hours at room temperature. Microplate wells were then washed 5 times with 300 ml of ice cold 1x PBS.
  • PBS phosphate-buffered saline
  • the membrane was then washed x 3 times (10 minutes each) and incubated for 1 hour with corresponding HRP-conjugated secondary antibody (Goat anti-Mouse IgG (H&L), F(ab')2 Frag Cross-adsorbed HRP (HRP-GaM F(ab’)2), ThermoFisher, Cat. No. A24524) diluted 1:5000 in 1%BSA-PBS.
  • the membrane was then washed x 3 times and incubated with Pierce ECL Plus Western Blotting Substrate (ThermoFisher, Cat. No. #32132) for 10 min before developing on a LICOR Scanner.
  • FIG. 1 The peptide-ELISA experiments shown in Figure 1 demonstrated selective and specific binding of monoclonal antibodies (mAb) NMC-103, NMC-204 and NMC-303 to their corresponding peptide antigens.
  • Figure 1A shows that NMC-103 bound to NMC-P1 peptide while NMC-204 did not bind to NMC-P1.
  • Figure 1B shows that mAb NMC-204 bound to NMC- P2 peptide while mAb NMC-103 did not bind to NMC-P2.
  • Figure 1C shows that mAb NMC- 303 bound to NMC-P3 peptide while NMC-204 did not bind to NMC-P3.
  • Lane 3 of the immunoblot shows the reactivity of mAb NMC-103 with a single band at approximately 83 kD, corresponding to the recombinant HDM2-GST protein.
  • Lane 4 of the immunoblot shows the binding of mAb NMC-204 to a single band at approximately 83 kD, corresponding to the recombinant HDM2-GST protein.
  • Lane 5 of the immunoblot shows reactivity of mAb NMC-303 with the recombinant HDM2-GST protein at 83 kD.
  • control mouse IgG (Abcam, Cat. No. ab18447) did not react with the recombinant HDM2-GST protein.
  • Cell-ELISA Methodology 8,000-10,000 cells/well of a 96-well microplate were grown overnight. The next day, unbound cells were washed off with sterile 1x PBS. The cells in each well were fixed with freshly prepared 4% buffered paraformaldehyde (pH 7.2) for 1 hour followed by 3 washes with 1x PBS. The wells were then blocked with 5% BSA in PBS (100 ml/well) for 2 hours at room temperature. Microplate wells were then washed 5 times with 300 ml of ice cold 1x PBS. MAbs NMC-103, NMC-204 or NMC-303 at 1 mg/mL in 1%BSA/PBS were then incubated with various cancer or normal untransformed cells for 2 hours at room
  • ThermoFisher, Cat. No.34028 was then added at 50 mL to each microplate well and incubated at room temperature for 30 minutes. The reaction was stopped by addition of 50 mL of stop solution (ThermoFisher, Cat. No. SS04) to each well and absorbance of each well was measured at OD450 nm. The absorbance value of each experimental well was corrected for the absorbance value obtained from wells treated with isotype-identical mAbs included in each experiment. The results are thus expressed as“relative binding”.
  • Figures 3-5 show the relative binding of mAbs NMC-103, NMC-204 and NMC-303 to intact cells of different types of human (A) and rodent (B) cancers.
  • Figure 3 shows reactivity of mAb NMC-103 to human breast cancer MCF-7 cells, human triple negative breast cancer HCC1806 cells, human pancreatic cancer MIA PaCa-2 cells, human ovarian cancer OVCAR-3 cells that are resistant to adriamycin, melphalan, and cisplatin, primary patient-derived human ovarian cancer OVCA4 cells, human melanoma A2058 cells, human uveal melanoma 92.1 cells, mouse colon cancer MC-38 cells, mouse Lewis Lung LL/2 cells and mouse pancreatic Panc02 cells.
  • Figure 4 presents the reactivity of NMC-204 monoclonal antibody to human breast cancer MCF-7 cells, human triple negative breast cancer HCC1806 cells, human pancreatic cancer MIA PaCa-2 cells, human ovarian cancer OVCAR-3 cells, primary patient-derived human ovarian cancer OVCA.4 cells, human melanoma A2058 cells, human uveal melanoma 92.1 cells, mouse colon cancer MC-38 cells, mouse Lewis Lung LL/2 cells and mouse pancreatic Panc02 cells.
  • Figure 5 shows reactivity of NMC-303 monoclonal antibody to human breast cancer MCF-7 cells, human triple negative breast cancer HCC1806 cells, human pancreatic cancer MIA PaCa-2 cells, human ovarian cancer OVCAR-3 cells, primary patient-derived human ovarian cancer OVCA.4 cells, human melanoma A2058 cells, human uveal melanoma 92.1 cells, mouse colon cancer MC-38 cells, mouse Lewis Lung LL/2 cells and mouse pancreatic Panc02 cells.
  • FIG. 6 demonstrates that NMC-204 did not react with normal human PBMCs. In contrast to cancer cells, neither mAb NMC-103, NMC-204 nor NMC-303 bound to normal intact cells (the data for NMC-103 and NMC-303 is not shown ). Data presented herein show that, while mAb NMC-204 reacted with human pancreatic cancer MIA PaCa-2 cells, no binding was seen above the background when mAb NMC-204 was incubated with freshly isolated normal human peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • Figure 6 demonstrates that while mAb NMC-204 did not react with normal human PBMCs (Figure 6 left; white bar graph), these cells showed strong reactivity with an mAb against CD3e, a cell surface marker for T cells ( Figure 6 left; shaded bar graph).
  • Figure 7 depicts mAb NMC-103 and NMC-204 saturation curves.
  • Figure 7A shows that cell-ELISA binding of mAb NMC-103 to intact MIA PaCa-2 cells increased as the concentration of the antibody increased. However, this binding reached a plateau at
  • FIG. 7B depicts the binding saturation curve of mAb NMC-204 to intact MIA PaCa-2 cells.
  • the binding of mAb NMC-204 to its antigen on the intact MIA PaCa-2 cells reached saturation at concentrations above 50 mg/mL of mAb NMC-204, demonstrating the binding saturation of antigen sites for mAb NMC- 204.
  • Example 3 mAb NMC-103, NMC-204 and NMC-303 were specific for
  • FIG. 8 shows the specificity of mAbs NMC-103, NMC- 204 and NMC-303 for NMC-P1, NMC-P2 and NMC-P3 sequences of HDM2 that are extracellularly accessible on the membrane of cancer cells.
  • Figure 8A shows the binding of mAb NMC-103 to intact human pancreatic cancer MIA PaCa-2 cells (left, solid bar).
  • Cytochrome-C (Santa Cruz, Cat. No. sc-13156) were unable to bind to their targets due to the inaccessibility of the intracellular compartments in the intact cells.
  • Example 5 Extracellular protease digestion of intact cancer cells removed extracellularly accessible sequences of M(H)DM2 on the plasma membrane of intact cancer cells.
  • MAbs NMC-103, NMC-204 and NMC-303 were raised against amino acids 1-15 (NMC-P1), 15-25 (NMC-P2) and 475-491 (NMC-P3) of HDM2, respectively, and were highly specific for HDM2. As shown in the immunoblot of Figure 2, all three mAbs NMC-103, NMC- 204 and NMC-303 recognized and bound to purified HDM2 protein, providing evidence that the unique antigenic epitopes recognized by the three mAbs in NMC-P1, NMC-P2 and NMC-P3, respectively, are de facto structures of intact HDM2.
  • Another set of the same cells was released with either EDTA or Trypsin and fixed with 4% buffered paraformaldehyde as above, then washed and blocked with 5% human serum albumin in PBS, and was treated separately with Triton X-100 (0.1% in PBS) (International Biotechnologies Inc.07100) for 5 minutes at room temperature for membrane permeabilization. After washing off the Triton X-100 solution with PBS the cells were adjusted to 10 6 cells/ml and stored in ice-cold PBS. Cells (10 6 /ml) from each preparation were then incubated either with 5 mg/ml of mAb to Na+/K+ ATPase a-1 (Abcam, Cat. No.
  • mAb to Cytochrome-C showed only minimal binding to EDTA-released cells (second bar from left), which is indicative of the inaccessibility of intracellular targets in intact cells to large molecules such as antibodies.
  • both mAbs NMC-103 and NMC-204 bound to intact, EDTA-released MIAPaCa-2 cells, establishing strong evidence for their interaction with their epitopes present in the extracellularly accessible sequences of HDM2.
  • EDTA-treated intact cells showed staining with mAb NMC-103 (70.4%), NMC-204 (51.2%) and anti- Na+/K+ ATPase a-1 antibody (29.9%).
  • NMC-103, NMC-204 and anti-Na+/K+ ATPase a-1 increased to 75.8%, 52.85 and 63.8%, respectively.
  • Figure 12 (A-D) demonstrates that human pancreatic cancer MIAPaCa-2 cells that were treated with mAb NMC-103 or NMC-204 exhibited a concentration-dependent growth inhibition, while an antibody against an intracellular marker (i.e., Cytochrome C) had no effect on the growth of these cancer cells.
  • MIAPaCa-2 cells treated with 1 mg/mL (top panel, left image), 5 mg/mL (to panel, middle image) and 10 mg/mL (top panel, left image) mAb NMC-103 showed an increasing effect in inhibition of their growth.
  • Figure 12B presents images of the cells treated with 1, 10 and 20 mg/mL of mAb NMC-204, showing a concentration-dependent inhibition of cell growth.
  • Figure 12C presents images of the cells treated with 1, 10 and 20 mg/mL of mAb NMC-204, showing a concentration-dependent inhibition of cell growth.
  • FIG. 12D quantifies the growth inhibitory effect of mAb NMC-103 at 1 mg/mL (65%), 5 mg/mL (84%) and 10 mg/mL (91%) (solid black line) and of NMC-204 at 1 mg/mL (65%), 5 mg/mL (73%) and 10 mg/mL (77%) (dashed black line).
  • treatment with an antibody to Cytochrome C an intracellular target; Santa Cruz, Cat. No.
  • the data presented herein show the ability of HDM2-specific antibodies to not only bind to the extracellularly accessible sequences of HDM2 on the surface membrane of cancer cells but also to initiate a potent cytotoxic effect in the presence of fresh normal human serum (NHS).
  • the cytotoxic effect was measured in pancreatic cancer cells as well as in normal human fibroblasts.
  • Data presented herein demonstrated one such cytotoxic effect when human pancreatic cancer MIAPaCa-2 cells were treated with mAb NMC-103 in the presence of NHS.
  • Figure 13 demonstrates the cytotoxic effect of HDM2-specific mAb NMC-103 against human pancreatic cells.
  • mAb NMC-103 in the presence of NHS triggered complement- mediated cytotoxicity in cancer cells, resulting in the death of the cancer cells as evident by the nuclear uptake of the cell-death marker Propidium Iodide (PI).
  • Figure 13C provides a quantitative representation of HDM2-specific antibody complement-dependent-cytotoxicity (CDC) against human pancreatic cancer cells.
  • Cells treated with mAb NMC-103 ( Figure 13B) in the presence of NHS demonstrated cytotoxicity over 2 hours post-treatment as compared with cells treated with NHS in the absence of any antibody ( Figure 13A).
  • Example 8 Evaluation of other anti-HDM2 mAb antibodies in their binding to NMC-P1, NMC-P2 and NMC-P3 and to intact cancer cells.
  • HDM2-specific monoclonal antibodies that are commercially available.
  • mAbs utilizing peptide- and cell-ELISA, we tested a number of such mAb for their binding to intact cancer cells.
  • These mAbs included antibodies that were raised against various regions from the N-terminus or C- terminus, or segments in the middle of the HDM2 protein, and were tested for their binding to newly identified extracellularly accessible NMC-P1 and NMC-P2 sequences as well as to intact cancer cells.
  • Table 3 summarizes the commercially available mAbs that were tested, the companies that generated these mAbs, and amino acid residues of HDM2 against which they had been raised.
  • Figures 14A and 14B demonstrated the lack of binding of any of these mAbs to either NMC-P1 or NMC-P2.
  • NMC-103 and NMC-204 showed strong binding to NMC-P1 and NMC-P2, respectively.
  • HDM-2 binding components such as peptides, for example PNC-27, interfered with the binding of mAb antibodies to NMC- P1 and NMC-P2.
  • HDM2-binding component of PNC-27 and PNC-28 peptides binds on HDM2
  • it has been reported to bind within amino acids 25-109 of HDM2 Do et al., 2003, Oncogene 22(10):1431-1444 (“Do 2003”); Chesse, 2003, Nat. Rev. Cancer 3(2):102-109).
  • HDM-2 binding components of PNC-27 and PNC-28 have no anti-cancer activity by themselves and are only active when attached to a membrane resident peptide (MRP or Penetratin sequence) (see Kanovsky 2001, Do 2003, and Bowne 2008).
  • MDM2 monoclonal antibody (M01), clone 1A7 was raised against amino acid 101 to 200 of full-length HDM2.
  • the data demonstrate that epitopes of HDM2 other than NMC-P1, NMC-P2 and NMC-P3 may be extracellularly accessible on cancer cells for binding.
  • two HDM2-specific monoclonal antibodies that did not react with intact cancer cells were identified ( Figure 15B).
  • One mAb was an anti- HDM2 antibody termed“Anti-MDM2 (Ab-4) Mouse mAb (2A9C1.18)” (EMD Millipore, Cat. No.
  • HDM2 there are select epitopes of HDM2 that are extracellularly accessible on cancer cells. Described herein are three (3) such extracellularly accessible segments of HDM2, namely NMC-P1 (SEQ ID NO:1), NMC-P2 (SEQ ID NO: 2) and NMC-P3 (SEQ ID NO: 3). Data herein demonstrated that select antibodies raised against these 3 segments selectively and specifically bound to various types of cancers but not normal health cells ( Figures 3-9). Moreover, results presented herein demonstrated the extracellular accessibility of these 3 segments of HDM2 on cancer cells ( Figure 10 and 11). Furthermore, data herein show that HDM2-specific antibodies had both growth inhibitory and cytotoxic effect against cancer cells in vitro. Finally, the anti-tumor activity of HDM2-specific antibodies was evaluated in vivo. As an example, efficacy of mAb NMC-103 and NMC-204 on lung, colon and pancreatic cancer was tested.
  • mice were micro-chipped and registered following tumor implantation. Tumor volume measurements and body weights were recorded and mice were randomized into groups that received: A) mAb NMC-103 at 0.4 mg/kg (3 times a week for 3 weeks), 2 mg/kg (2 times a week for 3 weeks), or 4 mg/kg (2 times a week for 3 weeks); B) NMC-204 at 0.4 mg/kg (3 times a week for 3 weeks); C) isotype control mouse IgG1 (Abcam, Cat. No. ab18447) or IgG3 (Abcam, Cat. No.
  • Figure 16 demonstrates the anti-tumor efficacy of mAb NMC-204 (0.4 mg/kg 3 times a week for 3 weeks; dashed line), in mice that were simultaneously inoculated with LL/2 Lewis Lung cancer cells.
  • NMC-204 reduced tumor volume (1446 mm 3 ) as compared with treatment with isotype control mouse IgG3 (2138 mm 3 ).
  • Figures 17 and 18 demonstrate the efficacy of mAb NMC-103 and NMC-204 against MC-38 syngeneic mouse model of colon cancer, respectively.
  • mice treated for 18 days with mAb NMC-103 (0.4mg/kg; 3 times a week for 2.5 weeks; dashed line) grew to 1268 mm 3 while mice treated with isotype control mouse IgG1 reached a tumor volume of 2205 mm 3 (solid line).
  • immunohistochemical staining for Ki67 protein a well- established cell proliferation marker (Li et al., 2015, Mol. Med.
  • mice treated with mAb NMC-103 had only 5% of tumor cells that stained positive for Ki67, while 80% of tumors in mice treated with isotype control antibody stained positive for Ki67 (Figure 17B). These results further confirm and are consistent with the growth inhibition result observed in vitro ( Figure 12). Furthermore, the anti-tumor efficacy of another HDM2-specific antibody, mAb NMC-204, that recognizes a different extracellularly accessible segment of membrane HDM2, namely NMC-P2 (SEQ ID NO:2), was evaluated.
  • mice treated with mAb NMC-204 grew to 1670 mm 3 while tumor in the control antibody-treated group reached 2555 mm 3 in volume.
  • Figure 18B shows that mice treated with NMC-204 had 30% of their tumor cells stained positively for Ki67 while mice in the control group had approximately 80% of their tumor cells staining positively for Ki67, demonstrating the anti-proliferative effect of NMC-204 treatment on tumor growth.
  • Figure 19 demonstrates the synergistic effect of NMC-103 in combination with pancreatic cancer standard-of-care treatment drugs: Gemcitabine (G) + nab-Paclitaxel (nP).
  • mice were then treated with a combination of NMC-103 (10mg/kg), Gemcitabine and nab-Paclitaxel two times a week for 2 weeks. As demonstrated in Figure 19, within 2 weeks tumors in both groups (C and D) reached the point where no measurable tumor was found.
  • mice in group A that had received isotype control antibody reached an average tumor size of 2028 mm 3 and were terminated.
  • mice that had previously received a combination of G + nP mice in group C (mice that had previously been treated with NMC-103) and group D (mice that had been treated with a combination of NMC-103 + G + nP), as described in Figure 27, were re-challenged by a second round of Panc-2 inoculation
  • mice were treated with pancreatic cancer standard of care (Gemcitabine (25 mg/kg) + nab- Paclitaxel (5 mg/kg)) for 19 days at which point the tumor reached a size of approximately 450 mm 3 .
  • pancreatic cancer standard of care Gemcitabine (25 mg/kg) + nab- Paclitaxel (5 mg/kg)
  • the mice were then randomly divided in 2 groups that received a single dose of: A) isotype control mouse IgG1 (10 mg/kg) or B) NMC-103 (10 mg/kg).
  • a single i.p. injection of NMC-103 reduced the tumor size by almost half 6 days post treatment (from 438 mm 3 to 233 mm 3 ).
  • Figure 17 mice treated with NMC-103 at 10 mg/kg, two times per week for three weeks resulted in a greater tumor reduction when compared to mice treated with NMC-103 at 0.4 mg/kg ( Figure 17).
  • mice The mouse Heavy and Light chain variable regions (SEQ ID NO:40 and SEQ ID NO:41, respectively) were cloned into a human Ig gamma-1 chain and human Ig kappa chain as constant region.
  • Figure 32A shows that by day 24 post tumor inoculation, mice treated with the chimeric version of monoclonal antibody NMC- 303 (10 mg/kg) reached an average tumor size of 726 mm 3 , while mice treated with control antibody (10 mg/kg) had an average tumor size of 1746 mm 3 . Furthermore, Figure 32B shows the individual mouse tumor sizes on day 24 post tumor inoculation. Due to the human constant region of the chimeric version of the monoclonal antibody NMC-303, the anti-tumor efficacy of this antibody might be improved if tested in mouse models with human immune background.
  • M(H)DM2/4-specific antibodies that target extracellularly accessible sequences of M(H)DM2/4 are in themselves effective anti-cancer agents.
  • M(H)DM2/4 antibodies used in combination with low concentrations of chemotherapeutics have a potently synergistic anti-tumor effect. This is particularly important, considering the frequently observed side-effects and limitations of chemotherapy used at their clinically effective concentrations.
  • these results demonstrate that M(H)DM2/4 antibodies lead to development of long-term anti-tumor immunity against cancer as demonstrated by prevention of recurrence of cancer and long-term survival of animals previously treated with M(H)DM2/4 antibodies.
  • Examples 10-12 below describe data obtained using other anti-HDM2 antibodies that bind to segments of HDM2 that are extracellularly accessible on cancer cells. 8.11 Example 10: Other HDM2-specific antibodies that also bind to intact cells from different human cancer cell lines and freshly isolated primary human cancer cells but not to normal cells. [00469] To further extend the therapeutic and diagnostic potentials of other antibodies raised against M(H)DM2/4 we evaluated several other available antibodies for their binding and anti- tumor activity. As described above, some of these antibodies (i.e. Calbiochem/Millipore OP-46 and OP-144) did not react with extracellularly accessible epitopes of M(H)DM2/4,
  • anti- M(H)DM2/4 antibodies demonstrate lack of epitope availability on cancer cell membranes, indicating that not all anti- M(H)DM2/4 antibodies can be used for the treatment of cancer.
  • anti- M(H)DM2/4 antibodies were shown to not only interact with extracellularly accessible epitopes on the cancer cell membrane, but also to have in vitro and/or in vivo anti-tumor activity.
  • anti-HDM2 antibodies were used in the experiments described in Examples 10-12: (i) polyclonal sc-813, N-20, rabbit IgG, from Santa Cruz (abbreviated throughout the specification as“N-20” or“sc-813 (N-20)”); (ii) monoclonal OP145, mouse IgG1, from Calbiochem (abbreviated throughout the specification as“OP145”); (iii) monoclonal OP46 (Ab-1), mouse IgG1, from Calbiochem (abbreviated throughout the specification as “OP46”); (iv) monoclonal OP144 (Ab-4), mouse IgG1, from Calbiochem (abbreviated throughout the specification as“OP144”); (v) polyclonal sc-812, C-18, rabbit IgG, from Santa Cruz (abbreviated throughout the specification as“C-18” or“sc-812 (C-18)”); and (vi) monoclonal 965 (SMP14), mouse IgG1, from Santa Cruz (abbrevi
  • Table 10 provides information regarding the HDM2 recognition sites of these antibodies (i.e., amino acids of HDM2 recognized by these antibodies), and whether or not these antibodies are cytotoxic to cancer cells.
  • OP145, N-20, C- 18 and SMP14 were cytotoxic to cancer cells tested, and OP46 and OP144 were not cytotoxic to cancer cells tested.
  • FACS Fluorescence-activated cell sorting
  • Figure 24 presents results of the FACS analysis of human melanoma, primary ovarian cancer, and normal mouse spleenocytes.
  • Figure 24A area under curve #1 represents cells incubated with goat anti-rabbit secondary antibody only; area under curve #2 represents cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #3 represents cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody.
  • Figure 24B area under curve #1 represents cells incubated with goat anti-mouse secondary antibody only; area under curve #2 represents cells incubated with anti-HDM2 monoclonal antibody OP145 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #3 represents cells incubated with anti-HDM2 monoclonal antibody OP145 followed by goat anti-mouse secondary antibody.
  • Figure 24C area under curve #1 represents cells incubated with goat anti-rabbit secondary antibody only; area under curve #2 represents trypsin-released cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody; area under curve #3 represents EDTA-released cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #4 represents EDTA-released cells incubated with anti-HDM2 polyclonal antibody N20 followed by goat anti-rabbit secondary antibody.
  • Figures 24D & E area under curve #1 represents cells incubated with goat anti-rabbit secondary antibody only; area under curve #2 represents cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #3 represents cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody.
  • Figure 24F area under curve #1 represents cells incubated with goat anti-rabbit secondary antibody only; area under curve #2 represents trypsin-released cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody; area under curve #3 represents EDTA-released cells incubated with anti-HDM2 polyclonal antibody N-20 pre-incubated with its blocking peptide followed by goat anti-rabbit secondary antibody; area under curve #4 represents EDTA-released cells incubated with anti-HDM2 polyclonal antibody N-20 followed by goat anti-rabbit secondary antibody.
  • the data presented herein show the ability of select HDM2-specific antibodies to not only bind to the surface membrane of cancer cells but also to initiate a cytotoxic effect in the presence of fresh normal human serum (NHS).
  • the cytotoxic effect was measured in various cancer cells such as human melanoma, pancreatic, breast and ovarian cancer cells as well as in normal human fibroblasts and blood cells.
  • Figure 25 demonstrates the cytotoxic effect of HDM2-specific antibodies against human pancreatic and ovarian cancer cells (Figure 25A), and rodent pancreatic cells (Figure 25B).
  • OP145, N-20 and C-18 antibodies in the presence of NHS trigger complement-mediated cytotoxicity in cancer cells incubated with such antibodies, resulting in the death of the cancer cells as evident by the nuclear uptake of the cell-death marker Propidium Iodide (PI) (see Figure 25A, panels b and e, for ovarian cancer cells incubated with OP145 and Figure 25B, panels b and c, for pancreatic cancer cells incubated with N-20 and C-18, respectively).
  • PI Propidium Iodide
  • cancer cell death is similar to that of control NHS alone, without any antibodies (see Figure 25A, panels a and d, and Figure 25B, panel a) or normal cells (Fibroblasts, Figure 25A, panel g) treated with HDM2-specific antibodies (i.e., there is no or minimal cell death as indicated by lack of PI staining, se Figures 25A, panels a, c, d, f and g, and Figure 25B, panel 1). Further, no cytotoxicity was observed when cells were treated with anti-HDM2 monoclonal OP46 antibody (see Figure 25B, panel d).
  • Figure 25C provides quantitative representations of HDM2-specific antibody-dependent complement cytotoxicity against human pancreatic cancer cells.
  • Cells treated with anti-HDM2 (C-18) antibody in the presence of NHS demonstrated cytotoxicity over 15-30 min. post-treatment, whereas anti-HDM2 OP46 shows no cytotoxic effect beyond that observed when cells were treated with control anti-Cytochrome C antibody or when cells were left untreated in the presence of NHS.
  • HDM2-specific antibodies in the presence of heat-inactivated human serum did not have any cytotoxic effect on cancer cells (i.e., did not result in the death of the cancer cells as was evident by PI staining), demonstrating that the cytotoxic effect is due to complement activity.
  • Table 10 lists antibodies tested by the inventors and summarizes results obtained relating to the in vitro cytotoxic effects of various anti-HDM2 antibodies against cancer cells.
  • Table 10 Antibodies used and their ability to induce a cytotoxic effect in cancer cells in vitro.
  • NMC-C103.VH0/VK0 chimeric NMC-103
  • humanized variants of NMC-C103 Humanized NMC-103” or “NMC-H103”
  • NMC-C303 chimeric NMC-303
  • Chimeric NMC-103 (“NMC-C103.VH0/VK0” or“NMC-C103”) monoclonal antibodies were generated by replacing the constant region of mouse IgG1 monoclonal antibody NMC-103 with a human IgG1 constant region.
  • NMC-103 is an antibody that binds to NMC-P1 (SEQ ID NO:1) (it is one of the NMC-100s series of antibodies).
  • Humanized variants of NMC-C103 (“Humanized NMC-103” or“NMC-H103”) were then designed comprising combinations of the following variable heavy and light chains: VH0, VH1, VH2, VH3, VH4, VH6, VH7 and VK0, VK1, VK2, VK3, VK4, VK5, VK6, VK7,
  • NMC-C303 chimeric NMC-303
  • a monoclonal antibody NMC-303 is an antibody that binds to NMC-P3 (SEQ ID NO:3) (it is one of the NMC-300s series of antibodies).
  • the heavy chain/light chain framework region sequences, complementarity determining region (CDR) sequences, and variable region sequences of these antibodies are listed in Section 11, below.
  • V murine antibody variable
  • CD4+ T Cell Epitope Avoidance Based upon the structural analysis, a large preliminary set of sequence segments were identified that could be used to create humanized variants. These segments were selected and analyzed using iTopeTM technology for in silico analysis of peptide binding to human MHC class II alleles (Perry et al., 2008) and using the TCEDTM of known antibody sequence-related T cell epitopes (Bryson et al., 2010). Sequence segments that were identified as significant non-human germline binders to human MHC class II or that scored significant hits against the TCEDTM were discarded.
  • VH1 to VH4, VH6, and VH7 Six heavy chain (VH1 to VH4, VH6, and VH7) and seven light chain (VK1 to VK7) sequences were chosen for gene expression in mammalian cells.

Abstract

La présente invention concerne, entre autres, certains anticorps anti-M(H)DM2/4 (y compris des anticorps chimériques et humanisés) ou des fragments de liaison à l'antigène de ceux-ci, des compositions pharmaceutiques comprenant des anticorps anti-M(H)DM2/4 ou des fragments de liaison à l'antigène de ceux-ci, des conjugués anticorps-médicament comprenant les anticorps anti-M(H)DM2/4 ou des fragments de liaison à l'antigène de ceux-ci liés à un médicament cytotoxique, et l'utilisation de ces anticorps, fragments, compositions et conjugués pour le traitement du cancer et/ou la prévention de métastases. L'invention concerne, à titre d'exemple, certains anticorps (y compris des anticorps chimériques et humanisés) ou des fragments de liaison à l'antigène de ceux-ci qui se lient de manière spécifique à des épitopes accessibles de manière extracellulaire de M(H)DM2/4 et inhibent la croissance tumorale in vivo, des compositions pharmaceutiques comprenant ces anticorps ou fragments, des conjugués anticorps-médicament comprenant ces anticorps ou fragments, et l'utilisation de ces anticorps, fragments, compositions et conjugués pour le traitement du cancer ou la prévention de métastases.
PCT/US2019/015900 2017-07-27 2019-01-30 Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer WO2020159504A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
PCT/US2019/015900 WO2020159504A1 (fr) 2019-01-30 2019-01-30 Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer
AU2019427766A AU2019427766A1 (en) 2019-01-30 2019-01-30 Antibodies to M(H)DM2/4 and their use in diagnosing and treating cancer
EP19705076.8A EP3917968A1 (fr) 2019-01-30 2019-01-30 Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer
US17/426,984 US20220098328A1 (en) 2017-07-27 2019-01-30 Antibodies to m(h)dm2/4 and their use in diagnosing and treating cancer
CA3127776A CA3127776A1 (fr) 2019-01-30 2019-01-30 Anticorps diriges contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2019/015900 WO2020159504A1 (fr) 2019-01-30 2019-01-30 Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer

Publications (1)

Publication Number Publication Date
WO2020159504A1 true WO2020159504A1 (fr) 2020-08-06

Family

ID=65409648

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/015900 WO2020159504A1 (fr) 2017-07-27 2019-01-30 Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer

Country Status (4)

Country Link
EP (1) EP3917968A1 (fr)
AU (1) AU2019427766A1 (fr)
CA (1) CA3127776A1 (fr)
WO (1) WO2020159504A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11407840B2 (en) 2017-07-27 2022-08-09 Nomocan Pharmaceuticals Llc Antibodies to M(H)DM2/4 and their use in diagnosing and treating cancer
EP3927362A4 (fr) * 2019-02-20 2022-11-23 Oncolyze, Inc. Anticorps anti-hdm2 destiné à être utilisé dans le traitement du cancer
WO2023194656A1 (fr) * 2022-04-08 2023-10-12 Tilt Biotherapeutics Oy Anticorps monoclonaux pd-l1

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1991009967A1 (fr) 1989-12-21 1991-07-11 Celltech Limited Anticorps humanises
WO1991010741A1 (fr) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation d'anticorps xenogeniques
WO1993017105A1 (fr) 1992-02-19 1993-09-02 Scotgen Limited Anticorps modifies, produits et procedes s'y rapportant
WO1993020238A2 (fr) * 1992-04-07 1993-10-14 The Johns Hopkins University Amplification du gene mdm2 dans des tumeurs chez l'homme
WO1995014233A2 (fr) * 1993-11-19 1995-05-26 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Procede d'identification d'anticorps specifiques contre l'hdm-2
WO1996033735A1 (fr) 1995-04-27 1996-10-31 Abgenix, Inc. Anticorps humains derives d'une xenosouris immunisee
WO1996034096A1 (fr) 1995-04-28 1996-10-31 Abgenix, Inc. Anticorps humains derives de xeno-souris immunisees
WO1998016654A1 (fr) 1996-10-11 1998-04-23 Japan Tobacco, Inc. Production de proteine multimere par procede de fusion cellulaire
WO1998024893A2 (fr) 1996-12-03 1998-06-11 Abgenix, Inc. MAMMIFERES TRANSGENIQUES POSSEDANT DES LOCI DE GENES D'IMMUNOGLOBULINE D'ORIGINE HUMAINE, DOTES DE REGIONS VH ET Vλ, ET ANTICORPS PRODUITS A PARTIR DE TELS MAMMIFERES
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
WO1998046645A2 (fr) 1997-04-14 1998-10-22 Micromet Gesellschaft Für Biomedizinische Forschung Mbh Nouveau procede de production de recepteurs d'anti-antigenes humains et leur utilisation
WO1998050433A2 (fr) 1997-05-05 1998-11-12 Abgenix, Inc. Anticorps monoclonaux humains contre le recepteur du facteur de croissance epidermique
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US7709226B2 (en) 2001-07-12 2010-05-04 Arrowsmith Technology Licensing Llc Method of humanizing antibodies by matching canonical structure types CDRs
WO2012010562A1 (fr) 2010-07-19 2012-01-26 International - Drug - Development - Biotech Anticorps anti-cd19 présentant des fonctions adcc et cdc, et un profil de glycosylation amélioré
US20120177566A1 (en) 2008-10-03 2012-07-12 Pincus Matthew R Membrane resident peptide in anti-cancer peptides causes tumor cell necrosis rather than apoptosis of cancer cells
US20140030319A1 (en) * 1995-09-04 2014-01-30 Bruno Tocque Antagonists Of The Oncongenic Activity Of The Protein MDM2, And Use Thereof In the Treatment of Cancers
WO2016054555A2 (fr) * 2014-10-03 2016-04-07 Novartis Ag Polythérapies
US9765117B2 (en) 2015-08-24 2017-09-19 Romek Figa Peptides for treating cancer
WO2019023460A1 (fr) * 2017-07-27 2019-01-31 Nomocan Pharmaceuticals Llc Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US6331415B1 (en) 1983-04-08 2001-12-18 Genentech, Inc. Methods of producing immunoglobulins, vectors and transformed host cells for use therein
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
WO1991009967A1 (fr) 1989-12-21 1991-07-11 Celltech Limited Anticorps humanises
WO1991010741A1 (fr) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation d'anticorps xenogeniques
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
WO1993017105A1 (fr) 1992-02-19 1993-09-02 Scotgen Limited Anticorps modifies, produits et procedes s'y rapportant
WO1993020238A2 (fr) * 1992-04-07 1993-10-14 The Johns Hopkins University Amplification du gene mdm2 dans des tumeurs chez l'homme
WO1995014233A2 (fr) * 1993-11-19 1995-05-26 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Procede d'identification d'anticorps specifiques contre l'hdm-2
WO1996033735A1 (fr) 1995-04-27 1996-10-31 Abgenix, Inc. Anticorps humains derives d'une xenosouris immunisee
WO1996034096A1 (fr) 1995-04-28 1996-10-31 Abgenix, Inc. Anticorps humains derives de xeno-souris immunisees
US20140030319A1 (en) * 1995-09-04 2014-01-30 Bruno Tocque Antagonists Of The Oncongenic Activity Of The Protein MDM2, And Use Thereof In the Treatment of Cancers
WO1998016654A1 (fr) 1996-10-11 1998-04-23 Japan Tobacco, Inc. Production de proteine multimere par procede de fusion cellulaire
WO1998024893A2 (fr) 1996-12-03 1998-06-11 Abgenix, Inc. MAMMIFERES TRANSGENIQUES POSSEDANT DES LOCI DE GENES D'IMMUNOGLOBULINE D'ORIGINE HUMAINE, DOTES DE REGIONS VH ET Vλ, ET ANTICORPS PRODUITS A PARTIR DE TELS MAMMIFERES
WO1998046645A2 (fr) 1997-04-14 1998-10-22 Micromet Gesellschaft Für Biomedizinische Forschung Mbh Nouveau procede de production de recepteurs d'anti-antigenes humains et leur utilisation
WO1998050433A2 (fr) 1997-05-05 1998-11-12 Abgenix, Inc. Anticorps monoclonaux humains contre le recepteur du facteur de croissance epidermique
US7709226B2 (en) 2001-07-12 2010-05-04 Arrowsmith Technology Licensing Llc Method of humanizing antibodies by matching canonical structure types CDRs
US20120177566A1 (en) 2008-10-03 2012-07-12 Pincus Matthew R Membrane resident peptide in anti-cancer peptides causes tumor cell necrosis rather than apoptosis of cancer cells
WO2012010562A1 (fr) 2010-07-19 2012-01-26 International - Drug - Development - Biotech Anticorps anti-cd19 présentant des fonctions adcc et cdc, et un profil de glycosylation amélioré
WO2016054555A2 (fr) * 2014-10-03 2016-04-07 Novartis Ag Polythérapies
US9765117B2 (en) 2015-08-24 2017-09-19 Romek Figa Peptides for treating cancer
WO2019023460A1 (fr) * 2017-07-27 2019-01-31 Nomocan Pharmaceuticals Llc Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer

Non-Patent Citations (204)

* Cited by examiner, † Cited by third party
Title
"Highlights of Prescribing Information", July 2015, CELGENE CORPORATION
"Monoclonal Antibodies: Principles and Practice"
"Monoclonal Antibodies: Principles and Practice", 1986, ACADEMIC PRESS, pages: 59 - 103
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO.
"Short Protocols in Molecular Biology", 2002, JOHN WILEY AND SONS
"UniProt", Database accession no. 015151
"UniProt", Database accession no. 035618
"UniProt", Database accession no. P23804
"UniProt", Database accession no. Q00987
A.K. SINGAV ET AL: "Predictive biomarkers for Hyper-progression (HP) in response to Immune Checkpoint Inhibitors (ICI) - Analysis of Somatic Alterations (SAs", ANN ONCOL, vol. 28, 2017
A.K. SINGAVI ET AL., ANN ONCOL, vol. 28, 2017
AHMAD ET AL., CLINICAL AND DEVELOPMENTAL IMMUNOLOGY, 2012
AL-LAZIKANI B ET AL., J MOL BIOL, vol. 273, 1997, pages 927 - 948
ALMAGRO; FRANSSON, FRONT. BIOSCI., vol. 13, 2008, pages 1619 - 1633
AMES RS ET AL., J IMMUNOL METHODS, vol. 184, 1995, pages 177 - 186
ARRUEBO ET AL., J. OF NANOMATER., 2009
BACA ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 10678 - 10684
BACA M ET AL., J BIOL CHEM, vol. 272, no. 16, 1997, pages 10678 - 84
BANDA NK; AK WOOD; K TAKAHASHI ET AL.: "Initiation of the alternative pathway of murine complement by immune complexes is dependent on N-glycans in IgG antibodies", ARTHRITIS RHEUM, vol. 58, 2008, pages 2081 - 3089
BARTEL ET AL., CANCER CELL, vol. 2, 2002, pages 9 - 15
BARTEL ET AL., MOL. CANCER RES., vol. 2, 2004, pages 29
BARTEL F.; HARRIS L.C.; WIIRL P.; TAUBERT T.: "MDM2 and Its Splice Variant Messenger RNAs: Expression in Tumors and Down-Regulation Using Antisense Oligonucleotides", MOL CANCER RES, vol. 2, 2004, pages 29
BARTEL F; TAUBERT H; HARRIS L.C.: "Alternative and aberrant splicing of MDM2 mRNA in human cancer", CANCER CELL, vol. 2, no. 1, 2002, pages 9 - 15
BARTEL F; TAUBERT H; HARRIS L.C.: "Alternative and aberrant splicing ofMDM2 mRNA in human cancer", CANCER CELL, vol. 2, no. 1, 2002, pages 9 - 15
BECHARA C; SAGAN S.: "Cell-penetrating peptides: 20 years later, where do we stand?", FEBS LETT., vol. 587, no. 12, 2013, pages 1693 - 702, XP028562950
BECHARA ET AL., FEBS LETT., vol. 587, 2013, pages 1693 - 1702
BELLMUNT ET AL., N. ENGL. J. MED., vol. 376, 2017, pages 1015 - 1026
BELLMUNT, J. ET AL.: "Pembrolizumab as second- line therapy for advanced urothelial carcinoma", N. ENGL. J. MED., vol. 376, 2017, pages 1015 - 1026, XP055518649
BESINGI RN; CLARK PL: "Extracellular protease digestion to evaluate membrane protein cell surface localization", NAT PROTOC., vol. 10, no. 12, December 2015 (2015-12-01), pages 2074 - 2080
BESINGI; CLARK, NAT. PROTOC., vol. 10, no. 12, 2015, pages 2074 - 2080
BOLHASSANI A.: "Potential efficacy of cell-penetrating peptides for nucleic acid and drug delivery in cancer", BIOCHIM BIOPHYS ACTA, vol. 1816, no. 2, 2011, pages 232 - 46, XP028392594
BOLHASSANI, BIOCHIM BIOPHYS. ACTA, vol. 1816, 2011, pages 232 - 246
BORGHAEI ET AL., N. ENGL. J. MED., vol. 373, 2015, pages 1627 - 1639
BORGHAEI, H. ET AL.: "Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer", N. ENGL. J. MED., vol. 373, 2015, pages 1627 - 1639
BOWNE ET AL., ANN SURG ONCOL., vol. 15, 2008, pages 3588 - 3600
BOWNE WB ET AL: "The penetratin sequence in the anticancer PNC-28 peptide causes tumor cell necrosis rather than apoptosis of human pancreatic cancer cells", ANN SURG ONCOL., vol. 15, no. 12, 2008, pages 3588 - 600, XP019638495
BRINKMAN U ET AL., J IMMUNOL METHODS, vol. 182, 1995, pages 41 - 50
BRODEUR ET AL.: "Monoclonal Antibody Production Techniques and Applications", 1987, MARCEL DEKKER, INC., pages: 51 - 63
BURTON DR; BARBAS CF, ADVAN IMMUNOL, vol. 57, 1994, pages 191 - 280
CALDAS C ET AL., PROTEIN ENG., vol. 13, no. 5, 2000, pages 353 - 60
CARDOSO ET AL., CURR. MED. CHEM., vol. 19, no. 19, 2012, pages 3103 - 27
CARTER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 4285
CHAMES ET AL., MABS, vol. 1, 2009, pages 539 - 547
CHAMES P; BATY D.: "Bispecific antibodies for cancer therapy: the light at the end of the tunnel?", MABS., vol. 1, no. 6, 2009, pages 539 - 47, XP002688758
CHAMPIAT ET AL., CLIN. CANCER RES., vol. 23, 2017, pages 1920 - 1928
CHAMPIAT, S. ET AL.: "Hyperprogressive disease is a new pattern of progression in cancer patients treated by anti- PD-l/PD- LI", CLIN. CANCER RES., vol. 23, 2017, pages 1920 - 1928
CHEN J ET AL: "MAPPING OF THE P53 AND MDM-2 INTERACTION DOMAINS", MOLECULAR AND CELLULAR BIOLOGY,, vol. 13, no. 7, 1 July 1993 (1993-07-01), pages 4107 - 4114, XP000571629, ISSN: 0270-7306 *
CHENE, NAT. REV. CANCER, vol. 3, no. 2, 2003, pages 102 - 109
CHOTHIA C ET AL., J MOL BIOL, vol. 227, 1992, pages 799 - 817
CHOTHIA C; LESK AM, J MOL BIOL, vol. 196, 1987, pages 901 - 917
CHOTHIA ET AL., J. MOL. BIOL., vol. 278, 1998, pages 457 - 479
CHOTHIA; LESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
COUTO JR ET AL., CANCER RES, vol. 55, no. 8, 1995, pages 1717 - 22
COUTO JR ET AL., CANCER RES., vol. 55, no. 23, 1995, pages 5973s - 5977s
DAVITT ET AL., ANNALS CLIN. LAB. SCI., vol. 44, 2014
DO ET AL., ONCOGENE, vol. 22, no. 10, 2003, pages 1431 - 1444
DO TN ET AL: "Preferential induction of necrosis in human breast cancer cells by a p53 peptide derived from the MDM2 binding site", ONCOGENE, vol. 22, no. 10, 2003, pages 1431 - 44, XP002573228
DUPONT E; PROCHIANTZ A; JOLIOT A.: "Penetratin Story: An Overview", METHODS MOL BIOL., vol. 1324, 2015, pages 29 - 37
DUPONT ET AL., METHODS MOL. BIOL., vol. 1324, 2015, pages 29 - 37
ELI LILLY, HIGHLIGHTS OF PRESCRIBING INFORMATION, March 2017 (2017-03-01), Retrieved from the Internet <URL:http://pi.lilly.com/us/gemzar.pdf>
EVANS ET AL., ONCOGENE, vol. 20, 2001, pages 4041 - 4049
EVANS S.C ET AL: "An alternatively spliced HDM2 product increases p53 activity by inhibiting HDM2", ONCOGENE, vol. 20, 2001, pages 4041 - 4049
FERRARA ET AL., JAMA ONCOL., 2018
FERRARA, R. ET AL.: "Hyperprogressive disease in patients with advanced non- small cell lung cancer treated with PD-l/PD- L1 inhibitors or with single agent chemotherapy", JAMA ONCOL., 2018, Retrieved from the Internet <URL:https://doi.org/10.1001/jamaoncol.2018.3676>
FERRIS ET AL., N. ENGL. J. MED., vol. 375, 2016, pages 1856 - 1867
FERRIS, R. L. ET AL.: "Nivolumab for recurrent squamous cell carcinoma of the head and neck", N. ENGL. J. MED., vol. 375, 2016, pages 1856 - 1867
FLATMAN ET AL., J. CHROMATOGR. B, vol. 848, 2007, pages 79 - 87
FRIDMAN JS ET AL: "Tumor promotion by Mdm2 splice variants unable to bind p53", CANCER RES., vol. 63, no. 18, 2003, pages 5703 - 6
GILLIES SD ET AL., J IMMUNOL METHODS, vol. 125, 1989, pages 191 - 202
GRAMER ET AL., MABS, vol. 5, 2013, pages 962 - 973
GRAMER MJ; ET VAN DEN BREMER; MD VAN KAMPEN; A KUNDU ET AL.: "Production of stable bispecific IgG1 by controlled Fab-arm exchange: scalability from bench to large-scale manufacturing by application of standard approaches", MABS, vol. 5, 2013, pages 962 - 973
HAMMERLING GJ ET AL.: "Monoclonal Antibodies and T-Cell Hybridomas", vol. 563, 1981, ELSEVIER, pages: 681
HARLOW E; LANE D: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY PRESS
HARLOW; LANE: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
IDUSOGIE EE ET AL.: "Engineered antibodies with increased activity to recruit complement", J IMMUNOL., vol. 166, no. 4, 2001, pages 2571 - 5, XP002781370
IDUSOGIE ET AL., J IMMUNOL., vol. 166, no. 4, 2001, pages 2571 - 5
IWAKUMA; LOZANO, MOL. CANCER RES., vol. 1, 2003, pages 993 - 1000
JAIN ET AL., CANCER RES., vol. 65, 2005, pages 7840 - 7846
JAIN M ET AL: "Penetratin improves tumor retention of single-chain antibodies: a novel step toward optimization of radioimmunotherapy of solid tumors", CANCER RES., vol. 65, no. 17, 2005, pages 7840 - 6, XP002585638
KABAT EA ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, NIH
KABAT EA; WU TT, ANN NY ACAD SCI, vol. 190, 1971, pages 382 - 391
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, NIH
KANOVSKY ET AL., PNAS, vol. 98, 2001, pages 12438 - 12443
KANOVSKY M ET AL: "Peptides from the amino terminal mdm-2-binding domain of p53, designed from conformational analysis, are selectively cytotoxic to transformed cells", PROC NATL ACAD SCI USA., vol. 98, no. 22, 2001, pages 12438 - 43, XP002391084
KATO ET AL., CLIN. CANCER RES., vol. 23, 2017, pages 4242 - 4250
KATO, S. ET AL.: "Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate", CLIN. CANCER RES., vol. 23, 2017, pages 4242 - 4250
KELLNER ET AL., METHODS, vol. 65, 2014, pages 105 - 113
KETTLEBOROUGH CA ET AL., EUR J IMMUNOL, vol. 24, 1994, pages 952 - 958
KLEEMANN ET AL., J. CONTROL RELEASE, vol. 109, 2005, pages 299 - 316
KLEEMANN ET AL: "Nano-carriers for DNA delivery to the lung based upon a TAT-derived peptide covalently coupled to PEG-PEI", J CONTROL RELEASE, vol. 109, no. 1-3, 2005, pages 299 - 316, XP005204221
KOHLER G; MILSTEIN C, NATURE, vol. 256, 1975, pages 495
KONTERMAN, MABS, vol. 4, 2012, pages 182 - 197
KONTERMAN, RE: "Dual targeting strategies with bispecific antibodies", MABS, vol. 4, 2012, pages 182 - 197
KOZBOR D, J IMMUNOL, vol. 133, 1984, pages 3001 - 5
LAFFLEUR ET AL., METHODS MOL. BIOL., vol. 901, 2012, pages 149 - 59
LAZAR ET AL., PNAS, vol. 103, 2006, pages 4005 - 4010
LAZAR ET AL., PNAS, vol. 103, no. 11, 2006, pages 4005 - 4010
LAZAR GA. ET AL.: "Engineered antibody Fc variants with enhanced effector function", PNAS, vol. 103, no. 11, 2006, pages 4005 - 4010
LEFRANC, M.-P. ET AL., NUCLEIC ACIDS RES., vol. 27, 1999, pages 209 - 212
LEFRANC, M.-P., THE IMMUNOLOGIST, vol. 7, 1999, pages 132 - 136
LENOS; JOCHEMSEN, J. BIOMED BIOTECHNOL., 2011
LI ET AL., MOL. MED. REP., vol. 11, no. 3, 2015, pages 1566 - 72
LI ET AL., SCI. REP., vol. 5, 2015, pages 7856
LI ET AL., VACCINES, vol. 2, 2014, pages 515 - 536
LI G ET AL.: "Enriched environment inhibits mouse pancreatic cancer growth and down-regulates the expression of mitochondria-related genes in cancer cells", SCI REP., vol. 5, 2015, pages 7856
LI G; GAN Y ET AL: "Enriched environment inhibits mouse pancreatic cancer growth and down- regulates the expression of mitochondria-related genes in cancer cells", SCI REP., vol. 5, 2015, pages 7856
LI, B; S SHI; W QUIAN; L ZHAO ET AL.: "Development of novel tetravalent anti-CD20 antibodies with potent tumor activity", CANCER RES, vol. 68, 2008, pages 2400 - 2408
LIANG H ET AL: "Genomic organization of the human MDM2 oncogene and relationship to its alternatively spliced mRNAs", GENE, vol. 338, 2004, pages 217 - 223, XP004527460
LIU ET AL., CA IMMUNOL RES., vol. 3, 2015, pages 173 - 183
LIU SD; C CHALOUNI; JC YOUNG ET AL.: "Afucosylated antobdies increase activation of FCgRIIIa-dependent signaling components to intensify processes promoting ADCC", CANCER IMMUNOL RES., vol. 3, 2015, pages 173 - 183
LUKAS ET AL., CANCER RES., vol. 61, 2001, pages 3212
LUKAS J ET AL: "Alternative and Aberrant Messenger RNA Splicing of the mdm2 Oncogene in Invasive Breast Cancer", CANCER RES, vol. 61, 2001, pages 3212, XP002300921
MACCALLUM RM ET AL., J MOL BIOL, vol. 5, 1996, pages 732 - 745
MACOR P ET AL: "Complment activated by chimeric anti-folate receptor antibodies is an efficient effector system to control ovarian carcinoma", CANCER RES., vol. 66, 2006, pages 3876 - 3883, XP002510868
MARINE ET AL., CELL CYCLE, vol. 3, 2004, pages 900 - 904
MARTIN A.: "Antibody Engineering", 2001, SPRINGER-VERLAG, article "Protein Sequence and Structure Analysis of Antibody Variable Domains", pages: 422 - 439
MARTIN, A.: "Antibody Engineering", 2001, SPRINGER-VERLAG, article "Protein Sequence and Structure Analysis of Antibody Variable Domains", pages: 422 - 439
MATSUMOTO ET AL., CANCER RES., vol. 58, 1998, pages 609 - 613
MATSUMOTO R ET AL: "Short alternative splice transcripts of the mdm2 oncogene correlate to malignancy in human astrocytic neoplasms", CANCER RES, vol. 58, 1998, pages 609 - 13, XP008071095
MCINTYRE R. M: "Mouse models of colorectal cancer as preclinical models", BIOASSAYS, vol. 37, no. 8, 2015, pages 909 - 920, XP055322925
MCINTYRE, BIOASSAYS, vol. 37, no. 8, 2015, pages 909 - 920
MCKAY BROWN ET AL: "Tolerance to single, but not multiple, amino acid replacements in antibody V-H CDR2: A means of minimizing B cell wastage from somatic hypermutation?", THE JOURNAL OF IMMUNOLOGY, THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS, INC, US, vol. 156, no. 9, 1 January 1996 (1996-01-01), pages 3285 - 3291, XP002649029, ISSN: 0022-1767 *
MOMAND ET AL., GENE, vol. 486, 2011, pages 23 - 30
MOREA V ET AL., METHODS, vol. 20, no. 3, 2000, pages 267 - 79
MORRISON SL, SCIENCE, vol. 229, 1985, pages 1202 - 7
MULLER S ET AL: "TransMabs: cell-penetrating antibodies, the next generation", EXPERT OPIN BIOL THER., vol. 5, no. 2, February 2005 (2005-02-01), pages 237 - 41, XP008141375
MUYLDERMANS S, J BIOTECHNOL, vol. 74, no. 4, 2001, pages 277 - 302
NAGANAWA Y ET AL., HUMAN ANTIBODIES, vol. 14, 2005, pages 27 - 31
NATSUME ET AL., DRUG DESIGN, DEVELOPMENT AND THERAPY, vol. 3, no. 3, 2009, pages 7 - 16
NUTTALL SD ET AL., CURR PHARM BIOTECHNOL, vol. 1, no. 3, 2000, pages 253 - 263
OI VT; MORRISON SL, BIOTECHNIQUES, vol. 4, 1986, pages 214 - 221
OLSON ES ET AL: "In vivo characterization of activatable cell penetrating peptides for targeting protease activity in cancer", INTEGR BIOL (CAMB, vol. 1, no. 5-6, 2009, pages 382 - 393, XP009163754
OLSON ET AL., INTEGR. BIOL (CAMB, vol. 1, 2009, pages 382 - 393
OLSON ET AL., ONCOGENE, vol. 8, 1993, pages 2353 - 2360
OLSON, D. C.; MARECHAL, V.; MOMAND, J.; CHEN, J.; ROMOCKI, C.; LEVINE, A. J.: "Identification and characterization of multiple mdm-2 proteins and mdm-2- p53 protein complexes", ONCOGENE, vol. 8, 1993, pages 2353 - 2360
PADLAN EA, MOL IMMUNOL, vol. 28, no. 4, 1991, pages 489 - 498
PAGE DB : "Immune modulation in cancer with antibodies", ANNU REV MED., vol. 65, 2014, pages 185 - 202, XP055492796
PANSRI ET AL., BMC BIOTECHNOLOGY, vol. 9, 2009, pages 6
PATERSON AM ET AL: "The programmed death-1 ligand 1:B7-1 pathway restrains diabetogenic effector T cells in vivo", J IMMUNOL., vol. 187, no. 3, 1 August 2011 (2011-08-01), pages 1097 - 105, XP055257409
PEDERSEN JT ET AL., J MOL BIOL, vol. 235, no. 3, 1994, pages 959 - 73
PERSIC L ET AL., GENE, vol. 187, 1997, pages 9 - 18
PETERS; BROWN, BIOSCI. REP., vol. 35, 2015, pages e00225
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623
RICHARDS ET AL., MOL.CANCER THER., vol. 7, 2008, pages 2517 - 27
RICHARDS JO ET AL.: "Optimization of antibody binding to FcgammaRIIa enhances macrophage phagocytosis of tumor cells", MOL CANCER THER., vol. 7, no. 8, 2008, pages 2517 - 27, XP002590014
RIECHMANN L; MUYLDERMANS S, J IMMUNOL, vol. 231, 1999, pages 25 - 38
ROGUSKA MA ET AL., PNAS, vol. 91, 1994, pages 969 - 973
ROGUSKA MA ET AL., PROTEIN ENG, vol. 9, no. 10, 1996, pages 895 904
ROSAL ET AL., ADV DRUG DELIV REV, vol. 57, 2005, pages 653 - 60
ROSAL R ET AL: "The role of alpha-helical structure in p53 peptides as a determinant for their mechanism of cell death: necrosis versus apoptosis", ADV DRUG DEVI REV, vol. 57, 2004, pages 653 - 660, XP025283898
ROSAL RL ET AL: "The role of alpha-helical structure in p53 peptides as a determinant for their mechanism of cell death: necrosis versus apoptosis", ADV DRUG DELIV REV., vol. 57, no. 4, 2005, pages 653 - 60, XP025283898
ROSOK ET AL., J. BIOL. CHEM., vol. 271, 1996, pages 22611 - 22618
ROSSO ET AL., SUBCELL BIOCHEM., vol. 85, 2014, pages 247 - 61
ROSSO M; OKORO DE; BARGONETTI J.: "Splice variants of MDM2 in oncogenesis", SUBCELL BIOCHEM., vol. 85, 2014, pages 247 - 61
RYAN MC. ET AL.: "Antibody targeting of B-cell maturation antigen on malignant plasma cells", MOL. CANCER THER., vol. 6, 2007, pages 3009 - 3018, XP002581270
SAADA- BOUZID ET AL., ANN. ONCOL., vol. 28, 2017, pages 1605 - 1611
SAADA- BOUZID, E. ET AL.: "Hyperprogression during anti- PD-l/PD- L1 therapy in patients with recurrent and/or metastatic head and neck squamous cell carcinoma", ANN. ONCOL., vol. 28, 2017, pages 1605 - 1611, XP055486761
SADELAIN ET AL., CANCER DISCOVERY, vol. 3, 2013, pages 388 - 398
SANDHU JS, GENE, vol. 150, no. 2, 1994, pages 409 - 10
SARAFRAZ-YAZDI E ET AL: "Anti-cancer peptide, PNC-27, adopts an HDM2-binding conformation and kills cancer cells by binding to HDM2 in their membranes", PNAS, vol. 107, 2010, pages 1918 - 1923, XP055519445
SARAFRAZ-YAZDI ET AL., PNAS, vol. 107, 2010, pages 1918 - 1923
SATOH ET AL., EXPERT OPIN BIOL THER., vol. 6, 2006, pages 1161 - 1173
SATOH MM; S IIDA; K SHITARA: "Non-fucosylated therapeutic antibodies as next-generation therapeutic antibodies", EXPERT OPIN BIOL THER, vol. 6, 2006, pages 1161 - 1173, XP008078583
SCHIILEIN R; RUTZ C; ROSENTHAL W.: "Membrane Targeting and Determination of Transmembrane Topology of the Human Vasopressin V2 Receptor", JBC, vol. 271, no. 46, 1996, pages 28844 - 28852
SCHOLZEN T; GERDES J: "The Ki-67 protein: from the known and the unknown", JOURNAL OF CELLULAR PHYSIOLOGY, vol. 182, no. 3, 2000, pages 311 - 22, XP055000935
SCHULEIN ET AL., J. BIOL. CHEM., vol. 271, no. 46, 1996, pages 28844 - 28852
SCHUSTER ET AL., MOL. CANCER RES., vol. 5, 2007, pages 403 - 412
SCHUSTER K.; FAN L.; HARRIS L.C.: "MDM2 Splice Variants Predominantly Localize to the Nucleoplasm Mediated by a COOH-Terminal Nuclear Localization Signal", MOL CANCER RESEARCH, vol. 5, no. 4, 2007, pages 403 - 412
SCHUSTER K; FAN L; HARRIS LC: "MDM2 splice variants predominantly localize to the nucleoplasm mediated by a COOH-terminal nuclear localization signal", MOL CANCER RES., vol. 5, no. 4, 2007, pages 403 - 12
SHARMA ET AL., J. IMMUNOL., vol. 163, no. 9, 1999, pages 5020 - 5028
SHERVEN SHARMA ET AL.: "T Cell-Derived IL-10 Promotes Lung Cancer Growth by Suppressing Both T Cell and APC Function", J IMMUNOL, vol. 163, no. 9, 1999, pages 5020 - 5028
SHIELDS RL. ET AL.: "High resolution mapping of the binding site on human IgGl for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgGl variants with improved binding to the Fc gamma R", J BIOL CHEM., vol. 276, no. 9, 2001, pages 6591 - 604
SHIN ET AL., J. BIOMED. MATER RES. A., vol. 102, 2014, pages 575 - 587
SHIN MC ET AL: "Cell-penetrating peptides: achievements and challenges in application for cancer treatment", J BIOMED MATER RES A., vol. 102, no. 2, 2014, pages 575 - 87
SHINMOTO H ET AL., CYTOTECHNOLOGY, vol. 46, 2004, pages 19 - 23
SHISUO DU ET AL., ONCOIMMUNOLOGY, vol. 7, no. 4, 2018, pages el408747
SHISUO DU ET AL: "Blockade of Tumor-Expressed PD-1 promotes lung cancer growth", ONCOIMMUNOLOGY, vol. 7, no. 4, 2018, pages e1408747
SHISUO ONCOIMMUNOLOGY, vol. 7, no. 4, 2018, pages el408747
SHULTZ LD ET AL: "Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells", J IMMUNOL., vol. 174, no. 10, 15 May 2005 (2005-05-15), pages 6477 - 89, XP008164911
SIGALAS ET AL., NAT. MED., vol. 2, 1996, pages 912 - 917
SIGALAS, A.H.; CALVERT, J.J.; ANDERSON, D.E. NEAL; J. LUNEC: "Alternatively spliced mdm2 transcripts with loss of p53 binding domain sequences: transforming ability and frequent detection in human cancer", NAT. MED., vol. 2, 1996, pages 912 - 917
SIMS ET AL., J. IMMUNOL., vol. 151, 1993, pages 2296
SOOKRAJ KA ET AL: "The anti-cancer peptide, PNC-27, induces tumor cell lysis as the intact peptide", CANCER CHEMOTHER PHARMACOL, vol. 66, no. 2, 2010, pages 325 - 31, XP019843311
STEINMAN ET AL., JBC, vol. 279, 2004, pages 4877 - 4886
STEINMAN H.A ET AL: "An Alternative Splice Form of Mdm2 Induces p53-independent Cell Growth and Tumorigenesis", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 279, 2004, pages 4877 - 4886
STEURER W. ET AL.: "Ex vivo coating of islet cell allografts with murine CTLA4/Fc promotes graft tolerance", J IMMUNOL., vol. 155, no. 3, 1995, pages 1165 - 74
STROHL, CURR OPIN BIOTECHNOL., vol. 20, no. 6, 2009, pages 685 - 91
STUDNICKA GM ET AL., PROT ENGINEERING, vol. 7, no. 6, 1994, pages 805 - 814
TAMBORINI E; DELLA TORRE G; LAVARINO C ET AL.: "Analysis of the molecular species generated by MDM2 gene amplification in liposarcomas", INT J CANCER, vol. 92, 2001, pages 790 - 6
TAMBORINI ET AL., INT. J. CANCER, vol. 92, 2001, pages 790 - 796
TAN P ET AL., J IMMUNOL, vol. 169, 2002, pages 1119 - 25
TAUBERT ET AL: "A MboII polymorphism in exon 11 of the human MDM2 gene occurring in normal blood donors and in soft tissue sarcoma patients: an indication for an increased cancer susceptibility?", MUTAT. RES., vol. 456, 2000, pages 39 - 44
TORCHILIN ET AL., PNAS, vol. 100, 2003, pages 1972 - 1977
TORCHILIN VP ET AL: "Cell transfection in vitro and in vivo with nontoxic TAT peptide-liposome-DNA complexes", PROC NATL ACAD SCI USA., vol. 100, no. 4, 2003, pages 1972 - 7
TRAMONTANO A ET AL., J MOL BIOL, vol. 215, no. 1, 1990, pages 175 - 82
VACCARO ET AL., NAT BIOTECHNOL., vol. 23, 2005, pages 1283 - 8128
VOLK, E. L.; FAN L.; SCHUSTER K.: "The MDM2-A splice variant of MDM2 alters transformation in vitro and the tumor spectrum in both Arf-null and p53-null models of tumorigenesis", MOL CANCER RES., vol. 7, no. 6, 2009, pages 863 - 869
VOLK, MOL CANCER RES., vol. 7, no. 6, 2009, pages 863 - 869
WADE ET AL, NAT REV. CANCER, vol. 13, 2013, pages 83 - 96
WANG ET AL., ANAL. CHEM., vol. 78, 2006, pages 997 - 1004
WILLAM C ET AL: "Peptide blockade of HIFalpha degradation modulates cellular metabolism and angiogenesis", PROC NATL ACAD SCI USA., vol. 99, no. 16, 2002, pages 10423 - 8, XP002265953
WILLIAM ET AL., PNAS, vol. 99, 2002, pages 10423 - 10428
YANG JY ET AL: "MDM2 promotes cell motility and invasiveness by regulating E-cadherin degradation", MOL CELL BIOL., vol. 26, no. 19, 2006, pages 7269 - 82
ZUAZO- IBARRA, M. ET AL.: "Highly differentiated CD4 T cells unequivocally identify primary resistance and risk of hyperprogression to PD- LI/IPD-1 immune checkpoint blockade in lung cancer", BIORXIV, 2018, Retrieved from the Internet <URL:https://doi.org/10.1101/320176>

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11407840B2 (en) 2017-07-27 2022-08-09 Nomocan Pharmaceuticals Llc Antibodies to M(H)DM2/4 and their use in diagnosing and treating cancer
EP3927362A4 (fr) * 2019-02-20 2022-11-23 Oncolyze, Inc. Anticorps anti-hdm2 destiné à être utilisé dans le traitement du cancer
WO2023194656A1 (fr) * 2022-04-08 2023-10-12 Tilt Biotherapeutics Oy Anticorps monoclonaux pd-l1

Also Published As

Publication number Publication date
CA3127776A1 (fr) 2020-08-06
AU2019427766A1 (en) 2021-09-16
EP3917968A1 (fr) 2021-12-08

Similar Documents

Publication Publication Date Title
US11845792B2 (en) Epitope specific antibodies that bind cell surface GRP78 and their use for cancer detection
US10202461B2 (en) Anti-human TROP-2 antibody having an antitumor activity in vivo
US9670287B2 (en) Anti-human TROP-2 antibody having anti-tumor activity in vivo
US11407840B2 (en) Antibodies to M(H)DM2/4 and their use in diagnosing and treating cancer
ES2527521T3 (es) Anticuerpos antihumanos frente a Dlk-1 que tienen actividad antitumoral
AU2016250900B2 (en) Therapeutic antibodies and uses thereof
WO2017026497A1 (fr) Anticorps
KR20220114559A (ko) Cd38에 결합하는 중쇄 항체
WO2020159504A1 (fr) Anticorps dirigés contre m(h)dm2/4 et leur utilisation dans le diagnostic et le traitement du cancer
WO2015076425A1 (fr) Nouvel anticorps monoclonal
KR20230057428A (ko) 어댑터를 포함하는 약물 치료제의 개발 및 이의 용도
US20220098328A1 (en) Antibodies to m(h)dm2/4 and their use in diagnosing and treating cancer
US20240043568A1 (en) Development of new tumor engager therapeutic drug and use thereof
EA042429B1 (ru) Композиции и способы для диагностики и лечения рака предстательной железы с использованием молекулы, связывающей прогастрин
NZ716839A (en) Anti-human trop-2 antibody having an antitumor activity in vivo
NZ716839B2 (en) Anti-human trop-2 antibody having an antitumor activity in vivo

Legal Events

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

Ref document number: 19705076

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3127776

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019427766

Country of ref document: AU

Date of ref document: 20190130

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019705076

Country of ref document: EP

Effective date: 20210830