WO2013177386A1 - Biomarkers for predicting response to tweak receptor (tweakr) agonist therapy - Google Patents

Biomarkers for predicting response to tweak receptor (tweakr) agonist therapy Download PDF

Info

Publication number
WO2013177386A1
WO2013177386A1 PCT/US2013/042409 US2013042409W WO2013177386A1 WO 2013177386 A1 WO2013177386 A1 WO 2013177386A1 US 2013042409 W US2013042409 W US 2013042409W WO 2013177386 A1 WO2013177386 A1 WO 2013177386A1
Authority
WO
WIPO (PCT)
Prior art keywords
tweakr
seq
cancer
antibody
mutation
Prior art date
Application number
PCT/US2013/042409
Other languages
French (fr)
Inventor
Patricia Culp
Keith Wilson
Shiming Ye
Original Assignee
Abbvie Biotherapeutics Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbvie Biotherapeutics Inc. filed Critical Abbvie Biotherapeutics Inc.
Publication of WO2013177386A1 publication Critical patent/WO2013177386A1/en

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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/01Phosphotransferases with an alcohol group as acceptor (2.7.1)
    • C12Y207/01153Phosphatidylinositol-4,5-bisphosphate 3-kinase (2.7.1.153), i.e. phosphoinositide 3-kinase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11001Non-specific serine/threonine protein kinase (2.7.11.1), i.e. casein kinase or checkpoint kinase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • 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
    • C07K2317/70Immunoglobulins 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • TWEAK RECEPTOR AGONIST THERAPY
  • TWEAK Tumor Necrosis Factor-Related Weak inducer of Apoptosis
  • TWEAK Tumor Necrosis Factor-Related Weak inducer of Apoptosis
  • TNF Tumor Necrosis Factor
  • TWEAK has a number of biological activities, including inducing angiogenesis, stimulating the release of cytokines or chemokines, stimulating proliferation of human smooth muscle and endothelial cell lines, inducing TNF-a-mediated cell death, cathepsin B-dependent necrosis, and, as its name implies, apoptosis. See Han et al, 2003, Bioch. Biophys. Res.
  • TWEAK binds to and activates a member of the TNF receptor family, TWEAKR (also known as Fibroblast Growth Factor-Inducible 14 or Fnl4). See id.
  • TWEAKR agonists induced cell death via apoptosis or necrosis in a variety of cancer cell lines.
  • Phosphoinositide 3-kinases are lipid kinases that function as signal transducers downstream of cell surface receptors and mediate pathways important for cell growth, proliferation, adhesion, survival and motility. Mutations in PI3K genes result in deregulation of one or more of these biological processes and have been associated with cancer pathogenesis (Samuels et al, Science 304:554; Bachman et al, Cancer Biol. Ther. 3:772; Campbell, et al, Cancer Res. 64:7678; Broderick et al, Cancer Res 64:5048; Lee et al, Oncogene 24:1477; Levin et al, Clin. Cancer Res.
  • PDKs are classified based on primary structure, regulation, and in vitro lipid substrate specificity. Studies of proliferation and tumorigenesis have focused on Class I A POKs, which are heterodimeric molecules composed of a catalytic subunit, referred to as pi 10a, and an associated regulatory subunit, referred to as p85.
  • Class IA PDKs are responsible for the production of phosphatidylinositol 3,4,5-triphosphate (PIP 3 ) from phosphatidylinositol 4,5-bisphopshate (PIP 2 ), while phosphatases, in particular the PTEN phosphatase, carry out the reverse reaction, dephosphorylating PIP 3 .
  • PIP 3 phosphatidylinositol 3,4,5-triphosphate
  • PTEN phosphatases in particular the PTEN phosphatase
  • Both pi 10a and PTEN have been linked to cancer: the gene encoding pi 10a, called PIK3CA, has been identified as an oncogene (Samuels, et al, Science 304:443) and PTEN has been classified as a tumor suppressor (Deane and Fruman, Annu. Rev. Immunol. 22:563; Okkenhaug et al, Sci. STKE 65:PE1).
  • TWEAKR agonists inhibit proliferation of some, but not all, tumor cells that express TWEAKR.
  • the ability of TWEAKR agonists to inhibit proliferation of TWEAKR positive tumors does not correlate with cancer type.
  • Table 1 and FIG. 1 in in vitro experiments carried out in tumor cell lines derived from a variety of different types of cancers, an anti- TWEAKR antibody having TWEAKR agonist activity, enavatuzumab (also known as, and referred to herein as, PDL192) inhibited the proliferation and/or survival of some, but not all, TWEAKR positive cell lines derived from the same type of cancer.
  • PIK3CA which encodes pi 10a, a PI3K lipid kinase, and in PTEN, a phosphatase
  • PIK3CA activating mutations mutations that result in increased activity of pi 10a
  • PTEN null mutations mutations that result in reduced activity of PTEN
  • the present disclosure provides methods of treating a subject having a TWEAKR-positive cancer comprising administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the TWEAKR agonist can be an anti-TWEAKR agonist antibody, including monoclonal and humanized antibodies.
  • the methods further comprise the step of testing the subject for one or more PIK3CA activating mutations.
  • PIK3CA activating mutations Exemplary methods for detecting a PIK3CA activating mutation are described in Section 5.3, below.
  • the testing of the subject is conducted prior to administration of the TWEAKR agonist, and can be conducted simultaneously with testing the subject for TWEAKR expression.
  • the present disclosure provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by testing a subject for one or more PIK3CA activating mutations, preferably concurrently with or after testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the present disclosure further provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by (i) testing a subject for one or more PIK3CA activating mutations and (ii) prior to or concurrently with testing the subject for one or more PIK3CA activating mutations, testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the present disclosure provides methods of treating a subject having a TWEAKR-positive cancer comprising administering to a subject who has tested negative for a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the TWEAKR agonist can be an anti-TWEAKR agonist antibody, including monoclonal and humanized antibodies.
  • the methods further comprise the step of testing the subject for a PTEN null mutation.
  • exemplary methods for detecting a PTEN null mutation are described in Section 5.3, below.
  • the testing of the subject is conducted prior to administration of the TWEAKR agonist, and can be conducted simultaneously with testing the subject for TWEAKR expression.
  • the present disclosure provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by testing a subject for a PTEN null mutation, preferably concurrently with or after testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR- positive cancer who has tested negative for a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the present disclosure further provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by (i) testing a subject for a PTEN null mutation and (ii) prior to or concurrently with testing the subject for a PTEN null mutation, testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the present disclosure provides methods of treating a subject having a TWEAKR-positive cancer comprising administering to a subject who has tested negative for a PTEN null mutation and for one or more PIK3CA activating mutations an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the methods of the disclosure further comprise the step of testing the subject for a PTEN null mutation and the one or more PIK3CA activating mutations, as described herein.
  • the testing of the subject is conducted prior to administration of the TWEAKR agonist, and can be conducted simultaneously with testing the subject for TWEAKR expression.
  • the present disclosure provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by testing a subject for a PTEN null mutation and one or more PIK3CA activating mutations, preferably concurrently with or after testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PTEN null mutation and one or more PIK3CA activating mutations an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the present disclosure further provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by (i) testing a subject for a PTEN null mutation and one or more PIK3CA activating mutations and (ii) prior to or concurrently with testing the subject for a PTEN null mutation and one or more PIK3CA activating mutations, testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PTEN null mutation and a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • the PIK3CA activating mutation can be any type of mutation that leads to increased kinase activity relative to wild type human PIK3CA, including a substitution mutation, deletion mutation, or insertion mutation.
  • the PIK3CA activating mutation is a substitution mutation, e.g., a mutation selected from the group consisting of: K111N, E542K, E545K, E545G, E545D, Q546K, Q546R, Ml 0431, M1043V, H1047R, H1047L, H1047Y, G1049R and G1049S.
  • substitution mutations are provided in Section 5.1, below.
  • the PIK3CA activating mutation can be detected at the nucleic acid level (e.g., DNA or mRNA) or at the protein level.
  • the subject is tested for one, two, three, four or more (or all) of Kl 1 IN, E542K, E545K, E545G, E545D, Q546K, Q546R, M1043I, M1043V, H1047R, H1047L, H1047Y, G1049R and G1049S, optionally in addition to further PIK3CA activating mutations, such as those provided in Section 5.1.
  • the PTEN null mutation can be any type of mutation that leads to decreased phosphatase activity compared to wild type human PTEN, including a substitution mutation, deletion mutation, or insertion mutation.
  • the PTEN null mutation can be detected at the nucleic acid level (e.g., DNA or mRNA) or at the protein level. Further PTEN null mutations are provided in Section 5.2, below.
  • TWEAKR agonists also referred to herein as TWEAKR agonist compounds, useful in the methods of the present disclosure are described in Section 5.4.
  • FIG. 1 provides a bar chart illustrating the percent growth inhibition for 38
  • FIG. 2 provides results of 91 cell lines treated with TWEAKR agonist PDL192 classified based on growth inhibition and presence (mutation) or absence (wild type) of a PIK3CA activating mutation. Cell lines that exhibited growth inhibition (responders) or lack of growth inhibition (nonresponders) when treated with PDL192 are indicated on the left.
  • FIGS. 3A-B provide the results of in vivo experiments in which mice bearing xenograft tumors derived from cell lines were treated with TWEAKR agonist PDL192.
  • the cell lines were classified based on growth inhibition response (responders or non-responders) and PIK3CA and PTEN genotype (mutation or wild type) (FIG. 3A).
  • a subset of cell lines were classified as being responsive via a non-ADCC mechanism or nonresponders and PIK3CA and PTEN genotype (mutation or wild type) (FIG. 3B).
  • FIG. 4A illustrates phospho-Akt (S473) levels in cell lines treated with PDL192 in vitro. Cell lines that were treated with PDL192 are indicated on the right hand side.
  • FIG. 5 provides the relative cell viability of cells transfected with control vector, wild type PIK3CA, or PIK3CA containing an activating mutation, as indicated, that were treated with TWEAKR agonist (PDL192, square data points) or control compound (MSL109, round data points).
  • FIG. 6 provides the results of treatment with TWEAKR agonist (PDL192, black bars) or control compound (MSL109, white bars) for DLD-1 and HCTl 16 cells.
  • DLD-1 cells have an E545K mutation in PIK3CA
  • HCTl 16 cells have an H1047R mutation in PIK3CA.
  • the cell lines were subjected to siRNA depletion using control or PIK3CA siR A.
  • TWEAKR also known as Fnl4 or TNFRSF12A
  • Fnl4 or TNFRSF12A tumor necrosis factor
  • TNF tumor necrosis factor
  • TWEAKR agonists are potentially useful in the treatment of some TWEAKR positive cancers, particularly if it is possible to determine whether the TWEAKR positive cancer is likely to be a responder or non-responder to TWEAKR agonist therapy.
  • the present disclosure is based in part on applicants' discovery that PIK3CA activating mutations and PTEN null mutations are indicative of non-responsiveness to TWEAKR agonist therapy in treating a TWEAKR positive cancer. As shown in the
  • the methods of treating a subject having a TWEAKR positive cancer comprise administering to a subject who has tested negative for a PIK3CA activating mutation or PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • methods of the disclosure may be employed to classify subjects as responders or non-responders to a TWEAKR agonist therapy. In some embodiments, the methods may be also used to predict whether or not a subject suffering from a TWEAKR positive cancer will respond to a TWEAKR agonist therapy.
  • PIK3CA encodes the catalytic subunit of a Class 1 A PI3 kinase, a class of enzymes that catalyzes the phosphorylation of inositol-containing lipids.
  • PI3 kinases PI3Ks
  • PI3CA sequence refers to a polynucleotide encoding the pi 10a kinase subunit.
  • a PIK3CA sequence can be a gene sequence (with or without introns), a cDNA, or an mRNA.
  • a representative example of a wild type human PIK3CA sequence is SEQ ID NO:55.
  • the corresponding polypeptide sequence for a human pi 10a is SEQ ID NO: 56.
  • Mutations in PIK3CA that correlate with proliferative disorders such as cancer are known to cluster in exon 9 (SEQ ID NO:57) and exon 20 (SEQ ID NO:58). See U.S. Patent Publication Nos. 2009/0208505, paragraph [0017] and Table 2 herein.
  • Exon 9 corresponds to the helical domain of PIK3CA
  • exon 20 corresponds to its kinase domain.
  • PIK3CA activating mutations cluster in these regions, known as mutational hotspots. Additional mutations found in human cancers are described in Table 3 of U.S. Patent Publication No. 2009/0208505.
  • PIK3CA activating mutations are PIK3CA activating mutations, a number of which have been described, including those described in Bader, et al. Nat. Rev. Cancer (2005) 5:921-929, Kang et al, PNAS (2005), 102:802, Samuels et al, Science (2004) 304:554, Zhang et al. , Breast Cancer Res. Treat. (2008) 112:217-227, and U.S. Patent Publication No. 2011/0060605.
  • a "PIK3CA activating mutation” can be any type of mutation, including a substitution mutation, a deletion mutation, or an insertion mutation, which results in increased kinase activity compared to wild type PIK3CA.
  • Increased kinase activity can be the result of increased enzymatic activity or increased levels of the pi 10a polypeptide.
  • Numerous assays for kinase activity are known, which can be used to determine whether a PIK3CA sequence results in a pi 10a with increased kinase activity relative to wild type pi 10a.
  • pi 10a kinase activity can be determined using an in vitro kinase assay.
  • PI3 kinase assays have been described (see Kang et ah, 2005, PNAS 102:802-807).
  • complexes of pi 10a and an anti- pi 10a antibody are prepared by incubating 400 ⁇ g of pi 10a with 5 ⁇ of anti-pl 10a antibody for 2 h at 4°C, followed by 2 h of incubation with Protein A-agarose. Protein A-agarose beads are washed three times with the lysis buffer and once with TNE (10 mM Tris, Ph 7.5/100 mM NaCl/1 mM EDTA).
  • the reaction is terminated by adding 80 ⁇ of 1 N HC1, and the phosphorylated lipids are extracted with 160 ⁇ of chloroform/methanol (1 : 1). After samples are dried down, they are dissolved in chloroform and spotted onto Silica Gel 60 TLC plates (Merck).
  • an increased kinase activity refers to a greater detection of radioactivity (indicating more incorporation of radiolabeled phosphate) per ⁇ g of pi 10a test protein, compared to incorporation per ⁇ g of pi 10a wild type protein, e.g., a protein having the sequence of SEQ ID NO-.56.
  • Mutations in the coding region of PIK3CA can be synonymous or non-synonymous mutations. Mutations in the noncoding or coding regions of a PIK3CA sequence can lead to altered RNA transcripts and/or altered protein as a result of differential post-transcriptional processing ⁇ e.g., mRNA splicing, nonsense-mediated decay, post-translational modification, etc.) as compared to wild type PIK3CA mRNA or pi 10a.
  • a PIK3CA sequence from a test sample e.g., a. sample containing cancer cells
  • a PIK3CA sequence from a different tissue in the same individual can be compared to a PIK3CA sequence from a different tissue in the same individual. A difference in the sequences from the two tissues indicates a mutation.
  • the PIK3CA sequence from a test sample can be compared to SEQ ID NO:55, with one or more differences between these two sequences indicative of a mutation.
  • PIK3CA activating mutations can be identified by reference to a wild type polypeptide or polynucleotide sequence where the number indicates the position within the polypeptide or polynucleotide sequence, the first letter indicates the identity of the residue or nucleotide in the wild type sequence and the second letter indicates the identity of the residue or nucleotide in the mutant sequence.
  • PIK3CA activating mutations are identified by reference to the wild type polypeptide sequence of SEQ ID NO: 56.
  • E545K refers to a polypeptide sequence for which the glutamate residue at position 545 has been mutated to lysine.
  • a PIK3CA activating mutation is selected from the group consisting of: Kl 1 IN, E542K, E545K, E545G, E545D, Q546K, Q546R, Ml 0431, Ml 043V, H1047R, H1047L, H1047Y, G1049R and G1049S. Subjects can be tested for one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, eleven or more, twelve or more, thirteen or more, or all fourteen of these mutations.
  • a PIK3CA activating mutation can also be identified by reference to a wild type polynucleotide sequence where the number indicates the position within the polynucleotide sequence, the first letter indicates the identity of the wild-type nucleotide, and the second letter indicates the identity of the mutant nucleotide.
  • the ">" character is placed between the first and second letter to denote that the change occurs from the nucleotide represented by the first letter to that of the second.
  • the underscore character indicates a range of nucleotides, "ins” indicates an insertion, and "del” indicates a deletion.
  • the number following "ins” or “del” indicates the number of nucleotides that are inserted or deleted, respectively.
  • a first number followed by a plus sign ("+”) or minus sign ("-"), followed by a second number indicates the presence of a mutation within an intron that is a number of positions downstream (for plus sign) or upstream (for minus sign) given by the second number with respect to the position in the coding sequence given by the first number.
  • a PIK3CA activating mutation is a mutation identified as PIK3CA (2188-7delT), with reference to SEQ ID NO:55, where the designation indicates that subjects bearing this mutation possess a PIK3CA gene in which the T nucleotide that occurs 7 positions upstream from position 2188 in the coding sequence is deleted.
  • a PIK3CA activating mutation is a mutation that gives rise to a higher expression level of pi 10a compared to wild type levels. Methods for detection and quantitation of pi 10a expression levels are known and have been described, for example, in U.S. Application Publication No. 2009/0192110.
  • Phosphatase and tensin homolog is encoded by the PTEN gene, and, like PIK3CA, is known to be mutated in the development of certain cancers.
  • PTEN has been identified as the phosphatase that catalyzes the reverse reaction of the reaction catalyzed by the PIK3CA-encoded protein (pi 10a), specifically the dephosphorylation of inositol polyphosphates.
  • a "PTEN polynucleotide sequence” refers to a
  • a PTEN polynucleotide encoding the PTEN protein can be a gene sequence (with or without introns), a cDNA, or an mRNA.
  • a polynucleotide sequence of a representative wild type human PTEN gene is SEQ ID NO:59.
  • the corresponding wild type PTEN polypeptide sequence is SEQ ID NO:60.
  • PTEN null mutation refers to any mutation or set of mutations within PTEN that leads to a decreased phosphatase activity compared to wild type PTEN, as a result of decreased activity or levels of the PTEN protein.
  • assays for determining phosphatase activity are known in the art.
  • PTEN phosphatase activity can be determined using an in vitro phosphatase assay. Such assays are known and described, for example, in U.S. Application Publication No. 2011/0150868, U.S. Patent No. US7745485, and Georgescu et al, PNAS (1999), 96(18): 10182-10187.
  • PTEN phosphatase activity can be measured with each assay performed in 50 ⁇ buffer containing 100 mM Tris-HCl (pH 8.0), 10 mM DTT, and 100 ⁇ water-soluble diC8-PIP3 (Echelon, Salt Lake City, Utah). Reactions containing PTEN can be
  • lysates can be precleared with protein G-agarose beads. After immunoprecipitation, the beads can be washed once in lysis buffer; five times in low stringency buffer containing 20 mM Hepes (pH 7.7), 50 mM NaCl, 0.1 mM EDTA, and 2.5 mM MgC12; and once in phosphatase assay buffer lacking PIP 3 . Reactions can be incubated for 40 min at 37° C and transferred to a 96- well plate. Release of phosphate from substrate may be measured using Biomol Green Reagent (Biomol Research Laboratories, Inc., Plymouth Meeting, Pa.).
  • PTEN phosphatase activity can be measured without
  • An exemplary reaction mixture for this assay includes 100 mM TrisHCl (pH 8), 10 mM dithiothreitol, 0.5 mM diC16 phosphatidylserine (PS), 25 ⁇ PIP 3 - diC16, BIOMOL PH-107 (Biomol Research Laboratories, Inc., Plymouth Meeting, Pa.) and 50 ⁇ g/ml purified recombinant wild type or mutant PTEN.
  • Lipids can be prepared in organic solvents and dispensed into 1.5 ml microfuge tubes followed by solvent removal under reduced pressure. Buffer can then be added and a lipid suspension formed by sonication.
  • PTEN phosphatase assays are initiated by the addition of PTEN and carried out at 37° C. At different time points, 15 ⁇ of 100 ⁇ NEM (N-ethylmaleimide) is added to 10 ⁇ of reaction mixture followed by rapid centrifugation at 18,000xg for 15 minutes at 4° C. Liberated inorganic phosphate is detected in twenty microliters of supernatant using the Malachite green assay with reference to an inorganic phosphate standard curve. Malachite green reaction with inorganic phosphate is detected spectrophotometrically at 620 nm wavelength.
  • NEM N-ethylmaleimide
  • a PTEN null mutation gives rise to a lowered expression level of PTEN compared to wild type levels.
  • Methods for detection and quantitation of PTEN expression levels are known and have been described, for example, in U.S. Application Publication No. 2010/0303809.
  • a PTEN null mutation can be any type of mutation, including substitution mutations, deletion mutations, and insertion mutations. Mutations in the coding region of PTEN can be synonymous or non-synonymous mutations. Mutations in the noncoding or coding regions of PTEN can lead to mutant RNA transcripts and/or mutant protein as a result of differential post-transcriptional processing ⁇ e.g., mRNA splicing, nonsense-mediated decay, post- translational modification, etc.) as compared to wild type PTEN. PTEN null mutations are known in the art, see Jhawer et al, Cancer Res (2008), 68:1953-1961, incorporated herein by reference in its entirety.
  • PTEN null mutations can be identified by reference to a wild type polypeptide sequence where the number indicates the position within the polypeptide sequence, the first letter indicates the identity of the residue in the wild type sequence and the second letter indicates the identity of the residue in the mutant sequence.
  • PTEN null mutations are identified by reference to the wild type polypeptide sequence of SEQ ID NO: 60.
  • "LI 12Q" refers to a polypeptide sequence for which the leucine residue at position 112 has been mutated to glutamine.
  • a PTEN null mutation is a mutation in which an amino acid is mutated to a stop codon, identified by reference to a wild type polypeptide sequence in which the number indicates the position within the sequence, the first letter indicates the identity of the wild type amino acid, and "*" indicates substitution of a stop codon at this position. For example, "Q149*” indicates that the glutamine at position 149 is mutated to a stop codon, resulting in a truncated polypeptide.
  • a PTEN null mutation is a frameshift mutation that introduces a stop codon, resulting in a truncated sequence.
  • Such mutations can be identified by reference to a wild type polypeptide where the number indicates the position where the frameshift occurs, "fs" indicates a frameshift mutation, "*” indicates introduction of a stop codon, and the final number indicates the position of the stop from the first amino acid affected by the frameshift.
  • a PTEN null mutation is selected, with reference to SEQ ID NO:60, from the group consisting of: Q149*, LI 12Q, F90fs*9, K267fs*9, V175fs*3, and P248fs*5. Subjects can be tested for one or more, two or more, three or more, four or more, five or more, or all six of these mutations.
  • a PTEN null mutation can also be identified by reference to a wild type
  • polynucleotide sequence where the number indicates the position within the polynucleotide sequence, the first letter indicates the identity of the wild-type nucleotide, and the second letter indicates the identity of the mutant nucleotide.
  • the ">" character is placed between the first and second letter to denote that the change occurs from the nucleotide represented by the first letter to that of the second.
  • the underscore character indicates a range of nucleotides, "ins” indicates an insertion, and “del” indicates a deletion. The number following "ins” or “del” indicates the number of nucleotides that are inserted or deleted, respectively.
  • 165_209del45 refers to a mutant in which the 45 nucleotides spanning from position 165 to position 209 are deleted.
  • a first number followed by a plus sign ("+”) or minus sign ("-"), followed by a second number indicates the presence of a mutation within an intron that is a number of positions downstream (for plus sign) or upstream (for minus sign) given by the second number with respect to the position in the coding sequence given by the first number.
  • a PTEN null mutation is selected, with reference to SEQ ID NO:59, from the group consisting of: l_164dell64, 165_209del45, 253+lOT, 270delT, 335T>A, 4450T, 524_558del35, 741_742insA, 800delA, and 1026+1 G>T. Subjects can be tested for one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or all ten of these mutations.
  • a PTEN polynucleotide sequence from a test sample e.g., a sample containing cancer cells
  • a PTEN polynucleotide sequence in a different tissue from the same individual can be compared to a PTEN polynucleotide sequence in a different tissue from the same individual. A difference in the sequences from the two tissues indicates a mutation.
  • the PTEN polynucleotide sequence determined in a PTEN gene in a test sample can be compared to SEQ ID NO:59.
  • a difference between the test sample sequence and SEQ ID NO:59 can be identified as a mutation.
  • Methods described herein for the detection of PIK3CA activating mutations also can be applied using the appropriate primers and probes to detect PTEN null mutations, and vice versa.
  • the Sanger Wellcome database contains information about PTEN deletions and truncations, and therefore may also be useful in determining a PTEN null mutation (see Wang et al, PNAS (2009) 106(15):6279-6284). 5.3. Detecting PIK3CA Activating and PTEN Null Mutations
  • the presence or absence of a PIK3CA activating mutation or a PTEN null mutation can be determined in a subject using a variety of methods known in the art.
  • a subject is tested for a PIK3CA activating mutation and/or a PTEN null mutation.
  • the testing can be performed in conjunction with T WEAKR agonist therapy in order to treat a T WEAKR-positive cancer.
  • the testing is preferably performed prior to treatment with the TWEAKR agonist, and can be carried out at the same time the subject's cancer is tested for TWEAKR expression, for example as described in Section 5.5.
  • the determination of a PIK3CA activating mutation or a PTEN null mutation can be made at the nucleic acid level and/or at the protein level.
  • the nucleic acid and/or proteins used for the determination can be obtained from biological samples, including, but not limited to tissues, cells, biological fluids and isolates thereof, including those isolated from a subject, as well as those present within a subject.
  • biological samples comprise cells, most preferably tumor cells, that are isolated from body samples, such as, but not limited to, smears, sputum, biopsies, secretions, cerebrospinal fluid, bile, blood, lymph fluid, urine and feces, or tissue which has been removed from organs, such as breast, lung, pancreas, intestine, skin, cervix, prostate, and stomach.
  • Tissues suspected of being cancerous can be tested for a PIK3CA activating mutation or a PTEN null mutation, as can body samples that may be expected to contain sloughed-off cells from tumors or cells of cancers.
  • An allele containing a PIK3CA activating mutation is referred to as a PIK3CA mutant allele, and the corresponding wild type region of DNA is referred to as a PIK3CA wild type allele.
  • An allele containing a PTEN null mutation is referred to as a PTEN mutant allele, and the corresponding wild type region of DNA is referred to as a PTEN wild type allele.
  • Methods for detecting mutations in DNA include genotyping, microarrays, direct sequencing, restriction mapping, Restriction Fragment Length Polymorphism,
  • primers flanking a PIK3CA or PTEN allele are selected and used to amplify that region.
  • the amplified region is then sequenced using DNA sequencing techniques known in the art and analyzed for the presence of a PIK3CA activating or PTEN null mutation.
  • a PIK3CA activating or PTEN null mutation in a nucleic acid can be detected using a probe.
  • an amplified region comprising a wild type or mutant allele is hybridized using a composition comprising a probe specific for the mutation under stringent hybridization conditions.
  • isolated nucleic acids that bind to mutant alleles at high stringency may be used as probes to determine the presence of the mutation.
  • Nucleic acids may be labeled with a detectable marker.
  • the marker or label is typically capable of producing, either directly or indirectly, a detectable signal.
  • the label may be radioopaque or a radioisotope, such as H, ' , "P, "S, " ⁇ , a fluorescent
  • fluorescein fluorescein
  • a PIK3CA activating or PTEN null mutation can also be detected using restriction enzymes.
  • amplified products can be digested with a restriction enzyme that specifically recognizes sequence comprising a mutant allele, but does not recognize the corresponding wild type allele or vice versa.
  • PCR may be used to amplify DNA comprising a PIK3CA or PTEN allele, and amplified PCR products are subjected to restriction enzyme digestion under suitable conditions and restriction products are assessed. If, for example, a PIK3CA or PTEN mutant allele corresponds to a sequence digested by the restriction enzyme, digestion is indicative of detecting that particular PIK3CA or PTEN mutant allele. Restriction products may be assayed electrophoretically as is common is the art.
  • PIK3CA and PTEN mutant alleles may also be detected by a variety of other methods known in the art.
  • PCR and RT-PCR and primers flanking a PIK3CA or PTEN allele can be employed to amplify sequences and transcripts, respectively, in a sample comprising DNA (for PCR) or RNA (for RT-PCR).
  • the amplified products are optionally sequenced to determine whether the PIK3CA or PTEN allele in the sample is mutant or wild type.
  • a PIK3CA or PTEN allele in a sample can be detected by a variety of other techniques known in the art including microarrays, hybridization assays, PCR based assays, molecular beacons, dynamic allele-specific hybridization (DASH) and/or
  • Mutant PIK3CA or PTEN polynucleotide sequences may also be identified by methods which detect particular mRNAs in cells. These include hybridization assays using complementary DNA probes (such as in situ hybridization using labeled mutant or wild type PIK3CA or PTEN riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for PIK3CA or PTEN polynucleotide sequences, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like).
  • complementary DNA probes such as in situ hybridization using labeled mutant or wild type PIK3CA or PTEN riboprobes, Northern blot and related techniques
  • nucleic acid amplification assays such as RT-PCR using complementary primers specific for PIK3CA or PTEN polynucleotide sequences, and other amplification type detection methods, such as,
  • Mutant pi 10a or PTEN protein bearing a PIK3CA activating mutation or PTEN null mutation can be detected using a specific agent, most preferably an antibody, that is itself detectably labeled, or using an unlabeled antibody specific for mutant pi 10a or PTEN protein and a second antibody that is detectably labeled and recognizes the unlabeled antibody specific for mutant pi 10a or PTEN protein.
  • a specific agent most preferably an antibody, that is itself detectably labeled, or using an unlabeled antibody specific for mutant pi 10a or PTEN protein and a second antibody that is detectably labeled and recognizes the unlabeled antibody specific for mutant pi 10a or PTEN protein.
  • any molecule that can be detectably labeled and that specifically binds to mutant pi 10a or PTEN protein can be used in the practice of the methods of the disclosure.
  • a two-component immunohistochemical staining system may be used to differentially stain mutant pi 10a or PTEN protein and the tissue or cell sample so that the stained mutant pi 10a or PTEN protein can be more readily distinguished from the counterstained tissue or cell sample.
  • pi 10a or PTEN protein can also be enriched by affinity purification using, for example, an anti-pl 10a or anti-PTEN antibody, accordingly, and the enriched protein can be sequenced by mass spectrometric methods known in the art to determine the presence of a PIK3CA activating or PTEN null mutation.
  • PIK3CA activating mutation or a PTEN null mutation in a given cell line can also be determined by reference to the Sanger Wellcome database of somatic mutations related to human cancers (see Wang et al, PNAS (2009) 106(15):6279-6284).
  • downstream pathway members can also be used as an indicator of PI3K pathway activation status as a result of upregulation of PIK3CA protein levels and/or the presence of activating mutations in PIK3CA.
  • downstream pathway members phosphorylated when the PI3K pathway is activated include PDK1 (S241, Y373 Y376, or S396), Akt (S473 or T308), FoxOl (T24, S249, S256, or S319), Fox04 (SI 93 or S262), Fox03a (T32, S253, S318, S321, or S644). Reagents specific for phosphorylated forms of these proteins are readily available.
  • a TWEAKR agonist is an agent capable of activating a
  • TWEAKR signaling cascade leading to one or more of the biological activities associated with TWEAKR such as killing TWEAKR positive cancer cells by apoptosis, inhibiting proliferation and/or survival of TWEAKR positive cancer cells, and inducing chemokine or cytokine release in TWEAKR positive cancer cells.
  • TWEAKR agonists induce apoptosis in cells expressing TWEAKR. Without being bound by theory, induction of apoptosis by TWEAKR agonists is postulated to occur through caspase activation.
  • induction of apoptosis by TWEAKR agonists can be assayed by measuring activation of caspase 3/7, using methods known in the art, including commercial kits such as Caspase-Glo® 3/7 Assay Systems from Promega®. Assays for apoptosis may also be performed by terminal deoxynucleotidyl transferase-mediated digoxigenin-11-dUTP nick end labeling (TUNEL) assay (Lazebnik et al. (1994), Nature: 371:346).
  • TUNEL terminal deoxynucleotidyl transferase-mediated digoxigenin-11-dUTP nick end labeling
  • a TWEAKR agonist inhibits proliferation ⁇ i.e. cell division) and/or survival of a cell, and in particular, a TWEAKR positive cancer cell.
  • TWEAKR agonists including TWEAK and anti-TWEAKR agonist antibodies, have been shown to inhibit proliferation and/or survival of TWEAKR positive cancer cell lines in in vitro and in vivo growth inhibition assays (Culp et al, 2010, Clin. Cancer Res. 16(2): 497-508).
  • Inhibition of proliferation and/or survival can be measured by an in vitro growth inhibition assay as described in Example 1 below.
  • a cancer cell line known to be sensitive to TWEAKR agonists such as BT549 or H358, is cultured at 500 cells per well in triplicate with 5-10 ⁇ g/ml of the proposed TWEAKR agonist for 5 days in 96 well plates.
  • Growth inhibition is determined using CellTiter-BlueTM (Promega): cells are incubated with the reagent and fluorescence emitted at 590 nm is measured and used to calculate the effect on viable cells of the proposed TWEAKR agonist relative to a control, such as an IgGl control.
  • a test compound is considered to be a TWEAKR agonist if it induces growth inhibition of at least about 20% relative to control treatment.
  • cells can be incubated with anti-TWEAKR antibody (10 ⁇ g/ml) and an antibody (5 ⁇ g/ml) or antibody fragment (3.5 ⁇ g/ml) that specifically binds to the anti-TWEAKR antibody.
  • the antibody or antibody fragment is preferably species- and isotype-specific.
  • the anti-TWEAKR antibody is a humanized IgG antibody
  • a F(ab') 2 antibody fragment that is a goat anti-human
  • Agonist activity can be determined by measuring the release of cytokines and/or chemokines in an in vitro cell growth assay. For example, in a typical assay the cells are incubated in vitro with a TWEAKR agonist. Twenty four hours later, the cell supernatant is assessed for the presence of cytokines and/or chemokines using an ELISA assay or a commercial fluorescent bead-based multiplex assay (e.g., Luminex®, Upstate). In some embodiments, the chemokine that is measured for release is IL-8.
  • TWEAKR agonists include the natural ligand of TWEAKR, TWEAK. TWEAK forms trimers and is thought to promote activation of TWEAKR via trimerization of the receptor. See Winkles, 2008, Nature Reviews Drug Discovery 7: 411-425. Therefore, it is expected that TWEAKR agonists can also be proteins that form multimers, e.g., trimers and are capable of binding to and promoting multimerization of TWEAKR. Accordingly, TWEAKR agonists include fusions of TWEAK, or a fragment of TWEAK capable of binding TWEAKR, to polypeptides comprising an oligomerization domain.
  • Polypeptides comprising suitable oligomerization domains include CD8 (Nakayama et al., 2000, J. Exp. Med. 192:1373), collagen (Frank et al., 2001, J. Mol. Bio. 308:1081-1089), bacteriophage T4 fibritin (Yang et al., 2002, J. Virology 76(9):4634-4642), engineered leucine/isoleucine zipper polypeptides (Harbury, 1994, Nature 371(6492):80-83), macrophage scavenger receptor (U.S. Pat. No.
  • TNF tumor necrosis factor
  • Methods of making fusion proteins are well known in the art, and involve combining coding sequences of two or more polypeptides to generate a coding sequence of a fusion protein.
  • Techniques for the manipulation of nucleic acids including techniques for the synthesis, isolation, cloning, detection, and identification are well known in the art and are well described in the scientific and patent literature. See, e.g., Sambrook et al, eds., Molecular Cloning: A Laboratory Manual (2nd Ed.), Vols. 1-3, Cold Spring Harbor
  • TWEAKR agonists also include polypeptides of TWEAK engineered or modified to have an increased serum half-life.
  • Suitable engineered TWEAK proteins include fusions of TWEAK, or a fragment of TWEAK capable of binding TWEAKR, to human
  • HSA human serum albumin
  • Modified TWEAK proteins also include conjugates of TWEAK, or a fragment of TWEAK capable of binding TWEAKR, and high molecular weight dextrans or polyethylene glycol (PEG), which can be made according to methods known in the art. See U.S. Pat. No. 5,177,059 and 7,587,286. PEG has been shown to prolong half-life and reduce
  • a specific embodiment of a TWEAKR agonist is an anti-TWEAKR agonist antibody.
  • Anti-TWEAKR agonist antibodies specifically bind to TWEAKR proteins.
  • An example of a TWEAKR protein is the human TWEAKR protein encoded by the nucleotide sequence given by SEQ ID NO:l, or the protein of the amino acid sequence given by SEQ ID NO:2. See Table 2 below.
  • the antibody preferably binds the TWEAKR from the same species as the subject.
  • an antibody to human TWEAKR is administered.
  • a TWEAKR agonist antibody need not be a pure agonist.
  • a TWEAKR agonist antibody can also have TWEAKR antagonist activity.
  • Antagonist activity refers to the ability of test agent (e.g. a TWEAKR agonist antibody) to inhibit the induction by TWEAK of one or more biological responses associated with TWEAKR.
  • test agent e.g. a TWEAKR agonist antibody
  • cytokine/chemokine release assay described above can be performed by incubating cells in vitro with an anti-TWEAKR antibody +/- TWEAK.
  • An anti-TWEAKR agonist antibody can bind to TWEAKR protein with a K D of at least about ⁇ ⁇ , at least about 0.1 ⁇ or better, at least about 0.01 ⁇ , and at least about 0.001 ⁇ or better.
  • antibody refers to an immunoglobulin molecule that specifically binds to, or is immunologically reactive with, a particular antigen, and includes polyclonal, monoclonal, chimeric, humanized, fully human, genetically engineered and otherwise modified forms of antibodies.
  • a full-length antibody contains two heavy chains and two light chains, each of which comprises complementarity determining regions (CDRs), also known as hypervariable regions, and more highly conserved framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs highly conserved framework regions
  • the amino acid position/boundary delineating a hypervariable region of an antibody can vary, depending on the context and the various definitions known in the art.
  • Some positions within a variable domain may be viewed as hybrid hypervariable positions in that these positions can be deemed to be within a hypervariable region under one set of criteria while being deemed to be outside a hypervariable region under a different set of criteria.
  • One or more of these positions can also be found in extended hypervariable regions.
  • the present disclosure includes antibodies comprising modifications in these hybrid hypervariable positions.
  • variable domains of native heavy and light chains each comprise four FR regions, largely by adopting a ⁇ -sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the target binding site of antibodies (See Kabat et al. ,
  • an anti-TWEAKR agonist antibody can be an antibody fragment.
  • antibody fragment refers to a portion of a full-length antibody capable of multivalent binding to the target, where multivalent refers to bivalent, trivalent, tetravalent, etc.
  • F(ab') 2 fragments examples include F(ab') 2 fragments.
  • F(ab') 2 fragments are bivalent, having two antigen-binding F(ab) portions linked together by disulfide bonds, and contain portions of VH and VL chains.
  • References to "VH” refer to the variable region of an immunoglobulin heavy chain of an antibody.
  • References to "VL” refer to the variable region of an immunoglobulin light chain. It is in this configuration that the three CDRs— typically denoted CDR1, CDR2, and CDR3— of each variable domain interact to define a target binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer target binding specificity to the antibody.
  • An F(ab') 2 fragment can be produced by pepsin cleavage of a whole antibody, removing most of the immunoglobulin constant region (Fc) portion of the antibody. Such F(ab') 2 fragments clear more rapidly from the circulation of the animal or plant, and may have less non-specific tissue binding than an intact antibody (Wahl et al. , 1983, J. Nucl. Med. 24:316). Multivalent antibody fragments can also be produced by linking CDRs using chemical couplings known to those of ordinary skill in the art.
  • the term "monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • Monoclonal antibodies useful with the present disclosure can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • the term monoclonal antibody (mAb) is meant to include both intact molecules, as well as, antibody fragments (such as, for example, F(ab') 2 fragments) which are capable of specifically binding to a protein.
  • the anti-TWEAKR agonist antibodies of the disclosure can be chimeric antibodies.
  • the term "chimeric" antibody as used herein refers to an antibody having variable sequences derived from a non-human immunoglobulins, such as rat or mouse antibody, and human immunoglobulins constant regions, typically chosen from a human immunoglobulin template. Methods for producing chimeric antibodies are known in the art. See, e.g., Morrison, 1985, Science 229(4719): 1202-7; Oi et al, 1986, BioTechniques 4:214-221; Gillies et al, 1985, J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715;
  • the anti-TWEAKR agonist antibodies of the disclosure can be humanized.
  • “Humanized” forms of non-human ⁇ e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as F(ab') 2 or other target-binding subsequences of antibodies) which contain minimal sequences derived from non-human immunoglobulin.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody can also comprise at least a portion of an Fc, typically that of a human
  • the anti-TWEAKR agonist antibodies of the disclosure can be fully human antibodies. Fully human anti-TWEAKR agonist antibodies can be desirable for therapeutic treatment of human patients.
  • "fully human antibodies” include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins.
  • Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See U.S. Pat. Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645; WO 98/50433; WO 98/24893; WO 98/16654; WO 96/34096; WO 96/33735; and WO 91/10741. Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes.
  • Fully human antibodies that recognize a selected epitope can be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a mouse antibody
  • is used to guide the selection of a fully human antibody recognizing the same epitope Jespers et al, 1988, Biotechnology 12:899-903.
  • anti-TWEAKR agonist antibodies can be of any of the recognized isotypes.
  • anti-TWEAKR agonist antibodies are one of the four human IgG isotypes, i.e., IgGl, IgG2, IgG3 and IgG4, or one of the four mouse IgG isotypes, i.e., murine IgGl, murine IgG2a, murine IgG2b, or murine IgG3.
  • the anti- TWEAKR antibodies are of the human IgGl isotype.
  • Anti-TWEAKR agonist antibodies useful in the present methods include antibodies that induce antibody-dependent cytotoxicity (ADCC) of TWEAKR-expressing cells.
  • ADCC antibody-dependent cytotoxicity
  • the ADCC of an anti-TWEAKR antibody can be improved by using antibodies that have low levels of or lack fucose.
  • Antibodies lacking fucose have been correlated with enhanced ADCC (antibody-dependent cellular cytotoxicity) activity, especially at low doses of antibody (Shields et al, 2002, J. Biol. Chem. 277:26733-26740; Shinkawa et al, 2003, J. Biol. Chem. 278:3466).
  • Methods of preparing fucose-less antibodies include growth in rat myeloma YB2/0 cells (ATCC CRL 1662).
  • YB2/0 cells express low levels of FUT8 mRNA, which encodes an enzyme (a-l,6-fucosyltransferase) necessary for fucosylation of polypeptides.
  • Alternative methods for increasing ADCC activity include mutations in the Fc portion of an anti-TWEAKR agonist antibody, particularly mutations which increase antibody affinity for an FcyR receptor.
  • a correlation between increased FcyR binding with mutated Fc has been demonstrated using targeted cytoxicity cell-based assays (Shields et al, 2001, J. Biol. Chem.
  • an anti-TWEAKR agonist antibody of the disclosure has a constant region that binds FcyRIIA, FcyRIIB and/or FcyRIIIA with greater affinity than the corresponding wild type constant region. Mutations in the Fc portion of an antibody that result in increased ADCC activity and methods of generating immunoglobulins with increased ADCC activity are known in the art and described in various references, e.g., U.S. Pat. No. 8,039,592, U.S. Pat. No. 7,632,497, U.S. Pat. No. 7,183,387, U.S. Pat. Pub. No. 2009/0074762, WO 00/42072.
  • ADCC activity can be monitored and quantified using a 51 Cr release assay.
  • Target cells are loaded with 5I Cr, which is released in culture supernatant upon damage to the plasma membrane in the presence of immune effector cells and an anti-TWEAKR agonist antibody.
  • the target cells can be derived from solid tumors, for example lung, pancreatic, breast, or renal cancer cells.
  • the anti-TWEAKR agonist antibodies induce at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, or at least 80% cytotoxicity in the target cells.
  • An example of an ADCC assay that can be used to measure ADCC of an anti-TWEAKR agonist antibody is provided in Culp et al, 2010, Clin. Cancer Res. 16(2):497-508.
  • Anti-TWEAKR agonist antibodies or fragments thereof as described herein can be antibodies or antibody fragments whose sequence has been modified to reduce at least one constant region-mediated biological effector function relative to the corresponding wild type sequence.
  • the immunoglobulin constant region segment of the antibody can be mutated at particular regions necessary for Fc receptor (FcR) interactions (see e.g., Canfield and Morrison, 1991, J. Exp. Med. 173:1483-1491; and Lund et al, 1991, J. Immunol.
  • Reduction in FcR binding ability of the antibody can also reduce other effector functions which rely on FcR interactions, such as opsonization and phagocytosis.
  • Anti-TWEAKR agonist antibodies or fragments thereof as described herein can be antibodies or antibody fragments that have been modified to increase or reduce their binding affinities to the fetal Fc receptor, FcRn, or serum half-life, for example by mutating the immunoglobulin constant region segment at particular regions involved in FcRn interactions (see, e.g., WO 2005/123780 and U.S. Pat. No. 5,739,277).
  • anti-TWEAKR agonist antibodies and sequences of VH, VL and CDRs which are described in U.S. Pat. Pub. No. 2009/0074762 and provided in Table 2 herein, include:
  • an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, comprising a heavy chain variable region corresponding to SEQ ID NO: 3 and a light chain variable region corresponding to SEQ ID NO: 4 (PDL192);
  • an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, comprising a heavy chain variable region corresponding to SEQ ID NO: 11 and a light chain variable region corresponding to SEQ ID NO: 12 (PDL400);
  • V H CDR1 is XiYWMX 2 (SEQ ID NO:49)
  • V H CDR2 is EIRX 3 KSX 4 NYATX 5 HYAESX 6 KG (SEQ ID NO:50)
  • V H CDR3 is X 7 X 8 ADX 9 X 10 DY (SEQ ID NO:51)
  • V L CDR1 is X n ASQSVSTSX 12 YSYMX 13 (SEQ ID NO:52);
  • V L CDR2 is YAX 14 X 15 LX 16 S (SEQ ID NO:53);
  • V L CDR3 is QHSWEX 17 PX 18 T (SEQ ID NO:54), wherein: Xj is selected from K, N, R, and S; X 2 is selected from N and S; X 3 is selected from L and V; j is selected from D and N; X 5 is T or no amino acid;
  • V H CDRl is SYWMS (SEQ ID NO: 13)
  • V H CDR2 is EIRLKSDNYATHYAESVKG (SEQ ID NO: 19)
  • V H CDR3 is
  • V L CDRl is RASQSVSTSSYSYMH (SEQ ID NO:31)
  • V L CDR2 is YASNLES (SEQ ID NO:37)
  • V L CDR3 is QHSWEIPYT (SEQ ID NO:43);
  • V H CDRl is NYWMS (SEQ ID NO: 15)
  • V H CDR2 is EIRLKSDNYATHYAESVKG (SEQ ID NO:21)
  • V H CDR3 is GFADYFDY (SEQ ID NO:27)
  • V L CDRl is RASQSVSTSSYSYMQ (SEQ ID NO:33)
  • V L CDR2 is YATNLDS (SEQ ID NO:39)
  • V L CDR3 is QHSWEIPYT (SEQ ID NO:45);
  • V H CDRl is KYWMN (SEQ ID NO: 16)
  • V H CDR2 is EIRLKSDNYATHYAESAKG (SEQ ID NO:22)
  • V H CDR3 is
  • V L CDRl is KASQSVSTSTYSYMQ (SEQ ID NO:34)
  • V L CDR2 is YASKLDS (SEQ ID NO:40)
  • V L CDR3 is QHSWELPYT (SEQ ID NO:46);
  • V H CDRl is RYWMS (SEQ ID NO: 17)
  • V H CDR2 is EIRVKSDNYATTHYAESVKG (SEQ ID NO:23)
  • V H CDR3 is
  • V L CDRl is RASQSVSTSSYSYMH (SEQ ID NO:35)
  • V L CDR2 is YASKLDS (SEQ ID NO:41)
  • V L CDR3 is QHSWEIPWT (SEQ ID NO:47);
  • V H CDR1 is NYWMN (SEQ ID NO: 17)
  • V H CDR2 is EIRLKSNNYATHYAESVKG (SEQ ID NO:24)
  • V H CDR3 is
  • V L CDR1 is RASQSVSTSTYSYMH (SEQ ID NO:36)
  • V L CDR2 is YASNLES (SEQ ID NO:42)
  • V L CDR3 is QHSWEIPYT (SEQ ID NO:48).
  • Anti-TWEAKR agonist antibodies of the present disclosure also include antibodies that compete for binding to TWEAKR with a reference antibody of interest. Any of the antibodies described herein can be used as a reference antibody in a competition assay.
  • a competition assay can be carried out between a test antibody and a reference antibody. The assay is conducted by first labeling the reference antibody with a detectable label, such as, biotin, or an enzymatic, radioactive, or fluorescent label to enable detection. The unlabeled test antibody is incubated (in fixed or increasing amounts) with a known amount of a detectable label, such as, biotin, or an enzymatic, radioactive, or fluorescent label to enable detection. The unlabeled test antibody is incubated (in fixed or increasing amounts) with a known amount of
  • TWEAKR forming an anti-TWEAKR antibody/ TWEAKR complex.
  • the labeled reference antibody is then added to the complex.
  • the intensity of the complexed label is measured. If the test antibody competes with the labeled antibody by binding to an overlapping epitope, the intensity will be decreased relative to the binding of the labeled reference antibody in the absence of the test antibody. Numerous methods for carrying out binding competition assays are known.
  • An antibody is considered to compete for binding TWEAKR with a reference anti-TWEAKR agonist antibody, and thus considered to bind approximately the same or overlapping epitope of TWEAKR as the reference antibody, if it reduces binding of the reference antibody by at least 50% at a test antibody concentration in the range of 0.01 to 100 ⁇ g/mL, although higher levels of reduction, for example, 60%, 70%, 80%, 90% or even 100%, may be desirable.
  • TWEAKR positive cancer is a cancer in which TWEAKR is overexpressed as compared to matched non-cancer tissue.
  • TWEAKR expression can be determined by detecting the presence of TWEAKR mRNA or TWEAKR protein, using standard techniques such as PCR, Northern blotting, microarray analysis (for mRNA detection) or Western blotting, ELISA, or immunohistochemistry (for protein detection).
  • TWEAKR has been shown to be overexpressed in a number of human cancers. See Culp et al, 2010, Clin. Cancer Res. 16(2):497-508; Feng et al, 2000, Am. J. Pathol. 156:1253-1261; Han et al, 2002, Cancer Res. 62:2890-2896; Wang et al, 2006, Oncogene 25:3346-3356.
  • TWEAKR positive cancers include breast cancer, lung cancer, melanoma, ovarian cancer, uterine cancer, colon cancer, renal cancer, pancreatic cancer, cervical cancer, bladder cancer, head and neck cancer, glioblastoma, esophageal cancer, sarcomas, and salivary gland cancer.
  • the present disclosure provides methods of treating a TWEAKR positive cancer.
  • a subject who has tested negative for at least one PIK3CA activating mutation is treated with an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • a subject who has tested negative for at least one PTEN null mutation is treated with an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
  • a subject who has tested negative for at least one PIK3CA activating mutation has also tested negative for at least one PTEN null mutation.
  • a subject who has tested negative for at least one PTEN null mutation may also have tested negative for at least one PIK3CA activating mutation.
  • the TWEAKR agonists are administered to a subject in an amount effective to promote a therapeutic benefit.
  • the amount effective to promote a therapeutic benefit refers to the amount of a pharmaceutical formulation or composition that is sufficient to cure, alleviate, attenuate or at least partially arrest the cancer and/or its symptoms, and/or complications.
  • Clinical methods for determining a therapeutic benefit are well known to those of ordinary skill in the art and may be determined empirically using routine
  • an amount effective to promote a therapeutic benefit is an amount capable of invoking one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, slowing down and complete growth arrest; (2) reduction in the number of cancer cells; (3) reduction in tumor size; (4) inhibition (i.e., reduction, slowing down or complete stopping) of cancer cell infiltration into peripheral organs; (5) inhibition (i.e., reduction, slowing down or complete stopping) of cancer cell metastasis; (6) enhancement of anti-cancer immune response, which may, but does not have to, result in the regression or rejection of a tumor; and/or (7) relief, to some extent, of one or more symptoms associated with the disorder.
  • Tumor size and/or number can be measured using various scanning techniques, such as, but not limited to CT, MRI, functional MRI, SPECT and PET, as well as other methods known to those skilled in the art.
  • the TWEAKR agonist can be used alone, as monotherapy, or in combination with or adjunctive to other therapies commonly used to treat the specific type (e.g., lung, breast, colon, etc.) of TWEAKR positive cancer.
  • Other therapies include surgery, radiation therapy, and treatment with conventional therapeutic agents, such as targeted agents, conventional chemotherapy agents, and hormonal therapy agents.
  • the conventional standard of care for a particular cancer is known to those skilled in the art and thus, one of skill in the art will be able to select suitable other therapy to combine with the TWEAKR agonist described herein.
  • the TWEAKR agonist and other therapy can be administered simultaneously, successively, or separately. Therapies that can be combined with TWEAKR agonist therapy are described in, e.g., U.S. Pub. No. US
  • the TWEAKR agonist can be used in combination with conventional therapeutic agents used to treat a particular cancer.
  • therapeutic agents that are presently used to treat cancer, and thus can be used in combination with the TWEAKR agonist described herein, include, but are not limited to, targeted agents, conventional chemotherapy agents, and hormonal therapy agents.
  • the TWEAKR agonist described herein can be used in combination with targeted agents.
  • Targeted agents include, but are not limited to, antiangiogenic agents (such as bevacizumab, sunitinib, sorafenib, temsirolimus, 2- methoxyestradiol or 2ME2, finasunate, PTK787, and vandetanib), EGFR inhibitors (such as erlotinib, cetuximab, panitumumab, gefinitib, lapatinib, and trastuzumab),
  • antiangiogenic agents such as bevacizumab, sunitinib, sorafenib, temsirolimus, 2- methoxyestradiol or 2ME2, finasunate, PTK787, and vandetanib
  • EGFR inhibitors such as erlotinib, cetuximab, panitumumab, gefinitib, lapatini
  • immunomodulators such as rituximab, alemtuzumab, and aldesleukine
  • proteasome inhibitors such as bortezomib, PR-171, and NPI-052
  • integral inhibitors such as natalizumab, volociximab, etaracizumab, and cilengitide
  • pro-apoptotic agents such as mapatumumab, lexatumumab, AMG951, ABT-737, oblimersen, and plitidepsin
  • agents with other mechanisms of action such as imatinib, dasatinib, lenalidomide, thalidomide, aldesleukin, and interferon alpha).
  • the TWEAKR agonist described herein can be used in combination with conventional chemotherapy agents.
  • Conventional chemotherapy agents include, but are not limited to, alkylating agents (such as oxaliplatin, carboplatin, cisplatin, cyclophosphamide, melphalan, ifosfamide, uramustine, chlorambucil, mechloethamine, thiotepa, busulfan, temozolomide, and dacarbazine), anti-metabolites (gemcitabine, cytosine arabinoside, Ara-C, capecitabine, 5FU (5-fluorouracil), azathioprinc, mercaplopurine (6- MP), 6-thioguanine, aminopterin, pemetrexed, and methotrexate), plant alkaloids and terpenoids (such as docetaxel, paclitaxel, protein-bound paclitaxel, vincristine, vinblastin,
  • the TWEAKR agonist described herein can be used in combination with hormonal therapy agents including, but not limited to, anastrozole, letrozole, goserelin, and tamoxifen.
  • concentration of a TWEAKR agonist in formulations administered by methods of the disclosure varies widely from about 0.1 to 100 mg/ml, but is often in the range 1 to 20 mg/ml.
  • the appropriate dosage of the antibody will depend on the severity and course of disease, the patient's clinical history and response, the toxicity of the antibodies, and the discretion of the attending physician.
  • the dose can range from about 0.1 to about 1 mg/kg, about 1 to about 5 mg/kg, about 1 to about 10 mg/kg, and about 10 to about 20 mg/kg.
  • the antibodies are suitably administered to the patient at one time or over a series of treatments.
  • the proper dosage and treatment regimen can be established by monitoring the progress of therapy using conventional techniques known to the people skilled of the art.
  • the methods described herein provide for administration of a TWEAKR agonist to a patient intravenously as a bolus or by continuous infusion over a period of time; or by intramuscular,
  • compositions to be administered are formulated for pharmaceutical use.
  • the compositions optionally further comprise a carrier.
  • compositions described herein typically comprise a TWEAKR agonist and a pharmaceutical carrier, and, commonly they comprise a solution of a TWEAKR agonist, or a cocktail thereof, dissolved in an acceptable carrier, preferably an aqueous carrier.
  • aqueous carriers can be used, e.g., water for injection (WFI), or water buffered with phosphate, citrate, acetate, etc. to a pH typically of 5.0 to 8.0, most often 6.0 to 7.0, and/or containing salts such as sodium chloride, potassium chloride, etc. to make the composition isotonic.
  • WFI water for injection
  • the carrier can also contain excipients such as human serum albumin, polysorbate 80, sugars or amino acids to protect the agonist.
  • Administration of a TWEAKR agonist can trigger killing of cancer cells.
  • the cancer cells can be present within a solid tumor, the lymph system, or in the bloodstream.
  • the therapeutic methods described herein are usually applied to human patients, but can be applied to other mammals.
  • TWEAKR agonist can induce apoptosis or cyto lysis of cells expressing TWEAKR.
  • induction of cytolysis is achieved via antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • the TWEAKR agonist can induce between 10% to greater than 80% cytotoxicity of cells expressing TWEAKR.
  • administration of the TWEAKR agonists induces at least 10%, 20%, 30%, 40%, 50%, 60%, or 80% or more cytotoxicity of cells expressing
  • TWEAKR administration of the TWEAKR agonist can reduce the size of a solid tumor by targeting TWEAKR on the cancer cell's surface with one or more of the antibodies described herein.
  • the tumor can be a primary tumor or a secondary tumor.
  • administration of the TWEAKR agonists can reduce the size of a solid tumor by at least 10%, 20%, 30%, 40%, 50%, 60%, or 80% or more.
  • administration of the TWEAKR agonists can completely inhibit or prevent the growth of a solid tumor. It will be appreciated that a range of different cytotoxicities and a range of reductions in tumor size are described for the TWEAKR agonists disclosed herein. The skilled person will appreciate that the TWEAKR agonists can have any one of the described cytotoxicities and any one of the described reductions in tumor size.
  • responder in the context of the present disclosure includes persons where the cancer/tumor(s) is eradicated, reduced or improved, or stabilized such that the disease is not progressing after treatment.
  • the period of stabilization is such that the quality of life and/or patients' life expectancy is increased (for example stable disease for more than 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more months) in comparison to a patient that does not receive treatment.
  • a non-responder includes persons for whom the cancer/tumor(s) does not show reduction or improvement after treatment.
  • the characterization of the patient as a responder or non-responder can be performed by reference to a standard or a training set.
  • the standard may be the profile of a person/patient who is known to be a responder or nonresponder or alternatively may be a numerical value.
  • Such pre-determined standards may be provided in any suitable form, such as a printed list or diagram, computer software program, or other media.
  • Example 1 TWEAKR agonist PDL192 inhibits growth of a wide range of cancers in vitro
  • This example shows that a monoclonal antibody that acts as a TWEAKR agonist, PDL192 (used interchangeable herein with enavatuzumab), inhibits the growth of a wide range of cancer cell lines, including breast cancer, pancreatic cancer, cervical cancer, colon cancer, bladder cancer, lung cancer and melanoma cancer cell lines, in vitro.
  • PDL192 used interchangeable herein with enavatuzumab
  • PDL192 is a humanized monoclonal antibody to TWEAKR described in US Pat. App. No. 2009/0074762 and Culp et al, 2010, Clin. Cancer Res. 16(2):497-508.
  • Human IgGl antibody to cytomegalovirus (MSL109) was used as a negative control.
  • Antibodies were used at 10 ⁇ g ml for in vitro studies, unless otherwise stated, and crosslinked with F(ab') 2 goat anti-human IgG (Fe y specific) from Jackson ImmunoResearch at 3.5 ⁇ g/ml.
  • [0104] Growth inhibition assay 102 cancer cell lines were cultured at 500 cells per well in triplicate with enavatuzumab or IgGl control in the presence of F(ab') 2 goat anti-human IgG, (Fey specific) for 5 days in 96 well plates. Relative cell viability was determined using CellTiter-BlueTM (Promega). Fluorescence emitted at 590 nm was used to calculate the growth effect relative to the IgGl control antibody treatment. Each cell line was tested twice with the average growth inhibition reported. Cell lines with at least about 20% growth inhibition were considered TWEAKR agonist sensitive.
  • Cancer Sensitive Resistant type at least 20% growth ( ⁇ 20% growth inhibition) inhibition
  • PDL192 displayed the ability to block proliferation in some but not other cell lines from each of the following tumor types: breast cancer, lung cancer, colon cancer, renal cancer, ovarian cancer, melanoma, and pancreatic cancer. PDL192 also blocked cell proliferation in cell lines derived from bladder cancer, cervical cancer, salivary cancer and oral cancer tumors.
  • Example 2 In vitro correlation of PIK3CA activating mutations with responsiveness to PDL192
  • This example shows a correlation between mutations activating the PI3K pathway and non-responsiveness to PDL192 treatment in vivo. Somatic mutation analysis was performed on cells from 26 tumors to determine if a PIK3CA activating mutation or a PTEN null mutation was present. The results are shown in FIG. 3A. Of the tumors that contained either a PIK3CA activating mutation or a PTEN null mutation, three of sixteen were responsive to PDL192 while thirteen were non-responsive. Response was defined as statistically significant decrease in tumor volumes in the PDL192-treated animals compared to control-treated tumors. In contrast, for tumors whose cells bore wild type PIK3CA and wild type PTEN, eleven of fourteen were responsive to PDL192 and three were non- responsive.
  • PDL192M had no anti-tumor activity, suggesting that ADCC is the predominant mechanism of action for PDL192 in these models.
  • PDL192M had activity equivalent to PDL192; thus, direct growth inhibitory activity is presumed to be the predominant mechanism of action for PDL192 in these lines.
  • FIG. 3B none of the 3 cell lines responding to PDL192 via direct growth inhibition in vivo harbored activating mutations in PIK3CA or PTEN null mutations; in contrast, 13 of 16 nonresponding cell lines carried such PIK3CA or PTEN mutations.
  • Wild type A375 cells were transfected with vector alone, wild type PIK3CA,
  • PIK3CA E545K mutant or PIK3CA H1047R mutant.
  • E545K and H1047R have been shown to be PIK3CA activating mutations.
  • the cells were treated with PDL 192 or control MSL109 compounds, and response was analyzed using the growth inhibition assay of Example 1.
  • FIG. 5 cells transfected with vector alone or with wild type PIK3CA showed a dose-dependent response to PDL 192.
  • cells that received vectors bearing PIK3CA activating mutations showed a decreased response to PDL 192.
  • This study showed that activation of PIK3CA results in decreased sensitivity to PDL 192 treatment.
  • Example 6 PIK3CA depletion studies
  • Two cell lines bearing PIK3CA activating mutations were subjected to siRNA knockdown of PIK3CA or control, and subsequently tested for PDL192 responsiveness.
  • the DLD-1 cell line bears an E545K PIK3CA activating mutation
  • the HCT116 cell line bears an H1047R PIK3CA activating mutation.
  • Each of the two cell lines was treated with 10 nM control siRNA or 10 nM PIK3CA siRNA (for DLD-1) or 2.5 nM PIK3CA siRNA (for HCT116).
  • FIG. 6 cell lines receiving control siRNA did not respond to PDL192 (compared to control compound, MSL109) while cell lines that had been subjected to PIK3CA siRNA depletion showed a dose-dependent response to PDL192.
  • Table 2 below provides sequences of SEQ ID Nos:l-60.

Abstract

The present disclosure is directed to methods of treating subjects having a TWEAKR-positive cancer comprising administering to a subject who has tested negative for a PIK3CA activating mutation or a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.

Description

BIOMARKERS FOR PREDICTING RESPONSE TO
TWEAK RECEPTOR (TWEAKR) AGONIST THERAPY
1. CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit under 35 U.S.C. § 119(e) of provisional application no. 61/651,252, filed May 24, 2012, the contents of which are incorporated herein in their entireties by reference thereto.
2. BACKGROUND
[0002] Tumor Necrosis Factor-Related Weak inducer of Apoptosis, or TWEAK, is a member of the Tumor Necrosis Factor (TNF) family of cytokines that can cause cell death (in some instances via apoptosis). TWEAK has a number of biological activities, including inducing angiogenesis, stimulating the release of cytokines or chemokines, stimulating proliferation of human smooth muscle and endothelial cell lines, inducing TNF-a-mediated cell death, cathepsin B-dependent necrosis, and, as its name implies, apoptosis. See Han et al, 2003, Bioch. Biophys. Res. Communications 305:789-796; Nakayama et al, 2003, J. Immunology 170:341-348. TWEAK binds to and activates a member of the TNF receptor family, TWEAKR (also known as Fibroblast Growth Factor-Inducible 14 or Fnl4). See id.
[0003] Studies of TWEAKR in cancer report both cancer-promoting effects and anti-cancer effects. For example, Tran et al, 2006, Cancer Res. 66(19):9535-9542, reported that increased levels of TWEAKR promote glioma cell invasion and correlate with poor patient outcomes, and Dai et al, 2009, Cancer Lett. 283(2):159-167, reported that signaling via TWEAKR promoted cell migration and invasion in an ovarian cancer cell line. Conversely, Culp et al, 2010, Clin. Cancer Res. 16(2):497-508, reported that anti-TWEAKR agonistic antibodies inhibited tumor growth in in vitro experiments with cancer cell lines and in vivo mouse xenograft experiments. Similarly, Nakayama et al, 2003, J. Immunology 170:341- 348, reported that TWEAKR agonists induced cell death via apoptosis or necrosis in a variety of cancer cell lines. Collectively, these data demonstrate that activation of TWEAKR in cancers that overexpress TWEAKR promotes positive anti-cancer responses in some, but not all, TWEAKR positive cancers. As a consequence, not all patients diagnosed with a TWEAKR positive cancer will respond positively to therapies that target and activate TWEAKR, such as TWEAKR agonists. It would be beneficial to have available tools to identify which TWEAKR positive cancers could benefit from TWEAKR agonist therapies.
[0004] Phosphoinositide 3-kinases (PBKs) are lipid kinases that function as signal transducers downstream of cell surface receptors and mediate pathways important for cell growth, proliferation, adhesion, survival and motility. Mutations in PI3K genes result in deregulation of one or more of these biological processes and have been associated with cancer pathogenesis (Samuels et al, Science 304:554; Bachman et al, Cancer Biol. Ther. 3:772; Campbell, et al, Cancer Res. 64:7678; Broderick et al, Cancer Res 64:5048; Lee et al, Oncogene 24:1477; Levin et al, Clin. Cancer Res. 11:2875; Li et al, BMC Cancer 5:29; Saal et al, Cancer Res. 65:2554; Wang et al, Hum. Mutat. 25:322; Hartmann et al, Acta Neuropathol. 109:639). PDKs are classified based on primary structure, regulation, and in vitro lipid substrate specificity. Studies of proliferation and tumorigenesis have focused on Class I A POKs, which are heterodimeric molecules composed of a catalytic subunit, referred to as pi 10a, and an associated regulatory subunit, referred to as p85. Class IA PDKs are responsible for the production of phosphatidylinositol 3,4,5-triphosphate (PIP3) from phosphatidylinositol 4,5-bisphopshate (PIP2), while phosphatases, in particular the PTEN phosphatase, carry out the reverse reaction, dephosphorylating PIP3. Both pi 10a and PTEN have been linked to cancer: the gene encoding pi 10a, called PIK3CA, has been identified as an oncogene (Samuels, et al, Science 304:443) and PTEN has been classified as a tumor suppressor (Deane and Fruman, Annu. Rev. Immunol. 22:563; Okkenhaug et al, Sci. STKE 65:PE1).
3. SUMMARY
[0005] As discussed in the Background Section, compounds that have TWEAKR agonist activity inhibit proliferation of some, but not all, tumor cells that express TWEAKR. The ability of TWEAKR agonists to inhibit proliferation of TWEAKR positive tumors does not correlate with cancer type. As illustrated in Table 1 and FIG. 1, in in vitro experiments carried out in tumor cell lines derived from a variety of different types of cancers, an anti- TWEAKR antibody having TWEAKR agonist activity, enavatuzumab (also known as, and referred to herein as, PDL192) inhibited the proliferation and/or survival of some, but not all, TWEAKR positive cell lines derived from the same type of cancer. These data indicate that not all tumors that express TWEAKR may respond to treatment with compounds having TWEAKR agonist activity.
[0006] Applicants have discovered that mutations in PIK3CA, which encodes pi 10a, a PI3K lipid kinase, and in PTEN, a phosphatase, are predictors of anti-tumorigenic efficacy of TWEAKR agonists. Specifically, Applicants have discovered that mutations that result in increased activity of pi 10a, referred to herein as PIK3CA activating mutations, and mutations that result in reduced activity of PTEN, referred to herein as PTEN null mutations, are predictive of a lack of response to treatment with a TWEAKR agonist.
[0007] The discovery that two enzymes that modulate phosphorylation of inositol polyphosphates are predictive for TWEAKR agonist response provides a new tool in selecting subjects to treat with TWEAKR agonist therapy. Accordingly, in one aspect, the present disclosure provides methods of treating a subject having a TWEAKR-positive cancer comprising administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit. The TWEAKR agonist can be an anti-TWEAKR agonist antibody, including monoclonal and humanized antibodies.
[0008] In certain embodiments, the methods further comprise the step of testing the subject for one or more PIK3CA activating mutations. Exemplary methods for detecting a PIK3CA activating mutation are described in Section 5.3, below. Preferably, the testing of the subject is conducted prior to administration of the TWEAKR agonist, and can be conducted simultaneously with testing the subject for TWEAKR expression. Accordingly, the present disclosure provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by testing a subject for one or more PIK3CA activating mutations, preferably concurrently with or after testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit. The present disclosure further provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by (i) testing a subject for one or more PIK3CA activating mutations and (ii) prior to or concurrently with testing the subject for one or more PIK3CA activating mutations, testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
[0009] In another aspect, the present disclosure provides methods of treating a subject having a TWEAKR-positive cancer comprising administering to a subject who has tested negative for a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit. The TWEAKR agonist can be an anti-TWEAKR agonist antibody, including monoclonal and humanized antibodies.
[0010] In certain embodiments, the methods further comprise the step of testing the subject for a PTEN null mutation. Exemplary methods for detecting a PTEN null mutation are described in Section 5.3, below. Preferably, the testing of the subject is conducted prior to administration of the TWEAKR agonist, and can be conducted simultaneously with testing the subject for TWEAKR expression. Accordingly, the present disclosure provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by testing a subject for a PTEN null mutation, preferably concurrently with or after testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR- positive cancer who has tested negative for a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit. The present disclosure further provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by (i) testing a subject for a PTEN null mutation and (ii) prior to or concurrently with testing the subject for a PTEN null mutation, testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
[0011] In yet other aspects, the present disclosure provides methods of treating a subject having a TWEAKR-positive cancer comprising administering to a subject who has tested negative for a PTEN null mutation and for one or more PIK3CA activating mutations an amount of a TWEAKR agonist effective to promote a therapeutic benefit. Optionally, the methods of the disclosure further comprise the step of testing the subject for a PTEN null mutation and the one or more PIK3CA activating mutations, as described herein. Preferably, the testing of the subject is conducted prior to administration of the TWEAKR agonist, and can be conducted simultaneously with testing the subject for TWEAKR expression.
Accordingly, the present disclosure provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by testing a subject for a PTEN null mutation and one or more PIK3CA activating mutations, preferably concurrently with or after testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PTEN null mutation and one or more PIK3CA activating mutations an amount of a TWEAKR agonist effective to promote a therapeutic benefit. The present disclosure further provides methods comprising (a) identifying a subject suitable for therapy with a TWEAKR agonist by (i) testing a subject for a PTEN null mutation and one or more PIK3CA activating mutations and (ii) prior to or concurrently with testing the subject for a PTEN null mutation and one or more PIK3CA activating mutations, testing the subject for TWEAKR expression, and (b) administering to a subject suffering from a TWEAKR-positive cancer who has tested negative for a PTEN null mutation and a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
[0012] The PIK3CA activating mutation can be any type of mutation that leads to increased kinase activity relative to wild type human PIK3CA, including a substitution mutation, deletion mutation, or insertion mutation. In certain embodiments, the PIK3CA activating mutation is a substitution mutation, e.g., a mutation selected from the group consisting of: K111N, E542K, E545K, E545G, E545D, Q546K, Q546R, Ml 0431, M1043V, H1047R, H1047L, H1047Y, G1049R and G1049S. Further PIK3CA activating mutations are provided in Section 5.1, below. The PIK3CA activating mutation can be detected at the nucleic acid level (e.g., DNA or mRNA) or at the protein level. In some embodiments, the subject is tested for one, two, three, four or more (or all) of Kl 1 IN, E542K, E545K, E545G, E545D, Q546K, Q546R, M1043I, M1043V, H1047R, H1047L, H1047Y, G1049R and G1049S, optionally in addition to further PIK3CA activating mutations, such as those provided in Section 5.1.
[0013] The PTEN null mutation can be any type of mutation that leads to decreased phosphatase activity compared to wild type human PTEN, including a substitution mutation, deletion mutation, or insertion mutation. The PTEN null mutation can be detected at the nucleic acid level (e.g., DNA or mRNA) or at the protein level. Further PTEN null mutations are provided in Section 5.2, below.
[0014] Methods of testing for T WEAKR expression are described in Section 5.5.
[0015] TWEAKR agonists, also referred to herein as TWEAKR agonist compounds, useful in the methods of the present disclosure are described in Section 5.4.
4. BRIEF DESCRIPTION OF THE FIGURES
[0016] FIG. 1 provides a bar chart illustrating the percent growth inhibition for 38
TWEAKR positive cancer cell lines treated with PDL192 (enavatuzumab) for 5 days;
[0017] FIG. 2 provides results of 91 cell lines treated with TWEAKR agonist PDL192 classified based on growth inhibition and presence (mutation) or absence (wild type) of a PIK3CA activating mutation. Cell lines that exhibited growth inhibition (responders) or lack of growth inhibition (nonresponders) when treated with PDL192 are indicated on the left.
[0018] FIGS. 3A-B provide the results of in vivo experiments in which mice bearing xenograft tumors derived from cell lines were treated with TWEAKR agonist PDL192. The cell lines were classified based on growth inhibition response (responders or non-responders) and PIK3CA and PTEN genotype (mutation or wild type) (FIG. 3A). A subset of cell lines were classified as being responsive via a non-ADCC mechanism or nonresponders and PIK3CA and PTEN genotype (mutation or wild type) (FIG. 3B).
[0019] FIG. 4A illustrates phospho-Akt (S473) levels in cell lines treated with PDL192 in vitro. Cell lines that were treated with PDL192 are indicated on the right hand side. [0020] FIG. 5 provides the relative cell viability of cells transfected with control vector, wild type PIK3CA, or PIK3CA containing an activating mutation, as indicated, that were treated with TWEAKR agonist (PDL192, square data points) or control compound (MSL109, round data points).
[0021] FIG. 6 provides the results of treatment with TWEAKR agonist (PDL192, black bars) or control compound (MSL109, white bars) for DLD-1 and HCTl 16 cells. DLD-1 cells have an E545K mutation in PIK3CA, and HCTl 16 cells have an H1047R mutation in PIK3CA. The cell lines were subjected to siRNA depletion using control or PIK3CA siR A.
Response to treatment with varying amounts of PDL192 is displayed.
5. DETAILED DESCRIPTION
[0022] TWEAKR (also known as Fnl4 or TNFRSF12A) is a tumor necrosis factor (TNF) superfamily member known to be expressed on the surface of cancer cells derived from a variety of solid tumors. Compounds that have TWEAKR agonist activity inhibit
proliferation of some, but not all, tumor cells that express TWEAKR. TWEAKR agonists are potentially useful in the treatment of some TWEAKR positive cancers, particularly if it is possible to determine whether the TWEAKR positive cancer is likely to be a responder or non-responder to TWEAKR agonist therapy.
[0023] The present disclosure is based in part on applicants' discovery that PIK3CA activating mutations and PTEN null mutations are indicative of non-responsiveness to TWEAKR agonist therapy in treating a TWEAKR positive cancer. As shown in the
Examples below, applicants have discovered that the presence of a PIK3CA activating mutation or a PTEN null mutation in TWEAKR-positive cancer cell lines correlates with a lack of response to a TWEAKR agonist. The disclosure makes use of these findings to provide methods of treating subjects having TWEAKR positive cancers. Generally, the methods of treating a subject having a TWEAKR positive cancer comprise administering to a subject who has tested negative for a PIK3CA activating mutation or PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit. [0024] Furthermore, methods of the disclosure may be employed to classify subjects as responders or non-responders to a TWEAKR agonist therapy. In some embodiments, the methods may be also used to predict whether or not a subject suffering from a TWEAKR positive cancer will respond to a TWEAKR agonist therapy.
5.1. PIK3CA Activating Mutations
[0025] PIK3CA encodes the catalytic subunit of a Class 1 A PI3 kinase, a class of enzymes that catalyzes the phosphorylation of inositol-containing lipids. PI3 kinases (PI3Ks) are known to be important regulators of signaling pathways. As used herein, "PIK3CA sequence" refers to a polynucleotide encoding the pi 10a kinase subunit. A PIK3CA sequence can be a gene sequence (with or without introns), a cDNA, or an mRNA. A representative example of a wild type human PIK3CA sequence is SEQ ID NO:55. The corresponding polypeptide sequence for a human pi 10a is SEQ ID NO: 56.
[0026] Mutations in PIK3CA that correlate with proliferative disorders such as cancer are known to cluster in exon 9 (SEQ ID NO:57) and exon 20 (SEQ ID NO:58). See U.S. Patent Publication Nos. 2009/0208505, paragraph [0017] and Table 2 herein. Exon 9 corresponds to the helical domain of PIK3CA, while exon 20 corresponds to its kinase domain. PIK3CA activating mutations cluster in these regions, known as mutational hotspots. Additional mutations found in human cancers are described in Table 3 of U.S. Patent Publication No. 2009/0208505.
[0027] Certain PIK3CA mutations are PIK3CA activating mutations, a number of which have been described, including those described in Bader, et al. Nat. Rev. Cancer (2005) 5:921-929, Kang et al, PNAS (2005), 102:802, Samuels et al, Science (2004) 304:554, Zhang et al. , Breast Cancer Res. Treat. (2008) 112:217-227, and U.S. Patent Publication No. 2011/0060605. As used herein, a "PIK3CA activating mutation" can be any type of mutation, including a substitution mutation, a deletion mutation, or an insertion mutation, which results in increased kinase activity compared to wild type PIK3CA. Increased kinase activity can be the result of increased enzymatic activity or increased levels of the pi 10a polypeptide. [0028] Numerous assays for kinase activity are known, which can be used to determine whether a PIK3CA sequence results in a pi 10a with increased kinase activity relative to wild type pi 10a. For certain applications, pi 10a kinase activity can be determined using an in vitro kinase assay. PI3 kinase assays have been described (see Kang et ah, 2005, PNAS 102:802-807). For example, complexes of pi 10a and an anti- pi 10a antibody are prepared by incubating 400 μg of pi 10a with 5 μΐ of anti-pl 10a antibody for 2 h at 4°C, followed by 2 h of incubation with Protein A-agarose. Protein A-agarose beads are washed three times with the lysis buffer and once with TNE (10 mM Tris, Ph 7.5/100 mM NaCl/1 mM EDTA). Subsequently, the complexes are incubated with 50 μΐ of kinase reaction buffer containing 20 mM Hepes (pH 7.5), 10 mM MgC12, 200 μg/ml phosphatidylinositol (sonicated), 60 μΜ ATP, and 200 μα/ιηΐ (1 Ci = 37 GBq) [γ-32 P]ATP for 5 min at room temperature. The reaction is terminated by adding 80 μΐ of 1 N HC1, and the phosphorylated lipids are extracted with 160 μΐ of chloroform/methanol (1 : 1). After samples are dried down, they are dissolved in chloroform and spotted onto Silica Gel 60 TLC plates (Merck). The plates are developed in a borate buffer system and visualized by autoradiography. Using this assay, an increased kinase activity refers to a greater detection of radioactivity (indicating more incorporation of radiolabeled phosphate) per μg of pi 10a test protein, compared to incorporation per μg of pi 10a wild type protein, e.g., a protein having the sequence of SEQ ID NO-.56.
[0029] Mutations in the coding region of PIK3CA can be synonymous or non-synonymous mutations. Mutations in the noncoding or coding regions of a PIK3CA sequence can lead to altered RNA transcripts and/or altered protein as a result of differential post-transcriptional processing {e.g., mRNA splicing, nonsense-mediated decay, post-translational modification, etc.) as compared to wild type PIK3CA mRNA or pi 10a.
[0030] For purposes of determining whether a PIK3CA sequence bears a mutation, a PIK3CA sequence from a test sample, e.g., a. sample containing cancer cells, can be compared to a PIK3CA sequence from a different tissue in the same individual. A difference in the sequences from the two tissues indicates a mutation. Alternatively, the PIK3CA sequence from a test sample can be compared to SEQ ID NO:55, with one or more differences between these two sequences indicative of a mutation.
[0031] Specific PIK3CA activating mutations can be identified by reference to a wild type polypeptide or polynucleotide sequence where the number indicates the position within the polypeptide or polynucleotide sequence, the first letter indicates the identity of the residue or nucleotide in the wild type sequence and the second letter indicates the identity of the residue or nucleotide in the mutant sequence. In certain embodiments, PIK3CA activating mutations are identified by reference to the wild type polypeptide sequence of SEQ ID NO: 56. For example, "E545K" refers to a polypeptide sequence for which the glutamate residue at position 545 has been mutated to lysine. In some embodiments, a PIK3CA activating mutation is selected from the group consisting of: Kl 1 IN, E542K, E545K, E545G, E545D, Q546K, Q546R, Ml 0431, Ml 043V, H1047R, H1047L, H1047Y, G1049R and G1049S. Subjects can be tested for one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, eleven or more, twelve or more, thirteen or more, or all fourteen of these mutations.
[0032] A PIK3CA activating mutation can also be identified by reference to a wild type polynucleotide sequence where the number indicates the position within the polynucleotide sequence, the first letter indicates the identity of the wild-type nucleotide, and the second letter indicates the identity of the mutant nucleotide. Typically, the ">" character is placed between the first and second letter to denote that the change occurs from the nucleotide represented by the first letter to that of the second. Furthermore, according to established sequence description conventions, the underscore character, indicates a range of nucleotides, "ins" indicates an insertion, and "del" indicates a deletion. The number following "ins" or "del" indicates the number of nucleotides that are inserted or deleted, respectively. A first number followed by a plus sign ("+") or minus sign ("-"), followed by a second number indicates the presence of a mutation within an intron that is a number of positions downstream (for plus sign) or upstream (for minus sign) given by the second number with respect to the position in the coding sequence given by the first number. [0033] In some embodiments, a PIK3CA activating mutation is a mutation identified as PIK3CA (2188-7delT), with reference to SEQ ID NO:55, where the designation indicates that subjects bearing this mutation possess a PIK3CA gene in which the T nucleotide that occurs 7 positions upstream from position 2188 in the coding sequence is deleted.
[0034] In some embodiments, a PIK3CA activating mutation is a mutation that gives rise to a higher expression level of pi 10a compared to wild type levels. Methods for detection and quantitation of pi 10a expression levels are known and have been described, for example, in U.S. Application Publication No. 2009/0192110.
5.2. PTEN Null Mutations
[0035] Phosphatase and tensin homolog (PTEN) is encoded by the PTEN gene, and, like PIK3CA, is known to be mutated in the development of certain cancers. PTEN has been identified as the phosphatase that catalyzes the reverse reaction of the reaction catalyzed by the PIK3CA-encoded protein (pi 10a), specifically the dephosphorylation of inositol polyphosphates. As used herein, a "PTEN polynucleotide sequence" refers to a
polynucleotide encoding the PTEN protein. A PTEN polynucleotide sequence can be a gene sequence (with or without introns), a cDNA, or an mRNA. A polynucleotide sequence of a representative wild type human PTEN gene is SEQ ID NO:59. The corresponding wild type PTEN polypeptide sequence is SEQ ID NO:60.
[0036] As used herein, the term "PTEN null mutation" refers to any mutation or set of mutations within PTEN that leads to a decreased phosphatase activity compared to wild type PTEN, as a result of decreased activity or levels of the PTEN protein. A variety of assays for determining phosphatase activity are known in the art. For certain applications, PTEN phosphatase activity can be determined using an in vitro phosphatase assay. Such assays are known and described, for example, in U.S. Application Publication No. 2011/0150868, U.S. Patent No. US7745485, and Georgescu et al, PNAS (1999), 96(18): 10182-10187. For example, PTEN phosphatase activity can be measured with each assay performed in 50 μΐ buffer containing 100 mM Tris-HCl (pH 8.0), 10 mM DTT, and 100 μΜ water-soluble diC8-PIP3 (Echelon, Salt Lake City, Utah). Reactions containing PTEN can be
immunoprecipitated from cell lysates on protein G-agarose beads using commercially- available anti-PTEN antibodies. Prior to immunoprecipitation with PTEN antibody, lysates can be precleared with protein G-agarose beads. After immunoprecipitation, the beads can be washed once in lysis buffer; five times in low stringency buffer containing 20 mM Hepes (pH 7.7), 50 mM NaCl, 0.1 mM EDTA, and 2.5 mM MgC12; and once in phosphatase assay buffer lacking PIP3. Reactions can be incubated for 40 min at 37° C and transferred to a 96- well plate. Release of phosphate from substrate may be measured using Biomol Green Reagent (Biomol Research Laboratories, Inc., Plymouth Meeting, Pa.).
[0037] Alternatively, PTEN phosphatase activity can be measured without
immunoprecipitation. An exemplary reaction mixture for this assay includes 100 mM TrisHCl (pH 8), 10 mM dithiothreitol, 0.5 mM diC16 phosphatidylserine (PS), 25 μΜ PIP3- diC16, BIOMOL PH-107 (Biomol Research Laboratories, Inc., Plymouth Meeting, Pa.) and 50 μg/ml purified recombinant wild type or mutant PTEN. Lipids can be prepared in organic solvents and dispensed into 1.5 ml microfuge tubes followed by solvent removal under reduced pressure. Buffer can then be added and a lipid suspension formed by sonication. PTEN phosphatase assays are initiated by the addition of PTEN and carried out at 37° C. At different time points, 15 μΐ of 100 μΜ NEM (N-ethylmaleimide) is added to 10 μΐ of reaction mixture followed by rapid centrifugation at 18,000xg for 15 minutes at 4° C. Liberated inorganic phosphate is detected in twenty microliters of supernatant using the Malachite green assay with reference to an inorganic phosphate standard curve. Malachite green reaction with inorganic phosphate is detected spectrophotometrically at 620 nm wavelength.
[0038] In other preferred embodiments, a PTEN null mutation gives rise to a lowered expression level of PTEN compared to wild type levels. Methods for detection and quantitation of PTEN expression levels are known and have been described, for example, in U.S. Application Publication No. 2010/0303809.
[0039] A PTEN null mutation can be any type of mutation, including substitution mutations, deletion mutations, and insertion mutations. Mutations in the coding region of PTEN can be synonymous or non-synonymous mutations. Mutations in the noncoding or coding regions of PTEN can lead to mutant RNA transcripts and/or mutant protein as a result of differential post-transcriptional processing {e.g., mRNA splicing, nonsense-mediated decay, post- translational modification, etc.) as compared to wild type PTEN. PTEN null mutations are known in the art, see Jhawer et al, Cancer Res (2008), 68:1953-1961, incorporated herein by reference in its entirety.
[0040] Specific PTEN null mutations can be identified by reference to a wild type polypeptide sequence where the number indicates the position within the polypeptide sequence, the first letter indicates the identity of the residue in the wild type sequence and the second letter indicates the identity of the residue in the mutant sequence. In certain embodiments, PTEN null mutations are identified by reference to the wild type polypeptide sequence of SEQ ID NO: 60. For example, "LI 12Q" refers to a polypeptide sequence for which the leucine residue at position 112 has been mutated to glutamine. In some embodiments, a PTEN null mutation is a mutation in which an amino acid is mutated to a stop codon, identified by reference to a wild type polypeptide sequence in which the number indicates the position within the sequence, the first letter indicates the identity of the wild type amino acid, and "*" indicates substitution of a stop codon at this position. For example, "Q149*" indicates that the glutamine at position 149 is mutated to a stop codon, resulting in a truncated polypeptide.
[0041] In some embodiments, a PTEN null mutation is a frameshift mutation that introduces a stop codon, resulting in a truncated sequence. Such mutations can be identified by reference to a wild type polypeptide where the number indicates the position where the frameshift occurs, "fs" indicates a frameshift mutation, "*" indicates introduction of a stop codon, and the final number indicates the position of the stop from the first amino acid affected by the frameshift.
[0042] In some embodiments, a PTEN null mutation is selected, with reference to SEQ ID NO:60, from the group consisting of: Q149*, LI 12Q, F90fs*9, K267fs*9, V175fs*3, and P248fs*5. Subjects can be tested for one or more, two or more, three or more, four or more, five or more, or all six of these mutations.
[0043] A PTEN null mutation can also be identified by reference to a wild type
polynucleotide sequence where the number indicates the position within the polynucleotide sequence, the first letter indicates the identity of the wild-type nucleotide, and the second letter indicates the identity of the mutant nucleotide. Typically, the ">" character is placed between the first and second letter to denote that the change occurs from the nucleotide represented by the first letter to that of the second. Furthermore, according to established sequence description conventions, the underscore character, indicates a range of nucleotides, "ins" indicates an insertion, and "del" indicates a deletion. The number following "ins" or "del" indicates the number of nucleotides that are inserted or deleted, respectively. For example, "165_209del45" refers to a mutant in which the 45 nucleotides spanning from position 165 to position 209 are deleted. A first number followed by a plus sign ("+") or minus sign ("-"), followed by a second number indicates the presence of a mutation within an intron that is a number of positions downstream (for plus sign) or upstream (for minus sign) given by the second number with respect to the position in the coding sequence given by the first number.
[0044] In some embodiments, a PTEN null mutation is selected, with reference to SEQ ID NO:59, from the group consisting of: l_164dell64, 165_209del45, 253+lOT, 270delT, 335T>A, 4450T, 524_558del35, 741_742insA, 800delA, and 1026+1 G>T. Subjects can be tested for one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or all ten of these mutations.
[0045] Methods of detecting PTEN null mutations are known in the art. For example, a PTEN polynucleotide sequence from a test sample, e.g., a sample containing cancer cells, can be compared to a PTEN polynucleotide sequence in a different tissue from the same individual. A difference in the sequences from the two tissues indicates a mutation.
Alternatively, the PTEN polynucleotide sequence determined in a PTEN gene in a test sample can be compared to SEQ ID NO:59. A difference between the test sample sequence and SEQ ID NO:59 can be identified as a mutation. Methods described herein for the detection of PIK3CA activating mutations also can be applied using the appropriate primers and probes to detect PTEN null mutations, and vice versa. The Sanger Wellcome database contains information about PTEN deletions and truncations, and therefore may also be useful in determining a PTEN null mutation (see Wang et al, PNAS (2009) 106(15):6279-6284). 5.3. Detecting PIK3CA Activating and PTEN Null Mutations
[0046] The presence or absence of a PIK3CA activating mutation or a PTEN null mutation can be determined in a subject using a variety of methods known in the art. In some embodiments, a subject is tested for a PIK3CA activating mutation and/or a PTEN null mutation. The testing can be performed in conjunction with T WEAKR agonist therapy in order to treat a T WEAKR-positive cancer. The testing is preferably performed prior to treatment with the TWEAKR agonist, and can be carried out at the same time the subject's cancer is tested for TWEAKR expression, for example as described in Section 5.5.
[0047] The determination of a PIK3CA activating mutation or a PTEN null mutation can be made at the nucleic acid level and/or at the protein level. The nucleic acid and/or proteins used for the determination can be obtained from biological samples, including, but not limited to tissues, cells, biological fluids and isolates thereof, including those isolated from a subject, as well as those present within a subject. Preferably, biological samples comprise cells, most preferably tumor cells, that are isolated from body samples, such as, but not limited to, smears, sputum, biopsies, secretions, cerebrospinal fluid, bile, blood, lymph fluid, urine and feces, or tissue which has been removed from organs, such as breast, lung, pancreas, intestine, skin, cervix, prostate, and stomach. Tissues suspected of being cancerous can be tested for a PIK3CA activating mutation or a PTEN null mutation, as can body samples that may be expected to contain sloughed-off cells from tumors or cells of cancers.
[0048] An allele containing a PIK3CA activating mutation is referred to as a PIK3CA mutant allele, and the corresponding wild type region of DNA is referred to as a PIK3CA wild type allele. An allele containing a PTEN null mutation is referred to as a PTEN mutant allele, and the corresponding wild type region of DNA is referred to as a PTEN wild type allele.
Methods for detecting mutations in DNA are known, and include genotyping, microarrays, direct sequencing, restriction mapping, Restriction Fragment Length Polymorphism,
Southern Blots, SSCP, dHPLC, single nucleotide primer extension, allele-specific
hybridization, allele-specific primer extension, oligonucleotide ligation assay, and invasive signal amplification, Matrix-assisted laser desorption/ionization time-of-flight (MALDI- TOF) mass spectrometry, and fluorescence polarization (FP). In one method of genotyping, primers flanking a PIK3CA or PTEN allele are selected and used to amplify that region. The amplified region is then sequenced using DNA sequencing techniques known in the art and analyzed for the presence of a PIK3CA activating or PTEN null mutation.
[0049] Commercial methods are available for detecting a mutation in a DNA sample, and include the SNaPshot® Assay (Applied Biosystems), TaqMan® SNP Genotyping Assay (Applied Biosystems), and GoldenGate® Genotyping Assay (Illumina). For a SNaPshot® Assay to detect PIK3CA activating mutations, see Hurst et al, BMC Research Notes, 2009, 2:66. Probes used to detect PIK3CA activating mutations are listed in Table 1 of Hurst et al, supra. One of ordinary skill in the art would be able to adapt methods used to detect PIK3CA activating mutations to detect PTEN null mutations.
[0050] A PIK3CA activating or PTEN null mutation in a nucleic acid can be detected using a probe. For example, an amplified region comprising a wild type or mutant allele is hybridized using a composition comprising a probe specific for the mutation under stringent hybridization conditions. For example, isolated nucleic acids that bind to mutant alleles at high stringency may be used as probes to determine the presence of the mutation. Nucleic acids may be labeled with a detectable marker. The marker or label is typically capable of producing, either directly or indirectly, a detectable signal. For example, the label may be radioopaque or a radioisotope, such as H, ' , "P, "S, "Ί, a fluorescent
(fluorophore) or chemiluminescent (chromophore) compound, such as fluorescein
isothiocyanate, rhodamine or luciferin; an enzyme, such as alkaline phosphatase, beta- galactosidase or horseradish peroxidase; an imaging agent; or a metal ion.
[0051] A PIK3CA activating or PTEN null mutation can also be detected using restriction enzymes. For example, amplified products can be digested with a restriction enzyme that specifically recognizes sequence comprising a mutant allele, but does not recognize the corresponding wild type allele or vice versa. PCR may be used to amplify DNA comprising a PIK3CA or PTEN allele, and amplified PCR products are subjected to restriction enzyme digestion under suitable conditions and restriction products are assessed. If, for example, a PIK3CA or PTEN mutant allele corresponds to a sequence digested by the restriction enzyme, digestion is indicative of detecting that particular PIK3CA or PTEN mutant allele. Restriction products may be assayed electrophoretically as is common is the art.
[0052] PIK3CA and PTEN mutant alleles may also be detected by a variety of other methods known in the art. For example, PCR and RT-PCR and primers flanking a PIK3CA or PTEN allele can be employed to amplify sequences and transcripts, respectively, in a sample comprising DNA (for PCR) or RNA (for RT-PCR). The amplified products are optionally sequenced to determine whether the PIK3CA or PTEN allele in the sample is mutant or wild type.
[0053] Alternatively, a PIK3CA or PTEN allele in a sample can be detected by a variety of other techniques known in the art including microarrays, hybridization assays, PCR based assays, molecular beacons, dynamic allele-specific hybridization (DASH) and/or
combinations of these.
[0054] Mutant PIK3CA or PTEN polynucleotide sequences may also be identified by methods which detect particular mRNAs in cells. These include hybridization assays using complementary DNA probes (such as in situ hybridization using labeled mutant or wild type PIK3CA or PTEN riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for PIK3CA or PTEN polynucleotide sequences, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like). Protocols for the detection of specific mRNAs in a sample are well known in the art (Sambrook et al, (1990) Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory Press; Ausubel et al, (1998) Current Protocols in Molecular Biology, Wiley).
[0055] Mutant pi 10a or PTEN protein bearing a PIK3CA activating mutation or PTEN null mutation can be detected using a specific agent, most preferably an antibody, that is itself detectably labeled, or using an unlabeled antibody specific for mutant pi 10a or PTEN protein and a second antibody that is detectably labeled and recognizes the unlabeled antibody specific for mutant pi 10a or PTEN protein. Alternatively, any molecule that can be detectably labeled and that specifically binds to mutant pi 10a or PTEN protein can be used in the practice of the methods of the disclosure. In a preferred embodiment of the methods of the present disclosure, a two-component immunohistochemical staining system may be used to differentially stain mutant pi 10a or PTEN protein and the tissue or cell sample so that the stained mutant pi 10a or PTEN protein can be more readily distinguished from the counterstained tissue or cell sample.
[0056] pi 10a or PTEN protein can also be enriched by affinity purification using, for example, an anti-pl 10a or anti-PTEN antibody, accordingly, and the enriched protein can be sequenced by mass spectrometric methods known in the art to determine the presence of a PIK3CA activating or PTEN null mutation.
[0057] The presence of a PIK3CA activating mutation or a PTEN null mutation in a given cell line can also be determined by reference to the Sanger Wellcome database of somatic mutations related to human cancers (see Wang et al, PNAS (2009) 106(15):6279-6284).
[0058] As PIK3CA drives downstream signaling events, assessing the phosphorylation status of downstream pathway members can also be used as an indicator of PI3K pathway activation status as a result of upregulation of PIK3CA protein levels and/or the presence of activating mutations in PIK3CA. Such downstream pathway members phosphorylated when the PI3K pathway is activated include PDK1 (S241, Y373 Y376, or S396), Akt (S473 or T308), FoxOl (T24, S249, S256, or S319), Fox04 (SI 93 or S262), Fox03a (T32, S253, S318, S321, or S644). Reagents specific for phosphorylated forms of these proteins are readily available.
5.4. TWEAKR Agonists
[0059] As described herein, the methods of the disclosure are useful for treating a subject with a TWEAKR agonist. A TWEAKR agonist is an agent capable of activating a
TWEAKR signaling cascade leading to one or more of the biological activities associated with TWEAKR, such as killing TWEAKR positive cancer cells by apoptosis, inhibiting proliferation and/or survival of TWEAKR positive cancer cells, and inducing chemokine or cytokine release in TWEAKR positive cancer cells. [0060] In some embodiments, TWEAKR agonists induce apoptosis in cells expressing TWEAKR. Without being bound by theory, induction of apoptosis by TWEAKR agonists is postulated to occur through caspase activation. Therefore, induction of apoptosis by TWEAKR agonists can be assayed by measuring activation of caspase 3/7, using methods known in the art, including commercial kits such as Caspase-Glo® 3/7 Assay Systems from Promega®. Assays for apoptosis may also be performed by terminal deoxynucleotidyl transferase-mediated digoxigenin-11-dUTP nick end labeling (TUNEL) assay (Lazebnik et al. (1994), Nature: 371:346).
[0061] In some embodiments, a TWEAKR agonist inhibits proliferation {i.e. cell division) and/or survival of a cell, and in particular, a TWEAKR positive cancer cell. TWEAKR agonists, including TWEAK and anti-TWEAKR agonist antibodies, have been shown to inhibit proliferation and/or survival of TWEAKR positive cancer cell lines in in vitro and in vivo growth inhibition assays (Culp et al, 2010, Clin. Cancer Res. 16(2): 497-508).
Inhibition of proliferation and/or survival can be measured by an in vitro growth inhibition assay as described in Example 1 below. Generally, a cancer cell line known to be sensitive to TWEAKR agonists, such as BT549 or H358, is cultured at 500 cells per well in triplicate with 5-10 μg/ml of the proposed TWEAKR agonist for 5 days in 96 well plates. Growth inhibition is determined using CellTiter-Blue™ (Promega): cells are incubated with the reagent and fluorescence emitted at 590 nm is measured and used to calculate the effect on viable cells of the proposed TWEAKR agonist relative to a control, such as an IgGl control. A test compound is considered to be a TWEAKR agonist if it induces growth inhibition of at least about 20% relative to control treatment.
[0062] In a specific exemplary assay where the proposed TWEAKR agonist is an anti- TWEAKR antibody as described below, cells can be incubated with anti-TWEAKR antibody (10 μg/ml) and an antibody (5 μg/ml) or antibody fragment (3.5 μg/ml) that specifically binds to the anti-TWEAKR antibody. The antibody or antibody fragment is preferably species- and isotype-specific. For example, where the anti-TWEAKR antibody is a humanized IgG antibody, a F(ab')2 antibody fragment that is a goat anti-human
immunoglobulin (Fey specific) or a full goat anti-human-IgG antibody can be used. [0063] Agonist activity can be determined by measuring the release of cytokines and/or chemokines in an in vitro cell growth assay. For example, in a typical assay the cells are incubated in vitro with a TWEAKR agonist. Twenty four hours later, the cell supernatant is assessed for the presence of cytokines and/or chemokines using an ELISA assay or a commercial fluorescent bead-based multiplex assay (e.g., Luminex®, Upstate). In some embodiments, the chemokine that is measured for release is IL-8.
[0064] Using data generated from a cytokine/chemokine release assay, the percent agonist activity can be calculated using the formula: % agonist activity= (a-c)/(b-c) in which a= quantity of cytokine/chemokine released from cells treated with TWEAKR agonist at 10 μg/ml, b= quantity of cytokine/chemokine released from cells treated with TWEAK at 300 ng/ml, and c= quantity of cytokine/chemokine released from untreated cells.
[0065] TWEAKR agonists include the natural ligand of TWEAKR, TWEAK. TWEAK forms trimers and is thought to promote activation of TWEAKR via trimerization of the receptor. See Winkles, 2008, Nature Reviews Drug Discovery 7: 411-425. Therefore, it is expected that TWEAKR agonists can also be proteins that form multimers, e.g., trimers and are capable of binding to and promoting multimerization of TWEAKR. Accordingly, TWEAKR agonists include fusions of TWEAK, or a fragment of TWEAK capable of binding TWEAKR, to polypeptides comprising an oligomerization domain. Polypeptides comprising suitable oligomerization domains include CD8 (Nakayama et al., 2000, J. Exp. Med. 192:1373), collagen (Frank et al., 2001, J. Mol. Bio. 308:1081-1089), bacteriophage T4 fibritin (Yang et al., 2002, J. Virology 76(9):4634-4642), engineered leucine/isoleucine zipper polypeptides (Harbury, 1994, Nature 371(6492):80-83), macrophage scavenger receptor (U.S. Pat. No. 7,238,499), and members of the tumor necrosis factor (TNF) superfamily including TNF, TRAIL, LIGHT, FasL, CD40L, OX40L, RANKL, GITRL (see Gravestein et l, 1998, Immunology 10:423-434).
[0066] Methods of making fusion proteins are well known in the art, and involve combining coding sequences of two or more polypeptides to generate a coding sequence of a fusion protein. Techniques for the manipulation of nucleic acids, including techniques for the synthesis, isolation, cloning, detection, and identification are well known in the art and are well described in the scientific and patent literature. See, e.g., Sambrook et al, eds., Molecular Cloning: A Laboratory Manual (2nd Ed.), Vols. 1-3, Cold Spring Harbor
Laboratory (1989); Ausubel et al, eds., Current Protocols in Molecular Biology, John Wiley & Sons, Inc., New York (1997); Tijssen, ed., Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I. Theory and Nucleic Acid Preparation, Elsevier, N.Y. (1993).
[0067] TWEAKR agonists also include polypeptides of TWEAK engineered or modified to have an increased serum half-life. Suitable engineered TWEAK proteins include fusions of TWEAK, or a fragment of TWEAK capable of binding TWEAKR, to human
immunoglobulin proteins or human serum albumin (HSA). Fusions of various proteins to HSA have been described, and are known to increase serum half-life, see U.S. Patents Nos. 5,876,969; 6,994,857; and 7,189,690.
[0068] Modified TWEAK proteins also include conjugates of TWEAK, or a fragment of TWEAK capable of binding TWEAKR, and high molecular weight dextrans or polyethylene glycol (PEG), which can be made according to methods known in the art. See U.S. Pat. No. 5,177,059 and 7,587,286. PEG has been shown to prolong half-life and reduce
immunogenicity of the conjugated protein. See Walsh et al, 2003, Antimicrob Agents Chemother. 47(2): 554-558 and Abuchowski et al, 1977, J Biol Chem 252:3582-3586. Similar increases in serum half-life and reductions in immunogenicity have been observed for dextran conjugates, see, e.g., Mehvar et al, 1992, J Pharm Sci, 81 :908-912.
[0069] A specific embodiment of a TWEAKR agonist is an anti-TWEAKR agonist antibody. Anti-TWEAKR agonist antibodies specifically bind to TWEAKR proteins. An example of a TWEAKR protein is the human TWEAKR protein encoded by the nucleotide sequence given by SEQ ID NO:l, or the protein of the amino acid sequence given by SEQ ID NO:2. See Table 2 below. For purposes of treating a subject using anti-TWEAKR agonist antibodies, the antibody preferably binds the TWEAKR from the same species as the subject. For example, when treating a human, an antibody to human TWEAKR is administered. [0070] A TWEAKR agonist antibody need not be a pure agonist. A TWEAKR agonist antibody can also have TWEAKR antagonist activity. Antagonist activity refers to the ability of test agent (e.g. a TWEAKR agonist antibody) to inhibit the induction by TWEAK of one or more biological responses associated with TWEAKR. For example, the
cytokine/chemokine release assay described above can be performed by incubating cells in vitro with an anti-TWEAKR antibody +/- TWEAK. Using the data generated from an cytokine/chemokine assay, the percent antagonist activity can be calculated using the formula: % antagonist=(b-d)/(b-a); in which a= quantity of cytokine/chemokine released from cells treated with antibody at 10 μg/ml, b= quantity of cytokine/chemokine released from cells treated with TWEAK at 300 ng/ml, c= quantity of cytokine/chemokine released from untreated cells, and d= quantity of cytokine/chemokine released from cells treated with TWEAK at 300 ng/ml and antibody at 10 μ^ιηΐ.
[0071] An anti-TWEAKR agonist antibody can bind to TWEAKR protein with a KD of at least about Ι Μ, at least about 0.1 μΜ or better, at least about 0.01 μΜ, and at least about 0.001 μΜ or better.
[0072] Unless indicated otherwise, the term "antibody" (Ab) refers to an immunoglobulin molecule that specifically binds to, or is immunologically reactive with, a particular antigen, and includes polyclonal, monoclonal, chimeric, humanized, fully human, genetically engineered and otherwise modified forms of antibodies.
[0073] A full-length antibody contains two heavy chains and two light chains, each of which comprises complementarity determining regions (CDRs), also known as hypervariable regions, and more highly conserved framework regions (FRs). As is known in the art, the amino acid position/boundary delineating a hypervariable region of an antibody can vary, depending on the context and the various definitions known in the art. Some positions within a variable domain may be viewed as hybrid hypervariable positions in that these positions can be deemed to be within a hypervariable region under one set of criteria while being deemed to be outside a hypervariable region under a different set of criteria. One or more of these positions can also be found in extended hypervariable regions. The present disclosure includes antibodies comprising modifications in these hybrid hypervariable positions. The variable domains of native heavy and light chains each comprise four FR regions, largely by adopting a β-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the β-sheet structure. The CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the target binding site of antibodies (See Kabat et al. ,
Sequences of Proteins of Immunological Interest (National Institute of Health, Bethesda, Md. 1987). As used herein, numbering of immunoglobulin amino acid residues is done according to the immunoglobulin amino acid residue numbering system of Kabat et al, unless otherwise indicated.
[0074] An anti-TWEAKR agonist antibody can be an antibody fragment. As used herein, the term "antibody fragment" refers to a portion of a full-length antibody capable of multivalent binding to the target, where multivalent refers to bivalent, trivalent, tetravalent, etc.
Examples of suitable antibody fragments include F(ab')2 fragments. F(ab')2 fragments are bivalent, having two antigen-binding F(ab) portions linked together by disulfide bonds, and contain portions of VH and VL chains. References to "VH" refer to the variable region of an immunoglobulin heavy chain of an antibody. References to "VL" refer to the variable region of an immunoglobulin light chain. It is in this configuration that the three CDRs— typically denoted CDR1, CDR2, and CDR3— of each variable domain interact to define a target binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer target binding specificity to the antibody. An F(ab')2 fragment can be produced by pepsin cleavage of a whole antibody, removing most of the immunoglobulin constant region (Fc) portion of the antibody. Such F(ab')2 fragments clear more rapidly from the circulation of the animal or plant, and may have less non-specific tissue binding than an intact antibody (Wahl et al. , 1983, J. Nucl. Med. 24:316). Multivalent antibody fragments can also be produced by linking CDRs using chemical couplings known to those of ordinary skill in the art.
[0075] The term "monoclonal antibody" as used herein is not limited to antibodies produced through hybridoma technology. The term "monoclonal antibody" refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. Monoclonal antibodies useful with the present disclosure can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. Moreover, unless otherwise indicated, the term monoclonal antibody (mAb) is meant to include both intact molecules, as well as, antibody fragments (such as, for example, F(ab')2 fragments) which are capable of specifically binding to a protein.
[0076] The anti-TWEAKR agonist antibodies of the disclosure can be chimeric antibodies. The term "chimeric" antibody as used herein refers to an antibody having variable sequences derived from a non-human immunoglobulins, such as rat or mouse antibody, and human immunoglobulins constant regions, typically chosen from a human immunoglobulin template. Methods for producing chimeric antibodies are known in the art. See, e.g., Morrison, 1985, Science 229(4719): 1202-7; Oi et al, 1986, BioTechniques 4:214-221; Gillies et al, 1985, J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715;
4,816,567; and 4,816397.
[0077] The anti-TWEAKR agonist antibodies of the disclosure can be humanized.
"Humanized" forms of non-human {e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as F(ab')2 or other target-binding subsequences of antibodies) which contain minimal sequences derived from non-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody can also comprise at least a portion of an Fc, typically that of a human
immunoglobulin consensus sequence. Methods of antibody humanization are known in the art. See, e.g., Riechmann et al, 1988, Nature 332:323-7; U.S. Pat. Nos: 5,530,101;
5,585,089; 5,693,761; 5,693,762; and 6,180,370 to Queen et al; EP239400; PCT publication WO 91/09967; U.S. Pat. No. 5,225,539; EP592106; EP519596; Padlan, 1991, Mol.
Immunol., 28:489-498; Studnicka et al, 1994, Prot. Eng. 7:805-814; Roguska et al, 1994, Proc. Natl. Acad. Sci. 91:969-973; and U.S. Pat. No. 5,565,332. [0078] The anti-TWEAKR agonist antibodies of the disclosure can be fully human antibodies. Fully human anti-TWEAKR agonist antibodies can be desirable for therapeutic treatment of human patients. As used herein, "fully human antibodies" include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See U.S. Pat. Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645; WO 98/50433; WO 98/24893; WO 98/16654; WO 96/34096; WO 96/33735; and WO 91/10741. Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. See, e.g., PCT publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; U.S. Patent Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825; 5,661,016; 5,545,806; 5,814,318; 5,885,793; 5,916,771; and 5,939,598. Fully human antibodies that recognize a selected epitope can be generated using a technique referred to as "guided selection." In this approach a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a fully human antibody recognizing the same epitope (Jespers et al, 1988, Biotechnology 12:899-903).
[0079] The anti-TWEAKR agonist antibodies can be of any of the recognized isotypes. In some embodiments, anti-TWEAKR agonist antibodies are one of the four human IgG isotypes, i.e., IgGl, IgG2, IgG3 and IgG4, or one of the four mouse IgG isotypes, i.e., murine IgGl, murine IgG2a, murine IgG2b, or murine IgG3. In other embodiments, the anti- TWEAKR antibodies are of the human IgGl isotype.
[0080] Anti-TWEAKR agonist antibodies useful in the present methods include antibodies that induce antibody-dependent cytotoxicity (ADCC) of TWEAKR-expressing cells. The ADCC of an anti-TWEAKR antibody can be improved by using antibodies that have low levels of or lack fucose. Antibodies lacking fucose have been correlated with enhanced ADCC (antibody-dependent cellular cytotoxicity) activity, especially at low doses of antibody (Shields et al, 2002, J. Biol. Chem. 277:26733-26740; Shinkawa et al, 2003, J. Biol. Chem. 278:3466). Methods of preparing fucose-less antibodies include growth in rat myeloma YB2/0 cells (ATCC CRL 1662). YB2/0 cells express low levels of FUT8 mRNA, which encodes an enzyme (a-l,6-fucosyltransferase) necessary for fucosylation of polypeptides. Alternative methods for increasing ADCC activity include mutations in the Fc portion of an anti-TWEAKR agonist antibody, particularly mutations which increase antibody affinity for an FcyR receptor. A correlation between increased FcyR binding with mutated Fc has been demonstrated using targeted cytoxicity cell-based assays (Shields et al, 2001, J. Biol. Chem. 276:6591-6604; Presta et al, 2002, Biochem Soc. Trans. 30:487- 490). In specific embodiments, an anti-TWEAKR agonist antibody of the disclosure has a constant region that binds FcyRIIA, FcyRIIB and/or FcyRIIIA with greater affinity than the corresponding wild type constant region. Mutations in the Fc portion of an antibody that result in increased ADCC activity and methods of generating immunoglobulins with increased ADCC activity are known in the art and described in various references, e.g., U.S. Pat. No. 8,039,592, U.S. Pat. No. 7,632,497, U.S. Pat. No. 7,183,387, U.S. Pat. Pub. No. 2009/0074762, WO 00/42072.
[0081] ADCC activity can be monitored and quantified using a 51Cr release assay. Target cells are loaded with 5ICr, which is released in culture supernatant upon damage to the plasma membrane in the presence of immune effector cells and an anti-TWEAKR agonist antibody. The target cells can be derived from solid tumors, for example lung, pancreatic, breast, or renal cancer cells. In various embodiments, the anti-TWEAKR agonist antibodies induce at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, or at least 80% cytotoxicity in the target cells. An example of an ADCC assay that can be used to measure ADCC of an anti-TWEAKR agonist antibody is provided in Culp et al, 2010, Clin. Cancer Res. 16(2):497-508.
[0082] Anti-TWEAKR agonist antibodies or fragments thereof as described herein can be antibodies or antibody fragments whose sequence has been modified to reduce at least one constant region-mediated biological effector function relative to the corresponding wild type sequence. To modify an antibody of the disclosure such that it exhibits reduced binding to the Fc receptor, the immunoglobulin constant region segment of the antibody can be mutated at particular regions necessary for Fc receptor (FcR) interactions (see e.g., Canfield and Morrison, 1991, J. Exp. Med. 173:1483-1491; and Lund et al, 1991, J. Immunol.
147:2657-2662). Reduction in FcR binding ability of the antibody can also reduce other effector functions which rely on FcR interactions, such as opsonization and phagocytosis.
[0083] Anti-TWEAKR agonist antibodies or fragments thereof as described herein can be antibodies or antibody fragments that have been modified to increase or reduce their binding affinities to the fetal Fc receptor, FcRn, or serum half-life, for example by mutating the immunoglobulin constant region segment at particular regions involved in FcRn interactions (see, e.g., WO 2005/123780 and U.S. Pat. No. 5,739,277).
[0084] Specific examples of anti-TWEAKR agonist antibodies and sequences of VH, VL and CDRs, which are described in U.S. Pat. Pub. No. 2009/0074762 and provided in Table 2 herein, include:
(a) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, comprising a heavy chain variable region corresponding to SEQ ID NO: 3 and a light chain variable region corresponding to SEQ ID NO: 4 (PDL192);
(b) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, comprising a heavy chain variable region corresponding to SEQ ID NO: 11 and a light chain variable region corresponding to SEQ ID NO: 12 (PDL400);
(c) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, wherein VH CDR1 is XiYWMX2 (SEQ ID NO:49), VH CDR2 is EIRX3KSX4NYATX5HYAESX6KG (SEQ ID NO:50), VH CDR3 is X7X8ADX9X10DY (SEQ ID NO:51); VL CDR1 is XnASQSVSTSX12YSYMX13 (SEQ ID NO:52); VL CDR2 is YAX14X15LX16S (SEQ ID NO:53); and VL CDR3 is QHSWEX17PX18T (SEQ ID NO:54), wherein: Xj is selected from K, N, R, and S; X2 is selected from N and S; X3 is selected from L and V; j is selected from D and N; X5 is T or no amino acid; X6 is selected from A and V; X7 is selected from A, G, T and Y; X8 is selected from F and Y; X9 is selected from A, T and Y; X10 is selected from F and M; Xn is selected from R and K; X12 is selected from S and T; X13 is selected from H and Q; X14 is selected from S and T; X15 is selected from N and K; X16 is selected from E and D; and X17 is selected from I and L; and X18 is selected from W and Y;
(d) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, wherein VH CDRl is SYWMS (SEQ ID NO: 13), VH CDR2 is EIRLKSDNYATHYAESVKG (SEQ ID NO: 19), VH CDR3 is
YYADAMDY (SEQ ID NO:25), VL CDRl is RASQSVSTSSYSYMH (SEQ ID NO:31), VL CDR2 is YASNLES (SEQ ID NO:37), and VL CDR3 is QHSWEIPYT (SEQ ID NO:43);
(e) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, wherein VH CDRl is NYWMS (SEQ ID NO: 15), VH CDR2 is EIRLKSDNYATHYAESVKG (SEQ ID NO:21), VH CDR3 is GFADYFDY (SEQ ID NO:27), VL CDRl is RASQSVSTSSYSYMQ (SEQ ID NO:33), VL CDR2 is YATNLDS (SEQ ID NO:39), and VL CDR3 is QHSWEIPYT (SEQ ID NO:45);
(f) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, wherein VH CDRl is KYWMN (SEQ ID NO: 16), VH CDR2 is EIRLKSDNYATHYAESAKG (SEQ ID NO:22), VH CDR3 is
TYADTMDY (SEQ ID NO:28), VL CDRl is KASQSVSTSTYSYMQ (SEQ ID NO:34), VL CDR2 is YASKLDS (SEQ ID NO:40), and VL CDR3 is QHSWELPYT (SEQ ID NO:46);
(g) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, wherein VH CDRl is RYWMS (SEQ ID NO: 17), VH CDR2 is EIRVKSDNYATTHYAESVKG (SEQ ID NO:23), VH CDR3 is
YYADAMDY (SEQ ID NO:29), VL CDRl is RASQSVSTSSYSYMH (SEQ ID NO:35), VL CDR2 is YASKLDS (SEQ ID NO:41), and VL CDR3 is QHSWEIPWT (SEQ ID NO:47); and
(h) an anti-TWEAKR antibody that is a monoclonal antibody or anti-TWEAKR antigen binding fragment, wherein VH CDR1 is NYWMN (SEQ ID NO: 17), VH CDR2 is EIRLKSNNYATHYAESVKG (SEQ ID NO:24), VH CDR3 is
AYADYFDY (SEQ ID NO:30), VL CDR1 is RASQSVSTSTYSYMH (SEQ ID NO:36), VL CDR2 is YASNLES (SEQ ID NO:42), and VL CDR3 is QHSWEIPYT (SEQ ID NO:48).
[0085] Anti-TWEAKR agonist antibodies of the present disclosure also include antibodies that compete for binding to TWEAKR with a reference antibody of interest. Any of the antibodies described herein can be used as a reference antibody in a competition assay. A competition assay can be carried out between a test antibody and a reference antibody. The assay is conducted by first labeling the reference antibody with a detectable label, such as, biotin, or an enzymatic, radioactive, or fluorescent label to enable detection. The unlabeled test antibody is incubated (in fixed or increasing amounts) with a known amount of
TWEAKR, forming an anti-TWEAKR antibody/ TWEAKR complex. The labeled reference antibody is then added to the complex. The intensity of the complexed label is measured. If the test antibody competes with the labeled antibody by binding to an overlapping epitope, the intensity will be decreased relative to the binding of the labeled reference antibody in the absence of the test antibody. Numerous methods for carrying out binding competition assays are known. An antibody is considered to compete for binding TWEAKR with a reference anti-TWEAKR agonist antibody, and thus considered to bind approximately the same or overlapping epitope of TWEAKR as the reference antibody, if it reduces binding of the reference antibody by at least 50% at a test antibody concentration in the range of 0.01 to 100 μg/mL, although higher levels of reduction, for example, 60%, 70%, 80%, 90% or even 100%, may be desirable.
5.5. TWEAKR Positive Cancers
[0086] The methods described herein are useful for treatment of subjects suffering from a TWEAKR positive cancer. The subject may be any animal, for example, a mammal, such as a farm animal or domesticated animal, or a human. A TWEAKR positive cancer is a cancer in which TWEAKR is overexpressed as compared to matched non-cancer tissue. TWEAKR expression can be determined by detecting the presence of TWEAKR mRNA or TWEAKR protein, using standard techniques such as PCR, Northern blotting, microarray analysis (for mRNA detection) or Western blotting, ELISA, or immunohistochemistry (for protein detection). Specific assays for detection of TWEAKR expression using microarray analysis and immunohistochemistry are described in Culp et al, 2010, Clin. Cancer Res. 16(2):497- 508. Using these and other assays, TWEAKR has been shown to be overexpressed in a number of human cancers. See Culp et al, 2010, Clin. Cancer Res. 16(2):497-508; Feng et al, 2000, Am. J. Pathol. 156:1253-1261; Han et al, 2002, Cancer Res. 62:2890-2896; Wang et al, 2006, Oncogene 25:3346-3356. As detailed in Example 1, a test of 102 cancer cell lines further shows that TWEAKR is expressed in a wide range of cancers, both primary and metastatic. Thus, TWEAKR positive cancers include breast cancer, lung cancer, melanoma, ovarian cancer, uterine cancer, colon cancer, renal cancer, pancreatic cancer, cervical cancer, bladder cancer, head and neck cancer, glioblastoma, esophageal cancer, sarcomas, and salivary gland cancer.
5.6. Methods of Treating a TWEAKR Positive Cancer
[0087] The present disclosure provides methods of treating a TWEAKR positive cancer. In methods of the disclosure, a subject who has tested negative for at least one PIK3CA activating mutation is treated with an amount of a TWEAKR agonist effective to promote a therapeutic benefit. In another aspect, a subject who has tested negative for at least one PTEN null mutation is treated with an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
[0088] Optionally, a subject who has tested negative for at least one PIK3CA activating mutation has also tested negative for at least one PTEN null mutation. A subject who has tested negative for at least one PTEN null mutation may also have tested negative for at least one PIK3CA activating mutation.
[0089] The TWEAKR agonists are administered to a subject in an amount effective to promote a therapeutic benefit. The amount effective to promote a therapeutic benefit refers to the amount of a pharmaceutical formulation or composition that is sufficient to cure, alleviate, attenuate or at least partially arrest the cancer and/or its symptoms, and/or complications. Clinical methods for determining a therapeutic benefit are well known to those of ordinary skill in the art and may be determined empirically using routine
experimentation. For example, in the context of cancer treatment, an amount effective to promote a therapeutic benefit is an amount capable of invoking one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, slowing down and complete growth arrest; (2) reduction in the number of cancer cells; (3) reduction in tumor size; (4) inhibition (i.e., reduction, slowing down or complete stopping) of cancer cell infiltration into peripheral organs; (5) inhibition (i.e., reduction, slowing down or complete stopping) of cancer cell metastasis; (6) enhancement of anti-cancer immune response, which may, but does not have to, result in the regression or rejection of a tumor; and/or (7) relief, to some extent, of one or more symptoms associated with the disorder. A complete cure, while desirable, is not required for therapeutic benefit to exist. Tumor size and/or number can be measured using various scanning techniques, such as, but not limited to CT, MRI, functional MRI, SPECT and PET, as well as other methods known to those skilled in the art.
[0090] The TWEAKR agonist can be used alone, as monotherapy, or in combination with or adjunctive to other therapies commonly used to treat the specific type (e.g., lung, breast, colon, etc.) of TWEAKR positive cancer. Other therapies include surgery, radiation therapy, and treatment with conventional therapeutic agents, such as targeted agents, conventional chemotherapy agents, and hormonal therapy agents. The conventional standard of care for a particular cancer is known to those skilled in the art and thus, one of skill in the art will be able to select suitable other therapy to combine with the TWEAKR agonist described herein. When used in combination with other treatments, the TWEAKR agonist and other therapy can be administered simultaneously, successively, or separately. Therapies that can be combined with TWEAKR agonist therapy are described in, e.g., U.S. Pub. No. US
2009/0074762.
[0091] In some embodiments, the TWEAKR agonist can be used in combination with conventional therapeutic agents used to treat a particular cancer. Examples of therapeutic agents that are presently used to treat cancer, and thus can be used in combination with the TWEAKR agonist described herein, include, but are not limited to, targeted agents, conventional chemotherapy agents, and hormonal therapy agents.
[0092] In some embodiments, the TWEAKR agonist described herein can be used in combination with targeted agents. Targeted agents include, but are not limited to, antiangiogenic agents (such as bevacizumab, sunitinib, sorafenib, temsirolimus, 2- methoxyestradiol or 2ME2, finasunate, PTK787, and vandetanib), EGFR inhibitors (such as erlotinib, cetuximab, panitumumab, gefinitib, lapatinib, and trastuzumab),
immunomodulators (such as rituximab, alemtuzumab, and aldesleukine), proteasome inhibitors (such as bortezomib, PR-171, and NPI-052), integral inhibitors (such as natalizumab, volociximab, etaracizumab, and cilengitide), pro-apoptotic agents (such as mapatumumab, lexatumumab, AMG951, ABT-737, oblimersen, and plitidepsin), and agents with other mechanisms of action (such as imatinib, dasatinib, lenalidomide, thalidomide, aldesleukin, and interferon alpha).
[0093] In some embodiments, the TWEAKR agonist described herein can be used in combination with conventional chemotherapy agents. Conventional chemotherapy agents include, but are not limited to, alkylating agents (such as oxaliplatin, carboplatin, cisplatin, cyclophosphamide, melphalan, ifosfamide, uramustine, chlorambucil, mechloethamine, thiotepa, busulfan, temozolomide, and dacarbazine), anti-metabolites (gemcitabine, cytosine arabinoside, Ara-C, capecitabine, 5FU (5-fluorouracil), azathioprinc, mercaplopurine (6- MP), 6-thioguanine, aminopterin, pemetrexed, and methotrexate), plant alkaloids and terpenoids (such as docetaxel, paclitaxel, protein-bound paclitaxel, vincristine, vinblastin, vinorelbine, vindesine, etoposide, VP- 16, teniposide, irinotecan, and topotecan), and antitumor antibiotics (such as dactinomycin, doxorubicin, liposomal doxorubicin, daunorubicin, daunomycin, epirubicin, mitoxantrone, adriamycin, bleomycin, plicamycin, mitomycin C, caminomycin, and esperamicins).
[0094] In some embodiments, the TWEAKR agonist described herein can be used in combination with hormonal therapy agents including, but not limited to, anastrozole, letrozole, goserelin, and tamoxifen. [0095] The concentration of a TWEAKR agonist in formulations administered by methods of the disclosure varies widely from about 0.1 to 100 mg/ml, but is often in the range 1 to 20 mg/ml. For the purpose of treatment of disease, the appropriate dosage of the antibody will depend on the severity and course of disease, the patient's clinical history and response, the toxicity of the antibodies, and the discretion of the attending physician. Where the TWEAKR agonist is an antibody, the dose can range from about 0.1 to about 1 mg/kg, about 1 to about 5 mg/kg, about 1 to about 10 mg/kg, and about 10 to about 20 mg/kg. The antibodies are suitably administered to the patient at one time or over a series of treatments. The proper dosage and treatment regimen can be established by monitoring the progress of therapy using conventional techniques known to the people skilled of the art. Typically, the methods described herein provide for administration of a TWEAKR agonist to a patient intravenously as a bolus or by continuous infusion over a period of time; or by intramuscular,
subcutaneous, intraperitoneal, or intra-cerebrospinal routes. Methods for preparing parentally administrable compositions are known or apparent to those skilled in the art and are described in more detail in, for example, Remington's Pharmaceutical Science (15th Ed., Mack Publishing Company, Easton, Pa., 1980), which is incorporated herein by reference.
[0096] The TWEAKR agonist compositions to be administered are formulated for pharmaceutical use. The compositions optionally further comprise a carrier. The
pharmaceutical compositions described herein typically comprise a TWEAKR agonist and a pharmaceutical carrier, and, commonly they comprise a solution of a TWEAKR agonist, or a cocktail thereof, dissolved in an acceptable carrier, preferably an aqueous carrier. A variety of aqueous carriers can be used, e.g., water for injection (WFI), or water buffered with phosphate, citrate, acetate, etc. to a pH typically of 5.0 to 8.0, most often 6.0 to 7.0, and/or containing salts such as sodium chloride, potassium chloride, etc. to make the composition isotonic. The carrier can also contain excipients such as human serum albumin, polysorbate 80, sugars or amino acids to protect the agonist.
[0097] Administration of a TWEAKR agonist can trigger killing of cancer cells. The cancer cells can be present within a solid tumor, the lymph system, or in the bloodstream. The therapeutic methods described herein are usually applied to human patients, but can be applied to other mammals.
[0098] Administration of a TWEAKR agonist can induce apoptosis or cyto lysis of cells expressing TWEAKR. In some embodiments, induction of cytolysis is achieved via antibody-dependent cellular cytotoxicity (ADCC). For example, the TWEAKR agonist can induce between 10% to greater than 80% cytotoxicity of cells expressing TWEAKR.
[0099] In some embodiments, administration of the TWEAKR agonists induces at least 10%, 20%, 30%, 40%, 50%, 60%, or 80% or more cytotoxicity of cells expressing
TWEAKR. Administration of the TWEAKR agonist can reduce the size of a solid tumor by targeting TWEAKR on the cancer cell's surface with one or more of the antibodies described herein. The tumor can be a primary tumor or a secondary tumor. By way of example, administration of the TWEAKR agonists can reduce the size of a solid tumor by at least 10%, 20%, 30%, 40%, 50%, 60%, or 80% or more. In other embodiments, administration of the TWEAKR agonists can completely inhibit or prevent the growth of a solid tumor. It will be appreciated that a range of different cytotoxicities and a range of reductions in tumor size are described for the TWEAKR agonists disclosed herein. The skilled person will appreciate that the TWEAKR agonists can have any one of the described cytotoxicities and any one of the described reductions in tumor size.
[0100] The term responder in the context of the present disclosure includes persons where the cancer/tumor(s) is eradicated, reduced or improved, or stabilized such that the disease is not progressing after treatment. In responders where the cancer is stabilized, the period of stabilization is such that the quality of life and/or patients' life expectancy is increased (for example stable disease for more than 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more months) in comparison to a patient that does not receive treatment. A non-responder includes persons for whom the cancer/tumor(s) does not show reduction or improvement after treatment. Optionally the characterization of the patient as a responder or non-responder can be performed by reference to a standard or a training set. The standard may be the profile of a person/patient who is known to be a responder or nonresponder or alternatively may be a numerical value. Such pre-determined standards may be provided in any suitable form, such as a printed list or diagram, computer software program, or other media.
6. EXAMPLES
Example 1: TWEAKR agonist PDL192 inhibits growth of a wide range of cancers in vitro
[0101] This example shows that a monoclonal antibody that acts as a TWEAKR agonist, PDL192 (used interchangeable herein with enavatuzumab), inhibits the growth of a wide range of cancer cell lines, including breast cancer, pancreatic cancer, cervical cancer, colon cancer, bladder cancer, lung cancer and melanoma cancer cell lines, in vitro.
[0102] Antibodies. PDL192 is a humanized monoclonal antibody to TWEAKR described in US Pat. App. No. 2009/0074762 and Culp et al, 2010, Clin. Cancer Res. 16(2):497-508. Human IgGl antibody to cytomegalovirus (MSL109) was used as a negative control.
Antibodies were used at 10 μg ml for in vitro studies, unless otherwise stated, and crosslinked with F(ab')2 goat anti-human IgG (Fey specific) from Jackson ImmunoResearch at 3.5 μg/ml.
[0103] Cell lines. All cell lines were obtained from the American Tissue Culture Collection (ATCC) or National Cancer Institute (NCI), except HSC-3, which was kindly provided by the Japan Health Science Foundation, and the MDA-MB231 variant cell line, which was derived from the MDA-MB-231 cell line and selected for consistent growth in vivo.
[0104] Growth inhibition assay. 102 cancer cell lines were cultured at 500 cells per well in triplicate with enavatuzumab or IgGl control in the presence of F(ab')2 goat anti-human IgG, (Fey specific) for 5 days in 96 well plates. Relative cell viability was determined using CellTiter-Blue™ (Promega). Fluorescence emitted at 590 nm was used to calculate the growth effect relative to the IgGl control antibody treatment. Each cell line was tested twice with the average growth inhibition reported. Cell lines with at least about 20% growth inhibition were considered TWEAKR agonist sensitive.
[0105] Results. Inhibition of proliferation by PDL192, as compared to an isotype control antibody, was tested in 102 cancer cell lines representing the majority of solid tumor types. Of these, 38 (or 37%) cell lines showed growth inhibition of at least 20% (relative to growth observed with the control antibody) in response to PDL192 (FIG. 1). Table 1 below summarizes the TWEAKR-agonist sensitive and TWEAKR-agonist resistant cell lines for each type of cancer tested.
Table 1
Cancer Sensitive Resistant
type (at least 20% growth (<20% growth
inhibition) inhibition)
Breast BT-549 BT-20
CAMA-1 BT-474
HCC1143 DU-4475
HCC1500 HCC-1428
HCC1569 Hs 578T
HCC1937 MCF-7
HCC38 MDA-MB-175-VII
HCC70 MDA-MB-361
MDA-MB-157 MDA-MB-435S
MDA-MB-231 MX-1
MDA-MB231 variant T47D
MDA-MB-453 ZR-75-1
MDA-MB-468 ZR-75-30
NCI-ADR-RES
SK-BR-3
Colon HT-29 COLO-205
SW480 HCC2998
HCT-116
HCT-15
KM12
LoVo
SW48
SW620
SW837
SW948 Table 1
Cancer Sensitive Resistant type (at least 20% growth (<20% growth inhibition) inhibition)
Lung A549 Calu-3
Calu-6 DMS-79
EKVX NCI-H1155
HOP-62 NCI-H146
HOP-92 NCI-H157
NCI-H1792 NCI-H1838
NCI-H358 NCI-H187
NCI-H209
NCI-H226
NCI-H23
NCI-H292
NCI-H322M
NCI-H345
NCI-H460
NCI-H522
NCI-H596
NCI-H69
NCI-H82
Renal ACHN 786-0
CAKI-1 A498
RXF393
SN12C
TK10
U031
Melanoma A375 A2058
Malme-3M C32
SK-MEL-2 HT-144
SK-MEL-28 LOX
M14
MDA-MB-435
SK-MEL-5
UACC-257
UACC-62
WM-115 Table 1
Cancer Sensitive Resistant
type (at least 20% growth (<20% growth
inhibition) inhibition)
Ovarian PA-1 IGROV-1
OVCAR-3 OVCAR-4
OVCAR-5
OVCAR-8
SK-OV-3
SW626 (metastasis
from colon)
Pancreatic AsPC-1 MIA-PaCa-2
BxPC3
Bladder SW780 —
Cervical HT3 ~
Salivary A253 —
Oral HSC-3 —
[0106] PDL192 displayed the ability to block proliferation in some but not other cell lines from each of the following tumor types: breast cancer, lung cancer, colon cancer, renal cancer, ovarian cancer, melanoma, and pancreatic cancer. PDL192 also blocked cell proliferation in cell lines derived from bladder cancer, cervical cancer, salivary cancer and oral cancer tumors.
Example 2: In vitro correlation of PIK3CA activating mutations with responsiveness to PDL192
[0107] This example shows that PDL192 does not have in vitro activity in the presence of PIK3CA activating mutations. 91 cell lines were classified based on the presence or absence of a PIK3CA activating mutation, using information from the Sanger Wellcome database (see Wang et al, PNAS (2009) 106(15):6279-6284).
[0108] The mutation data were then correlated with responsiveness or non-responsiveness of the cell line to treatment with PDL192, in a growth inhibition assay as described in Example 1. The results are summarized in FIG. 2. Of cell lines bearing a PIK3CA activating mutation, one responded to PDL192 and nine did not respond. In contrast, of 81 cell lines that did not carry a PIK3CA activating mutation, 34 were responders to PDL192 while 47 were nonresponders.
Example 3: In vivo correlation of PIK3CA activating mutations with
non-responsiveness to PDL192
[0109] This example shows a correlation between mutations activating the PI3K pathway and non-responsiveness to PDL192 treatment in vivo. Somatic mutation analysis was performed on cells from 26 tumors to determine if a PIK3CA activating mutation or a PTEN null mutation was present. The results are shown in FIG. 3A. Of the tumors that contained either a PIK3CA activating mutation or a PTEN null mutation, three of sixteen were responsive to PDL192 while thirteen were non-responsive. Response was defined as statistically significant decrease in tumor volumes in the PDL192-treated animals compared to control-treated tumors. In contrast, for tumors whose cells bore wild type PIK3CA and wild type PTEN, eleven of fourteen were responsive to PDL192 and three were non- responsive.
[0110] Analysis of a subset of the responsive cells for action of ADCC showed that some of the responders were killed through ADCC. A version of PDL192, PDL192M, containing two mutations in the Fc domain critical for interaction with FcR's, L234A/L235A, was tested in 9 cell line xenograft models known to be responsive to PDL192. In 6 of the cell lines,
PDL192M had no anti-tumor activity, suggesting that ADCC is the predominant mechanism of action for PDL192 in these models. In 3 of the cell lines (A375, HT29, and H358), PDL192M had activity equivalent to PDL192; thus, direct growth inhibitory activity is presumed to be the predominant mechanism of action for PDL192 in these lines. As shown in FIG. 3B, none of the 3 cell lines responding to PDL192 via direct growth inhibition in vivo harbored activating mutations in PIK3CA or PTEN null mutations; in contrast, 13 of 16 nonresponding cell lines carried such PIK3CA or PTEN mutations.
[0111] Further cell lines with wild-type PIK3CA and PTEN that responded to PDL192 included Lox, MiaPaCa2, SKMEL5, SN12C, A253, A549, Calu6, and HT1376. Of these, Lox, MiaPaCa2, SKMEL5, and SN12C were tested for and showed an ADCC-dependent response. The MCF7 cell line carries a PIK3CA mutation (E545K), and the HCC70 cell line carries a PTEN mutation (F90fs*9). Both MCF7 and HCC70 displayed an ADCC-dependent response to PDL 192.
Example 4: Phosphoprotein profiling analysis following PDL 192
administration
[0112] Six cell lines (HT3, SW780, MB468, HT144, T47D, and UACC62) were treated with PDL 192 for 0 h, 0.5 h, 4 h, 8 h, and 24 h, and the cells were analyzed for phosphorylation of protein biomarkers following treatment using Reverse Phase Protein Array (RPPA).
[0113] Phosphoprotein status was compared to the baseline of 0 h treatment. HT3, SW780, and MB468 cells were responsive to PDL192, while HT144, T47D and UACC62 were non- responsive. The results of the analysis are shown in FIG. 4. The analysis indicated that Ser473 phosphorylation of Akt, a known member of the PI3K pathway to which PIK3CA belongs, is decreased in 2 of 3 PDL192-responsive cells upon PDL 192 treatment, while in non-responsive cells, phosphorylation of Akt is increased in one cell line, unchanged in a second line, and decreased in a third line upon PDL 192 treatment.
Example 5: PIK3CA overexpression studies
[0114] Wild type A375 cells were transfected with vector alone, wild type PIK3CA,
PIK3CA E545K mutant, or PIK3CA H1047R mutant. E545K and H1047R have been shown to be PIK3CA activating mutations. Following transfection, the cells were treated with PDL 192 or control MSL109 compounds, and response was analyzed using the growth inhibition assay of Example 1. As shown in FIG. 5, cells transfected with vector alone or with wild type PIK3CA showed a dose-dependent response to PDL 192. In contrast, cells that received vectors bearing PIK3CA activating mutations showed a decreased response to PDL 192. This study showed that activation of PIK3CA results in decreased sensitivity to PDL 192 treatment. Example 6: PIK3CA depletion studies
[0115] Two cell lines bearing PIK3CA activating mutations were subjected to siRNA knockdown of PIK3CA or control, and subsequently tested for PDL192 responsiveness. The DLD-1 cell line bears an E545K PIK3CA activating mutation, while the HCT116 cell line bears an H1047R PIK3CA activating mutation. Each of the two cell lines was treated with 10 nM control siRNA or 10 nM PIK3CA siRNA (for DLD-1) or 2.5 nM PIK3CA siRNA (for HCT116). As shown in FIG. 6, cell lines receiving control siRNA did not respond to PDL192 (compared to control compound, MSL109) while cell lines that had been subjected to PIK3CA siRNA depletion showed a dose-dependent response to PDL192.
7. SEQUENCE LISTING
[0116] Table 2 below provides sequences of SEQ ID NOs:l-60.
TABLE 2
SEQ ID Description SEQUENCE
NO:
TCGACCCACG CGTCCGCCCA CGCGTCCGCC CACGCGTCCG GGCGCAGGAC GTGCACTATG
1 Human TWEAK-R GCTCGGGGCT CGCTGCGCCG GTTGCTGCGG CTCCTCGTGC TGGGGCTCTG GCTGGCGTTG nucleic acid CTGCGCTCCG TGGCCGGGGA GCAAGCGCCA GGCACCGCCC CCTGCTCCCG CGGCAGCTCC
TGGAGCGCGG ACCTGGACAA GTGCATGGAC TGCGCGTCTT GCAGGGCGCG ACCGCACAGC GACTTCTGCC TGGGCTGCGC TGCAGCACCT CCTGCCCCCT TCCGGCTGCT TTGGCCCATC CTTGGGGGCG CTCTGAGCCT GACCTTCGTG CTGGGGCTGC TTTCTGGCTT TTTGGTCTGG AGACGATGCC GCAGGAGAGA GAAGTTCACC ACCCCCATAG AGGAGACCGG CGGAGAGGGC TGCCCAGCTG TGGCGCTGAT CCAGTGACAA TGTGCCCCCT GCCAGCCGGG GCTCGCCCAC TCATCATTCA TTCATCCATT CTAGAGCCAG TCTCTGCCTC CCAGACGCGG CGGGAGCCAA GCTCCTCCAA CCACAAGGGG GGTGGGGGGC GGTGAATCAC CTCTGAGGCC TGGGCCCAGG GTTCAGGGGA ACCTTCCAAG GTGTCTGGTT GCCCTGCCTC TGGCTCCAGA ACAGAAAGGG AGCCTCACGC TGGCTCACAC AAAACAGCTG ACACTGACTA AGGAACTGCA GCATTTGCAC AGGGGAGGGG GGTGCCCTCC TTCCTAGAGG CCCTGGGGGC CAGGCTGACT TGGGGGGCAG ACTTGACACT AGGCCCCACT CACTCAGATG TCCTGAAATT CCACCACGGG GGTCACCCTG GGGGGTTAGG GACCTATTTT TAACACTAGG GGGCTGGCCC ACTAGGAGGG CTGGCCCTAA GATACAGACC CCCCCAACTC CCCAAAGCGG GGAGGAGATA TTTATTTTGG GGAGAGTTTG GAGGGGAGGG AGAATTTATT AATAAAAGAA TCTTTAACTT TAAAAAAAAA AAAAAAAAAG GGCGGCCGCT CTAGAGGATC CCTC
Human TWEAK-R MARGSLRRLL RLLVLGLWLA LLRSVAGEQA PGTAPCSRGS SWSADLDKCM DCASCRARPH protein SDFCLGCAAA PPAPFRLLWP ILGGALSLTF VLGLLSGFLV WRRCRRREKF TTPIEETGGE
GCPAVALIQ
3 PDL192 VH EVQLVESGGG LVQPGGSLRL SCAASGFTFS SYWMSWVRQA PGKGLEWVAE IRLKSDNYAT
HYAESVKGRF TISRDDSKNS LYLQMNSLRA EDTAVYYCTG YYADAMDYWG QGTLVTVSS
4 PDL192 VL DIQMTQSPSS LSASVGDRVT ITCRASQSVS TSSYSYMHWY QQKPGKAPKL LIKYASNLES
GVPSRFSGSG SGTDFTLTIS SLQPEDFATY YCQHSWEIPY TFGGGTKVEI KR
5 19.2.1 mVH EVKLEESGGG LVQPGGSMKL SCVASGFTFS SYWMSWVRQS PEKGLEWVAE IRLKSDNYAT
HYAESVKGKF TISRDDSKSR LYLQMNSLRA EDTGIYYCTG YYADAMDYWG QGTSVTVSS
6 19.2.1 mVL DIVLTQSPAS LAVSLGQRAT ISCRASQSVS TSSYSYMHWY QQKPGQPPKL LIKYASNLES
GVPARFSGSG SGTDFTLNIH PVEEEDTATY YCQHSWEIPY TFGGGTKLEI KR
Figure imgf000044_0001
Figure imgf000045_0001
TABLE 2
SEQ ID Description SEQUENCE
NO:
42 VL CDR2 - ITEM-1 YASNLES
43 VL CDR3 - PDL192 QHSWEIPYT
44 VL CDR3- 19.2.1 QHSWEIPYT
45 VL CDR3 - 136.1 QHSWEIPYT
46 VL CDR3 - 18.3.3 QHSWELPYT
47 VL CDR3 - ITEM-3 QHSWEIPWT
48 VL CDR3 - ITEM-1 QHSWEIPYT
49 VH CDRl XiY MXj
50 VHCDR2 EIRX3KSX4NYATX=HYAESXeKG
51 VH CDR3 X7X8ADX9X10DY
52 VL CDRl XnASQSVSTSX12YSYMX:
53 VL CDR2 YAX X T X S
54 VL CDR3 QHSWEX1VPX18T
55 PIK3CA coding region atgcctccaa gaccatcatc aggtgaactg tggggcatcc acttgatgcc cccaagaatc ctagtggaat gtttactacc aaatggaatg atagtgactt tagaatgcct ccgtgaggct acattagtaa ctataaagca tgaactattt aaagaagcaa gaaaataccc tctccatcaa cttcttcaag atgaatcttc ttacattttc gtaagtgtta cccaagaagc agaaagggaa gaattttttg atgaaacaag acgactttgt gatcttcggc tttttcaacc atttttaaaa gtaattgaac cagtaggcaa ccgtgaagaa aagatcctca atcgagaaat tggttttgct atcggcatgc cagtgtgcga atttgatatg gttaaagatc ctgaagtaca ggacttccga agaaatattc ttaatgtttg taaagaagct gtggatctta gggatcttaa ttcacctcat agtagagcaa tgtatgtcta tccgccacat gtagaatctt caccagagct gccaaagcac atatataata aattggatag aggccaaata atagtggtga tttgggtaat agtttctcca aataatgaca agcagaagta tactctgaaa atcaaccatg actgtgtgcc agaacaagta
TABLE 2
SEQ ID Description SEQUENCE
NO:
attgctgaag caatcaggaa aaaaactaga agtatgttgc tatcatctga acaattaaaa ctctgtgttt tagaatatca gggcaagtac attttaaaag tgtgtggatg tgatgaatac ttcctagaaa aatatcctct gagtcagtat aagtatataa gaagctgtat aatgcttggg aggatgccca atttgaagat gatggctaaa gaaagccttt attctcaact gccaatggac tgttttacaa tgccatctta ttccagacgc atttccacag ctacaccata tatgaatgga gaaacatcta caaaatccct ttgggttata aatagagcac tcagaataaa aattctttgt gcaacctacg tgaatctaaa tattcgagac attgacaaga tttatgttcg aacaggtatc taccatggag gagaaccctt atgtgacaat gtgaacactc aaagagtacc ttgttccaat cccaggtgga atgaatggct gaattatgat atatacattc ctgatcttcc tcgtgctgct cgactttgcc tttccatttg ctctgttaaa ggccgaaagg gtgctaaaga ggaacactgt ccattggcat ggggaaatat aaacttgttt gattacacag acactctagt atctggaaaa atggctttga atctttggcc agtacctcat ggattagaag atttgctgaa ccctattggt gttactggat caaatccaaa taaagaaact ccatgcttag agttggagtt tgactggttc agcagtgtgg taaagttccc agatatgtca gtgattgaag agcatgccaa ttggtctgta tcccgagaag caggatttag ctattcccac gcaggactga gtaacagact agctagagac aatgaattaa gggaaaatga caaagaacag ctcaaagcaa tttctacacg agatcctctc tctgaaatca ctgagcagga gaaagatttt ctatggagtc acagacacta ttgtgtaact atccccgaaa ttctacccaa attgcttctg tctgttaaat ggaattctag agatgaagta gcccagatgt attgcttggt aaaagattgg cctccaatca aacctgaaca ggctatggaa cttctggact gtaattaccc agatcctatg gttcgaggtt ttgctgttcg gtgcttggaa aaatatttaa cagatgacaa actttctcag tatttaattc agctagtaca ggtcctaaaa tatgaacaat atttggataa cttgcttgtg agatttttac tgaagaaagc attgactaat caaaggattg ggcacttttt cttttggcat ttaaaatctg agatgcacaa taaaacagtt agccagaggt ttggcctgct tttggagtcc tattgtcgtg catgtgggat gtatttgaag cacctgaata ggcaagtcga ggcaatggaa aagctcatta acttaactga cattctcaaa caggagagga aggatgaaac acaaaaggta cagatgaagt ttttagttga gcaaatgagg cgaccagatt tcatggatgc cctacagggc ttgctgtctc ctctaaaccc tgctcatcaa ctaggaaacc tcaggcttaa agagtgtcga attatgtctt ctgcaaaaag gccactgtgg ttgaattggg agaacccaga catcatgtca gagttactgt ttcagaacaa tgagatcatc tttaaaaatg gggatgattt acggcaagat atgctaacac ttcaaattat tcgtattatg gaaaatatct ggcaaaatca aggtcttgat cttcgaatgt taccttatgg ttgtctgtca atcggtgact gtgtgggact tattgaggtg gtgcgaaatt ctcacactat tatgcaaatt cagtgcaaag gcggcttgaa aggtgcactg cagttcaaca gccacacact acatcagtgg ctcaaagaca agaacaaagg agaaatatat gatgcagcca ttgacctgtt tacacgttca
TABLE 2
SEQ ID Description SEQUENCE
NO:
tgtgctggat actgtgtagc taccttcatt ttgggaattg gagatcgtca caatagtaac atcatggtga aagacgatgg acaactgttt catatagatt ttggacactt tttggatcac aagaagaaaa aatttggtta taaacgagaa cgtgtgccat ttgttttgac acaggatttc ttaatagtga ttagtaaagg agcccaagaa tgcacaaaga caagagaatt tgagaggttt caggagatgt gttacaaggc ttatctagct attcgacagc atgccaatct cttcataaat cttttctcaa tgatgcttgg ctctggaatg ccagaactac aatcttttga tgacattgca tacattcgaa agaccctagc cttagataaa actgagcaag aggctttgga gtatttcatg aaacaaatga atgatgcaca tcatggtggc tggacaacaa aaatggattg gatcttccac acaattaaac agcatgcatt gaactgaaag ataactgaga aaatgaaagc tcactctgga ttccacactg cactgttaat aactctcagc aggcaaagac cgattgcata ggaattgcac aatccatgaa cagcattaga tttacagcaa gaacagaaat aaaatactat ataatttaaa taatgtaaac gcaaacaggg tttgatagca cttaaactag ttcatttcaa aa
56 PIK3CA protein MPPRPSSGEL WGIHLMPPRI LVECLLPNGM IVTLECLREA TLVTIKHELF KEARKYPLHQ sequence LLQDESSYIF VSVTQEAERE EFFDETRRLC DLRLFQPFLK VIEPVGNREE KILNREIGFA
IGMPVCEFDM VKDPEVQDFR RNILNVCKEA VDLRDLNSPH SRAMYVYPPH VESSPELPKH IYNKLDRGQI IWIWVIVSP NNDKQKYTLK INHDCVPEQV IAEAIRKKTR SMLLSSEQLK LCVLEYQGKY ILKVCGCDEY FLEKYPLSQY KYIRSCIMLG RMPNLKMMAK ESLYSQLPMD CFTMPSYSRR ISTATPYM G ETSTKSLWVI NRALRIKILC ATYVNLNIRD IDKIYVRTGI YHGGEPLCDN VNTQRVPCSN PRWNEWLNYD IYIPDLPRAA RLCLSICSVK GRKGAKEEHC PLAWGNINLF DYTDTLVSGK MALNLWPVPH GLEDLLNPIG VTGSNPNKET PCLELEFDWF SSWKFPDMS VIEEHANWSV SREAGFSYSH AGLSNRLARD NELRENDKEQ LKAISTRDPL SEITEQE DF LWSHRHYCVT IPEILPKLLL SVKWNSRDEV AQMYCLVKDW PPIKPEQAME LLDC YPDPM VRGFAVRCLE KYLTDDKLSQ YLIQLVQVLK YEQYLDNLLV RFLLKKALTN QRIGHFFFWH LKSEMHNKTV SQRFGLLLES YCRACGMYLK HLNRQVEAME KLINLTDILK QERKDETQKV QMKFLVEQMR RPDFMDALQG LLSPLNPAHQ LGNLRLKECR IMSSAKRPLW LNWENPDIMS ELLFQNNEII FKNGDDLRQD MLTLQIIRIM ENIWQNQGLD LRMLPYGCLS IGDCVGLIEV VRNSHTIMQI QCKGGLKGAL QFNSHTLHQW LKDKNKGEIY DAAIDLFTRS CAGYCVATFI LGIGDRHNSN IMVKDDGQLF HIDFGHFLDH KKKKFGYKRE RVPFVLTQDF LIVISKGAQE CTKTREFERF QEMCYKAYLA IRQHANLFIN LFSMMLGSGM PELQSFDDIA YIRKTLALDK TEQEALEYFM KQMNDAHHGG WTTKMDWIFH TIKQHALN
57 Exon 9 agtaacagac tagctagaga caatgaatta agggaaaatg acaaagaaca gctcaaagca
atttctacac gagatcctct ctctgaaatc actgagcagg agaaagattt tctatggagt
TABLE 2
Figure imgf000049_0001
cacag
58 Exon 20 gtttcaggag atgtgttaca aggcttatct agctattcga cagcatgcca atctcttcat aaatcttttc tcaatgatgc ttggctctgg aatgccagaa ctacaatctt ttgatgacat tgcatacatt cgaaagaccc tagccttaga taaaactgag caagaggctt tggagtattt catgaaacaa atgaatgatg cacatcatgg tggctggaca acaaaaatgg attggatctt ccacacaatt aaacagcatg cattgaactg aaaagataac tgagaaaatg aaagctcact ctggattcca cactgcactg ttaataactc tcagcaggca aagaccgatt gcataggaat tgcacaatcc atgaacagca ttagaattta cagcaagaac agaaataaaa tactatataa tttaaataat gtaaacgcaa acagggtttg atagcactta aactagttca tttcaaaatt aagctttaga ataatgcgca atttcatgtt atgccttaag tccaaaaagg taaactttga agattgtttg tatctttttt taaaaaacaa aacaaaacaa aaatccccaa aatatataga aatgatggag aaggaaaaag tgatggtttt ttttgtcttg caaatgttct atgttttgaa atgtggacac aacaaaggct gttattgcat taggtgtaag taaactggag tttatgttaa attacattga ttggaaaaga atgaaaattt cttatttttc cattgctgtt caatttatag tttgaagtgg gtttttgact gcttgtttaa tgaagaaaaa tgcttggggt ggaagggact cttgagattt caccagagac tttttctttt taataaatca aaccttttga tgatttgagg ttttatctgc agttttggaa gcagtcacaa atgagacctg ttataaggtg gtattttttt ttttcttctg gacagtattt aaaggatctt attcttattt cccagggaaa ttctgggctc ccacaaagta aaaaaaaaaa aaaatcatag aaaaagaatg agcaggaata gttcttattc cagaattgta cagtattcac cttaagttga ttttttttct ccttctgcaa ttgaactgaa tacatttttc atgcatgttt tccagaaaat agaagtatta atgtta
59 PTEN coding region atgacagcca tcatcaaaga gatcgttagc agaaacaaaa ggagatatca agaggatgga ttcgacttag acttgaccta tatttatcca aacattattg ctatgggatt tcctgcagaa agacttgaag gcgtatacag gaacaatatt gatgatgtag taaggttttt ggattcaaag cataaaaacc attacaagat atacaatctt tgtgctgaaa gacattatga caccgccaaa tttaattgca gagttgcaca atatcctttt gaagaccata acccaccaca gctagaactt atcaaaccct tttgtgaaga tcttgaccaa tggctaagtg aagatgacaa tcatgttgca gcaattcact gtaaagctgg aaagggacga actggtgtaa tgatatgtgc atatttatta catcggggca aatttttaaa ggcacaagag gccctagatt tctatgggga agtaaggacc agagacaaaa agggagtaac tattcccagt cagaggcgct atgtgtatta ttatagctac ctgttaaaga atcatctgga ttatagacca gtggcactgt tgtttcacaa gatgatgttt gaaactattc caatgttcag tggcggaact tgcaatcctc agtttgtggt ctgccagcta
TABLE 2
SEQ ID Description SEQUENCE
NO:
aaggtgaaga tatattcctc caattcagga cccacacgac gggaagacaa gttcatgtac tttgagttcc ctcagccgtt acctgtgtgt ggtgatatca aagtagagtt cttccacaaa cagaacaaga tgctaaaaaa ggacaaaatg tttcactttt gggtaaatac attcttcata ccaggaccag aggaaacctc agaaaaagta gaaaatggaa gtctatgtga tcaagaaatc gatagcattt gcagtataga gcgtgcagat aatgacaagg aatatctagt acttacttta acaaaaaatg atcttgacaa agcaaataaa gacaaagcca accgatactt ttctccaaat tttaaggtga agctgtactt cacaaaaaca gtagaggagc cgtcaaatcc agaggctagc agttcaactt ctgtaacacc agatgttagt gacaatgaac ctgatcatta tagatattct gacaccactg actctgatcc agagaatgaa ccttttgatg aagatcagca tacacaaatt acaaaagtct ga
60 PTEN protein sequence MTAIIKEIVS RNKRRYQEDG FDLDLTYIYP NIIAMGFPAE RLEGVYRNNI DDWRFLDSK
HKNHYKIYNL CAERHYDTAK FNCRVAQYPF EDHNPPQLEL IKPFCEDLDQ WLSEDDNHVA
AIHCKAGKGR TGVMICAYLL HRGKFLKAQE ALDFYGEVRT RDKKGVTIPS QRRYVYYYSY
LLKNHLDYRP VALLFHKMMF ETIPMFSGGT CNPQFWCQL KVKIYSSNSG PTRREDKFMY
FEFPQPLPVC GDIKVEFFHK QNKMLKKDKM FHFWVNTFFI PGPEETSEKV ENGSLCDQEI
DSICSIERAD NDKEYLVLTL TKNDLDKANK DKANRYFSPN FKVKLYFTKT VEEPSNPEAS
SSTSVTPDVS DNEPDHYRYS DTTDSDPENE PFDEDQHTQI TKV
[0117] All publications, patents, patent applications and other documents cited in this application are hereby incorporated by reference in their entireties for all purposes to the same extent as if each individual publication, patent, patent application or other document were individually indicated to be incorporated by reference for all purposes.
[0118] While various specific embodiments have been illustrated and described, it will be appreciated that various changes can be made without departing from the spirit and scope of the invention(s).

Claims

WHAT IS CLAIMED IS:
1. A method of treating a subject having a T EAKR-positive cancer comprising
administering to a subject who has tested negative for a PIK3CA activating mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
2. The method of claim 1, in which the PIK3CA mutation is a substitution mutation.
3. The method of claim 1, in which the PIK3CA mutation is selected from the group consisting of: Kl 1 IN, E542K, E545K, E545G, E545D, Q546K, Q546R, Ml 0431, M1043V, H1047R, H1047L, H1047Y, G1049R and G1049S.
4. The method of claim 1, wherein the subject has tested negative for two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen mutations selected from the group consisting of: Kl 1 IN, E542K, E545K, E545G, E545D, Q546K, Q546R, M1043I, Ml 043V, H1047R, H1047L, H1047Y, G1049R and G1049S.
5. The method of claim 1 , wherein the subject has further tested negative for a PTEN null mutation.
6. The method of any one of the preceding claims, wherein the TWEAKR agonist is an anti-TWEAKR agonist antibody.
7. The method of claim 6, wherein the anti-TWEAKR agonist antibody is a monoclonal antibody.
8. The method of claim 6, wherein the anti-TWEAKR agonist antibody is a humanized antibody.
9. The method of claim 6, wherein the anti-TWEAKR agonist antibody is a monoclonal antibody or anti-TWEAKR antigen binding fragment, comprising the following six CDRs: VH CDR 1 is: XJ Y W M X2 (SEQ ID NO:49);
VH CDR 2is:EIRX3KSX4NYATX5HYAESX6KG (SEQ ID NO:50);
VH CDR3 is: X7X8 A D X9 X10 D Y (SEQ ID NO:51);
VL CDR1 is: Xn A S Q S V S T S X12 Y S Y M X13 (SEQ ID NO:52);
VL CDR2 is: Y A X14 X15 L X16 S (SEQ ID NO:53); and
VL CDR3 is: Q H S W E X,7 P X18 T (SEQ ID NO:54), wherein:
X\ is selected from K, N, R, and S; X2 is selected from N and S; X3 is selected from L and V; X4 is selected from D and N; X5 is T or no amino acid; X6 is selected from A and V; X7 is selected from A, G, T and Y; X8 is selected from F and Y; X9 is selected from A, T and Y; X10 is selected from F and M; Xn is selected from R and K; X12 is selected from S and T; X13 is selected from H and Q; X14 is selected from S and T; X15 is selected from N and K; X16 is selected from E and D; and X17 is selected from I and L; and X18 is selected from W and Y.
The method of claim 9, wherein the anti-TWEAKR agonist antibody is a monoclonal antibody or anti-T EAKR antigen binding fragment, comprising the following six CDRs:
VH CDR 1 is: S Y W M S (SEQ ID NO: 13);
VH CDR 2is:EIRLKSDNYATHYAESVKG (SEQ ID NO: 19);
VH CDR3 is: Y Y A D AMD Y (SEQ ID NO:25);
VL CDRl is: AASQSVSTSSYSYMH (SEQ ID NO:31);
VL CDR2 is: Y A S N L E S (SEQ ID NO:37); and
VL CDR3 is:QHSWEIPYT (SEQ ID NO:43).
The method of claim 6, wherein the anti-TWEAKR agonist antibody comprises a VH region having a sequence selected from SEQ ID NO:3 and SEQ ID NO:l 1 and a VL region having a sequence selected from SEQ ID NO:4 and SEQ ID NO: 12. The method of claim 6, wherein the anti-TWEAKR agonist antibody is PDL192.
The method of claim 1, wherein the TWEAKR positive cancer is selected from the group consisting of: breast cancer, lung cancer, melanoma, ovarian cancer, uterine cancer, colon cancer, renal cancer, pancreatic cancer, cervical cancer, bladder cancer, head and neck cancer, glioblastoma, esophageal cancer, sarcomas, and salivary gland cancer.
A method of treating a subject having a TWEAKR-positive cancer comprising administering to a subject who has tested negative for a PTEN null mutation an amount of a TWEAKR agonist effective to promote a therapeutic benefit.
The method of claim 14, wherein the TWEAKR positive cancer is selected from the group consisting of: breast cancer, lung cancer, melanoma, ovarian cancer, uterine cancer, colon cancer, renal cancer, pancreatic cancer, cervical cancer, bladder cancer, head and neck cancer, glioblastoma, esophageal cancer, sarcomas, and salivary gland cancer.
The method of claim 14, in which the PTEN null mutation is a substitution mutation.
The method of claim 14, in which the PTEN null mutation is a frameshift mutation that introduces a stop codon.
The method of claim 14, in which the PTEN null mutation results in lower expression of PTEN.
The method of claim 14, wherein the subject has further tested negative for a
PIK3CA activating mutation.
The method of any one of the preceding claims, wherein the TWEAKR agonist is an anti-TWEAKR agonist antibody.
21. The method of claim 20, wherein the anti-TWEAKR agonist antibody is a monoclonal antibody.
22. The method of claim 20, wherein the anti-TWEAKR agonist antibody is a humanized antibody.
23. The method of claim 20, wherein the anti-TWEAKR agonist antibody is a
monoclonal antibody or anti-TWEAKR antigen binding fragment, comprising the following six CDRs:
VH CDR 1 is: Xl Y W M X2 (SEQ ID NO:49);
VH CDR 2 is: E I RX3K S X4N YATX5HYAESX6KG (SEQ ID NO:50);
VH CDR3 is: X7X8 A D X9X10D Y (SEQ ID NO:51);
VL CDRl is: X„ A S Q S V S T S X12 Y S Y M X13 (SEQ ID NO:52);
VL CDR2 is: Y A X14 X15 L X16 S (SEQ ID NO:53); and
VL CDR3 is: Q H S W E X17P X]8T (SEQ ID NO:54), wherein:
Xi is selected from K, N, R, and S; X2 is selected from N and S; X3 is selected from L and V; X4 is selected from D and N; X5 is T or no amino acid; X6 is selected from A and V; X7 is selected from A, G, T and Y; X8 is selected from F and Y; X9 is selected from A, T and Y; X10 is selected from F and M; Xn is selected from R and K; X12 is selected from S and T; X13 is selected from H and Q; Xn is selected from S and T; Xj5 is selected from N and K; X16 is selected from E and D; and X17 is selected from I and L; and X18 is selected from W and Y.
24. The method of claim 23, wherein the anti-TWEAKR agonist antibody is a
monoclonal antibody or anti-TWEAKR antigen binding fragment, comprising the following six CDRs:
VH CDR 1 is: S Y W M S (SEQ ID NO: 13);
VH CDR 2is:EIRLKSDNYATHYAESVKG (SEQ ID NO:19); VH CDR3 is: YYAD AMD Y (SEQ ID NO:25);
VL CDRl is: AASQSVSTSSYSYMH (SEQ ID NO:31);
VL CDR2 is: Y A S N L E S (SEQ ID NO:37); and
VL CDR3 is:QHSWEIPYT (SEQ ID NO:43).
25. The method of claim 20, wherein the anti-TWEAKR agonist antibody which
comprises a VH region having a sequence selected from SEQ ID NO:3 and SEQ ID NO:l 1 and a VL region having a sequence selected from SEQ ID NO:4 and SEQ ID NO:12.
26. The method of claim 20, wherein the anti-TWEAKR agonist antibody is PDL192.
PCT/US2013/042409 2012-05-24 2013-05-23 Biomarkers for predicting response to tweak receptor (tweakr) agonist therapy WO2013177386A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261651252P 2012-05-24 2012-05-24
US61/651,252 2012-05-24

Publications (1)

Publication Number Publication Date
WO2013177386A1 true WO2013177386A1 (en) 2013-11-28

Family

ID=48577920

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/042409 WO2013177386A1 (en) 2012-05-24 2013-05-23 Biomarkers for predicting response to tweak receptor (tweakr) agonist therapy

Country Status (1)

Country Link
WO (1) WO2013177386A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10556007B2 (en) 2015-02-27 2020-02-11 Ucl Business Ltd Antibody which binds Lrg1 and methods of use

Citations (48)

* 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
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
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 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
US5177059A (en) 1989-11-15 1993-01-05 Sandoz Ltd. Polymyxin B conjugates
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
WO1998016654A1 (en) 1996-10-11 1998-04-23 Japan Tobacco, Inc. Production of a multimeric protein by cell fusion method
WO1998024893A2 (en) 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
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
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
WO1998046645A2 (en) 1997-04-14 1998-10-22 Micromet Gesellschaft Für Biomedizinische Forschung Mbh Method for the production of antihuman antigen receptors and uses thereof
WO1998050433A2 (en) 1997-05-05 1998-11-12 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US5876969A (en) 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
WO2005123780A2 (en) 2004-04-09 2005-12-29 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
US6994857B2 (en) 2000-04-12 2006-02-07 Human Genome Sciences, Inc. Albumin fusion proteins
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US7189690B2 (en) 2001-12-21 2007-03-13 Human Genome Sciences, Inc. Albumin fusion proteins
US7238499B2 (en) 2001-12-21 2007-07-03 Immunex Corporation Trimeric macrophage scavenger receptor fusion proteins and encoding nucleic acids
WO2009020933A2 (en) * 2007-08-03 2009-02-12 Facet Biotech Corporation Therapeutic use of anti-tweak receptor antibodies
US20090192110A1 (en) 2007-12-03 2009-07-30 Santaris Pharma A/S Rna antagonist compounds for the modulation of pik3ca expression
US20090208505A1 (en) 2004-03-02 2009-08-20 The Johns Hopkins University Mutations of the pik3ca gene in human cancers
US7587286B2 (en) 2003-03-31 2009-09-08 Xencor, Inc. Methods for rational pegylation of proteins
WO2009140177A2 (en) * 2008-05-15 2009-11-19 Biogen Idec Ma Inc. Anti-fn14 antibodies and uses thereof
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
US7745485B2 (en) 2000-05-30 2010-06-29 Indiana University Research And Technology Corporation Compositions and methods for identifying agents which modulate PTEN function and PI-3 kinase pathways
US20100303809A1 (en) 2009-06-01 2010-12-02 Targeted Molecular Diagnostics, Llc Methods for the detection and quantitation of pten
US20110060605A1 (en) 2009-09-09 2011-03-10 Quintiles Transnational Corp. Methods for predicting responsiveness of a disease or disorder to a receptor tyrosine kinase inhibitor by analysis of mutations in pik3ca
US20110150868A1 (en) 2003-11-05 2011-06-23 Board Of Regents Of The University Of Texas System Diagnostic and therapeutic methods and compositions involving pten and breast cancer
US8039592B2 (en) 2002-09-27 2011-10-18 Xencor, Inc. Optimized Fc variants and methods for their generation

Patent Citations (55)

* 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
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
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5693761A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Polynucleotides encoding improved humanized immunoglobulins
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
US5177059A (en) 1989-11-15 1993-01-05 Sandoz Ltd. Polymyxin B conjugates
WO1991009967A1 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
US5939598A (en) 1990-01-12 1999-08-17 Abgenix, Inc. Method of making transgenic mice lacking endogenous heavy chains
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
WO1992001047A1 (en) 1990-07-10 1992-01-23 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5876969A (en) 1992-01-31 1999-03-02 Fleer; Reinhard Fusion polypeptides comprising human serum albumin, nucleic acids encoding same, and recombinant expression thereof
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1998016654A1 (en) 1996-10-11 1998-04-23 Japan Tobacco, Inc. Production of a multimeric protein by cell fusion method
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
WO1998024893A2 (en) 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
WO1998046645A2 (en) 1997-04-14 1998-10-22 Micromet Gesellschaft Für Biomedizinische Forschung Mbh Method for the production of antihuman antigen receptors and uses thereof
WO1998050433A2 (en) 1997-05-05 1998-11-12 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US6994857B2 (en) 2000-04-12 2006-02-07 Human Genome Sciences, Inc. Albumin fusion proteins
US7745485B2 (en) 2000-05-30 2010-06-29 Indiana University Research And Technology Corporation Compositions and methods for identifying agents which modulate PTEN function and PI-3 kinase pathways
US7189690B2 (en) 2001-12-21 2007-03-13 Human Genome Sciences, Inc. Albumin fusion proteins
US7238499B2 (en) 2001-12-21 2007-07-03 Immunex Corporation Trimeric macrophage scavenger receptor fusion proteins and encoding nucleic acids
US8039592B2 (en) 2002-09-27 2011-10-18 Xencor, Inc. Optimized Fc variants and methods for their generation
US7587286B2 (en) 2003-03-31 2009-09-08 Xencor, Inc. Methods for rational pegylation of proteins
US20110150868A1 (en) 2003-11-05 2011-06-23 Board Of Regents Of The University Of Texas System Diagnostic and therapeutic methods and compositions involving pten and breast cancer
US20090208505A1 (en) 2004-03-02 2009-08-20 The Johns Hopkins University Mutations of the pik3ca gene in human cancers
WO2005123780A2 (en) 2004-04-09 2005-12-29 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
WO2009020933A2 (en) * 2007-08-03 2009-02-12 Facet Biotech Corporation Therapeutic use of anti-tweak receptor antibodies
US20090074762A1 (en) 2007-08-03 2009-03-19 Pdl Biopharma, Inc. Therapeutic use of anti-tweak receptor antibodies
US20090192110A1 (en) 2007-12-03 2009-07-30 Santaris Pharma A/S Rna antagonist compounds for the modulation of pik3ca expression
WO2009140177A2 (en) * 2008-05-15 2009-11-19 Biogen Idec Ma Inc. Anti-fn14 antibodies and uses thereof
US20100303809A1 (en) 2009-06-01 2010-12-02 Targeted Molecular Diagnostics, Llc Methods for the detection and quantitation of pten
US20110060605A1 (en) 2009-09-09 2011-03-10 Quintiles Transnational Corp. Methods for predicting responsiveness of a disease or disorder to a receptor tyrosine kinase inhibitor by analysis of mutations in pik3ca

Non-Patent Citations (67)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Science 15th Ed.,", 1980, MACK PUBLISHING COMPANY
ABUCHOWSKI ET AL., J BIOL CHEM, vol. 252, 1977, pages 3582 - 3586
AUSUBEL ET AL.,: "Current Protocols in Molecular Biology", 1997, JOHN WILEY & SONS, INC.
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1998, WILEY
BACHMAN ET AL., CANCER BIOL. THER., vol. 3, pages 772
BADER ET AL., NAT. REV. CANCER, vol. 5, 2005, pages 921 - 929
BRODERICK, CANCER RES, vol. 64, pages 5048
CAMPBELL ET AL., CANCER RES., vol. 64, pages 7678
CANFIELD; MORRISON, J. EXP. MED., vol. 173, 1991, pages 1483 - 1491
CULP ET AL., CLIN. CANCER RES., vol. 16, no. 2, 2010
CULP ET AL., CLIN. CANCER RES., vol. 16, no. 2, 2010, pages 497 - 508
DAI ET AL., CANCER LETT., vol. 283, no. 2, 2009, pages 159 - 167
DE PLATER L ET AL: "PDL192, a humanized anti-Tweak receptor monoclonal antibody, mediates antitumor effects in primary human breast carcinoma xenografts", EJC SUPPLEMENTS, vol. 8, no. 7, November 2010 (2010-11-01), & 22ND EORTC-NCI-AACR SYMPOSIUM ON MOLECULAR TARGETS AND CANCER THERAPEUTICS; BERLIN, GERMANY; NOVEMBER 16 -19, 2010, pages 37, XP002711462, ISSN: 1359-6349 *
DEANE; FRUMAN, ANNU. REV. IMMUNOL., vol. 22, pages 563
FENG ET AL., AM. J. PATHOL., vol. 156, 2000, pages 1253 - 1261
FRANK, J. MOL. BIO., vol. 308, 2001, pages 1081 - 1089
GEORGESCU ET AL., PNAS, vol. 96, no. 18, 1999, pages 10182 - 10187
GILLIES ET AL., J. IMMUNOL. METHODS, vol. 125, 1985, pages 191 - 202
GRAVESTEIN ET AL., IMMUNOLOGY, vol. 10, 1998, pages 423 - 434
HAN ET AL., BIOCH. BIOPHYS. RES. COMMUNICATIONS, vol. 305, 2003, pages 789 - 796
HAN ET AL., CANCER RES., vol. 62, 2002, pages 2890 - 2896
HARBURY, NATURE, vol. 371, no. 6492, 1994, pages 80 - 83
HARTMANN ET AL., ACTA NEUROPATHOL., vol. 109, pages 639
HYLANDER B L ET AL: "64 PDL192, a humanized antibody to TweakR exhibits potent anti-tumor activity in non-small cell lung cancer models", EUROPEAN JOURNAL OF CANCER. SUPPLEMENT, PERGAMON, OXFORD, GB, vol. 8, no. 7, 1 November 2010 (2010-11-01), pages 29, XP027497752, ISSN: 1359-6349, [retrieved on 20101101], DOI: 10.1016/S1359-6349(10)71769-9 *
JESPERS, BIOTECHNOLOGY, vol. 12, 1988, pages 899 - 903
JHAWER, CANCER RES, vol. 68, 2008, pages 1953 - 1961
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1987, NATIONAL INSTITUTE OF HEALTH
KANG ET AL., PNAS, vol. 102, 2005, pages 802 - 807
KANG, PNAS, vol. 102, 2005, pages 802
LAZEBNIK ET AL., NATURE, vol. 371, 1994, pages 346
LEE ET AL., ONCOGENE, vol. 24, pages 1477
LEVIN ET AL., CLIN. CANCER RES., vol. 11, pages 2875
LI ET AL., BMC CANCER, vol. 5, pages 29
LIANG XIAOHUI ET AL: "Mutational hotspot in exon 20 of PIK3CA in breast cancer among Singapore Chinese.", CANCER BIOLOGY & THERAPY MAY 2006, vol. 5, no. 5, May 2006 (2006-05-01), pages 544 - 548, XP002711464, ISSN: 1538-4047 *
LUND ET AL., J. IMMUNOL., vol. 147, 1991, pages 2657 - 2662
MEHVAR ET AL., J PHARM SCI, vol. 81, 1992, pages 908 - 912
MORRISON, SCIENCE, vol. 229, no. 4719, 1985, pages 1202 - 7
NAKAYAMA ET AL., J. EXP. MED., vol. 192, 2000, pages 1373
NAKAYAMA ET AL., J. IMMUNOLOGY, vol. 170, 2003, pages 341 - 348
OI ET AL., BIOTECHNIQUES, vol. 4, 1986, pages 214 - 221
OKKENHAUG ET AL., SCI. STKE, vol. 65, pages PE1
PADLAN, MOL. IMMUNOL., vol. 28, 1991, pages 489 - 498
PRESTA ET AL., BIOCHEM SOC. TRANS., vol. 30, 2002, pages 487 - 490
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 7
ROGUSKA ET AL., PROC. NATL. ACAD. SCI., vol. 91, 1994, pages 969 - 973
SAAL ET AL., CANCER RES., vol. 65, pages 2554
SAMBROOK ET AL.,: "Molecular Cloning: A Laboratory Manual(2nd Ed.)", vol. 1-3, 1989, COLD SPRING HARBOR LABORATORY
SAMBROOK: "Molecular Cloning A Laboratory Manual", 1990, COLD SPRING HARBOR LABORATORY PRESS
SAMUELS ET AL., SCIENCE, vol. 304, 2004, pages 554
SAMUELS ET AL., SCIENCE, vol. 304, pages 443
SAMUELS ET AL., SCIENCE, vol. 304, pages 554
SHIELDS ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 26733 - 26740
SHIELDS, J. BIOL. CHEM., vol. 276, 2001, pages 6591 - 6604
SHINKAWA ET AL., J. BIOL. CHEM., vol. 278, 2003, pages 3466
STUDNICKA ET AL., PROT. ENG., vol. 7, 1994, pages 805 - 814
TIJSSEN: "Laboratory Techniques in Biochemistry and Molecular Biology: Hybridization With Nucleic Acid Probes, Part I. Theory and Nucleic Acid Preparation", 1993, ELSEVIER
TRAN ET AL., CANCER RES., vol. 66, no. 19, 2006, pages 9535 - 9542
WAHL ET AL., J. NUCL. MED., vol. 24, 1983, pages 316
WALSH ET AL., ANTIMICROB AGENTS CHEMOTHER, vol. 47, no. 2, 2003, pages 554 - 558
WANG ET AL., HUM. MUTAT., vol. 25, pages 322
WANG ET AL., ONCOGENE, vol. 25, 2006, pages 3346 - 3356
WANG ET AL., PNAS, vol. 106, no. 15, 2009, pages 6279 - 6284
WHYTE ET AL: "Correlation of PIK3Ca mutations with gene expression and drug sensitivity in NCI-60 cell lines", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS INC. ORLANDO, FL, US, vol. 340, no. 2, 10 February 2006 (2006-02-10), pages 469 - 475, XP002711465, ISSN: 0006-291X, DOI: 10.1016/J.BBRC.2005.12.025 *
WINKLES, NATURE REVIEWS DRUG DISCOVERY, vol. 7, 2008, pages 411 - 425
YANG ET AL., J. VIROLOGY, vol. 76, no. 9, 2002, pages 4634 - 4642
ZHANG ET AL., BREAST CANCER RES. TREAT., vol. 112, 2008, pages 217 - 227
ZHOU HONG ET AL: "Development and characterization of a potent immunoconjugate targeting the Fn14 receptor on solid tumor cells.", MOLECULAR CANCER THERAPEUTICS JUL 2011, vol. 10, no. 7, July 2011 (2011-07-01), pages 1276 - 1288, XP002711463, ISSN: 1538-8514 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10556007B2 (en) 2015-02-27 2020-02-11 Ucl Business Ltd Antibody which binds Lrg1 and methods of use

Similar Documents

Publication Publication Date Title
CN111213059B (en) Diagnostic and therapeutic methods for cancer
TWI775781B (en) Therapeutic and diagnostic methods for cancer
ES2850428T3 (en) Cancer monitoring and treatment procedures
EP2182982B1 (en) Therapeutic use of anti-tweak receptor antibodies
KR102500659B1 (en) Therapeutic and diagnostic methods for cancer
US10202454B2 (en) Anti-PD-L1 monoclonal antibodies and fragments thereof
KR20190134631A (en) How to diagnose and treat cancer
KR20180008449A (en) Treatment and Diagnosis Methods for Cancer
CN111699196A (en) Antibodies to LILRB2
JP2019515670A (en) Methods for monitoring and treating cancer
KR20210079311A (en) Diagnosis and treatment methods for sarcoma renal cancer
US8962808B2 (en) EGFR-related polypeptides and methods of use
WO2013177386A1 (en) Biomarkers for predicting response to tweak receptor (tweakr) agonist therapy
CN116568824A (en) Method for diagnosing and treating lymphoma
AU2008284025B2 (en) Therapeutic use of anti-tweak receptor antibodies
WO2013166290A1 (en) P21 biomarker assay

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: 13727751

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13727751

Country of ref document: EP

Kind code of ref document: A1