EP0787187A1 - Her4-menslicher rezeptor tyrosine kinase von der epidermalen wachstumsfaktor rezeptor familie - Google Patents

Her4-menslicher rezeptor tyrosine kinase von der epidermalen wachstumsfaktor rezeptor familie

Info

Publication number
EP0787187A1
EP0787187A1 EP95937555A EP95937555A EP0787187A1 EP 0787187 A1 EP0787187 A1 EP 0787187A1 EP 95937555 A EP95937555 A EP 95937555A EP 95937555 A EP95937555 A EP 95937555A EP 0787187 A1 EP0787187 A1 EP 0787187A1
Authority
EP
European Patent Office
Prior art keywords
her4
leu
pro
ser
val
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP95937555A
Other languages
English (en)
French (fr)
Inventor
Gregory D. Plowman
Mohammed Shoyab
Clay Siegall
Jean-Michel Culouscou
Ingegerd Hellstrom
Karl E. Hellstrom
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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 Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Publication of EP0787187A1 publication Critical patent/EP0787187A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6425Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/4756Neuregulins, i.e. p185erbB2 ligands, glial growth factor, heregulin, ARIA, neu differentiation factor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/485Epidermal growth factor [EGF] (urogastrone)
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/4756Neuregulins, i.e. p185erbB2 ligands, glial growth factor, heregulin, ARIA, neu differentiation factor

Definitions

  • the present invention is generally directed to a novel receptor tyrosine kinase related to the epidermal growth factor receptor, termed HER4/pl80"* lM ("HER4"), and to novel diagnostic and therapeutic compositions comprising HER4-derived or HER4-related biological components.
  • HER4/pl80"* lM HER4
  • novel diagnostic and therapeutic compositions comprising HER4-derived or HER4-related biological components.
  • the invention is based in part upon applicants discovery of human HER4, its complete nucleotide coding sequence, and functional properties of the HER4 receptor protein.
  • the invention is directed to HER4 biologies comprising, for example, polynucleotide molecules encoding HER4, HER4 polypeptides, anti-HER4 antibodies which recognize epitopes of HER4 polypeptides, ligands'which interact with HER4, and diagnostic and therapeutic compositions and methods based fundamentally upon such molecules.
  • HER4 HER4 biologies comprising, for example, polynucleotide molecules encoding HER4, HER4 polypeptides, anti-HER4 antibodies which recognize epitopes of HER4 polypeptides, ligands'which interact with HER4, and diagnostic and therapeutic compositions and methods based fundamentally upon such molecules.
  • the present invention provides a framework upon which effective biological therapies may be designed.
  • the invention is hereinafter described in detail, in part by way of experimental examples specifically illustrating various aspects of the invention and particular embodiments thereof.
  • RTKs receptor tyrosine kinases
  • EGF epidermal growth factor
  • PDGF platelet-derived growth factor
  • NGF neurotrophins
  • FGF fibroblast growth factor
  • ligands for several previously-characterized receptors have been identified, including ligands for c-kit (steel factor) , met (hepatocyte growth factor) , trk (nerve growth factor) (see, respectively, Zsebo et al . , 1990, Cell 63:195-201; Bottardo et al . , 1991, Science 251:802-04; Kaplan et al . , 1991, Nature 350:158-160).
  • the soluble factor NDF, or heregulin- alpha (HRG- ⁇ ) has been identified as the ligand for HER2, a receptor which is highly related to HER4 (Hen et al . , 1992, Cell 69:559-72; Holmes et al . , 1992, Science 256:1205-10).
  • the heregulins are a family of molecules that were first isolated as specific ligands for HER2 (Wen, et al . , 1992, Cell. 69:559-572; Holmes et al . , 1992, Science 256:1205-1210; Falls et al . , 1993, Cell 72:801-815; and Marchionni et al . , 1993, Nature 362:312-318).
  • a rat homologue was termed Neu differentiation factor (NDF) based on its ability to induce differentiation of breast cancer cells through its interaction with HER2/Neu (Wen et al., supra) .
  • NDF Neu differentiation factor
  • Heregulin also appears to play an important role in development and maintenance of the nervous system based on its abundant expression in cells of neuronal
  • GGF glial growth factor
  • HER2-neutral!zing antibodies fail to block heregulin activation of human breast cancer cells. Heregulin only activates tyrosine phosphorylation of HER2 in cells of breast, colon, and neuronal origin, and not in fibroblasts or ovarian cell lines that overexpress recombinant HER2 (Peles et al . , 1993, EMBO J. 12:961-971).
  • EGFR EGF receptor
  • HER2/pl85- rt * J HER3/pl60 a * M1
  • EGFR-family members have also been determined from other organisms including: the drosophila EGFR ("DER”: Livneh et al . , 1985, Cell 40:599-607), nematode EGFR ( M let-23": Aroian et al . , 1990, Nature 348:693-698), chicken EGFR ( M CER”: Lax et al . , 1988, Mol. Cell. Biol. 8:1970-1978), rat EGFR (Petch et al . , 1990, Mol. Cell. Biol. 10:2973-2982), rat HER2/Neu (Bargmann et al . , 1986, Nature.
  • DER Livneh et al . , 1985, Cell 40:599-607
  • nematode EGFR M let-23”: Aroian et al . , 1990, Nature 348:693-698
  • chicken EGFR M CER”:
  • HER2 has been associated with a wide variety of human malignancies, particularly breast and ovarian carcinomas, for which a strong correlation between HER2 overexpression and poor clinical prognosis and/or increased relapse probability have been established (see, for example.
  • EGF transforming growth factor-alpha
  • AR amphiregulin
  • HB-EGF heparin-binding EGF
  • VVF vaccinia virus growth factor
  • FBMP-2SS4C.1 Recently, several groups have reported the identification of specific ligands for HER2. Some of these ligands, such as gp30 (Lupu et al . , 1990, Science 249:1552-55; Bacus et al., 1992, Cell Growth and Differentiation 3:401-11) interact with both EGFR and HER2, while others are reported to bind specifically to HER2 (Wen et al . , 1992, Cell 69:559- 72; Peles et al. , 1992, Cell 69:205-16; Holmes et al. , 1992, Science 256:1205-10; Lupu et al . , 1992, Proc. Natl. Acad. Sci.
  • NDF neu differentiation factor
  • Both of these proteins are similar size (44- 45 kDa) , increase tyrosine phosphorylation of HER2 in MDA-MB-453 cells and not the EGF-receptor, and have been reported to bind to HER2 in cross-linking studies on human breast cancer cells.
  • NDF has been shown to induce differentiation of human mammary tumor cells to milk-producing, growth-arrested cells, whereas the heregulin family have been reported to stimulate proliferation of cultured human breast cancers cell monolayers.
  • F-MF-2S94C.1 existence of other receptors for heregulin responsible for the activation of HER2.
  • Such cross-activation between members of the receptor tyrosine kinase family has been already reported and is believed to arise from a ligand induced receptor heterodimerization event (Wada et al . , 1990, Cell 61:1339-1347).
  • HER3 binds heregulin (Carraway et al . , 1994, J. Biol. Chem. 269:14303-14306), and in fact, this receptor seems to be involved in the heregulin-mediated tyrosine kinase activation of HER2 (Carraway et al . , supra ; Sliwkowski et al . , 1994, J. Biol. Chem. 269:14661- 14665) .
  • receptor polypeptides transduce regulatory signals in response to ligand binding
  • important components of the process have been uncovered, including the understanding that phosphorylation of and by cell surface receptors hold fundamental roles in signal transduction.
  • the intracellular phenomena of receptor dimerization and receptor crosstalk function as primary components of the circuit through which ligand binding triggers a resulting cellular response.
  • Ligand binding to transmembrane receptor tyrosine kinases induces receptor dimerization, leading to activation of kinase function through the interaction of adjacent cytoplas ic domains.
  • Receptor crosstalk refers to intracellular communication between two or more proximate receptor molecules mediated by, for example, activation of one receptor through a mechanism involving the kinase activity of the other.
  • HER4 is the fourth member of the EGFR-family of receptor tyrosine kinases and is likely to be involved not only in regulating normal cellular function but also in the loss of normal growth control associated with certain human cancers.
  • HER4 appears to be closely connected with certain carcinomas of epithelial origin, such as adenocarcino a of the breast.
  • carcinomas of epithelial origin such as adenocarcino a of the breast.
  • the invention includes embodiments directly involving the production and use of HER4 polynucleotide molecules.
  • the invention provides HER4 polypeptides, such as the prototype HER4 polypeptide disclosed and characterized in the sections which follow. Polypeptides sharing nearly equivalent structural characteristics with the prototype HER4 molecule are also included within the scope of this invention.
  • the invention includes polypeptides which interact with HER4 expressed on the surface of certain cells thereby
  • the invention is also directed to anti-HER4 antibodies, which have a variety of uses including but not limited to their use as components of novel biological approaches to human cancer diagnosis and therapy provided by the invention.
  • the invention also relates to the identification of HER4 ligands and methods for their purification.
  • the invention also relates to the discovery of an apparent functional relationship between HER4 and HER2, and the therapeutic aspects of the invention include those which are based on applicants' preliminary understanding of this relationship.
  • Applicants' data strongly suggests that HER4 interacts with HER2 either by heterodimer formation or receptor crosstalk, and that such interaction appears to be one mechanism by which the HER4 receptor mediates effects on cell behavior.
  • the reciprocal consequence is that HER2 activation is in some circumstances mediated through HER4.
  • heregulin induces phosphorylation of HER2 in cells expressing HER2 and HER4.
  • Heregulin does not directly stimulate HER2 but acts by stimulating tyrosine phosphorylation of HER4.
  • HER4 as a primary component of the heregulin signal transduction pathway opens a number of novel approaches to the diagnosis and treatment of human cancers in which the aberrant expression and/or function of heregulin and/or HER4 are involved.
  • the therapeutic aspects of this invention thus include mediating a ligand's affect on HER4 and HER2 through antagonists, agonists or antibodies to HER4 ligands or HER4 receptor itself.
  • the invention also relates to chimeric proteins that specifically target and kill HER4 expressing tumor cells, polynucleotides encoding such chimeric proteins, and methods cf using both in the therapeutic treatment of cancer and other human malignancies.
  • Applicants' data demonstrate that such recombinant chimeric proteins specifically bind to the HER4 receptor and are cytotoxic against tumor cells that express HER4 on their surface.
  • the bifunctional retention of both the specificity of the cell-binding portion of the molecule and the cytotoxic potential of the toxin portion makes for a very potent and targeted reagent.
  • the invention further relates to a method allowing determination of the cytotoxic activity of HER4 directed cytotoxic substances on cancer cells, thereby providing a powerful diagnostic tool; this will be of particular interest for prognosis of the effectiveness of these substances on an individual malignancy prior their therapeutic use.
  • FIG. IA and IB Nucleotide sequence [SEQ ID No:l] and deduced amino acid sequence of HER4 of the coding sequence from position 34 to 3961 (1308 amino acid residues) [SEQ ID No:2]. Nucleotides are numbered on the left, and amino acids are numbered above the sequence.
  • FIG. 2A and 2B Nucleotide sequence [SEQ ID No:3] and deduced amino acid sequence ([SEQ ID No:4] of cDNAs encoding HER4 with alternate 3' end and without autophosphorylation domain. This sequence is identical with that of HER4 shown in FIG. IA and IB up to nucleotide 3168, where the sequence diverges and the open reading frame stops after 13 amino acids.
  • FIG. 3 Nucleotide sequence [SEQ ID No:5] and deduced amino acid sequence [SEQ ID No:6] of cDNA encoding HER4 with a N-terminal truncation. This sequence contains the 3'-portion of the HER4 sequence where nucleotide position 156 of the truncated sequence aligns with position 2335 of the complete HER4 sequence shown in FIG. IA and IB (just downstream from the region encoding the ATP-binding site of the HER4 kinase) .
  • the first 155 nucleotides of the truncated sequence are unique from HER4 and may represent the 5'-untranslated region of a transcript derived from a cryptic promoter within an intron of the HER4 gene. (Section 6.2.2., infra) .
  • FIGS. 4 and 5 The deduced amino acid sequence of two variant forms of human HER4 aligned with the full length HER4 receptor as represented in FIG. IA and IB. Sequences are displayed using the single-letter code and are numbered on the right with the complete HER4 sequence on top and the variant sequences below. Identical residues are indicated by a colon between the aligned residues.
  • FIG. 4. HER4 with alternate 3'-end, lacking an autophosphorylation domain [SEQ ID No. 4]. This sequence is identical with that of HER4, shown in FIG. IA and IB, up to amino acid 1045, where the sequence diverges and continues for 13 amino acids before reaching a stop codon.
  • FIG. 5. HER4 with N-terminal truncation [SEQ ID No. 6]. This sequence is identical to the 3'-portion of the HER4 shown in FIG. IA and IB beginning at amino acid 768. (Section 6.2.2., infra).
  • FIG. 6A and 6B Deduced amino acid sequence of human HER4 and alignment with other human EGFR-family
  • PENP-2S946.1 members (EGFR [SEQ ID No:7]; HER2 [SEQ ID No:8]; HER3 [SEQ ID No:9]). Sequences are displayed using the single-letter code and are numbered on the left. Identical residues are denoted with dots, gaps are introduced for optimal alignment, cysteine residues are marked with an asterisk, and N-linked glycosylation sites are denoted with a plus (+) . Potential protein kinase C phosphorylation sites are indicated by arrows (HER4 amino acid positions 679, 685, and 699).
  • the predicted ATP-binding site is shown with 4 circled crosses, C-terminal tyrosines are denoted with open triangles, and tyrosines in HER4 that are conserved with the major autophosphorylation sites in the EGFR are indicated with black triangles.
  • the predicted extracellular domain extends from the boundary of the signal sequence marked by an arrow at position 25, to the hydrophobic transmembrane domain which is overlined from amino acid positions 650 through 675.
  • Various subdomains are labeled on the right: I, II, III, and IV - extracellular subdomains (domains II and IV are cysteine-rich) ; TM ⁇ transmembrane domain; TK « tyrosine kinase domain. Domains I, III, TK are boxed.
  • FIG. 7 Hydropathy profile of HER4, aligned with a comparison of protein domains for HER4 (1308 amino acids) , EGFR (1210 amino acids) , HER2 (1255 amino acids), and HER3 (1342 amino acids).
  • the signal peptide is represented by a stippled box, the cysteine-rich extracellular subdomains are hatched, the transmembrane domain is filled, and the cytoplas ic tyrosine kinase domain is stippled. The percent amino acid sequence identities between HER4 and other EGFR-family members are indicated.
  • Sig signal peptide; I, II, III, and IV, extracellular domains; TM, transmembrane domain; JM, juxtamembrane
  • P_HP-2S94C.l domain Cain, calcium influx and internalization domain; 3'UTR, 3' untranslated region.
  • FIG. 8 Northern blot analysis from human tissues hybridized to HER4 probes. RNA size markers (in kilobases) are shown on the left. Lanes 1 through 8 represent 2 ⁇ g of poly(A)+ mRNA from pancreas, kidney, skeletal muscle, liver, lung, placenta, brain, and heart, respectively.
  • FIG. 8 (Panel 1) , Northern blot analysis of mRNA from human tissues hybridized to HER4 probes from the 3'-autophosphorylation domain;
  • FIG. 8 (Panel 2), Northern blot analysis from human tissues hybridized to HER4 probes from the 5'- extracellular domain (see Section 6.2.3., infra) .
  • FIG. 9 Immunoblot analysis of recombinant HER4 stably expressed in CHO-KI cells, according to procedure outlined in Section 7.1.3, infra .
  • Membrane preparations from CHO-KI cells expressing recombinant HER4 were separated on 7% SDS-polyacrylamide gels and transferred to nitrocellulose.
  • FIG. 9 (Panel 1) , blots were hybridized with a monoclonal antibody to the C-terminus of HER2 (Ab3, Oncogene Science, Uniondale, NY) that cross-reacts with HER4.
  • FIG. 9 Panel 1
  • blots were hybridized with a monoclonal antibody to the C-terminus of HER2 (Ab3, Oncogene Science, Uniondale, NY) that cross-reacts with HER4.
  • FIG. 9 (Panel 1) , blots were hybridized with a monoclonal antibody to the C-terminus of HER2 (Ab3, Oncogen
  • FIG. 10 Specific activation of HER4 tyrosine kinase by a breast cancer differentiation factor (see Section 8., infra) .
  • FIG. 10 (Panell) , CHO/HER4 #3 cells; FIG. 10 (Panel 2), CHO/HER2 cells; FIG. 10 (Panel 3), NRHER5 cells; and FIG. 10 (Panel 4), 293/HER3 cells.
  • the size (in kilodaltons) of the prestained molecular weight markers are labeled on the left of each panel.
  • FIG. 11 Biological and biochemical properties of the MDA-MB-453-cell differentiation activity * purified from the conditioned media of HepG2 cells (Section 9., infra) .
  • FIG. 11 (Panel 1 and 2) show induction of morphologic differentiation. Conditioned media from HepG2 cells was subjected to ammonium sulfate fractionation, followed by dialysis against PBS. Dilutions of this material were added to MDA-MB- 453 monolayer at the indicated protein concentrations.
  • FIG. 11 Panel 1, control; FIG. 11 (Panel 2), 80 ng per well; FIG. 11 (Panel 3), 2.0 ⁇ g per well; FIG. 11 (Panel 4), Phenyl-5PW column elution profile monitored at 230 nm absorbance; FIG. 11 (Panel 5) , Stimulation of MDA-MB-453 tyrosine autophosphorylation
  • PBMP-2S94C.1 with the following ligand preparations: None (control with no factor added); TGF-o (50 ng/ml); CM (16-fold concentrated HepG2 conditioned medium tested at 2 ⁇ l and 10 ⁇ l per well) ; fraction (phenyl column fractions 13 to 20, 10 ⁇ l per well).
  • FIG. 11 Panel 6
  • FIG. 12 NDF-induced tyrosine phosphorylation.
  • FIG. 12 Panel 1
  • MDA-MB-453 cells lane 1, mock transfected COS cell supernatant; lane 2, NDF transfected COS cell supernatant
  • FIG. 12 Panel 2
  • CH0/HER4 21-2 cells lanes 1 and 2, mock transfected COS cell supernatant; lanes 3 and 4, NDF transfected COS cell supernatant.
  • Tyrosine phosphorylation was determined by the tyrosine kinase stimulation assay described in Section 8.2., infra .
  • FIG. 13 Regional location of the HER4 gene to human chromosome 2 band q33.
  • FIG. 13 Panel 1
  • FIG. 13 Panel 2
  • FIG. 13 Panel 2
  • FIG. 13 Panel 2
  • FIG. 14 Amino acid sequence of HER4-Ig fusion protein [SEQ ID No:10] (Section 5.4., infra).
  • FIG. 15. Recombinant heregulin induces tyrosine phosphorylation of HER4. Tyrosine phosphorylated receptors were detected by Western blotting with an anti-phosphotyrosine Mab. Arrows indicate the HER2 and HER4 proteins.
  • FIG. 15 (Panel 1), Monolayers of MDA-MB453 or FIG. 15 (Panel 2) , CH0/HER4 cells were incubated with media from COS-1 cells transfected with a rat heregulin expression plasmid (HRG) , or with a cDM8 vector control (-) . The media was either applied directly (lx) or after concentrating 20-fold (20x, and vector control) . Solubilized cells were
  • FIG. 15 (Panel 3), Monolayers of CHO/HER2 cells were incubated as above with transfected Cos-1 cell supernatants or with two stimulatory Mabs to HER2 (Mab 28 and 29) . Solubilized cells were immunoprecipitated with anti-HER2 Mab.
  • FIG. 16 Expression of recombinant HER2 and HER4 in human CEM cells. Transfected CEM cells were selected that stably express either HER2, HER4, or both recombinant receptors.
  • recombinant HER2 was detected by immunmoprecipitation of cell lysates with anti-HER2 Mab (Ab-2) and Western blotting with another anti-HER2 Mab (Ab-3).
  • FIG. 16 (Panel 2)
  • Recombinant HER4 was detected by immunoprecipitation of 3S S-labeled cell lysates with HER4-specific rabbit anti-peptide antisera.
  • FIG. 17 Heregulin induces tyrosine phosphorylation in CEM cells expressing HER4.
  • P-HP-2S946.1 1 HER2-specific anti-HER2 Mab (Ab-2) ; in FIG. 17 (Panel 2), HER4-specific Mab (6-4); in FIG. 17 (Panel 3) , in each case tyrosine phosphorylated receptors were detected by Western blotting with anti- phosphotyrosine Mab.
  • the size in kilodaltons of prestained molecular weight markers (BioRad) is shown on the left and arrows indicate the HER2 and HER4 proteins.
  • HRG recombinant rat heregulin.
  • FIG. 18 Covalent cross-linking of iodinated heregulin to HER4. 12S I-heregulin was added to
  • CHO/HER4 or CHO/HER2 cells for 2 h at 4° C. Washed cells were cross-linked with BS 3 , lysed, and the proteins separated using 7% PAGE. Labeled bands were detected on the phosphorimager. Molecular weight markers are shown on the left.
  • FIG. 19 Purification of p45 from HepG2 conditioned media. Column fractions were tested for their potential to induce differentiation of MDA-MB- 453 cells. Active fractions were pooled as indicated by an horizontal bar.
  • FIG. 19 (Panel 1) , Concentrated HepG2 conditioned medium was subjected to 50% ammonium sulfate precipitation. Supernatant resulting from this step was subjected to hydrophobic interaction chromatography using phenyl-Sepharo ⁇ e. Pooled fractions were then loaded on a DEAE-Sepharose column.
  • FIG. 19 Panel 2
  • the DEAE-Sepharose column flow- through was subjected to CM-Sepharo ⁇ e chromatography.
  • FIG. 19 Panel 2
  • FIG. 19 (Panel 3) , Affinity Chromatography of the MDA- MB-453 differentiation factor using heparin-5PW column. Fractions 35-38 eluting around 1.3M NaCl were pooled.
  • FIG. 19 (Panel 4), Size Exclusion chromatography of the differentiation factor. The molecular masses of calibration standards are indicated in kilodaltons.
  • FIG. 20 Aliguots (25 microliter) of the active size exclusion column fractions (30 and 32) were electrophoresed under reducing conditions on a 12.5% polyacryla ide gel. The gel was silver-stained. Molecular masses of Bio-Rad silver stain standards are indicated in kilodaltons.
  • FIG. 21 Stimulation of tyrosine phosphorylation by p45.
  • FIG. 21 Panel 1
  • Size exclusion column fractions were tested on MDA-MB-453 cells for the induction of tyrosine phosphorylation.
  • Cell lysates were then electrophoresed on a 4-15% polyacrylamide gel. After transfer to nitrocellulose, proteins were probed with a phosphotyrosine antibody and phosphoproteins detected by chemiluminescence. The molecular mass of the predominantly phosphorylated protein is indicated.
  • FIG. 21 Panel 2
  • the experiments were performed on cells that had been transfected with expression plasmids for either HER4 (CH0/HER4) or HER2 (CH0/HER2) .
  • FIG. 21 Cell monolayers were incubated in the absence or the presence of p45 (size exclusion column fraction 32, 100 ng/ml). Samples were then processed as indicated in FIG. 21 (Panel 1) except that a 7.5% polyacrylamide gel was used to separate the CH0/HER2 cell lysates.
  • FIG. 21 (Panel 3) , CH0/HER2 cells were incubated in the presence or the absence of N29 monoclonal antibody to the extracellular domain of pl ⁇ S** 4 * 2 .
  • Cell lysates were immunoprecipitated with the Ab-3 monoclonal antibody to pl85 ⁇ rt ⁇ 2 . Precipitated proteins were subjected to SDS-PAGE, and phosphoproteins were detected as indicated under Section 13.4. , supra .
  • FIG. 22 Binding and cross-linking of 15 I-p45 to CHO-KI, CH0-HER2 and CH0/HER4 cells.
  • FIG. 22 (Panel 1) , Scatchard analysis of the binding of 135 I-p45 to CHO/HER4 cells. Increasing concentrations of 125 I-p45
  • FIG. 22 (Panel 2) , Covalent cross-linking. 12S I-p45 was added to the cells in the presence or absence of an excess of unlabeled p45 for 2 h at 4° C. After washing of the cells to remove unbound iodinated material, the cross-linking reagent bis- (sulfosuccinimidyl)-suberate was added to the cells for 45 min. at 4 ⁇ C. Cells were lysed and proteins separated by electrophoresis on a 7.5% polyacrylamide gel. Molecular masses of protein standards are indicated in kilodaltons. A Molecular Dynamics Phospholmager was used to visualize the radioactive species.
  • FIG. 23 Construction of the HAR-TX ⁇ 2 expression plasmid, encoding the hydrophilic leader sequence of amphiregulin (AR) , heregulin ,92, and PE40, under control of the IPTG inducible T7 promoter;
  • FIG. 23 (Panel A) schematic diagram of the expression plasmid pSE 8.4, encoding HAR-TX 02;
  • FIG. 23 (Panel B) amino acid sequence of HAR ⁇ 2 , the ligand portion of HAR-TX ,32, composed of the AR leader sequence and rat heregulin ⁇ 2 [SEQ ID No:40].
  • FIG. 24A and 24B cDNA sequence [SEQ ID No:41] and deduced amino acid sequence [SEQ ID No:42] of the chimera HAR-TX 02, comprising the amphiregulin (AR) leader sequence and the coding sequences of rat heregulin Pseudomonas exotoxin PE40.
  • the linker sequence between the two portions is indicated by a bar above the sequence, the ligand portion is located at the 5' (N-terminal) , the PE40 exotoxin portion is located at the 3' (C-terminal) part of the sequence.
  • PBMP-2594C.1 Nucleotides are numbered on the right side, and amino acids are numbered below the sequence.
  • FIG. 25 Purification of the chimeric HAR-TX 02 protein: shown is a Coomassie brilliant blue stained SDS-PAGE (4-20%) of the different purification steps. Lanes 1 - 5 have been loaded under reducing conditions. Lane 1, MW standards; lane 2 , refolded HAR-TX 02, 2Ox concentrated; lane 3, POROS HS flow- through, 2Ox concentrated; lane 4, POROS HS eluate; lane 5, Source 15S eluate (pure HAR-TX 02, 2 ⁇ g) ; lane 6, 2 ⁇ g HAR-TX 02, loaded under non-reducing conditions.
  • FIG. 26 Membrane-based ELISA binding analysis, performed to determine the binding activity of the purified HAR-TX 02 protein. Binding of HAR-TX 02 (O) and PE40 (•) to membranes prepared from the HER4 expressing human breast carcinoma cell line.
  • FIG. 27 HAR-TX 02 induced tyrosine phosphorylation in transfected CEM cells.
  • the arrow indicates the phosphorylated receptor band, the molecular weight is indicated in kDA.
  • FIG. 28 Cytotoxic effect of HAR-TX 02 on tumor cell lines.
  • FIG. 28 Panel 1
  • FIG. 28 (Panel 1), following 48 hours incubation with HAR-TX 02, the cell killing effect of HAR-TX 02 on the tumor cell lines LNCaP ( ⁇ ) , AU565 (0) , SKBR3 (•), and SK0V3 (D) by quantification of fluorescent calcein cleaved from calcein-AM.
  • FIG. 28 (Panel 2) , Competitive cytotoxicity of HAR-TX 02 with heregulin 02-Ig. LNCaP cells were co-incubated with 50 ng/ml HAR-TX 02 and increasing concentrations (2-5000
  • PBMP-2594..1 ng/ml of either heregulin 02-Ig (D) or L6-Ig ( ⁇ ) .
  • the data represent the mean of triplicate assays.
  • FIG. 29 HAR-TX 02 induced tyrosine phosphorylation in tumor cells expressing HER3 (L2987) or co-expressing HER2 and HER3 (H3396) .
  • Cells were incubated in the presence (+) or in tb ⁇ absence (-) of HAR-TX 02, solubilized, and immunoblotted with the monoclonal anti-phosphotyrosine antibody PY20.
  • Phosphorylated receptors are indicated by an arrow, the molecular weight is indicated in kDa.
  • the present invention is directed to HER4/pl80 ⁇ rtS4 ("HER4"), a closely related yet distinct member of the Human EGF Receptor (HER)/neu subfamily of receptor tyrosine kinases, as well as HER4-encoding polynucleotides (e.g.
  • HER4 polynucleotide coding sequence a HER4 polynucleotide coding sequence
  • HER4 polynucleotide coding sequence recombinant HER4 expression vectors, HER4 analogues and derivatives, anti-HER4 antibodies, HER4 ligands, and diagnostic and therapeutic uses of HER4 polynucleotides, polypeptides, ligands, and" antibodies in the field of human oncology and neurobiology.
  • HER2 has been reported to be associated with a wide variety of human malignancies, thus the understanding of its activation mechanisms as well as the identification of molecules involved are of particular clinical interest.
  • This invention uncovers an apparent functional relationship between the HER4 and HER2 receptors involving HER4- mediated phosphorylation of HER2, potentially via intracellular receptor crosstalk or receptor dimerization.
  • the invention also
  • HER4 ligands capable of inducing cellular differentiation in breast carcinoma cells that appears to involve HER4-mediated phosphorylation of HER2.
  • heregulin mediates biological effects on such cells not directly through HER2, as has been reported (Peles et al . , 1992, Cell 69:205-216), but instead by means of a direct interaction with HER4, and/or through an interaction with a HER2/ HER4 complex.
  • binding of heregulin to HER4 may stimulate HER2 either by heterodimer formation of these two related receptors or by intracellular receptor crosstalk.
  • HER3 has been reported to bind heregulin (see Section 2, supra) .
  • various observations indicate that the heregulin-mediated activation of HER3 varies considerably, depending on the context of expression, suggesting that other cellular components may be involved in the modulation of HER3 activity (reviewed in: Carraway and Cantley, 1994, Cell 78:5-8).
  • the practice of the present invention utilizes standard techniques of molecular biology and molecular cloning, microbiology, immunology, and recombinant DNA known in the art.
  • One aspect of the present invention is directed to HER4 polynucleotides, including recombinant polynucleotides encoding the prototype HER4
  • polypeptide shown in FIG. IA and IB polynucleotides which are related or are complementary thereto, and recombinant vectors and cell lines incorporating such recombinant polynucleotides.
  • recombinant polynucleotide refers to a polynucleotide of genomic, cDNA, synthetic or se isynthetic origin which, by virtue of its origin or manipulation, is not associated with any portion of the polynucleotide with which it is associated in nature, and may be linked to a polynucleotide other than that to which it is linked in nature, and includes single or double stranded polymers of ribonucleotides, deoxyribonucleotides, nucleotide analogs, or combinations thereof.
  • the term also includes various modifications known in the art, including but not limited to radioactive and chemical labels, methylation, caps, internucleotide modifications such as those with charged linkages (e . g. , phosphorothothioates, phosphorodithothioates, etc.) and uncharged linkages ⁇ e.g. , methyl phosphonates, phosphotriester ⁇ , phosphoamidites , carbamites, etc.), as well as those containing pendant moeties, intercalcator ⁇ , chelators, alkylators, etc.
  • charged linkages e . g. , phosphorothothioates, phosphorodithothioates, etc.
  • uncharged linkages ⁇ e.g. , methyl phosphonates, phosphotriester ⁇ , phosphoamidites , carbamites, etc.
  • HER4 polynucleotides are those having a contiguous stretch of about 200 or more nucleotides and sharing at least about 80% homology to a corresponding sequence of nucleotides within the nucleotide sequence disclosed in FIG. IA and IB.
  • HER4 polynucleotides and vectors are provided in example Sections 6 and 7, infra .
  • HER4 polynucleotides may be obtained using a variety of general techniques known in the art, including molecular cloning and chemical synthetic methods. One method by which the molecular cloning of cDNAs encoding the prototype HER4 polypeptide of the
  • P-KF-25946.1 invention (FIG. IA and I B), as well as several HER4 polypeptide variants, is described by way of example in Section 6. , infra. conserveed regions of the sequences of EGFR, HER2, HER3, and Xmrk are used for selection of the degenerate oligonucleotide primers which are then used to isolate HER4. Since many of these sequences have extended regions of amino acid identity, it is difficult to determine if a short PCR fragment represents a unique molecule or merely the species-specific counterpart of EGFR, HER2, or HER3. Often the species differences for one protein are as great as the differences within species for two distinct proteins.
  • fish Xmrk has regions of 47/55 (85%) amino acid identity to human EGFR, suggesting it might be the fish EGFR, however isolation of another clone that has an amino acid sequence identical to Xmrk in this region (57/57) shows a much higher homology to human EGFR in its flanking sequence (92% amino acid homology) thereby suggesting that it, and not Xmrk, is the fish EGFR (Wittbrodt et al . , 1989, Nature 342:415-421).
  • HER4/er_>B4 PCR fragment was indeed a unique gene, and not the murine homolog of EGFR, HER2, or HER3, by isolating genomic fragments corresponding to murine EGFR, erbB2 and eri>B3. Sequence analysis of these clones confirmed that this fragment was a novel member of the EGFR family. Notably a region of the murine clone had a stretch of 60/64 amino acid identity to human HER2, but comparison with the amino acid and DNA sequences of the other EGFR homologs from the same species (mouse) firmly established it encoded a novel transcript.
  • HER4 polynucleotides may be obtained from a variety of cell sources which produce HER4-like
  • HER4 polynucleotides including but not limited to brain, cerebellum, pituitary, heart, skeletal muscle, and a variety of breast carcinoma cell lines (see Section 6. , infra) .
  • polynucleotides encoding HER4 polypeptides may be obtained by cDNA cloning from RNA isolated and purified from such cell sources or by genomic cloning. Either cDNA or genomic libraries of clones may be prepared using techniques well known in the art and may be screened for particular HER4- encoding DNAs with nucleotide probes which are substantially complementary to any portion of the HER4 gene. Various PCR cloning techniques may also be used to obtain the HER4 polynucleotides of the invention. A number of PCR cloning protocols suitable for the isolation of HER4 polynucleotides have been reported in the literature (see, for example, PCR protocols: A Guide to Methods and Applications. Eds. Inis et al . , Academic Press, 1990) .
  • polynucleotides containing the entire coding region of the desired HER4 may be isolated as-full length clones or prepared by splicing two or more polynucleotides together.
  • HER4-encoding DNAs may be synthesized in whole or in part by chemical synthesis using techniques standard in the art. Due to the inherent degeneracy of nucleotide coding sequences, any polynucleotide encoding the desired HER4 polypeptide may be used for recombinant expression.
  • IA and IB may be altered by substituting nucleotides such that the same HER4 product is obtained.
  • the invention also provides a number of useful applications of the HER4 polynucleotides of the invention, including but not limited to their use in the preparation of HER4 expression vectors, primers and probes to detect and/or clone HER4, and diagnostic reagents. Diagnostics based upon HER4 polynucleotides include various hybridization and PCR assays known in the art, utilizing HER4 polynucleotides as primers or probes, as appropriate.
  • One particular aspect of the invention relates to a PCR kit comprising a pair of primers capable of priming cDNA synthesis in a PCR reaction, wherein each of the primers is a HER4 polynucleotide of the invention.
  • Such a kit may be useful in the diagnosis of certain human cancers which are characterized by aberrant HER4 expression.
  • certain human carcinomas may overexpress HER4 relative to their normal cell counterparts, such as human carcinomas of the breast.
  • detection of HER4 overexpression mRNA in breast tissue may be an indication of neopla ⁇ ia.
  • human carcinomas characterized by overexpression of HER2 and expression or overexpression of HER4 may be diagnosed by a polynucleotide-based assay kit capable of detecting both HER2 and HER4 RNAs, such a kit comprising, for example, a set of PCR primer pairs derived from divergent sequences in the HER2 and HER4 genes, respectively.
  • Another aspect of the invention is directed to HER4 polypeptides, including the prototype HER4 polypeptide provided herein, as well as polypeptides
  • PEMP-25946.1 derived from or having substantial homology to the amino acid sequence of the prototype HER4 molecule.
  • polypeptide in this context refers to a polypeptide prepared by synthetic or recombinant means, or which is isolated from natural sources.
  • substantially homologous in this context refers to polypeptides of about 80 or more amino acids sharing greater than about 90% amino acid homology to a corresponding contiguous amino acid sequence in the prototype HER4 primary structure (FIG. IA and IB) .
  • prototype HER4" refers to a polypeptide having the amino acid sequence of precursor or mature HER4 as provided in FIG. IA and IB, which is encoded by the consensus cDNA nucleotide sequence also provided therein, or by any polynucleotide sequence which encodes the same amino acid sequence.
  • HER4 polypeptides of the invention may contain deletions, additions or substitutions of amino acid residues relative to the sequence of the prototype HER4 depicted in FIG. IA and IB which result in silent changes thus producing a bioactive product.
  • amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the resides involved.
  • negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; amino acids with uncharged polar head groups or nonpolar head groups having similar hydrophilicity values include the following: leucine, i ⁇ oleucine, valine; glycine, alanine; asparagine, glutamine; serine, threonine; phenylalanine, tyrosine.
  • the HER4 polypeptide depicted in FIG. IA and IB has all of the fundamental structural features
  • P-MP-25946 characterizing the EGFR-family of receptor tyrosine kinases (Hanks et al . , 1988, Science 241:42-52).
  • the precursor contains a single hydrophobic stretch of 26 amino acids characteristic of a transmembrane region that bisects the protein into a 625 amino acid extracellular ligand binding domain, and a 633 amino acid C-terminal cytoplasmic domain.
  • the ligand binding domain can be further divided into 4 subdomains (I - IV) , including two cysteine-rich regions (II, residues 186-334; and IV, residues 496- 633), and two flanking domains (I, residues 29-185; and III, residues 335-495) that may define specificity for ligand binding (Lax et al . , 1988, Mol. Cell. Biol. 8:1970-78).
  • the extracellular domain of HER4 is most similar to HER3, where domains II-IV of HER4 share 56- 67% identity to the respective domains of HER3.
  • the same regions of EGFR and HER2 exhibit 43-51% and 34-46% homology to HER4, respectively (FIG.
  • HER4 also conserves all 50 cysteines present in the extracellular portion of EGFR, HER2, and HER3, except that the HER2 protein lacks the fourth cy ⁇ teine in domain IV.
  • HER4 Following the transmembrane domain of HER4 is a cytoplasmic juxtamembrane region of 37 amino acids. This region shares the highest degree of homology with EGFR (73% amino acid identity) and contains two consensus protein kinase C phosphorylation sites at amino acid residue numbers 679 (Serine) and 699 (Threonine) in the FIG. IA and IB sequence, the latter of which is present in EGFR and HER2. Notably, HER4 lacks a site analogous to Thr654 of EGFR.
  • HER4 also contains Thr692 analogous to Thr694 of HER2. This threonine is absent in EGFR and HER3 and has been proposed to impart cell-type specificity to the mitogenic and transforming activity of the HER2 kinase (DiFiore et al . 1992, EMBO J. 11:3927-33).
  • the juxtamembrane region of HER4 also contains a MAP kinase consensus phosphorylation site at amino acid number 699 (Threonine) , in a position homologous to Thr699 of EGFR which is phosphorylated by MAP kinase in response to EGF stimulation (Takishima et al . , 1991, Proc. Natl. Acad. Sci. U.S.A. 88:2520-25).
  • the remaining cytoplasmic portion of HER4 consists of a 276 amino acid tyrosine kinase domain, an acidic helical structure of 38 amino acids that is homologous to a domain required for ligand-induced internalization of the EGFR (Chen et al . , 1989, Cell 59:33-43), and a 282 amino acid region containing 18 tyrosine residues characteristic of the autophosphorylation domains of other EGFR-related proteins (FIG. 6A and 6B) .
  • the 276 amino acid tyrosine kinase domain conserves all the diagnostic structural motifs of a tyrosine kinase, and is most related to the catalytic domains of EGFR (79% identity) and HER2 (77% identity), and to a lesser degree, HER3 (63% identity) . In this same region, EGFR and HER2 share 83% identity.
  • Examples of the various conserved structural motifs include the following: the ATP-binding motif (GXGXXG) [SEQ ID No:11] with a distal lysine residue that is predicted to be involved in the pho ⁇ photransfer reaction (Hanks et al . , 198, Science 241:42-52; Hunter and Cooper, in
  • the C- terminal 282 amino acids of HER4 has limited homology with HER2 (27%) and EGFR (19%) .
  • the C- terminal domain of each EGFR-family receptor is proline-rich and conserves stretches of 2-7 amino acids that are generally centered around a tyrosine residue. These residues include the major tyrosine autophosphorylation sites of EGFR at Tyrl068, Tyrl086, Tyrll48, and Tyrll73 (FIG. 6A and 6B, filled triangles; Margolis et al., 1989, J. Biol. Chem. 264:10667-71).
  • the HER4 polypeptides of the invention may be produced by the cloning and expression of DNA encoding the desired HER4 polypeptide. Such DNA may be ligated into a number of expression vectors well known in the art and suitable for use in a number of acceptable host organisms, in fused or mature form, and may contain a signal sequence to permit secretion. Both prokaryotic and eukaryotic host expression systems may be employed in the production of recombinant HER4 polypeptides.
  • the prototype HER4 precursor coding sequence or its functional equivalent may be used in a host cell capable of processing the precursor correctly.
  • HER4 precursor coding sequence for mature HER4 may be used to directly express the mature HER4 molecule.
  • Functional equivalents of the HER4 precursor coding sequence include any DNA sequence which, when expressed inside the appropriate host cell, is capable of directing the synthesis, processing and/or export of HER4.
  • Production of a HER4 polypeptide using recombinant DNA technology may be divided into a four- step process for the purposes of description: (1) isolation or generation of DNA encoding the desired HER4 polypeptide; (2) construction of an expression vector capable of directing the synthesis of the desired HER4 polypeptide; (3) transfection or transformation of appropriate host cells capable of replicating and expressing the HER4 coding sequence and/or processing the initial product to produce the desired HER4 polypeptide; and (4) identification and purification of the desired HER4 product.
  • HER4-encoding DNA may be used to construct recombinant • > expression vectors which will direct the expression of the desired HER4 polypeptide product.
  • DNA encoding the prototype HER4 polypeptide (FIG. IA and IB), or fragments or functional equivalents thereof, may be used to generate the recombinant molecules which will direct the expression of the recombinant HER4 product in appropriate host cells.
  • HER4-encoding nucleotide sequences may be obtained from a variety of cell sources which produce HER4-like activities and/or which express HER4-encoding mRNA.
  • HER4- encoding cDNAs may be obtained from the breast adenocarcino a cell line MDA-MB-453 (ATCC HTB131) as
  • HER4 cDNAs include but not limited to various epidermoid and breast carcinoma cells, and normal heart, kidney, and brain cells (see Section 6.2.3., infra) .
  • the HER4 coding sequence may be obtained by molecular cloning from RNA isolated and purified from such cell sources or by genomic cloning. Either cDNA or genomic libraries of clones may be prepared using techniques well known in the art and may be screened for particular HER4-encoding DNAs with nucleotide probes which are substantially complementary to any portion of the HER4 gene. Alternatively, cDNA or genomic DNA may be used as templates for PCR cloning with suitable oligonucleotide primers. Full length clones, i.e., those containing the entire coding region of the desired HER4 may be selected for constructing expression vectors, or overlapping cDNAs can be ligated together to form a complete coding sequence. Alternatively, HER4-encoding DNAs may be synthesized in whole or in part by chemical synthesis using techniques standard in the art.
  • HER4 polypeptides may be utilized equally well by those skilled in the art for the recombinant expression of HER4 polypeptides.
  • Such systems include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing the desired HER4 coding sequence; yeast transformed with recombinant yeast expression vectors containing the desired HER4 coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g..).
  • P-MP-2594C.1 baculovirus containing the desired HER4 coding sequence
  • plant cell systems infected with recombinant virus expression vectors e.g., cauliflower mosaic virus CaMV; tobacco mosaic virus, TMV
  • recombinant plasmid expression vectors e.g., Ti plasmid
  • animal cell systems infected with recombinant virus expression vectors e.g., adenovirus, vaccinia virus
  • the expression elements of these vectors vary in their strength and specificities.
  • any one of a number of suitable transcription and translation elements may be used.
  • promoters isolated from the genome of mammalian cells e.g., mouse metallothionein promoter
  • viruses that grow in these cells e.g., vaccinia virus 7.5K promoter or Moloney murine sarcoma virus long terminal repeat
  • Promoters produced by recombinant DNA or synthetic techniques may also be used to provide for transcription of the inserted sequences.
  • Specific initiation signals are also required for sufficient translation of inserted protein coding sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where the entire HER4 gene including its own initiation codon and adjacent sequences are inserted into the appropriate expression vectors, no additional translational control signals may be needed. However, in cases where only a portion of the coding sequence
  • exogenous translational control signals including the ATG initiation codon must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the HER4 coding sequences to ensure translation of the entire insert.
  • exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of transcription attenuation sequences, enhancer elements, etc.
  • the desired HER4 coding sequence may be ligated to an adenovirus transcription/translation control complex, e .g. , the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e .g. , region E3 or E4) will result in a recombinant virus that is viable and capable of expressing HER4 in infected hosts.
  • the vaccinia 7.5K promoter may be used.
  • An alternative expression system which could be used to express'HER4 is an insect system.
  • Autographa californica nuclear polyhidrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • the HER4 coding sequence may be cloned into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter) .
  • Successful insertion of the HER4 coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat encoded by the polyhedrin gene) .
  • PBKP-2594S.1 These recombinant viruses are then used to infect Spodoptera frugiperda cells in which the inserted gene is expressed.
  • Yet another approach uses retroviral vectors prepared in amphotropic packaging cell lines, which permit high efficiency expression in numerous cells types. This method allows one to assess cell- type specific processing, regulation or function of the inserted protein coding sequence.
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers (e.g., zinc and cadmium ions for metallothionein promoters). Therefore, expression of the recombinant HER4 polypeptide may be controlled. This is important if the protein product of the cloned foreign gene is lethal to host cells. Furthermore, modifications (e.g., phosphorylation) and processing (e.g., cleavage) of protein products are important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of protein. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • inducers e.g., zinc and cadmium ions for metallothionein promoters. Therefore, expression of the recombinant HER4 polypeptide may be controlled
  • the host cells which contain the recombinant coding sequence and which express the desired HER4 polypeptide product may be identified by at least four general approaches (a) DNA-DNA, DNA-RNA or RNA- antisense RNA hybridization; (b) the presence or absence of "marker" gene functions; (c) assessing the level of transcription as measured by the expression
  • HER4 coding sequences inserted into expression vectors can be detected by DNA-DNA hybridization using hybridization probes and/or primers for PCR reactions comprising polynucleotides that are homologous to the HER4 coding sequence.
  • the recombinant expression vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, resistance to methotrexate (MTX) , resistance to methionine sulfoximine (MSX) , transformation phenotype, occlusion body formation in baculovirus, etc.).
  • certain "marker” gene functions e.g., thymidine kinase activity, resistance to antibiotics, resistance to methotrexate (MTX) , resistance to methionine sulfoximine (MSX) , transformation phenotype, occlusion body formation in baculovirus, etc.
  • a marker gene can be placed in tandem with the HER4 sequence under the control of the same or different promoter used to control the expression of the HER4 coding sequence. Expression of the marker in response to induction or selection indicates expression of the HER4 coding sequence.
  • a HER4 expression vector incorporating glutamine synthetase as a selectable marker is constructed, used to transfect CHO cells, and amplified expression of HER4 in CHO cells is obtained by selection with increasing concentration of MSX.
  • transcriptional activity for the HER4 coding region can be assessed by hybridization assays.
  • polyadenylated RNA can be isolated and analyzed by Northern blot using a probe homologous to the HER4 coding sequence or particular portions thereof.
  • total nucleic acids of the host cell may be extracted and assayed for hybridization to such probes.
  • the expression of HER4 can be assessed immunologically, for example by Western blots, immunoassays such as radioimmunoprecipitation, enzyme-linked immunoassays and the like.
  • expression of HER4 may be assessed by detecting a biologically active product. Where the host cell secretes the gene product the cell free media obtained from the cultured transfectant host cell may be assayed for HER4 activity. Where the gene product is not secreted, cell lysates may be assayed for such activity. In either case, assays which measure ligand binding to
  • HER4 may be used.
  • anti-HER4 antibodies are expected to have a variety of useful applications in the field of oncology, several of which are described generally below. More detailed and specific descriptions of various uses for anti-HER4 antibodies are provided in the sections and subsections which follow. Briefly, anti-HER4 antibodies may be used for the detection and quanti ication of HER4 polypeptide expression in cultured cells, tissue samples, and in vivo. Such
  • PKMP-2594C.1 immunological detection of HER4 may be used, for example, to identify, monitor, and assist in the prognosis of neoplasms characterized by aberrant or attenuated HER4 expression and/or function. Additionally, monoclonal antibodies recognizing epitopes from different parts of the HER4 structure may be used to detect and/or distinguish between native HER4 and various subcomponent and/or mutant forms of the molecule. Anti-HER4 antibody preparations are also envisioned as useful biomodulatory agents capable of effectively treating particular human cancers.
  • anti-HER4 antibodies In addition to the various diagnostic and therapeutic utilities of anti-HER4 antibodies, a number of industrial and research applications will be obvious to those skilled in the art, including, for example, the use of anti-HER4 antibodies as affinity reagents for the purification of HER4 polypeptides, and as immunological probes for elucidating the biosynthesis, metabolism and biological functions of HER4.
  • Anti-HE 4 antibodies may be useful for influencing cell functions and behaviors which are directly or indirectly mediated by HER4.
  • modulation of HER4 biological activity with anti-HER4 antibodies may influence HER2 activation and, as a consequence, modulate intracellular signals generated by HER2.
  • anti-HER4 antibodies may be useful to effectively block ligand- induced, HER4-mediated activation of HER2, thereby affecting HER2 biological activity.
  • anti- HER4 antibodies capable of acting as HER4 ligands may be used to trigger HER4 biological activity and/or initiate a ligand-induced, HER4-mediated effect on HER2 biological activity, resulting in a cellular
  • PBNP-2594C.1 response such as differentiation, growth inhibition, etc.
  • anti-HER4 antibodies conjugated to cytotoxic compounds may be used to selectively target such compounds to tumor cells expressing HER4, resulting in tumor cell death and reduction or eradication of the tumor.
  • toxin-conjugated antibodies having the capacity to bind to HER4 and internalize into such cells are administered systemically for targeted cytotoxic effect.
  • the preparation and use of radionuclide and toxin conjugated anti-HER4 antibodies are further described in Section 5.5., infra .
  • HER4 is expressed in certain human carcinomas in which HER2 overexpression is present. Therefore, anti-HER4 antibodies may have growth and differentiation regulatory effects on cells which overexpress HER2 in combination with HER4 expression, including but not limited to breast adenocarcinoma cells. Accordingly, this invention includes antibodies capable of binding to the HER4 receptor and modulating HER2 or HER2-HER4 functionality, thereby affecting a response in the target cell.
  • agents capable of selectively and specifically affecting the intracellular molecular interaction between these two receptors may be conjugated to internalizing anti-HER4 antibodies. The specificity of such agents may result in biological effects only in cells which co-express HER2 and HER4, such as breast cancer cells.
  • HER4 PBNP-2594-.1 epitopes of HER4.
  • a number of host animals are acceptable for the generation of anti-HER4 antibodies by immunization with one or more injections of a HER4 polypeptide preparation, including but not limited to rabbits, mice, rats, etc.
  • adjuvants may be used to increase the immunological response in the host animal, depending on the host species, including but not limited to Freund's (complete and incomplete) , mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, oil emulsions, keyhole lympet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • Corynebacterium parvum bacille Calmette-Guerin
  • a monoclonal antibody to an epitope of HER4 may be prepared by using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975, Nature 256, 495-497) , and the more recent human B-cell hybridoma technique (Kosbor et al . , 1983, 4:72) and EBV-hybridoma technique (Cole et al . , 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). In addition, techniques developed for the production of "chimeric antibodies" by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity may be used (Morrison et al . ,
  • PEMP-2S94C.1 antibodies (U.S. Patent 4,946,778) can be adapted to produce HER4-specific single chain antibodies.
  • Recombinant human or humanized versions of anti-HER4 monoclonal antibodies are a preferred embodiment for human therapeutic applications.
  • Humanized antibodies may be prepared according to procedures in the literature (e.g., Jones et al . , 1986, Nature 321:522- 25; Reichman et al . , 1988, Nature 332:323-27; Verhoeyen et al . , 1988, Science 239:1534-36).
  • anti-HER4 monoclonal antibodies may be generated by immunization of mice with cells selectively overexpressing HER4 (e.g., CH0/HER4 21-2 cells as deposited with the ATCC) or with partially purified recombinant HER4 polypeptides.
  • the full length HER4 polypeptide (FIG. IA and IB) may be expressed in Baculovirus systems, and membrane fractions of the recombinant cells used to immunize mice.
  • Hybridomas are then screened on CH0/HER4 cells (e.g., CHO HER4 21-2 cells as deposited with the ATCC) to identify monoclonal antibodies reactive with the extracellular domain of HER4.
  • Such monoclonal antibodies may be evaluated for their ability to block NDF, or HepG2-differentiating factor, binding to HER4; for their ability to bind and stay resident on the cell surface, or to internalize into
  • monoclonal antibodies N28 and N29 directed to HER2, specifically bind HER2 with high affinity.
  • monoclonal N29 binding results in receptor internalization and downregulation, morphologic differentiation, and inhibition of HER2 expressing tumor cells in athymic mice.
  • monoclonal N28 binding to HER2 expressing cells results in stimulation of autophosphorylation, and an acceleration of tumor cell growth both in vitro and in vivo (Bacus et al .
  • a soluble recombinant HER4-Immunoglobulin (HER4-Ig) fusion protein is expressed and purified on a Protein A affinity column.
  • the amino acid sequence of one such HER4-Ig fusion protein is provided in FIG. 14.
  • the soluble HER4-Ig fusion protein may then be used to screen phage libraries designed so that all available combinations of a variable domain of the antibody binding site are presented on the surfaces of the phages in the library.
  • Recombinant anti-HER4 antibodies may be propagated from phage which specifically recognize the HER4-Ig fusion protein.
  • Antibody fragments which contain the idiotype of the molecule may be generated by known techniques.
  • such fragments include but are not limited to: the F(ab) 'E2 fragment which can be produced by pepsin digestion of the intact antibody molecule; the Fab' fragments which can be generated by reducing the disulfide bridges of the F(ab')2 fragment, and the two Fab fragments which can be
  • F-MP-2594C.1 generated by treating the antibody molecule with papain and a reducing agent.
  • Fab expression libraries may be constructed (Huse et al . , 1989, Science. 246:1275-1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity to HER4 protein.
  • HER4 ligands are capable of binding to the 18OK transmembrane protein, HER4/pl80 erbB4 or functional analogues thereof, and activating tyrosine kinase activity.
  • Functional analogues of HER4/pl80" bB4 -ligands are capable of activating HER4 tyrosine kinase activity.
  • Activation of the tyrosine kinase activity may stimulate autophosphorylation and may affect a biological activity mediated by HER4. It has been observed in systems described in Section 12 and 13 that binding of HER4 ligands to HER4 triggers tyrosine phosphorylation and affects differentiation of breast cancer cells.
  • the HER4 ligands of the present invention include NDF, a 44 kDa glycoprotein isolated from ras- transformed rat fibroblasts (Wen et al . , 1992, Cell 69:559-572); heregulin, its human homologue, which exists as multiple isoforms (Peles et al . , 1992, Cell 69:205-218 and Holmes et al .
  • HER4 ligands of the present invention can be prepared by synthetic or recombinant means, or can be 5 isolated from natural sources.
  • the HER4 ligand of the present invention may contain deletions, additions or substitutions of amino acid residues relative to the sequence of NDF, p45 or other heregulins or any HER4 ligand known in the art as long as the ligand 0 maintains HER4 receptor binding and tyrosine kinase activation capacity.
  • Such amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the resides involved.
  • negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; amino acids with uncharged polar head groups or nonpolar head groups having similar hydrophilicity values include 0 the following: leucine, isoleucine, valine; glycine, alanine; asparagine, glutamine; serine, threonine; phenylalanine, tyrosine.
  • the HER4 ligands of the present invention may be produced by the cloning and expression of DNA encoding the desired HER4 ligand.
  • DNA may be ligated into a number of expression vectors well known in the art 0 and suitable for use in a number of acceptable host organisms, in fused or mature form, and may contain a signal sequence to permit secretion.
  • Both prokaryotic and eukaryotic host expression systems may be employed in the production of recombinant HER4 ligands.
  • a HER4 ligand precursor coding sequence or its functional equivalent may be used in a host cell
  • the coding sequence for a mature HER4 ligand may be used to directly express the mature HER4 ligand molecule.
  • Functional equivalents of the HER4 ligand precursor coding sequence include any DNA sequence which, when expressed inside the appropriate host cell, is capable of directing the synthesis, processing and/or export of the HER4 ligand.
  • Production of a HER4 ligand using recombinant DNA technology may be divided into a four-step process for the purposes of description: (1) isolation or generation of DNA encoding the desired HER4 ligand; (2) construction of an expression vector capable of directing the synthesis of the desired HER4 ligand; (3) transfection or transformation of appropriate host cells capable of replicating and expressing the HER4 ligand coding sequence and/or processing the initial product to produce the desired HER4 ligand; and (4) identification and purification of the desired HER4 ligand product.
  • HER4 ligand-encoding nucleic acid sequences' may be obtained from human hepatocellular carcinoma cell lines, specifically the HepG2 cells available from the ATCC, accession number HB 8065.
  • a number of human cell sources are suitable for obtaining HER4 ligand nucleic acids, including MDA-MB-231 cells available from the ATCC, accession number HTB 26, brain tissue (Falls et al . , 1993, Cell 72:801-815 and Marchionni et al .
  • Anti-HER4 Ligand Antibodies The present invention is also directed to polyclonal and monoclonal antibodies which recognize eptitopes of HER4 ligand polypeptides.
  • Anti-HER4 ligand antibodies are expected to have a variety of useful applications in the field of oncology. Briefly, anti-HER4 ligand antibodies may be used for the detection and quantification of HER4 ligand polypeptide expression in cultured cells, tissue samples, and in vivo.
  • monoclonal antibodies recognizing epitopes from different parts of the HER4 ligand structure may be used to detect and/or distinguish binding from non-binding regions of the ligand.
  • Anti-HER4 ligand antibody preparations are also envisioned as useful biomodulatory agents capable of effectively treating particular human cancers.
  • An anti-HER4 ligand antibody could be used to block signal transduction mediated through HER4, thereby inhibiting undesirable biological responses.
  • anti-HER4 ligand antibodies a number of industrial and research applications will be obvious to those skilled in the art, including, for example, the use of anti-HER4 ligand antibodies as affinity reagents for the purification of HER4 ligand polypeptides, and as immunological probes for elucidating the biosynthesis, metabolism and biological functions of HER4 ligands.
  • Anti-HER4 ligand antibodies may be useful for influencing cell functions and behaviors which are directly or indirectly mediated by HER4. As an example, modulation of HER4 biological activity with anti-HER4 ligand antibodies may influence HER2 activation and, as a consequence, modulate intracellular signals generated by HER2. In this regard, anti-HER4 ligand antibodies may be useful to effectively block ligand-induced, HER4-mediated activation of HER2, thereby affecting HER2 biological activity.
  • anti-HE 4 ligand antibodies capable of acting as HER4 ligands may be used to trigger HER4 biological activity and/or initiate a ligand-induced, HER4-mediated effect on HER2 biological activity, resulting in a cellular response such as differentiation, growth inhibition, etc.
  • anti-HER4 ligand antibodies conjugated to cytotoxic compounds may be used to selectively target such compounds to tumor cells expressing HER4, resulting in tumor cell death and reduction or eradication of the tumor.
  • the invention also relates to the detection of human neoplastic conditions, particularly carcinomas of epithelial origin, and more particularly human breast carcinomas.
  • oligomer ⁇ corresponding to portions of the consensus HER4 cDNA sequence provided in FIG. IA and IB are used for the quantitative detection of HER4 mRNA levels in a human biological sample, such as blood, serum, or tissue biopsy samples, using a suitable hybridization or PCR
  • detection of HER4 mRNA may be combined with the detection HER2 mRNA overexpression, using appropriate HER2 sequences, to identify neoplasias in which a functional relationship between HER2 and HER4 may exist.
  • labeled anti-HER4 antibodies or antibody derivatives are used to detect the presence of HER4 in biological samples, using a variety of immunoassay formats well known in the art, and may be used for in situ diagnostic radioimmunoimaging. Current diagnostic and staging techniques do not routinely provide a comprehensive scan of the body for metastatic tumors. Accordingly, anti-HER4 antibodies labeled with, for example, fluorescent, chemiluminescent, and radioactive molecules may overcome this limitation.
  • a gamma-emitting diagnostic radionuclide is attached to a monoclonal antibody which is specific for an epitope of HER4, but not significantly cross-reactive with other EGFR-fam'ily members.
  • the labeled antibody is then injected into a patient systemically, and total body imaging for the distribution and density of HER4 molecules is performed using gamma cameras, followed by localized imaging using computerized tomography or magnetic resonance imaging to confirm and/or evaluate the condition, if necessary.
  • Preferred diagnostic radionuclides include but are not limited to technetium-99m, indium-Ill, iodine-123, and iodine- 131.
  • Ab-MTs Recombinant antibody-metallothionein chimeras
  • EGFR-family can be generated by transfection of a variety of parental cell types with an appropriate expression vector as described in Section 7., infra .
  • Candidate ligands, or partially purified preparations may be applied to such cells and assayed for receptor binding and/or activation.
  • a CHO-KI cell line transfected with a HER4 expression plasmid and lacking detectable EGFR, HER2, or HER3 may be used to screen for HER4-specific ligands.
  • a particular embodiment of such a cell line is described in Section 7., infra, and has been deposited with the ATCC (CHO/HER4 21-2) .
  • Ligands may be identified by ' detection of HER4 autophosphorylation, stimulation of DNA synthesis, induction of morphologic differentiation, relief from serum or growth factor requirements in the culture media, and direct binding of labeled purified growth factor.
  • the invention also relates to a bioassay for testing potential analogs of HER4 ligands based on a capacity to affect a biological activity mediated by the HER4 receptor.
  • the invention is also directed to methods for the treatment of human cancers involving abnormal expression and/or function of HER4 and cancers in which HER2 overexpression is combined with the proximate expression of HER4, including but not limited to human breast carcinomas and other neoplasms overexpressing HER4 or overexpressing HER2 in combination with expression of HER4.
  • the cancer therapy methods of the invention are generally based on treatments with unconjugated, toxin- or radionuclide- conjugated HER4 antibodies, ligands, and derivatives or fragments thereof.
  • such HER4 antibodies or ligands may be used for systemic and targeted therapy of certain cancers overexpressing HER2 and/or HER4, such as metastatic breast cancer, with minimal toxicity to normal tissues and organs.
  • an anti-HER2 monoclonal antibody has been shown to inhibit the growth of human tumor cells v overexpressing HER2 (Bacus et al . , 1992, Cancer Res. 52:2580-89).
  • modulation of heregulin signaling through HER4 provides a means to affect the growth and differentiation of cells overexpressing HER2, such as certain breast cancer cells, using HER4-neutralizing monoclonal antibodies, NDF/HER4 antagonists, monoclonal antibodies or ligands which act as super- agonists for HER4 activation, or agents which block the interaction between HER2 and HER4, either by disrupting heterodimer formation or by blocking HER- mediated phosphorylation of the HER2 substrate.
  • ricin a cytotoxin from the Ricinis communis plant may be conjugated to an anti-HER4 antibody using methods known in the art (e.g., Blakey et al . , 1988, Pro ⁇ . Aller ⁇ v 45:50-90; Marsh and Neville, 1988, J. Immunol. 140:3674-78).
  • ricin Once ricin is inside the cell cytoplasm, its A chain inhibits protein synthesis by inactivating the 60S ribosomal subunit (March et al . , 1989, EMBO J.
  • Immunotoxins of ricin are therefore extremely cytotoxic.
  • ricin immunotoxins are not ideally specific because the B chain can bind to virtually all cell surface receptors, and immunotoxins made with ricin A chain alone have increased specificity. Recombinant or deglycosylated forms of the ricin A chain may result in improved survival (i.e., slower clearance from circulation) of the immunotoxins.
  • Methods for conjugating ricin A chain to antibodies are known (e.g., Vitella and Thorpe, in: Seminars in Cell Biolo ⁇ v. pp 47-58; Saunder ⁇ , Philadelphia 1991) .
  • Additional toxins which may ' be used in the formulation of immunotoxins include but are not limited to daunorubicin, methotrexate, ribosome inhibitors (e.g., trichosanthin, trichokirin, gelonin, saporin, mormordin, and pokeweed antiviral protein) and various bacterial toxins (e.g., Pseudomonas exotoxin) .
  • Immunotoxins for targeted cancer therapy may be administered by any route which will result in antibody interaction with the target cancer cells, including systemic administration and injection directly to the site of tumor.
  • Another therapeutic strategy may be the administration of immunotoxins by sustained-release systems, such as
  • PBMP-2594C.1 semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials Various of sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release immunotoxic molecules for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
  • preferred radionuclides for labeling include alpha, beta, and Auger electron emitters.
  • alpha emitters include astatine 211 and bismuth 212; beta emitters include iodine 131, rhenium 188, copper 67 and yttrium 90; and iodine 125 is an example of an Auger electron emitter.
  • purified ligand molecules may be chemically conjugated to cytotoxic substances.
  • recombinant chimeric polypeptides comprising a HER4 binding (-ligand) portion fused to all or'part of a cytotoxin may be engineered by constructing vectors comprising DNA encoding the ligand in reading frame with DNA encoding the toxin or part thereof.
  • recombinant ligand-toxins may be used to specifically target HER4 expressing cancer cells.
  • a particular embodiment of such a ligand-toxin is disclosed herein and described in more detail in Sections 5.8.2., infra , and Section 15, infra .
  • HER4 expressing tumor cells may be specifically targeted and killed by contacting such tumor cells with a fusion protein comprising a cytotoxic polypeptide covalently linked to a polypeptide which is capable of activating HER4 expressed on such cells.
  • a fusion protein comprising a chimeric heregulin 02 ligand and the cytotoxic substance PE40 is generated by expression of the corresponding chimeric coding sequence.
  • PE40 is a derivative of the Pseudomonas exotoxin PE, a potent cell killing agent made by Pseudomonas aeruginosa (Fitzgerald et al . , 1980, Cell 21:867-873).
  • the wildtype protein PE contains three domains whose functions are cell recognition, membrane translocation, and ADP ribosylation of elongation factor 2.
  • PE40 kills cells by binding to a cell surface receptor, entering the cell via an endocytotic vesicle and catalyzing ADP-ribosylation of elongation factor 2.
  • the derivative PE40 lacks the cell binding function of the wildtype protein, but still exhibits strong cytotoxic activity. Generation of PE40 fusion proteins with specific cell targeting molecules have been described (Kondo et al . , 1988, J. Biol. Chem.
  • the chimeric heregulin-toxin protein HAR-TX 02 described in Section 15, infra contains the amphiregulin (AR) leader sequence thereby facilitating the purification of the recombinant protein. As confirmed by applicants' data, the AR leader has no influence on the binding specificity of the recombinant heregulin-toxin.
  • Related embodiments include, for example, PE40 linked to other members of the heregulin family, like heregulin-01 and heregulin- ⁇ , and other molecules capable of activating HER4.
  • the applicants demonstrate specificity of the cytotoxic effect of the chimeric heregulin-PE40 protein to HER4 expressing cancer cells; they include but are not limited to prostate carcinoma, bladder carcinoma, and a considerable number of different breast cancer types, including breast carcinoma cells with amplified HER2 expression.
  • the bifunctional retention of both the specificity of the cell binding portion of the molecule and the cytotoxic potential of PE40 provides a very potent and targeted reagent.
  • An effective therapeutic amount of heregulin- toxin will depend upon the therapeutic objectives, the route of administration, and the condition of the patient. Accordingly, dosages should be titrated and the route of administration modified as required to obtain the optimal therapeutic effect.
  • a typical daily dosage may be in the range of 0.1 mg/kg - 1 mg/kg, preferably between 0.1 and 0.5 mg/kg, with intravenous administration. For regression of solid tumors, it may take 3-5 doses, with schedules such as 3 doses, each four days apart. Also the use of sustained-release preparations (see Section 5.8.1., supra) may be considered for administration of the reagent.
  • P-HP-2594C.1 may be between 2 and 10, which means that a tumor regression effect would be expected between 2- and 10- fold below the toxic dose (see Section 15, infra) .
  • the heregulin-toxin will be administered at a dose and frequency that achieves the desired therapeutic effect, which can be monitored using conventional assays.
  • Cancer therapy with heregulin-toxins of the invention may be combined with chemotherapy, surgery, and radiation therapy, depending on the type of tumor.
  • One advantage of using a low molecular weight toxin drug is that they are capable of targeting metastatic lesions that cannot be located and removed by surgery.
  • Heregulin-toxins may also be particularly useful on patients that are MDR (Ifulti ⁇ rug ⁇ esistance) positive since their mechanism of action is not inhibited by the p-glycoprotein pump of MDR positive cells as are many standard cancer therapeutic drugs.
  • HER4 ligands may include other diseases caused by deficient HER4 receptor tyrosine kinase activation rather than by hyperactivation.
  • type II diabetes mellitus is the consequence of deficient insulin- mediated signal transduction, caused by mutations in the insulin-receptor, including mutations in the ligand-binding domain (Taira et al . , 1889, Science 245:63-66; Odawara et al . , 1989, Science 245:66-68; Obermeier-Kusser et al . , 1989, J. Biol. Chem.
  • Such diseases might be treated by administration of modified ligands or ligand-analogues which re-establish a functional ligand-receptor interaction.
  • derivatives, analogues and peptides related to HER4 are also envisioned and are within the scope of the invention.
  • Such derivatives, analogues and peptides may be used to compete with native HER4 for binding of HER4 specific ligand, thereby inhibiting HER4 signal transduction and function.
  • the inhibition of HER4 function may be utilized in several applications, including but not limited to the treatment of cancers in which HER4 biological activity is involved.
  • a series of deletion mutants in the HER4 nucleotide coding sequence depicted in FIG. IA and IB may be constructed and analyzed to determine the minimum amino acid sequence requirements for binding of a HER4 ligand.
  • Deletion mutants of the HER4 coding sequence may be constructed using methods known in the art which include but are not limited to use of nucleases and/or restriction enzymes; site-directed mutagenesis techniques, PCR, etc. The mutated polypeptides expressed may be assayed for their ability to bind HER4 ligand.
  • the DNA sequence encoding the desired HER4 analogue may then be cloned into an appropriate expression vector for overexpression in either bacteria or eukaryotic cells.
  • Peptides may be purified from cell extracts in a number of ways including but not limited to ion-exchange chromatography or affinity chromatography using HER4 ligand or antibody. Alternatively, polypeptides may be synthesized by solid phase techniques followed by cleavage from resin and purification by high performance liquid chromatography.
  • EGFR and the related proteins, HER2, HER3, and Xmrk exhibit extensive amino acid homology in their tyrosine kinase domains (Kaplan et ai., 1991, Nature 350:158-160; Wen et al . , 1992, Cell 69:559-72; Holmes et al . , 1992, Science 256:1205-10; Hirai et al . , Science 1987 238:1717-20).
  • there is strict conservation of the exon-intron boundaries within the genomic regions that encode these catalytic domains (Wen et al . , supra; Lindberg and Hunter, 1990, Mol. Cell. Biol. 10:6316-24; and unpublished observations) .
  • oligonucleotide primers were designed based on conserved amino acids encoded by a single exon or adjacent exons from the kinase domains of these four proteins. These primers were used in a polymerase chain reaction (PCR) to isolate genomic fragments corresponding to murine EGFR, erJbB2 and erbB3. In addition, a highly related DNA fragment (designated MER4) was identified as distinct from these other genes. A similar strategy was used to obtain a cDNA clone corresponding to the human homologue of MER4 from the breast cancer cell line, MDA-MB-453. Using this fragment as a probe, several breast cancer cell lines and human heart were found to be an abundant source of the EGFR-related transcript. cDNA libraries were constructed using RNA from human heart and MDA-MB-453 cells, and overlapping clones were isolated spanning the complete open reading frame of HER4/eribB4.
  • MER4-85 one clone was identified that contained a 144 nucleotide insert corresponding to murine er_>B4.
  • This 32P-labeled insert was used to isolate a 17-kiloba ⁇ e fragment from a murine T-cell genomic library (Stratagene, La Jolla, CA) that was found to contain two exons of the murine eri>B4 gene.
  • a specific oligonucleotide (4M3070) was synthesized based on the DNA sequence of an er_>B4 exon, and used in a PCR protocol with a degenerate 5'-oligonucleotide (H4PIKWMA) on a template of single stranded MDA-MB-453 cDNA. This reaction generated a 260 nucleotide fragment (pMDAPIK) corresponding to human HER4.
  • cDNA libraries were constructed in lambda ZAP II (Stratagene) from oligo(dT)- and specific-primed MDA- MB453 and human heart RNA (Plowman et al . , supra; Plowman et al . , 1990, Mol. Cell. Biol. 10:1969-81).
  • HER4-specific clones were isolated by probing the libraries with the 32 P-labeled insert from pMDAPIK. To complete the cloning of the 5'-portion of HER4, we used a PCR strategy to allow for rapid amplification of cDNA ends (Plowman et al . , supra ; Frohman et al . , 1988, Proc. Natl. Acad. Sci. U.S.A. 85:8998-9002). All cDNA clones and several PCR generated clones were sequenced on both strands using T7 polymerase with oligonucleotide primers (Tabor and Richardson, 1987, Proc. Natl. Acad. Sci. U.S.A. 84:4767-71..
  • 3'- and 5'-HER4 specific [ ⁇ M P]DTP-labeled antisense RNA probes were synthesized from the linearized plasmids pHtlB1.6 (containing an 800 bp HER4 fragment beginning at nucleotide 3098) and p5'H4E7 (containing a 1 kb fragment from the 5'-end of the HER4 sequence) , respectively.
  • the Northern blot (Clontech, Palo Alto, CA) contained 2 Mg
  • the filter was prehybridized at 60° C for several hours in RNA hybridization mixture (50% formamide, 5x SSC, 0.5% SDS, lOx Denhardt's solution, 100 ⁇ g/ml denatured herring sperm DNA, 100 ⁇ g/ml tRNA, and 10 ⁇ g/ml polyadenosine) and hybridized in the same buffer at 60° C, overnight with 1-1.5 x 106 cpm/ml of 32P- labeled antisense RNA probe.
  • the filters were washed in O.lXSSC/0.1% SDS, 65° C, and exposed overnight on a Phospholmager (Molecular Dynamics, Sunnyvale, CA) .
  • RNA was isolated from a variety of human cell lines, fresh frozen tissues, and primary tumors. Single stranded cDNA was synthesized from 10 ⁇ g of each RNA by priming with an oligonucleotide containing a T17 track on its 3'-end
  • reaction products were electrophoresed on 2% agarose gels, stained with ethidium bromide and photographed on a UV light box. The relative intensity of the 291- bp HER4-specific bands were estimated for each sample as shown in Table II.
  • a 25 amino acid hydrophobic signal sequence follows a consensus initiating methionine at position number 1 in the amino acid sequence depicted in FIG. IA and IB.
  • the mature HER4 polypeptide would be predicted to begin at amino acid residue number 26 in the sequence depicted in FIG. IA and IB (Gin) , followed by the next 1283 amino acids in the sequence.
  • the prototype mature HER4 of the invention is a polypeptide of 1284 amino acids, having a calculated Mr of 144,260 daltons and an amino acid sequence corresponding to residues 26 through 1309 in FIG. IA and IB.
  • HER4 nucleotide sequence is unique, and revealed a 60/64 amino acid identity with HER2 and a 54/54 amino acid identity to a fragment of a rat EGFR homolog, tyro-2.
  • P-MP-25946.1 also isolated from the MDA-MB-453 cDNA library and comprised two forms.
  • the first alternative type of cDNA was identical to the consensus HER4 nucleotide sequence up to nucleotide 3168 (encoding Arg at amino acid position 1045 in the FIG. IA and IB) and then abruptly diverges into an apparently unrelated sequence (FIG. 2A and 2B, FIG. 4) . Downstream from this residue the open reading frame continues for another 13 amino acids before reaching a stop codon followed by a 2 kb 3'- untranslated sequence and poly(A) tail. This cDNA would be predicted to result in a HER4 variant having the C-terminal autophosphorylation domain of the prototype HER4 deleted.
  • a second type of cDNA was isolated as 4 independent clones each with a 3'-sequence identical to the HER4 consensus, but then diverging on the 5'- side of nucleotide 2335 (encoding Glu at amino acid position 768 in the FIG. IA and IB) , continuing upstream for only another 114-154 nucleotides (FIG. 3, FIG. 5) .
  • Nucleotide 2335 is the precise location of an intron-exon junction in the HER2 gene (Coussens et al . , 1985, Science 230:1132-39; Semba et al . , 1985, Proc. Natl. Acad. Sci. U.S.A.
  • cDNAs could be derived from mRNAs that have initiated from a cryptic promoter within the flanking intron.
  • These 5'-truncated transcripts contain an open reading frame identical to that of the HER4 cDNA sequence of FIG. IA and IB, beginning with the codon for Met at amino acid position 772 in FIG. IA and IB.
  • These cDNAs would be predicted to encode a cytoplasmic HER4 variant polypeptide that initiates just downstream from the ATP-binding domain of the HER4 kinase.
  • Northern blots of poly(A)+ mRNA from human tissue samples were hybridized with antisense RNA probes to the 3'-end of HER4, encoding the autophosphorylation domain, as described in Section 6.1.2., supra.
  • a HER4 mRNA transcript of approximately 6kb was identified, and was found to be most abundant in the heart and skeletal muscle (FIG. 8, Panel 1).
  • An mRNA of greater than approximately 15 kb was detected in the brain, with lower levels also detected in heart, skeletal muscle, kidney, and pancreas tissue samples.
  • HER4 mRNA samples were also examined for the presence of HER4 mRNA using the semi-quantitative PCR assay described in Section 6.1.3., supra . The results are shown in Table II, together with results of the assay on primary tumor samples and neoplastic cell lines (Section 6.2.4., immediately below). These results correlate well with the Northern and solution hybridization analysis results on the selected RNA samples. The highest levels of HER4 transcript expression were found in heart, kidney, and brain tissue samples. In addition, high levels of HER4 mRNA expression were found in parathyroid, cerebellum,
  • P8MP-2594C.1 pituitary, spleen, testis, and breast tissue samples. Lower expression levels were found in thymus, lung, salivary gland, and pancreas tissue samples. Finally, low or negative expression was observed in liver, prostate, ovary, adrenal, colon, duodenum, epidermis, and bone marrow samples.
  • HER4 mRNA expression profiles in several primary tumors and a number of cell lines of diverse neoplastic origin were determined with the semi- quantitative PCR assay (Section 6.1.3, supra) using primers from sequences in the HER4 kinase domain. The results are included in Table II. This analysis detected the highest expression of HER4 RNA in 4 human mammary adenocarcinoma cell lines (T-47D, MDA-MB-453, BT-474, and H3396) , and in neuroblastoma (SK-N-MC) , and pancreatic carcinoma (Hs766T) cell lines.
  • SK-N-MC (neural) HEPM (palate) Wilrns Tumor (kidney) 458(medullablastoma) Breast Carcinoma Kidney Brain Skeletal Muscle Heart Cerebellum Thymus Parathyroid Pituitary Pancreas
  • MDB-MB-231 (breast) MDA-MB-468 (breast) MDA-MB-157 (breast) G-401 (kidney) SK-BR-3 (breast) H ⁇ pG2 (liver) A-431 (vulva) PANC-1 (pancreas) caki-1 (kidney) A ⁇ PC-1(pancrea ) Caki-2 (kidney) Capan-1 (pancreas) SK-HEP-1 (liver) HT-29 (colon) THP-1 (macrophage) CaSki (cervix) PA-1 (ovary)
  • Adrenal SK-MEL-28 (melanoma)
  • CHO-KI cells were obtained from the ATCC
  • Transfected cell colonies expressing HER4 were selected in gluta ine-free Glasgow modified Eagle's medium (GMEM-S, Gibco) supplemented with 10% dialyzed fetal bovine serum an increasing concentrations of methionine sulfoximine (Bebbington, 1991, in Methods: A Companion to Methods in Enzvmolo ⁇ v 2:136-145 Academic Press).
  • GMEM-S Glasgow modified Eagle's medium
  • HER4 The complete 4 kilobase coding sequence of prototype HER4 was reconstructed and inserted into a glutamine synthetase expression vector, pEE14, under the control of the cytomegalovirus immediate-early promoter (Bebbington, supra) to generate the HER4 expression vector pEEHER4.
  • This construct (pEEHER4) was linearized with Mlul and transfected into CHO-KI cells by calcium phosphate precipitation using standard techniques.
  • HER4 Expression Im unoassay Confluent cell monolayers were scraped into hypotonic lysis buffer (10 mM Tris pH7.4, 1 mM KC1, 2 mM MgCl2) at 4 ⁇ C, dounce homogenized with 30 strokes, and the cell debris was removed by centrifugation at 3500 x g, 5 min. Membrane fractions were collected by centrifugation at 100,000 x g, 20 min, and the pellet was resuspended in hot Laemmli sample buffer with 2- ercaptoethanol.
  • HER4 polypeptide was detected by immunoblot analysis on solubilized cells or membrane preparations using HER2 immunoreagents generated to either a 19 amino acid region of the HER2 kinase domain, which coincidentally is identical to the HER4 sequence (residues 927-945) , or to the C-terminal 14 residues of HER2, which share a stretch of 7 consecutive residues with a region near the C-terminus of HER4.
  • HER4 was detected from solubilized cell extracts by immunoblot analysis with PY20 anti-phosphotyrosine antibody (ICN Biochemicals) , presumably reflecting autoactivation and autophosphorylation of HER4 due to receptor aggregation resulting from abberantly high receptor density. More specifically, expression'was detected by immunobloting with a primary murine monoclonal antibody to HER2 (Neu-Ab3, Oncogene
  • HER4 expressed in CHO/HER4 cells migrated with an apparent Mr of 180,000, slightly less than HER2, whereas the parental CHO cells showed no cross-reactive bands (FIG. 9) .
  • a 130 kDa band was also detected in the CHO/HER4 cells, and presumably represents a degradation product of the 180 kDa mature protein.
  • CHO/HER4 cells were used to identify ligand specific binding and autophosphorylation of the HER4 tyrosine kinase (see Section 9., et seq., infra) .
  • CHO/HER2 cells (clone 1-2500) were selected to express high levels of recombinant human pl85 ⁇ rbB2 by dihydrofolate reductase-induced gene amplification in dhfr-deficient CHO cells.
  • the HER2 expression plasmid, cDNeu was generated by insertion of a full length HER2 coding sequence into a modified pCDM ⁇ (Invitrogen, San Diego, CA) expression vector (Seed and Aruffo, 1987, Proc. Natl. Adad. Sci. U.S.A.
  • the cell line 293/HER3 was selected for high level expression of pl ⁇ O- 1 ** 3 .
  • the parental cell line 293 human embryonic kidney cells, constitutively expresses adenovirus Ela and have low levels of EGFR expression. This line was established by cotran ⁇ fection of linearized cHER3 (Plowman et al . , 1990, Proc. Natl. Acad. Sci. U.S.A. 87:4905-09) and pMClneoPolyA (neomycin selectable marker with an Herpes simplex thymidine kinase promoter, Stratagene) , with selection in DMEM/F12 media containing 500 ⁇ g/ml G418.
  • Cells were plated in 6-well tissue culture plates (Falcon), and allowed to attach at 37° C for 18-24 hr. Prior to the assay, the cells were changed to serum- free media for at least 1 hour. Cell monolayers were then incubated with the amounts of ligand preparations indicated in Section 7.3., below for 5 min at 37° C.
  • PBSTDS phosphatase inhibitors
  • phosphatase inhibitors 10 mM NaHP04, 7.25, 150 mM NaCl, 1% Triton X-100, 0.5% deoxycholate, 0.1% SDS, 0.2% sodium azide, 1 mM NaF, 1 mM EGTA, 4 mM sodium orthovanadate, 1% aprotinin, 5 mg/ml leupeptin
  • EGFR were able to stimulate tyrosine phosphorylation of EGFR expressed in recombinant NIH3T3 cells (for EGF, see FIG. 10, Panel 3, lane 2), but not HER4, HER2, or HER3 expressed in recombinant CHO or 293 cells (FIG. 10, Panel 1, 2, 4, lanes 2 and 3).
  • the assay identified a HepG2-derived preparation (fraction 17) as a HER4 ligand capable of specifically stimulating tyrosine phoshorylation of HER4 expressed in CHO/HER4 cells alone.
  • HER2, HER3 or HER4 the receptor expression profile of MDA-MB-453 cells offers an excellent indicator for morphologic differentiation inducing activity.
  • This cell line is known to express HER2 and HER3, but contains no detectable EGFR.
  • the results of the semi- quantitative PCR assays indicated high level expression of HER4 in MDA-MB-453 cells.
  • cDNA encoding the prototype HER4 polypeptide of the invention was first isolated from this cell line (Section 6., supra) .
  • MDA-MB-453 cells (7500/well) were grown in 50 ml DMEM supplemented with 5% FBS and lx essential amino acids. Cells were allowed to adhere to 96-well plates for 24 hr. Samples were diluted in the above medium, added to the cell monolayer in 50 ml final volume, and the incubation continued for an additional 3 days. Cells were then examined by inverted light microscopy for morphologic changes.
  • Serum free media from a panel of cultures of human cancer cells were screened for growth regulatory activity on MDA-MB-453 cells.
  • a human hepatocarcinoma cell line, HepG2 was identified as a source of a factor which induced dramatic morphologic differentiation of the MDA-MB-453 cells.
  • Section 10.1.1., supra was used throughout the purification procedure to monitor the column fractions that induce morphological changes in MDA-MB-453 cells.
  • HepG2 cells were cultured in DMEM containing 10% fetal bovine serum using Nunc cell factories. At about 70% confluence, cells were washed then incubated with serum-free DMEM.
  • Conditioned medium HepG2-CM was collected 3 days later, and fresh serum-free medium added to the cells. Two additional harvests of HepG2- CM were collected per cell factory. The medium was centrifuged and stored at -20° C in the presence of 500 mM PMSF.
  • HepG2-CM Ten litres of HepG2-CM were concentrated 16-fold using an Amicon ultrafiltration unit (10,000 molecular weight cutoff membrane) , and subjected to sequential precipitation with 20% and 60% ammonium sulfate. After centrifugation at 15,000 x g, the supernatant was extensively dialyzed against PBS and passed through a DEAE-sepharose (Pharmacia) column pre- equilibrated with PBS. The flow-through fraction was then applied onto a 4 ml heparin-acrylic (Bio-Rad) column equilibrated with PBS. Differentiation inducing activity eluted from the heparin column between 0.4 and 0.8 M NaCl. Active heparin fractions
  • PY20 horseradish peroxidase-conjugated goat F(ab')2 anti-mouse Ig (Cappell) and chemiluminescence were used for detection. Phosphorylation signals were analyzed using the Molecular Dynamics personal densitometer.
  • Semi-purified HepG2-derived factor demonstrated a capacity to induce differentiation in MDA-MB-453 cells (FIG. 11, Panel 1-3) .
  • untreated MDA-MB-453 cells are moderately adherent and show a rounded morphology (FIG. 11, Panel 1) .
  • the addition of semi-purified HepG2-derived factor induces these cells to display a noticeably flattened morphology with larger nuclei and increased cytoplasm (FIG. 11, Panel 2 and 3).
  • This HepG2-derived factor preparation also binds to heparin, a property which was utilized for purifying the activity.
  • FIG. 11 Panel 4 shows the phenyl column elution profile. Tyrosine phosphorylation assays of the phenyl column fractions revealed that the same fractions found to induce
  • PBMP-25946.1 differentiation of the human breast carcinoma cells are also able to stimulate tyrosine phosphorylation of a 185 kDa protein in MDA-MB-453 cells (FIG. 11, Panel 5).
  • fraction 16 induced a 4.5-fold increase in the phosphorylation signal compared to the baseline signal observed in unstimulated cells, as determined by densitometry analysis (FIG. 11, Panel 6).
  • Adjacent fraction 14 was used as a control and had no effect on the phosphorylation of any of the EGFR- family receptors (FIG. 10, Panel 1-4, lane 5). Further purification and analysis of the factor present in fraction 17 indicates that it is a glycoprotein of 40 to 45 kDa, approximately the same size as NDF and HRG.
  • the HepG2-derived factor also has functional properties similar to NDF and HRG, inasmuch as it stimulates tyrosine phosphorylation of HER2/pl85 in MDA-MB-453 cells, but not EGFR in NR5 cells, and induces morphologic differentiation of HER2 overexpressing human breast cancer cells.
  • HepG2-derived factor to stimulate phosphorylation of MDA-MD-453 cells a cell line known to overexpress HER2 and HER3 and the source from which HER4 was cloned. Since EGFR and HER2 have been shown to act 5 synergistically, it is conceivable that HER4 may also interact with other EGFR-family members. In this connection, these results suggest that NDF may bind to HER4 in MDA-MB-453 cells resulting in the activation of HER2. The results described in Section 10., 0 immediately below, provide evidence that NDF interacts directly with HER4, resulting in activation of HER2.
  • rat NDF cDNA was isolated from o normal rat kidney RNA and inserted into a cDM8-based expression vector to generate cNDFl.6.
  • This construct was transiently expressed in COS cells, and conditioned cell supernatants were tested for NDF activity using the tyrosine kinase stimulation assay 5 described in Section 8.2., supra.
  • Supernatants from cNDFl.6 transfected cells upregulated tyrosine phosphorylation in MDA-MB-453 cells relative to mock transfected COS media FIG. 12, Panel 1. Phosphorylation peaked 10-15 minutes after addition on 0 TM*'
  • the NDF preparation had no effect on phosphorylation of EGFR, or HER2 containing cells, but induced a 2.4 to 4 fold increase in tyrosine
  • PKMP-25946 phosphorylation of HER4 after 15 minutes incubation (see FIG. 12, Panel 2).
  • a HER4 cDNA probe corresponding to the 5' portion of the gene was used for in situ hybridization mapping of the HER4 gene.
  • in situ hybridization to metaphase chromosomes from lymphocytes of two normal male donors was conducted using the HER4 probe labeled with 3 H to a specific activity of 2.6 x 10 7 cpm/ ⁇ g as described (Marth et al . , 1986, Proc. Natl. Acad. Sci. U.S.A. 83:7400-04). The final probe concentration was 0.05 ⁇ g/ ⁇ l of hybridization mixture. Slides were exposed for one month. Chromosomes were identified by Q banding.
  • Results A total of 58 metaphase cells with autoradiographic grains were examined. Of the 124 hybridization sites scored, 38 (31%) were located on the distal portion of the long arm of chromosome 2 (FIG. 13). The greatest number of grains (21 grains) was located at band q33, with significant numbers of
  • CHO cells expressing recombinant HER4 or HER2 were generated as previously described in Section 8.
  • MDA-MB453 were seeded in 24 well plates and cultured
  • Biochemicals or anti-HER2 Mab (c-neu Ab-2, Oncogene
  • Recombinant rat heregulin was produced as follows. A 1.6 kb fragment encoding the entire open reading frame of rat heregulin (and 324 bp of 5'- untranslated sequence) was obtained by PCR using normal rat kidney RNA as a template. This fragment was inserted into a CDM8-based expression vector
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS Dulbecco's Modified Eagle Medium
  • Clarified conditioned medium was either used directly or was dialyzed against 0.1 M acetic acid for 2 days, dried, and resuspended as a 20-fold concentrate in DMEM.
  • HER Tyrosine Phosphorylation As shown in FIG. 15, recombinant heregulin induces tyrosine phosphorylation of HER4. Tyrosine phosphorylated receptors were detected by Western blotting with an anti-phosphotyrosine Mab a, Monolayers of MDA-MB453 or CHO/HER4 cells were incubated with media from COS-1 cells transfected with a rat heregulin expression plasmid (HRG) , or with a cDM8 vector control (-) . The media was either applied directly (lx) or after concentrating 20-fold (20x, and vector control) . Solubilized cells were immunoprecipitated with anti-phosphotyrosine Mab.
  • HRG rat heregulin expression plasmid
  • - cDM8 vector control
  • HER4 is involved in signaling by heregulin
  • recombinant rat heregulin to stimulate tyrosine phosphorylation in a panel of Chinese hamster ovary (CHO) cells that ectopically express human HER2 or HER4 was examined.
  • the activity of recombinant heregulin was first confirmed by its ability to stimulate differentiation of human breast cancer cells (data not shown) and to
  • FSKP-25946.1 induce tyrosine phosphorylation of a high molecular weight protein in MDA-MB453 cells (FIG. 15, Panel 1).
  • Heregulin had no effect on CHO cells expressing only HER2 (FIG. 15, Panel 3) , yet these cells were shown to have a functional receptor since their tyrosine kinase activity could be stimulated by either of two antibodies specific to the extracellular domain of HER2 (FIG. 15, Panel 3).
  • heregulin was able to induce tyrosine phosphorylation of a 180K protein in CHO cells expressing HER4 (FIG. 15, Panel 2) .
  • Species differences in ligand-receptor interactions have been reported for EGF receptor (Lax et al .
  • rat heregulin does not directly interact with rat HER2/neu (Peles et al . , supra) .
  • rat, rabbit, and human heregulin share high sequence homology and have been shown to induce tyrosine phosphorylation in their target cells of human origin (Wen D. et al . , supra; Holmes et al . , supra; and Falls et al . , supra) .
  • CNHER2 and CNHER4 expression plasmids were generated by insertion of the complete coding sequences of human HER2 and HER4 into cNEO, an expression vector that contains an SV2-NEO expression unit inserted at a unique BamHI site of CDM8. These constructs were linearized and transfected into CEM cells by electroporation with a Bio-Rad Gene Pulser apparatus essentially as previously described (Wen et al . , supra) . Stable clones were selected in RPMI/10%
  • HER2 immunoprecipitations were as described in FIG. 15, using 5 x 10 6 cells per reaction, and the HER2 Western blots were performed with a second anti-HER2 Mab (c-neu Ab-3, Oncogence Sciences).
  • MEM methionine and cysteine-free Minimal Essential Medium
  • Lysates were then incubated for 6 h, 4 * C with 3 ⁇ l each of two rabbit antisera raised against synthetic peptides corresponding to two regions of the cytoplasmic domain of human HER4 (• « _ _RLLEGDEKEYNADGG M [SEQ ID No:31] and
  • FIG. 16 Expression of recombinant HER2 and HER4 in human CEM cells is shown in FIG. 16. Transfected CEM cells were selected that stably express either HER2, HER4, or both recombinant receptors. In FIG. 16, Panel l.
  • recombinant HER2 was detected by immunmoprecipitation of cell lysates with anti-HER2 Mab (Ab-2) and Western blotting with another anti-HER2 Mab (Ab-3) .
  • Ab-2 anti-HER2 Mab
  • Ab-3 Western blotting with another anti-HER2 Mab
  • Panel 2 recombinant HER4 was detected by immunoprecipitation of 3S S-labeled cell lysates with HER4-specific rabbit anti-peptide antisera.
  • Panel 3 CEM cell lines were selected that express one or both recombinant receptors and aliquots of each were incubated with media control (-) , with two HER2-stimulatory Mabs (Mab 28 and 29) , or with an isotype matched control Mab (18.4).
  • Solubilized cells were immunoprecipitated with anti-HER2 Mab (Ab-2) and tyrosine phosphorylated HER2 was detected by Western blotting with an anti-phosphotyrosine Mab.
  • the size in kilodaltons of prestained high molecular weight markers (Bio-Rad) is shown on the left and arrows indicate the HER2 and HER4 proteins.
  • Example 12 supports the earlier observation that HER2 alone is not sufficient to transduce the heregulin signal.
  • a panel of human CEM cells that express the recombinant receptors either alone or in combination was established.
  • the desired model system was of human origin, since many of the reagents against erbB family members are specific to the human homologues.
  • CEM cells are a human T lymphoblastoid cell line and were found to lack expression of EGF receptor, HER2, HER3, or HER4, by a variety of immunologic, biologic, and genetic analyses (data not shown) .
  • FIG. 16 demonstrates the selection of three CEM cell lines that express only HER2 (CEM 1-3), only HER4 (CEM 3-13), or both HER2 and HER4 (CEM 2-9). The presence of a functionally and structurally intact
  • HER2 in the appropriate cells was confirmed by the induction of HER2 tyrosine phosphorylation by each of the two antibodies specific to the extracellular domain of HER2, but not by an isotype matched control antibody (FIG. 16, Panel 3).
  • Recombinant rat heregulin was prepared as in FIG. 15, and diluted to 7x in RPMI.
  • the HER4-specific Mab was prepared by immunization of mice with recombinant HER4 (manuscript in preparation) .
  • CEM cells (5 x 10 6 ) were treated with the concentrated supernatants for 10 min, room temperature and precipitated with PY20 or anti-HER2 Mab (Ab-2) as described in FIG. 15.
  • Immunoprecipitation with anti-HER4 Mab was performed by incubation of cells lysates with a 1:5 dilution of hybridoma supernatent for several hours followed by 2 ⁇ g rabbit anti-mouse Ig (cappel) and Protein A Sepharose CL-4B (Pharmacia) . PY20 Westerns as described in FIG. 15.
  • heregulin induces tyrosine phosphorylation in CEM cells expressing HER4.
  • CEM cell lines that express either HER2 or HER4 alone (CEM 1-3 and CEM 3-13) or together (CEM 2-9) were incubated with 7x concentrated supernatants from mock- (-) or heregulin-transfected (+) COS-1 cells. Solubilized cells were immunoprecipitated (IP) with anti-phosphotyrosine Mab (PY20) (FIG. 17, Panel 1); HER2-specific anti-HER2 Mab (Ab-2) (FIG. 17, Panel 2); or HER4-specific Mab (6-4) (FIG. 17, Panel 3) .
  • IP immunoprecipitated
  • the panel of CEM cells were then analyzed by phosphotyrosine Western blots of cells lysates following treatment with heregulin and immunoprecipitation with three different monoclonal antibodies (Mabs) .
  • Precipitation with an anti- phosphotyrosine antibody (PY20) again demonstrates that heregulin is able to stimulate tyrosine phosphorylation in cells expressing HER4, but not in cells expressing only HER2 (FIG. 17, Panel 1).
  • precipitation with an antibody specific to the extracellular domain of HER2 demonstrates that HER2 is tyrosine phosphorylated in response to heregulin in cells that co-express HER4 (FIG. 17, Panel 2).
  • recombinant heregulin was produced as an epitope-tagged fusion with amphiregulin.
  • the 63 amino acid EGF-structural motif of rat heregulin (Wen et al . , supra) from serine 177 to tyrosine 239 was fused to the N-terminal 141 amino
  • Purified heregulin was iodinated with 250 ⁇ Ci of X 5 I- labeled Bolton-Hunter reagent (NEN) . CH0/HER4 or CHO/HER2 cells were incubated with 125 I-heregulin (10 s - cpm) for 2 h at 4° C. Monolayers were washed in PBS and 3 mM Bi ⁇ ( ⁇ ulfosuccinimidyl) suberate (BS 3 , Pierce) was added for 30 min on ice. The cells were washed in tris-buffered saline, dissolved in SDS sample buffer, run on a 7% polyacrylamide gel, and visualized on the phosphorimager.
  • heregulin induces tyrosine phosphorylation of HER4 in the absence of HER2. In contrast, heregulin does not directly stimulate HER2. However, in the presence of HER4, heregulin induces phosphorylation of HER2, presumably either by transphosphorylation or through receptor heterodimerization. Together, these experiments suggest that HER4 is the receptor for heregulin. Most breast cancer cells that overexpress HER2 have been shown to be responsive to heregulin, whereas HER2-positive ovarian and fibroblast lines do not respond to the ligand. This observation could be explained by the fact that HER4 is co-expressed with HER2 in most or all of the breast cancer cell lines studied, but not in the ovarian carcinomas. Furthermore, overexpression of HER2 in heregulin- responsive breast cancer cells leads to increased binding, whereas expression of HER2 in heregulin- unresponsive ovarian or fibroblast cells has no effect (Peles et al . , supra) .
  • Northern and in situ hybridization analyses localizes HER4 to the white matter and glial cells of the central and peripheral nervous system, as well as to cardiac, skeletal, and smooth muscle. This distribution is consistent with HER4 being involved in signaling by the neurotropic factors, GGF, and ARIA. Recognition of HER4 as a primary component of the heregulin signal transduction pathway will assist in deciphering the molecular mechanisms that results in its diverse biologic effects.
  • HepG2 cells were obtained from Dr. S. Radka and cultured in 10% fetal 0 bovine serum containing DMEM. For large scale production of serum-free conditioned medium, HepG2 cells were propagated in Nunc cell factories. Chinese hamster ovary cells (CHO-KI) expressing high levels of either recombinant human pl ⁇ - 1 * 82 (CH0/HER2) or 5 recombinant human pl80* rbB4 (CHO/HER4) were generated and cultured as described in Section 8.
  • DMEM Dulbecco's modified Eagle's medium
  • fetal bovine serum and amino acids Life Technologies, Inc.
  • HepG2 cells were obtained from Dr. S. Radka and cultured in 10% fetal 0 bovine serum containing DMEM.
  • Nunc cell factories Chinese hamster ovary cells (CHO-KI) expressing high levels of either recombinant human pl ⁇ - 1 * 82 (CH0/HER2) or 5 recombinant human pl80* rb
  • N29 monoclonal antibody to the extracellular portion of the human HER2 receptor was a gift from Dr. Y. Yarden.
  • Ab-3 c- neu monoclonal antibody that reacts with the human 0 pi85* ri ⁇ 2 was from Oncogene Science Inc.
  • MDA-MB-453 human breast cancer cells overexpress 5 pl85 ⁇ rtB2 but do not express the EGFR at their surface
  • HepG2-CM Medium conditioned by HepG2 cells (HepG2-CM, 60 liters) was concentrated 26-fold using an Amicon ultrafiltration unit (10,000 molecular weight cutoff _ membranes) and then subjected to 50% ammonium sulfate ((NH «) 2 S04 4 ) precipitation. After centrifugation at
  • Active fractions were pooled, concentrated, dialyzed against PBS, and then applied (three separate runs) to a DEAE- Sepharose column (2.5 x 25 cm, Pharmacia) equilibrated with PBS, pH 7.3. The flow rate was 1 ml/min. The column flow-through was then loaded (two separate runs) on a CM-Sepharose Fast Flow column (2.5 x 13.5 cm, Pharmacia) pre-equilibr ted with PBS, pH 7.3. Proteins were eluted at 1 ml/min. with a 330-ml gradient from PBS to 1 M NaCl in PBS. Fractions of 5 ml were collected.
  • the active material was loaded on a TSKgel heparin-5PW HPLC column (7.5 x 75 mm, TosoHaas) equilibrated with PBS. The flow rate was 0.5 ml/min. A 50-ml linear NaCl gradient (PBS to 2 M in PBS) followed by an isocratic elution with 2 M NaCl was used to elute the bound proteins. Fractions' of l ml were collected. Active fractions corresponding to the 1.3 M NaCl peak of protein were pooled and concentrated. A Protein Pak SW-200 size exclusion chromatography column (8 x 300 mm.
  • the membrane was blocked for 2 h at room temperature with 6% hovine serum albumin in 10 mM Tria-HCl, pH 8.0, 150 mM NaCl, 0.05% Tween 20.
  • PY20 monoclonal anti-phosphotyrosine antibody (ICN, 2 h at 22° C) and horseradish peroxidase-conjugated goat anti- mouse IgG F(ab') 3 (Cappel, lh at 22° C) were used as primary and secondary probing reagents, respectively. Proteins phosphorylated on tyrosine residues were detected with a chemiluminescence reagent (Amersham Corp.).
  • CHO/HER2 stimulation Assay CH0/HER2 cells were seeded in 24-well plates at 1 x 10 s cells/well and cultured 24 h. Monoclonal antibody N29 specific to the extracellular domain of p 185 . r __ 2 (stancovski et al . , 1991, PNAS 88:8691-8695) was added at 25 ⁇ g/ml. Following a 20-min. incubation at room temperature, media were removed and cells were solubilized for 10 min.
  • PSMP-25946 extracts were incubated for 2 h at 4° C with an antip- 185 ⁇ r _ B2 an tibody (Ab-3 c-neu, Oncogene Science Inc.). Rabbit anti-mouse IgG (Cappel) and protein A-Sepharose were then added, and samples were incubated an additional 30 min. Immune complexes were washed 3 times with PBS-TDS, resolved on a 7% polyacrylamide gel, and electrophoretically transferred to nitrocellulose. Phosphorylation of the receptor was assessed by Western blot using a 1:1000 dilution of PY20 phosphotyrosine primary antibody (ICN
  • HPLC-purified p45 (1.5 ⁇ g) was iodinated with 250 ⁇ Ci of " 4 1-labeled Bolton-Hunter reagent obtained from Du Pont-New England Nuclear. 125 I-p45 was purified by filtration through a Pharmacia PD-10 column. The specific activity was 10 4 cpm/ng. 1 S I-p45 retained its biological activity as confirmed in a differentiation assay as well as a kinase stimulation assay (data not shown) . Binding of radiolabeled p45 was performed on 2 x 10 s CHO/HER4 cells and 4 X 10 s CHO-KI or CHO/HER2 cells in 12-well plates.
  • Cell monolayers were washed twice with 1 ml of ice-cold binding buffer (DMEM supplemented with 44 mM sodium bicarbonate, 50 mM BES [N-, N-Bis (2-hydroxyethyl) -2-aminoethan-sulfonic acid], pH 7.0, 0.1% bovine serum albumin) and then incubated on ice for 2 h with 50 ng/ml 12S I-p45 in the absence or the presence of 250 ng/ml unlabeled p45. The monolayers were washed twice with PBS and then incubated in the presence of 1 mM bis( ⁇ ulfosuccinimidyl)suberate (BS 3 , Pierce) in PBS for 45 min. on ice. Supernatants were discarded, and the reaction was quenched by adding 0.2 M glycine in PBS.
  • DMEM ice-cold binding buffer
  • BES N-, N-Bis (2-hydroxyethyl) -2
  • CH0/HER2 cells (2 x 10 s cells/well) were seeded in 24- well plates. After 48 h, cells were washed with binding buffer and then incubated with increasing concentrations of 15 I-p45. Nonspecific binding was determined in the presence of excess unlabeled p45. After a 2-h incubation at 4° C, the cells were washed three times with binding buffer and then lysed in 500 ⁇ l of 0.5M NaOH, 0.1% SDS. Cell-associated radioactivity was determined by using a ⁇ -counter. Scatchard analysis was performed using the computerized LIGAND program (Munson and Rodbard, 1980, Anal. Biochem 107:220-239).
  • N-terminal Amino Acid Sequence The N-terminal sequence analysis of p45 (25 pmol) was performed as previously described (Shoyab et al . , 1990, Proc. Natl. Acad. Sci. 87:7912-7916).
  • PBMP-25946.1 active material (1010 mg of protein) was loaded on a phenyl-Sepharose column (FIG. 19, Panel 1). Column fractions 40-85 (348 mg of protein eluting between 1M ammonium sulfate and 0M ammonium sulfate) were found to induce morphological changes in MDA-MB-453 cells.
  • the biologically active column flow-through (174 mg of protein) was subjected to a cation-exchange chro otography (FIG. 19, Panel 2) with activity eluting between 0.35 and 0.48 M NaCl.
  • the active fractions were pooled (1.5 mg of protein) and applied to an analytical heparin column (FIG. 19, Panel 3).
  • the differentiation activity eluted from the heparin column between 0.97 and 1.45 M NaCl fractions 27-38.
  • Size exclusion chromatography of the heparin column fractions 35-38 achieved a homogeneous preparation of the human breast cancer cell differentiation factor.
  • a major protein peak eluted with a molecular weight greater than 70,000 (FIG. 19, Panel 4).
  • Fractions 30 and 32 assayed at 30 ng/ml confirmed the bioactivity of this protein with phenotypic changes being apparent after 24 hours.
  • SDS-PAGE analysis of these column fractions followed by silver staining of the gel showed that the biologically active peak contained a single protein migrating around 45 kDA (FIG. 20) .
  • the faint 67 kDa band corresponds to a staining artifact, as evidenced by the left lane of the gel, which contained no sample.
  • the amount of pure protein recovered in fractions 30-33 was estimated to be 6 micrograms.
  • the difference in the molecular weight estimated by size exclusion chromatography and SDS- PAGE indicates that this protein may form dimers or oligomers under non-denaturing conditions.
  • FIG. 21, Panel 1 shows the stimulatory effect of sequential fractions from the size exclusion chromatography column on tyrosine phosphorylation in MDA-MB-453 cells. Densitometric analysis of the autoradiogram revealed that fractions 30-34 were essentially equipotent. Homogeneously purified p45 specifically stimulated tyrosine phosphorylation of pl80 « * ⁇ 4 (FIG. 21, Panel 2). p45 was not able to stimulate phosphorylation in CHO/HER2 cells, and the cell were found to express functional pl85 ⁇ rtB2 receptor as evidenced by immunoreactivity with 5 monoclonal antibodies specific to different regions of pl85 ⁇ rtB2 . p45 has an N-terminal amino acid sequence similar to the recently isolated pl85 ⁇ rbB2 ligand.
  • Binding and cross-linking studies were performed in order to confirm that p45 was able to bind to pl ⁇ o**** 4 . Binding studies revealed that while no
  • the 45 kDa band represents uncross-linked yet pl80 ⁇ rbB4 associated 125 I- p45.
  • the 210 kDa band corresponds to the p45-pl80 ⁇ rbB4 complex (assuming an equimolar stoichiometry of ligand and receptor) , whereas the high molecular weight band is presumed to be a dimerized form of the receptor- ligand complex.
  • the 100 kDa band could represent a truncated portion of the extracellular domain of the pi80 #rtB4 receptor complexed to 125 I-p45 or a covalently associated p45 dimer.
  • the c-kit ligand provides precedence for cross-linked dimers (Williams et al . , 1990, Cell 63:167-174).
  • the HER4 ligand, p45 purified from medium conditioned by HepG2, induces differentiation of breast cancer cells and activates tyrosine phosphorylation of a 185 kDa protein in MDA-MB-453 cells.
  • p45 is not capable of directly binding to pl85 ⁇ rt ⁇ 2 but shows specificity to HER4/pl80 ,rtB4 .
  • Heregulin 02-Ig and the mouse monoclonal antibody directed against the Pseudomonas exotoxin (PE) was supplied by Dr. J.-M. Colusco and by Dr. Tony Siadek, respectively (Bristol-Myers-Squibb, Seattle, WA) .
  • the cell lines BT474, MDA-MB-453, T47D, SKBR-3, and MCF-7 (all breast carcinoma) , LNCaP (prostate carcinoma) , CEM (T-cell leukemia) and SKOV3 (ovarian carcinoma) were obtained from ATCC (Rockville, MD) .
  • the H3396 breast carcinoma cell line and the L2987 lung carcinoma cell line were established at Bristol-Myers- Squibb (Seattle, WA) .
  • the AU565 breast carcinoma cell line was purchased from the Cell Culture laboratory. Naval Biosciences Laboratory (Naval Supply Center, Oakland, CA) . All cell lines were of human origin.
  • BT474 and T47D cells were cultured in IMDM supplemented with 10% fetal bovine serum (FBS) and 10 ⁇ g/ml insulin.
  • MCF-7, H3396, LNCaP and L2987 were cultured in IMDM supplemented with 10% FBS.
  • SKBR3 and SKOV3 cells were grown in McCoys media supplemented with 10% FBS and 0.5% non-essential amino acids.
  • AU565 cells were cultured in RPMI 1640 media supplemented with 15% FBS and CEM transfectants (see section 15.1.5., infra) were cultured in RPMI 1640 supplemented with 10% FBS and 500 ⁇ g/ml G418.
  • Rat heregulin cDNA (Wen et al . , 1994, Mol. Cell. ____2l_.14:1909-1919) was isolated by RT-PCR using mRNA from rat kidney cells as template. The cDNA was
  • P-MP-25946 prepared in chimeric form with the AR leader sequence by a two-step PCR insertional cloning protocol using cARP (Plowman et al . , 1990, Mol. Cell. Biol. 10:1969- 1981) as template to amplify the 5' end of the chimeric ligand using the oligonucleotide primers CARP5: (5'-CGGAAGCTTCTAGAGATCCCTCGAC-3') [SEQ ID No:34] and
  • ANSHLIK2 (3'CCGCACAC 'lTATGTGTTGGCTTGTGTTTCTTCTATTTTTTCCA TTTTTG-5') [SEQ ID No:35].
  • the EGF-like domain PCR was amplified from cNDFl.6 (Plowman et al . , 1993, Nature 366:473-475) using the oligonucleotide primers ANSHLIKl:
  • XNDF1053 (3'-GTCTCTCTAGATTAGTAGAGTTCCTCCGCTTTTTCTTG-5') [SEQ ID No:37].
  • the products were combined and reamplified using the oligonucleotide primers CARP5 and XNDF1053.
  • HAR la ⁇ eregulin-amphi__egulin construct
  • NARPl (5'-GTCAGAGTTCATATGGTAGTTAAGCCCCCCCAAAAC-3') [SEQ ID No:38] and NARP4:
  • the resulting 287 bp DNA fragment was digested with Nde I and Hind III, followed by ligation into the compatibly digested expression plasmid pBW 7.0 which contained, in frame at the 5' fusion site, the nucleotide sequence encoding for of PE40 (Friedman et al . , 1993, Cancer Res. 53:334-339).
  • the resulting expression plasmid pSE 8.4 then contained the gene fusion encoding the chimeric heregulin-toxin protein, under the control of a IPTG-inducible T7 promoter.
  • the plasmid pSE 8.4 encoding the chimeric protein HAR-TX ⁇ 2 was transformed into the ⁇ . coli strain BL21 ( ⁇ DE3) .
  • Cells were grown by fermentation in T broth containing 100 ⁇ g/ml ampicillin at 37°C to a optical density of A 650 ⁇ 4.8, followed by induction of protein expression with 1 mM isopropyl-l-thio-3-D- galactopyrano ⁇ ide (IPTG) . After 90 minutes the cells were harvested by centrifugation.
  • IPTG isopropyl-l-thio-3-D- galactopyrano ⁇ ide
  • the cell pellet was frozen at -70°C, then thawed and resuspended at 4°C in solubilization buffer (50 mM Tris-HCl (pH 8.0), 10 mM EDTA, 1 ug/ml leupeptin, 2 ug/ l aprotinin, 1 ug/ml pepstatin-A, 0.5 mM PMSF) containing 1% tergitol by homogenization and sonication.
  • solubilization buffer 50 mM Tris-HCl (pH 8.0), 10 mM EDTA, 1 ug/ml leupeptin, 2 ug/ l aprotinin, 1 ug/ml pepstatin-A, 0.5 mM PMSF
  • the resulting pellet containing pre-purifled inclusion bodies was dissolved in 6.5 M guanidine-HCl , 0.1 M Tris-HCl (pH 8.0), 5 mM EDTA; sonicated; and refolded by rapid dilution (100-fold) into 0.1 M Tris- HCl (pH 8.0), 1.3 M urea, 5 mM EDTA, 1 mM glutathione,
  • HAR-TX ⁇ 2 protein was diluted 2-fold with 50 mM sodium phosphate (pH 7.0) and applied to a cation-exchange resin (POROS 50 HS, PerSeptive Biosystems, Cambridge, MA) , pre- equilibrated in the same buffer.
  • POROS 50 HS PerSeptive Biosystems, Cambridge, MA
  • the HAR-TX ⁇ 2 protein was eluted with a 450 nM NaCl step gradient in 50 mM sodium phosphate (pH 7.0) and fractions were analyzed using SDS-PAGE and Coomassie blue staining. Final purification of pooled fractions was performed by chromatography using Source 15S cation-exchange media (Pharmacia, Uppsala, Sweden) equilibrated with 50 mM sodium phosphate (pH 6.0). Chimeric HAR-TX ⁇ 2 protein was eluted with a gradient of 0-1 M NaCl in the same buffer and analyzed by SDS-PAGE.
  • Membranes from 5 x 10 7 MDA-MB-453 cells were prepared and coated to 96 well plates as previously described for H3396 human breast carcinoma cells (Siegall et al . , 1994, J. Immunol. 152:2377-2384). Subsequently, the membranes were incubated with titrations of either HAR-TX ⁇ 2 or PE40 ranging from 0.3 - 300 ug/ml and the mouse monoclonal anti-PE antibody EXA2-1H8 as the secondary reagent (Siegall et al . , supra) . The isolate of the toxin portion PE40 alone was used to determine unspecific binding activity to the membrane preparations, in comparison with the specific binding activity of HAR-TX ⁇ 2.
  • EGF-R family (1-5 x 10 6 cells) were stimulated with 500 ng/ml HAR-TX ⁇ 2 for 5 minutes at room temperature.
  • the cells were pelleted and resuspended in 0.1 ml lysis buffer (50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 5 mM MgCl 2 , 1% NP40, 0.5% deoxycholate, 0.1% sodium dodecylsulfate, 1 mM sodium orthovanadate) at 4°C.
  • 0.1 ml lysis buffer 50 mM Tris-HCl, pH 7.4, 150 mM NaCl, 5 mM MgCl 2 , 1% NP40, 0.5% deoxycholate, 0.1% sodium dodecylsulfate, 1 mM sodium orthovanadate
  • cytotoxicity assays For cytotoxicity assays, tumor cells (10 s cells/ml) in growth medium were added to 96-well flat bottom tissue culture plates (0.1 ml/well) and incubated at 37°C for 16 h. Cells were incubated with HAR-TX ⁇ 2 for 48 h at 37°C, washed twice with phosphate buffered saline (PBS) , followed by addition of 200 ⁇ l/well of 1.5 ⁇ M calcein-AM (Molecular Probes Inc., Eugene, OR) .
  • PBS phosphate buffered saline
  • HER4 expressed in baculovirus, was used as the immunogen for subcutaneous injection into 4-6 week old female BALB/c mice. Immunization was performed 4 times (approximately 1 month apart) with 20 ⁇ g of HER4 protein given each time. Spleen cells from immunized mice were removed four days after the final immunization and fused with the mouse myeloma line P2x63-Ag8.653 as previously described (Siegall et al . , supra) . Positive hybridoma supernatants were selected by ELISA screening on plates coated with HER4 transfected CHO cells (Plowman et al., 1993, Nature 366:473-475) and selected against parental CHO cells and human fibroblasts.
  • PBMP-25946.1 also selected and found to bind to the cytoplasmic domain of HER4.
  • HER2 staining was performed by using mouse anti-HER2 mAb 24.7 (Stancovski et al . , 1991, Proc. Natl. Acad. Sci. USA 88:8691-8695) as primary, and biotinylated goat anti-mouse IgG (Jackson Labs, West Grove, PA) as secondary antibody as previously described (Bacus et al . , 1993, Cancer Res. 53:5251- 5261) .
  • the primary antibodies used were, respectively, mouse anti-HER3 mAb RTJ2 (Santa Cruz Biotech, Santa Cruz, CA) at 2.5 ⁇ g/ml concentration or mouse anti-HER4 mAb 6-4-11 at 15 ⁇ g/ml concentration followed by incubation wi_h biotinylated rabbit anti-mouse IgG (Zymed Labs, South San Francisco, CA) .
  • the staining procedure was performed at RT as follows: cells were fixed in 10% neutral buffered formalin for 60 minutes, washed with H 2 0 and rinsed with Tris buffered saline (TBS; 0.05 M Tris, 0.15 M NaCl, pH 7.6). Unspecific binding sites were blocked by incubation with 10% goat serum (for HER2) or rabbit serum (for HER3 and HER4) in 0.1% bovine serum albumin/TBS for 15 minutes. Subsequently, cells were incubated with primary and secondary antibodies for 30 and 20 minutes, respectively, followed by incubation with alkaline phosphatase conjugated streptavidin
  • the HAR-TX ⁇ 2 expression plasmid encoding the hydrophilic leader sequence from amphiregulin (AR) , heregulin ⁇ 2 , and PE40, under control of the IPTG inducible T7 promoter, was constructed as described in Section 15.1.2., supra , and is diagrammatically shown in FIG. 23, Panel 1.
  • the AR leader sequence was added to the N-terminus of heregulin to facilitate the purification procedure (FIG. 23, Panel 2).
  • FIG. 24A and 24B show the nucleotide sequence and the deduced amino acid sequence of the cDNA encoding HAR-TX ⁇ 2 Chimeric HAR-TX ⁇ 2 protein was expressed in E. coli of inclusion bodies.
  • Recombinant protein was denatured and refolded as described in Section 15.1.2., supra, and applied to cation-exchange chromatography on a POROS HS column.
  • Semi-purified HAR-TX ⁇ 2 protein was detected by PAGE and Cooma ⁇ sie blue staining as major band migrating at 51 kDa (FIG. 25, lane 2) .
  • the column flow-through from POROS HS contained only small amounts of HAR-TX ⁇ 2 (FIG. 25, lane 3) .
  • POROS HS chromatography resulted in >50% purity of HAR-TX ⁇ 2 (FIG. 25, lane 4).
  • HAR-TX ⁇ 2 To determine the ⁇ pecific binding activity of HAR-TX ⁇ 2 , an ELISA assay was performed using membranes of the HER4 positive human breast carcinoma cell line MDA-MB-453 as the target for binding. HAR- TX ⁇ 2 was found to bind to the immobilized cell membranes in a dose-dependent fashion up to 300 ⁇ g/ml (FIG. 26). PE40, the toxin component of HAR-TX ⁇ 2 used as negative control, was unable to bind to MDA- MB-453 membranes.
  • HAR-TX ⁇ 2 a HER4 receptor phosphorylation assay was performed as previously described for heregulin (Carraway et al . , 1994, J. Biol. Chem. 269:14303-14306). CEM cells expressing different HER family members were exposed to HAR-TX ⁇ 2 and stimulation of tyrosine phosphorylation was analyzed by phosphotyrosine v immunoblot analysis (Section 4, supra; Section 15.1.5., supra) . As shown in FIG. 27, HAR-TX ⁇ 2 induced tyrosine phosphorylation in CEM cells expressing HER4 either alone or together with HER2, but not in cells expressing only HER2 or HERl.
  • heregulin moiety in its selective interaction between receptor family members.
  • the cell killing activity of HAR-TX -52 was determined against a variety of human cancer cell lines.
  • AU565 and SKBR3 breast carcinomas and LNCaP prostate carcinoma were sensitive to HAR-TX ⁇ 2 with EC 50 values of 25, 20, 4.5 ng/ml, respectively, while SK0V3 ovarian carcinoma cells were insensitive to HAR- TX ⁇ 2 (EC S0 >2000 ng/ml) (FIG. 28, Panel 1).
  • Addition of heregulin ,52-Ig to LNCaP cells reduced the cytotoxic activity of HAR-TX ⁇ 2 (FIG. 28, Panel 2).
  • L6-Ig a chimeric mouse-human antibody with a non-related specificity but matching human Fc domains (Hellstr ⁇ m et al . , supra)
  • L6-Ig did not inhibit the HAR-TX ⁇ 2 cytotoxic activity (FIG. 28, Panel 2).
  • the cytotoxic effect of HAR-TX ⁇ 2 was due to specific heregulin-mediated binding. Similar data were obtained using MDA-MB-453 cells (not shown) .
  • HAR-TX ⁇ 2 was found to induce tyrosine phosphorylation in both tumor cell types (FIG. 29) similar to that previously seen in COS-7 cells transfected with HER2 and HER3 (Sliwkowski et al . , supra) .
  • SKOV3 cells were found to exhibit the same tyrosine phosphorylation pattern in the presence or absence of heregulin and thus direct interaction between receptors and heregulin could not be established (data not shown) .
  • heregulin does not bind to'these cells (Peles et al . , supra) .
  • HAR-TX ⁇ 2 was administered as described in section 15.1.10. In mice, 2/2 animals died at 2 mg/kg, 2/2 died at 1 mg/kg, 1/2 died at 0.75 mg/kg, and 0/2 died at 0.5 mg/kg, thus the LD S0 is about 0.75 mg/kg (Table V). In rats the determined LD S0 was slightly higher, as 50% of the animals died at 1 mg/kg (Table V) .
  • Cyto cytoplasmic domain
  • BCD extracellular domain
  • FACS fluorescence-activated cell sorter analysis
  • fibro fibroblasts
  • ICC immunocytochemi ⁇ try
  • RIP receptor immunoprecipitation
  • MOLECULE TYPE DNA (genomic)
  • AATTGTCAGC ACGGGATCTG AGACTTCCAA AAA ATG AA3 CCG GCG ACA GGA CTT 54
  • TTT TTA AAC T ⁇ C AGA AAA GAT GGA AAC TTT GGA CTT CAA GAA CTT GGA 438 Phe Leu Asn Tyr ⁇ rg Lys ⁇ p Gly ⁇ sn Phe Gly Leu Gin Glu Leu ⁇ ly 120 125 130 135
  • TGT GTC CAG ATA GCT A ⁇ G GG ⁇ ⁇ TG ⁇ TG T ⁇ C CTG G ⁇ GAA AGA CGA CTC 2550 Cys Val Gin He Ala Lys Gly Met Met Tyr Leu Glu Glu Arg Arg Leu 825 830 835
  • MOLECULE TYPE DNA (genomic)
  • AATTGTCAGC ACGGGATCTG AGACTTCCAA A ⁇ ⁇ T ⁇ ⁇ G CCG GC ⁇ ⁇ C ⁇ ⁇ CTT 54
  • MOLECULE TYPE protein
  • GAG TAT GTC CAC GAG CAC ⁇ G G ⁇ T ⁇ C ⁇ TT GG ⁇ TC ⁇ C ⁇ CT ⁇ CT ⁇ CTT 317 ⁇ lu Tyr Val Hie Glu Hie Lye ⁇ p ⁇ n He Gly Ser ⁇ ln Leu Leu Leu 45 50 v
  • GAG AAA GGA GAA CGT TTG CCT CAG CCT CCC ATC TGC ACT ATT GAC GTT 701 Glu Lys Gly Glu Arg Leu Pro Gin Pro Pro He Cys Thr He Asp Val 170 175 180
  • GTCTG ⁇ TT GAG ⁇ TCC ⁇ TTTCTTTCCC C ⁇ GC ⁇ GTTTC TGTCCT ⁇ C ⁇ ⁇ GT ⁇ TG 2348 ⁇ CC ⁇ CTC ⁇ CTTTC ⁇ T ⁇ T TTAAAAATCT CCATTAAAGT TATAACTAGT ⁇ TT ⁇ TGTTT 2408
  • AAAATAAGAA AGG ⁇ CT ⁇ ⁇ T ⁇ TTTTTC ⁇ TGCTATCAAA TTATCTTC ⁇ C CCTC ⁇ TCCTT 2648
EP95937555A 1994-10-14 1995-10-10 Her4-menslicher rezeptor tyrosine kinase von der epidermalen wachstumsfaktor rezeptor familie Withdrawn EP0787187A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US32344294A 1994-10-14 1994-10-14
PCT/US1995/013524 WO1996012019A2 (en) 1994-10-14 1995-10-10 Her4 human receptor tyrosine kinase or the epidermal growth factor receptor family
US323442 2002-12-18

Publications (1)

Publication Number Publication Date
EP0787187A1 true EP0787187A1 (de) 1997-08-06

Family

ID=23259216

Family Applications (1)

Application Number Title Priority Date Filing Date
EP95937555A Withdrawn EP0787187A1 (de) 1994-10-14 1995-10-10 Her4-menslicher rezeptor tyrosine kinase von der epidermalen wachstumsfaktor rezeptor familie

Country Status (9)

Country Link
EP (1) EP0787187A1 (de)
JP (1) JPH10507362A (de)
AU (1) AU3963295A (de)
CA (1) CA2202533A1 (de)
FI (1) FI971532A (de)
IL (1) IL115642A0 (de)
MX (1) MX9702664A (de)
NO (1) NO971686L (de)
WO (1) WO1996012019A2 (de)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999019488A1 (en) * 1997-10-15 1999-04-22 Children's Medical Center Corporation Novel human egf receptors and use thereof
AU1518799A (en) * 1997-10-31 1999-05-24 Georgetown University Medical Center Erbb-4 targeted ribozymes
JP2006517109A (ja) 2003-02-07 2006-07-20 プロテイン デザイン ラブス インコーポレイテッド アンフィレグリン抗体ならびに癌および乾癬を処置するためのその使用
US8652787B2 (en) 2008-11-12 2014-02-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Use of ERBB4 as a prognostic and therapeutic marker for melanoma

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2103323A1 (en) * 1992-11-24 1994-05-25 Gregory D. Plowman Her4 human receptor tyrosine kinase

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9612019A2 *

Also Published As

Publication number Publication date
AU3963295A (en) 1996-05-06
JPH10507362A (ja) 1998-07-21
WO1996012019A2 (en) 1996-04-25
NO971686L (no) 1997-06-16
WO1996012019A3 (en) 1996-08-15
FI971532A (fi) 1997-06-11
NO971686D0 (no) 1997-04-11
CA2202533A1 (en) 1996-04-25
MX9702664A (es) 1997-06-28
IL115642A0 (en) 1996-01-19
FI971532A0 (fi) 1997-04-11

Similar Documents

Publication Publication Date Title
US5811098A (en) Antibodies to HER4, human receptor tyrosine kinase
AU676502B2 (en) HER4 human receptor tyrosine kinase
US5916755A (en) Methods of characterizing ligands for the erbB-3 receptor, methods of influencing erbB-3 activities and methods of diagnosing erbB-3-related neoplasm
US6696290B2 (en) ErbB2 and ErbB4 Chimeric Heteromultimeric Adhesins
Modjtahedi et al. The receptor for EGF and its ligands-expression, prognostic value and target for therapy in cancer
US8226935B2 (en) ErbB2 and ErbB3 chimeric heteromultimer receptors
AU2014249405B2 (en) Fusion immunomodulatory proteins and methods for making same
ES2230537T3 (es) Una tirosina quinasa de tipo receptor (quinasa de tipo eph/elk hunaba-henki y uso de la misma.
Jeschke et al. Targeted inhibition of tumor‐cell growth by recombinant heregulin‐toxin fusion proteins
Schmidt et al. Targeted inhibition of tumour cell growth by a bispecific single-chain toxin containing an antibody domain and TGF α
WO1993022424A1 (en) RECOMBINANT STIMULATING FACTOR OF THE neu RECEPTOR
US20020002276A1 (en) Chimeric heteromultimer adhesins
CA2354846A1 (en) Method of enhancing the biological activity of ligands
AU2004226162A1 (en) Peptabody for cancer treatment
McGrath et al. Bifunctional fusion between nerve growth factor and a transferrin receptor antibody
EP0828843A1 (de) Rekombinante hereguline und ihre biologische funktionen nach rezeptoraktivierung
EP0787187A1 (de) Her4-menslicher rezeptor tyrosine kinase von der epidermalen wachstumsfaktor rezeptor familie
WO1996012019A9 (en) Her4 human receptor tyrosine kinase or the epidermal growth factor receptor family
QIAN et al. Identification of pl85neu sequences required for monoclonal antibody-or ligand-mediated receptor signal attenuation
Wels et al. Recombinant fusion toxins targeted to members of the ErbB family of receptor tyrosine kinases
Peles The NEU oncogene—Biochemistry of transformation

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19970414

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

17Q First examination report despatched

Effective date: 20020805

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20021216