WO1991008214A1 - Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor - Google Patents

Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor Download PDF

Info

Publication number
WO1991008214A1
WO1991008214A1 PCT/US1990/007025 US9007025W WO9108214A1 WO 1991008214 A1 WO1991008214 A1 WO 1991008214A1 US 9007025 W US9007025 W US 9007025W WO 9108214 A1 WO9108214 A1 WO 9108214A1
Authority
WO
WIPO (PCT)
Prior art keywords
erbb
dna segment
human
dna
gene
Prior art date
Application number
PCT/US1990/007025
Other languages
French (fr)
Inventor
Matthias H. Kraus
Stuart A. Aaronson
Original Assignee
The United States Of America, Represented By The Secretary, United States Department Of Commerce
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 The United States Of America, Represented By The Secretary, United States Department Of Commerce filed Critical The United States Of America, Represented By The Secretary, United States Department Of Commerce
Priority to EP91900108A priority Critical patent/EP0502927B1/en
Priority to DE69032133T priority patent/DE69032133T2/en
Priority to ES91900108T priority patent/ES2113877T3/en
Priority to CA002069900A priority patent/CA2069900C/en
Priority to DK91900108.1T priority patent/DK0502927T3/en
Priority to JP50088591A priority patent/JP3196079B2/en
Priority to AU68899/91A priority patent/AU654805B2/en
Publication of WO1991008214A1 publication Critical patent/WO1991008214A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/6843Medicinal 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 material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/5748Immunoassay; Biospecific binding assay; Materials therefor for cancer involving oncogenic proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/02Screening involving studying the effect of compounds C on the interaction between interacting molecules A and B (e.g. A = enzyme and B = substrate for A, or A = receptor and B = ligand for the receptor)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the present invention relates to genes which 5 encode novel proteins related to a family of receptor proteins typified by two related membrane scanning tyro- sine inases: the Epidermal Growth Factor receptor (EGF- R), which is encoded by the eriaB gene, the normal human counterpart of an oncogene (v-e_riB) that was first recog- 10. nized in the proviral DNA of avian erythroblastosis virus; and the receptor encoded by the related gene erbB-2 .
  • the present invention relates to a DNA segment encoding the coding sequence, or a unique portion thereof, for a third member of this receptor gene family, herein 15 designated erJbB-3.
  • Proto-oncogenes encoding growth factor receptors constitute several distinct families with close overall structural homology. The highest degree of homology is 0 observed in their catalytic domains, essential for the intrinsic tyrosine kinase activity of these proteins.
  • Examples of such receptor families include: the EGF-R and the related product of the e JbB-2 oncogene; the Colony
  • CSF-l-R Stimulating Factor 1 receptor
  • PDGF-R Platelet-Derived Growth Factor receptor
  • IFN-R insulin receptor
  • IGF-l-R Growth factor 1 receptor
  • Drosophila EGF-R homologue The genetic locus of the Drosophila EGF-R homologue, designated DER, has recently been identified as being allelic to the zygotic embryonic lethal faint little ball exhibiting a complex phenotype with deterioration of multiple tissue components of ectodermal origin.
  • Fur ⁇ thermore other mutants appear to lack DER function either in the egg or the surrounding maternal tissue.
  • the DER receptor may play an important role in the ligand- receptor interaction between egg and follicle cells necessary for determination of correct shape of eggshell and embryo. It is not yet known whether DER represents the sole of the Drosophila counterpart of both known mammalian e JbB-related genes.
  • both the erJbB and erbB-2 genes have been shown to be activated as oncogenes by mechanisms involving over- expression or mutations that constitutively activate the catalytic activity of their encoded receptor proteins (Bargmann, C. I., Hung, M. C. & Weinberg, R. A., 1986, Cell 45:649-657; Di Fiore, P. P., Pierce, J. H., Kraus, M. H., Segatto, 0., King, C. R. & Aaronson, S. A., 1987, Science 237:178-182; Di Fiore, P. P., Pierce, J. H.
  • e bB gene amplification or overexpression, or a combination of both has been demonstrated in squamous cell carcinomas and glioblastomas (Libermann, T. A., Nusbaum, H. R. , Razon, N., Kris, R., Lax, I., Soreq, H., Whittle, N. , Waterfield, M. D., Ullrich, A. & Schlessinger, J., 1985, Nature 313:144-147).
  • erbB-2 amplification and overexpression have been observed in human breast and ovarian carcinomas (King, C. R. , Kraus, M. H. & Aaronson, S. A., 1985, Science 229:974-976; Slamon, D.
  • this DNA fragment is distinct from those known to encode the epidermal growth factor receptor (EGF-R) (i.e., the erJbB gene) and from the related erJB-2 gene. Characterization of this DNA fragment after partial purification and molecular cloning showed that the region of v-erJbB homology mapped to three exons that encode a ino acid sequences having homologies of 64% and 67% to contig ⁇ uous regions within the tyrosine kinase domains of the EGF-R and erbB-2 proteins, respectively.
  • EGF-R epidermal growth factor receptor
  • a probe derived from the genomic DNA clone identified cDNA clones of the related mRNA which encode a predicted 148 kd transmembrane polypeptide with structural features identifying it as a member of the eriB family, prompting designation of the new gene as eri?B-3.
  • This gene was mapped to human chromo ⁇ some 12qll-13 and was shown to be expressed as a 6.2 kb transcript in a variety of normal tissues of epithelial origin. Markedly elevated eriB-3 irtRNA levels were demon- strated in certain human tumor cell lines.
  • the present invention relates to a DNA segment having a nucleotide sequence that encodes an erJB-3 gene or a unique portion thereof.
  • This portion of an erbB-3 gene includes at least about 12 to 14 nucleotides which are sufficient to allow formation of a stable duplex with a DNA or RNA segment having sequences complementary to those in this portion of an erJbB-3 gene.
  • this unique portion of an erbB-3 gene has a sequence not present in an erbB or an eri?B-2 gene. In other words, the sequence of this portion of an erbB-3 gene differs in at least one nucleotide from the sequence of any other DNA segment.
  • this DNA segment is exempli ⁇ fied by a human genomic DNA fragment that is produced by cleavage with the Sacl restriction enzyme, has a size of about 90 kbp, and is detectable by nucleic acid hybridiza ⁇ tion with a probe derived from the v-erJbB gene only under reduced stringency hybridization conditions, as described in Example 1.
  • nucleic acid hybrid- ization and cloning methods described in the present disclosure without undue experimentation, one of ordinary skill in the art of recombinant DNA is enabled to identify and isolate DNA fragments related to the present human DNA fragment comprising a nucleotide sequence that encodes at least a portion of a mammalian erjB-3 gene other than the human eri?B-3 gene.
  • Application of the genomic DNA frag ⁇ ment of the er_t>B-3 gene as a probe in hybridization methods also enables one of ordinary skill in the art to obtain an entire er2?B-3 gene, by sequential isolation of overlapping fragments adjoining the present fragment, i.e. , by an approach known in the art as chromosome walking.
  • the present disclosure describes the partial nucleotide sequence of the human genomic 9 kbp Sacl DNA fragment, within the region of homology of the v-erjbB gene; however, the methods in the present disclosure further enable the isolation and determination of the sequence of the entire 9 kbp human genomic DNA fragment according to the present invention. Accordingly, the present invention further relates to a DNA segment having the nucleotide sequence, or a unique portion thereof, of a human genomic DNA fragment that is produced by cleavage with the Sacl restriction enzyme, has a size of about 9 kbp, and is detectable by nucleic acid hybridization with a probe derived from the v-erJbB gene only under reduced stringency hybridization conditions, as described in Example 1.
  • the present invention further enables one of ordinary skill in the art to determination of the sequences of related DNA fragments comprising the complete human erJbB-3 gene as well as erJbB-3 genes of, for example, mammals other than human.
  • the fragment is amplified, for example, by the in vitro polymerase chain reaction method (PCR; see U.S. Patent 4,683,202; U.S. Patent 4,683,195; and Saiki et al., 1985, Science 230:1350-54) or by standard methods of molecular cloning.
  • PCR in vitro polymerase chain reaction method
  • a clone of the human erjB-3 gene DNA segment according to the present invention is exemplified by a recombinant clone of a normal human thymus DNA fragment, herein designated as the E3-1 genomic clone, having the partial restriction enzyme map defined in Figure 2 and the partial DNA sequence defined in Figure 3 of the present application. Isolation and characteriza ⁇ tion of genomic clone E3-1 is described in Example 2, below.
  • nucleotide sequences of the human genomic DNA segment reveals that the nucleotide sequence encodes three open reading frames bordered by splice junction consensus sequences which define the boundaries between non- translated intron sequences and the translated exons (Fig. 2) .
  • the predicted amino acid sequences of the three exons are highly similar to three regions which are contiguous in the tyrosine kinase domains of V-erJbB, as well as human EGF-R and erJbB-2 proteins.
  • the predicted amino acid sequences of this human genomic clone are included in a larger open reading frame in complementary DNA (cDNA) clones of an mRNA species that is detected by hybridiza ⁇ tion of a probe derived from the human genomic DNA clone.
  • the present invention also relates to a DNA segment having a nucleotide sequence of an er B-3 gene in which that nucleotide sequence encodes the amino acid sequence of an erJbB-3 gene or a unique portion thereof.
  • the sequence of this portion of an ez ⁇ »B-3 amino acid sequence differs in at least one amino acid residue from the amino acid sequence encoded by any other DNA segment.
  • This portion of an erbB-3 amino acid sequence includes at least about 4 to 6 amino acids which are sufficient to provide a binding site for an antibody specific for this portion of the er * i?B-3 polypep ⁇ tide.
  • this unique portion of an erJbB-3 amino acid sequence includes sequences not present in an erbB or an er2?B-2 gene.
  • the present invention relates to such a DNA segment for which this amino acid sequence or unique portion thereof is that of the polypeptide product of the human e.rJB-3 gene.
  • This DNA segment is exemplified by the human genomic DNA clone E3-1, above, as well as by human cDNA closed designated E3-6, E3-8, E3-9, E3-11 and E3-16, which are described in Example 3 below.
  • a preferred embodiment of this DNA segment that encodes the amino acid sequence of the entire polypeptide product of the human eri?B-3 gene is human cDNA clone E3-16 having the nucleotide sequence defined in Figure 4 and having the predicted amino acid sequence defined in Figure 4.
  • the present invention relates to a bioassay for detecting erJbB-3 mRNA in a biological sample comprising the steps of: i) contacting that biological sample with a DNA segment of this inven- tion under conditions such that a DNA:RNA hybrid molecular containing this DNA segment and complementary RNA can be formed; and ii) determining the amount of that DNA segment present in the resulting hybrid molecule.
  • the present invention also includes DNA segments having DNA sequences other than those in the present examples that also encode the amino acid sequence of the polypeptide product of an erjbB-3 gene.
  • DNA segments having DNA sequences other than those in the present examples that also encode the amino acid sequence of the polypeptide product of an erjbB-3 gene.
  • standard genetic engineering methods can be used to produce synthetic DNA segments having various sequences that encode any given amino acid sequence.
  • Such synthetic DNA segments encoding at least a portion of the amino acid sequence of the polypeptide product of the human erJbB-3 gene also fall within the scope of the present invention.
  • mutant or variant genes encode polypeptide products having amino acid sequences which differ among individuals without affecting the essential function of the polypep ⁇ tide product. Still further, it is also known that many amino acid substitutions can be made in a polypeptide product by genetic engineering methods without affecting the essential function of that polypeptide.
  • the present invention further relates to a DNA segment having a nucleotide sequence that encodes an amino acid sequence differing in at least one amino acid from the the present invention further relates to a DNA segment having a nucleotide sequence that encodes an amino acid sequence differing in at least one amino acid from the amino acid sequence of human e_rJB-3, or a unique portion thereof, and having greater overall similarity to the amino acid sequence of human erJbB-3 than to that of any other polypeptide.
  • the amino acid sequence of this DNA segment includes at least about 4 to 6 amino acids which are sufficient to provide a binding site for an antibody specific for the portion of a polypeptide containing this sequence.
  • this DNA segment encodes an amino acid sequence having substantially the function of the human erJbB-3 polypeptide.
  • the predicted ex ⁇ .B-3 polypeptide is a 148 Kd transmembrane polypeptide with structural features identifying it as a member of the erbB receptor family.
  • the similarity of the amino acid sequence of the present invention with that of an erJbB-3 amino acid sequence is determined by the method of analysis defined by the sequence alignment and comparison algorithms described by Pearson and Lipman (Pearson, W.R. & Lipman, D. J., 1988, Proc. Nat. Acad. Sci . U. S.A. 55:2444-48).
  • This comparison contemplates not only precise homology of amino acid sequences, but also substitutions of one residue for another which are known to occur frequently in families of evolutionarily related proteins sharing a conserved function.
  • the present invention further relates to a recom- binant DNA molecule comprising DNA segment of this inven- tion and a vector.
  • the present invention relates to culture of cells transformed with a DNA segment according to this invention.
  • These host cells transformed with DNAs of the invention include both higher eukaryotes, including animal, plant and insect cells, and lower eukaryotes, such as yeast cells, as well as prokary- otic hosts including bacterial cells such as those of E. coli and Bacillus subtilis.
  • mammalian cells transformed with the pSV2 gpt vector carrying the E3-16 cDNA, are prepared according to well-known methods, such as those described in U.S. Patent Application 07/308,302 of Matsui et al., filed February 9, 1989; see also Pierce, J. H. et al., 1988, Science 239:628-631; and Matsui, T., Heinies, M. , Miki, T., Vietnamese, N. , La Rochelle, W. , Kraus, M. , Pierce, J. & Aaronson, S., 1989, Science 243:800-804).
  • cDNA expression plasmids are constructed by introducing the erJbB-3-related cDNA encompassing all- the nucleotides in the open reading frame into the pSV2 gpt vector into which the simian sarcoma virus long-terminal-repeat (LTR) had been engineered as the promoter, as previously described in detail.
  • Transient expression an eri?B-3 gene in such recombinant vectors is achieved by transection into COS-1 cells.
  • Stable expression of an erJbB-3 gene can also be obtained with mammalian expression vectors such as the pZIPNEOSVX vector (Cepko, C. L., Roberts, B.E. and Mulli ⁇ gan, R.
  • a eukaryotic expression vector was engineered by cloning the full-length erJbB-3 coding sequence derived from cDNA clone E3-16 into the BamHI site of the pZIPNEOSVX vector DNA adapting the DNA fragments with synthetic oligonu- cleotides.
  • NIH3T3 cells were transfected with 1 ⁇ g of recombinant expression vector DNA (LTRerJB-3) and selected with the resistance marker antibiotic G418.
  • a polyclonal rabbit antiserum was raised against a synthetic peptide (amino acid positions 1191-1205) within the predicted carboxyl terminus of the erJbB-3 coding sequence.
  • immuno- blotting analysis led to detection of the ejrJbB-3 protein (Fig. 8A) .
  • the specificity of erJbB-3 protein detection was demonstrated by preincubating the antiserum with the homologous peptide (Fig. 8B) .
  • the normal 180 kD erbB-3 protein was specifically detected with the poly- clonal antiserum only in cells transfected with the recom ⁇ binant erbB-3 expression vector, while control NIH3T3 cells that were not transfected with the vector were negative.
  • the stably transfected NIH3T3 cells are useful as erbB-3 receptor protein sources for testing potential candidates for an erbB-3-specific ligand, analysis of the biological activity, as well as generation of monoclonal antibodies raised against the native erbB-3 protein.
  • An erbB-3-specific liquid is identified by detection of autophosphorylation of the erJbB-3 receptor protein, stimulation of DNA synthesis or induction of the trans ⁇ formed phenotype of the LTRerJbB-3 transfected' NIH3T3 cells.
  • other transformed cell systems are available for functional expression of receptors of the erbB receptor family, for example, a system based on the 32D cell line, a mouse hematopoietic cell line normally dependent on interleukin-3 (11-3) for survival and prolif- eration.
  • the present invention also relates to a bioassay for testing potential analogs of ligands of erbB-3 receptors for the ability to affect an activity mediated by erbB-3 receptors, comprising the steps of: i) contacting a molecule suspected of being a ligand with erbB-3 receptors produced by a cell producing functional erbB-3 receptors; and ii) determining the amount of a biological activity mediated by those erbB-3 receptors.
  • the present invention also encompasses an isolated polypeptide having at least a portion of the amino acid sequence defined in Figure 4.
  • This invention further comprises an antibody specific for a unique portion of the human erbB-3 polypep ⁇ tide having the amino acid sequence defined in Figure 4, or a unique portion thereof.
  • the antibodies are monoclonal or polyclonal in origin, and are generated using erbB-3 receptor-related polypeptides or peptides from natural, recombinant or synthetic chemistry sources.
  • These antibodies specifical- ly bind to an erbB-3 protein which includes the sequences of such polypeptide.
  • these antibodies bind only to erbB-3 receptor proteins and not to erbB (EGF-R) or erbB-2 proteins.
  • preferred antibodies of this invention bind to an erbB-3 protein when that protein is in its native (biologically active) conformation.
  • fragments of antibodies of this invention such as Fab or F(ab)' fragments, which retain antigen binding activity and can be prepared by methods well known in the art, also fall within the scope of the present invention.
  • this invention comprises a pharmaceutical compo ⁇ sition of the antibodies of this invention, or an active fragment thereof, which can be prepared using materials and methods for preparing pharmaceutical compositions for administration of polypeptides that are well known in the art and can be adapted readily for administration of the present antibodies without undue experimentation.
  • antibodies and active fragments thereof can be used, for example, for specific detection or purifica ⁇ tion of the novel erbB-3 receptor.
  • Such antibodies could also be used in various methods known in the art for targeting drugs to tissues with high levels of erbB-3 receptors, for example, in the treatment of appropriate tumors with conjugates of such antibodies and cell killing agents.
  • the present invention further relates to a method for targeting a therapeutic drug to cells having high levels of erbB-3 receptors, comprising the steps of i) conjugating an antibody specific for an erbB-3 receptor, or an active fragment of that antibody, to the therapeutic drug; and ii) administering the result ⁇ ing conjugate to an individual with cells having high levels of erbB-3 receptors in an effective amount and by an effective route such that the antibody is able to bind to the erbB-3 receptors on those cells.
  • the antibody of this invention is exemplified by rabbit antisera containing antibodies which specifically bind to erbB-3 protein.
  • receptor specific antisera are raised to synthetic peptides representing a unique portion of the erbB-3 amino acid sequence, having six or more amino acids in sequences which are sufficient to provide a binding site for an antibody specific for this portion of the erbB-3 polypeptide.
  • this unique portion of an erbB-3 amino acid sequence includes sequences not present in an erbB or an erbB-2 amino acid sequence, as predicted by the respective cDNA sequences.
  • the erbB-3 specific anti-peptide antibody of the present invention is exemplified by an anti-peptide antibody in polyclonal rabbit antiserum raised against the synthetic peptide having the sequence (in single letter amino acid code) EDEDEEYEYMNRRRR representing amino acid positions 1191-1205 in the predicted sequence of the erbB- 3 polypeptide.
  • the specific detection of erbB-3 polypep- tide with this antiserum is illustrated in mammalian cells transformed with an expression vector carrying a human erbB-3 cDNA (see Figures 8A and 8B) .
  • Antibodies to peptides are prepared by chemically synthesizing the peptides, conjugating them to a carrier protein, and injecting the conjugated peptides into rabbits with complete Freund's adjuvant, according to standard methods of peptide immunization.
  • the peptide is synthesized by standard methods (Merrifield, R. B., 1963, J. Amer. Soc. , 85:2149) on a solid phase synthesizer.
  • the crude peptide is purified by HPLC and conjugated to the carrier, keyhole limpet hemocy- anin or bovine thyroglobulin, for example by coupling the amino terminal cysteine to the carrier through a malei ido linkage according to well known methods (e.g., Lerner, R. A. et al., 1981, Proc. Nat . Acad. Sci . USA, 78:3403).
  • rabbits are immunized with 100 ⁇ g of the erbB-3 peptide-carrier conjugate (1 mg/ml) in an equal volume of complete Freund's adjuvant and then boosted at 10-14 day intervals with 100 ⁇ g of conjugated peptide in incomplete Freund's adjuvant. Additional boosts with similar doses at 10-14 day intervals are continued until anti-peptide antibody titer, as determined, for example, by routine ELISA assays, reaches a plateau.
  • the present invention also relates to a bioassay for detecting an erbB-3 antigen in a biological sample comprising the steps of: i) contacting that sample with an antibody of the present invention specific for an erbB-3 polypeptide, under conditions such that a specific complex of that antibody and that antigen can be formed; and ii) determin- ing the amount of that antibody present in the form of those complexes.
  • Figures 1A and IB show detection of v-erbB-related DNA fragments in DNAs from normal human thymus (lane 1), human mammary tumor lines MDA-MB468 (lane 2), and SK-BR-3 (lane 3). Hybridization was conducted at reduced (Fig. 2A). i or intermediate (Fig. 2B) stringency conditions.
  • the arrow denotes a novel 9 kilobase pair (kbp) erbB-related restriction fragment distinct from those of the EGF-R gene (erbB) and erbB-2.
  • Figure 2 shows genomic and cDNA cloning of erbB-3.
  • the region of v-erbB homology within the genomic 9 kbp Sacl insert of ⁇ E3-l was subcloned into the plasmid pUC (pE3-l) and subjected to nucleotide sequence analysis.
  • the three predicted exons are depicted as solid boxes.
  • erbB-3 cDNA clones were isolated from oligo dT-primed libraries of mRNAs from normal human placenta (shaded bars) and the breast tumor cell line MCF-7 (open bar).
  • the entire nucleotide sequence was determined for both strands on erbB-3 complementary DNA from normal human placenta and upstream of the 5' Xhol site on pE3-16.
  • Figure 3 shows the nucleotide sequence of the region of v-erbB homology in the human erbB-3 gene derived from human genomic DNA clone E3-1, in the 1.5 kbp region from the Ec ⁇ R ⁇ to the Pstl sites. This region contains three open reading frames bordered by splice junction consensus sequences (underlined) . The predicted amino acid sequences of the three exons are shown in three letter code above the relevant DNA sequences.
  • Figure 4 shows the nucleotide sequence of the cDNA encoding the erbB-3 polypeptide and the predicted amino acid sequence of that polypeptide.
  • Figure 5 shows comparison of the predicted amino acid sequence of the erbB-3 polypeptide with other recep ⁇ tor-like tyrosine kinases.
  • the amino acid sequence is shown in single letter code and is numbered on the left.
  • the putative extracellular domain extends between the predicted signal sequence (solid box) at the amino-terminus and a single hydrophobic transmembrane region (solid box) within the polypeptide.
  • the two cysteine clusters (Cys) in the extracellular domain and the predicted tyrosine kinase domain (TK) within the cytoplasmic portion of the polypeptides are outlined by dark shading.
  • the putative ATP-binding site at the amino- terminus of the TK domain is circled.
  • Figure 6 shows the assignment of the genomic locus of erbB-3 was assigned to human chromosomal locus 12ql3. A total of 142 grains were localized on the 400-band ideogram. As depicted in the diagram, specific labeling of chromosome 12 was observed, where 38 out of 51 grains were localized to band ql3.
  • Figures 7A and 7B show the elevated erbB-3 transcript levels in human mammary tumor cell lines.
  • a Northern blot containing 10 ⁇ g total cellular RNA from AB589 mammary epithelial cells (lane 1), as well as mammary tumor cell lines MDA-MB415 (lane 2) and MDA-MB453 (lane 3) was hybridized with an erbB-3 cDNA probe (Fig. 7A) .
  • Fig. 7A was hybridized with an erbB-3 cDNA probe.
  • Fig. 7A erbB-3 cDNA probe
  • Figures 8A and 8B show the expression of a human erbB-3 polypeptide in cells transformed by a cDNA segment as detected by an erbB-3-specific antipeptide antiserum.
  • Cellular lysates 100 ⁇ g of each sample) were electropho- resed and transferred to nitrocellulose membranes for analysis by Western blotting.
  • Figure 8A shows the detec ⁇ tion of erbB-3 polypeptide with the antiserum.
  • Figure 8B shows the preincubation of the antiserum with homologous peptide.
  • Antibody blocking indicates binding specificity.
  • Lane 1 Selected cultures of NIH3T3 cells transfected with 1 ⁇ g LTRerbB-3 expression vector.
  • Lane 2 control NIH3T3 cells.
  • the erbB-3 gene is expected to play important roles in both normal and neoplastic processes, as is known for the EGF-R and erbB-2 genes.
  • tyrosine kinase domain which represents the most conserved region of the predicted erbB-3 protein, a short stretch of 29 amino acids closer to the carboxyl terminus than the ATP binding site differed from regions of the predicted erbB-2 and EGF-R coding sequence in 28 and 25 positions, respectively. Such regions of higher diver ⁇ gence in their cytoplasmic domains are likely to confer different functional specificity to these closely related receptor-like molecules. Thus, mutations ,o .other alter ⁇ ations in expression of the erbB-3 gene are likely to cause cancers or genetic disorders different from those associated with such defects in the erbB and erbB-2 genes.
  • EGF-R and erbB-2 genes are located at chromosomal sites 7pl2-13 and 17pl2-21.3, respectively. Thus, each gene appears to be localized to a region containing a different homeobox and a different collagen chain gene locus. Keratin type I and type II genes also map to regions of 12 and 17, respectively, consistent with the different localizations of erbB-3 and erbB-2, respectively.
  • the DNA segments of the present invention represent novel probes to aid in genetic mapping of any heritable diseases which are associated with chromosomal aberrations in the vicinity of the 12qll-13 locus. There is evidence for autocrine as well as para- crine effectors of normal cell proliferation.
  • the former are factors that are produced by the same cells upon which they stimulate cell proliferation, whereas the latter factors are secreted by cells other than those that are affected by those factors.
  • the inherent trans ⁇ forming potential of autocrine growth factors suggests that growth factors most commonly act on their target cell populations by a paracrine route.
  • the present survey of erbB-3 gene expression indicates its normal expression in cells of epithelial and neuroectodermal derivation. Comparative analysis of the three erbB receptor-like genes in different cell types of epidermal tissue revealed that keratinocytes expressed all three genes. In contrast, melanocytes and stromal fibroblasts specifically lacked EGF-R and erbB-3 transcripts, respectively.
  • melano ⁇ cytes and stromal fibroblasts may be sources of paracrine growth factors for EGF-R and erbB-3 products, respective- ly, that are expressed by the other cell types residing in close proximity in epidermal tissues.
  • Example 1 Identification of a human DNA fragment related to the erbB proto-onco ⁇ ene family.
  • human genomic DNA was cleaved with a variety of restriction endonucleases and subjected to Southern blot analysis with v-erbB as a probe.
  • Normal mammary epithelial cells AB589 Wang, K. H. & Stampfer, M. R., 1989, Cancer. Genet. Cytogenet . 37:249-261
  • immortalized keratinocytes RHEK have been described previously (Rhim, J. S. , Jay, G. , Arnstein, P., Price, F. M. , Sanford, K. K.
  • NHEM human epidermal melanocytes
  • NHEK keratinocytes
  • Sources for human embryo fibroblasts (Rubin, J. S., Osada, H. , Finch, P. W. , Taylor, W. G. , Rudikoff, S., & Aaronson, S. A., 1989, Proc. Nat. Acad. Sci . USA 86:802-806) or mammary tumor cell lines SK-BR-3, MDA- MB468, MDA-MB453, and MDA-MB415 (Kraus, M. H. ,ffy, N.
  • RNA hybridiza ⁇ tion DNA and RNA were transferred to nitrocellulose membranes as previously described (Kraus, K. H. , et al. 1987, supra) .
  • High stringency hybridization was conducte in 50% formamide and 5xSSC at 42°C. Filters were washe at 50°C in O.lxSSC.
  • Reduced stringency hybridization o DNA was carried out in 30% formamide followed by washes i O. ⁇ xSSC, while intermediate stringency was achieved b hybridization in 40% formamide and washing in 0.25xSSC
  • lane 1,2 known to contain EGF-R gene amplification and one as a erbB-2 specific gene fragment due to its increased signa intensity in another mammary tumor cell line, SK-BR-3 known to have erbB-2 amplified (Fig. 1A, lane 1,3).
  • a single 9 kbp Sacl fragment exhibited equa signal intensities in DNAs from normal human thymus, SK BR-3 and a line with high levels of EGF-R, A431 (Fig. 1A) .
  • thi fragment did not hybridize, whereas EGF-R and erbB- specific restriction fragments were still detected with v erbB as a probe (Fig. IB).
  • these finding suggested the specific detection of a novel v-erbB-relate DNA sequence within the 9 kbp Sacl fragment.
  • Example 2 Cloning of the human DNA fraqmen related to erbB.
  • a norma human genomic library was prepared from Sacl cleave thymus DNA enriched for 8 to 12 kbp fragments.
  • bacteriophage ⁇ sep6-lac5 was obtained from L. Prestidge and D. Hogness (Stanford University); many other cloning vectors derived from phage ⁇ or other genomes ca be used for cloning this DNA fragment according to stan ⁇ dard recombinant DNA methods that are well known in the art.
  • Purified phage DNA was subjected to cos-end liga- tion, restriction with Sacl , and fractionation in a continuous 10-40% sucrose gradient.
  • a genomic library was prepared by ligating Sacl restriction fragments of normal human thymus DNA in the molecular weight range of 8 kbp to 12 kbp (isolated by sucrose gradient sedimentation) with the purified phage arms. Ten recombinant clones detected by v-erbB under reduced stringency conditions did not hybridize with human EGF-R or erbB-2 cDNA probes at high stringency. As shown in the restriction map of a repre ⁇ sentative clone with a 9 kbp insert,:- region of v-erbB homology was localized by hybridization analysis to a 1.5 kbp segment spanning from the Eco ⁇ Xl to the downstream Pstl site.
  • the nucleotide sequence of a portion of a clone of the novel human genomic DNA fragment related to erbB was determined for both DNA strands by the dideoxy chain termination method (Sanger, F., Nicklen, S. & Coulson, A. R., 1977, Proc. Nat. Acad. Sci . USA. 74:5463-67) using supercoiled plasmid DNA as template.
  • the nucleotide sequence was assembled and translated using IntelliGenetics software. Amino acid sequence comparison was performed with the alignment program by Pearson and Lipman (Pearson, W. R. & Lipman, D. J. , 1988, supra) as implemented on the computers of the NCI Advanced Scientif ⁇ ic Computing Laboratory.
  • Example 3 Cloning and characterization of cDNAs for the mRNA of the human erbB-3 gene.
  • overlap ⁇ ping cDNA clones were isolated from oligo dT-primed cDNA libraries from sources with known erbB-3 expression, utilizing gene-specific genomic exons or cDNA fragments as probes.
  • an oligo dT-primed human placenta cDNA library in ⁇ gtll was obtained from Clontech.
  • MCF-7 cDNA was prepared by first strand synthesis from 5 ⁇ g poly A * RNA using an oligo dT containing linker-primer and Mo-MuLV reverse transcriptase, followed by second strand synthesis with DNA polymerase I, RNaseH, and subsequent T4 DNA polymerase treatment. Double-stranded cDNA was direction- ally cloned into the Sfil site of ⁇ pCEV9 using specific linker adapter oligonucleotides (Miki, T., Matsui, T., Heiong, M. A. & Aaronson, S. A. , 1989, Gene 83:137-146; see also, U.S. Application Ser. No.
  • Clones designated pE3-6, pE3-8, pE3-9, and pE3-ll carrying inserts with molecular weights ranging from 1.3 kpb to 4.3 kbp were isolated from a human placenta library, whereas the pE3-16 clone con ⁇ taining a 5 kbp insert was obtained by screening the MCF-7 cDNA library with the upstream most coding sequence of pE3-ll as a probe.
  • the clones pE3-8, pE3-9, pE3-ll, and pE3-16 contained identical 3' ends terminating in a poly A stretch (Fig. 2).
  • the complete coding sequence of erbB-3 was con- tained within a single long open reading frame of 4080 nucleotides extending from position 46 to an in-frame termination codon at position 4126.
  • the most upstream ATG codon at -position 100 was the likely initiation codon, as it was preceded by an in-frame stop codon at nucleotide position 43 and fulfilled the criteria of Kozak for an authentic initiation codon.
  • the open reading frame comprised 1342 codons predicting a 148 kd polypeptide. Downstream from the termination codon, multiple stop codons were present in all frames. As shown in Fig.
  • the deduced amino acid sequence of the erbB-3 polypeptide predicted a transmembrane receptor tyrosine kinase most closely related to EGF-R and erbB-2.
  • a hydrophobic signal sequence of erbB-3 was predicted to comprise the 19 amino- terminal amino acid residues. Cleavage of this signal sequence between glycine at position 19 and serine at position 20 would generate a processed polypeptide of 1323 amino acids with an estimated molecular weight of 145 kd.
  • a single hydrophobic membrane spanning domain encompassing 21 amino acids was identified within the coding sequence separating an extracellular domain of 624 amino acids from a cytoplasmic domain comprising 678 amino acids (Fig. 5).
  • the putative erbB-3 ligand-binding domain was 43% and 45% identical in amino acid residues with the predict ⁇ ed erbB-2 and EGF-R protein, respectively. Within the extracellular domain, all 50 cysteine residues of the processed erbB-3 polypeptide were conserved and similarly spaced when compared to the EGF-R and erbB-2.
  • cysteine residues were organized in two clusters contain- ing.22 and 25 cysteines respectively, a structural hall ⁇ mark of this tyrosine kinase receptor subfamily (see, for example, Yamamoto, T., Ikawa, S., Akiyama, T., Semba, K., Nomura, N., Miyajima, N., Saito, T. & Toyoshima, K. , 1986, Nature 319:230-234).
  • Ten potential N-linked glycosylation sites were localized within the erbB-3 extracellular domain. In comparison with the EGF-R and erbB-2 proteins, five and two of these glycosylation sites were conserved, respectively. Among these, the site proximal to the transmembrane domain was conserved among all three pro ⁇ teins (Fig. 5) .
  • Example 4 Chromosomal mapping of the human erbB- 3 gene.
  • the chromosomal location of the erbB-3 gene was determined by in situ hybridization (Popescu, N. C, King, C. R. & Kraus, M. H. , 1989, Genomics 4:362-366) with a 3 H- labeled plasmid containing the amino-terminal erbB-3 coding sequence.
  • a total of 110 human chromosome spreads were examined prior and subsequent to G banding for identification of individual chromosomes.
  • a total of 142 grains were localized on a 400-band ideogram. Specific labeling of chromosome 12 was observed, where 38 out of 51 grains were localized to band ql3 (Fig. 6).
  • the genomic locus of erbB-3 was assigned to 12ql3.
  • several genes have previously been mapped including the melanoma-associated antigen ME491, histone genes and the gene for lactalbumin.
  • two proto-oncogenes, int-1 and gli are located in close proximity to erbB-3.
  • Example 5 ErbB-3 expression in normal and malignant human cells. To investigate its pattern of expression, a number of human tissues were surveyed for the erbB-3 transcript. The 6.2 kb erbB-3 specific mRNA was observed in term placenta, postnatal skin, stomach, lung, kidney, and brain, while it was not detectable in skin fibroblasts, skeletal muscle or lymphoid cells. Among the fetal tissues analyzed, the erbB-3 transcript was expressed in liver, kidney, and brain, but not in fetal heart or embryonic lung fibroblasts. These observa ⁇ tions indicate the preferential expression of erbB-3 in epithelial tissues and brain.
  • iSrbB-3 expression was also investigated in indi ⁇ vidual cell populations derived from normal human epithe ⁇ lial tissues including keratinocytes, glandular epithelial cells, melanocytes, and fibroblasts.
  • EGF-R and erbB-2 transcripts were analyzed.
  • erbB-3 mRNA levels were relatively high in keratinocytes, comparable with those of erbB-2 and EGF- R in these cells.
  • Lower, but similar expression levels of each transcript were detected in cells derived from glandular epithelium.
  • Table 1 Normal expression pattern of human erbB gene family members.
  • NHEK Primary keratinocyte
  • Glandular epithelium (AB589)
  • erbB-3 mRNA levels in a series of human tumor cell lines were surveyed.
  • the erbB-3 tran ⁇ script was detected in 36 of 38 carcinomas and 2 of 12 sarcomas while 7 tumor cell lines of hematopoietic origin lacked measurable erbB-3 mRNA.
  • Markedly elevated levels of a normal-sized transcript were observed in 6 out of 17 tumor cell lines derived from human mammary carcinomas.
  • Southern blot analysis neither gross gene rearrange ⁇ ment nor amplification was detected in the cell lines.
  • Figure 7A shows the results of Northern blot analysis with control AB589 nonmalignant human mammary epithelial cells (lane 1) and two representative human mammary tumor lines, MDA-MB415 (lane 2) and MDA-MB453 (lane 3).
  • Hybridization of the same filter with a human ⁇ -actin probe (Fig. 7B) verified actual levels of mRNA in each lane. Densito- metric scanning indicated that the erbB-3 transcript in each tumor cell line was elevated more than 100 fold above that of the control cell line.
  • overexpression of this new member of the erbB family is likely to play an important role in some human malignancies.

Abstract

A DNA fragment distinct from the epidermal growth factor receptor (EGF-R) and erbB-2 genes was detected by reduced stringency hybridization of v-erbB to normal genomic human DNA. Characterization of the cloned DNA fragment mapped the region of v-erbB homology to three exons with closest homology of 64 % and 67 % to a contiguous region within the tyrosine kinase domains of theEGF-R and erbB-2 proteins, respectively. cDNA cloning revealed a predicted 148 kd transmembrane polypeptide with structural features identifying it as a member of the erbB family, prompting designation of the new gene as erbB-3. It was mapped to human chromosome 12q11-13 and was shown to be expressed as 6.2 kb transcript in a variety of normal tissues of epithelial origin. Markedly elevated erbB-3 mRNA levels were demonstrated in certain human mammary tumor cell lines. These findings indicate that increased erbB-3 expression, as in the case of EGF-R and erbB-2, plays a role in some human malignancies.

Description

DNA SEGMENT ENCODING A GENE FOR A RECEPTOR RELATED TO THE EPIDERMAL GROWTH FACTOR RECEPTOR
FIELD OF THE INVENTION The present invention relates to genes which 5 encode novel proteins related to a family of receptor proteins typified by two related membrane scanning tyro- sine inases: the Epidermal Growth Factor receptor (EGF- R), which is encoded by the eriaB gene, the normal human counterpart of an oncogene (v-e_riB) that was first recog- 10. nized in the proviral DNA of avian erythroblastosis virus; and the receptor encoded by the related gene erbB-2 . In particular, the present invention relates to a DNA segment encoding the coding sequence, or a unique portion thereof, for a third member of this receptor gene family, herein 15 designated erJbB-3.
BACKGROUND OF THE INVENTION
Proto-oncogenes encoding growth factor receptors constitute several distinct families with close overall structural homology. The highest degree of homology is 0 observed in their catalytic domains, essential for the intrinsic tyrosine kinase activity of these proteins.
Examples of such receptor families include: the EGF-R and the related product of the e JbB-2 oncogene; the Colony
Stimulating Factor 1 receptor (CSF-l-R) and the related 5 Platelet-Derived Growth Factor receptor (PDGF-R); the insulin receptor (IF-R) and the related Insulin-like
Growth factor 1 receptor (IGF-l-R); and the receptors encoded by the related oncogenes eph and elk.
It is well established that growth f ctor recep- 0 tors in several of these families play critical roles in regulation of normal growth and development. Recent studies in Drosophila have emphasized how critical and multifunctional are developmental processes mediated by ligand-receptor interactions. An increasing number of 5 Drosophila mutants with often varying phenotypes have now been identified as being due to lesions in genes encoding such proteins. The genetic locus of the Drosophila EGF-R homologue, designated DER, has recently been identified as being allelic to the zygotic embryonic lethal faint little ball exhibiting a complex phenotype with deterioration of multiple tissue components of ectodermal origin. Fur¬ thermore, other mutants appear to lack DER function either in the egg or the surrounding maternal tissue. Thus, the DER receptor may play an important role in the ligand- receptor interaction between egg and follicle cells necessary for determination of correct shape of eggshell and embryo. It is not yet known whether DER represents the sole of the Drosophila counterpart of both known mammalian e JbB-related genes.
Some of these receptor molecules have been impli¬ cated in the neoplastic process as well. In particular, both the erJbB and erbB-2 genes have been shown to be activated as oncogenes by mechanisms involving over- expression or mutations that constitutively activate the catalytic activity of their encoded receptor proteins (Bargmann, C. I., Hung, M. C. & Weinberg, R. A., 1986, Cell 45:649-657; Di Fiore, P. P., Pierce, J. H., Kraus, M. H., Segatto, 0., King, C. R. & Aaronson, S. A., 1987, Science 237:178-182; Di Fiore, P. P., Pierce, J. H. , Fleming, T. P., Hazan, R. , Ullrich, A., King, C. R. , Schlessinger, J. & Aaronson, S. A., 1987, Cell 51:1063- 1070; Velu, T. J., Beguinot, L., Vass, W. C, Willingham, M. C, Merlino, G. T., Pastan, I. & Lowy, D. R., 1987, Science 238:1408-1410). Both erbB and erjB-2 have been casually implicated in human malignancy. e bB gene amplification or overexpression, or a combination of both, has been demonstrated in squamous cell carcinomas and glioblastomas (Libermann, T. A., Nusbaum, H. R. , Razon, N., Kris, R., Lax, I., Soreq, H., Whittle, N. , Waterfield, M. D., Ullrich, A. & Schlessinger, J., 1985, Nature 313:144-147). erbB-2 amplification and overexpression have been observed in human breast and ovarian carcinomas (King, C. R. , Kraus, M. H. & Aaronson, S. A., 1985, Science 229:974-976; Slamon, D. J., Godolphin, W., Jones, L. A., Holt, J. A., Wong, S. G., Keith, D. E., Levin, W. J., Stuart, S. G. , Udove, J., Ullrich, A. & Press, M. F., 1989, Science 244:707-712), and eri?B-2 overexpression has been reported to be an important prognostic indicator of particularly aggressive tumors (Slamon, D. J., et al., 1989, supra ) . Yet, not all such tumors have been found to overexpress e_ri?B-2, and many human tumors have not yet been associated with any known oncogene. Thus, there has been a continuing need to search for additional oncogenes which would provide knowledge and methods for diagnosis and, ultimately, for rational molecular therapy of human cancers.
SUMMARY OF THE INVENTION It is an object of the present invention to provide a DNA segment encoding a receptor protein related to the erbB protό-oncogene family which previously has not been known or even suspected to exist. Further, it is an object of the present invention to develop assays for expression of the RNA and protein products of such genes to enable determining whether abnormal expression of such genes is involved in human cancers. In pursuit of the above objects, the present inventors have discovered a human genomic DNA fragment that is produced by cleavage with the Sacl restriction enzyme, has a size of about 9 kbp, and is detectable by nucleic acid hybridization with a probe derived from the v-erJB gene only under reduced stringency hybridization conditions. Thus, this DNA fragment is distinct from those known to encode the epidermal growth factor receptor (EGF-R) (i.e., the erJbB gene) and from the related erJB-2 gene. Characterization of this DNA fragment after partial purification and molecular cloning showed that the region of v-erJbB homology mapped to three exons that encode a ino acid sequences having homologies of 64% and 67% to contig¬ uous regions within the tyrosine kinase domains of the EGF-R and erbB-2 proteins, respectively. A probe derived from the genomic DNA clone identified cDNA clones of the related mRNA which encode a predicted 148 kd transmembrane polypeptide with structural features identifying it as a member of the eriB family, prompting designation of the new gene as eri?B-3. This gene was mapped to human chromo¬ some 12qll-13 and was shown to be expressed as a 6.2 kb transcript in a variety of normal tissues of epithelial origin. Markedly elevated eriB-3 irtRNA levels were demon- strated in certain human tumor cell lines.
Accordingly, in a principal embodiment, the present invention relates to a DNA segment having a nucleotide sequence that encodes an erJB-3 gene or a unique portion thereof. This portion of an erbB-3 gene includes at least about 12 to 14 nucleotides which are sufficient to allow formation of a stable duplex with a DNA or RNA segment having sequences complementary to those in this portion of an erJbB-3 gene. Further, this unique portion of an erbB-3 gene, of course, has a sequence not present in an erbB or an eri?B-2 gene. In other words, the sequence of this portion of an erbB-3 gene differs in at least one nucleotide from the sequence of any other DNA segment. In one embodiment, this DNA segment is exempli¬ fied by a human genomic DNA fragment that is produced by cleavage with the Sacl restriction enzyme, has a size of about 90 kbp, and is detectable by nucleic acid hybridiza¬ tion with a probe derived from the v-erJbB gene only under reduced stringency hybridization conditions, as described in Example 1. By application of the nucleic acid hybrid- ization and cloning methods described in the present disclosure, without undue experimentation, one of ordinary skill in the art of recombinant DNA is enabled to identify and isolate DNA fragments related to the present human DNA fragment comprising a nucleotide sequence that encodes at least a portion of a mammalian erjB-3 gene other than the human eri?B-3 gene. Application of the genomic DNA frag¬ ment of the er_t>B-3 gene as a probe in hybridization methods also enables one of ordinary skill in the art to obtain an entire er2?B-3 gene, by sequential isolation of overlapping fragments adjoining the present fragment, i.e. , by an approach known in the art as chromosome walking. The present disclosure describes the partial nucleotide sequence of the human genomic 9 kbp Sacl DNA fragment, within the region of homology of the v-erjbB gene; however, the methods in the present disclosure further enable the isolation and determination of the sequence of the entire 9 kbp human genomic DNA fragment according to the present invention. Accordingly, the present invention further relates to a DNA segment having the nucleotide sequence, or a unique portion thereof, of a human genomic DNA fragment that is produced by cleavage with the Sacl restriction enzyme, has a size of about 9 kbp, and is detectable by nucleic acid hybridization with a probe derived from the v-erJbB gene only under reduced stringency hybridization conditions, as described in Example 1. By extension of the qhromosome walking ap¬ proach noted above, the present invention further enables one of ordinary skill in the art to determination of the sequences of related DNA fragments comprising the complete human erJbB-3 gene as well as erJbB-3 genes of, for example, mammals other than human.
In the application of the present Sacl DNA frag- ment or any portion thereof as a probe for nucleic acid hybridization, the fragment is amplified, for example, by the in vitro polymerase chain reaction method (PCR; see U.S. Patent 4,683,202; U.S. Patent 4,683,195; and Saiki et al., 1985, Science 230:1350-54) or by standard methods of molecular cloning. For example, a clone of the human erjB-3 gene DNA segment according to the present invention is exemplified by a recombinant clone of a normal human thymus DNA fragment, herein designated as the E3-1 genomic clone, having the partial restriction enzyme map defined in Figure 2 and the partial DNA sequence defined in Figure 3 of the present application. Isolation and characteriza¬ tion of genomic clone E3-1 is described in Example 2, below.
Analysis of the nucleotide sequences of the human genomic DNA segment according to the present invention reveals that the nucleotide sequence encodes three open reading frames bordered by splice junction consensus sequences which define the boundaries between non- translated intron sequences and the translated exons (Fig. 2) . The predicted amino acid sequences of the three exons are highly similar to three regions which are contiguous in the tyrosine kinase domains of V-erJbB, as well as human EGF-R and erJbB-2 proteins. Moreover, the predicted amino acid sequences of this human genomic clone are included in a larger open reading frame in complementary DNA (cDNA) clones of an mRNA species that is detected by hybridiza¬ tion of a probe derived from the human genomic DNA clone. Accordingly, the present invention also relates to a DNA segment having a nucleotide sequence of an er B-3 gene in which that nucleotide sequence encodes the amino acid sequence of an erJbB-3 gene or a unique portion thereof. In other words, the sequence of this portion of an ez±»B-3 amino acid sequence differs in at least one amino acid residue from the amino acid sequence encoded by any other DNA segment. This portion of an erbB-3 amino acid sequence includes at least about 4 to 6 amino acids which are sufficient to provide a binding site for an antibody specific for this portion of the er*i?B-3 polypep¬ tide. Further, this unique portion of an erJbB-3 amino acid sequence, of course, includes sequences not present in an erbB or an er2?B-2 gene. In particular, the present invention relates to such a DNA segment for which this amino acid sequence or unique portion thereof is that of the polypeptide product of the human e.rJB-3 gene. This DNA segment is exemplified by the human genomic DNA clone E3-1, above, as well as by human cDNA closed designated E3-6, E3-8, E3-9, E3-11 and E3-16, which are described in Example 3 below. A preferred embodiment of this DNA segment that encodes the amino acid sequence of the entire polypeptide product of the human eri?B-3 gene is human cDNA clone E3-16 having the nucleotide sequence defined in Figure 4 and having the predicted amino acid sequence defined in Figure 4.
The DNA segments according to this invention are useful for detection of expression of erJbB-3 genes in normal and tumor tissues, as described in Example 5 below. Therefore, in yet another aspect, the present invention relates to a bioassay for detecting erJbB-3 mRNA in a biological sample comprising the steps of: i) contacting that biological sample with a DNA segment of this inven- tion under conditions such that a DNA:RNA hybrid molecular containing this DNA segment and complementary RNA can be formed; and ii) determining the amount of that DNA segment present in the resulting hybrid molecule. Findings described in Example 5, below, indicate that increased erJbB-3 expression, as detected by this method of this invention, plays a role in some human malignancies, as is the case for the EGF-R (erjB) and erbB-2 genes.
Of course, it will be understood by one skilled in the art of genetic engineering that in relation to produc- tion of erJbB-3 polypeptide products, the present invention also includes DNA segments having DNA sequences other than those in the present examples that also encode the amino acid sequence of the polypeptide product of an erjbB-3 gene. For example, it is known that by reference to the universal genetic code, standard genetic engineering methods can be used to produce synthetic DNA segments having various sequences that encode any given amino acid sequence. Such synthetic DNA segments encoding at least a portion of the amino acid sequence of the polypeptide product of the human erJbB-3 gene also fall within the scope of the present invention. Further, it is known that different individuals may have slightly different DNA sequences for any given human gene and, in some cases, such mutant or variant genes encode polypeptide products having amino acid sequences which differ among individuals without affecting the essential function of the polypep¬ tide product. Still further, it is also known that many amino acid substitutions can be made in a polypeptide product by genetic engineering methods without affecting the essential function of that polypeptide. Accordingly, the present invention further relates to a DNA segment having a nucleotide sequence that encodes an amino acid sequence differing in at least one amino acid from the the present invention further relates to a DNA segment having a nucleotide sequence that encodes an amino acid sequence differing in at least one amino acid from the amino acid sequence of human e_rJB-3, or a unique portion thereof, and having greater overall similarity to the amino acid sequence of human erJbB-3 than to that of any other polypeptide. The amino acid sequence of this DNA segment includes at least about 4 to 6 amino acids which are sufficient to provide a binding site for an antibody specific for the portion of a polypeptide containing this sequence. In a preferred embodiment, this DNA segment encodes an amino acid sequence having substantially the function of the human erJbB-3 polypeptide. As noted above, the predicted ex±.B-3 polypeptide is a 148 Kd transmembrane polypeptide with structural features identifying it as a member of the erbB receptor family.
The similarity of the amino acid sequence of the present invention with that of an erJbB-3 amino acid sequence is determined by the method of analysis defined by the sequence alignment and comparison algorithms described by Pearson and Lipman (Pearson, W.R. & Lipman, D. J., 1988, Proc. Nat. Acad. Sci . U. S.A. 55:2444-48). This comparison contemplates not only precise homology of amino acid sequences, but also substitutions of one residue for another which are known to occur frequently in families of evolutionarily related proteins sharing a conserved function.
The present invention further relates to a recom- binant DNA molecule comprising DNA segment of this inven- tion and a vector. In yet another aspect, the present invention relates to culture of cells transformed with a DNA segment according to this invention. These host cells transformed with DNAs of the invention include both higher eukaryotes, including animal, plant and insect cells, and lower eukaryotes, such as yeast cells, as well as prokary- otic hosts including bacterial cells such as those of E. coli and Bacillus subtilis. These aspects of the inven- transformed with a DNA of the invention, wherein the transforming DNA is capable of being expressed to produce the functional polypeptide of an erJbB-3 gene. For exam¬ ple, mammalian cells (COS-1) transformed with the pSV2 gpt vector carrying the E3-16 cDNA, are prepared according to well-known methods, such as those described in U.S. Patent Application 07/308,302 of Matsui et al., filed February 9, 1989; see also Pierce, J. H. et al., 1988, Science 239:628-631; and Matsui, T., Heidaran, M. , Miki, T., Popescu, N. , La Rochelle, W. , Kraus, M. , Pierce, J. & Aaronson, S., 1989, Science 243:800-804). Briefly, cDNA expression plasmids are constructed by introducing the erJbB-3-related cDNA encompassing all- the nucleotides in the open reading frame into the pSV2 gpt vector into which the simian sarcoma virus long-terminal-repeat (LTR) had been engineered as the promoter, as previously described in detail. Transient expression an eri?B-3 gene in such recombinant vectors is achieved by transection into COS-1 cells. Stable expression of an erJbB-3 gene can also be obtained with mammalian expression vectors such as the pZIPNEOSVX vector (Cepko, C. L., Roberts, B.E. and Mulli¬ gan, R. C, 1984, Cell 37:1053-62). For example, a eukaryotic expression vector was engineered by cloning the full-length erJbB-3 coding sequence derived from cDNA clone E3-16 into the BamHI site of the pZIPNEOSVX vector DNA adapting the DNA fragments with synthetic oligonu- cleotides. NIH3T3 cells were transfected with 1 μg of recombinant expression vector DNA (LTRerJB-3) and selected with the resistance marker antibiotic G418. To detect expression of eri?B-3, a polyclonal rabbit antiserum was raised against a synthetic peptide (amino acid positions 1191-1205) within the predicted carboxyl terminus of the erJbB-3 coding sequence. As shown in Figure 8, immuno- blotting analysis led to detection of the ejrJbB-3 protein (Fig. 8A) . The specificity of erJbB-3 protein detection was demonstrated by preincubating the antiserum with the homologous peptide (Fig. 8B) . Moreover, the normal 180 kD erbB-3 protein was specifically detected with the poly- clonal antiserum only in cells transfected with the recom¬ binant erbB-3 expression vector, while control NIH3T3 cells that were not transfected with the vector were negative. The stably transfected NIH3T3 cells are useful as erbB-3 receptor protein sources for testing potential candidates for an erbB-3-specific ligand, analysis of the biological activity, as well as generation of monoclonal antibodies raised against the native erbB-3 protein. An erbB-3-specific liquid is identified by detection of autophosphorylation of the erJbB-3 receptor protein, stimulation of DNA synthesis or induction of the trans¬ formed phenotype of the LTRerJbB-3 transfected' NIH3T3 cells. Alternatively, other transformed cell systems are available for functional expression of receptors of the erbB receptor family, for example, a system based on the 32D cell line, a mouse hematopoietic cell line normally dependent on interleukin-3 (11-3) for survival and prolif- eration. Recent studies have established that introduc¬ tion of an expression vector for the EGF-R in these cells leads to effective coupling with EGF mitogenic signal transduction pathways, thereby allowing a ligand of the EGF-R to replace 11-3 in supporting survival and growth of the 32D cells. By employing the known methods described for the EGF-R, for example (Pierce, J. H. et al., 1988, supra) , the E3-16 cDNA of the present invention is ex¬ pressed to produce functional receptors in 32D cells which are then useful for examining the biological function of these erbB-3 receptors, for instance, the specificity of their ligand binding capacity and coupling capacities to secondary messenger systems. Thus, by so using gene expression methods described herein with the DNAs of the present invention, especially the preferred E3-16 cDNA clone, one of ordinary skill in the art, without undue experimentation, can construct cell systems which fall within the scope of this invention, for determining the mechanisms of erbB-3 regulatory processes. Accordingly, the present invention also relates to a bioassay for testing potential analogs of ligands of erbB-3 receptors for the ability to affect an activity mediated by erbB-3 receptors, comprising the steps of: i) contacting a molecule suspected of being a ligand with erbB-3 receptors produced by a cell producing functional erbB-3 receptors; and ii) determining the amount of a biological activity mediated by those erbB-3 receptors.
Various standard recombinant systems, such as those cited above as well as others known in the art, are suitable as well for production of large amounts of the novel erbB-3 receptor protein using methods of isolation for receptor proteins that are well known in the art. Therefore, the present invention also encompasses an isolated polypeptide having at least a portion of the amino acid sequence defined in Figure 4.
This invention further comprises an antibody specific for a unique portion of the human erbB-3 polypep¬ tide having the amino acid sequence defined in Figure 4, or a unique portion thereof. In this embodiment of the invention, the antibodies are monoclonal or polyclonal in origin, and are generated using erbB-3 receptor-related polypeptides or peptides from natural, recombinant or synthetic chemistry sources. These antibodies specifical- ly bind to an erbB-3 protein which includes the sequences of such polypeptide. In other words, these antibodies bind only to erbB-3 receptor proteins and not to erbB (EGF-R) or erbB-2 proteins. Also, preferred antibodies of this invention bind to an erbB-3 protein when that protein is in its native (biologically active) conformation.
Fragments of antibodies of this invention, such as Fab or F(ab)' fragments, which retain antigen binding activity and can be prepared by methods well known in the art, also fall within the scope of the present invention. Further, this invention comprises a pharmaceutical compo¬ sition of the antibodies of this invention, or an active fragment thereof, which can be prepared using materials and methods for preparing pharmaceutical compositions for administration of polypeptides that are well known in the art and can be adapted readily for administration of the present antibodies without undue experimentation.
These antibodies and active fragments thereof, can be used, for example, for specific detection or purifica¬ tion of the novel erbB-3 receptor. Such antibodies could also be used in various methods known in the art for targeting drugs to tissues with high levels of erbB-3 receptors, for example, in the treatment of appropriate tumors with conjugates of such antibodies and cell killing agents. Accordingly, the present invention further relates to a method for targeting a therapeutic drug to cells having high levels of erbB-3 receptors, comprising the steps of i) conjugating an antibody specific for an erbB-3 receptor, or an active fragment of that antibody, to the therapeutic drug; and ii) administering the result¬ ing conjugate to an individual with cells having high levels of erbB-3 receptors in an effective amount and by an effective route such that the antibody is able to bind to the erbB-3 receptors on those cells.
The antibody of this invention is exemplified by rabbit antisera containing antibodies which specifically bind to erbB-3 protein. Such receptor specific antisera are raised to synthetic peptides representing a unique portion of the erbB-3 amino acid sequence, having six or more amino acids in sequences which are sufficient to provide a binding site for an antibody specific for this portion of the erbB-3 polypeptide. Further, this unique portion of an erbB-3 amino acid sequence, of course, includes sequences not present in an erbB or an erbB-2 amino acid sequence, as predicted by the respective cDNA sequences. The erbB-3 specific anti-peptide antibody of the present invention is exemplified by an anti-peptide antibody in polyclonal rabbit antiserum raised against the synthetic peptide having the sequence (in single letter amino acid code) EDEDEEYEYMNRRRR representing amino acid positions 1191-1205 in the predicted sequence of the erbB- 3 polypeptide. The specific detection of erbB-3 polypep- tide with this antiserum is illustrated in mammalian cells transformed with an expression vector carrying a human erbB-3 cDNA (see Figures 8A and 8B) .
Antibodies to peptides are prepared by chemically synthesizing the peptides, conjugating them to a carrier protein, and injecting the conjugated peptides into rabbits with complete Freund's adjuvant, according to standard methods of peptide immunization. For example, the peptide is synthesized by standard methods (Merrifield, R. B., 1963, J. Amer. Soc. , 85:2149) on a solid phase synthesizer. The crude peptide is purified by HPLC and conjugated to the carrier, keyhole limpet hemocy- anin or bovine thyroglobulin, for example by coupling the amino terminal cysteine to the carrier through a malei ido linkage according to well known methods (e.g., Lerner, R. A. et al., 1981, Proc. Nat . Acad. Sci . USA, 78:3403). In one standard method of peptide immunology, rabbits are immunized with 100 μg of the erbB-3 peptide-carrier conjugate (1 mg/ml) in an equal volume of complete Freund's adjuvant and then boosted at 10-14 day intervals with 100 μg of conjugated peptide in incomplete Freund's adjuvant. Additional boosts with similar doses at 10-14 day intervals are continued until anti-peptide antibody titer, as determined, for example, by routine ELISA assays, reaches a plateau.
Thus, by following the teachings of the present disclosure, including application of generally known immunological methods cited herein, one of ordinary skill in the art is able to obtain erbB-3-specific antibodies and use them in a variety of immunological assays, for example, for diagnostic detection of unusually high or low expression in normal or tumor tissues. Thus, the present invention also relates to a bioassay for detecting an erbB-3 antigen in a biological sample comprising the steps of: i) contacting that sample with an antibody of the present invention specific for an erbB-3 polypeptide, under conditions such that a specific complex of that antibody and that antigen can be formed; and ii) determin- ing the amount of that antibody present in the form of those complexes.
* * * *
The present invention may be understood more readily by reference to the following detailed description of specific embodiments and the Examples and Figures included therein.
BRIEF DESCRIPTION OF THE DRAWINGS Figures 1A and IB show detection of v-erbB-related DNA fragments in DNAs from normal human thymus (lane 1), human mammary tumor lines MDA-MB468 (lane 2), and SK-BR-3 (lane 3). Hybridization was conducted at reduced (Fig. 2A).ior intermediate (Fig. 2B) stringency conditions. The arrow denotes a novel 9 kilobase pair (kbp) erbB-related restriction fragment distinct from those of the EGF-R gene (erbB) and erbB-2.
Figure 2 shows genomic and cDNA cloning of erbB-3. The region of v-erbB homology within the genomic 9 kbp Sacl insert of λE3-l was subcloned into the plasmid pUC (pE3-l) and subjected to nucleotide sequence analysis. The three predicted exons are depicted as solid boxes. erbB-3 cDNA clones were isolated from oligo dT-primed libraries of mRNAs from normal human placenta (shaded bars) and the breast tumor cell line MCF-7 (open bar). The entire nucleotide sequence was determined for both strands on erbB-3 complementary DNA from normal human placenta and upstream of the 5' Xhol site on pE3-16. The coding sequence is shown as a solid bar and splice junc¬ tions of the three characterized genomic exons are indi- cated by vertical white lines. Solid lines in the cDNA map represent untranslated sequences. Restriction sites: A=AccI, Av=AvaI, B=SazπHI, Bg=SgIII, E=£coRI, H=ffϊ.ndIII,
Figure imgf000016_0001
Figure 3 shows the nucleotide sequence of the region of v-erbB homology in the human erbB-3 gene derived from human genomic DNA clone E3-1, in the 1.5 kbp region from the EcόRΪ to the Pstl sites. This region contains three open reading frames bordered by splice junction consensus sequences (underlined) . The predicted amino acid sequences of the three exons are shown in three letter code above the relevant DNA sequences.
Figure 4 shows the nucleotide sequence of the cDNA encoding the erbB-3 polypeptide and the predicted amino acid sequence of that polypeptide.
Figure 5 shows comparison of the predicted amino acid sequence of the erbB-3 polypeptide with other recep¬ tor-like tyrosine kinases. The amino acid sequence is shown in single letter code and is numbered on the left. The putative extracellular domain (light shading) extends between the predicted signal sequence (solid box) at the amino-terminus and a single hydrophobic transmembrane region (solid box) within the polypeptide. The two cysteine clusters (Cys) in the extracellular domain and the predicted tyrosine kinase domain (TK) within the cytoplasmic portion of the polypeptides are outlined by dark shading. The putative ATP-binding site at the amino- terminus of the TK domain is circled. Potential auto- phosphorylation sites within the carboxyl-terminal domain (COOH) are indicated by asterisks. Potential N-linked glycosylation sites (•—— ) are marked above the amino acid sequence. The percentage of amino acid homology of erbB-3 in individual domains with erbB-2, EGF-R, met, eph, insulin receptor (IR), and fms is listed below. Less than 16% identity is denoted by (-).
Figure 6 shows the assignment of the genomic locus of erbB-3 was assigned to human chromosomal locus 12ql3. A total of 142 grains were localized on the 400-band ideogram. As depicted in the diagram, specific labeling of chromosome 12 was observed, where 38 out of 51 grains were localized to band ql3.
Figures 7A and 7B show the elevated erbB-3 transcript levels in human mammary tumor cell lines. A Northern blot containing 10 μg total cellular RNA from AB589 mammary epithelial cells (lane 1), as well as mammary tumor cell lines MDA-MB415 (lane 2) and MDA-MB453 (lane 3) was hybridized with an erbB-3 cDNA probe (Fig. 7A) . Following signal decay the same blot was rehybrid- ized with a human β-actin cDNA probe (Gunning, P., Ponte, P., Okayama, H., Engel, J., Blau, H. & Kedes, L., 1983, Mol . Cell Biol . 3:787-795). Figures 8A and 8B show the expression of a human erbB-3 polypeptide in cells transformed by a cDNA segment as detected by an erbB-3-specific antipeptide antiserum. Cellular lysates (100 μg of each sample) were electropho- resed and transferred to nitrocellulose membranes for analysis by Western blotting. Figure 8A shows the detec¬ tion of erbB-3 polypeptide with the antiserum. Figure 8B shows the preincubation of the antiserum with homologous peptide. Antibody blocking indicates binding specificity. Lane 1: Selected cultures of NIH3T3 cells transfected with 1 μg LTRerbB-3 expression vector. Lane 2: control NIH3T3 cells.
DESCRIPTION OF SPECIFIC EMBODIMENTS The identification of a third member of the erbBEGF receptor family of membrane spanning tyrosine kinases and the cloning of its full length coding sequence is described in the Examples herein. The presence of apparent structural domains resembling those of the EGF receptor suggests the existence of an extracellular binding site for a ligand. The structural relatedness of the extracellular domain of the erbB-3 receptor with that of the EGF receptor indicates that one or more of an increasing number of EGF-like ligands (Shoyab, M. , Plow¬ man, G. D., McDonald, V. L., Bradley, J. G. & Todaro, G. J., 1989, Science 243:1074-1076) interacts with the erbB-3 product. Accordingly, the erbB-3 gene is expected to play important roles in both normal and neoplastic processes, as is known for the EGF-R and erbB-2 genes.
Despite extensive collinear homology with the EGF receptor and erbB-2, distinct regions within the predicted erbB-2, coding sequence revealed relatively higher degrees of divergence. For example, its carboxyl terminal domain failed to exhibit significant collinear identity scores with either erbB-2 or EGF-R. The divergence at the carboxyl terminus also accounts for minor size differences among the three polypeptides of erbB-3, erbB-2, and EGF-R, which possess estimated molecular weights of 148 kilo- daltons (kd), 138 kd, and 131 kd, respectively. Within the tyrosine kinase domain, which represents the most conserved region of the predicted erbB-3 protein, a short stretch of 29 amino acids closer to the carboxyl terminus than the ATP binding site differed from regions of the predicted erbB-2 and EGF-R coding sequence in 28 and 25 positions, respectively. Such regions of higher diver¬ gence in their cytoplasmic domains are likely to confer different functional specificity to these closely related receptor-like molecules. Thus, mutations ,o .other alter¬ ations in expression of the erbB-3 gene are likely to cause cancers or genetic disorders different from those associated with such defects in the erbB and erbB-2 genes.
Chromosomal mapping localized erbB-3 to human chromosome 12 at the qll-13 locus, whereas the related
EGF-R and erbB-2 genes are located at chromosomal sites 7pl2-13 and 17pl2-21.3, respectively. Thus, each gene appears to be localized to a region containing a different homeobox and a different collagen chain gene locus. Keratin type I and type II genes also map to regions of 12 and 17, respectively, consistent with the different localizations of erbB-3 and erbB-2, respectively. Thus, the DNA segments of the present invention represent novel probes to aid in genetic mapping of any heritable diseases which are associated with chromosomal aberrations in the vicinity of the 12qll-13 locus. There is evidence for autocrine as well as para- crine effectors of normal cell proliferation. The former are factors that are produced by the same cells upon which they stimulate cell proliferation, whereas the latter factors are secreted by cells other than those that are affected by those factors. However, the inherent trans¬ forming potential of autocrine growth factors suggests that growth factors most commonly act on their target cell populations by a paracrine route. The present survey of erbB-3 gene expression indicates its normal expression in cells of epithelial and neuroectodermal derivation. Comparative analysis of the three erbB receptor-like genes in different cell types of epidermal tissue revealed that keratinocytes expressed all three genes. In contrast, melanocytes and stromal fibroblasts specifically lacked EGF-R and erbB-3 transcripts, respectively. Thus, melano¬ cytes and stromal fibroblasts may be sources of paracrine growth factors for EGF-R and erbB-3 products, respective- ly, that are expressed by the other cell types residing in close proximity in epidermal tissues.
Given that both erbB and erbB-2 have been casually implicated in human malignancy, the present, findings (Example 5) that the erbB-3 transcript is overexpressed in a significant fraction of human mammary tumor cell lines indicates that this new member of the EGF-R receptor family also plays an important role in some human malig¬ nancies.
Example 1. Identification of a human DNA fragment related to the erbB proto-oncoσene family. In an effort to detect novel erbB-related genes, human genomic DNA was cleaved with a variety of restriction endonucleases and subjected to Southern blot analysis with v-erbB as a probe. Normal mammary epithelial cells AB589 (Walen, K. H. & Stampfer, M. R., 1989, Cancer. Genet. Cytogenet . 37:249-261) and immortalized keratinocytes RHEK have been described previously (Rhim, J. S. , Jay, G. , Arnstein, P., Price, F. M. , Sanford, K. K. & Aaronson, S. A., 1985, Science 227:1250-52). Normal human epidermal melanocytes (NHEM) and keratinocytes (NHEK) were obtained from Clonetics. Sources for human embryo fibroblasts (Rubin, J. S., Osada, H. , Finch, P. W. , Taylor, W. G. , Rudikoff, S., & Aaronson, S. A., 1989, Proc. Nat. Acad. Sci . USA 86:802-806) or mammary tumor cell lines SK-BR-3, MDA- MB468, MDA-MB453, and MDA-MB415 (Kraus, M. H. , Popescu, N. C, Amsbaugh, S. C. & King, C. R., 1987 EMBO. J. 6:605- 610) have been described. For nucleic acid RNA hybridiza¬ tion, DNA and RNA were transferred to nitrocellulose membranes as previously described (Kraus, K. H. , et al. 1987, supra) . High stringency hybridization was conducte in 50% formamide and 5xSSC at 42°C. Filters were washe at 50°C in O.lxSSC. Reduced stringency hybridization o DNA was carried out in 30% formamide followed by washes i O.δxSSC, while intermediate stringency was achieved b hybridization in 40% formamide and washing in 0.25xSSC For the specific results depicted in Fig. 1, DNAs wer restricted with Sacl and hybridized with probe specifi for an oncogenic viral form of the erbB gene, v-erbB spanning from the upstream BamHI site to the EcoRl site i the avian erythroblastosis proviral DNA (Vennstrom, B. Franshier, L., Moscovici, C; <- & Bishop, J. M. , 1980, J Virol . 36:575-585). Under reduced stringency hybridization, four Sac restriction fragments were detected. Two were identifie as EGF-R gene fragments by their amplifications in th mammary tumor cell line MDA-MB468 (Fig. 1A, lane 1,2 known to contain EGF-R gene amplification and one as a erbB-2 specific gene fragment due to its increased signa intensity in another mammary tumor cell line, SK-BR-3 known to have erbB-2 amplified (Fig. 1A, lane 1,3). However, a single 9 kbp Sacl fragment exhibited equa signal intensities in DNAs from normal human thymus, SK BR-3 and a line with high levels of EGF-R, A431 (Fig. 1A) . When the hybridization stringency was raised by 7°C, thi fragment did not hybridize, whereas EGF-R and erbB- specific restriction fragments were still detected with v erbB as a probe (Fig. IB). Taken together, these finding suggested the specific detection of a novel v-erbB-relate DNA sequence within the 9 kbp Sacl fragment.
Example 2.* Cloning of the human DNA fraqmen related to erbB. For further characterization a norma human genomic library was prepared from Sacl cleave thymus DNA enriched for 8 to 12 kbp fragments. Fo convenience, bacteriophage λsep6-lac5 was obtained from L. Prestidge and D. Hogness (Stanford University); many other cloning vectors derived from phage λ or other genomes ca be used for cloning this DNA fragment according to stan¬ dard recombinant DNA methods that are well known in the art. Purified phage DNA was subjected to cos-end liga- tion, restriction with Sacl , and fractionation in a continuous 10-40% sucrose gradient. A genomic library was prepared by ligating Sacl restriction fragments of normal human thymus DNA in the molecular weight range of 8 kbp to 12 kbp (isolated by sucrose gradient sedimentation) with the purified phage arms. Ten recombinant clones detected by v-erbB under reduced stringency conditions did not hybridize with human EGF-R or erbB-2 cDNA probes at high stringency. As shown in the restriction map of a repre¬ sentative clone with a 9 kbp insert,:-
Figure imgf000022_0001
region of v-erbB homology was localized by hybridization analysis to a 1.5 kbp segment spanning from the EcoϊXl to the downstream Pstl site.
The nucleotide sequence of a portion of a clone of the novel human genomic DNA fragment related to erbB was determined for both DNA strands by the dideoxy chain termination method (Sanger, F., Nicklen, S. & Coulson, A. R., 1977, Proc. Nat. Acad. Sci . USA. 74:5463-67) using supercoiled plasmid DNA as template. The nucleotide sequence was assembled and translated using IntelliGenetics software. Amino acid sequence comparison was performed with the alignment program by Pearson and Lipman (Pearson, W. R. & Lipman, D. J. , 1988, supra) as implemented on the computers of the NCI Advanced Scientif¬ ic Computing Laboratory. Hydrophobic and hydrophilic regions in the predicted protein were identified according to Kyte and Doolittle (Kyte, J. & Doolittle, R. F., 1982, J. Mol . Biol . 157:105-132). Nucleotide sequence analysis revealed that the region of v-erbB homology in the 1.5 kbp segment from the EcόRl to the Pstl contained three open reading frames bordered by splice junction consensus sequences (Fig. 2). Computerized comparisons of the predicted amino acid sequence of these three open reading frames with other known proteins revealed the highest identity scores of 64% to 67% to three regions which are contiguous in the tyrosine kinase domains of v-erbB, as well as human EGF-R and erbB-2 proteins. Furthermore, all splice junctions of the three characterized exons in the new gene were conserved with erbB-2. Amino acid sequence homology to other known tyrosine kinases was significantly lower, ranging from 39% to 46%.
A single 6.2 kb specific mRNA was identified by Northern blot analysis of human epithelial cells using the 150 bp Spel-Accl exon-containing fragment as probe (Fig. 2). Under the stringent hybridization conditions em¬ ployed, this probe detected neither the 5 kb erbB-2 mRNA nor the 6 kb and 10 kb EGF-R mRNAs. All of these findings suggested that the present work has identified a new functional member of the erbB proto-oncogene family, which tentatively has been designated as erbB-3.
Example 3. Cloning and characterization of cDNAs for the mRNA of the human erbB-3 gene. In an effort to characterize the entire erbB-3 coding sequence, overlap¬ ping cDNA clones were isolated from oligo dT-primed cDNA libraries from sources with known erbB-3 expression, utilizing gene-specific genomic exons or cDNA fragments as probes. In brief, an oligo dT-primed human placenta cDNA library in λgtll was obtained from Clontech. MCF-7 cDNA was prepared by first strand synthesis from 5 μg poly A* RNA using an oligo dT containing linker-primer and Mo-MuLV reverse transcriptase, followed by second strand synthesis with DNA polymerase I, RNaseH, and subsequent T4 DNA polymerase treatment. Double-stranded cDNA was direction- ally cloned into the Sfil site of λpCEV9 using specific linker adapter oligonucleotides (Miki, T., Matsui, T., Heidaran, M. A. & Aaronson, S. A. , 1989, Gene 83:137-146; see also, U.S. Application Ser. No. 07/386,053 of Miki et al., filed July 28, 1989). Following plaque purification, phage DNA inserts were subcloned into pUC-based plasmid vectors for further characterization. The clones were initially characterized by restriction analysis and hybridization to the mRNA, and were subsequently subjected to nucleotide sequence analysis. Clones designated pE3-6, pE3-8, pE3-9, and pE3-ll carrying inserts with molecular weights ranging from 1.3 kpb to 4.3 kbp were isolated from a human placenta library, whereas the pE3-16 clone con¬ taining a 5 kbp insert was obtained by screening the MCF-7 cDNA library with the upstream most coding sequence of pE3-ll as a probe. The clones pE3-8, pE3-9, pE3-ll, and pE3-16 contained identical 3' ends terminating in a poly A stretch (Fig. 2).
The complete coding sequence of erbB-3 was con- tained within a single long open reading frame of 4080 nucleotides extending from position 46 to an in-frame termination codon at position 4126. The most upstream ATG codon at -position 100 was the likely initiation codon, as it was preceded by an in-frame stop codon at nucleotide position 43 and fulfilled the criteria of Kozak for an authentic initiation codon. The open reading frame comprised 1342 codons predicting a 148 kd polypeptide. Downstream from the termination codon, multiple stop codons were present in all frames. As shown in Fig. 5, the deduced amino acid sequence of the erbB-3 polypeptide predicted a transmembrane receptor tyrosine kinase most closely related to EGF-R and erbB-2. A hydrophobic signal sequence of erbB-3 was predicted to comprise the 19 amino- terminal amino acid residues. Cleavage of this signal sequence between glycine at position 19 and serine at position 20 would generate a processed polypeptide of 1323 amino acids with an estimated molecular weight of 145 kd. A single hydrophobic membrane spanning domain encompassing 21 amino acids was identified within the coding sequence separating an extracellular domain of 624 amino acids from a cytoplasmic domain comprising 678 amino acids (Fig. 5).
The putative erbB-3 ligand-binding domain was 43% and 45% identical in amino acid residues with the predict¬ ed erbB-2 and EGF-R protein, respectively. Within the extracellular domain, all 50 cysteine residues of the processed erbB-3 polypeptide were conserved and similarly spaced when compared to the EGF-R and erbB-2. Forty-seven cysteine residues were organized in two clusters contain- ing.22 and 25 cysteines respectively, a structural hall¬ mark of this tyrosine kinase receptor subfamily (see, for example, Yamamoto, T., Ikawa, S., Akiyama, T., Semba, K., Nomura, N., Miyajima, N., Saito, T. & Toyoshima, K. , 1986, Nature 319:230-234). Ten potential N-linked glycosylation sites were localized within the erbB-3 extracellular domain. In comparison with the EGF-R and erbB-2 proteins, five and two of these glycosylation sites were conserved, respectively. Among these, the site proximal to the transmembrane domain was conserved among all three pro¬ teins (Fig. 5) .
Within the cytoplasmic domain, a core of 277 amino acids from position 702 through 978 revealed the most extensive homology with the tyrosine kinase domains of EGF-R and erbB-2. In this region 60% or 62% of amino acid residues were identical and 90% or 89% were conserved, respectively. This stretch of amino acid homology coin¬ cides with the minimal catalytic domain of tyrosine kinases (Hanks, S. K., Quinn, A. M. & Hunter, T., 1988, Science 241:42-52). There was significantly lower homolo¬ gy with other tyrosine kinases (Fig. 5). The consensus sequence for an ATP-binding site GxGxxG (Hanks, S. K. et al., 1988, supra ) was identified at amino acid positions 716 through 721. This sequence as well as a lysine residue located 21 amino acid residues further toward the carboxyl terminus were conserved between the three erbB- related receptors. Taken together these findings defined the region between amino acid position 702 and 978 as the putative catalytic domain of the erbB-3 protein (Fig. 5). The most divergent region of erbB-3 compared to either EGF-R or erbB-2 was its carboxyl terminus compris¬ ing 364 amino acids. This region showed a high degree of hydrophilicity and the frequent occurrence of proline and tyrosine residues. Among these tyrosine residues, those at positions 1197, 1199, and 1262 matched closest with the consensus sequence for putative phosphorylation sites. The peptide sequence YEYMN, encompassing tyrosine 1197 and 1199, was repeated at positions 1260-1264 and was at both locations surrounded by charged residues, providing an environment of high local hydrophilicity. These observa¬ tions render tyrosines 1197, 1199 and 1262 likely candidates for autophosphorylation sites of the erbB3 protein.
Example 4. Chromosomal mapping of the human erbB- 3 gene. The chromosomal location of the erbB-3 gene was determined by in situ hybridization (Popescu, N. C, King, C. R. & Kraus, M. H. , 1989, Genomics 4:362-366) with a 3H- labeled plasmid containing the amino-terminal erbB-3 coding sequence. A total of 110 human chromosome spreads were examined prior and subsequent to G banding for identification of individual chromosomes. A total of 142 grains were localized on a 400-band ideogram. Specific labeling of chromosome 12 was observed, where 38 out of 51 grains were localized to band ql3 (Fig. 6). Thus, the genomic locus of erbB-3 was assigned to 12ql3. In this region of chromosome 12, several genes have previously been mapped including the melanoma-associated antigen ME491, histone genes and the gene for lactalbumin. In addition, two proto-oncogenes, int-1 and gli are located in close proximity to erbB-3.
Example 5. ErbB-3 expression in normal and malignant human cells. To investigate its pattern of expression, a number of human tissues were surveyed for the erbB-3 transcript. The 6.2 kb erbB-3 specific mRNA was observed in term placenta, postnatal skin, stomach, lung, kidney, and brain, while it was not detectable in skin fibroblasts, skeletal muscle or lymphoid cells. Among the fetal tissues analyzed, the erbB-3 transcript was expressed in liver, kidney, and brain, but not in fetal heart or embryonic lung fibroblasts. These observa¬ tions indicate the preferential expression of erbB-3 in epithelial tissues and brain. iSrbB-3 expression was also investigated in indi¬ vidual cell populations derived from normal human epithe¬ lial tissues including keratinocytes, glandular epithelial cells, melanocytes, and fibroblasts. For comparison levels of EGF-R and erbB-2 transcripts were analyzed. As shown in Table 1, erbB-3 mRNA levels were relatively high in keratinocytes, comparable with those of erbB-2 and EGF- R in these cells. Lower, but similar expression levels of each transcript were detected in cells derived from glandular epithelium. These findings are consistent with growth regulatory roles of all three receptor-like mole¬ cules in squamous and glandular epithelium. Whereas erbB-
2 and EGF-R transcripts were also readily observed in normal fibroblasts, the same cells lacked detectable erbB-
3 mRNA. In contrast, normal human melanocytes, which expressed both erbB-3 and erbB-2 at levels comparable with human.keratinocytes, lacked detectable EGF-R transcripts.
Thus, the expression patterns of these receptor-like molecules were different in specialized cell populations derived from epidermal tissues.
Table 1: Normal expression pattern of human erbB gene family members.
Cell Source of Transcripts Embryonic fibroblast (M426)
Skin fibroblast (501T)
Immortal keratinocyte (RHEK)
Primary keratinocyte (NHEK)
Glandular epithelium (AB589)
Melanocyte (NHEM)
Figure imgf000027_0001
Replicate Northern blots were hybridized with equal amounts (in cpm) of probes of similar specific activities for erbB-3, erbB-2, and EGF-R, respectively. Relative signal intensities were estimated: - not detectable, (+) weakly positive, + positive, ++ strongly positive.
To search for evidence of erbB-3 involvement in the neoplastic process, erbB-3 mRNA levels in a series of human tumor cell lines were surveyed. The erbB-3 tran¬ script was detected in 36 of 38 carcinomas and 2 of 12 sarcomas while 7 tumor cell lines of hematopoietic origin lacked measurable erbB-3 mRNA. Markedly elevated levels of a normal-sized transcript were observed in 6 out of 17 tumor cell lines derived from human mammary carcinomas. By Southern blot analysis, neither gross gene rearrange¬ ment nor amplification was detected in the cell lines. Figure 7A shows the results of Northern blot analysis with control AB589 nonmalignant human mammary epithelial cells (lane 1) and two representative human mammary tumor lines, MDA-MB415 (lane 2) and MDA-MB453 (lane 3). Hybridization of the same filter with a human β-actin probe (Fig. 7B) verified actual levels of mRNA in each lane. Densito- metric scanning indicated that the erbB-3 transcript in each tumor cell line was elevated more than 100 fold above that of the control cell line. Thus, overexpression of this new member of the erbB family, as in the case of the EGF-R and erbB-2 genes, is likely to play an important role in some human malignancies.
* * * *
For purposes of completing the background descrip¬ tion and present disclosure, each of the published arti- cles, patents and patent applications heretofore identi¬ fied in this specification are hereby incorporated by reference into the specification.
The foregoing invention has been described in some detail for purposes of clarity and understanding. It will also be obvious that various changes and combinations in form and detail can be made without departing from the scope of the invention.

Claims

WHAT IS CLAIMED IS:
1. A DNA segment having a nucleotide sequence that encodes an erbB-3 gene or a unique portion thereof.
2. The DNA segment according to claim 1, wherein said gene is a mammalian erbB-3 gene.
3. The DNA segment according to claim 2, wherein said mammalian gene is a human erbB-3 gene.
4. A DNA segment having the nucleotide sequence, or a unique portion thereof, of a genomic DNA fragment that is produced by cleavage with the Sacl restriction enzyme, has a size of about 9 kbp, and is detectable by nucleic acid hybridization with a probe derived from the v-erbB gene only under reduced stringenpy hybridization conditions.
5. The DNA segment according to claim 4, wherein said segment is the human genomic DNA clone E3-1, or a unique portion thereof, said clone having the partial restriction enzyme map defined in Figure 2 and the partial DNA sequence defined in Figure 3.
6. The DNA segment according to claim 1, wherein said nucleotide sequence encodes the amino acid sequence of an erbB-3 gene or a unique portion thereof.
7. The DNA segment according to claim 6, wherein said amino acid sequence is that defined in Figure 4.
8. The DNA segment according to claim 7, compris¬ ing human cDNA clone E3-16 having the nucleotide sequence defined in Figure 4.
9. A DNA segment having a nucleotide sequence that encodes an amino acid sequence differing in at least one amino acid from the amino acid sequence of human erbB- 3, or a unique portion thereof, and having greater overall similarity to the amino acid sequence of human erbB-3 than to that of any other polypeptide.
10. The DNA segment according to claim 9 that encodes an amino acid sequence having substantially the function of the human erbB-3 polypeptide.
11. An isolated polypeptide having an amino acid sequence encoded by the DNA segment according to claim 9.
12. A recombinant DNA molecule comprising the DNA segment to claim 1 and a vector.
13. A culture of cells transformed with the DNA segment according to claim 1.
14. An isolate polypeptide having the amino acid sequence defined in Figure 4, or a unique portion thereof.
15. A bioassay for detecting erbB-3 mRNA in a biological sample comprising the steps of: i) contacting said biological sample with a DNA segment according to claim 1 under conditions such that a DNA:RNA hybrid molecule containing said DNA segment and complementary RNA can be formed; and i^ .... ii) determining the amount of said DNA segment present in said hybrid molecule.
16. A bioassay for testing potential analogs of ligands of erbB-3 receptors for the ability to affect an activity mediated by said erbB-3 receptors, comprising the steps of: i) contacting a molecule suspected of being a ligand with erbB-3 receptors produced by a cell according to claim 11; and ii) determining the amount of a biological activity mediated by said erbB-3 receptors in said cells.
17. An antibody specific for a unique portion of the polypeptide according to claim 14.
18. A bioassay for detecting an erbB-3 antigen in a biological sample comprising the steps of: i) contacting said sample with an antibody according to claim 17, under conditions such that a specific complex of said antibody and said antigen can be formed; and ii) determining the amount of said antibody present as said complexes.
19. A method for targeting a therapeutic drug to cells having high levels of erbB-3 receptors, comprising the steps of: i) conjugating an antibody according to claim 17, or an active fragment thereof, to said drug; and ii) administering the resulting conjugate to an individual with cells having high levels of erbB-3 receptors in an effective amount and by an effective route such that said antibody is able to bind to said receptor on said cells.
20. Use of the antibody of claim 17, or an active fragment thereof, conjugated to a therapeutic drug to target said therapeutic drug to cells having high levels of erbB-3 receptors.
PCT/US1990/007025 1989-12-01 1990-11-30 Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor WO1991008214A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP91900108A EP0502927B1 (en) 1989-12-01 1990-11-30 Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
DE69032133T DE69032133T2 (en) 1989-12-01 1990-11-30 DNA SECTION ENCODING A GENE FOR THE RECEPTOR RELATED TO THE EPIDERMAL GROWTH FACTOR RECEPTOR
ES91900108T ES2113877T3 (en) 1989-12-01 1990-11-30 DNA SEGMENT CODING A GENE FOR A RECEPTOR RELATED TO THE RECEPTOR OF THE EPIDERMIC GROWTH FACTOR.
CA002069900A CA2069900C (en) 1989-12-01 1990-11-30 Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
DK91900108.1T DK0502927T3 (en) 1989-12-01 1990-11-30 DNA segment encoding a gene for a receptor related to the epidermal growth factor receptor
JP50088591A JP3196079B2 (en) 1989-12-01 1990-11-30 DNA segment encoding a gene for a receptor related to the epidermal growth factor receptor
AU68899/91A AU654805B2 (en) 1989-12-01 1990-11-30 DNA segment encoding a gene for a receptor related to the epidermal growth factor receptor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US07/444,406 US5183884A (en) 1989-12-01 1989-12-01 Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
US444,406 1989-12-01

Publications (1)

Publication Number Publication Date
WO1991008214A1 true WO1991008214A1 (en) 1991-06-13

Family

ID=23764745

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/007025 WO1991008214A1 (en) 1989-12-01 1990-11-30 Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor

Country Status (10)

Country Link
US (6) US5183884A (en)
EP (1) EP0502927B1 (en)
JP (2) JP3196079B2 (en)
AT (1) ATE163970T1 (en)
AU (1) AU654805B2 (en)
CA (1) CA2069900C (en)
DE (1) DE69032133T2 (en)
DK (1) DK0502927T3 (en)
ES (1) ES2113877T3 (en)
WO (1) WO1991008214A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997035885A1 (en) * 1996-03-27 1997-10-02 Genentech, Inc. ErbB3 ANTIBODIES
US5811098A (en) * 1992-11-24 1998-09-22 Bristol-Myers Squibb Company Antibodies to HER4, human receptor tyrosine kinase
US5968511A (en) * 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
WO2000078347A1 (en) * 1999-06-18 2000-12-28 The Victor Chang Cardiac Research Institute Limited Cell growth inhibition
WO2003080835A1 (en) * 2002-03-26 2003-10-02 Zensun (Shanghai) Sci-Tech. Ltd. Erbb3 based methods and compositions for treating neoplasms
US8859737B2 (en) 2009-12-22 2014-10-14 Roche Glycart Ag Anti-HER3 antibodies and uses thereof
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US9180185B2 (en) 2013-01-11 2015-11-10 Hoffman-La Roche Inc. Combination therapy of anti-HER3 antibodies
EP3325005A4 (en) * 2015-07-17 2019-03-27 Brian J. Czerniecki Identification of immunogenic mhc class ii peptides for immune-based therapy
US10829538B2 (en) 2014-07-17 2020-11-10 The Trustees Of The University Of Pennsylvania Identification of immunogenic MHC class II peptides for immune-based therapy

Families Citing this family (172)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK0474727T3 (en) * 1989-05-19 1998-01-12 Genentech Inc HER2 extracellular domain
US5183884A (en) 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
US5578482A (en) * 1990-05-25 1996-11-26 Georgetown University Ligand growth factors that bind to the erbB-2 receptor protein and induce cellular responses
US5776755A (en) * 1992-10-09 1998-07-07 Helsinki University Licensing, Ltd. FLT4, a receptor tyrosine kinase
US6824777B1 (en) 1992-10-09 2004-11-30 Licentia Ltd. Flt4 (VEGFR-3) as a target for tumor imaging and anti-tumor therapy
US6107046A (en) * 1992-10-09 2000-08-22 Orion Corporation Antibodies to Flt4, a receptor tyrosine kinase and uses thereof
JPH09503381A (en) * 1993-07-09 1997-04-08 ジ・インスティテュート・オブ・キャンサー・リサーチ Protein tyrosine kinase and its ligand
JP4145953B2 (en) 1993-10-27 2008-09-03 ザ・トラスティーズ・オブ・コランビア・ユニバーシティー・イン・ザ・シティー・オブ・ニューヨーク MDA-7 gene
US5518885A (en) * 1994-04-19 1996-05-21 The United States Of America As Represented By The Department Of Health & Human Services ERBB2 promoter binding protein in neoplastic disease
US7175995B1 (en) 1994-10-27 2007-02-13 Thomas Jefferson University TCL-1 protein and related methods
US5985598A (en) * 1994-10-27 1999-11-16 Thomas Jefferson University TCL-1 gene and protein and related methods and compositions
US5635351A (en) * 1995-03-14 1997-06-03 The Regents Of The University Of California Genetic gain and loss in gliomas
US6685940B2 (en) 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
IL127891A0 (en) 1996-07-12 1999-10-28 Genentech Inc Chimeric heteromultimeter adhesins
IL127892A0 (en) * 1996-07-12 1999-10-28 Genentech Inc Gamma-heregulin
US5710137A (en) 1996-08-16 1998-01-20 The Trustees Of Columbia University In The City Of New York Use of a melanoma differentiation associated gene (mda 7) for reversing a cancerous phenotype
US7371376B1 (en) * 1996-10-18 2008-05-13 Genentech, Inc. Anti-ErbB2 antibodies
US6136558A (en) * 1997-02-10 2000-10-24 Genentech, Inc. Heregulin variants
ATE427353T1 (en) 1997-02-10 2009-04-15 Genentech Inc HEREGULIN VARIANTS
ATE323756T1 (en) * 1997-04-30 2006-05-15 Hans Klingemann NATURAL KILLER CELL LINES AND METHODS OF USE THEREOF
US7052692B1 (en) 1997-09-02 2006-05-30 Advanced Research & Technology Institute Role of tyrosine phosphorylation of a cellular protein in adeno-associated virus 2-mediated transgene expression
EP2033970A3 (en) 1997-10-29 2009-06-17 Genentech, Inc. Polypeptides and nucleic acids encoding the same
US6387657B1 (en) 1997-10-29 2002-05-14 Genentech, Inc. WISP polypeptides and nucleic acids encoding same
ZA9811162B (en) * 1997-12-12 2000-06-07 Genentech Inc Treatment with anti-ERBB2 antibodies.
US20010023241A1 (en) * 1998-02-04 2001-09-20 Sliwkowski Mark X. Use of heregulin as a growth factor
EP1941905A1 (en) 1998-03-27 2008-07-09 Genentech, Inc. APO-2 Ligand-anti-her-2 antibody synergism
EP1119371B1 (en) * 1998-10-09 2010-11-24 Vegenics Limited Flt4 (VEGFR-3) AS A TARGET FOR TUMOR IMAGING AND ANTI-TUMOR THERAPY
DK1165113T3 (en) * 1999-04-06 2008-09-08 Genentech Inc Use of ErbB receptor ligands in the treatment of diabetes
US20030086924A1 (en) * 1999-06-25 2003-05-08 Genentech, Inc. Treatment with anti-ErbB2 antibodies
WO2001000238A1 (en) * 1999-06-25 2001-01-04 Genentech, Inc. TREATING PROSTATE CANCER WITH ANTI-ErbB2 ANTIBODIES
AU784157B2 (en) 1999-06-25 2006-02-16 Genentech Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
HU226742B1 (en) 1999-06-25 2009-08-28 Genentech Inc Humanized anti-erbb2 antibodies and treatment with anti-erbb2 antibodies
US20040013667A1 (en) * 1999-06-25 2004-01-22 Genentech, Inc. Treatment with anti-ErbB2 antibodies
US6949245B1 (en) 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
US7041292B1 (en) 1999-06-25 2006-05-09 Genentech, Inc. Treating prostate cancer with anti-ErbB2 antibodies
NZ517150A (en) 1999-08-27 2005-01-28 Genentech Inc Dosages for treatment with anti-ErbB2 antibodies
US7390632B2 (en) * 1999-09-30 2008-06-24 Tumor Biology Investment Group, Inc. Soluble ErbB3 receptor isoforms
US7892541B1 (en) 1999-09-30 2011-02-22 Tumor Biology Investment Group, Inc. Soluble epidermal growth factor receptor isoforms
WO2001024763A2 (en) 1999-10-01 2001-04-12 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
US7097840B2 (en) 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
LT2857516T (en) 2000-04-11 2017-09-11 Genentech, Inc. Multivalent antibodies and uses therefor
WO2001083781A2 (en) * 2000-04-28 2001-11-08 Millennium Pharmaceuticals, Inc. 14094, a novel human trypsin family member and uses thereof
PL400669A1 (en) * 2000-05-19 2012-11-05 Genentech, Inc. Genetic probe for detecting a response to ErbB antagonist for improving the likelihood and efficiency of cancer treatment
CN1204142C (en) * 2000-06-01 2005-06-01 冯宝章 Antigene V-erb B oligonucleotide and its use
US6277640B1 (en) 2000-07-31 2001-08-21 Isis Pharmaceuticals, Inc. Antisense modulation of Her-3 expression
US20020119148A1 (en) * 2000-09-01 2002-08-29 Gerritsen Mary E. ErbB4 antagonists
EP1332368A2 (en) * 2000-11-03 2003-08-06 Board of Regents, The University of Texas System Methods for detecting the efficacy of anticancer treatments
AU2002248372B8 (en) * 2001-01-19 2008-03-20 Vegenics Limited FLT4(VEGFR-3) as a target for tumor imaging and anti-tumor therapy
EP1228766A1 (en) * 2001-01-31 2002-08-07 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. PYK2 phosphorylation by HER3 induces tumor invasion
WO2002081649A2 (en) * 2001-04-06 2002-10-17 The Trustees Of The University Of Pennsylvania ErbB INTERFACE PEPTIDOMIMETICS AND METHODS OF USE THEREOF
WO2002088363A2 (en) * 2001-05-02 2002-11-07 Bayer Healthcare Ag Regulation of novel human prolyl 4-hydroxylases
US7745398B2 (en) * 2001-05-31 2010-06-29 Tumor Biology Investment Group, Inc. Soluble ErbB3 and treatment of cancer
US7744882B2 (en) 2001-05-31 2010-06-29 Tumor Biology Investment Group, Inc. Soluble ErbB3 methods of detection and antibodies
US7638302B2 (en) * 2001-05-31 2009-12-29 Tumor Biology Investment Group, Inc. Soluble ErbB3 receptor isoforms
DE10131187C2 (en) * 2001-06-28 2003-06-18 Infineon Technologies Ag Exposure mask with repaired blind structure
WO2003011897A1 (en) 2001-07-27 2003-02-13 The Regents Of The University Of California Modulation of heregulin and her3 interaction
EP1283053A1 (en) * 2001-08-09 2003-02-12 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Inhibitors of HER3 activity
AU2002326910A1 (en) * 2001-09-14 2003-04-01 Incyte Genomics, Inc. Receptors and membrane-associated proteins
JP2003110360A (en) * 2001-09-28 2003-04-11 Seiko Epson Corp Voltage-controlled oscillator, receiving apparatus, and communications equipment
US20040229294A1 (en) 2002-05-21 2004-11-18 Po-Ying Chan-Hui ErbB surface receptor complexes as biomarkers
US7402397B2 (en) 2002-05-21 2008-07-22 Monogram Biosciences, Inc. Detecting and profiling molecular complexes
US20040229380A1 (en) * 2002-05-21 2004-11-18 Po-Ying Chan-Hui ErbB heterodimers as biomarkers
CA2490542C (en) 2002-05-23 2013-07-16 Mark I. Greene Fas peptide mimetics and uses thereof
JP5069843B2 (en) 2002-07-15 2012-11-07 ジェネンテック, インコーポレイテッド Method for identifying tumors responsive to treatment with anti-ErbB2 antibodies
US8505468B2 (en) * 2002-11-19 2013-08-13 Sharp Kabushiki Kaisha Substrate accommodating tray
JP2006516117A (en) * 2002-11-21 2006-06-22 ジェネンテック・インコーポレーテッド Treatment of non-malignant diseases or disorders using anti-ErbB2 antibodies
AU2004225439A1 (en) 2003-04-01 2004-10-14 Monogram Biosciences, Inc. Intracellular complexes as biomarkers
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
JP2007525187A (en) * 2003-05-16 2007-09-06 レセプター・バイオロジクス・インコーポレイテッド Intron fusion proteins and methods for identifying and using the same
US7402398B2 (en) * 2003-07-17 2008-07-22 Monogram Biosciences, Inc. Measuring receptor homodimerization
EP1673399B1 (en) * 2003-08-11 2017-04-05 Monogram BioSciences, Inc. Detecting and profiling molecular complexes
EP1681983A4 (en) * 2003-10-14 2008-12-10 Monogram Biosciences Inc Receptor tyrosine kinase signaling pathway analysis for diagnosis and therapy
BR122018071808B8 (en) 2003-11-06 2020-06-30 Seattle Genetics Inc conjugate
CA2563341C (en) * 2004-04-08 2014-10-21 David B. Agus Erbb antagonists for pain therapy
WO2005113596A2 (en) * 2004-05-14 2005-12-01 Receptor Biologix, Inc. Cell surface receptor isoforms and methods of identifying and using the same
BRPI0510883B8 (en) 2004-06-01 2021-05-25 Genentech Inc drug-antibody conjugate compound, pharmaceutical composition, method of manufacturing a drug-antibody conjugate compound, and uses of a formulation, a drug-antibody conjugate and a chemotherapeutic agent, and a combination
GT200500155A (en) * 2004-06-16 2006-05-15 PLATINUM-RESISTANT CANCER THERAPY
JP2008507520A (en) * 2004-07-22 2008-03-13 ジェネンテック・インコーポレーテッド HER2 antibody composition
RU2412947C2 (en) 2004-09-23 2011-02-27 Дженентек, Инк. Antibodies, constructed on cysteine basis and their conjugates
US20100111856A1 (en) 2004-09-23 2010-05-06 Herman Gill Zirconium-radiolabeled, cysteine engineered antibody conjugates
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US7939267B2 (en) * 2004-11-04 2011-05-10 Laboratory Corporation Of America Holdings Detection of activation of endothelial cells as surrogate marker for angiogenesis
AU2005314127A1 (en) * 2004-12-07 2006-06-15 Genentech, Inc. Selecting patients for therapy with a HER inhibitor
KR20190110637A (en) 2005-01-21 2019-09-30 제넨테크, 인크. Fixed dosing of her antibodies
KR20070100968A (en) * 2005-02-09 2007-10-15 제넨테크, 인크. Inhibiting her2 shedding with matrix metalloprotease antagonists
RU2404806C2 (en) * 2005-02-23 2010-11-27 Дженентек, Инк. Extension of time to progression of disease or lifetime of oncologic patients with application of her dimerisation inhibitors
US20060204505A1 (en) * 2005-03-08 2006-09-14 Sliwkowski Mark X Methods for identifying tumors responsive to treatment with HER dimerization inhibitors (HDIs)
JP2006316040A (en) 2005-05-13 2006-11-24 Genentech Inc Herceptin(r) adjuvant treatment
US20090170769A1 (en) * 2005-05-13 2009-07-02 Pei Jin Cell surface receptor isoforms and methods of identifying and using the same
AR056857A1 (en) 2005-12-30 2007-10-24 U3 Pharma Ag DIRECTED ANTIBODIES TO HER-3 (RECEIVER OF THE HUMAN EPIDERMAL GROWTH FACTOR-3) AND ITS USES
DK2511301T3 (en) 2006-08-04 2018-03-12 Medimmune Ltd HUMAN ANTIBODIES AGAINST ERBB 2
EP2097754B2 (en) * 2006-11-28 2018-01-24 Daiichi Sankyo Europe GmbH Activated her3 as a marker for predicting therapeutic efficacy
EP3248617A3 (en) 2007-02-16 2018-02-21 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
PE20090681A1 (en) 2007-03-02 2009-06-10 Genentech Inc PREDICTION OF RESPONSE TO A HER INHIBITOR
KR101540822B1 (en) 2007-03-27 2015-07-30 씨 레인 바이오테크놀로지스, 엘엘씨 constructs and libraries comprising antibody surrogate light chain sequences
WO2008154249A2 (en) 2007-06-08 2008-12-18 Genentech, Inc. Gene expression markers of tumor resistance to her2 inhibitor treatment
WO2009070772A1 (en) * 2007-11-27 2009-06-04 Monogram Biosciences, Inc. Enhanced method for detecting and/or quantifying an analyte in a sample
EP2235536A4 (en) * 2007-12-20 2011-05-04 Lab Corp America Holdings Her-2 diagnostic methods
BRPI0822012A2 (en) 2008-01-18 2015-07-21 Natco Pharma Ltd 6,7-Dialkoxy quinazoline derivatives useful for treating cancer-related disorders
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
ES2629405T3 (en) 2008-03-18 2017-08-09 Genentech, Inc. Combinations of a drug-antibody anti-HER2 and docetaxel conjugate
BRPI0812682A2 (en) 2008-06-16 2010-06-22 Genentech Inc metastatic breast cancer treatment
WO2009157919A1 (en) * 2008-06-23 2009-12-30 Tumor Biology Investment Group, Inc. SOLUBLE ErbB3 DETECTION, REGULATION AND TREATMENT OF CANCER
UA104868C2 (en) * 2008-08-15 2014-03-25 Меррімак Фармасьютікалз, Інк. Method for treating patient having neoplastic tumor according to predicted reaction
CA2764386C (en) 2008-12-01 2018-05-01 Laboratory Corporation Of America Holdings P95-her2 antibodies and uses thereof
WO2010083470A1 (en) 2009-01-15 2010-07-22 Laboratory Corporation Of America Holdings Methods of determining patient response by measurement of her-3
TW201544123A (en) 2009-03-20 2015-12-01 Genentech Inc Anti-HER antibodies
EP2425009A4 (en) 2009-04-29 2013-01-23 Trellis Bioscience Llc Improved antibodies immunoreactive with heregulin-coupled her3
CN102482345A (en) 2009-05-13 2012-05-30 航道生物技术有限责任公司 Neutralizing molecules to influenza viruses
CA2761280A1 (en) 2009-05-29 2010-12-02 F. Hoffmann-La Roche Ag Modulators for her2 signaling in her2 expressing patients with gastric cancer
US9050341B2 (en) * 2009-07-14 2015-06-09 Natco Pharma Limited Methods of treating drug resistant and other tumors by administering 6,7-dialkoxy quinazoline derivatives
EP2488199A1 (en) 2009-10-14 2012-08-22 Merrimack Pharmaceuticals, Inc. Bispecific binding agents targeting igf-1r and erbb3 signalling and uses thereof
LT3351558T (en) * 2009-11-13 2020-06-10 Daiichi Sankyo Europe Gmbh Material and methods for treating or preventing her-3 associated diseases
CA2781682A1 (en) 2009-12-04 2011-06-09 Genentech, Inc. Multispecific antibodies, antibody analogs, compositions, and methods
WO2011112953A2 (en) 2010-03-11 2011-09-15 Merrimack Pharmaceuticals, Inc. Use of erbb3 inhibitors in the treatment of triple negative and basal-like breast cancers
US8481687B2 (en) 2010-04-09 2013-07-09 Aveo Pharmaceuticals, Inc. Anti-ErbB3 antibodies
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
US8741288B2 (en) * 2010-07-07 2014-06-03 Chang Gung Medical Foundation, Linkou Branch Protein markers for detecting liver cancer and method for identifying the markers thereof
BR112013004012B1 (en) 2010-08-20 2021-03-23 Novartis Ag ISOLATED MONOCLONAL ANTIBODY OR ANTIGEN BINDING FRAGMENT OF THE SAME TO THE HER3 RECEPTOR, ITS USE AND PHARMACEUTICAL COMPOSITION
JP5766296B2 (en) 2010-12-23 2015-08-19 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Polypeptide-polynucleotide complexes and their use in targeted delivery of effector components
CA2828075A1 (en) 2011-03-11 2012-09-20 Merrimack Pharmaceuticals, Inc. Use of inhibitors of egfr-family receptors in the treatment of hormone refractory breast cancers
KR101517320B1 (en) 2011-04-19 2015-05-28 메리맥 파마슈티컬즈, 인크. Monospecific and bispecific anti-igf-1r and anti-erbb3 antibodies
US9346883B2 (en) 2011-05-13 2016-05-24 Institut National De La Sante Et De La Recherche Medicale (Inserm) Antibodies against HER3
JP6468838B2 (en) 2011-05-19 2019-02-13 ラボラトリー コーポレイション オブ アメリカ ホールディングス Methods for determining the survival potential of cancer patients and for predicting the likelihood of metastasis in cancer patients
CA2840461A1 (en) 2011-06-20 2012-12-27 Kyowa Hakko Kirin Co., Ltd. Anti-erbb3 antibody
US10300140B2 (en) 2011-07-28 2019-05-28 I2 Pharmaceuticals, Inc. Sur-binding proteins against ERBB3
EP2751285B2 (en) 2011-08-31 2020-04-01 Genentech, Inc. Method for sensitivity testing of a tumour for a egfr kinase inhibitor
US9273143B2 (en) 2011-09-30 2016-03-01 Regeneron Pharmaceuticals, Inc. Methods and compositions comprising a combination of an anti-ErbB3 antibody and an anti-EGFR antibody
IN2014CN03042A (en) 2011-09-30 2015-07-03 Regeneron Pharma
CN103959065B (en) 2011-10-06 2018-07-17 Aveo制药公司 Predict the response of tumour confrontation ERBB3 antibody
CN116271013A (en) 2011-10-14 2023-06-23 霍夫曼-拉罗奇有限公司 Use of the HER2 dimerization inhibitor pertuzumab and articles of manufacture comprising the HER2 dimerization inhibitor pertuzumab
DK2797957T3 (en) 2011-11-23 2019-09-23 Medimmune Llc BINDING MOLECULES SPECIFIC TO HER3 AND APPLICATIONS THEREOF
WO2013081645A2 (en) 2011-11-30 2013-06-06 Genentech, Inc. Erbb3 mutations in cancer
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
JP2015500829A (en) 2011-12-05 2015-01-08 ノバルティス アーゲー HER3 antibody against domain II of epidermal growth factor receptor 3 (HER3)
WO2013083810A1 (en) 2011-12-09 2013-06-13 F. Hoffmann-La Roche Ag Identification of non-responders to her2 inhibitors
WO2013096828A1 (en) 2011-12-22 2013-06-27 Sea Lane Biotechnologies, Llc Surrogate binding proteins
WO2013124419A1 (en) 2012-02-23 2013-08-29 U3 Pharma Gmbh Her3 inhibitor for modulating radiosensitivity
RU2014136886A (en) 2012-03-27 2016-05-20 Дженентек, Инк. DIAGNOSTIC AND TREATMENT TYPES RELATED TO HER3 INHIBITORS
MX2014011925A (en) 2012-04-02 2015-05-11 Merrimack Pharmaceuticals Inc Dosage and administration of monospecific and bispecific anti-igf-1r and anti-erbb3 antibodies.
MX363188B (en) 2012-11-30 2019-03-13 Hoffmann La Roche Identification of patients in need of pd-l1 inhibitor cotherapy.
EP2954068B8 (en) 2013-02-05 2018-10-31 Pierian Holdings, Inc. Drug selection for non-small cell lung cancer therapy
RS60026B1 (en) 2013-02-18 2020-04-30 Vegenics Pty Ltd Ligand binding molecules and uses thereof
CN105264382A (en) 2013-04-05 2016-01-20 美国控股实验室公司 Systems and methods for facilitating diagnosis, prognosis and treatment of cancer based on detection of HER3 activation
CA2910945A1 (en) 2013-05-08 2014-11-13 Zymeworks Inc. Bispecific her2 and her3 antigen binding constructs
US11305012B2 (en) 2013-09-24 2022-04-19 Medimmune, Llc Binding molecules specific for HER3 and uses thereof
EP3087394A2 (en) 2013-12-27 2016-11-02 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with erbb3 inhibitors and/or chemotherapies
MA39776A (en) 2014-03-24 2017-02-01 Hoffmann La Roche Cancer treatment with c-met antagonists and correlation of the latter with hgf expression
DK3129063T3 (en) 2014-04-10 2021-04-06 Daiichi Sankyo Co Ltd ANTI-HER3 ANTIBODY-MEDICINE CONJUGATE
WO2015157634A1 (en) 2014-04-11 2015-10-15 Kolltan Pharmaceuticals, Inc. Anti-erbb antibodies and methods of use thereof
EP3539990B1 (en) 2014-07-16 2021-09-08 Dana-Farber Cancer Institute, Inc. Her3 inhibition in low-grade serous cancers
US10689459B2 (en) 2014-12-12 2020-06-23 Novartis Ag Treatment of breast cancer brain metastases
WO2016196377A1 (en) 2015-05-29 2016-12-08 Merrimack Pharmaceuticals, Inc. Combination cancer therapies
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
AU2016286898B2 (en) 2015-06-29 2022-12-08 Daiichi Sankyo Company, Limited Method for selectively manufacturing antibody-drug conjugate
US20170233491A1 (en) 2016-01-19 2017-08-17 Merrimack Pharmaceuticals, Inc. Dosage and administration of combination therapies comprising istiratumab, uses and methods of treatment
MX2018011054A (en) 2016-03-15 2019-01-21 Merrimack Pharmaceuticals Inc Methods for treating er+, her2-, hrg+ breast cancer using combination therapies comprising an anti-erbb3 antibody.
EP3454863A1 (en) 2016-05-10 2019-03-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Combinations therapies for the treatment of cancer
TW201828993A (en) 2016-12-12 2018-08-16 日商第一三共股份有限公司 Combination of antibody-drug conjugate and immune checkpoint inhibitor
TW201827077A (en) 2016-12-28 2018-08-01 美商建南德克公司 Treatment of advanced her2 expressing cancer
LT3570884T (en) 2017-01-17 2020-12-10 Genentech, Inc. Subcutaneous her2 antibody formulations
KR102537651B1 (en) 2017-01-17 2023-05-26 다이이찌 산쿄 가부시키가이샤 Anti-GPR20 Antibodies and Anti-GPR20 Antibody-Drug Conjugates
AU2018227788B2 (en) 2017-03-02 2021-04-22 F. Hoffmann-La Roche Ag Adjuvant treatment of HER2-positive breast cancer
WO2018200505A1 (en) 2017-04-24 2018-11-01 Genentech, Inc. Erbb2/her2 mutations in the transmbrane or juxtamembrane domain
TW202330036A (en) 2017-05-15 2023-08-01 日商第一三共股份有限公司 Manufacturing method of antibody-drug conjugates
CA3063747C (en) * 2017-06-02 2023-01-03 Etubics Corporation Compositions and methods for tumor vaccination and immunotherapy involving her antigens
CN117838881A (en) 2017-08-31 2024-04-09 第一三共株式会社 New method for preparing antibody-drug conjugate
KR20200041993A (en) 2017-08-31 2020-04-22 다이이찌 산쿄 가부시키가이샤 Method for improved production of antibody-drug conjugates
CN117815404A (en) 2018-05-18 2024-04-05 第一三共株式会社 anti-MUC 1 antibody-drug conjugates
AU2020456731A1 (en) 2020-06-29 2023-01-05 F. Hoffmann-La Roche Ag Pertuzumab plus trastuzumab fixed dose combination
EP4180061A1 (en) 2021-11-10 2023-05-17 Nerviano Medical Sciences S.r.l. Anthracycline derivative linker reagents, antibody-drug conjugates and methods

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4867973A (en) * 1984-08-31 1989-09-19 Cytogen Corporation Antibody-therapeutic agent conjugates

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4480968A (en) * 1984-04-05 1984-11-06 General Motors Corporation Two-cycle engine compressor
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
GB8810400D0 (en) * 1988-05-03 1988-06-08 Southern E Analysing polynucleotide sequences
GB8910977D0 (en) 1989-05-12 1989-06-28 Rolls Royce Plc Metal matrix composite material
US5183884A (en) * 1989-12-01 1993-02-02 United States Of America Dna segment encoding a gene for a receptor related to the epidermal growth factor receptor
CA2037440A1 (en) * 1990-03-02 1991-09-03 Gregory D. Plowman Her3: a novel egf receptor homolog

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4867973A (en) * 1984-08-31 1989-09-19 Cytogen Corporation Antibody-therapeutic agent conjugates

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Nature, Vol. 319, issued 16 January 1986, YAMAMOTO et al., "Similarity of Protein Encoded by the Human c-erbB-2 Gene to Epidermal Growth Factor Receptor", pages 230-234. *
Proceedings of the National Academy of Sciences, Vol. 86, issued December 1989, KRAUS et al., "Isolation and Characterization of ERBB3. A Third Member of the ERBB/Epidermal Growth Factor Receptor Family: Evidence for Overexpression in a Subset of Human Mammary Tumors", pages 9193-9197. *
Science, Vol. 230, issued 06 December 1985, COUSSENS et al., "Tyrosine Kinase Receptor with Extensive Homology to EGF Receptor Shares Chromosomal Location with neu Oncogene", pages 1132-1139. *
Science, Vol. 237, issued 10 July 1987, DIFIORE et al., "erbB-2 Is a Potent Oncogene When Overexpressed in NIH/3T3 cells", pages 178-182. *
Sciences, Vol. 229, issued 06 September 1985, KING et al., "Amplification of a Novel v-erbB -Related Gene in a Human Mammary Carcinoma", pages 974-978. *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811098A (en) * 1992-11-24 1998-09-22 Bristol-Myers Squibb Company Antibodies to HER4, human receptor tyrosine kinase
WO1997035885A1 (en) * 1996-03-27 1997-10-02 Genentech, Inc. ErbB3 ANTIBODIES
US5968511A (en) * 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
EP1728802A2 (en) * 1996-03-27 2006-12-06 Genentech, Inc. ErbB3 antibodies
EP1728802A3 (en) * 1996-03-27 2006-12-13 Genentech, Inc. ErbB3 antibodies
US7285649B2 (en) 1996-03-27 2007-10-23 Genentech, Inc. Isolated nucleic acids, vectors and host cells encoding ErbB3 antibodies
WO2000078347A1 (en) * 1999-06-18 2000-12-28 The Victor Chang Cardiac Research Institute Limited Cell growth inhibition
US9783456B1 (en) 1999-06-18 2017-10-10 Zensun (Shanghai) Science & Technology, Co., Ltd. Method for inhibiting cell growth using anti-ErbB-3 and anti-ErbB-2 antibodies
AU2003218600C1 (en) * 2002-03-26 2009-12-17 Zensun (Shanghai) Science & Technology Co., Ltd. ERBB3 based methods and compositions for treating neoplasms
AU2003218600B2 (en) * 2002-03-26 2009-05-21 Zensun (Shanghai) Science & Technology Co., Ltd. ERBB3 based methods and compositions for treating neoplasms
CN100424175C (en) * 2002-03-26 2008-10-08 上海泽生科技开发有限公司 Methods and compositions for treating neoplasms by ERBB3
US7919098B2 (en) 2002-03-26 2011-04-05 Zensun ( Shanghai ) Sci & Tech Co., Ltd. ErbB-3 based methods and compositions for treating neoplasms
WO2003080835A1 (en) * 2002-03-26 2003-10-02 Zensun (Shanghai) Sci-Tech. Ltd. Erbb3 based methods and compositions for treating neoplasms
US8859737B2 (en) 2009-12-22 2014-10-14 Roche Glycart Ag Anti-HER3 antibodies and uses thereof
US9611331B2 (en) 2009-12-22 2017-04-04 Roche Glycart Ag Anti-HER3 antibodies and uses thereof
US10196456B2 (en) 2009-12-22 2019-02-05 Roche Glycart Ag Anti-HER3 antibodies and uses thereof
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US9180185B2 (en) 2013-01-11 2015-11-10 Hoffman-La Roche Inc. Combination therapy of anti-HER3 antibodies
US10829538B2 (en) 2014-07-17 2020-11-10 The Trustees Of The University Of Pennsylvania Identification of immunogenic MHC class II peptides for immune-based therapy
EP3325005A4 (en) * 2015-07-17 2019-03-27 Brian J. Czerniecki Identification of immunogenic mhc class ii peptides for immune-based therapy

Also Published As

Publication number Publication date
EP0502927A4 (en) 1993-05-26
DE69032133T2 (en) 1998-07-02
EP0502927A1 (en) 1992-09-16
ES2113877T3 (en) 1998-05-16
JPH06503467A (en) 1994-04-21
AU654805B2 (en) 1994-11-24
US20040063140A1 (en) 2004-04-01
JP3632171B2 (en) 2005-03-23
AU6889991A (en) 1991-06-26
US5916755A (en) 1999-06-29
EP0502927B1 (en) 1998-03-11
JP3196079B2 (en) 2001-08-06
JP2001299373A (en) 2001-10-30
US5820859A (en) 1998-10-13
CA2069900A1 (en) 1991-06-02
US5480968A (en) 1996-01-02
ATE163970T1 (en) 1998-03-15
US6639060B1 (en) 2003-10-28
DE69032133D1 (en) 1998-04-16
DK0502927T3 (en) 1998-06-02
US5183884A (en) 1993-02-02
CA2069900C (en) 2004-07-27

Similar Documents

Publication Publication Date Title
AU654805B2 (en) DNA segment encoding a gene for a receptor related to the epidermal growth factor receptor
Gullick et al. Expression of the c‐erbB‐2 protein in normal and transformed cells
Rodrigues et al. Dimerization mediated through a leucine zipper activates the oncogenic potential of the met receptor tyrosine kinase
ES2230537T3 (en) A TYPE REINFORCING TYPE KINASE (EPH / ELK HUNABA-HENKI TYPE KINASE AND USE OF IT.
AU670704B2 (en) Peptide inhibitors of mitogenesis and motogenesis
JP2004043486A (en) DETECTION OF neu GENE PRODUCT
JPH05508550A (en) Fibroblast growth factor receptor
US6608181B2 (en) Fibroblast growth factor receptor activating gene I and related compositions and methods
US6238918B1 (en) DNA sequence encoding the tumor suppressor gene ING1
Lorenzi et al. FRAG1, a gene that potently activates fibroblast growth factor receptor by C-terminal fusion through chromosomal rearrangement.
US8703917B2 (en) Epidermal growth factor receptor variants and pharmaceutical compositions thereof
EP0646129A1 (en) Receptor-type tyrosine kinase-like molecules
US8796421B2 (en) Human epidermal growth factor receptor variant lacking an exon
US5716782A (en) Nucleic acid encoding a signal mediator protein that induces cellular morphological alterations
EP0797662A1 (en) Methods for treatment or diagnosis of diseases or conditions associated with abnormal signal transduction
US5807989A (en) Methods for treatment or diagnosis of diseases or disorders associated with an APB domain
Ihle et al. Phenotypes and mechanisms in the transformation of hematopoietic cells
US6673911B1 (en) Human polyhomeotic 2 (hph2) acts as an oncogene
JP2001527418A (en) Neurotrophic factor receptor from glial cell line
US6335169B1 (en) Nucleic acids encoding hBub1, a cell cycle checkpoint gene
US6747133B1 (en) Antibodies against the tumor suppressor gene ING1
AU4050593A (en) Receptor-type tyrosine kinase-like molecules

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

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

COP Corrected version of pamphlet

Free format text: PAGES 1/8-8/8,DRAWINGS,REPLACED BY NEW PAGES 1/18-18/18

WWE Wipo information: entry into national phase

Ref document number: 2069900

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1991900108

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1991900108

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1991900108

Country of ref document: EP