US20110229478A1 - Erbb3 based methods and compositions for treating neoplasms - Google Patents

Erbb3 based methods and compositions for treating neoplasms Download PDF

Info

Publication number
US20110229478A1
US20110229478A1 US13/035,244 US201113035244A US2011229478A1 US 20110229478 A1 US20110229478 A1 US 20110229478A1 US 201113035244 A US201113035244 A US 201113035244A US 2011229478 A1 US2011229478 A1 US 2011229478A1
Authority
US
United States
Prior art keywords
antibody
erbb
protein
amino acid
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/035,244
Inventor
Mingdong Zhou
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zensun Shanghai Science and Technology Ltd
Original Assignee
Zensun Shanghai Science and Technology Ltd
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
Priority claimed from CNB02116259XA external-priority patent/CN1219882C/en
Application filed by Zensun Shanghai Science and Technology Ltd filed Critical Zensun Shanghai Science and Technology Ltd
Priority to US13/035,244 priority Critical patent/US20110229478A1/en
Publication of US20110229478A1 publication Critical patent/US20110229478A1/en
Assigned to ZENSUN (SHANGHAI) SCIENCE & TECHNOLOGY, CO., LTD. reassignment ZENSUN (SHANGHAI) SCIENCE & TECHNOLOGY, CO., LTD. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ZENSUN (SHANGHAI) SCIENCE & TECHNOLOGY, LTD.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4747Apoptosis related proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/179Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001103Receptors for growth factors
    • A61K39/001106Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to compositions and methods for treating neoplasms in mammals, particularly humans. More particularly, the present invention provides for methods for preventing, treating or delaying neoplasm in a mammal using an ErbB-3 protein, a nucleic acid encoding an ErbB-3 protein or a functional fragment thereof.
  • the present invention also provides for isolated nucleic acids encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, substantially purified extracellular domain of the ErbB-3 protein, or a functional fragment thereof and antibodies that bind to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof.
  • the present invention further provides for pharmaceutical compositions and/or vaccines comprising the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, or nucleic acids encoding and antibodies binding to such extracellular domain or functional fragments thereof.
  • Cancer is a major lethal disease for humans and is caused by physiologically-uncontrolled cell proliferation which affects normal physiological conditions of human body resulting in serious pathological reactions often leading to death. Although tremendous efforts on cancer studies and treatments have been made, presently, cancer is still the major cause of death to humans. There are multiple approaches to treat cancer patients including surgery, radiation therapy and chemotherapy. As the first two methods are not able to completely eliminate cancer cells in patients, the latter approach is commonly used to control cancer cell growth with or without other treatments. Anti-cancer compounds used in patients are often targeting prevention of cancer cell proliferation or killing dividing cells.
  • ErbBs are class one receptor protein tyrosine kinases. ErbB-mediated cell signaling plays a critical role in embryo development and adult organ function. On a cellular level, ErbB receptors have been shown to mediate signals for cell proliferation, differentiation, migration, and cell structure reorganization. There are four structurally similar ErbB members, ErbB-1, ErbB-2, ErbB-3 and ErbB-4.
  • the epidermal growth factor (EGF) is one of several ligands that bind ErbB-1, ErbB-3 or ErbB-4 also bind several ligands, including neuregulin-1 (NRG-1). To date, no ligand for ErbB-2 has been identified.
  • ErbB-2 serves as a heterodimer partner for ErbB-3, ErbB-4 or ErbB-1 and is critically involved in NRG-1-activated cell signaling.
  • In vivo studies using gene targeting experiments indicate that developmental defects resulting from inactivation of ErbB-2 are similar to those observed in NRG-1-inactivated animals. Both animals show defects in the neural crania ganglia and heart trabeculae development.
  • ErbB-3 or ErbB-4 gene-inactivated mice have similar or overlapping phenotypes to NRG-1 or ErbB-2 knockout mice.
  • ErbB-2 In addition to its role in development, the human ErbB-2 gene is frequently amplified and its encoded protein is over-expressed in a variety of human carcinomas.
  • ErbB-2 Early research on ErbB-2 discovered that an oncogenic point mutation resulted in the formation of ErbB-2 homodimers that in turn caused significant phosphorylation of the tyrosine residues on the intracellular domain. While no corresponding point mutation has been found in ErbB-2 over expressing human carcinomas, the upregulation of ErbB-2 results in the formation of homodimers that in turn increases the tyrosine phosphorylation of its intracellular domain. This process is hypothesized to be the start of a signal cascade that triggers cell transformation and/or growth, and thus initiate tumorigenesis.
  • ErbB-2 acts as a heterodimer partner for the ligand-binding ErbB-3 or ErbB-4 receptors.
  • the ligand, NRG-1 has been identified to have two independent receptor binding sites: one that has a high affinity for ErbB-3 or ErbB-4, and the other that has a low but non-specific affinity for all ErbB members.
  • the exposure of NRG-1 to cells expressing ErbB-3/4 and ErbB-2 would result in heterodimers of ErbB-2 and ErbB-3/4.
  • ErbB-3 is unique because: i) ErbB-2 preferentially forms heterodimers with ErbB-3; ii) cotransfection of NIH3T3 cells with ErbB-2 and ErbB-3 results in much higher levels of cell transformation than that of transfection with ErbB-2 alone; iii) in ErbB-2 over-expression-associated breast cancer cells, ErbB-3 is also highly expressed; and iv) ErbB-3 is also over expressed in ErbB-2-over expressing tumour cells from ErbB-2 transgenic mice.
  • WO 00/78347 discloses methods for arresting or inhibiting cell growth, particularly cancer cell growth, comprising preventing or reducing ErbB-2/ErbB-3 heterodimer formation, or interfering with ErbB-2/ErbB-3 heterodimer conformation in a cell and agents which prevent or reduce ErbB-2/ErbB-3 heterodimer formation or interfere with ErbB-2/ErbB-3 heterodimer conformation in a cell thereby arresting or inhibiting the growth of the cell.
  • 5,578,482 relates to erbB-2 ligands and functional derivatives thereof which are capable of binding to the erbB-2 oncogene product.
  • U.S. Pat. No. 5,820,859 relates to a method of targeting a therapeutic agent to cells expressing the erb B-3 receptor.
  • U.S. Pat. No. 5,968,511 relates to ErbB3 antibodies.
  • the present invention is directed to a method for preventing, treating or delaying neoplasm in a mammal, which method comprises administering to a mammal, to which such prevention, treatment or delay is needed or desirable, an effective amount of an ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, whereby an immune response is generated against said neoplasm and said neoplasm is prevented, treated or delayed.
  • the present invention is directed to an isolated nucleic acid fragment, which isolated nucleic acid fragment comprises a sequence of nucleotides encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 ( FIG. 5 ) or SEQ ID NO:3 ( FIG. 11 ) or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the present invention is directed to a substantially purified protein or peptide, which comprises an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the present invention is directed to a conjugate, which conjugate comprises: a) a protein or peptide comprising an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.; and b) a facilitating agent linked to the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, directly or via a linker, wherein the agent facilitates: i) affinity isolation or purification of a conjugate; ii) attachment of a conjugate to a surface; or iii) detection of a conjugate.
  • the present invention is directed to an antibody, which antibody binds to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • compositions and/or vaccines comprising the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, or nucleic acids encoding and antibodies binding to such extracellular domain or functional fragments thereof are also provided.
  • FIG. 1 depicts B3 cDNA sequence (SEQ ID NO:4).
  • FIG. 2 illustrates restriction enzyme digestion of B3 plasmid.
  • Lane 10 pCDNA3 vector alone digested with BamHI/XbaI.
  • FIG. 3 illustrates B3 plasmid construction.
  • FIG. 4 illustrates isolation and/or purification and SDS-PAGE analysis of B3 protein.
  • Lane 1-4 BSA control, 10 ⁇ g, 5 ⁇ g, 3 ⁇ g, 1 ⁇ g/lane respectively.
  • Lane 5 Protein marker, 7708S NEB.
  • Lane 6-7 B3 protein expressed for COST.
  • FIG. 5 depicts B3 amino acid sequence (SEQ ID NO:2).
  • FIG. 6 depicts DE3-1 cDNA sequence (SEQ ID NO:5).
  • FIG. 7 illustrates DE3-1 plasmid construction.
  • FIG. 8 illustrates restriction enzyme digestion of DE3-1 plasmid.
  • Lane 1 DE3-1 in pGEX4T-1, cut with BamHI/XhoI.
  • Lane 2 DE3-1 in pET32a, cut with BamHI/XhoI.
  • Lane 3 1 Kd ladder (NEB).
  • FIG. 9 illustrates SDS-PAGE analysis of DE3-1 expression. Lane 1: before induction. Lane 2: after induction. Lane 3: inclusion body. Lane 4: supernatant after sonication.
  • FIG. 10 illustrates isolation and/or purification and SDS-PAGE analysis of DE3-1 protein.
  • Lane 1 Flow through.
  • Lane 2-8 Eluates from NTA His tag affinity column.
  • FIG. 11 depicts DE3-1 amino acid sequence (SEQ ID NO:3).
  • FIG. 12 illustrates the effect of various vaccines on incidence of FVB/N transgenic mice.
  • FIG. 13 illustrates the effect of various drugs on tumor growth in mice (5 weeks).
  • FIG. 14 illustrates the tumor-inhibitory effect of various drugs against tumor growth (5 weeks).
  • FIG. 15 illustrates the effect of DE3-1 on the growth of breast cancer in mice (5 weeks).
  • FIG. 16 illustrates the tumor-inhibitory rate of DE3-1 against tumor growth (5 weeks).
  • FIG. 17 illustrates experiment on cross immunity between B2 and B3 antigen (B3 protein wrapped).
  • FIG. 18 illustrates experiment on cross immunity between B2 and B3 antigen (B2 protein wrapped).
  • FIG. 19 illustrates Result of PCR amplification
  • Lane 2 192 bp ErbB3-f12 gene obtained by RT-PCR; Lane 1: DNA marker
  • FIG. 20 illustrates Screening for expression engineering strain.
  • FIG. 21 illustrates Experimental results of anti-tumor effect of rhErbB3-f12.
  • FIG. 22 illustrates Experimental results of anti-tumor effect of rhErbB3-f78.
  • FIG. 23 depicts ErbB3-f12 amino acid sequence (SEQ ID NO:14) and cDNA sequence (SEQ ID NO:17).
  • FIG. 24 depicts ErbB3-f78 cDNA sequence (SEQ ID NO:15).
  • FIG. 25 depicts ErbB3-f78 amino acid sequence (SEQ ID NO:16).
  • neoplasm neoplasia
  • neoplastic proliferation persists even in the absence of the original stimulus.
  • cancer refers to a general term for diseases caused by any type of malignant tumor.
  • malignant refers to primary tumors that have the capacity of metastasis with loss of both growth control and positional control.
  • erb refers to two oncogenes, erb A and erb B, associated with erythroblastosis virus (an acute transforming retrovirus).
  • immune response refers to alteration in the reactivity of an organism's immune system in response to an antigen; in vertebrates, this may involve antibody production, induction of cell-mediated immunity, complement activation or development of immunological tolerance.
  • immune response potentiator refers to a substance that enhances an antigen's effect in eliciting an immune response.
  • vaccine refers to any compositions intended for active immunological prophylaxis.
  • a vaccine may be used therapeutically to treat a disease, or to prevent development of a disease or to decrease the severity of a disease either proactively or after infection.
  • Exemplary vaccines include, but are not limited to, preparations of killed microbes of virulent strains or living microbes of attenuated (variant or mutant) strains, or microbial, fungal, plant, protozoa, or metazoa derivatives or products.
  • Vaccine also encompasses protein/peptide and nucleic acid/oligonucleotides based vaccines.
  • anti-neoplasm agent (used interchangeably with anti-neoplastic agent, anti-tumor or anti-cancer agent) refers to any agents used in the anti-neoplasm treatment. These include any agents, that when used alone or in combination with other compounds, can alleviate, reduce, ameliorate, prevent, or place or maintain in a state of remission of clinical symptoms or diagnostic markers associated with neoplasm, tumor or cancer, and can be used in methods, combinations and compositions provided herein.
  • Anti-neoplastic agents include, but are not limited to, anti-angiogenic agents, alkylating agents, antimetabolite, certain natural products, platinum coordination complexes, anthracenediones, substituted ureas, methylhydrazine derivatives, adrenocortical suppressants, certain hormones and antagonists, anti-cancer polysaccharides and certain herb extracts such as Chinese herb extracts.
  • an “anti-neoplastic treatment” refers to any treatment designed to treat the neoplasm, tumor or cancer by lessening or ameliorating its symptoms. Treatments that prevent the occurrence of neoplasm, tumor or cancer or lessen its severity are also contemplated.
  • anti-neoplasm agent or anti-tumor or anti-cancer agent or anti-neoplasm treatment
  • an effective amount of a compound for treating a particular disease is an amount that is sufficient to ameliorate, or in some manner reduce the symptoms associated with the disease. Such amount may be administered as a single dosage or may be administered according to a regimen, whereby it is effective. The amount may cure the disease but, typically, is administered in order to ameliorate the symptoms of the disease. Repeated administration may be required to achieve the desired amelioration of symptoms.
  • treatment means any manner in which the symptoms of a conditions, disorder or disease are ameliorated or otherwise beneficially altered. Treatment also encompasses any pharmaceutical use of the compositions herein.
  • amelioration of the symptoms of a particular disorder by administration of a particular pharmaceutical composition refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the composition.
  • antibody is used in the broadest sense and specifically covers intact monoclonal antibodies, polyclonal antibodies, multi-specific antibodies, e.g., bi-specific antibodies, formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • the antibody may be an IgM, IgG, e.g., IgG 1 , IgG 2 , IgG 3 or IgG 4 , IgD, IgA or IgE, for example.
  • antibody fragments comprise a portion of an intact antibody, generally the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab′, F(ab′) 2 , and Br fragments; diabodies; single-chain antibody molecules; and multi-specific antibodies formed from antibody fragments.
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • polyclonal antibody refers to antibodies produced by several clones of B-lymphocytes as would be the case in a whole animal. Usually refers to antibodies raised in immunized animals, whereas a monoclonal antibody is the product of a single clone of B-lymphocytes, usually maintained in vitro.
  • hybridoma refers to a cell hybrid in which a tumour cell forms one of the original source cells.
  • exemplary hybridoma are hybrids between T- or B-lymphocytes and appropriate myeloma cell lines that produce a monoclonal antibody.
  • humanized antibodies refer to antibodies that are modified to include “human” sequences of amino acids so that administration to a human will not provoke an immune response. Methods for preparation of such antibodies are known. For example, the hybridoma that expresses the monoclonal antibody is altered by recombinant DNA techniques to express an antibody in which the amino acid composition of the non-variable regions is based on human antibodies. Computer programs have been designed to identify such regions.
  • production by recombinant means refers to production methods that use recombinant nucleic acid methods that rely on well known methods of molecular biology for expressing proteins encoded by cloned nucleic acids.
  • complementary when referring to two nucleic acid molecules, means that the two sequences of nucleotides are capable of hybridizing, preferably with less than 25%, more preferably with less than 15%, even more preferably with less than 5%, most preferably with no mismatches between opposed nucleotides. Preferably the two molecules will hybridize under conditions of high stringency.
  • medium stringency 0.2 ⁇ SSPE, 0.1% SDS, 50° C. (also referred to as moderate stringency);
  • vector refers to discrete elements that are used to introduce heterologous DNA into cells for either expression or replication thereof. Selection and use of such vehicles are well known within the skill of the artisan.
  • An expression vector includes vectors capable of expressing DNA's that are operatively linked with regulatory sequences, such as promoter regions, that are capable of effecting expression of such DNA fragments.
  • an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid, a phage, recombinant virus or other vector that, upon introduction into an appropriate host cell, results in expression of the cloned DNA.
  • Appropriate expression vectors are well known to those of skill in the art and include those that are replicable in eukaryotic cells and/or prokaryotic cells and those that remain episomal or those which integrate into the host cell genome.
  • a promoter region or promoter element refers to a segment of DNA or RNA that controls transcription of the DNA or RNA to which it is operatively linked.
  • the promoter region includes specific sequences that are sufficient for RNA polymerase recognition, binding and transcription initiation. This portion of the promoter region is referred to as the promoter.
  • the promoter region includes sequences that modulate this recognition, binding and transcription initiation activity of RNA polymerase. These sequences may be cis acting or may be responsive to trans acting factors. Promoters, depending upon the nature of the regulation, may be constitutive or regulated. Exemplary promoters contemplated for use in prokaryotes include the bacteriophage T7 and T3 promoters, and the like.
  • operatively linked or operationally associated refers to the functional relationship of DNA with regulatory and effector sequences of nucleotides, such as promoters, enhancers, transcriptional and translational stop sites, and other signal sequences.
  • operative linkage of DNA to a promoter refers to the physical and functional relationship between the DNA and the promoter such that the transcription of such DNA is initiated from the promoter by an RNA polymerase that specifically recognizes, binds to and transcribes the DNA.
  • protein binding sequence refers to a protein or peptide sequence that is capable of specific binding to other protein or peptide sequences generally, to a set of protein or peptide sequences or to a particular protein or peptide sequence.
  • epitope tag refers to a short stretch of amino acid residues corresponding to an epitope to facilitate subsequent biochemical and immunological analysis of the “epitope tagged” protein or peptide. “Epitope tagging” is achieved by appending the sequence of the “epitope tag” to the protein-encoding sequence in an appropriate expression vector. “Epitope tagged” proteins can be affinity purified using highly specific antibodies raised against the tags.
  • Protein A or Protein G refers to proteins that can bind to Fc region of most IgG isotypes. Protein A or Protein G are typically found in the cell wall of some strains of staphylococci . It is intended to encompass Protein A or Protein G with conservative amino acid substitutions that do not substantially alter its activity.
  • nucleotide binding sequence refers to a protein or peptide sequence that is capable of specific binding to nucleotide sequences generally, to a set of nucleotide sequences or to a particular nucleotide sequence.
  • lipid binding sequence refers to a protein or peptide sequence that is capable of specific binding to lipids generally, to a set of lipids or to a particular lipid.
  • polysaccharide binding sequence refers to a protein or peptide sequence that is capable of specific binding to polysaccharides generally, to a set of polysaccharides or to a particular polysaccharide.
  • metal binding sequence refers to a protein or peptide sequence that is capable of specific binding to metal ions generally, to a set of metal ions or to a particular metal ion.
  • the present invention is directed to a method for preventing, treating or delaying neoplasm in a mammal, which method comprises administering to a mammal, to which such prevention, treatment or delay is needed or desirable, an effective amount of an ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, whereby an immune response is generated against said neoplasm and said neoplasm is prevented, treated or delayed.
  • the present method can be used for preventing, treating or delaying neoplasm in any mammals, such as mice, rats, rabbits, cats, dogs, pigs, cows, ox, sheep, goats, horses, monkeys and other non-human primates.
  • the present method can be used for preventing, treating or delaying neoplasm in humans.
  • Any suitable ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, that can elicit an immune response to the neoplasm to be treated, prevented or delayed, can be used in the present method.
  • the ErbB-3 elicited immune response can be cellular, humoral or both.
  • ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, disclosed in U.S. Pat. No. 5,820,859 can be used in the present method.
  • ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, derived from human ErbB-3 is used in the present method.
  • Any ErbB-3 protein, or a functional fragment thereof, with conservative amino acid substitutions that do not substantially alter its activity can be used in the present method.
  • an effective amount of an extracellular domain of an ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an extracellular domain of an ErbB-3 protein, or a functional fragment thereof is administered.
  • an effective amount of the ErbB-3 protein comprising an amino acid sequence set forth in SEQ ID NO:1 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14, or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16 is administered.
  • an effective amount of the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 is administered.
  • the present method can further comprise administering an immune response potentiator to the mammal.
  • the immune response potentiator can be administered prior to, concurrently with, or subsequent to the administration of the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid an ErbB-3 protein, or a functional fragment thereof.
  • Exemplary immune response potentiators include Bacille Calmette-Guerin (BCG) (Ratliff, Eur.
  • the formulation, dosage and route of administration of ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, preferably in the form of pharmaceutical compositions can be determined according to the methods known in the art (see e.g., Remington: The Science and Practice of Pharmacy , Alfonso R. Gennaro (Editor) Mack Publishing Company, April 1997 ; Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems , Banga, 1999; and.
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof can be formulated for oral, rectal, topical, inhalational, buccal (e.g., sublingual), parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous), transdermal administration or any other suitable route of administration.
  • buccal e.g., sublingual
  • parenteral e.g., subcutaneous, intramuscular, intradermal, or intravenous
  • transdermal administration e.g., transdermal administration or any other suitable route of administration.
  • the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, which is being used.
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof can be administered to any suitable place in the mammal.
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof is administered to the neoplasm in situ, i.e., administered to the place where the neoplasm is located or the vicinity thereof.
  • the present method can further comprise administering an immune response potentiator to the neoplasm in situ.
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof can be administered alone.
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof is co-administered with a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutically acceptable carrier or excipient can be used in the present method (See e.g., Remington: The Science and Practice of Pharmacy , Alfonso R. Gennaro (Editor) Mack Publishing Company, April 1997).
  • the present method can be used alone. Alternatively, the present method can be used in combination with other anti-neoplasm treatment, e.g., radiationtherapy, chemotherapy or surgery. The present method can also be used in combination with other anti-neoplasm agent.
  • Such other anti-neoplasm treatment or agent can be used before, with or after the administration of ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof.
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof can be co-administered with an anti-neoplasm agent.
  • anti-neoplasm agents include an anti-angiogenic agent (See e.g., Auerbach and Auerbach, Pharmacol. Ther., 63(3):265-311 (1994)), an alkylating agent, an antimetabolite, a natural product, a platinum coordination complex, an anthracenedione, a substituted urea, a methylhydrazine derivative, an adrenocortical suppressant, a hormone, an antagonist, an oncogene inhibitor, a tumor suppressor gene or protein, an anti-oncogene antibody and an anti-oncogene antisense oligonucleotide.
  • the nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, or any tumor suppressor gene can be used in the form of naked DNA, complexed DNA, cDNA, plasmid DNA, RNA or other mixtures thereof as components of the gene delivery system.
  • the nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, or the tumor suppressor gene is included in a viral vector.
  • Any viral vectors that are suitable for gene therapy can used in the combination.
  • an adenovirus vector U.S. Pat. No. 5,869,305
  • a simian virus vector U.S. Pat. No. 5,962,274
  • a conditionally replicating human immunodeficiency viral vector U.S.
  • retrovirus retrovirus
  • SV40 Herpes simplex viral amplicon vectors
  • Vaccinia virus vectors can be used.
  • the genes can be delivered in a non-viral vector system such as a liposome wherein the lipid protects the DNA or other biomaterials from oxidation during the coagulation.
  • the present method can be used to treat, prevent or delay any suitable neoplasms or cancers.
  • the present method is used to treat, prevent or delay any suitable neoplasms or cancers wherein the interaction between ErbB-2 and ErbB-3 is critical for causing or sustaining the neoplasms or cancers.
  • the present method can be used to treat, prevent or delay adrenal gland, anus, auditory nerve, bile ducts, bladder, bone, brain, breast, bruccal, central nervous system, cervix, colon, ear, endometrium, esophagus, eye, eyelids, fallopian tube, gastrointestinal tract, head and neck, heart, kidney, larynx, liver, lung, mandible, mandibular condyle, maxilla, mouth, nasopharynx, nose, oral cavity, ovary, pancreas, parotid gland, penis, pinna, pituitary, prostate gland, rectum, retina, salivary glands, skin, small intestine, spinal cord, stomach, testes, thyroid, tonsil, urethra, uterus, vagina, vestibulocochlear nerve and vulva neoplasm.
  • the present method is used to treat, prevent or delay breast, ovary, stomach, prostate,
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents, carriers or excipients may be formulated for any suitable administration route, such as intracavernous injection, subcutaneous injection, intravenous injection, intramuscular injection, intradermal injection, oral or topical administration.
  • the method may employ formulations for injectable administration in unit dosage form, in ampoules or in multidose containers, with an added preservative.
  • the formulations may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, sterile pyrogen-free water or other solvents, before use.
  • Topical administration in the present invention may employ the use of a foam, gel, cream, ointment, transdermal patch, or paste.
  • compositions and methods for their administration that may be employed for use in this invention include, but are not limited to those described in U.S. Pat. Nos. 5,736,154; 6,197,801 B1; 5,741,511; 5,886,039; 5,941,868; 6,258,374 B1; and 5,686,102.
  • the magnitude of a therapeutic dose in the treatment or prevention will vary with the severity of the condition to be treated and the route of administration.
  • the dose, and perhaps dose frequency will also vary according to age, body weight, condition and response of the individual patient.
  • the attending physician would know how to and when to terminate, interrupt or adjust therapy to lower dosage due to toxicity, or adverse effects. Conversely, the physician would also know how to and when to adjust treatment to higher levels if the clinical response is not adequate (precluding toxic side effects).
  • Dosage forms include tablets, troches, cachet, dispersions, suspensions, solutions, capsules, patches, and the like. See, Remington's Pharmaceutical Sciences.
  • the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents may be combined as the active in intimate admixture with a pharmaceutical carrier or excipient, such as beta-cyclodextrin and 2-hydroxy-propyl-beta-cyclodextrin, according to conventional pharmaceutical compounding techniques.
  • a pharmaceutical carrier or excipient such as beta-cyclodextrin and 2-hydroxy-propyl-beta-cyclodextrin, according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide form of preparation desired for administration, topical or parenteral.
  • compositions for parenteral dosage form such as intravenous injection or infusion
  • similar pharmaceutical media may be employed, water, glycols, oils, buffers, sugar, preservatives, liposomes, and the like known to those of skill in the art.
  • parenteral compositions include, but are not limited to dextrose 5% w/v, normal saline or other solutions.
  • the total dose of the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents to be administered may be administered in a vial of intravenous fluid, ranging from about 1 ml to 2000 ml.
  • the volume of dilution fluid will vary according to the total dose administered.
  • kits for carrying out the therapeutic regimens of the invention comprise in one or more containers therapeutically effective amounts of the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents, in pharmaceutically acceptable form.
  • Preferred pharmaceutical forms would be in combination with sterile saline, dextrose solution, or buffered solution, or other pharmaceutically acceptable sterile fluid.
  • the composition may be lyophilized or dessicated; in this instance, the kit optionally further comprises in a container a pharmaceutically acceptable solution, preferably sterile, to reconstitute the complex to form a solution for injection purposes.
  • Exemplary pharmaceutically acceptable solutions are saline and dextrose solution.
  • kits of the invention further comprises a needle or syringe, preferably packaged in sterile form, for injecting the composition, and/or a packaged alcohol pad. Instructions are optionally included for administration of composition by a physician or by the patient.
  • the present invention is directed to an isolated nucleic acid fragment, which isolated nucleic acid fragment hybridizes, under low, middle or high stringency, with a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the isolated nucleic acid fragment hybridizes, under high stringency, with a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the isolated nucleic acid fragment comprises a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the isolated nucleic acid fragment comprises a sequence of nucleotides, or a complementary strand thereof, set forth in SEQ ID NO:4 ( FIG. 1 ) or SEQ ID NO:5 ( FIG. 6 ) or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the isolated nucleic acid fragments can be in any suitable form.
  • the isolated nucleic acid fragment can comprise DNA, RNA, PNA or a derivative thereof.
  • the isolated nucleic acid fragment can comprise both DNA and RNA or derivatives thereof.
  • the isolated nucleic acid fragment can be single-stranded and be ready to be used in a hybridization analysis.
  • the isolated nucleic acid fragment can be double-stranded and be denatured into single-stranded prior to the hybridization analysis.
  • the isolated nucleic acid fragment can comprise any kind of oligonucleotide or nucleic acid strand(s) containing genetically-coded and/or naturally occurring structures.
  • the isolated nucleic acid fragments can comprise non-natural elements such as non-natural bases, e.g., inosine and xanthine, non-natural sugars, e.g., 2′-methoxy ribose, or non-natural phosphodiester linkages, e.g., methylphosphonates, phosphorothioates and peptides.
  • the isolated nucleic acid fragments can be produced by any suitable methods.
  • the isolated nucleic acid fragments can be chemically synthesized (See generally, Ausubel (Ed.) Current Protocols in Molecular Biology, 2.11 . Synthesis and purification of oligonucleotides , John Wiley & Sons, Inc. (2000)), isolated from a natural source, produced by recombinant methods or a combination thereof.
  • the isolated nucleic acid fragments are produced by recombinant methods.
  • the isolated nucleic acid fragment can be labeled for various purposes, e.g., facilitating detection, purification and/or attachment to a surface.
  • the label can be a chemical, an enzymatic, an immunogenic, a radioactive, a fluorescent, a luminescent or a FRET label.
  • a plasmid, which plasmid comprises the above nucleic acid fragment is also provided.
  • a cell, which cell comprises the above plasmid is further provided. Any suitable cells can be used, e.g., bacterial cells, yeast cells, fungal cells, plant cells, insect cells, animal cells and human cells.
  • the present invention is directed to a method for producing an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, which method comprises growing the above cells under conditions whereby the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, is expressed by the cells, and recovering the expressed extracellular domain of the ErbB-3 protein, or a functional fragment thereof.
  • the present invention is directed to a substantially purified protein or peptide, which comprises an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the extracellular domain of the ErbB-3 protein, or a functional fragment thereof can be produced by any suitable methods.
  • the extracellular domain of the ErbB-3 protein, or a functional fragment thereof can be chemically synthesized, isolated from a natural source, produced by recombinant methods or a combination thereof.
  • the extracellular domains of the ErbB-3 protein, or functional fragments thereof are produced by recombinant methods.
  • the present invention is directed to a conjugate, which conjugate comprises: a) a protein or peptide comprising an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3; an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16; and b) a facilitating agent linked to the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, directly or via a linker, wherein the agent facilitates: i) affinity isolation or purification of a conjugate; ii) attachment of a conjugate to a surface; or iii) detection of a conjugate.
  • the conjugate can be a fusion protein.
  • the ErbB-3 protein, or a functional fragment thereof, and the facilitating agent can be linked by other means.
  • the conjugate is a fusion protein, a nucleic acid encoding the conjugate is also provided.
  • the conjugates can be produced by chemical conjugation, such as via thiol linkages, but are preferably produced by recombinant means as fusion proteins.
  • the peptide or fragment thereof is linked to either the N-terminus or C-terminus of the ErbB-3 protein, or a functional fragment thereof.
  • chemical conjugates the peptide or fragment thereof may be linked anywhere that conjugation can be effected, and there may be a plurality of such peptides or fragments linked to a single the ErbB-3 protein, or a functional fragment thereof, or to a plurality thereof.
  • Conjugation can be effected by any method known to those of skill in the art. As described below, conjugation can be effected by chemical means, through covalent, ionic or any other suitable linkage. For example, the reagents and methods for conjugation as disclosed in WO 01/02600 can be used.
  • the conjugate is a fusion protein, which can be isolated or purified through affinity binding between the protein or peptide fragment of the fusion protein and an affinity binding moiety. Any kind of affinity interaction can be used for isolating or purifying the fusion protein.
  • the affinity interactions such as those described herein, but not limited to, are protein/protein, protein/nucleotide, protein/lipid, protein/polysaccharide, or protein/metal interactions.
  • the conjugate can be attached to a surface. More preferably, the conjugate can be attached to the surface through affinity binding between the facilitating agent of conjugate and an affinity binding moiety on the surface. Any kind of affinity interaction can be used for attaching the conjugate, including the protein/protein, protein/nucleotide, protein/lipid, protein/polysaccharide, or protein/metal interactions.
  • the present invention is directed to a pharmaceutical composition, which pharmaceutical composition comprises an isolated nucleic acid fragment which isolated nucleic acid fragment hybridizes, under low, middle or high stringency, with a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16 and a pharmaceutically acceptable carrier or excipient.
  • the isolated nucleic acid comprises a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the pharmaceutical composition can further comprise an immune response potentiator and/or an anti-neoplasm agent.
  • Vaccines comprising the above isolated nucleic acid fragments, alone or in combination with an immune response potentiator, are also provided.
  • the present invention is directed to a pharmaceutical composition, which pharmaceutical composition comprises a substantially purified protein or peptide, which comprises an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16 and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition can further comprise an immune response potentiator and/or an anti-neoplasm agent.
  • Vaccines comprising the above substantially purified proteins or peptides, alone or in combination with an immune response potentiator, are also provided.
  • the present invention is directed to an antibody, which antibody binds to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the antibody binds specifically to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • the antibody can be in any suitable form.
  • the antibody can a polyclonal, monoclonal, chimeric, single chain, human or humanized antibody (See e.g., U.S. Pat. No. 5,968,511).
  • the antibody in various forms, can be made according to any methods known in the art (See, e.g., Coligan et al. (Ed.), Current Protocols in Immunology, John Wiley & Sons, Inc. (2000)).
  • Pharmaceutical compositions, comprising the above antibodies, alone or in combination with anti-neoplasm agent, and a pharmaceutically acceptable carrier or excipient are also provided.
  • the inventor provided a method of B3, DE3-1, rhErbB3-f12 and rhErbB3-f78 as an anti-tumor vaccine significantly lowering the incidence of the development of human cancer such as breast cancer in high-risk population.
  • the inventor discovered that there was significant effect of B3, DE3-1, DE3-1, rhErbB3-f12 and rhErbB3-f78 as an anti-tumor vaccine on postponing the tumor development such as human breast cancer.
  • the inventor discovered that there was significant inhibitory effect of B3, DE3-1, rhErbB3-f12 and rhErbB3-f78 as an anti-tumor vaccine on the tumor development such as human breast cancer.
  • the inventor discovered a method of inhibiting cancerous growth such as breast cancer and that was achieved through inducing immune responses.
  • the aforementioned cells may be a tumor cells, much probably they are human breast cancer cells and other cancerous cells with Erb2/ErbB3 over-expression.
  • ErbB3 protein antigen expressed through genetic engineering It is ErbB3 protein antigen expressed through genetic engineering that made the aforementioned method come true; De3-1, rhErbB3-f12 and rhErbB3-178 is a protein expressed by E. Coli ; B3 was an antigen of protein expressed by Eucaryotic cells or ErbB3 antigen produced by other methods, ErbB3 antigen may be ErbB3 molecule or part of a segment of the molecule.
  • ErbB3 vaccine which is produced by different methods can inhibit tumor growth under certain dosage level.
  • the aforementioned cancers included breast cancer, ovary carcinoma, gastric carcinoma, and prostate carcinoma and lung cancer.
  • the vaccine involved in the present experiment included protein molecule in the extra-cell membranes region of ErbB3 and part of the protein segment of the extra-cell membranes, they are named as B2 and SD32.
  • the protein molecule in the extra-cell membranes region of ErbB3 and part of the protein of the extra-cell membranes serves as an experiment sample, they are named as B3, De3-1, rhErbB3-f12 and rhErbB3-f78 here; the aforementioned vaccines are manufactured by Zensun (Shanghai) Science and Technology Development Co Ltd.
  • the preparation of B3 and DE3-1, rhErbB3-f12 and rhErbB3-f78 is as follow:
  • B3 gene is the encoded cDNA sequence of protein of ErbB3 extra-cell membrane region ( FIG. 1 ); amplified with PCR, sequence of the primer was:
  • Primer1 (SEQ ID NO: 6) 5′TCTGCGGAGTCATGAGGGC Primer2, (SEQ ID NO: 7) 3′TCACTTGTCGTCATCGTCCTTGTAGTCTTTGCCGATCAGCACCAGTGT
  • the italics are flag sequence.
  • the targeting gene was cloned into pMD-18T vector; the transformer will be cut by BamHI/Sa1I after enzyme digestion and identified of the sequence to be correct, then connected to pCDNA3BamHI/xhol.
  • FIG. 4 illustrated the B3 protein purification, affinity chromatography purification.
  • FIG. 5 showed the targeting protein and amino acid sequence of B3 purified protein after amino acid sequencing.
  • FIG. 6 showed cDNA sequence of encoded extra-cell membrane ErbB3 protein segment of PCR amplified targeting gene.
  • Structure of the expressed plasmid targeting gene segment was cut out with BamHI/XhoI from pGEX4T-1 vector (Phamacia company), connecting into pET32a vector (Novagen company) BabHI/XhoI, the protein was expressed by T7 promoter, N end fused with Trx Tag, His Tag and S-Tag,
  • FIG. 7 illustrates the diagram.
  • FIG. 8 illustrates the identification of the plasmid composition.
  • rhErbB3-f12(SEQ ID NO:14) gene is the encoded cDNA sequence of protein of ErbB3 extra-cell membrane region; amplified with PCR, sequence of the primer was:
  • FVB/N transgenic mice 8-10 weeks old FVB/N transgenic mice (bought from Jackson Lab USA) was selected as experiment animals, the mice were divided into 5 groups with 40 mice each, they were control group, B2, B3 and DE3-1 group; BSA, B2, SD32, B3, DE3-1 was mixed with Freud's adjuvant (CFA, complete Freud's adjuvant, bought from Sigma company) and injected abdominally every 20 days for 7 injections respectively to various groups.
  • the dosage of BSA, B2, SD32, B3 and DE3-1 vaccine was 10, 5, 10, 1 and 10 ⁇ g/mouse/injection. Weekly monitor tumour development. The tumour development was verified and analysed statistically.
  • Transplanted tumour model after immunohistological screening test, about 1000 mm 3 tumour mass was cut down from spontaneous tumour of neu protein over-expressed FVB/N transgenic mice. The tumour mass was abraded into single cells with nylon net, the amount injected under the breast of each FVB/N transgenic mice was 5 ⁇ 10 6 cells. About 10-14 days after inoculation, tumor was palpable (>5 mm) in the control group, demonstrating that the animal model was established successfully.
  • inhibitory rate [(tumor weight of control group ⁇ tumor weight of experiment groups)/tumor weight of control group] ⁇ 100.
  • Preparing animal and transplanted animal tumor model The same as (Experiment on therapeutic effect of B3 and DE3-1 rhErbB3-f12 and rhErbB3-f78 vaccine on immune therapy against tumor). No treatment was given to the control group, histag protein was injected to the negative control group, and Adriamycin (Santou MingZhi Pharmaceuticals) was administered for the positive control group, 5 ⁇ g, 20 ⁇ g and 80 ⁇ g was given to DE3-1 group respectively.
  • Adriamycin 2.2 mg/kg was injected abdominally for consecutive 7 days in mice of the positive control group; histag protein+Al(OH) 3 was injected abdominally for mice of the negative control group;
  • the vaccine was absorbed on 0.1 mg of Al(OH) 3 and multi-points subcutaneous injection every 2 weeks for a total of 3 injections were carried out in mice.
  • the experiment completed in 14 days after the third injection. Tumor development was monitored weekly, tumor size was measured with vernier and the size was expressed as (length diameter ⁇ short diameter 2/2), curve of tumor growth was protracted and analyzed statistically.
  • tumor inhibitory rate [(tumor weight of control group ⁇ tumor weight of experiment groups)/tumor weight of control group] ⁇ 100.
  • FVB transgenic mice were immuned with B2 protein and B3 protein respectively, 10 days thereafter, blood was withdrawn and antibody titer was tested with ELISA.
  • 0.3 ⁇ g/hole of B2 and B3 was wrapped, 1:1000 B2 and B3 on each plate were titrated with standard serum respectively, cultured at 37° C. for 30 minutes, sealed with 1% BSA, added double antibody, color development for 15 minutes with DAD, tested with Bio-Rad 450 nm enzyme labeled device.
  • Table 1 and FIG. 12 illustrate the experimental results of tumor inhibitory effect of B3 and DE3-1.
  • the inventor carried out experiment on immune therapy with B3 in transplanted tumor model.
  • Table 2 and FIG. 13-14 illustrate the effect of various vaccines on tumor growth in mice, demonstrating that the tumor-inhibitory rate of SD32 and B3 were 46% and 33% respectively, and that both of them had significant tumor-inhibitory effect (P ⁇ 0.01; t testing).
  • Tumor-inhibitory rate and measured tumor size was consistent among groups with various dosage of DE3-1, the best tumor-inhibitory effect was seen in 20 ⁇ g level of DE3-1, reaching about 28.9%.
  • the mice were killed, took out the tumor and measured their weight; there were significant difference (P ⁇ 0.001, t test) between the positive control group, groups with various dosage level, negative control group and placebo control group.
  • the tumor-inhibitory rate of 5 ⁇ g, 20 ⁇ g and 80 ⁇ g dosage level group were 26.3%, 22.4% and 28.9% respectively.
  • FIG. 17-18 showed the experimental results demonstrating that there isn't any cross immunity between B2 and B3 antigen.
  • ErbB2 receptor existed in part of adenocarcinoma discovered in the previous studies was considered to be associated with cancer development after formation of homogenous dimer.
  • Over-expression of ErbB2 was considered to be the major cause of adenocarcinoma development, it is due to: 1) the amplification of ErbB2 gene existed in tumor cells such as breast cancer and ovary carcinoma was the cause of over-expression of ErbB2; 2) Over-expression of ErbB2 leads to phosphorylation in its cellular functional area and affects the interaction between intracellular signal molecule Shc and ErbB2; 3) the transfection of wild type ErbB2 into fibroblast can lead to cell transformation; 4) the enhancement of the formation of ErbB2 variants from ErbB2 homogenous dimer can also enhance its activity of cell transformation.
  • ErbB3 As another new anti-tumor target in addition to ErbB2.
  • the inventors clarify that over-expression of ErbB2 receptor leads to the formation of heterogenous dimer from ErbB2 and ErbB3, and that was the cause of cancer development.
  • Discovery of this new target provides us with new concept of anti-cancer therapeutical method: use extra-cell membrane protein of ErbB3 cells for cancer prevention and treatment, to lower the incidence of breast cancer and produce effect against tumor growth.

Abstract

The present invention relates to compositions and methods for treating neoplasms in mammals, particularly humans. More particularly, the present invention provides for methods for preventing, treating or delaying neoplasm in a mammal using an ErbB-3 protein, a nucleic acid encoding an ErbB-3 protein or a functional fragment thereof. The present invention also provides for an isolated nucleic acids encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, substantially purified extracellular domain of the ErbB-3 protein, or a functional fragment thereof and antibodies that bind to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof. The present invention further provides for pharmaceutical compositions and/or vaccines comprising the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, or nucleic acids encoding and antibodies binding to such extracellular domain or functional fragments thereof.

Description

  • This application is a divisional application of U.S. application Ser. No. 10/516,759, filed on Mar. 3, 2006, which is a national phase of PCT/CN03/00217, having an international filing date of Mar. 26, 2003, which claims priority from the Chinese Patent Application No. 02116259.x, filed Mar. 26, 2002, now Chinese Patent No. 03806762.5. The disclosures of the above applications are incorporated herein by reference in their entirety.
  • TECHNICAL FIELD
  • The present invention relates to compositions and methods for treating neoplasms in mammals, particularly humans. More particularly, the present invention provides for methods for preventing, treating or delaying neoplasm in a mammal using an ErbB-3 protein, a nucleic acid encoding an ErbB-3 protein or a functional fragment thereof. The present invention also provides for isolated nucleic acids encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, substantially purified extracellular domain of the ErbB-3 protein, or a functional fragment thereof and antibodies that bind to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof. The present invention further provides for pharmaceutical compositions and/or vaccines comprising the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, or nucleic acids encoding and antibodies binding to such extracellular domain or functional fragments thereof.
  • BACKGROUND ART
  • Cancer is a major lethal disease for humans and is caused by physiologically-uncontrolled cell proliferation which affects normal physiological conditions of human body resulting in serious pathological reactions often leading to death. Although tremendous efforts on cancer studies and treatments have been made, presently, cancer is still the major cause of death to humans. There are multiple approaches to treat cancer patients including surgery, radiation therapy and chemotherapy. As the first two methods are not able to completely eliminate cancer cells in patients, the latter approach is commonly used to control cancer cell growth with or without other treatments. Anti-cancer compounds used in patients are often targeting prevention of cancer cell proliferation or killing dividing cells.
  • When the compounds are toxic to cancer cells, they may also severely affect normal dividing cells which are necessary for human life. Therefore, one of main directions in cancer studies is to find methods to specifically block or kill cancer cells without affecting normal cell proliferation. There is a demand now for such treatment on cancer patients.
  • ErbBs are class one receptor protein tyrosine kinases. ErbB-mediated cell signaling plays a critical role in embryo development and adult organ function. On a cellular level, ErbB receptors have been shown to mediate signals for cell proliferation, differentiation, migration, and cell structure reorganization. There are four structurally similar ErbB members, ErbB-1, ErbB-2, ErbB-3 and ErbB-4. The epidermal growth factor (EGF) is one of several ligands that bind ErbB-1, ErbB-3 or ErbB-4 also bind several ligands, including neuregulin-1 (NRG-1). To date, no ligand for ErbB-2 has been identified. However, ErbB-2 serves as a heterodimer partner for ErbB-3, ErbB-4 or ErbB-1 and is critically involved in NRG-1-activated cell signaling. In vivo studies using gene targeting experiments indicate that developmental defects resulting from inactivation of ErbB-2 are similar to those observed in NRG-1-inactivated animals. Both animals show defects in the neural crania ganglia and heart trabeculae development. Furthermore, ErbB-3 or ErbB-4 gene-inactivated mice have similar or overlapping phenotypes to NRG-1 or ErbB-2 knockout mice.
  • In addition to its role in development, the human ErbB-2 gene is frequently amplified and its encoded protein is over-expressed in a variety of human carcinomas. Early research on ErbB-2 discovered that an oncogenic point mutation resulted in the formation of ErbB-2 homodimers that in turn caused significant phosphorylation of the tyrosine residues on the intracellular domain. While no corresponding point mutation has been found in ErbB-2 over expressing human carcinomas, the upregulation of ErbB-2 results in the formation of homodimers that in turn increases the tyrosine phosphorylation of its intracellular domain. This process is hypothesized to be the start of a signal cascade that triggers cell transformation and/or growth, and thus initiate tumorigenesis. There is evidence, however, to contradict the hypothesis that ErbB-2 homodimers are responsible for the initiation of tumorigenesis: i) some ErbB-2 mutants that are engineered to enhanced dimerization and self-phosphorylation have no effect on cell transformation; ii) antibodies that bind to the extracellular domain of ErbB-2 and presumably promote homodimerization result in ErbB2-expressing cancer cell growth promotion, whereas others inhibit cancer cell growth. These data indicate that homodimerization of ErbB-2 is insufficient for cell growth promotion or cell transformation, and other conditions, possibly involving specific dimer orientation or conformation, are required.
  • ErbB-2 acts as a heterodimer partner for the ligand-binding ErbB-3 or ErbB-4 receptors. The ligand, NRG-1, has been identified to have two independent receptor binding sites: one that has a high affinity for ErbB-3 or ErbB-4, and the other that has a low but non-specific affinity for all ErbB members. Thus, the exposure of NRG-1 to cells expressing ErbB-3/4 and ErbB-2 would result in heterodimers of ErbB-2 and ErbB-3/4. In the absence of the ligand, however, it is unclear whether ErbB-2 has an affinity with other ErbB receptors, and it is possible that such an interaction could be involved in the initiation of cancer. Amongst all the ErbB receptors, ErbB-3 is unique because: i) ErbB-2 preferentially forms heterodimers with ErbB-3; ii) cotransfection of NIH3T3 cells with ErbB-2 and ErbB-3 results in much higher levels of cell transformation than that of transfection with ErbB-2 alone; iii) in ErbB-2 over-expression-associated breast cancer cells, ErbB-3 is also highly expressed; and iv) ErbB-3 is also over expressed in ErbB-2-over expressing tumour cells from ErbB-2 transgenic mice.
  • A number of patents and patent applications disclose ErbB-2 and/or ErbB-3 related neoplasm or cancer treatment. For example, WO 00/78347 discloses methods for arresting or inhibiting cell growth, particularly cancer cell growth, comprising preventing or reducing ErbB-2/ErbB-3 heterodimer formation, or interfering with ErbB-2/ErbB-3 heterodimer conformation in a cell and agents which prevent or reduce ErbB-2/ErbB-3 heterodimer formation or interfere with ErbB-2/ErbB-3 heterodimer conformation in a cell thereby arresting or inhibiting the growth of the cell. U.S. Pat. No. 5,578,482 relates to erbB-2 ligands and functional derivatives thereof which are capable of binding to the erbB-2 oncogene product. U.S. Pat. No. 5,820,859 relates to a method of targeting a therapeutic agent to cells expressing the erb B-3 receptor. U.S. Pat. No. 5,968,511 relates to ErbB3 antibodies.
  • There exists a need in the art for more efficient and/or cost effective ErbB-3 related neoplasm treatments. The present invention addresses this and other related needs in the art.
  • DISCLOSURE OF THE INVENTION
  • In one aspect, the present invention is directed to a method for preventing, treating or delaying neoplasm in a mammal, which method comprises administering to a mammal, to which such prevention, treatment or delay is needed or desirable, an effective amount of an ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, whereby an immune response is generated against said neoplasm and said neoplasm is prevented, treated or delayed.
  • In another aspect, the present invention is directed to an isolated nucleic acid fragment, which isolated nucleic acid fragment comprises a sequence of nucleotides encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 (FIG. 5) or SEQ ID NO:3 (FIG. 11) or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • In still another aspect, the present invention is directed to a substantially purified protein or peptide, which comprises an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • In yet another aspect, the present invention is directed to a conjugate, which conjugate comprises: a) a protein or peptide comprising an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.; and b) a facilitating agent linked to the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, directly or via a linker, wherein the agent facilitates: i) affinity isolation or purification of a conjugate; ii) attachment of a conjugate to a surface; or iii) detection of a conjugate.
  • In yet another aspect, the present invention is directed to an antibody, which antibody binds to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • Pharmaceutical compositions and/or vaccines comprising the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, or nucleic acids encoding and antibodies binding to such extracellular domain or functional fragments thereof are also provided.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 depicts B3 cDNA sequence (SEQ ID NO:4).
  • FIG. 2 illustrates restriction enzyme digestion of B3 plasmid. Lane 1:1 KB ladder (NEB). Lane 2-9: DNA for diagnostic digestion with BamHI/XbaI. All are correct clones except the colony on Lane 5. Lane 10: pCDNA3 vector alone digested with BamHI/XbaI.
  • FIG. 3 illustrates B3 plasmid construction.
  • FIG. 4 illustrates isolation and/or purification and SDS-PAGE analysis of B3 protein. Lane 1-4: BSA control, 10 μg, 5 μg, 3 μg, 1 μg/lane respectively. Lane 5: Protein marker, 7708S NEB. Lane 6-7: B3 protein expressed for COST.
  • FIG. 5 depicts B3 amino acid sequence (SEQ ID NO:2).
  • FIG. 6 depicts DE3-1 cDNA sequence (SEQ ID NO:5).
  • FIG. 7 illustrates DE3-1 plasmid construction.
  • FIG. 8 illustrates restriction enzyme digestion of DE3-1 plasmid. Lane 1: DE3-1 in pGEX4T-1, cut with BamHI/XhoI. Lane 2: DE3-1 in pET32a, cut with BamHI/XhoI. Lane 3: 1 Kd ladder (NEB).
  • FIG. 9 illustrates SDS-PAGE analysis of DE3-1 expression. Lane 1: before induction. Lane 2: after induction. Lane 3: inclusion body. Lane 4: supernatant after sonication.
  • FIG. 10 illustrates isolation and/or purification and SDS-PAGE analysis of DE3-1 protein. Lane 1: Flow through. Lane 2-8: Eluates from NTA His tag affinity column.
  • FIG. 11 depicts DE3-1 amino acid sequence (SEQ ID NO:3).
  • FIG. 12 illustrates the effect of various vaccines on incidence of FVB/N transgenic mice.
  • FIG. 13 illustrates the effect of various drugs on tumor growth in mice (5 weeks).
  • FIG. 14 illustrates the tumor-inhibitory effect of various drugs against tumor growth (5 weeks).
  • FIG. 15 illustrates the effect of DE3-1 on the growth of breast cancer in mice (5 weeks).
  • FIG. 16 illustrates the tumor-inhibitory rate of DE3-1 against tumor growth (5 weeks).
  • FIG. 17 illustrates experiment on cross immunity between B2 and B3 antigen (B3 protein wrapped).
  • FIG. 18 illustrates experiment on cross immunity between B2 and B3 antigen (B2 protein wrapped).
  • FIG. 19 illustrates Result of PCR amplification
  • Lane 2,3: 192 bp ErbB3-f12 gene obtained by RT-PCR; Lane 1: DNA marker
  • FIG. 20 illustrates Screening for expression engineering strain.
  • FIG. 21 illustrates Experimental results of anti-tumor effect of rhErbB3-f12.
  • FIG. 22 illustrates Experimental results of anti-tumor effect of rhErbB3-f78.
  • FIG. 23 depicts ErbB3-f12 amino acid sequence (SEQ ID NO:14) and cDNA sequence (SEQ ID NO:17).
  • FIG. 24 depicts ErbB3-f78 cDNA sequence (SEQ ID NO:15).
  • FIG. 25 depicts ErbB3-f78 amino acid sequence (SEQ ID NO:16).
  • MODES OF CARRYING OUT THE INVENTION
  • For clarity of disclosure, and not by way of limitation, the detailed description of the invention is divided into the subsections that follow.
  • A. Definitions
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belongs. All patents, applications, published applications and other publications referred to herein are incorporated by reference in their entirety. If a definition set forth in this section is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, published applications and other publications that are herein incorporated by reference, the definition set forth in this section prevails over the definition that is incorporated herein by reference.
  • As used herein, “a” or “an” means “at least one” or “one or more.”
  • As used herein, “neoplasm (neoplasia)” refers to abnormal new growth, and thus means the same as tumor, which may be benign or malignant. Unlike hyperplasia, neoplastic proliferation persists even in the absence of the original stimulus.
  • As used herein, “cancer” refers to a general term for diseases caused by any type of malignant tumor.
  • As used herein, “malignant,” as applies to tumors, refers to primary tumors that have the capacity of metastasis with loss of both growth control and positional control.
  • As used herein, “erb” refers to two oncogenes, erb A and erb B, associated with erythroblastosis virus (an acute transforming retrovirus).
  • As used herein, “immune response” refers to alteration in the reactivity of an organism's immune system in response to an antigen; in vertebrates, this may involve antibody production, induction of cell-mediated immunity, complement activation or development of immunological tolerance.
  • As used herein, “immune response potentiator” refers to a substance that enhances an antigen's effect in eliciting an immune response.
  • As used herein, “vaccine” refers to any compositions intended for active immunological prophylaxis. A vaccine may be used therapeutically to treat a disease, or to prevent development of a disease or to decrease the severity of a disease either proactively or after infection. Exemplary vaccines include, but are not limited to, preparations of killed microbes of virulent strains or living microbes of attenuated (variant or mutant) strains, or microbial, fungal, plant, protozoa, or metazoa derivatives or products. “Vaccine” also encompasses protein/peptide and nucleic acid/oligonucleotides based vaccines.
  • As used herein, “anti-neoplasm agent (used interchangeably with anti-neoplastic agent, anti-tumor or anti-cancer agent)” refers to any agents used in the anti-neoplasm treatment. These include any agents, that when used alone or in combination with other compounds, can alleviate, reduce, ameliorate, prevent, or place or maintain in a state of remission of clinical symptoms or diagnostic markers associated with neoplasm, tumor or cancer, and can be used in methods, combinations and compositions provided herein. Anti-neoplastic agents include, but are not limited to, anti-angiogenic agents, alkylating agents, antimetabolite, certain natural products, platinum coordination complexes, anthracenediones, substituted ureas, methylhydrazine derivatives, adrenocortical suppressants, certain hormones and antagonists, anti-cancer polysaccharides and certain herb extracts such as Chinese herb extracts.
  • As used herein, an “anti-neoplastic treatment” refers to any treatment designed to treat the neoplasm, tumor or cancer by lessening or ameliorating its symptoms. Treatments that prevent the occurrence of neoplasm, tumor or cancer or lessen its severity are also contemplated.
  • As used herein, “anti-neoplasm agent (or anti-tumor or anti-cancer agent) or anti-neoplasm treatment” does not encompass an ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, or use thereof for treatment, but encompasses all agents and treatment modalities known to those of skill in the art to ameliorate the symptoms in some manner of a neoplasm, tumor or cancer.
  • As used herein, “an effective amount of a compound for treating a particular disease” is an amount that is sufficient to ameliorate, or in some manner reduce the symptoms associated with the disease. Such amount may be administered as a single dosage or may be administered according to a regimen, whereby it is effective. The amount may cure the disease but, typically, is administered in order to ameliorate the symptoms of the disease. Repeated administration may be required to achieve the desired amelioration of symptoms.
  • As used herein, “treatment” means any manner in which the symptoms of a conditions, disorder or disease are ameliorated or otherwise beneficially altered. Treatment also encompasses any pharmaceutical use of the compositions herein.
  • As used herein, “amelioration” of the symptoms of a particular disorder by administration of a particular pharmaceutical composition refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the composition.
  • As used herein, “antibody” is used in the broadest sense and specifically covers intact monoclonal antibodies, polyclonal antibodies, multi-specific antibodies, e.g., bi-specific antibodies, formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity. The antibody may be an IgM, IgG, e.g., IgG1, IgG2, IgG3 or IgG4, IgD, IgA or IgE, for example.
  • As used herein, “antibody fragments” comprise a portion of an intact antibody, generally the antigen binding or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab′, F(ab′)2, and Br fragments; diabodies; single-chain antibody molecules; and multi-specific antibodies formed from antibody fragments.
  • As used herein, “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • As used herein, “polyclonal antibody” refers to antibodies produced by several clones of B-lymphocytes as would be the case in a whole animal. Usually refers to antibodies raised in immunized animals, whereas a monoclonal antibody is the product of a single clone of B-lymphocytes, usually maintained in vitro.
  • As used herein, “hybridoma” refers to a cell hybrid in which a tumour cell forms one of the original source cells. Exemplary hybridoma are hybrids between T- or B-lymphocytes and appropriate myeloma cell lines that produce a monoclonal antibody.
  • As used herein, “humanized antibodies” refer to antibodies that are modified to include “human” sequences of amino acids so that administration to a human will not provoke an immune response. Methods for preparation of such antibodies are known. For example, the hybridoma that expresses the monoclonal antibody is altered by recombinant DNA techniques to express an antibody in which the amino acid composition of the non-variable regions is based on human antibodies. Computer programs have been designed to identify such regions.
  • As used herein, “production by recombinant means” refers to production methods that use recombinant nucleic acid methods that rely on well known methods of molecular biology for expressing proteins encoded by cloned nucleic acids.
  • As used herein, “complementary” when referring to two nucleic acid molecules, means that the two sequences of nucleotides are capable of hybridizing, preferably with less than 25%, more preferably with less than 15%, even more preferably with less than 5%, most preferably with no mismatches between opposed nucleotides. Preferably the two molecules will hybridize under conditions of high stringency.
  • As used herein: “stringency of hybridization” in determining percentage mismatch is as follows:
  • 1) high stringency: 0.1×SSPE, 0.1% SDS, 65° C.;
  • 2) medium stringency: 0.2×SSPE, 0.1% SDS, 50° C. (also referred to as moderate stringency); and
  • 3) low stringency: 1.0×SSPE, 0.1% SDS, 50° C.
  • It is understood that equivalent stringencies may be achieved using alternative buffers, salts and temperatures.
  • As used herein, “vector (or plasmid)” refers to discrete elements that are used to introduce heterologous DNA into cells for either expression or replication thereof. Selection and use of such vehicles are well known within the skill of the artisan. An expression vector includes vectors capable of expressing DNA's that are operatively linked with regulatory sequences, such as promoter regions, that are capable of effecting expression of such DNA fragments. Thus, an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid, a phage, recombinant virus or other vector that, upon introduction into an appropriate host cell, results in expression of the cloned DNA. Appropriate expression vectors are well known to those of skill in the art and include those that are replicable in eukaryotic cells and/or prokaryotic cells and those that remain episomal or those which integrate into the host cell genome.
  • As used herein, “a promoter region or promoter element” refers to a segment of DNA or RNA that controls transcription of the DNA or RNA to which it is operatively linked. The promoter region includes specific sequences that are sufficient for RNA polymerase recognition, binding and transcription initiation. This portion of the promoter region is referred to as the promoter. In addition, the promoter region includes sequences that modulate this recognition, binding and transcription initiation activity of RNA polymerase. These sequences may be cis acting or may be responsive to trans acting factors. Promoters, depending upon the nature of the regulation, may be constitutive or regulated. Exemplary promoters contemplated for use in prokaryotes include the bacteriophage T7 and T3 promoters, and the like.
  • As used herein, “operatively linked or operationally associated” refers to the functional relationship of DNA with regulatory and effector sequences of nucleotides, such as promoters, enhancers, transcriptional and translational stop sites, and other signal sequences. For example, operative linkage of DNA to a promoter refers to the physical and functional relationship between the DNA and the promoter such that the transcription of such DNA is initiated from the promoter by an RNA polymerase that specifically recognizes, binds to and transcribes the DNA. In order to optimize expression and/or in vitro transcription, it may be necessary to remove, add or alter 5′ untranslated portions of the clones to eliminate extra, potential inappropriate alternative translation initiation (i.e., start) codons or other sequences that may interfere with or reduce expression, either at the level of transcription or translation. Alternatively, consensus ribosome binding sites (see, e.g., Kozak, J. Biol. Chem., 266:19867-19870 (1991)) can be inserted immediately 5′ of the start codon and may enhance expression. The desirability of (or need for) such modification may be empirically determined.
  • As used herein, “protein binding sequence” refers to a protein or peptide sequence that is capable of specific binding to other protein or peptide sequences generally, to a set of protein or peptide sequences or to a particular protein or peptide sequence.
  • As used herein, “epitope tag” refers to a short stretch of amino acid residues corresponding to an epitope to facilitate subsequent biochemical and immunological analysis of the “epitope tagged” protein or peptide. “Epitope tagging” is achieved by appending the sequence of the “epitope tag” to the protein-encoding sequence in an appropriate expression vector. “Epitope tagged” proteins can be affinity purified using highly specific antibodies raised against the tags.
  • As used herein, “Protein A or Protein G” refers to proteins that can bind to Fc region of most IgG isotypes. Protein A or Protein G are typically found in the cell wall of some strains of staphylococci. It is intended to encompass Protein A or Protein G with conservative amino acid substitutions that do not substantially alter its activity.
  • As used herein, “nucleotide binding sequence” refers to a protein or peptide sequence that is capable of specific binding to nucleotide sequences generally, to a set of nucleotide sequences or to a particular nucleotide sequence.
  • As used herein, “lipid binding sequence” refers to a protein or peptide sequence that is capable of specific binding to lipids generally, to a set of lipids or to a particular lipid.
  • As used herein, “polysaccharide binding sequence” refers to a protein or peptide sequence that is capable of specific binding to polysaccharides generally, to a set of polysaccharides or to a particular polysaccharide.
  • As used herein, “metal binding sequence” refers to a protein or peptide sequence that is capable of specific binding to metal ions generally, to a set of metal ions or to a particular metal ion.
  • B. Methods for Preventing, Treating or Delaying Neoplasm Using ErbB-3
  • In one aspect, the present invention is directed to a method for preventing, treating or delaying neoplasm in a mammal, which method comprises administering to a mammal, to which such prevention, treatment or delay is needed or desirable, an effective amount of an ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, whereby an immune response is generated against said neoplasm and said neoplasm is prevented, treated or delayed.
  • The present method can be used for preventing, treating or delaying neoplasm in any mammals, such as mice, rats, rabbits, cats, dogs, pigs, cows, ox, sheep, goats, horses, monkeys and other non-human primates. Preferably, the present method can be used for preventing, treating or delaying neoplasm in humans.
  • Any suitable ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, that can elicit an immune response to the neoplasm to be treated, prevented or delayed, can be used in the present method. The ErbB-3 elicited immune response can be cellular, humoral or both. For example, ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, disclosed in U.S. Pat. No. 5,820,859 can be used in the present method. In other examples, ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, derived from rat ErbB-3 (GenBank Accession No. U29339; and Hellyer et al., Gene, 165(2):279-284 (1995)), Fugu rubripes ErbB-3 (GenBank Accession No. AF056116; and Gellner and Brenner, Genome Res., 9(3):251-258 (1999)) and human ErbB-3 (GenBank Accession No. M29366; and Kraus et al., Proc. Natl. Acad. Sci. U.S.A., 86:9193-9197 (1989)) can be used in the present method. Preferably, ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, derived from human ErbB-3 is used in the present method. Any ErbB-3 protein, or a functional fragment thereof, with conservative amino acid substitutions that do not substantially alter its activity can be used in the present method.
  • In a preferred embodiment, an effective amount of an extracellular domain of an ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an extracellular domain of an ErbB-3 protein, or a functional fragment thereof, is administered. In another preferred embodiment, an effective amount of the ErbB-3 protein comprising an amino acid sequence set forth in SEQ ID NO:1 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14, or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16 is administered. In still another preferred embodiment, an effective amount of the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3, is administered.
  • The present method can further comprise administering an immune response potentiator to the mammal. The immune response potentiator can be administered prior to, concurrently with, or subsequent to the administration of the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid an ErbB-3 protein, or a functional fragment thereof. Exemplary immune response potentiators include Bacille Calmette-Guerin (BCG) (Ratliff, Eur. Urol., 2:17-21 (1992)), Corynebacterium Parvum (Lillehoj et al., Avian Dis., 37(3):731-40 (1993)), Brucella abortus extract, glucan, levamisole, tilorone, an enzyme, a non-virulent virus, polysaccharides, and herb extracts such as Chinese herb extracts.
  • The formulation, dosage and route of administration of ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, preferably in the form of pharmaceutical compositions, can be determined according to the methods known in the art (see e.g., Remington: The Science and Practice of Pharmacy, Alfonso R. Gennaro (Editor) Mack Publishing Company, April 1997; Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems, Banga, 1999; and. Pharmaceutical Formulation Development of Peptides and Proteins, Hovgaard and Frkjr (Ed.), Taylor & Francis, Inc., 2000; Medical Applications of Liposomes, Lasic and Papahadjopoulos (Ed.), Elsevier Science, 1998; Textbook of Gene Therapy, Jain, Hogrefe & Huber Publishers, 1998; Adenoviruses: Basic Biology to Gene Therapy, Vol. 15, Seth, Landes Bioscience, 1999; Biopharmaceutical Drug Design and Development, Wu-Pong and Rojanasakul (Ed.), Humana Press, 1999; Therapeutic Angiogenesis: From Basic Science to the Clinic, Vol. 28, Dole et al. (Ed.), Springer-Verlag New York, 1999). The ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, can be formulated for oral, rectal, topical, inhalational, buccal (e.g., sublingual), parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous), transdermal administration or any other suitable route of administration. The most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, which is being used.
  • The ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, can be administered to any suitable place in the mammal. Preferably, the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, is administered to the neoplasm in situ, i.e., administered to the place where the neoplasm is located or the vicinity thereof. Also, preferably, the present method can further comprise administering an immune response potentiator to the neoplasm in situ.
  • The ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, can be administered alone. Alternatively and preferably, the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, is co-administered with a pharmaceutically acceptable carrier or excipient. Any suitable pharmaceutically acceptable carrier or excipient can be used in the present method (See e.g., Remington: The Science and Practice of Pharmacy, Alfonso R. Gennaro (Editor) Mack Publishing Company, April 1997).
  • The present method can be used alone. Alternatively, the present method can be used in combination with other anti-neoplasm treatment, e.g., radiationtherapy, chemotherapy or surgery. The present method can also be used in combination with other anti-neoplasm agent. Such other anti-neoplasm treatment or agent can be used before, with or after the administration of ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof. For example, the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, can be co-administered with an anti-neoplasm agent.
  • Any suitable anti-neoplasm agent can be used in the present method. Exemplary anti-neoplasm agents include an anti-angiogenic agent (See e.g., Auerbach and Auerbach, Pharmacol. Ther., 63(3):265-311 (1994)), an alkylating agent, an antimetabolite, a natural product, a platinum coordination complex, an anthracenedione, a substituted urea, a methylhydrazine derivative, an adrenocortical suppressant, a hormone, an antagonist, an oncogene inhibitor, a tumor suppressor gene or protein, an anti-oncogene antibody and an anti-oncogene antisense oligonucleotide.
  • The nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, or any tumor suppressor gene can be used in the form of naked DNA, complexed DNA, cDNA, plasmid DNA, RNA or other mixtures thereof as components of the gene delivery system. In another embodiment, the nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, or the tumor suppressor gene is included in a viral vector. Any viral vectors that are suitable for gene therapy can used in the combination. For example, an adenovirus vector (U.S. Pat. No. 5,869,305), a simian virus vector (U.S. Pat. No. 5,962,274), a conditionally replicating human immunodeficiency viral vector (U.S. Pat. No. 5,888,767), retrovirus, SV40, Herpes simplex viral amplicon vectors and Vaccinia virus vectors can be used. In addition, the genes can be delivered in a non-viral vector system such as a liposome wherein the lipid protects the DNA or other biomaterials from oxidation during the coagulation.
  • The present method can be used to treat, prevent or delay any suitable neoplasms or cancers. Preferably, the present method is used to treat, prevent or delay any suitable neoplasms or cancers wherein the interaction between ErbB-2 and ErbB-3 is critical for causing or sustaining the neoplasms or cancers. For example, the present method can be used to treat, prevent or delay adrenal gland, anus, auditory nerve, bile ducts, bladder, bone, brain, breast, bruccal, central nervous system, cervix, colon, ear, endometrium, esophagus, eye, eyelids, fallopian tube, gastrointestinal tract, head and neck, heart, kidney, larynx, liver, lung, mandible, mandibular condyle, maxilla, mouth, nasopharynx, nose, oral cavity, ovary, pancreas, parotid gland, penis, pinna, pituitary, prostate gland, rectum, retina, salivary glands, skin, small intestine, spinal cord, stomach, testes, thyroid, tonsil, urethra, uterus, vagina, vestibulocochlear nerve and vulva neoplasm. Preferably, the present method is used to treat, prevent or delay breast, ovary, stomach, prostate, colon and lung cancer. More preferably, the present method is used to treat, prevent or delay breast cancer.
  • According to the present invention, the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents, carriers or excipients, may be formulated for any suitable administration route, such as intracavernous injection, subcutaneous injection, intravenous injection, intramuscular injection, intradermal injection, oral or topical administration. The method may employ formulations for injectable administration in unit dosage form, in ampoules or in multidose containers, with an added preservative. The formulations may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, sterile pyrogen-free water or other solvents, before use. Topical administration in the present invention may employ the use of a foam, gel, cream, ointment, transdermal patch, or paste.
  • Pharmaceutically acceptable compositions and methods for their administration that may be employed for use in this invention include, but are not limited to those described in U.S. Pat. Nos. 5,736,154; 6,197,801 B1; 5,741,511; 5,886,039; 5,941,868; 6,258,374 B1; and 5,686,102.
  • The magnitude of a therapeutic dose in the treatment or prevention will vary with the severity of the condition to be treated and the route of administration. The dose, and perhaps dose frequency, will also vary according to age, body weight, condition and response of the individual patient.
  • It should be noted that the attending physician would know how to and when to terminate, interrupt or adjust therapy to lower dosage due to toxicity, or adverse effects. Conversely, the physician would also know how to and when to adjust treatment to higher levels if the clinical response is not adequate (precluding toxic side effects).
  • Any suitable route of administration may be used. Dosage forms include tablets, troches, cachet, dispersions, suspensions, solutions, capsules, patches, and the like. See, Remington's Pharmaceutical Sciences.
  • In practical use, the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents, may be combined as the active in intimate admixture with a pharmaceutical carrier or excipient, such as beta-cyclodextrin and 2-hydroxy-propyl-beta-cyclodextrin, according to conventional pharmaceutical compounding techniques. The carrier may take a wide form of preparation desired for administration, topical or parenteral. In preparing compositions for parenteral dosage form, such as intravenous injection or infusion, similar pharmaceutical media may be employed, water, glycols, oils, buffers, sugar, preservatives, liposomes, and the like known to those of skill in the art. Examples of such parenteral compositions include, but are not limited to dextrose 5% w/v, normal saline or other solutions. The total dose of the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents to be administered may be administered in a vial of intravenous fluid, ranging from about 1 ml to 2000 ml. The volume of dilution fluid will vary according to the total dose administered.
  • The invention also provides for kits for carrying out the therapeutic regimens of the invention. Such kits comprise in one or more containers therapeutically effective amounts of the ErbB-3 protein, or a functional fragment thereof, or a nucleic acid encoding an ErbB-3 protein, or a functional fragment thereof, alone or in combination with other agents, in pharmaceutically acceptable form. Preferred pharmaceutical forms would be in combination with sterile saline, dextrose solution, or buffered solution, or other pharmaceutically acceptable sterile fluid. Alternatively, the composition may be lyophilized or dessicated; in this instance, the kit optionally further comprises in a container a pharmaceutically acceptable solution, preferably sterile, to reconstitute the complex to form a solution for injection purposes. Exemplary pharmaceutically acceptable solutions are saline and dextrose solution.
  • In another embodiment, a kit of the invention further comprises a needle or syringe, preferably packaged in sterile form, for injecting the composition, and/or a packaged alcohol pad. Instructions are optionally included for administration of composition by a physician or by the patient.
  • C. Extracellular Domains of the ErbB-3 Protein and Nucleic Acids Encoding the Extracellular Domains of the ErbB-3 Protein and Uses thereof
  • In another aspect, the present invention is directed to an isolated nucleic acid fragment, which isolated nucleic acid fragment hybridizes, under low, middle or high stringency, with a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • In a preferred embodiment, the isolated nucleic acid fragment hybridizes, under high stringency, with a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16. In another preferred embodiment, the isolated nucleic acid fragment comprises a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16. In still another preferred embodiment, the isolated nucleic acid fragment comprises a sequence of nucleotides, or a complementary strand thereof, set forth in SEQ ID NO:4 (FIG. 1) or SEQ ID NO:5 (FIG. 6) or an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • The isolated nucleic acid fragments can be in any suitable form. For example, the isolated nucleic acid fragment can comprise DNA, RNA, PNA or a derivative thereof. Alternatively, the isolated nucleic acid fragment can comprise both DNA and RNA or derivatives thereof. The isolated nucleic acid fragment can be single-stranded and be ready to be used in a hybridization analysis. Alternatively, the isolated nucleic acid fragment can be double-stranded and be denatured into single-stranded prior to the hybridization analysis.
  • The isolated nucleic acid fragment can comprise any kind of oligonucleotide or nucleic acid strand(s) containing genetically-coded and/or naturally occurring structures. The isolated nucleic acid fragments can comprise non-natural elements such as non-natural bases, e.g., inosine and xanthine, non-natural sugars, e.g., 2′-methoxy ribose, or non-natural phosphodiester linkages, e.g., methylphosphonates, phosphorothioates and peptides.
  • The isolated nucleic acid fragments can be produced by any suitable methods. For example, the isolated nucleic acid fragments can be chemically synthesized (See generally, Ausubel (Ed.) Current Protocols in Molecular Biology, 2.11. Synthesis and purification of oligonucleotides, John Wiley & Sons, Inc. (2000)), isolated from a natural source, produced by recombinant methods or a combination thereof. Preferably, the isolated nucleic acid fragments are produced by recombinant methods.
  • The isolated nucleic acid fragment can be labeled for various purposes, e.g., facilitating detection, purification and/or attachment to a surface. The label can be a chemical, an enzymatic, an immunogenic, a radioactive, a fluorescent, a luminescent or a FRET label.
  • A plasmid, which plasmid comprises the above nucleic acid fragment is also provided. A cell, which cell comprises the above plasmid is further provided. Any suitable cells can be used, e.g., bacterial cells, yeast cells, fungal cells, plant cells, insect cells, animal cells and human cells.
  • In still another aspect, the present invention is directed to a method for producing an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, which method comprises growing the above cells under conditions whereby the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, is expressed by the cells, and recovering the expressed extracellular domain of the ErbB-3 protein, or a functional fragment thereof.
  • In yet another aspect, the present invention is directed to a substantially purified protein or peptide, which comprises an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16. The extracellular domain of the ErbB-3 protein, or a functional fragment thereof, can be produced by any suitable methods. For example, the extracellular domain of the ErbB-3 protein, or a functional fragment thereof can be chemically synthesized, isolated from a natural source, produced by recombinant methods or a combination thereof. Preferably, the extracellular domains of the ErbB-3 protein, or functional fragments thereof, are produced by recombinant methods.
  • In yet another aspect, the present invention is directed to a conjugate, which conjugate comprises: a) a protein or peptide comprising an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3; an amino acid sequence comprising at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14; or an amino acid sequence comprising at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16; and b) a facilitating agent linked to the extracellular domain of the ErbB-3 protein, or a functional fragment thereof, directly or via a linker, wherein the agent facilitates: i) affinity isolation or purification of a conjugate; ii) attachment of a conjugate to a surface; or iii) detection of a conjugate. The conjugate can be a fusion protein. Alternatively, the ErbB-3 protein, or a functional fragment thereof, and the facilitating agent can be linked by other means. When the conjugate is a fusion protein, a nucleic acid encoding the conjugate is also provided.
  • The conjugates can be produced by chemical conjugation, such as via thiol linkages, but are preferably produced by recombinant means as fusion proteins. In the fusion protein, the peptide or fragment thereof is linked to either the N-terminus or C-terminus of the ErbB-3 protein, or a functional fragment thereof. In chemical conjugates the peptide or fragment thereof may be linked anywhere that conjugation can be effected, and there may be a plurality of such peptides or fragments linked to a single the ErbB-3 protein, or a functional fragment thereof, or to a plurality thereof.
  • Conjugation can be effected by any method known to those of skill in the art. As described below, conjugation can be effected by chemical means, through covalent, ionic or any other suitable linkage. For example, the reagents and methods for conjugation as disclosed in WO 01/02600 can be used.
  • In some embodiments, the conjugate is a fusion protein, which can be isolated or purified through affinity binding between the protein or peptide fragment of the fusion protein and an affinity binding moiety. Any kind of affinity interaction can be used for isolating or purifying the fusion protein. The affinity interactions, such as those described herein, but not limited to, are protein/protein, protein/nucleotide, protein/lipid, protein/polysaccharide, or protein/metal interactions.
  • In other embodiments, the conjugate can be attached to a surface. More preferably, the conjugate can be attached to the surface through affinity binding between the facilitating agent of conjugate and an affinity binding moiety on the surface. Any kind of affinity interaction can be used for attaching the conjugate, including the protein/protein, protein/nucleotide, protein/lipid, protein/polysaccharide, or protein/metal interactions.
  • In yet another aspect, the present invention is directed to a pharmaceutical composition, which pharmaceutical composition comprises an isolated nucleic acid fragment which isolated nucleic acid fragment hybridizes, under low, middle or high stringency, with a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16 and a pharmaceutically acceptable carrier or excipient. Preferably, the isolated nucleic acid comprises a sequence of nucleotides, or a complementary strand thereof, encoding an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16. The pharmaceutical composition can further comprise an immune response potentiator and/or an anti-neoplasm agent. Vaccines, comprising the above isolated nucleic acid fragments, alone or in combination with an immune response potentiator, are also provided.
  • In yet another aspect, the present invention is directed to a pharmaceutical composition, which pharmaceutical composition comprises a substantially purified protein or peptide, which comprises an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16 and a pharmaceutically acceptable carrier or excipient. The pharmaceutical composition can further comprise an immune response potentiator and/or an anti-neoplasm agent. Vaccines, comprising the above substantially purified proteins or peptides, alone or in combination with an immune response potentiator, are also provided.
  • In yet another aspect, the present invention is directed to an antibody, which antibody binds to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16. Preferably, the antibody binds specifically to an epitope in an extracellular domain of the ErbB-3 protein, or a functional fragment thereof, comprising an amino acid sequence set forth in SEQ ID NO:2 or SEQ ID NO:3 or at least amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO:14 or at least amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16.
  • The antibody can be in any suitable form. For example, the antibody can a polyclonal, monoclonal, chimeric, single chain, human or humanized antibody (See e.g., U.S. Pat. No. 5,968,511). The antibody, in various forms, can be made according to any methods known in the art (See, e.g., Coligan et al. (Ed.), Current Protocols in Immunology, John Wiley & Sons, Inc. (2000)). Pharmaceutical compositions, comprising the above antibodies, alone or in combination with anti-neoplasm agent, and a pharmaceutically acceptable carrier or excipient are also provided.
  • D. Examples
  • The following are exemplary embodiments provided for illustrative purposes only. The inventor discovered the effect and method of B3, DE3-1, rhErbB3412 and rhErbB3-f78 as an anti-tumor vaccine in the treatment of human cancer such as breast cancer.
  • The inventor discovered that B3, DE3-1, rhErbB3412 and rhErbB3-f78 as an anti-tumor vaccine can significantly lower the incidence of the development of human cancer such as breast cancer in high-risk population.
  • The inventor provided a method of B3, DE3-1, rhErbB3-f12 and rhErbB3-f78 as an anti-tumor vaccine significantly lowering the incidence of the development of human cancer such as breast cancer in high-risk population.
  • The inventor discovered that there was significant effect of B3, DE3-1, DE3-1, rhErbB3-f12 and rhErbB3-f78 as an anti-tumor vaccine on postponing the tumor development such as human breast cancer.
  • The inventor discovered that there was significant inhibitory effect of B3, DE3-1, rhErbB3-f12 and rhErbB3-f78 as an anti-tumor vaccine on the tumor development such as human breast cancer.
  • The inventor discovered a method of inhibiting cancerous growth such as breast cancer and that was achieved through inducing immune responses.
  • The aforementioned cells may be a tumor cells, much probably they are human breast cancer cells and other cancerous cells with Erb2/ErbB3 over-expression.
  • It is ErbB3 protein antigen expressed through genetic engineering that made the aforementioned method come true; De3-1, rhErbB3-f12 and rhErbB3-178 is a protein expressed by E. Coli; B3 was an antigen of protein expressed by Eucaryotic cells or ErbB3 antigen produced by other methods, ErbB3 antigen may be ErbB3 molecule or part of a segment of the molecule.
  • Under a typical condition of cancer treatment such as breast cancer, ErbB3 vaccine, which is produced by different methods can inhibit tumor growth under certain dosage level.
  • The aforementioned cancers included breast cancer, ovary carcinoma, gastric carcinoma, and prostate carcinoma and lung cancer.
  • The following description will make the aforementioned invention more clear.
  • 1. Experimental Material and Methods
  • Preparation of B3, De3-1, rhErbB3-f12 and rhErbB3-f78
  • The vaccine involved in the present experiment included protein molecule in the extra-cell membranes region of ErbB3 and part of the protein segment of the extra-cell membranes, they are named as B2 and SD32. The protein molecule in the extra-cell membranes region of ErbB3 and part of the protein of the extra-cell membranes serves as an experiment sample, they are named as B3, De3-1, rhErbB3-f12 and rhErbB3-f78 here; the aforementioned vaccines are manufactured by Zensun (Shanghai) Science and Technology Development Co Ltd. The preparation of B3 and DE3-1, rhErbB3-f12 and rhErbB3-f78 is as follow:
  • Preparing B3
  • B3 gene is the encoded cDNA sequence of protein of ErbB3 extra-cell membrane region (FIG. 1); amplified with PCR, sequence of the primer was:
  • Primer1,
    (SEQ ID NO: 6)
    5′TCTGCGGAGTCATGAGGGC
    Primer2,
    (SEQ ID NO: 7)
    3′TCACTTGTCGTCATCGTCCTTGTAGTCTTTGCCGATCAGCACCAGTGT
  • The italics are flag sequence.
  • After PCR amplification, the targeting gene was cloned into pMD-18T vector; the transformer will be cut by BamHI/Sa1I after enzyme digestion and identified of the sequence to be correct, then connected to pCDNA3BamHI/xhol.
  • Establishment and screening of high performance engineering bacteria: After identification through PCR and enzyme digestion, the engineering bacteria went through 15% SDS-PAGE electrophoresis, thin layer scanning analysis, affinity chromatography, Western-blotting identification and repeated screening a stable high expressive targeting protein engineering bacteria. FIG. 4 illustrated the B3 protein purification, affinity chromatography purification. FIG. 5 showed the targeting protein and amino acid sequence of B3 purified protein after amino acid sequencing.
  • DE3-1 Preparation
  • FIG. 6 showed cDNA sequence of encoded extra-cell membrane ErbB3 protein segment of PCR amplified targeting gene. Structure of the expressed plasmid: targeting gene segment was cut out with BamHI/XhoI from pGEX4T-1 vector (Phamacia company), connecting into pET32a vector (Novagen company) BabHI/XhoI, the protein was expressed by T7 promoter, N end fused with Trx Tag, His Tag and S-Tag, FIG. 7 illustrates the diagram. FIG. 8 illustrates the identification of the plasmid composition.
  • DE3-1 protein expression: Transferring the plasmid into BL21 strain, inoculated the strain into 5 ml of LB+AP, over night; 1:100 inoculated into pre-warmed LB+AP, 37° C. for 2.5-3 hours (OD=0.6); induction with IPTG at 37° C. for 3 hours or 30° C. for 8 hours; centrifuge at 4° C., 6K for 10 minutes, take out the supernatant, put the sediment on ice; PBS suspension was made with cold, 1/20 bacteria solution, then crashed by ultrasound; centrifuge at 4° C., 12K for 10 minutes, large amount of 34 KD targeting gene is harvested (FIG. 9). Purified the DE3-1 protein; DE3-1 emerges in the inclusion bodies, dissolved with 6M guanidine hydrochloride, dialyzed in NTA-O buffer solution (Histag purified solution), good duplicated condition, purified with Histag affinity chromatography (bought from Bo-Cai Company) FIG. 10, after amino acid sequencing, the purified DE3-1 protein was in consistent with the targeting protein sequence, FIG. 11 showed the amino acid sequence.
  • rhErbB3-f12 and rhErbB3-f78 (SEQ ID NO:16) Preparation
  • rhErbB3-f12(SEQ ID NO:14) gene is the encoded cDNA sequence of protein of ErbB3 extra-cell membrane region; amplified with PCR, sequence of the primer was:
  • P1: 5′-TGG  CCA TGG  ACA TCA AGC ATA ATC GGC C-3′ (1645-1664) (SEQ ID NO: 12)
                Ncol
    P2: 3′-GTG  CTC GAG  AGG CTC CCC ATT CAG AAA G-5′ (1800-1818) (SEQ ID NO: 13)
                Xhol
  • Experiment on the Anti-Tumor Effect of B3, DE3-1
  • The Preventive Effect of B3, DE3-1 on Tumor Development.
  • 8-10 weeks old FVB/N transgenic mice (bought from Jackson Lab USA) was selected as experiment animals, the mice were divided into 5 groups with 40 mice each, they were control group, B2, B3 and DE3-1 group; BSA, B2, SD32, B3, DE3-1 was mixed with Freud's adjuvant (CFA, complete Freud's adjuvant, bought from Sigma company) and injected abdominally every 20 days for 7 injections respectively to various groups. The dosage of BSA, B2, SD32, B3 and DE3-1 vaccine was 10, 5, 10, 1 and 10 μg/mouse/injection. Weekly monitor tumour development. The tumour development was verified and analysed statistically.
  • Therapeutic Effect of B3, DE3-1, against Tumour
  • Transplanted tumour model, after immunohistological screening test, about 1000 mm3 tumour mass was cut down from spontaneous tumour of neu protein over-expressed FVB/N transgenic mice. The tumour mass was abraded into single cells with nylon net, the amount injected under the breast of each FVB/N transgenic mice was 5×106 cells. About 10-14 days after inoculation, tumor was palpable (>5 mm) in the control group, demonstrating that the animal model was established successfully.
  • Nothing was administrated in the control group; 24 hours after the inoculation, SD32 and B3 vaccine injection started in SD32 and B3 experiment groups, the aforementioned vaccines were absorbed on 0.1 mg/ml of Al(OH)3 respectively, and injected multi-pointedly every 2 weeks for a total of three injections; the experiment was completed in 14 days after the third injection. Morbidity was monitored weekly, tumor size was measured weekly with vernier. Volume (length diameter×short diameter 2/2) of the tumor was used to represent their size, and curve of tumor growth was protracted,
  • Tumor weight was measured after completion of the experiment and tumor-inhibitory rate was calculated, inhibitory rate=[(tumor weight of control group−tumor weight of experiment groups)/tumor weight of control group]×100.
  • Experiment on the therapeutic effect of various dosage of DE3-1 rhErbB3412 and rhErbB3-f78 on immune therapy against tumor.
  • Preparing animal and transplanted animal tumor model: The same as (Experiment on therapeutic effect of B3 and DE3-1 rhErbB3-f12 and rhErbB3-f78 vaccine on immune therapy against tumor). No treatment was given to the control group, histag protein was injected to the negative control group, and Adriamycin (Santou MingZhi Pharmaceuticals) was administered for the positive control group, 5 μg, 20 μg and 80 μg was given to DE3-1 group respectively.
  • One day after the inoculation, Adriamycin 2.2 mg/kg was injected abdominally for consecutive 7 days in mice of the positive control group; histag protein+Al(OH)3 was injected abdominally for mice of the negative control group; In DE3-1 group, the vaccine was absorbed on 0.1 mg of Al(OH)3 and multi-points subcutaneous injection every 2 weeks for a total of 3 injections were carried out in mice. The experiment completed in 14 days after the third injection. Tumor development was monitored weekly, tumor size was measured with vernier and the size was expressed as (length diameter×short diameter 2/2), curve of tumor growth was protracted and analyzed statistically.
  • Tumor weight was measured after completion of the experiment and tumor-inhibitory rate was calculated, tumor inhibitory rate=[(tumor weight of control group−tumor weight of experiment groups)/tumor weight of control group]×100.
  • Experiment on Cross Immunity of B2 and B3 Antigen
  • FVB transgenic mice were immuned with B2 protein and B3 protein respectively, 10 days thereafter, blood was withdrawn and antibody titer was tested with ELISA. 0.3 μg/hole of B2 and B3 was wrapped, 1:1000 B2 and B3 on each plate were titrated with standard serum respectively, cultured at 37° C. for 30 minutes, sealed with 1% BSA, added double antibody, color development for 15 minutes with DAD, tested with Bio-Rad 450 nm enzyme labeled device.
  • 2. Experiment Results and Discussion
  • Table 1 and FIG. 12 illustrate the experimental results of tumor inhibitory effect of B3 and DE3-1.
  • TABLE 1
    Experimental results of tumor inhibitory
    effect of B3 and DE3-1 vaccine
    Dosage Time of Incidence
    (μg/ tumor of tumor
    Case animal/ occurrence growth
    Grouping number Treatment dose) (weeks) (%)
    Negative 40 BSA + CFA 10 19 37.5
    control group
    B2 experiment
    40 B2 + CFA 5 21 12.5
    group
    SD32
    40 SD32 + CFA 10 22 10
    experiment
    group
    B3 experiment
    40 B3 + CFA 5 20 12.5
    group
    DE3-1 40 DE3-1 + 10 23 35
    experiment CFA
    group
  • Objective of the present experiment is to explore whether there is preventive effect of B3 or DE3-1 vaccine on tumor development. The reason to choose this type of transgenic mice as experiment animal model, is because rat wild type neu cDNA controlled by mice breast virus promotor was transferred into the body of mice and produce over-expression of neu protein and spontaneous breast cancer occurred within 5-8 months in half of the mice. Natural course of tumor in the transgenic mice and its pathologic pattern is similar to that of human breast cancer. Therefore, it may have better therapeutic effect when used clinically. The sample contains 40 animals in each group, the aim of selecting such large sample is to ensure the number of cases which have the disorders will be greater than 10 animals, thus will be of greater implication statistically. The selection of dosage is based on the results of pre-experiments.
  • Transgenic mice were immunized with BSA, B2, B3, SD32 and DE3-1 respectively, as we can see from the tables and figures, the tumor incidence of 37.5% began from the 19th week on in the negative control group; whereas the time of tumor development in SD32, B3 and B2 group was 21, 22, and 20 weeks with their incidence of 10%, 12.5% and 12.5% respectively, demonstrating that there were significant tumor-inhibitory effect of SD32, B3 and B2 vaccine against the development of tumor (P<0.025; ×2 testing); at the same time, they can postpone the time of tumor development. The occurrence of tumor in DE3-1 group is later than that in the control group, however the tumor incidence of 35% was not significantly different from that of the control group (P<0.05; ×2 testing).
  • Experimental Results of Anti-Tumor Effect of B3 and DE3-1 Vaccine
  • Table 2 and FIG. 13-14 show experimental results of anti-tumor effect of B3 vaccine
  • TABLE 2
    Experimental results of anti-tumor effect of B3 and DE3-1
    Inhib-
    Tumor Tumor itory
    size weight rate
    Grouping Treatment (mm3) (g) (%)
    Negative histag 7849.8 ± 849.8 5.76 ± 0.55
    control protein +
    group A1(OH)3
    SD32 SD32 + A1(OH)3 4246.5 ± 540.6 3.28 ± 0.36 46
    experiment
    group
    B3 B3 + A1(OH)3 5271.8 ± 658.9 3.13 ± 0.33 33
    experiment
    group
  • In order to identify the anti-tumor therapeutic effect of B3, the inventor carried out experiment on immune therapy with B3 in transplanted tumor model.
  • Table 2 and FIG. 13-14 illustrate the effect of various vaccines on tumor growth in mice, demonstrating that the tumor-inhibitory rate of SD32 and B3 were 46% and 33% respectively, and that both of them had significant tumor-inhibitory effect (P<0.01; t testing).
  • Experimental Results of Anti-Tumor Effect of DE3-1, rhErbB3412 and rhErbB3478
  • Dosage of 5 μg, 20 μg and 80 μg/animal were used to immunized mice in the experiment group, table 3 and FIG. 15-16 showed the experimental results.
  • TABLE 3
    Experimental results of anti-tumor effect of DE3-1
    Case Tumor size Tumor weight Inhibitory rate
    Grouping number Treatment (mm3) (g) (%)
    Control group 8 6742.9 ± 657.8 4.769 ± 0.56
    Negative 8 Histag 6476.9 ± 567.9 4.461 ± 0.52
    control group protein + A1(OH)3
    Positive control 8 ADR 2.2 mg/kg 4603.1 ± 478.3 3.564 ± 0.42 25.3
    group
    DE3-1 experiment 8 80 μg DE3-1 + A1(OH)3 4810.8 ± 460.5 3.658 ± 0.37 26.3
    group
    DE3-1 experiment 8 20 μg DE3-1 + A1(OH)3 4715.0 ± 434.8 3.455 ± 0.41 28.9
    group
    DE3-1 experiment 8 5 μg DE3-1 + A1(OH)3 5563.7 ± 600.6 3.687 ± 0.45 22.4
    group
  • Tumor-inhibitory rate and measured tumor size was consistent among groups with various dosage of DE3-1, the best tumor-inhibitory effect was seen in 20 μg level of DE3-1, reaching about 28.9%. After completion of the experiment, the mice were killed, took out the tumor and measured their weight; there were significant difference (P<0.001, t test) between the positive control group, groups with various dosage level, negative control group and placebo control group. The tumor-inhibitory rate of 5 μg, 20 μg and 80 μg dosage level group were 26.3%, 22.4% and 28.9% respectively.
  • TABLE 4
    Experimental results of anti-tumor effect of rhErbB3-f12
    Inhib-
    Tumor itory
    Case Dosage weight rate
    Grouping number (mg/kg) Treatment (g) (%)
    Negative 14 sc X 3 q14d 5.55 ± 1.25
    control
    Positive
    7 10 ip X 7 qd 3.09 ± 1.08* 44.32
    control (Taxol)
    rhErbB3-f12 7 1 sc X 3 q14d 2.40 ± 0.49* 56.76
    rhErbB3-f12 7 0.5 sc X 3 q14d 2.62 ± 0.67* 52.61
    rhErbB3-f12 7 0.25 sc X 3 q14d 2.31 ± 0.40* 58.39
  • TABLE 5
    Experimental results of anti-tumor effect of rhErbB3-f78
    Inhib-
    Tumor itory
    Case Dosage weight rate
    Grouping number (mg/kg) Treatment (g) (%)
    Negative 14 2.5 sc X 3 q14d 1.098 ± 0.17
    control
    Positive
    7 2 ip X 7 qd 0.648 ± 0.27* 40.98
    control
    (ADM)
    rhErbB3-f78 7 2.5 sc X 3 q14d 0.435 ± 0.12* 60.38
  • Experiment on Cross Immunity between B2 and B3 Antigen
  • The objective of experiment on cross immunity between B2 and B3 antigen is to explore whether there exists cross immunity between B2 and B3 antigen. FIG. 17-18 showed the experimental results demonstrating that there isn't any cross immunity between B2 and B3 antigen.
  • 3. Summary
  • In this research, we discovered new promising vaccines of B3 and DE3-1, which are designed on the basis of a new anti-tumor targeting ErbB3, and have preventive effect on tumor development and immune therapeutical effect against tumor.
  • The over-expression of ErbB2 receptor existed in part of adenocarcinoma discovered in the previous studies was considered to be associated with cancer development after formation of homogenous dimer. Over-expression of ErbB2 was considered to be the major cause of adenocarcinoma development, it is due to: 1) the amplification of ErbB2 gene existed in tumor cells such as breast cancer and ovary carcinoma was the cause of over-expression of ErbB2; 2) Over-expression of ErbB2 leads to phosphorylation in its cellular functional area and affects the interaction between intracellular signal molecule Shc and ErbB2; 3) the transfection of wild type ErbB2 into fibroblast can lead to cell transformation; 4) the enhancement of the formation of ErbB2 variants from ErbB2 homogenous dimer can also enhance its activity of cell transformation.
  • Prior to the present discovery, the inventors have discovered ErbB3 as another new anti-tumor target in addition to ErbB2. The inventors clarify that over-expression of ErbB2 receptor leads to the formation of heterogenous dimer from ErbB2 and ErbB3, and that was the cause of cancer development. Discovery of this new target provides us with new concept of anti-cancer therapeutical method: use extra-cell membrane protein of ErbB3 cells for cancer prevention and treatment, to lower the incidence of breast cancer and produce effect against tumor growth.
  • The tremendous success of humanized monoclonal antibody-herceptin targeted on ErbB2 is based on the relativity between over-expression of ErbB2 and occurrence of various tumors. However, the co-expression of ErbB2 and ErbB4 receptors in myocardial cells leads to the formation of heterogenous dimer from ErbB2 receptor and ErbB4 receptor; the dimer was very important in the maintain of normal structure of myocardial cells, thus, anti-cancer medicine targeting on ErbB2 receptor has damages on myocardial cells and leads to heart failure; however, anti-cancer drug targeting at ErbB3 receptor doesn't have this adverse reaction. Therefore, the use of ErbB3 as a specific anti-tumor vaccine against breast cancer, ovary carcinoma, gastrocarcinoma, prostate cancer, rectal cancer and lung cancer will play a very important role in the prevention and treatment of these cancers.
  • The above examples are included for illustrative purposes only and are not intended to limit the scope of the invention. Many variations to those described above are possible. Since modifications and variations to the examples described above will be apparent to those of skill in this art, it is intended that this invention be limited only by the scope of the appended claims.

Claims (18)

1-28. (canceled)
29. An antibody which binds to an ErbB-3 protein, wherein the ErbB-3 protein is a fragment of the extracellular domain of ErbB-3 and comprises:
a) the amino acid sequence set forth in SEQ ID NO:3; or;
b) amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO: 14; or
c) amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16
wherein the ErbB-3 protein is not the entire extracellular domain of ErbB-3.
30. The antibody of claim 29, which is a polyclonal or monoclonal antibody.
31. The antibody of claim 29, which is a human or humanized antibody.
32-43. (canceled)
44. The antibody of claim 29, further comprising administering an immune response potentiator to the mammal.
45. The antibody of claim 29, wherein the antibody is co-administered with a pharmaceutically acceptable carrier or excipient.
46. The antibody of claim 29, wherein the antibody is co-administered with an anti-neoplasm agent.
47. The antibody of claim 46, wherein the anti-neoplasm agent is selected from the group consisting of an anti-angiogenic agent, an alkylating agent, an antimetabolite, a natural product, a platinum coordination complex, an anthracenedione, a substituted urea, a methylhydrazine derivative, an adrenocortical suppressant, a hormone, an antagonist, an oncogene inhibitor, a tumor suppressor gene or protein, an anti-oncogene antibody and an anti-oncogene antisense oligonucleotide.
48. The antibody of claim 46, wherein the neoplasm to be treated is selected from the group consisting of adrenal gland, anus, auditory nerve, bile ducts, bladder, bone, brain, breast, bruccal, central nervous system, cervix, colon, ear, endometrium, esophagus, eye, eyelids, fallopian tube, gastrointestinal tract, head and neck, heart, kidney, larynx, liver, lung, mandible, mandibular condyle, maxilla, mouth, nasopharynx, nose, oral cavity, ovary, pancreas, parotid gland, penis, pinna, pituitary, prostate gland, rectum, retina, salivary glands, skin, small intestine, spinal cord, stomach, testes, thyroid, tonsil, urethra, uterus, vagina, vestibulocochlear nerve and vulva neoplasm.
49. The antibody of claim 46, wherein the neoplasm to be treated is selected from the group consisting of breast, ovary, stomach, prostate, colon and lung cancer.
50. The antibody of claim 46, wherein the neoplasm to be treated is breast cancer.
51. The antibody of claim 29, wherein the antibody is administered by intracavernous injection, subcutaneous injection, intravenous injection, intramuscular injection, intradermal injection, oral administration or topical administration.
52. The antibody of claim 29, wherein the antibody is administered by subcutaneous injection.
53. The antibody of claim 29, wherein the antibody is administered to the neoplasm in situ.
53. The antibody of claim 44, wherein the immune response potentiator is administered to the neoplasm in situ.
54. A method for treating neoplasm which express ErbB-3 in a mammal using the antibody of claim 29.
55. A method for preparing the antibody of claim 29, wherein the method comprises:
a. providing an antigen comprising the amino acid sequence set forth in SEQ ID NO:3, amino acid residues 24-81 of the amino acid sequence set forth in SEQ ID NO: 14, or amino acid residues 2-139 of the amino acid sequence set forth in SEQ ID NO:16;
b. exposing an antibody repertoire or potential antibody repertoire to one of the above amino acid sequences; and
c. selecting from the repertoire an antibody which specifically binds to one of the above amino acid sequences.
US13/035,244 2002-03-26 2011-02-25 Erbb3 based methods and compositions for treating neoplasms Abandoned US20110229478A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/035,244 US20110229478A1 (en) 2002-03-26 2011-02-25 Erbb3 based methods and compositions for treating neoplasms

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN02116259.X 2002-03-26
CNB02116259XA CN1219882C (en) 2002-03-18 2002-03-26 Method and combination for treating tumors based on ERBB-3
PCT/CN2003/000217 WO2003080835A1 (en) 2002-03-26 2003-03-26 Erbb3 based methods and compositions for treating neoplasms
US10/516,759 US7919098B2 (en) 2002-03-26 2003-03-26 ErbB-3 based methods and compositions for treating neoplasms
US13/035,244 US20110229478A1 (en) 2002-03-26 2011-02-25 Erbb3 based methods and compositions for treating neoplasms

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2003/000217 Division WO2003080835A1 (en) 2002-03-26 2003-03-26 Erbb3 based methods and compositions for treating neoplasms
US11/516,759 Division US20080233410A1 (en) 2005-11-17 2006-09-07 Transition metal complex compound

Publications (1)

Publication Number Publication Date
US20110229478A1 true US20110229478A1 (en) 2011-09-22

Family

ID=28048642

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/516,759 Active 2025-07-26 US7919098B2 (en) 2002-03-26 2003-03-26 ErbB-3 based methods and compositions for treating neoplasms
US13/035,244 Abandoned US20110229478A1 (en) 2002-03-26 2011-02-25 Erbb3 based methods and compositions for treating neoplasms

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/516,759 Active 2025-07-26 US7919098B2 (en) 2002-03-26 2003-03-26 ErbB-3 based methods and compositions for treating neoplasms

Country Status (7)

Country Link
US (2) US7919098B2 (en)
EP (2) EP1495123B1 (en)
JP (2) JP4660094B2 (en)
CN (1) CN100424175C (en)
AU (1) AU2003218600C1 (en)
CA (1) CA2480099C (en)
WO (1) WO2003080835A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US9220775B2 (en) 2011-11-23 2015-12-29 Medimmune Llc Binding molecules specific for HER3 and uses thereof
WO2017014810A1 (en) * 2015-07-17 2017-01-26 Czerniecki Brian J Identification of immunogenic mhc class ii peptides for immune-based therapy
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
US10745490B2 (en) 2014-04-11 2020-08-18 Celldex Therapeutics, Inc. Anti-ErbB antibodies and methods of use thereof
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
US11305012B2 (en) 2013-09-24 2022-04-19 Medimmune, Llc Binding molecules specific for HER3 and uses thereof

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1940461B1 (en) * 2005-11-02 2014-01-08 Duke University Concurrent chemotherapy and immunotherapy
ES2664086T3 (en) 2005-12-30 2018-04-18 Zensun (Shanghai) Science & Technology, Co., Ltd. Extended release of neurregulin to improve cardiac function
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
EP3248617A3 (en) 2007-02-16 2018-02-21 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
US20090156488A1 (en) * 2007-09-12 2009-06-18 Zensun (Shanghai) Science & Technology Limited Use of neuregulin for organ preservation
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
CN102231987A (en) * 2008-11-28 2011-11-02 上海泽生科技开发有限公司 Neuregulin and cardiac stem cells
US8609620B2 (en) 2008-11-28 2013-12-17 Zensun (Shanghai) Science & Technology Ltd. Neuregulin peptides and their use
LT3351558T (en) 2009-11-13 2020-06-10 Daiichi Sankyo Europe Gmbh Material and methods for treating or preventing her-3 associated diseases
HUE029026T2 (en) 2009-12-22 2017-01-30 Roche Glycart Ag ANTI-HER3 Antibodies and uses thereof
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
IT1402149B1 (en) * 2010-10-04 2013-08-28 Ist Fisioterap Ospitalroma USE OF A PHOSPHOPEPTIDE WHICH IS ABLE TO BLOCK THE HER3 / P85 INTERACTION FOR THE TREATMENT OF HER2 EXPRESSION TUMORS.
ITRM20100577A1 (en) 2010-11-02 2012-05-03 Takis Srl IMMUNOTHERAPY AGAINST THE ERBB-3 RECEPTOR
TWI418362B (en) * 2010-11-19 2013-12-11 Univ Nat Pingtung Sci & Tech Recombinant outer membrane lipoprotein e of pasteurella haemolytica and vaccine composition containing the same
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
US20150231238A1 (en) 2011-03-15 2015-08-20 Merrimack Pharmaceuticals, Inc. Overcoming resistance to erbb pathway inhibitors
WO2013053076A1 (en) 2011-10-10 2013-04-18 Zensun (Shanghai)Science & Technology Limited Compositions and methods for treating heart failure
CA2862306C (en) * 2012-01-19 2019-08-27 Duke University Vaccines against antigens involved in therapy resistance and methods of using same
AR094403A1 (en) 2013-01-11 2015-07-29 Hoffmann La Roche ANTI-HER3 ANTIBODY COMBINATION THERAPY
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
CA2954279C (en) 2014-07-07 2023-11-14 Duke University Vaccines against an oncogenic isoform of esr1 and methods of using the same
US20170196953A1 (en) 2014-07-07 2017-07-13 Duke University VACCINES AGAINST AN ONCOGENIC ISOFORM OF HER2 (ErbB2) AND METHODS OF USING THE SAME
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
US11090284B2 (en) 2015-09-02 2021-08-17 The Cleveland Clinic Foundation Ovarian cancer vaccines
CN108697779B (en) 2016-01-07 2023-09-19 杜克大学 Cancer vaccine and delivery method
WO2018050848A1 (en) * 2016-09-15 2018-03-22 Universität Stuttgart Antigen binding protein against her3
US10487143B2 (en) 2016-10-05 2019-11-26 Duke University Vaccines against HER3 antigens and methods of using the same
US11224665B2 (en) 2016-10-05 2022-01-18 Duke University Mitochondrial antiviral signaling (MAVS) protein compositions and methods of using the same
AU2018341578B2 (en) * 2017-09-27 2024-02-08 L2 Diagnostics, Llc ErbB peptide pharmaceutical and vaccine compositions and therapeutics uses thereof for cancer

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US5686102A (en) * 1992-06-26 1997-11-11 Lancaster Group Ag Pharmacological composition for topical administration
US5736154A (en) * 1996-03-11 1998-04-07 Fuisz Technologies Ltd. Transdermal delivery system
US5741511A (en) * 1995-04-12 1998-04-21 Sam Yang Co., Ltd. Transdermal drug delivery device for treating erectile dysfunction
US5869305A (en) * 1992-12-04 1999-02-09 The University Of Pittsburgh Recombinant viral vector system
US5886039A (en) * 1988-09-02 1999-03-23 Kock; Nils G. Method and composition for treating erectile dysfunction
US5888767A (en) * 1996-11-27 1999-03-30 The Johns Hopkins University School Of Medicine Method of using a conditionally replicating viral vector to express a gene
US5941868A (en) * 1995-12-22 1999-08-24 Localmed, Inc. Localized intravascular delivery of growth factors for promotion of angiogenesis
US5962274A (en) * 1998-03-13 1999-10-05 Wake Forest University Viral vector directed to predetermined target cells
US5968511A (en) * 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
US6197801B1 (en) * 1998-01-14 2001-03-06 Usa Doctors Products, Inc. Injectable pharmaceutical composition for treatment and reversal of erectile dysfunction
US6258374B1 (en) * 1997-09-08 2001-07-10 Astra Aktiebolag Foam-forming pharmaceutical composition
US20040197332A1 (en) * 2001-08-09 2004-10-07 Axel Ullrich Inhibitors of her3 activity

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3859695B2 (en) 1989-08-04 2006-12-20 バーレックス ラボラトリーズ インコーポレイテッド C-erbB-2 foreign domain: GP75
DK0896586T4 (en) 1996-03-27 2015-08-24 Genentech Inc ErbB3 antibodies.
IL127891A0 (en) * 1996-07-12 1999-10-28 Genentech Inc Chimeric heteromultimeter adhesins
NZ509480A (en) 1996-10-18 2005-05-27 Genentech Inc Use of anti-ErbB2 antibodies which bind to domain1 of ErbB2 for treating a tumour or cancer
US5981201A (en) 1997-01-08 1999-11-09 Beth Israel Deaconess Medical Center Methods of detection and treatment of breast cancer
ATE427353T1 (en) 1997-02-10 2009-04-15 Genentech Inc HEREGULIN VARIANTS
AUPQ105799A0 (en) 1999-06-18 1999-07-08 Victor Chang Cardiac Research Institute, The Cell growth inhibition
AU5781800A (en) 1999-07-06 2001-01-22 General Atomics Methods and compositions for assaying analytes
AU784617B2 (en) 2000-05-15 2006-05-18 Pharmacia & Upjohn Company Aromatase inhibitors and monoclonal anti-HER2 antibodies as antitumors agents
AU2005327973A1 (en) 2005-02-23 2006-08-31 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
EP3248617A3 (en) 2007-02-16 2018-02-21 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5886039A (en) * 1988-09-02 1999-03-23 Kock; Nils G. Method and composition for treating erectile dysfunction
US5480968A (en) * 1989-12-01 1996-01-02 The United States Of America As Represented By The Department Of Health And Human Services Isolated polypeptide erbB-3, related to the epidermal growth factor receptor and antibody thereto
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
US5820859A (en) * 1989-12-01 1998-10-13 The United States Of America As Represented By The Department Of Health And Human Services Method of targeting a therapeutic agent to cells expressing the erb B-3 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
US5686102A (en) * 1992-06-26 1997-11-11 Lancaster Group Ag Pharmacological composition for topical administration
US5869305A (en) * 1992-12-04 1999-02-09 The University Of Pittsburgh Recombinant viral vector system
US5741511A (en) * 1995-04-12 1998-04-21 Sam Yang Co., Ltd. Transdermal drug delivery device for treating erectile dysfunction
US5941868A (en) * 1995-12-22 1999-08-24 Localmed, Inc. Localized intravascular delivery of growth factors for promotion of angiogenesis
US5736154A (en) * 1996-03-11 1998-04-07 Fuisz Technologies Ltd. Transdermal delivery system
US5968511A (en) * 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
US20050136494A1 (en) * 1996-03-27 2005-06-23 Genentech, Inc. Isolated nucleic acids, vectors and host cells encoding ErbB3 antibodies
US7285649B2 (en) * 1996-03-27 2007-10-23 Genentech, Inc. Isolated nucleic acids, vectors and host cells encoding ErbB3 antibodies
US5888767A (en) * 1996-11-27 1999-03-30 The Johns Hopkins University School Of Medicine Method of using a conditionally replicating viral vector to express a gene
US6258374B1 (en) * 1997-09-08 2001-07-10 Astra Aktiebolag Foam-forming pharmaceutical composition
US6197801B1 (en) * 1998-01-14 2001-03-06 Usa Doctors Products, Inc. Injectable pharmaceutical composition for treatment and reversal of erectile dysfunction
US5962274A (en) * 1998-03-13 1999-10-05 Wake Forest University Viral vector directed to predetermined target cells
US20040197332A1 (en) * 2001-08-09 2004-10-07 Axel Ullrich Inhibitors of her3 activity

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Kraus et al (PNAS, 1989, 86:9193-9197, IDS) *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US9220775B2 (en) 2011-11-23 2015-12-29 Medimmune Llc Binding molecules specific for HER3 and uses thereof
US10040857B2 (en) 2011-11-23 2018-08-07 Medimmune, Llc Binding molecules specific for HER3 and uses thereof
US11091554B2 (en) 2011-11-23 2021-08-17 Medlmmune, Llc Binding molecules specific for HER3 and uses thereof
US11305012B2 (en) 2013-09-24 2022-04-19 Medimmune, Llc Binding molecules specific for HER3 and uses thereof
US10745490B2 (en) 2014-04-11 2020-08-18 Celldex Therapeutics, Inc. Anti-ErbB antibodies and methods of use thereof
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
WO2017014810A1 (en) * 2015-07-17 2017-01-26 Czerniecki Brian J Identification of immunogenic mhc class ii peptides for immune-based therapy

Also Published As

Publication number Publication date
US7919098B2 (en) 2011-04-05
AU2003218600A1 (en) 2003-10-08
EP2400021B1 (en) 2016-11-30
AU2003218600B2 (en) 2009-05-21
JP2005533488A (en) 2005-11-10
CA2480099C (en) 2019-01-08
JP4660094B2 (en) 2011-03-30
JP5249282B2 (en) 2013-07-31
WO2003080835A1 (en) 2003-10-02
CN100424175C (en) 2008-10-08
EP2400021A2 (en) 2011-12-28
EP1495123A1 (en) 2005-01-12
EP1495123A4 (en) 2009-06-24
CA2480099A1 (en) 2003-10-02
EP1495123B1 (en) 2013-10-30
CN1650011A (en) 2005-08-03
EP2400021A3 (en) 2012-03-21
JP2010213710A (en) 2010-09-30
AU2003218600C1 (en) 2009-12-17
US20080057064A1 (en) 2008-03-06

Similar Documents

Publication Publication Date Title
US7919098B2 (en) ErbB-3 based methods and compositions for treating neoplasms
AU2016201712B2 (en) ActRIIb antagonists and dosing and uses thereof
US11235043B2 (en) Vaccines against antigens involved in therapy resistance and methods of using same
WO2010105573A1 (en) Anti-angiogenic fusion proteins
JP2007297398A (en) Reagent and method for targeting mutant epidermal growth factor receptor
US11883464B2 (en) Nerve growth factor fusion protein, preparation method and use thereof
JP2005533000A (en) Method for enhancing immunity induction involving MDA-7
JPH01502669A (en) Purified platelet-derived growth factor and its purification method
JP2003524021A (en) Compositions and methods for diagnosis and treatment of malignant mesothelioma
JP2001526040A (en) Anti-angiogenic protein using Pichia yeast expression system: Method for producing endostatin, angiostatin or restin
AU2002322762B2 (en) Hybrid proteins with neuregulin heparin-binding domain for targeting to heparan sulfate proteoglycans
AU2002322762A1 (en) Hybrid proteins with neuregulin heparin-binding domain for targeting to heparan sulfate proteoglycans
WO2022100585A1 (en) Anti-her-2 antibody-chemokine fusion protein, preparation method therefor and application thereof
US20080095789A1 (en) Vaccine
CN115814105A (en) Application of antibody drug conjugate, combined drug and application thereof
EP1832604A1 (en) Use of IEX-1 for the treatment of glioma tumors
CA2541840C (en) P185neu-encoding dna and therapeutical uses thereof
US20240131114A1 (en) Nerve growth factor fusion protein, preparation method and use thereof
WO2023077924A1 (en) Vaccine against pancreatic cancer, and medical use thereof
KR20230120543A (en) Cancer Vaccine Comprising Epitopes of c-Met and Its Uses
WO2018021892A1 (en) Composition for treating or sensitizing interferon beta resistant cancer disease comprising cflip sirna
CN1444992A (en) Method and combination for treating tumors based on ERBB-3

Legal Events

Date Code Title Description
AS Assignment

Owner name: ZENSUN (SHANGHAI) SCIENCE & TECHNOLOGY, CO., LTD., CHINA

Free format text: CHANGE OF NAME;ASSIGNOR:ZENSUN (SHANGHAI) SCIENCE & TECHNOLOGY, LTD.;REEL/FRAME:038925/0398

Effective date: 20150728

Owner name: ZENSUN (SHANGHAI) SCIENCE & TECHNOLOGY, CO., LTD.,

Free format text: CHANGE OF NAME;ASSIGNOR:ZENSUN (SHANGHAI) SCIENCE & TECHNOLOGY, LTD.;REEL/FRAME:038925/0398

Effective date: 20150728

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION