WO2007144893A2 - Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof - Google Patents

Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof Download PDF

Info

Publication number
WO2007144893A2
WO2007144893A2 PCT/IL2007/000732 IL2007000732W WO2007144893A2 WO 2007144893 A2 WO2007144893 A2 WO 2007144893A2 IL 2007000732 W IL2007000732 W IL 2007000732W WO 2007144893 A2 WO2007144893 A2 WO 2007144893A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
fgfr2
molecule
fragment
affinity
Prior art date
Application number
PCT/IL2007/000732
Other languages
French (fr)
Other versions
WO2007144893A3 (en
Inventor
Avner Yayon
Eran Rom
Original Assignee
Fibron 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
Application filed by Fibron Ltd. filed Critical Fibron Ltd.
Priority to EP07766788A priority Critical patent/EP2046384A4/en
Priority to CA002655504A priority patent/CA2655504A1/en
Priority to US12/304,638 priority patent/US8101721B2/en
Publication of WO2007144893A2 publication Critical patent/WO2007144893A2/en
Publication of WO2007144893A3 publication Critical patent/WO2007144893A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention is related in general to antibodies with binding affinity to fibroblast growth factor receptors (FGFRs) which block both ligand-dependent and constitutive ligand independent receptor activation.
  • FGFRs fibroblast growth factor receptors
  • the present invention relates to antibodies specific for more than one receptor subtype, and fragments thereof, useful for treating diseases and disorders, including cell proliferative diseases.
  • Fibroblast Growth Factor ligands constitute a family of over twenty structurally related polypeptides that are developmentally regulated and expressed in a wide variety of tissues. FGFs stimulate proliferation, cell migration and differentiation and play a major role in skeletal and limb development, wound healing, tissue repair, hematopoiesis, angiogenesis, and tumorigenesis (reviewed in Ornitz and Itoh, 2001).
  • FGFs The biological action of FGFs is mediated by specific cell surface receptors belonging to the receptor protein tyrosine kinase (RPTK) family of protein kinases. These proteins consist of an extracellular ligand binding domain, a single transmembrane domain and an intracellular tyrosine kinase domain that undergoes phosphorylation upon binding of FGF.
  • the FGF receptor (FGFR) extracellular region contains three immunoglobulin-like (Ig-like) domains (Dl, D2 and D3), an acidic box, and a heparin- binding domain. Five FGFR genes encoding for multiple receptor variants have been identified to date. Alternative splicing further increases the diversity of the FGFR family.
  • the second half of the third Ig-like domain in FGFRl, 2 and 3 is encoded by one of two exons, HIb or IHc, generating receptors with different ligand affinities and specificities.
  • FGFR3 is the most frequently mutated oncogene in transitional cell carcinoma (TCC) of the bladder where it is mutated in more than 30% of the cases (Cappeln
  • FGFR3 mutations seem to have a central role in the early development of papillary bladder tumors and can serve as a target for treatment (Billerey et al., 2001; Cappeln et al., 1999). These tumors follow a common molecular pathway, which is different from tumors with concomitant carcinoma in situ (CIS). However, FGFR3 mutations do not seem to play a role in bladder cancer progression (Zieger, et al., 2005).
  • FGFR2 mRNAs were found to be overexpressed in both human pancreatic cancer cells and the adjacent pancreatic parenchyma (Ishiwata et al., 1998,) and Kurban et al., (2004) identified FGFR2 IHb (KGFR) expression in cervical cancer cells. Lorenzi et al. (1996) have identifies a constitutively active form of FGFR2 in rat osteosarcoma cells.
  • FGFR2 exhibits expression of the IHb isoform in epithelial type tissues and the uroepithelium and the IHc isoform in the mesenchyme.
  • IHb (FGFR2-IIIb) was shown to have tumor suppressive properties, i.e. be downregulated in a subset of transitional cell carcinomas of the bladder (Bernard-Pierrot et al., 2004; Ricol et al. 1999).
  • SNPs single nucleotide polymorphisms
  • PCT Publication WO 2006/048877 co-assigned to the assignee of the present invention teaches a method of treating multiple myeloma comprising administering to an individual in need thereof an anti-FGFR3 antibody which is specific for wild type FGFR3.
  • PCT Publication WO 2004/110487 assigned to the assignee of the present invention provides a method of treating a T cell mediated disease comprising administering to a subject in need thereof an FGFR3 antagonist.
  • FGFR polypeptides and methods of use thereof for treating tumors. That application provides tables disclosing human tumors, which express the various FGFR proteins but there is no teaching of cross reactive antibodies. Martinez-Torrecuadrada et al. (2005) teach anti-FGFR3 antibodies which inhibit bladder carcinoma cell proliferation.
  • Kan et al. (1993) disclose that heparin interacts with a specific region in the extracellular domain of the FGFR, and is essential for FGFR activation. They synthesized a peptide corresponding to the heparin binding domain of FGFRl and raised polyclonal antibodies specific to this peptide. Both the peptide and the antibodies were antagonistic to FGFl -stimulated cell growth. However, Kan et al. did not demonstrate blocking of other FGF receptors nor do they show inhibition of constitutive receptor activation.
  • the present invention provides antibodies having high specificity to fibroblast growth factors of more than one subtype.
  • the present invention further provides antibodies having high affinity to fibroblast growth factor receptor 2 (FGFR2) having cross-reactivity to other fibroblast growth factor receptors (FGFRs) useful in blocking both ligand-dependent and constitutive ligand-independent receptor activation.
  • FGFR2 fibroblast growth factor receptor 2
  • FGFRs fibroblast growth factor receptors
  • the present invention is related to molecules comprising at least the antigen-binding portion of an antibody having affinity for fibroblast growth factor receptor 2 (FGFR2) optionally with cross-reactivity to other FGF receptors, which block both ligand-dependent and constitutive ligand-independent receptor activation.
  • the antibody which blocks activation of fibroblast growth factor receptors (FGFR) has high affinity to both FGFR2 and FGFR3.
  • the antibody has affinity of at least 5OnM (KD ⁇ 50 nM), to both FGFR2 and FGFR3.
  • the antibody of the invention has an affinity of at least 1OnM to both FGFR2 and FGFR3.
  • the antibody is substantially devoid of affinity to FGFRl .
  • the antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors is selected from a monoclonal antibody, and a fragment of a monoclonal antibody including but not limited to Fab, F(ab') 2 and single chain Fv (scFv).
  • Additional embodiments include chimeric antibodies; human and humanized antibodies; recombinant and engineered antibodies, and fragments thereof.
  • a preferred antibody species is a single chain antibody.
  • Single chain antibodies can be single chain composite polypeptides having antigen binding capabilities and comprising amino acid sequences homologous or analogous to the variable regions of an immunoglobulin light and heavy chain i.e. linked VR-V L or single chain Fv (scFv).
  • the molecule comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors consists of a V H -CDR3 region and a V L -CDR3 region having SEQ ID NO: 1 and SEQ ID NO:2, respectively.
  • the molecule comprising at least the antigen- binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors is a single chain Fv molecule (ScFv) having SEQ ID NO:5, having corresponding polynucleotide sequence SEQ ID NO: 6.
  • the fragment of the antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors is modified.
  • the modification is PEGylation.
  • the present invention is related to a pharmaceutical composition useful for preventing, attenuating or treating a disease or disorder associated with FGFR2 comprising a therapeutically effective amount of a molecule comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors; and a pharmaceutically acceptable carrier.
  • the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder.
  • the cell proliferative disease or disorder is of epithelial origin.
  • cell proliferative diseases or disorders include but are not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
  • the present invention is related to a method of preventing, attenuating or treating a disease or disorder associated with FGFR2, comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of an antibody having affinity to FGFR2 with cross- reactivity to other FGF receptors; and a pharmaceutically acceptable carrier.
  • the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder of epithelial origin selected from bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
  • Another aspect of the present invention relates to the use of a molecule comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 optionally with cross-reactivity to other FGF receptors, for the manufacture of a therapeutic composition for the treatment of a cell proliferative disease or disorder including but not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
  • Yet another aspect of the present invention is related to the use of a pharmaceutical composition comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors for the treatment of a cell proliferative disease or disorder, wherein the cell proliferative disease or disorder is selected from bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
  • treatment of a cell proliferative disease includes treatment of a primary tumor as well as prevention or treatment of metastases.
  • FGFR according to the present invention is preferably human, however other mammalian FGFR proteins are within the scope of the invention.
  • PRO-007 inhibits proliferation of FDCP cells transfected with either FGFR2 or FGFR3, but not of FDCP cells transfected with either FGFRl or FGFR4.
  • PRO-OOl and PRO-007 inhibit proliferation of FDCP cells transfected with either wild-type (ligand-dependent) FGFR3 (WT) or constitutively activated
  • PRO-007 single chain (sc) blocks cell proliferation in 5637 bladder carcinoma cells (A) and in Jon cells (B) but not in SW780 and RT4 cell lines
  • Figure 7 Neutralizing activity of PRO-007 scFv and PEGylated PRO-007 scFv.
  • Figure 8. Expression of FGFRs in bladder carcinoma cell lines. The indicated cell lines were lysed and probed with either anti-FGFRl, anti-FGFR2, anti- FGFR3 or anti-FGFR4.
  • PRO-007 scFv blocks FGF-induced MAPK in RTl 12 cells.
  • PRO-007 inhibits proliferation of Osteosarcoma SAOA2 cells (A).
  • PRO-007 and PRO-OOl inhibit proliferation of SAOA2 cells (B) Figure 11.
  • PRO-007 and PRO-OOl inhibit proliferation of SAOA2 cells (A).
  • PRO-007 and PRO-OOl inhibit proliferation of Osteosarcoma SJSAl cells (B).
  • PRO-008 Fab control antibody
  • PRO-007 In addition to FGFR3 PRO-007 also binds to FGFR2 in the heparin binding domain and blocks heparin binding.
  • Figure 13 PRO-007 induces dose dependent inhibition of ERK phosphorylation in RCJ cells specifically expressing FGFR2 or FGFR3.
  • the present invention provides for the first time antibodies with high affinity to fibroblast growth factor receptor 2 (FGFR2) optionally having cross-reactivity to other fibroblast growth factor receptors (FGFRs); and fragments thereof, which are efficient in blocking both ligand-dependent and constitutive ligand-independent receptor activation.
  • FGFR2 fibroblast growth factor receptor 2
  • FGFRs fibroblast growth factor receptors
  • FGF receptor subtype may be involved in the pathogenesis of many cell proliferative diseases.
  • cross-reactivity of the antibodies to more than one FGFR subtype may be advantageous for clinical treatment.
  • the present invention is related to a composition
  • a composition comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 optionally with cross-reactivity to other FGF receptors, which blocks both ligand-dependent and constitutive ligand-independent receptor activation.
  • the present invention involves a pharmaceutical composition for the prevention, attenuation or treatment of a disease or disorder associated with FGFR2, comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 with cross-reactivity to other FGF receptors; and a pharmaceutically acceptable carrier.
  • the present invention relates to a method for the prevention, attenuation or treatment of cell proliferative diseases comprising administering to a subject in need thereof a therapeutically effective amount of a molecule comprising the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors; and a pharmaceutically acceptable carrier.
  • the present invention relates to a method for the prevention, attenuation or treatment of bladder cancer comprising administering to a subject in need thereof a therapeutically effective amount of a molecule comprising the antigen-binding portion of an isolated antibody having affinity for FGFR2 and/or FGFR3; and a pharmaceutically acceptable carrier.
  • Another aspect relates to the use of a molecule comprising the antigen-binding portion of an isolated antibody having affinity for FGFR2 with cross-reactivity to other
  • FGF receptors for the manufacture of a therapeutic composition for the treatment of cell proliferative diseases.
  • a pharmaceutical composition comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 with cross-reactivity to other FGF receptors, for the treatment of a cell proliferative disease or disorder including but not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
  • the antibody which blocks both ligand- dependent and constitutive (ligand-independent) receptor activation, has binding affinity of at least 5OnM to both FGFR2 and FGFR3.
  • the antibody will have affinity of at least 1OnM for both FGFR2 and FGFR3.
  • the antigen-binding portion of an isolated antibody having affinity for FGFR2 and/or FGFR3 is selected from monoclonal antibodies, a monoclonal antibody fragment or an antibody- fusion protein. The affinity of a given antibody to various receptor subtypes can be easily measured using methods known to one of skill in the art.
  • antibody affinities to receptor subtypes may conveniently be measured using the BIACORE® technology (AB Corporation, Sweden), among others. Using this technology, antibodies of the present invention were found to have affinities of less than 1OnM to both FGFR2 and FGFR3, whereas affinities to FGFRl were undetectable using this method.
  • the antibody of the invention binds to the heparin binding site of at least one or more subtype of FGF receptor. According to another embodiment the antibody of the invention prevents heparin from binding to the heparin binding site of the FGFRs. According to yet another embodiment the antibody of the invention blocks both ligand-dependent and constitutive ligand-independent receptor activation. According to some embodiments the antibody binds to the heparin binding site of a plurality of FGFR subtypes.
  • fibroblast growth factor receptor or "FGFR” denotes a receptor specific for FGF which is necessary for transducing the signal exerted by FGF to the cell interior, typically comprising an extracellular ligand-binding domain, a single transmembrane helix, and a cytoplasmic domain having tyrosine kinase activity.
  • FGFR extracellular domain consists of three immunoglobulin-like (Ig-like) domains (Dl,
  • FGFR2 and D3 a heparin binding domain and an acidic box.
  • Five FGFR genes that encode for multiple receptor protein variants are known.
  • Alternative splicing of the FGFR2 mRNAs generates at least two known isoforms of the receptors, FGFR2IIIb and
  • FGFR2 specific refers to any effector that has higher affinity (K D ⁇ 50 nM) or activity or binding to FGFR2 polypeptide or to the polynucleotide encoding same, than to another FGF receptor protein or polynucleotide.
  • the effector can be any molecule including a ligand, an inhibitor, an antibody, a polypeptide, a polynucleotide or a small organic molecule such as a tyrosine kinase inhibitor. It is to be explicitly understood that the term “FGFR2 specific” does not exclude or preclude situations wherein the effector has some activity on another FGF receptor subtype. It is further to be understood that if the activity mediated via direct or indirect interaction with another receptor subtype is clinically important for the therapeutic utility observed, this is explicitly encompassed within the scope of the claimed invention.
  • affinity refers to the attraction between an antigen and an antibody which induces their binding.
  • antibody which has affinity for fibroblast growth factor receptor 2 (FGFR2) optionally with cross-reactivity to other FGFR2
  • FGF receptors is meant that the antibody possesses high affinity to FGFR2 (KD ⁇ 50 nM), but may also have affinity to other FGFRs.
  • the antibody has an affinity of at least 1OnM for both FGFR2 and FGFR3. In some embodiments the affinity for
  • FGFR2 and FGFR3 is approximately equal. In other embodiments the particular antibody may have a greater affinity for one or the other of these receptors.
  • substantially devoid of affinity to FGFRl is meant that the antibody has low affinity to FGFRl (KD > 100 nM).
  • a non-limiting example for an antibody which has high affinity for FGFR2 with cross-reactivity to other FGF receptors but is substantially devoid of affinity to FGFRl is PRO-007 which has high affinity (KD ⁇ 50 nM) to both FGFR2 and FGFR3 but low affinity to FGFRl .
  • inhibitor and “antagonist” are interchangeable and refer to a molecule, which attenuates, reduces or inhibits the activity or expression of at least one FGFR receptor subtype.
  • ligand-dependent receptor activation refers to activation of the FGF receptor that is dependent on the amount of ligand presented to the receptor.
  • ligand-independent receptor activation or “constitutive receptor activation” refers to activation of the FGF receptor that is independent of ligand presentation.
  • Receptors are usually activated by their corresponding ligand in a dose dependent manner. Certain mutations cause receptors to be constitutively activated independent of their ligand. For example the achondroplastic mutated form of FGFR3 causes ligand independent cell proliferation.
  • antibodies block only ligand-dependent receptor activation but not ligand- independent receptor activation.
  • the antibodies of the present invention are unique in that they block both ligand-dependent and ligand-independent receptor activation.
  • One embodiment of the present invention is directed to molecules comprising an antigen binding domain which blocks both ligand-dependent and ligand-independent activation of FGFR2 and/or FGFR3. 2
  • bladder cancer refers to abnormal and or malignant cell growth in the bladder.
  • Urothelial, or bladder cancer is the fourth most common cancer in the United States.
  • About 90 percent of bladder cancers are transitional cell carcinomas, cancers that begin in the cells lining the bladder (Billerey et al., 2001).
  • cancer that begins in the transitional cells develops into invasive bladder cancer when it spreads through the lining of the bladder and invades the muscular wall of the bladder. Invasive cancer may grow through the bladder wall and spread to nearby organs.
  • the muscle invasive tumors are associated with a high risk of metastases and a poor prognosis.
  • osteosarcoma refers to abnormal and or malignant bone growth.
  • Osteosarcoma (osteogenic sarcoma) is the second most common primary bone tumor and is highly malignant. It is most common in people aged 10 to 20, although it can occur at any age. Osteosarcoma usually develops around the knee or in other long bones, particularly the metaphyses. It can metastasize, usually to lung or bone.
  • breast cancer refers to abnormal and or malignant cell growth in the breast. Most breast cancers are epithelial tumors that develop from cells lining ducts or lobules. Breast cancer invades locally and spreads initially through the regional lymph nodes, bloodstream, or both. Metastatic breast cancer may affect almost any organ in the body most commonly, lungs, liver, bone, brain, and skin. Most skin metastases occur in the region of the breast surgery; scalp metastases also are common.
  • the present invention is related to a method of preventing, attenuating or treating a disease or disorder associated with FGFR2, comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of an antibody specific to FGFR2 with cross-reactivity to other FGF receptors ; and a pharmaceutically acceptable carrier.
  • the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder including but not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
  • Antibodies, or immunoglobulins comprise two heavy chains linked together by disulfide bonds and two light chains, each light chain being linked to a respective heavy chain by disulfide bonds in a "Y" shaped configuration.
  • Proteolytic digestion of an antibody yields Fv (Fragment variable) and Fc (fragment crystalline) domains.
  • the antigen binding domains, Fab include regions where the polypeptide sequence varies.
  • F(ab') 2 represents two Fab 1 arms linked together by disulfide bonds.
  • the central axis of the antibody is termed the Fc fragment.
  • Each heavy chain has at one end a variable domain (V H ) followed by a number of constant domains (C H ).
  • Each light chain has a variable domain (V L ) at one end and a constant domain (CL) at its other end, the light chain variable domain being aligned with the variable domain of the heavy chain and the light chain constant domain being aligned with the first constant domain of the heavy chain (CHl).
  • variable domains of each pair of light and heavy chains form the antigen- binding site.
  • the domains on the light and heavy chains have the same general structure and each domain comprises four framework regions, whose sequences are relatively conserved, joined by three hypervariable domains known as complementarity determining regions (CDRl -3). These domains contribute specificity and affinity of the antigen-binding site.
  • the present invention provides antibodies which block ligand-dependent and constitutive ligand-independent FGF receptor activation, comprising a VH-CDR3 region (complementarity determining region 3 of the heavy chain) having a polypeptide sequence as set forth in SEQ ID NO: 1 and a corresponding
  • VL-CDR3 region (complementarity determining region 3 of the light chain) having a polypeptide sequence as set forth in SEQ ID NO:2.
  • the corresponding polynucleotide sequences of the V H -CDR3 and V L -CDR3 regions are set forth in SEQ ID NO: 3 and
  • CDR complementarity determining region
  • the isotype of the heavy chain determines immunoglobulin class (IgG, IgA, IgD, IgE or IgM, respectively).
  • the light chain is either of two isotypes (kappa, K or lambda, ⁇ ) found in all antibody classes.
  • antibody or “antibodies”
  • this is intended to include intact antibodies, such as polyclonal antibodies or monoclonal antibodies (niAbs), as well as proteolytic fragments thereof such as the Fab or F(ab') 2 fragments.
  • niAbs polyclonal antibodies or monoclonal antibodies
  • proteolytic fragments thereof such as the Fab or F(ab') 2 fragments.
  • chimeric antibodies human and humanized antibodies; recombinant and engineered antibodies, and fragments thereof.
  • the DNA encoding the variable region of the antibody can be inserted into the DNA encoding other antibodies to produce chimeric antibodies (see, for example, US patent 4,816,567). Single chain antibodies fall within the scope of the present invention.
  • single chain variable fragment scFv
  • Single chain antibodies can be single chain composite polypeptides having antigen binding capabilities and comprising amino acid sequences homologous or analogous to the variable regions of an immunoglobulin light and heavy chain (linked V H -VL or single chain Fv (scFv)).
  • VH and VL may copy natural monoclonal antibody sequences or one or both of the chains may comprise a CDR-FR construct of the type described in US patent 5,091,513, the entire contents of which are incorporated herein by reference.
  • the antigen-binding portion of an antibody which has affinity for FGFR2 with cross-reactivity to other FGF receptors comprises a fragment of a monoclonal antibody such as the Fab, F(ab') 2 or a scFv fragments.
  • the antibody fragment is a single chain Fv molecule (scFv) set forth in SEQ ID NO:5, having corresponding polynucleotide sequence SEQ ID NO:6.
  • the respective polypeptide and polynucleotide sequences are presented in Example 1.
  • the amino acid sequence (SEQ ID NO:7) and polynucleotide sequences (SEQ ID NO: 8) of the scFv for PRO-OOl are included herein for reference. Accordingly, the amino acid sequences for VH-CDR3 and VL-CDR3 of
  • PRO-008 (SEQ ID NO:9 and SEQ ID NO: 10, respectively) and the corresponding polynucleotide sequences SEQ ID NO:11 and SEQ ID NO:12 are also included herein only for reference.
  • PEGylation is a process of attaching one or more chains of a fat polymer called polyethylene glycol (PEG) to a protein molecule. This process is intended to lengthen the life time of a substance in the bloodstream (without being metabolized and excreted by the body).
  • PEG polyethylene glycol
  • the term "antibody” also includes modified formats of an antibody or its fragments i.e. PEGylated scFv or an antibody (or an antibody fragment) conjugated to a toxin molecule.
  • a "molecule having the antigen-binding portion of an antibody” as used herein is intended to include not only intact immunoglobulin molecules of any isotype and generated by any animal cell line or microorganism, but also the antigen-binding reactive fraction thereof, including, but not limited to, the Fab fragment, the Fab' fragment, the F(ab') 2 fragment, the variable portion of the heavy and/or light chains thereof, Fab mini- antibodies (see WO 93/15210, US patent application 08/256,790, WO 96/13583, US patent application 08/817,788, WO 96/37621, US patent application 08/999,554, the entire contents of which are incorporated herein by reference), dimeric bispecific miniantibodies (see Muller et al., 1998) and chimeric or single-chain antibodies incorporating such reactive fraction, as well as any other type of molecule or cell in which such antibody reactive fraction has been physically inserted, such as a chimeric T- cell receptor or a T-cell having such a receptor,
  • Such molecules may be provided by any known technique, including, but not limited to, enzymatic cleavage, peptide synthesis or recombinant techniques.
  • the term "Fc" as used herein refers to the constant portion of an immunoglobulin molecule (Fragment crystallizable) that mediates phagocytosis, triggers inflammation and targets Ig to particular tissues; the Fc portion is also important in complement activation.
  • a chimera comprising a fusion of the extracellular domain of the RPTK and an immunoglobulin constant domain can be constructed useful in assaying for ligands of the receptor and in screening for antibodies and fragments thereof.
  • extracellular domain when used herein refers to the polypeptide sequence of the FGFR2 disclosed herein which are normally positioned to the outside of the cell.
  • the extracellular domain encompasses polypeptide sequences in which part of or all of the adjacent (C-terminal) hydrophobic transmembrane and intracellular sequences of the mature FGFR2 have been deleted.
  • the extracellular domain-containing polypeptide can comprise the extracellular domain and a part of the transmembrane domain.
  • the polypeptide comprises only the extracellular domain of the FGFR2.
  • the truncated extracellular domain is generally soluble.
  • the skilled practitioner can readily determine the extracellular and transmembrane domains of the FGFR2 by aligning it with known RPTK (receptor protein tyrosine kinases) amino acid sequences for which these domains have been delineated.
  • the hydrophobic transmembrane domain can be readily delineated based on a hydrophobicity plot of the polypeptide sequence.
  • the extracellular domain is N-terminal to the transmembrane domain.
  • epitope is meant to refer to that portion of any molecule capable of being bound by an antibody or a fragment thereof which can also be recognized by that antibody.
  • Epitopes or antigenic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and have specific three- dimensional structural characteristics as well as specific charge characteristics.
  • an "antigen” is a molecule or a portion of a molecule capable of being bound by an antibody.
  • An antigen may have one or more than one epitope.
  • the specific reaction referred to above is meant to indicate that the antigen will react, in a highly selective manner, with its corresponding antibody and not with the multitude of other antibodies which may be evoked by other antigens.
  • a “neutralizing antibody” as used herein refers to a molecule having an antigen- binding site to a specific receptor capable of reducing or inhibiting (blocking) activity or signaling through a receptor, as determined by in vivo or in vitro assays, as per the specification.
  • a “monoclonal antibody” or “mAb” is a substantially homogeneous population of antibodies to a specific antigen. mAbs may be obtained by methods known to those skilled in the art. See, for example Kohler et al (1984) and US patent 4,376,110 the contents of which are incorporated entirely herein by reference.
  • the mAbs of the present invention may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and any subclass thereof.
  • a hybridoma producing a mAb may be cultivated in vitro or in vivo.
  • High titers of mAbs can be obtained by in-vivo production where cells from the individual hybridomas are injected intraperitoneally into pristine-primed Balb/c mice to produce ascites fluid containing high concentrations of the desired mAbs.
  • mAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.
  • Chimeric antibodies are molecules, the different portions of which are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region.
  • Antibodies which have variable region framework residues substantially from human antibody (termed an acceptor antibody) and complementarity determining regions substantially from a mouse antibody (termed a donor antibody) are also referred to as humanized antibodies.
  • Chimeric antibodies are primarily used to reduce immunogenicity in application and to increase yields in production, for example, where murine mAbs have higher yields from hybridomas but higher immunogenicity in humans, such that human/murine chimeric mAbs are used.
  • antibodies can be generated in vitro using phage display technology.
  • Such a production of recombinant antibodies is much faster compared to conventional antibody production and they can be generated against an enormous number of antigens.
  • many antigens prove to be non-immunogenic or extremely toxic, and therefore cannot be used to generate antibodies in animals.
  • affinity maturation i.e., increasing the affinity and specificity
  • large numbers of different antibodies against a specific antigen can be generated in one selection procedure.
  • To generate recombinant monoclonal antibodies one can use various methods all based on display libraries to generate a large pool of antibodies with different antigen recognition sites.
  • Such a library can be made in several ways: One can generate a synthetic repertoire by cloning synthetic CDR3 regions in a pool of heavy chain germline genes and thus generating a large antibody repertoire, from which recombinant antibody fragments with various specificities can be selected.
  • the present invention provides molecules comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 optionally with cross-reactivity to other FGF receptors .
  • These molecules include antibodies specific to FGFR2 and/or FGFR3, peptide analogs of such antibodies with binding affinity to the extracellular portion of FGFR2 and/or FGFR3, and peptidomimetics based on the structure of such peptides.
  • the peptidomimetics of the invention may be similar in structure to AHNP, the small molecule form of an anti- HER2/neu peptidomimetic that has activity similar to the full monoclonal antibody Herceptin (Zhang et al., 2000). Pharmacology
  • the present invention also contemplates pharmaceutical formulations, both for veterinary and for human medical use, which comprise as the active agent one or more molecules comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 optionally with cross-reactivity to other FGF receptors , for the manufacture of a therapeutic composition for the treatment or prophylaxis of the conditions variously described herein.
  • the active agent is preferably utilized together with one or more pharmaceutically acceptable carrier(s) and optionally any other therapeutic ingredients.
  • the carrier(s) must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the formulation and not unduly deleterious to the recipient thereof.
  • the active agent is provided in an amount effective to achieve the desired pharmacological effect, as described above, and in a quantity appropriate to achieve the desired daily dose.
  • the molecules of the present invention comprising the antigen binding portion of an antibody or comprising another polypeptide including a peptidomimetic will be suspended in a sterile saline solution for therapeutic uses.
  • compositions may alternatively be formulated to control release of active ingredient (molecule comprising the antigen binding portion of an antibody) or to prolong its presence in a patient's system.
  • active ingredient molecule comprising the antigen binding portion of an antibody
  • suitable drug delivery systems include, e.g., implantable drug release systems, hydrogels, hydroxymethylcellulose, microcapsules, liposomes, microemulsions, microspheres, and the like. Controlled release preparations can be prepared through the use of polymers to complex or adsorb the molecule according to the present invention.
  • biocompatible polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a polyanhydride copolymer of a stearic acid dimer and sebaric acid (Sherwood et al, 1992).
  • the rate of release of the molecule according to the present invention, i.e., of an antibody or antibody fragment, from such a matrix depends upon the molecular weight of the molecule, the amount of the molecule within the matrix, and the size of dispersed particles (Saltzman et al., 1989 and Sherwood et al., 1992).
  • composition of this invention may be administered by any suitable means, such as orally, topically, intranasally, subcutaneously, intramuscularly, intravenously, intra-arterially, intraarticulary, intralesionally or parenterally. Ordinarily, intravenous (i.v.), intraarticular, topical or parenteral administration will be preferred.
  • the therapeutically effective amount of the molecule according to the present invention will depend, inter alia upon the administration schedule, the unit dose of molecule administered, whether the molecule is administered in combination with other therapeutic agents, the immune status and health of the patient, the therapeutic activity of the molecule administered and the judgment of the treating physician.
  • a "therapeutically effective amount” refers to the amount of a molecule required to alleviate one or more symptoms associated with a disorder being treated over a period of time.
  • the daily dosage can generally be between about O.Olmg to about 500 mg, preferably about O.Olmg to about 50 mg, more preferably about 0.1 mg to about 10 mg, per kg body weight.
  • the daily dosage can generally be between about O.OOlmg to about 100 mg, preferably about O.OOlmg to about 10 mg, more preferably about O.Olmg to about 1 mg, per kg body weight.
  • the daily dosage can be administered, for example in regimens typical of 1-4 individual administration daily.
  • Other preferred methods of administration include intraarticular administration of about O.Olmg to about 100 mg per kg body weight.
  • Various considerations in arriving at an effective amount are described, e.g., in Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press, 1990; and Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa., 1990.
  • the molecules of the present invention as active ingredients are dissolved, dispersed or admixed in an excipient that is pharmaceutically acceptable and compatible with the active ingredient as is well known.
  • Suitable excipients are, for example, water, saline, phosphate buffered saline (PBS), dextrose, glycerol, ethanol, or the like and combinations thereof.
  • PBS phosphate buffered saline
  • dextrose glycerol
  • ethanol ethanol
  • Other suitable carriers are well known to those skilled in the art.
  • the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents.
  • the combined treatment of one or more of the molecules of the invention with an anti-inflammatory drug such as methotrexate or glucocorticoids may provide a more efficient treatment for inhibiting FGFR2 activity.
  • the pharmaceutical composition comprises the antibody, an anti-inflammatory drug and a pharmaceutically
  • nucleic acid and “polynucleotides” refers to molecules such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • the term should also be understood to include, as equivalents, analogs of either RNA or DNA
  • nucleotide 10 made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
  • nucleic acid molecule encoding an antibody having affinity for FGFR2 optionally with cross-reactivity to other FGF receptors, which block receptor activation.
  • the nucleic acid molecule contains a
  • nucleic acid molecule contains a nucleotide sequence having at least 75% sequence identity, preferably about 90%, and more preferably about 95% identity to the above encoding nucleotide sequence set forth in any one of SEQ ID NO:3, SEQ ID NO:4 or SEQ ID NO:6 as would be well understood by those of skill in the art.
  • the nucleic acid molecule contains a nucleotide sequence having at least
  • the invention also provides nucleic acids that hybridize under high stringency conditions to polynucleotides set forth in any one of SEQ ID NO:3, SEQ ID NO:4 or SEQ ID NO:6. or the complement thereof.
  • highly stringent conditions are used herein, highly stringent conditions
  • Examples of highly stringent (-10 0 C below the calculated Tm (temperature midpoint) of the hybrid) conditions use a wash solution of 0.1 X SSC (standard saline citrate) and 0.5% SDS at the appropriate incubation temperature (Ti) below the calculated Tm of the hybrid.
  • 0.1 X SSC standard saline citrate
  • Ti incubation temperature
  • a common hybridization condition that can be used with the highly stringent to moderately stringent wash conditions described above is hybridization in a solution of 6 X SSC (or 6 X SSPE), 5 X Denhardt's reagent, 0.5% SDS, 100 ⁇ g/ml denatured, fragmented salmon sperm DNA at an appropriate incubation temperature Ti. See generally Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d edition, Cold Spring Harbor Press (1989)) for suitable high stringency conditions.
  • Stringency conditions are a function of the temperature used in the hybridization experiment and washes, the molarity of the monovalent cations in the hybridization solution and in the wash solution(s) and the percentage of formamide in the hybridization solution.
  • sensitivity by hybridization with a probe is affected by the amount and specific activity of the probe, the amount of the target nucleic acid, the detectability of the label, the rate of hybridization, and the duration of the hybridization.
  • the hybridization rate is maximized at a Ti (incubation temperature) of 20-25 0 C below Tm for DNA:DNA hybrids and 10-15 0 C below Tm for DNA:RNA hybrids. It is also maximized by an ionic strength of about 1.5M Na + .
  • the rate is directly proportional to duplex length and inversely proportional to the degree of mismatching.
  • Hybrid stability is a function of duplex length, base composition, ionic strength, mismatching, and destabilizing agents (if any).
  • Tm of a perfect hybrid may be estimated for DNA:DNA hybrids using the equation of Meinkoth et al (1984), as
  • Tm is reduced by 0.5-1.5 0 C (an average of I 0 C can be used for ease of calculation) for each 1% mismatching.
  • the Tm may also be determined experimentally. As increasing length of the hybrid (L) in the above equations increases the Tm and enhances stability, the full-length rat gene sequence can be used as the probe.
  • Filter hybridization is typically carried out at 68°C, and at high ionic strength
  • Tm (e.g., 5 - 6 X SSC), which is non-stringent, and followed by one or more washes of increasing stringency, the last one being of the ultimately desired high stringency.
  • the equations for Tm can be used to estimate the appropriate Ti for the final wash, or the Tm of the perfect duplex can be determined experimentally and Ti then adjusted accordingly.
  • the invention also provides for conservative amino acid variants of the molecules. Variants according to the invention also may be made that conserve the overall molecular structure of the encoded proteins. Given the properties of the individual amino acids comprising the disclosed protein products, some rational substitutions will be recognized by the skilled worker. Amino acid substitutions, i.e. "conservative substitutions,” may be made, for instance, on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
  • Example 1 Generation and sequences of Antibodies Screening for Antibodies
  • the antibodies PRO-OOl (which binds only FGFR3 with high affinity) and PRO-
  • PRO-001 and PRO-008 serve merely as reference to PRO-007 (which binds both FGFR2 and FGFR3 with high affinity).
  • the generation of PRO-001 and PRO-008 was described in WO 2002/102972.
  • Soluble dimeric forms of a FGFR can be constructed and prepared in a number of different ways. For instance, the extracellular domain of a FGFR joined to Fragment crystalline (Fc) and expressed as a fusion polypeptide that 5 dimerizes naturally by means of the Fc portion of the FGFR-Fc fusion.
  • Fragment crystalline Fc
  • PRO-007 single chain (sc) presented hereafter as SEQ ID NO: 5 was generated from PRO-001 sc (SEQ ID NO: 7) by point mutations in the complementarity determining region 3 of the variable domain in the light chain (VL- CDR3). The antibody fragment was then tested for receptor specificity and inhibition of cell proliferation.
  • PRO-007 SINGLE CHAIN SEQ ID NO: 5
  • PRO-OOL SINGLE CHAIN SEQ ID NO: 7
  • AssayMaxiSorp ELISA plates were coated with 100 ⁇ l anti-human Fc (10 ⁇ g/ml) in bicarbonate overnight at 4 0 C. Wells were washed five consecutive times with a PBS solution containing 0.1% Tween 20 (PBST). The well surface was blocked with 250 ⁇ l PBST+3%BSA (blocking solution) for 1 hour at 37 0 C. This was followed by capturing 1 ⁇ g of fragment crystalline (Fc) fusion of the extracellular domain of FGF receptor (FGFR/Fc) for 1 hour at room temperature. To assess the antibody binding to the captured FGFR/Fc, l ⁇ g each of the tested Fabs was incubated in 100 ⁇ l blocking solution per well for 1 hour at room temperature.
  • PBST PBS solution containing 0.1% Tween 20
  • PRO-OOl binds only FGFR3 with high affinity (and FGFR2 with low affinity).
  • PRO-008 binds both FGFRl and FGFR3 with high affinity (fig.lC
  • Example 3 Inhibition of mitogen-activated protein kinase (MAPKinase) by various antibodies mKlotho stable clone 09 HEK293 cells (Human Embryonic Kidney cells) were grown to a final concentration of 200,000cells/well in a 24 well plate and incubated at
  • lysates were transferred to tube and heated for 5 minutes at 95 0 C. Lysate samples were analyzed by 13% sodium dodecyl sulfate — polyacrylamide gel electrophoresis (SDS PAGE).
  • the FDCP cell line is a murine immortalized, interleukin 3 (IL3) dependent cell line of myelocytic bone marrow origin, which does not express endogenous FGF
  • FGFR FGFR Receptors
  • FDCP cells expressing FGFRl, FGFR2, FGFR3 or FGFR4 were grown in "full medium” (Iscove's Medium containing 2ml glutamine, 100 ⁇ g/ml penicillin, lOO ⁇ g/ml streptomycin, 12.5 ⁇ g/ml Nystatin and 10% FBS, Gibco, California, USA) supplemented with 5 ⁇ g/ml heparin and 10ng/ml FGF9. Cells were split every 3 days and kept in culture no more than one month. One day prior to the experiment, the cells were split. Before the experiment, the cells were washed 3 times (1000 rpm, 6 min) with full medium.
  • the cells were resuspended and counted with Trypan Blue (Sigma). Twenty thousand (20,000) cells /well were added to wells in a 96-well plate in 50 ⁇ l in full medium containing 5 ⁇ g/ml heparin. Conditioned medium was added in an additional volume of 50 ⁇ l full medium containing FGF9 at varying concentrations to a final volume of lOO ⁇ l. A primary stock solution of the antibody (containing twice the amount of the highest antibody concentration) was prepared in Iscove's medium containing 5 ⁇ g/ml heparin and 2.5ng/ml FGF or IL-3.
  • the stock solution was filtered through a pre-blocked 0.2 ⁇ m syringe nitrocellulose filter, and serial 2 fold antibody dilutions were prepared. Dilutions were kept on ice until use. 50 ⁇ l of the antibody containing solution was added to each well and the plate was incubated at 37 0 C for either 40 hours or 64 hours.
  • reaction was developed as follows: 100 ⁇ l of activator solution was added to 5 ml XTT reagent and mixed gently. 50 ⁇ l of mixture was added to each well. Optical density (OD) at 490 nm at this point gave the zero time reading.
  • the assay is successful when the O.D. of untreated control growing with saturated amounts of FGF (10 and 20 ng/ml) is at least 1.3 O.D. units. Furthermore, it is noted that the background of wells with no cells should be 0.2-0.35 O.D. units and that the O.D. absorbance of 1.25 ng/ml FGF9 should not be less than 40% of the O.D. absorbance achieved with saturated FGF 9 concentration (10 and 20 ng/ml). Specific inhibition of FGF and FGF receptor mediated proliferation should always be accompanied with lack of any inhibition of the same antibody concentration on IL-3 dependent cell proliferation.
  • FDCP-FRl FDCP cells transfected with the human wild-type FGFRl .
  • FDCP-FR2 FDCP cells transfected with the human wild-type FGFR2.
  • FDCP-FR3 FDCP cells transfected with the human wild-type FGFR3.
  • FDCP-FR4 FDCP cells transfected with the human wild-type FGFR4.
  • the neutralizing activity of the antibodies was measured by the aforementioned cell proliferation analysis in FDCP-FRl, FDCP-FR2, FDCP-FR3 and FDCP-FR4 cell lines.
  • Increasing amounts of the PRO-007 scFv were added to FDCP-FRl (diamond ⁇ ), FDCP-FR2 (square ⁇ ), FDCP-FR3 (closed triangle A) 3 and FDCP-FR4 (X) grown in the presence of FGF9 (Fig.2). Two days later, an XTT proliferation assay was performed.
  • Figure 2 demonstrates that PRO-007 scFv inhibited FDCP-FR2 and FDCP-FR3 cell proliferation, however, it had no effect on FDCP-FRl and FDCP-FR4 cell proliferation.
  • Example 5 Inhibition of ligand-dependent and constitutive ligand-independent receptor activation
  • FDCP cells do not express endogenous FGF Receptors (FGFR).
  • FDCP cells were transfected with either wild type FGFR3 (WT) which has ligand-dependent activation, or with mutant FGFR3 (S249C and E650K) which are activated independently of the ligand (ligand-independent activation).
  • WT wild type FGFR3
  • S249C and E650K mutant FGFR3
  • DSMZ human bladder carcinoma
  • grade II SW780 human transitional cell carcinoma grade I RTl 12
  • DMZ human transitional bladder carcinoma
  • grade II J82 human transitional bladder carcinoma RT-4 (DSMZ) human transitional bladder carcinoma established from a recurrent well-differentiated transitional papillary tumor of the urinary bladder (clinical stage T2, histological grade Gl.
  • Example 7 Anti-FGFR2 antibody inhibits proliferation in human bladder cancer cell lines
  • DSMZ Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH - German Collection of Microorganisms and Cell Cultures.
  • the cells were seeded at 1000 or 2000 cells/well in a 96 well plate and cultured with 10%FCS or with FGFl/heparin. RTl 12 cells were then exposed to either of the antibodies PRO-007 scFv or PRO-OOl scFv. In contrast to the slight inhibition observed with PRO-OOl, PRO-007 scFv completely blocked RTl 12 cell proliferation at 150 ⁇ g/ml (Fig. 4). When PRO-OOl scFv was added to the cells at the time of cell seeding, there was still no effect on the RTl 12 proliferation rate (not shown).
  • PRO-007 scFv to the J82 bladder carcinoma cell line that harbors a FGFR3 K650E activated receptor.
  • PRO-007 had no effect on J82 cell proliferation demonstrating the specificity of its action on RTl 12 cells (Fig. 5B).
  • SW780, 5637, RT4 and Jon TCC cell lines were seeded at a concentration of about 2000 cells/well in a 96 well plate.
  • PRO-007 scFv or a control scFv were added at increasing concentrations.
  • cell proliferation was measured by XTT showing a dose dependent proliferation arrest of 5637 and Jon but not of SW780 and RT4 cell lines (Fig. 6A-D).
  • RTl 12 cells were incubated with PEGylated or non-PEGylated PRO-007 scFv.
  • the inhibitory effect of the PEGylated PRO-007 scFv was demonstrated to match that of the non-modified single chain (Fig. 7). Relative expression of FGFRs in bladder carcinoma cell lines
  • Serum starved RTl 12 cells were pre-incubated 20 minutes with 150 ⁇ g/ml PRO- 007 scFv and then stimulated 5 minutes with 100ng/ml FGFl. Pre-incubation with PRO- 007 scFv completely blocked ERKl and 2 activation by FGFl as judged by Western with anti-phospho MAPK antibodies (Fig. 9).
  • Example 8 inhibition of Osteosarcoma cell proliferation
  • SA0A2 and SJSA are two Osteosarcoma cell lines.
  • Osteosarcoma SAOA2 cells were cultured in Dulbecco Modified Eagle's Minimal Essential Medium (DMEM- Gibco, California, USA)+10% FCS and SJSA cells were cultured in Roswell Park Memorial Institute medium (RPMI Gibco, California, USA) +10% FCS.
  • DMEM- Gibco, California, USA Dulbecco Modified Eagle's Minimal Essential Medium
  • SJSA cells were cultured in Roswell Park Memorial Institute medium (RPMI Gibco, California, USA) +10% FCS.
  • Cells were seeded in a concentration of 2000 cells per well in a 96 well plate and incubated with the antibodies for 4 days. Antibody containing medium was replaced once. Cell proliferation was measured using the XTT assay).
  • FIG. 10A Relative to untreated cells, treatment with single chain Fv of PRO-007 significantly inhibited cell proliferation in SAOA2 cell line (fig. 10A).
  • Figure 1OB demonstrates that PRO-007 scFv, PRO-001 scFv and PRO-001 Fab all inhibit cell proliferation in SASO2 cells.
  • Figure HA again shows a marked inhibition of SASO2 cell proliferation by PRO-007 scFv and an even more pronounced inhibition by PRO- 001 scFv.
  • SJSAl cells there was significant inhibition of cell proliferation with a stronger inhibition by PRO-001 scFv (fig. 1 IB).
  • PRO-007, PRO-001 and PRO-008 were examined for interference with the interaction between FGFR2 or FGFR3 and heparin.
  • Heparin binding to the fragment crystalline fusion of the extracellular domain of FGFR3 (FGFR3/Fc) or FGFR2 (FGFR2/Fc) was measured in the presence of PRO-007, PRO-001 and PRO-008 antibodies.
  • Heparin Sepharose anchored FGFR3/Fc or FGFR2/Fc were incubated with the indicated antibodies.
  • Bound (B) and unbound (U) fractions were then analyzed using Coomassie-stained SDS-PAGE. As shown in figure 12, FGFR3 was located entirely in the unbound fraction in the presence of either PRO-001 or PRO-007.
  • Example 10 inhibition of ERK phosphorylation by PRO-007 in RCJ cells RCJ cell assay RCJ cells (fetal rat calvaria-derived mesenchymal cells, RCJ 3.1C5.18;
  • RCJ cells that stably express either FGFRl, FGFR2 or FGFR3 were stimulated for 5 minutes with 20ng/ml of either FGF2 (stimulating FGFRl and FGFR2, ProChon Biotech Ltd., Rehovot, Israel) or FGF9 (stimulating FGFR3, ProChon Biotech Ltd., Rehovot, Israel).
  • Parallel cultures were pre- incubated for 20 minutes with either 12.5 ⁇ g/ml, 25 ⁇ g/ml or 50 ⁇ g/ml of PRO-007.
  • Example 11 Examining PRO-007 in additional bladder cancer cell lines
  • 97-7 bladder cancer cell lines are also examined for the effect of PRO-007.
  • MGHU3 and the 97-7 cells are seeded at a concentration of about 2000 cells/well in a 96 well plate.
  • PRO-007sc or a control scFv are added at increasing concentrations. Five days latter, cell proliferation is measured by XTT.
  • Example 12 Examining PRO-007 in bladder cancer cells from human patients
  • the aim in this study is to test the effects of neutralizing anti-FGFR antibodies on primary cultures of human bladder cancer and to compare them to normal bladder cells.
  • HBSS Hanks' balanced salt solution
  • PBS phosphate-buffered saline
  • Urothelial sheets are washed twice in HBSS and digested to a single-cell suspension with 200 U/ml of collagenase type IV for 10 min at 37 0 C.
  • Cells are washed in keratinocyte serum-free medium containing 30 ng/ml cholera toxin and 100 ⁇ g/ml of penicillin/streptomycin solution (KSFM).
  • KSFM penicillin/streptomycin solution
  • Cells are resuspended in fresh KSFM and placed in 25 cm2 culture flasks at a density of 4 ⁇ lO4/cm2 and incubated at 37°C in 5% CO2.
  • Cells are passaged by incubating with 0.02% EDTA at 37°C for 10 min, followed by trypsin-EDTA (Sigma) for a further 60 s. Cells are disaggregated by gentle agitation, and then serum containing medium is added to the flask to neutralize residual trypsin.
  • Cell viability is assessed by XTT dye absorbance.
  • Cells are seeded in 96-well plates at a density of 3,000-10,000 cells per well in culture medium. Cells are incubated in the absence or presence of increasing concentrations of the PRO-007 antibody. Plates are incubated for 48 or 72 hours at 37 0 C, 5% CO2. XTT assays are performed according to the manufacturer's instruction.
  • FGFRs Fibroblast growth factor receptors

Abstract

The present invention is related to antibodies with binding affinity to fibroblast growth factor receptor 2 (FGFR2) optionally with binding affinity to other FGF receptors, which block both ligand-dependent and constitutive ligand independent receptor activation. Specifically, the present invention relates to antibodies with high affinity to more than one FGF receptor subtype, and fragments thereof, useful in treating disorders including cell proliferative diseases.

Description

ANTIBODIES BLOCKING FIBROBLAST GROWTH FACTOR RECEPTOR ACTIVATION AND METHODS OF USE THEREOF
FIELD OF THE INVENTION The present invention is related in general to antibodies with binding affinity to fibroblast growth factor receptors (FGFRs) which block both ligand-dependent and constitutive ligand independent receptor activation. In particular, the present invention relates to antibodies specific for more than one receptor subtype, and fragments thereof, useful for treating diseases and disorders, including cell proliferative diseases.
BACKGROUND OF THE INVENTION
Fibroblast Growth Factor Receptors
Fibroblast Growth Factor ligands (FGFs) constitute a family of over twenty structurally related polypeptides that are developmentally regulated and expressed in a wide variety of tissues. FGFs stimulate proliferation, cell migration and differentiation and play a major role in skeletal and limb development, wound healing, tissue repair, hematopoiesis, angiogenesis, and tumorigenesis (reviewed in Ornitz and Itoh, 2001).
The biological action of FGFs is mediated by specific cell surface receptors belonging to the receptor protein tyrosine kinase (RPTK) family of protein kinases. These proteins consist of an extracellular ligand binding domain, a single transmembrane domain and an intracellular tyrosine kinase domain that undergoes phosphorylation upon binding of FGF. The FGF receptor (FGFR) extracellular region contains three immunoglobulin-like (Ig-like) domains (Dl, D2 and D3), an acidic box, and a heparin- binding domain. Five FGFR genes encoding for multiple receptor variants have been identified to date. Alternative splicing further increases the diversity of the FGFR family. The second half of the third Ig-like domain in FGFRl, 2 and 3 is encoded by one of two exons, HIb or IHc, generating receptors with different ligand affinities and specificities.
Fibroblast Growth Factor Receptors and Malignancy
Certain FGFRs have been implicated in certain malignancies and proliferative diseases. FGFR3 is the most frequently mutated oncogene in transitional cell carcinoma (TCC) of the bladder where it is mutated in more than 30% of the cases (Cappellen
1999). Yee et al. (2000) identified a mutation in FGFR3 linked to cervical carcinoma. van Rhijn et al. (2002) disclosed FGFR3 mutations in bladder cancer which were previously identified in skeletal disorders.
FGFR3 mutations seem to have a central role in the early development of papillary bladder tumors and can serve as a target for treatment (Billerey et al., 2001; Cappellen et al., 1999). These tumors follow a common molecular pathway, which is different from tumors with concomitant carcinoma in situ (CIS). However, FGFR3 mutations do not seem to play a role in bladder cancer progression (Zieger, et al., 2005).
FGFR2 mRNAs were found to be overexpressed in both human pancreatic cancer cells and the adjacent pancreatic parenchyma (Ishiwata et al., 1998,) and Kurban et al., (2004) identified FGFR2 IHb (KGFR) expression in cervical cancer cells. Lorenzi et al. (1996) have identifies a constitutively active form of FGFR2 in rat osteosarcoma cells.
In general, FGFR2 exhibits expression of the IHb isoform in epithelial type tissues and the uroepithelium and the IHc isoform in the mesenchyme. FGFR2 subtype
IHb (FGFR2-IIIb) was shown to have tumor suppressive properties, i.e. be downregulated in a subset of transitional cell carcinomas of the bladder (Bernard-Pierrot et al., 2004; Ricol et al. 1999).
Johnston et al. (1995) reported that FGFR4 and FGFR2 are expressed at higher levels in breast cancer cell lines than in normal epithelial cells. Khnykin D et al. (2006) found that in the majority of cases FGFR2-FGFR4, but not FGFRl, were expressed by malignant cells. FGFR4 was shown to be associated with pituitary tumors (Ezzat, et al,
2002) and breast cancer progression (Bange, et al., 2002).
Recent findings implicate that single nucleotide polymorphisms (SNPs) in FGFR2 were highly associated with breast cancer (Hunter et al., 2007). An additional study demonstrated that SNPs in five novel independent loci including FGFR2 exhibited strong and consistent evidence of association with breast cancer (Easton et al., 2007).
These and other findings implicate the involvement of both FGFR2 and FGFR3 in the pathogenesis of various malignancies rendering these FGF receptors potential targets for therapeutic intervention in these cell proliferative diseases.
Fibroblast Growth Factor Receptor inhibitors International Patent Publication WO 03/024987 discloses antisense compounds useful for modulating FGFR2. International Patent Publication WO 03/ 023004 discloses antisense compounds useful for modulating FGFR3. US Patent No. 6,900,053 teaches compositions comprising antisense oligo-nucleotides and methods for modulating the expression of FGFR2. Those compositions were found useful in the treatment of diseases associated with overexpression of FGFR2. Small molecule tyrosine kinase inhibitors, which have been shown to inhibit the activity of certain tyrosine kinase receptors, are disclosed in US Patent Nos. 6,987,113; 6,683,082 and others.
International Patent Publication WO 02/102972, co-assigned to the assignee of the present invention and incorporated by reference herein, discloses antibodies to receptor protein tyrosine kinases, specifically anti-fibroblast growth factor receptor 3 (FGFR3) antibodies. Certain antibodies shown to be specific for FGFR3 neutralize FGFR3 activity and are potentially useful for treating skeletal dysplasias such as achondroplasia and proliferative diseases such as multiple myeloma. That disclosure notes bladder cancer in a list of proliferative diseases in which FGFR3 is known to be involved but does not teach the method of treating or attenuating bladder cancer using an anti-FGFR2 antibody.
PCT Publication WO 2006/048877, co-assigned to the assignee of the present invention teaches a method of treating multiple myeloma comprising administering to an individual in need thereof an anti-FGFR3 antibody which is specific for wild type FGFR3. PCT Publication WO 2004/110487 assigned to the assignee of the present invention, provides a method of treating a T cell mediated disease comprising administering to a subject in need thereof an FGFR3 antagonist.
International Patent Publication WO 2005/066211 teaches antibodies directed to
FGFR polypeptides, and methods of use thereof for treating tumors. That application provides tables disclosing human tumors, which express the various FGFR proteins but there is no teaching of cross reactive antibodies. Martinez-Torrecuadrada et al. (2005) teach anti-FGFR3 antibodies which inhibit bladder carcinoma cell proliferation.
Kan et al. (1993) disclose that heparin interacts with a specific region in the extracellular domain of the FGFR, and is essential for FGFR activation. They synthesized a peptide corresponding to the heparin binding domain of FGFRl and raised polyclonal antibodies specific to this peptide. Both the peptide and the antibodies were antagonistic to FGFl -stimulated cell growth. However, Kan et al. did not demonstrate blocking of other FGF receptors nor do they show inhibition of constitutive receptor activation.
Fortin et al. (2005) used antibodies specific to a single receptor subtype, namely either FGFRl, FGFR2 or FGFR3 to examine FGF/FGFR interactions during oligodendrocyte development. In this article there is no teaching of an antibody that binds both FGFR2 and FGFR3 with high affinity.
Nowhere in the art is it suggested that anti-FGFR antibodies cross-reactive with more than one receptor subtype would be particularly useful for treating neoplasms. Nowhere in the prior art was it taught or suggested that antibodies capable of blocking heparin binding to the heparin binding site of an FGF receptor could exhibit cross- reactivity to multiple receptor subtypes. Thus, there is an unmet need for anti-FGFR antibodies capable of blocking both ligand-dependent and aberrant constitutive ligand- independent FGF receptor activation, thereby modulating various biological abnormalities.
SUMMARY OF THE INVENTION
The present invention provides antibodies having high specificity to fibroblast growth factors of more than one subtype. The present invention further provides antibodies having high affinity to fibroblast growth factor receptor 2 (FGFR2) having cross-reactivity to other fibroblast growth factor receptors (FGFRs) useful in blocking both ligand-dependent and constitutive ligand-independent receptor activation. It is now disclosed that antibodies cross-reactive to FGFR2 and/or FGFR3 are useful in the prevention, attenuation or treatment of cell proliferative diseases of epithelial origin including but not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma. Without wishing to be bound by any particular theory or mechanism of action, these antibodies may block receptor activation by blocking either FGF binding or heparin binding or both.
In one aspect the present invention is related to molecules comprising at least the antigen-binding portion of an antibody having affinity for fibroblast growth factor receptor 2 (FGFR2) optionally with cross-reactivity to other FGF receptors, which block both ligand-dependent and constitutive ligand-independent receptor activation. According to certain embodiments of the present invention, the antibody which blocks activation of fibroblast growth factor receptors (FGFR) has high affinity to both FGFR2 and FGFR3. According to particular embodiments the antibody has affinity of at least 5OnM (KD < 50 nM), to both FGFR2 and FGFR3. Preferably the antibody of the invention has an affinity of at least 1OnM to both FGFR2 and FGFR3. According to some embodiments the antibody is substantially devoid of affinity to FGFRl .
According to some embodiments of the present invention, the antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors is selected from a monoclonal antibody, and a fragment of a monoclonal antibody including but not limited to Fab, F(ab')2 and single chain Fv (scFv). Additional embodiments include chimeric antibodies; human and humanized antibodies; recombinant and engineered antibodies, and fragments thereof. A preferred antibody species is a single chain antibody. Single chain antibodies can be single chain composite polypeptides having antigen binding capabilities and comprising amino acid sequences homologous or analogous to the variable regions of an immunoglobulin light and heavy chain i.e. linked VR-VL or single chain Fv (scFv).
In one embodiment, the molecule comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors, consists of a VH-CDR3 region and a VL-CDR3 region having SEQ ID NO: 1 and SEQ ID NO:2, respectively. In another embodiment the molecule comprising at least the antigen- binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors, is a single chain Fv molecule (ScFv) having SEQ ID NO:5, having corresponding polynucleotide sequence SEQ ID NO: 6.
According to some embodiments of the present invention the fragment of the antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors is modified. According to other embodiments the modification is PEGylation.
In another aspect the present invention is related to a pharmaceutical composition useful for preventing, attenuating or treating a disease or disorder associated with FGFR2 comprising a therapeutically effective amount of a molecule comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors; and a pharmaceutically acceptable carrier. According to certain embodiments the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder. According to other embodiments the cell proliferative disease or disorder is of epithelial origin. According to additional embodiments cell proliferative diseases or disorders include but are not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
In yet another aspect the present invention is related to a method of preventing, attenuating or treating a disease or disorder associated with FGFR2, comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of an antibody having affinity to FGFR2 with cross- reactivity to other FGF receptors; and a pharmaceutically acceptable carrier. According to some embodiments the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder of epithelial origin selected from bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma. Another aspect of the present invention relates to the use of a molecule comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 optionally with cross-reactivity to other FGF receptors, for the manufacture of a therapeutic composition for the treatment of a cell proliferative disease or disorder including but not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
Yet another aspect of the present invention is related to the use of a pharmaceutical composition comprising at least the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors for the treatment of a cell proliferative disease or disorder, wherein the cell proliferative disease or disorder is selected from bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
It is to be understood that treatment of a cell proliferative disease includes treatment of a primary tumor as well as prevention or treatment of metastases.
The FGFR according to the present invention is preferably human, however other mammalian FGFR proteins are within the scope of the invention. These and other aspects of the present invention will be apparent from the figures, description, and claims that follow. BRIEF DESCRIPTION OF THE FIGURES
Figure 1. Binding specificity of three antibodies to FGF receptors. A) PRO-007 binds
FGFR2 with high affinity; B) Both PRO-007 and PRO-OOl bind FGFR3 with high affinity; C) PRO-OOl binds FGFR3 with high affinity but FGFR2 with low affinity; D) PRO-008 binds both FGFRl and FGFR3 with high affinity. Figure 2. PRO-007 inhibits proliferation of FDCP cells transfected with either FGFR2 or FGFR3, but not of FDCP cells transfected with either FGFRl or FGFR4. Figure 3. PRO-OOl and PRO-007 inhibit proliferation of FDCP cells transfected with either wild-type (ligand-dependent) FGFR3 (WT) or constitutively activated
FGFR3 mutant (S249C), but not of FDCP cells transfected with the intracellular mutant (E650K). Figure 4. Proliferation of bladder carcinoma RTl 12 cells was completely blocked upon treatment with PRO-007 scFv (antibody with affinity to both FGFR2 and FGFR3) but only partially inhibited by treatment with PRO-OOl Fab or scFv (an anti-FGFR3 specific antibody). Figure 5. PRO -007 specifically blocks proliferation of bladder carcinoma RTl 12 cells
(A), but not of bladder carcinoma J82 cells (B).
Figure 6. PRO-007 single chain (sc) blocks cell proliferation in 5637 bladder carcinoma cells (A) and in Jon cells (B) but not in SW780 and RT4 cell lines
(C and D5 respectively).
Figure 7. Neutralizing activity of PRO-007 scFv and PEGylated PRO-007 scFv. Figure 8. Expression of FGFRs in bladder carcinoma cell lines. The indicated cell lines were lysed and probed with either anti-FGFRl, anti-FGFR2, anti- FGFR3 or anti-FGFR4.
Figure 9. PRO-007 scFv blocks FGF-induced MAPK in RTl 12 cells.
Figure 10. PRO-007 inhibits proliferation of Osteosarcoma SAOA2 cells (A). Both
PRO-007 and PRO-OOl inhibit proliferation of SAOA2 cells (B) Figure 11. PRO-007 and PRO-OOl inhibit proliferation of SAOA2 cells (A). PRO-007 and PRO-OOl inhibit proliferation of Osteosarcoma SJSAl cells (B).
Figure 12. FGFR3 appears only in the heparin Sepharose unbound fraction (U) when either PRO-001 or PRO-007 are present, indicating that these antibodies compete with heparin; FGFR3 remains heparin bound (B) in the presence of 000732
control antibody PRO-008 Fab. In addition to FGFR3 PRO-007 also binds to FGFR2 in the heparin binding domain and blocks heparin binding. Figure 13. PRO-007 induces dose dependent inhibition of ERK phosphorylation in RCJ cells specifically expressing FGFR2 or FGFR3.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides for the first time antibodies with high affinity to fibroblast growth factor receptor 2 (FGFR2) optionally having cross-reactivity to other fibroblast growth factor receptors (FGFRs); and fragments thereof, which are efficient in blocking both ligand-dependent and constitutive ligand-independent receptor activation. These antibodies were found highly effective in blocking cell proliferation of abnormal cell types including bladder cancer and osteosarcoma cell lines.
Various indications support the notion that more than one FGF receptor subtype may be involved in the pathogenesis of many cell proliferative diseases. Without wishing to be bound by theory, cross-reactivity of the antibodies to more than one FGFR subtype may be advantageous for clinical treatment.
According to one aspect the present invention is related to a composition comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 optionally with cross-reactivity to other FGF receptors, which blocks both ligand-dependent and constitutive ligand-independent receptor activation. According to another aspect the present invention involves a pharmaceutical composition for the prevention, attenuation or treatment of a disease or disorder associated with FGFR2, comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 with cross-reactivity to other FGF receptors; and a pharmaceutically acceptable carrier.
In one aspect the present invention relates to a method for the prevention, attenuation or treatment of cell proliferative diseases comprising administering to a subject in need thereof a therapeutically effective amount of a molecule comprising the antigen-binding portion of an antibody having affinity for FGFR2 with cross-reactivity to other FGF receptors; and a pharmaceutically acceptable carrier. In a non-limiting example, the present invention relates to a method for the prevention, attenuation or treatment of bladder cancer comprising administering to a subject in need thereof a therapeutically effective amount of a molecule comprising the antigen-binding portion of an isolated antibody having affinity for FGFR2 and/or FGFR3; and a pharmaceutically acceptable carrier.
Another aspect relates to the use of a molecule comprising the antigen-binding portion of an isolated antibody having affinity for FGFR2 with cross-reactivity to other
FGF receptors, for the manufacture of a therapeutic composition for the treatment of cell proliferative diseases. Yet another aspect concerns the use of a pharmaceutical composition comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 with cross-reactivity to other FGF receptors, for the treatment of a cell proliferative disease or disorder including but not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
According to certain embodiments, the antibody, which blocks both ligand- dependent and constitutive (ligand-independent) receptor activation, has binding affinity of at least 5OnM to both FGFR2 and FGFR3. Preferably the antibody will have affinity of at least 1OnM for both FGFR2 and FGFR3. According to other embodiments, the antigen-binding portion of an isolated antibody having affinity for FGFR2 and/or FGFR3 is selected from monoclonal antibodies, a monoclonal antibody fragment or an antibody- fusion protein. The affinity of a given antibody to various receptor subtypes can be easily measured using methods known to one of skill in the art. By way of example, antibody affinities to receptor subtypes may conveniently be measured using the BIACORE® technology (AB Corporation, Sweden), among others. Using this technology, antibodies of the present invention were found to have affinities of less than 1OnM to both FGFR2 and FGFR3, whereas affinities to FGFRl were undetectable using this method.
According to one embodiment the antibody of the invention binds to the heparin binding site of at least one or more subtype of FGF receptor. According to another embodiment the antibody of the invention prevents heparin from binding to the heparin binding site of the FGFRs. According to yet another embodiment the antibody of the invention blocks both ligand-dependent and constitutive ligand-independent receptor activation. According to some embodiments the antibody binds to the heparin binding site of a plurality of FGFR subtypes.
PCT publication WO 02/102972, co-assigned to some of the assignees of the present invention, discloses monoclonal antibodies to receptor protein tyrosine kinases, including specific anti-Fibroblast Growth Factor Receptor antibodies, the contents of which are incorporated in their entirety. A soluble dimeric form of the extracellular domain of the FGFR3 receptor was utilized to screen for antibodies (e.g., Fabs) from a phage display antibody library. This screening yielded numerous high affinity antibodies (Fabs, KD < 50 nM) that bind FGFR3 and interfere with ligand binding, thereby blocking ligand-dependent activation of FGFR3. Certain antibodies were shown to be specific for FGFR3 and useful to neutralize FGFR3 activity and for the treatment of skeletal dysplasias such as achondroplasia and proliferative diseases such as bladder cancer and multiple myeloma. Definitions
For convenience certain terms employed in the specification, examples and claims are described herein.
Fibroblast growth factor receptors
The term "fibroblast growth factor receptor" or "FGFR" denotes a receptor specific for FGF which is necessary for transducing the signal exerted by FGF to the cell interior, typically comprising an extracellular ligand-binding domain, a single transmembrane helix, and a cytoplasmic domain having tyrosine kinase activity. The
FGFR extracellular domain consists of three immunoglobulin-like (Ig-like) domains (Dl,
D2 and D3), a heparin binding domain and an acidic box. Five FGFR genes that encode for multiple receptor protein variants are known. Alternative splicing of the FGFR2 mRNAs generates at least two known isoforms of the receptors, FGFR2IIIb and
FGFR2IIIC.
Throughout the specification and the claims that follow, the term "FGFR2 specific" refers to any effector that has higher affinity (KD < 50 nM) or activity or binding to FGFR2 polypeptide or to the polynucleotide encoding same, than to another FGF receptor protein or polynucleotide. The effector can be any molecule including a ligand, an inhibitor, an antibody, a polypeptide, a polynucleotide or a small organic molecule such as a tyrosine kinase inhibitor. It is to be explicitly understood that the term "FGFR2 specific" does not exclude or preclude situations wherein the effector has some activity on another FGF receptor subtype. It is further to be understood that if the activity mediated via direct or indirect interaction with another receptor subtype is clinically important for the therapeutic utility observed, this is explicitly encompassed within the scope of the claimed invention.
The term "affinity" refers to the attraction between an antigen and an antibody which induces their binding. As used herein, by the term "antibody which has affinity for fibroblast growth factor receptor 2 (FGFR2) optionally with cross-reactivity to other
FGF receptors" is meant that the antibody possesses high affinity to FGFR2 (KD < 50 nM), but may also have affinity to other FGFRs. Preferably the antibody has an affinity of at least 1OnM for both FGFR2 and FGFR3. In some embodiments the affinity for
FGFR2 and FGFR3 is approximately equal. In other embodiments the particular antibody may have a greater affinity for one or the other of these receptors.
By the term "substantially devoid of affinity to FGFRl" is meant that the antibody has low affinity to FGFRl (KD > 100 nM). A non-limiting example for an antibody which has high affinity for FGFR2 with cross-reactivity to other FGF receptors but is substantially devoid of affinity to FGFRl is PRO-007 which has high affinity (KD < 50 nM) to both FGFR2 and FGFR3 but low affinity to FGFRl .
As used herein, the terms "inhibitor" and "antagonist" are interchangeable and refer to a molecule, which attenuates, reduces or inhibits the activity or expression of at least one FGFR receptor subtype.
As used herein the term "ligand-dependent receptor activation" refers to activation of the FGF receptor that is dependent on the amount of ligand presented to the receptor. The term "ligand-independent receptor activation" or "constitutive receptor activation" refers to activation of the FGF receptor that is independent of ligand presentation. Receptors are usually activated by their corresponding ligand in a dose dependent manner. Certain mutations cause receptors to be constitutively activated independent of their ligand. For example the achondroplastic mutated form of FGFR3 causes ligand independent cell proliferation.
Many antibodies block only ligand-dependent receptor activation but not ligand- independent receptor activation. The antibodies of the present invention are unique in that they block both ligand-dependent and ligand-independent receptor activation. One embodiment of the present invention is directed to molecules comprising an antigen binding domain which blocks both ligand-dependent and ligand-independent activation of FGFR2 and/or FGFR3. 2
As used herein "bladder cancer" or "urothelial cancer" refers to abnormal and or malignant cell growth in the bladder. Urothelial, or bladder cancer is the fourth most common cancer in the United States. About 90 percent of bladder cancers are transitional cell carcinomas, cancers that begin in the cells lining the bladder (Billerey et al., 2001). In some cases, cancer that begins in the transitional cells develops into invasive bladder cancer when it spreads through the lining of the bladder and invades the muscular wall of the bladder. Invasive cancer may grow through the bladder wall and spread to nearby organs. The muscle invasive tumors are associated with a high risk of metastases and a poor prognosis. As used herein "osteosarcoma" refers to abnormal and or malignant bone growth.
Osteosarcoma (osteogenic sarcoma) is the second most common primary bone tumor and is highly malignant. It is most common in people aged 10 to 20, although it can occur at any age. Osteosarcoma usually develops around the knee or in other long bones, particularly the metaphyses. It can metastasize, usually to lung or bone. As used herein "breast cancer" refers to abnormal and or malignant cell growth in the breast. Most breast cancers are epithelial tumors that develop from cells lining ducts or lobules. Breast cancer invades locally and spreads initially through the regional lymph nodes, bloodstream, or both. Metastatic breast cancer may affect almost any organ in the body most commonly, lungs, liver, bone, brain, and skin. Most skin metastases occur in the region of the breast surgery; scalp metastases also are common.
In the US, cumulative risk of developing breast cancer is 12% (1 in 8) by age 95, and risk of dying of it is about 4%. Family history of breast cancer in a lst-degree relative (mother, sister or daughter) doubles or triples risk of developing the cancer. About 5% of women with breast cancer carry a mutation in one of the 2 known breast cancer genes, BRCAl or BRCA2. Men who carry a BRCA2 mutation also have an increased risk of developing breast cancer. Recent studies have demonstrated association between breast cancer and single nucleotide polymorphisms (SNPs) in FGFR2 (Hunter et al., 2007).
According to one aspect the present invention is related to a method of preventing, attenuating or treating a disease or disorder associated with FGFR2, comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of an antibody specific to FGFR2 with cross-reactivity to other FGF receptors ; and a pharmaceutically acceptable carrier. According to some embodiments the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder including but not limited to bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma. Antibodies
Antibodies, or immunoglobulins, comprise two heavy chains linked together by disulfide bonds and two light chains, each light chain being linked to a respective heavy chain by disulfide bonds in a "Y" shaped configuration. Proteolytic digestion of an antibody yields Fv (Fragment variable) and Fc (fragment crystalline) domains. The antigen binding domains, Fab, include regions where the polypeptide sequence varies. The term F(ab')2 represents two Fab1 arms linked together by disulfide bonds. The central axis of the antibody is termed the Fc fragment. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains (CH). Each light chain has a variable domain (VL) at one end and a constant domain (CL) at its other end, the light chain variable domain being aligned with the variable domain of the heavy chain and the light chain constant domain being aligned with the first constant domain of the heavy chain (CHl).
The variable domains of each pair of light and heavy chains form the antigen- binding site. The domains on the light and heavy chains have the same general structure and each domain comprises four framework regions, whose sequences are relatively conserved, joined by three hypervariable domains known as complementarity determining regions (CDRl -3). These domains contribute specificity and affinity of the antigen-binding site.
In certain embodiments the present invention provides antibodies which block ligand-dependent and constitutive ligand-independent FGF receptor activation, comprising a VH-CDR3 region (complementarity determining region 3 of the heavy chain) having a polypeptide sequence as set forth in SEQ ID NO: 1 and a corresponding
VL-CDR3 region (complementarity determining region 3 of the light chain) having a polypeptide sequence as set forth in SEQ ID NO:2. The corresponding polynucleotide sequences of the VH-CDR3 and VL-CDR3 regions are set forth in SEQ ID NO: 3 and
SEQ ID NO: 4, respectively. The amino acid and nucleotide sequences are presented in
Example 1. Additionally, complementarity determining region (CDR) grafting may be performed to alter certain properties of the antibody molecule including affinity or specificity. A non-limiting example of CDR grafting is disclosed in US patent 5,225,539.
The isotype of the heavy chain (gamma, alpha, delta, epsilon or mu) determines immunoglobulin class (IgG, IgA, IgD, IgE or IgM, respectively). The light chain is either of two isotypes (kappa, K or lambda, λ) found in all antibody classes.
It should be understood that when the terms "antibody" or "antibodies" are used, this is intended to include intact antibodies, such as polyclonal antibodies or monoclonal antibodies (niAbs), as well as proteolytic fragments thereof such as the Fab or F(ab')2 fragments. Further included within the scope of the invention are chimeric antibodies; human and humanized antibodies; recombinant and engineered antibodies, and fragments thereof. Furthermore, the DNA encoding the variable region of the antibody can be inserted into the DNA encoding other antibodies to produce chimeric antibodies (see, for example, US patent 4,816,567). Single chain antibodies fall within the scope of the present invention.
By the term "single chain variable fragment (scFv)" is meant a fusion of the variable regions of the heavy and light chains of immunoglobulin, linked together with a short (usually serine, glycine) linker. Single chain antibodies can be single chain composite polypeptides having antigen binding capabilities and comprising amino acid sequences homologous or analogous to the variable regions of an immunoglobulin light and heavy chain (linked VH-VL or single chain Fv (scFv)). Both VH and VL may copy natural monoclonal antibody sequences or one or both of the chains may comprise a CDR-FR construct of the type described in US patent 5,091,513, the entire contents of which are incorporated herein by reference. The separate polypeptides analogous to the variable regions of the light and heavy chains are held together by a polypeptide linker. Methods of production of such single chain antibodies, particularly where the DNA encoding the polypeptide structures of the VH and VL chains are known, may be accomplished in accordance with the methods described, for example, in US patents 4,946,778, 5,091,513 and 5,096,815, the entire contents of each of which are incorporated herein by reference.
According to certain embodiments of the present invention, the antigen-binding portion of an antibody which has affinity for FGFR2 with cross-reactivity to other FGF receptors comprises a fragment of a monoclonal antibody such as the Fab, F(ab')2 or a scFv fragments.
According to other embodiments of the present invention, the antibody fragment is a single chain Fv molecule (scFv) set forth in SEQ ID NO:5, having corresponding polynucleotide sequence SEQ ID NO:6. The respective polypeptide and polynucleotide sequences are presented in Example 1. The amino acid sequence (SEQ ID NO:7) and polynucleotide sequences (SEQ ID NO: 8) of the scFv for PRO-OOl are included herein for reference. Accordingly, the amino acid sequences for VH-CDR3 and VL-CDR3 of
PRO-008 (SEQ ID NO:9 and SEQ ID NO: 10, respectively) and the corresponding polynucleotide sequences SEQ ID NO:11 and SEQ ID NO:12 are also included herein only for reference.
PEGylation is a process of attaching one or more chains of a fat polymer called polyethylene glycol (PEG) to a protein molecule. This process is intended to lengthen the life time of a substance in the bloodstream (without being metabolized and excreted by the body). The term "antibody" also includes modified formats of an antibody or its fragments i.e. PEGylated scFv or an antibody (or an antibody fragment) conjugated to a toxin molecule.
A "molecule having the antigen-binding portion of an antibody" as used herein is intended to include not only intact immunoglobulin molecules of any isotype and generated by any animal cell line or microorganism, but also the antigen-binding reactive fraction thereof, including, but not limited to, the Fab fragment, the Fab' fragment, the F(ab')2 fragment, the variable portion of the heavy and/or light chains thereof, Fab mini- antibodies (see WO 93/15210, US patent application 08/256,790, WO 96/13583, US patent application 08/817,788, WO 96/37621, US patent application 08/999,554, the entire contents of which are incorporated herein by reference), dimeric bispecific miniantibodies (see Muller et al., 1998) and chimeric or single-chain antibodies incorporating such reactive fraction, as well as any other type of molecule or cell in which such antibody reactive fraction has been physically inserted, such as a chimeric T- cell receptor or a T-cell having such a receptor, or molecules developed to deliver therapeutic moieties by means of a portion of the molecule containing such a reactive fraction. Such molecules may be provided by any known technique, including, but not limited to, enzymatic cleavage, peptide synthesis or recombinant techniques. The term "Fc" as used herein refers to the constant portion of an immunoglobulin molecule (Fragment crystallizable) that mediates phagocytosis, triggers inflammation and targets Ig to particular tissues; the Fc portion is also important in complement activation. In one embodiment of the invention, a chimera comprising a fusion of the extracellular domain of the RPTK and an immunoglobulin constant domain can be constructed useful in assaying for ligands of the receptor and in screening for antibodies and fragments thereof.
The "extracellular domain" when used herein refers to the polypeptide sequence of the FGFR2 disclosed herein which are normally positioned to the outside of the cell.
The extracellular domain encompasses polypeptide sequences in which part of or all of the adjacent (C-terminal) hydrophobic transmembrane and intracellular sequences of the mature FGFR2 have been deleted. Thus, the extracellular domain-containing polypeptide can comprise the extracellular domain and a part of the transmembrane domain. According to some embodiments, the polypeptide comprises only the extracellular domain of the FGFR2. The truncated extracellular domain is generally soluble. The skilled practitioner can readily determine the extracellular and transmembrane domains of the FGFR2 by aligning it with known RPTK (receptor protein tyrosine kinases) amino acid sequences for which these domains have been delineated. Alternatively, the hydrophobic transmembrane domain can be readily delineated based on a hydrophobicity plot of the polypeptide sequence. The extracellular domain is N-terminal to the transmembrane domain.
The term "epitope" is meant to refer to that portion of any molecule capable of being bound by an antibody or a fragment thereof which can also be recognized by that antibody. Epitopes or antigenic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and have specific three- dimensional structural characteristics as well as specific charge characteristics.
An "antigen" is a molecule or a portion of a molecule capable of being bound by an antibody. An antigen may have one or more than one epitope. The specific reaction referred to above is meant to indicate that the antigen will react, in a highly selective manner, with its corresponding antibody and not with the multitude of other antibodies which may be evoked by other antigens. A "neutralizing antibody" as used herein refers to a molecule having an antigen- binding site to a specific receptor capable of reducing or inhibiting (blocking) activity or signaling through a receptor, as determined by in vivo or in vitro assays, as per the specification. A "monoclonal antibody" or "mAb" is a substantially homogeneous population of antibodies to a specific antigen. mAbs may be obtained by methods known to those skilled in the art. See, for example Kohler et al (1984) and US patent 4,376,110 the contents of which are incorporated entirely herein by reference. The mAbs of the present invention may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and any subclass thereof. A hybridoma producing a mAb may be cultivated in vitro or in vivo. High titers of mAbs can be obtained by in-vivo production where cells from the individual hybridomas are injected intraperitoneally into pristine-primed Balb/c mice to produce ascites fluid containing high concentrations of the desired mAbs. mAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.
Chimeric antibodies are molecules, the different portions of which are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region. Antibodies which have variable region framework residues substantially from human antibody (termed an acceptor antibody) and complementarity determining regions substantially from a mouse antibody (termed a donor antibody) are also referred to as humanized antibodies. Chimeric antibodies are primarily used to reduce immunogenicity in application and to increase yields in production, for example, where murine mAbs have higher yields from hybridomas but higher immunogenicity in humans, such that human/murine chimeric mAbs are used. Chimeric antibodies and methods for their production are known in the art (Better et al, 1988; Cabilly et al, 1984; European Patent Applications 125023, 171496, 173494, 184187, 173494, PCT patent applications WO 86/01533, WO 97/02671, WO 90/07861, WO 92/22653 and US patents 5,693,762, 5,693,761, 5,585,089, 5,530,101 and 5,225,539). These references are hereby incorporated by reference. 007/000732
Besides the conventional method of raising antibodies in vivo, antibodies can be generated in vitro using phage display technology. Such a production of recombinant antibodies is much faster compared to conventional antibody production and they can be generated against an enormous number of antigens. Furthermore, when using the conventional method, many antigens prove to be non-immunogenic or extremely toxic, and therefore cannot be used to generate antibodies in animals. Moreover, affinity maturation (i.e., increasing the affinity and specificity) of recombinant antibodies is very simple and relatively fast. Finally, large numbers of different antibodies against a specific antigen can be generated in one selection procedure. To generate recombinant monoclonal antibodies one can use various methods all based on display libraries to generate a large pool of antibodies with different antigen recognition sites. Such a library can be made in several ways: One can generate a synthetic repertoire by cloning synthetic CDR3 regions in a pool of heavy chain germline genes and thus generating a large antibody repertoire, from which recombinant antibody fragments with various specificities can be selected. One can use the lymphocyte pool of humans as starting material for the construction of an antibody library. It is possible to construct naive repertoires of human IgM antibodies and thus create a human library of large diversity. This method has been widely used successfully to select a large number of antibodies against different antigens. Protocols for bacteriophage library construction and selection of recombinant antibodies are provided in the well-known reference text Current Protocols in Immunology, Colligan et al (Eds.), John Wiley & Sons, Inc. (1992-2000), Chapter 17, Section 17.1.
In yet another embodiment, the present invention provides molecules comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 optionally with cross-reactivity to other FGF receptors . These molecules include antibodies specific to FGFR2 and/or FGFR3, peptide analogs of such antibodies with binding affinity to the extracellular portion of FGFR2 and/or FGFR3, and peptidomimetics based on the structure of such peptides. The peptidomimetics of the invention may be similar in structure to AHNP, the small molecule form of an anti- HER2/neu peptidomimetic that has activity similar to the full monoclonal antibody Herceptin (Zhang et al., 2000). Pharmacology
The present invention also contemplates pharmaceutical formulations, both for veterinary and for human medical use, which comprise as the active agent one or more molecules comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 optionally with cross-reactivity to other FGF receptors , for the manufacture of a therapeutic composition for the treatment or prophylaxis of the conditions variously described herein.
In such pharmaceutical and medicament formulations, the active agent is preferably utilized together with one or more pharmaceutically acceptable carrier(s) and optionally any other therapeutic ingredients. The carrier(s) must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the formulation and not unduly deleterious to the recipient thereof. The active agent is provided in an amount effective to achieve the desired pharmacological effect, as described above, and in a quantity appropriate to achieve the desired daily dose. Typically, the molecules of the present invention comprising the antigen binding portion of an antibody or comprising another polypeptide including a peptidomimetic will be suspended in a sterile saline solution for therapeutic uses. The pharmaceutical compositions may alternatively be formulated to control release of active ingredient (molecule comprising the antigen binding portion of an antibody) or to prolong its presence in a patient's system. Numerous suitable drug delivery systems are known and include, e.g., implantable drug release systems, hydrogels, hydroxymethylcellulose, microcapsules, liposomes, microemulsions, microspheres, and the like. Controlled release preparations can be prepared through the use of polymers to complex or adsorb the molecule according to the present invention. For example, biocompatible polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a polyanhydride copolymer of a stearic acid dimer and sebaric acid (Sherwood et al, 1992). The rate of release of the molecule according to the present invention, i.e., of an antibody or antibody fragment, from such a matrix depends upon the molecular weight of the molecule, the amount of the molecule within the matrix, and the size of dispersed particles (Saltzman et al., 1989 and Sherwood et al., 1992).
The pharmaceutical composition of this invention may be administered by any suitable means, such as orally, topically, intranasally, subcutaneously, intramuscularly, intravenously, intra-arterially, intraarticulary, intralesionally or parenterally. Ordinarily, intravenous (i.v.), intraarticular, topical or parenteral administration will be preferred.
It will be apparent to those of ordinary skill in the art that the therapeutically effective amount of the molecule according to the present invention will depend, inter alia upon the administration schedule, the unit dose of molecule administered, whether the molecule is administered in combination with other therapeutic agents, the immune status and health of the patient, the therapeutic activity of the molecule administered and the judgment of the treating physician. As used herein, a "therapeutically effective amount" refers to the amount of a molecule required to alleviate one or more symptoms associated with a disorder being treated over a period of time.
Although an appropriate dosage of a molecule of the invention varies depending on the administration route, type of molecule (polypeptide, polynucleotide, organic molecule etc.) age, body weight, sex, or conditions of the patient, and should be determined by the physician in the end, in the case of oral administration, the daily dosage can generally be between about O.Olmg to about 500 mg, preferably about O.Olmg to about 50 mg, more preferably about 0.1 mg to about 10 mg, per kg body weight. In the case of parenteral administration, the daily dosage can generally be between about O.OOlmg to about 100 mg, preferably about O.OOlmg to about 10 mg, more preferably about O.Olmg to about 1 mg, per kg body weight. The daily dosage can be administered, for example in regimens typical of 1-4 individual administration daily. Other preferred methods of administration include intraarticular administration of about O.Olmg to about 100 mg per kg body weight. Various considerations in arriving at an effective amount are described, e.g., in Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press, 1990; and Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa., 1990.
The molecules of the present invention as active ingredients are dissolved, dispersed or admixed in an excipient that is pharmaceutically acceptable and compatible with the active ingredient as is well known. Suitable excipients are, for example, water, saline, phosphate buffered saline (PBS), dextrose, glycerol, ethanol, or the like and combinations thereof. Other suitable carriers are well known to those skilled in the art. In addition, if desired, the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents. The combined treatment of one or more of the molecules of the invention with an anti-inflammatory drug such as methotrexate or glucocorticoids may provide a more efficient treatment for inhibiting FGFR2 activity. In one embodiment, the pharmaceutical composition comprises the antibody, an anti-inflammatory drug and a pharmaceutically
5 acceptable carrier.
Polynucleotides
The term "nucleic acid" and "polynucleotides" refers to molecules such as deoxyribonucleic acid (DNA), and, where appropriate, ribonucleic acid (RNA). The term should also be understood to include, as equivalents, analogs of either RNA or DNA
10 made from nucleotide analogs, and, as applicable to the embodiment being described, single (sense or antisense) and double-stranded polynucleotides.
Within the scope of the present invention is a nucleic acid molecule encoding an antibody having affinity for FGFR2 optionally with cross-reactivity to other FGF receptors, which block receptor activation. The nucleic acid molecule contains a
15 nucleotide sequence having at least 75% sequence identity, preferably about 90%, and more preferably about 95% identity to the above encoding nucleotide sequence set forth in any one of SEQ ID NO:3, SEQ ID NO:4 or SEQ ID NO:6 as would be well understood by those of skill in the art. In the hypervariable regions of the heavy chain and light chain, the nucleic acid molecule contains a nucleotide sequence having at least
20 50% sequence identity, preferably about 70% and more preferably about 80% identity to the molecule set forth in SEQ ID NO:6.
The invention also provides nucleic acids that hybridize under high stringency conditions to polynucleotides set forth in any one of SEQ ID NO:3, SEQ ID NO:4 or SEQ ID NO:6. or the complement thereof. As used herein, highly stringent conditions
25 are those which are tolerant of up to about 5%-25% sequence divergence, preferably about 5%-15%. Without limitation, examples of highly stringent (-100C below the calculated Tm (temperature midpoint) of the hybrid) conditions use a wash solution of 0.1 X SSC (standard saline citrate) and 0.5% SDS at the appropriate incubation temperature (Ti) below the calculated Tm of the hybrid. The ultimate stringency of the
30 conditions is primarily due to the washing conditions, particularly if the hybridization conditions used are those which allow less stable hybrids to form along with stable hybrids. The wash conditions at higher stringency then remove the less stable hybrids. A common hybridization condition that can be used with the highly stringent to moderately stringent wash conditions described above is hybridization in a solution of 6 X SSC (or 6 X SSPE), 5 X Denhardt's reagent, 0.5% SDS, 100 μg/ml denatured, fragmented salmon sperm DNA at an appropriate incubation temperature Ti. See generally Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d edition, Cold Spring Harbor Press (1989)) for suitable high stringency conditions.
Stringency conditions are a function of the temperature used in the hybridization experiment and washes, the molarity of the monovalent cations in the hybridization solution and in the wash solution(s) and the percentage of formamide in the hybridization solution. In general, sensitivity by hybridization with a probe is affected by the amount and specific activity of the probe, the amount of the target nucleic acid, the detectability of the label, the rate of hybridization, and the duration of the hybridization. The hybridization rate is maximized at a Ti (incubation temperature) of 20-250C below Tm for DNA:DNA hybrids and 10-150C below Tm for DNA:RNA hybrids. It is also maximized by an ionic strength of about 1.5M Na+. The rate is directly proportional to duplex length and inversely proportional to the degree of mismatching.
Specificity in hybridization, however, is a function of the difference in stability between the desired hybrid and "background" hybrids. Hybrid stability is a function of duplex length, base composition, ionic strength, mismatching, and destabilizing agents (if any).
The Tm of a perfect hybrid may be estimated for DNA:DNA hybrids using the equation of Meinkoth et al (1984), as
Tm = 81.50C + 16.6 (log M) + 0.41 (%GC) - 0.61 (% form) - 500/L and for DNA:RNA hybrids, as Tm = 79.8°C + 18.5 (log M) + 0.58 (%GC) - 11.8 (%GC)2 - 0.56(% form) - 820/L where M, molarity of monovalent cations, 0.01 -0.4 M NaCl,
%GC, percentage of G and C nucleotides in DNA, 30%-75%, % form, percentage formamide in hybridization solution, and L, length hybrid in base pairs. Tm is reduced by 0.5-1.50C (an average of I0C can be used for ease of calculation) for each 1% mismatching. The Tm may also be determined experimentally. As increasing length of the hybrid (L) in the above equations increases the Tm and enhances stability, the full-length rat gene sequence can be used as the probe.
Filter hybridization is typically carried out at 68°C, and at high ionic strength
(e.g., 5 - 6 X SSC), which is non-stringent, and followed by one or more washes of increasing stringency, the last one being of the ultimately desired high stringency. The equations for Tm can be used to estimate the appropriate Ti for the final wash, or the Tm of the perfect duplex can be determined experimentally and Ti then adjusted accordingly.
The invention also provides for conservative amino acid variants of the molecules. Variants according to the invention also may be made that conserve the overall molecular structure of the encoded proteins. Given the properties of the individual amino acids comprising the disclosed protein products, some rational substitutions will be recognized by the skilled worker. Amino acid substitutions, i.e. "conservative substitutions," may be made, for instance, on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
Having now fully described this invention, it will be appreciated by those skilled in the art that the same can be performed within a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation. The principles of the invention may be better understood with reference to the non-limiting examples below.
EXAMPLES
Example 1: Generation and sequences of Antibodies Screening for Antibodies The antibodies PRO-OOl (which binds only FGFR3 with high affinity) and PRO-
008 (which binds both FGFRl and FGFR3 with high affinity) serve merely as reference to PRO-007 (which binds both FGFR2 and FGFR3 with high affinity). The generation of PRO-001 and PRO-008 was described in WO 2002/102972. Fabs from the Human Combinatorial Antibody Library (HuCAL®) developed at MorphoSys (Munich, Germany), were screened for affinity to a soluble dimeric form of the extracellular domain of the FGFR3 receptor (screening strategies are disclosed in WO 97/08320, US patent 6,300,064, and Knappik et al., (2000), the entire contents of which are incorporated herein by reference). Soluble dimeric forms of a FGFR can be constructed and prepared in a number of different ways. For instance, the extracellular domain of a FGFR joined to Fragment crystalline (Fc) and expressed as a fusion polypeptide that 5 dimerizes naturally by means of the Fc portion of the FGFR-Fc fusion.
PRO-007 single chain (sc) presented hereafter as SEQ ID NO: 5 was generated from PRO-001 sc (SEQ ID NO: 7) by point mutations in the complementarity determining region 3 of the variable domain in the light chain (VL- CDR3). The antibody fragment was then tested for receptor specificity and inhibition of cell proliferation.
10 Additional antibodies generated from PRO-007 by point mutations also demonstrated high binding affinity to both FGFR2 and FGFR3. For example antibody IBl, the VH of which is presented hereafter in SEQ ID NO: 13 (generated by three point mutations in the VH of PRO-007), binds both FGFR2 and FGFR3 with high affinity similar to PRO-007. CDRs are bold and underlined, mutations are designated with larger
15 letters, the X symbol represents a stop codon.
Antigen binding sequences of the antibodies
Figure imgf000025_0001
PRO-007 SINGLE CHAIN (SEQ ID NO: 5)
MVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPGRGLEWLGRTYYRSKW 0 YNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCARSYYPDFDYWGQGTLVTV SSAGGGSGGGGSGGGGSGGGGSDIELTQPPSVSVAPGQTARISCSGDALGDKYASWYQQ KPGQAPVLVIYDDSDRPSGIPERFSGSNSGNTATLTISGTQAEDEADYYCQSYASQGIH YVFGGGTKLTVLGQ Polynucleotide sequence encoding PRO-007 SINGLE CHAIN (SEQ ID NO: 6)
ATGGTGCAATTGCAACAGTCTGGTCCGGGCCTGGTGAAACCGAGCCAAACCCTGAGCCT GACCTGTGCGATTTCCGGAGATAGCGTGAGCAGCAACAGCGCGGCGTGGAACTGGATTC GCCAGTCTCCTGGGCGTGGCCTCGAGTGGCTGGGCCGTACCTATTATCGTAGCAAATGG TATAACGATTATGCGGTGAGCGTGAAAAGCCGGATTACCATCAACCCGGATACTTCGAA AAACCAGTTTAGCCTGCAACTGAACAGCGTGACCCCGGAAGATACGGCCGTGTATTATT GCGCGCGTTCTTATTATCCTGATTTTGATTATTGGGGCCAAGGCACCCTGGTGACGGTT AGCTCAGCGGGTGGCGGTTCTGGCGGCGGTGGGAGCGGTGGCGGTGGTTCTGGCGGTGG TGGTTCCGATATCGAACTGACCCAGCCGCCTTCAGTGAGCGTTGCACCAGGTCAGACCG CGCGTATCTCGTGTAGCGGCGATGCGCTGGGCGATAAATACGCGAGCTGGTACCAGCAG AAACCCGGGCAGGCGCCAGTTCTGGTGATTTATGATGATTCTGACCGTCCCTCAGGCAT CCCGGAACGCTTTAGCGGATCCAACAGCGGCAACACCGCGACCCTGACCATTAGCGGCA CTCAGGCGGAAGACGAAGCGGATTATTATTGCCAGTCTTATGCTTCTCAGGGTATTCAT TATGTGTTTGGCGGCGGCACGAAGTTAACCGTTCTTGGCCAGTGA
PRO-OOL SINGLE CHAIN (SEQ ID NO: 7)
MVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPGRGLEWLGRTYYRSKW YNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCARSYYPDFDYWGQGTLVTV SSAGGGSGGGGSGGGGSGGGGSDIELTQPPSVSVAPGQTARISCSGDALGDKYASWYQQ KPGQAPVLVIYDDSDRPSGIPERFSGSNSGNTATLTISGTQAEDEADYYCQSYDGPDLW VFGGGTKLTVLGQ
Polynucleotide sequence encoding PRO-001 SINGLE CHAIN (SEQ ID NO: 8)
ATGGTGCAATTGCAACAGTCTGGTCCGGGCCTGGTGAAACCGAGCCAAACCCTGAGCCT GACCTGTGCGATTTCCGGAGATAGCGTGAGCAGCAACAGCGCGGCGTGGAACTGGATTC GCCAGTCTCCTGGGCGTGGCCTCGAGTGGCTGGGCCGTACCTATTATCGTAGCAAATGG TATAACGATTATGCGGTGAGCGTGAAAAGCCGGATTACCATCAACCCGGATACTTCGAA AAACCAGTTTAGCCTGCAACTGAACAGCGTGACCCCGGAAGATACGGCCGTGTATTATT GCGCGCGTTCTTATTATCCTGATTTTGATTATTGGGGCCAAGGCACCCTGGTGACGGTT AGCTCAGCGGGTGGCGGTTCTGGCGGCGGTGGGAGCGGTGGCGGTGGTTCTGGCGGTGG TGGTTCCGATATCGAACTGACCCAGCCGCCTTCAGTGAGCGTTGCACCAGGTCAGACCG CGCGTATCTCGTGTAGCGGCGATGCGCTGGGCGATAAATACGCGAGCTGGTACCAGCAG AAACCCGGGCAGGCGCCAGTTCTGGTGATTTATGATGATTCTGACCGTCCCTCAGGCAT CCCGGAACGCTTTAGCGGATCCAACAGCGGCAACACCGCGACCCTGACCATTAGCGGCA CTCAGGCGGAAGACGAAGCGGATTATTATTGCCAGAGCTATGACGGTCCTGATCTTTGG GTGTTTGGCGGCGGCACGAAGTTAACCGTTCTTGGCCAGTGA
Figure imgf000027_0001
PRO-IBl VH (SEQ ID NQ: 13)
VQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAΆWSWIRQSPGRGLEWLGRTYYRSKWY
NDYAVSVKSRi T INPDTSKNQFSLQLNGVTPEDTAVYYCARSYYPDFDXWGQGTLVTV SS
Example 2 : Antibody - Receptor Binding
AssayMaxiSorp ELISA plates were coated with 100 μl anti-human Fc (10 μg/ml) in bicarbonate overnight at 40C. Wells were washed five consecutive times with a PBS solution containing 0.1% Tween 20 (PBST). The well surface was blocked with 250 μl PBST+3%BSA (blocking solution) for 1 hour at 370C. This was followed by capturing 1 μg of fragment crystalline (Fc) fusion of the extracellular domain of FGF receptor (FGFR/Fc) for 1 hour at room temperature. To assess the antibody binding to the captured FGFR/Fc, lμg each of the tested Fabs was incubated in 100 μl blocking solution per well for 1 hour at room temperature. Wells were then washed 5 times with PBST. Reaction was initiated with the addition of 100 μl of 0.8μg/ml goat anti-human Fab-HRP in blocking solution, subsequently washed and detected with TMB substrate (Pierce). The absorbance was measured at 450 nm. Fortin et al. (2005) teach that the antibody PRO-007 binds only FGFR2 with high affinity, however, figures IA and IB demonstrate that PRO-007 binds both FGFR2 and
FGFR3 with high affinity. PRO-OOl binds only FGFR3 with high affinity (and FGFR2 with low affinity). PRO-008 binds both FGFRl and FGFR3 with high affinity (fig.lC
5 and D).
Example 3: Inhibition of mitogen-activated protein kinase (MAPKinase) by various antibodies mKlotho stable clone 09 HEK293 cells (Human Embryonic Kidney cells) were grown to a final concentration of 200,000cells/well in a 24 well plate and incubated at
10 370C 5% CO2 until they attached to the plate. The following day, cells were starved in Dulbecco Modified Eagle's Minimal Essential Medium (DMEM- Gibco-California, USA) for 4 hours and then incubated with 0.5 ml DMEM containing 20μg/ml of either one of the antibodies PRO-001, PRO-007 and PRO-008 (the antigen binding sequences of which are presented in Example 1) or PRO-000 (a control antibody which lacks the
15 VL-CDR3) for 20 minutes at 370C 5% CO2. FGF23 (50 μl of a solution of 10 μg/ml ProChon Biotech Ltd., Rehovot, Israel) was then added to each well to reach a final concentration of 10ng/ml, and the reaction was incubated for 5 additional minutes at 370C 5% CO2. Then, the reaction was stopped by draining all the media and lysing the cells with 200 μl 2X Loading buffer (Tris ICN Glycine SDS BioLab, Jerusalem, Israel).
20 The lysates were transferred to tube and heated for 5 minutes at 950C. Lysate samples were analyzed by 13% sodium dodecyl sulfate — polyacrylamide gel electrophoresis (SDS PAGE).
Table 1: Antibod induced inhibition of MAPKinase Phos hor lation:
Figure imgf000028_0001
Example 4: Inhibition of Cell Proliferation using PRO-007 FDCP cell line
The FDCP cell line is a murine immortalized, interleukin 3 (IL3) dependent cell line of myelocytic bone marrow origin, which does not express endogenous FGF
Receptors (FGFR). Upon transfection with FGFR cDNA and stable expression of the encoded receptor, the FDCP cell line exhibits an FGF dose dependent proliferative response that can replace the dependence on IL3. FDCP cell lines, expressing FGFRs can therefore be used to screen for specific inhibitors or activators of FGFR, as well as for analyzing FGFR signaling.
FDCP Cell Proliferation Assay
FDCP cells expressing FGFRl, FGFR2, FGFR3 or FGFR4 were grown in "full medium" (Iscove's Medium containing 2ml glutamine, 100 μg/ml penicillin, lOOμg/ml streptomycin, 12.5 μg/ml Nystatin and 10% FBS, Gibco, California, USA) supplemented with 5μg/ml heparin and 10ng/ml FGF9. Cells were split every 3 days and kept in culture no more than one month. One day prior to the experiment, the cells were split. Before the experiment, the cells were washed 3 times (1000 rpm, 6 min) with full medium. Later, the cells were resuspended and counted with Trypan Blue (Sigma). Twenty thousand (20,000) cells /well were added to wells in a 96-well plate in 50μl in full medium containing 5 μg/ml heparin. Conditioned medium was added in an additional volume of 50μl full medium containing FGF9 at varying concentrations to a final volume of lOOμl. A primary stock solution of the antibody (containing twice the amount of the highest antibody concentration) was prepared in Iscove's medium containing 5 μg/ml heparin and 2.5ng/ml FGF or IL-3. The stock solution was filtered through a pre-blocked 0.2 μm syringe nitrocellulose filter, and serial 2 fold antibody dilutions were prepared. Dilutions were kept on ice until use. 50 μl of the antibody containing solution was added to each well and the plate was incubated at 370C for either 40 hours or 64 hours.
After incubation, the reaction was developed as follows: 100 μl of activator solution was added to 5 ml XTT reagent and mixed gently. 50 μl of mixture was added to each well. Optical density (OD) at 490 nm at this point gave the zero time reading.
Cells were then incubated at 37°C for 4 hours (in the case of 40 hours incubation) or 2 hours (for the 64 hours incubation) and proliferation was measured by O.D. at 490 nm (A490).
It is noted that the assay is successful when the O.D. of untreated control growing with saturated amounts of FGF (10 and 20 ng/ml) is at least 1.3 O.D. units. Furthermore, it is noted that the background of wells with no cells should be 0.2-0.35 O.D. units and that the O.D. absorbance of 1.25 ng/ml FGF9 should not be less than 40% of the O.D. absorbance achieved with saturated FGF 9 concentration (10 and 20 ng/ml). Specific inhibition of FGF and FGF receptor mediated proliferation should always be accompanied with lack of any inhibition of the same antibody concentration on IL-3 dependent cell proliferation.
The following FDCP cell lines were used:
FDCP-FRl : FDCP cells transfected with the human wild-type FGFRl . FDCP-FR2: FDCP cells transfected with the human wild-type FGFR2. FDCP-FR3: FDCP cells transfected with the human wild-type FGFR3. FDCP-FR4: FDCP cells transfected with the human wild-type FGFR4.
Neutralizing activity of PRO-007
The neutralizing activity of the antibodies was measured by the aforementioned cell proliferation analysis in FDCP-FRl, FDCP-FR2, FDCP-FR3 and FDCP-FR4 cell lines. Increasing amounts of the PRO-007 scFv were added to FDCP-FRl (diamond ♦), FDCP-FR2 (square ■), FDCP-FR3 (closed triangle A)3 and FDCP-FR4 (X) grown in the presence of FGF9 (Fig.2). Two days later, an XTT proliferation assay was performed. Figure 2 demonstrates that PRO-007 scFv inhibited FDCP-FR2 and FDCP-FR3 cell proliferation, however, it had no effect on FDCP-FRl and FDCP-FR4 cell proliferation.
Example 5: Inhibition of ligand-dependent and constitutive ligand-independent receptor activation
FDCP cell lines expressing wild type and mutant FGFR3
As previously described, FDCP cells do not express endogenous FGF Receptors (FGFR). FDCP cells were transfected with either wild type FGFR3 (WT) which has ligand-dependent activation, or with mutant FGFR3 (S249C and E650K) which are activated independently of the ligand (ligand-independent activation). Neutralizing activity of PRO-007 vs. PRO-OOl
Cell proliferation assay was performed as previously described in example 4. In FDCP cells transfected with wild type FGFR3 (WT), both PRO-OOl and PRO-007 markedly inhibited cell proliferation, however, PRO-007 had a stronger effect. In FDCP cells transfected with constitutively activated FGFR3 mutant (S249C), again both PRO- 001 and PRO-007 significantly inhibited cell proliferation, with PRO-007 having a stronger effect. However, in FDCP cells transfected with a different constitutively activated FGFR3 mutant (E650K), PRO-OOl and PRO-007 had no effect on cell proliferation (fϊg.3) Example 6: Bladder carcinoma cell lines
5637 (DSMZ) human bladder carcinoma; grade II SW780 (DSMZ) human transitional cell carcinoma grade I RTl 12 (DSMZ) human transitional bladder carcinoma, grade II J82 (ATCC) human transitional bladder carcinoma RT-4 (DSMZ) human transitional bladder carcinoma established from a recurrent well-differentiated transitional papillary tumor of the urinary bladder (clinical stage T2, histological grade Gl.
Example 7: Anti-FGFR2 antibody inhibits proliferation in human bladder cancer cell lines The bladder carcinoma RTl 12 cell line, which expresses the wild-type receptor, was obtained from the DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH - German Collection of Microorganisms and Cell Cultures). The ability of PRO-OOl antibody (with high affinity only to FGFR3), and PRO-007 antibody (with high affinity to both FGFR2 and FGFR3) to inhibit RTl 12 cell proliferation was tested.
The cells were seeded at 1000 or 2000 cells/well in a 96 well plate and cultured with 10%FCS or with FGFl/heparin. RTl 12 cells were then exposed to either of the antibodies PRO-007 scFv or PRO-OOl scFv. In contrast to the slight inhibition observed with PRO-OOl, PRO-007 scFv completely blocked RTl 12 cell proliferation at 150 μg/ml (Fig. 4). When PRO-OOl scFv was added to the cells at the time of cell seeding, there was still no effect on the RTl 12 proliferation rate (not shown).
To determine the effective dose of PRO-007 scFv, increasing levels of the single chain were added to RT12 cells. A clear dose response on cell proliferation was observed with minimal effect at 9 μg/ml and complete block at 150 μg/ml (Fig. 5A).
To show the specificity of the observed effect, we added PRO-007 scFv to the J82 bladder carcinoma cell line that harbors a FGFR3K650E activated receptor. PRO-007 had no effect on J82 cell proliferation demonstrating the specificity of its action on RTl 12 cells (Fig. 5B). PRO-007 sc activity in other bladder carcinoma cell lines
The SW780, 5637, RT4 and Jon TCC cell lines were seeded at a concentration of about 2000 cells/well in a 96 well plate. The next day, PRO-007 scFv or a control scFv were added at increasing concentrations. Five days later, cell proliferation was measured by XTT showing a dose dependent proliferation arrest of 5637 and Jon but not of SW780 and RT4 cell lines (Fig. 6A-D).
Activity of PEGylated PRO-007 scFv
RTl 12 cells were incubated with PEGylated or non-PEGylated PRO-007 scFv. The inhibitory effect of the PEGylated PRO-007 scFv was demonstrated to match that of the non-modified single chain (Fig. 7). Relative expression of FGFRs in bladder carcinoma cell lines
The relative expression levels of FGFRs in RTl 12, 5637, RT4, Jon and SW780 were analyzed by Western blot analysis (Fig. 8). This demonstrated low or undetectable FGFRl expression and moderate or high expression of FGFR2 and FGFR3 in all lines as compared to primary articular chondrocytes. FGF-induced signal transduction in RTl 12 cells
Serum starved RTl 12 cells were pre-incubated 20 minutes with 150μg/ml PRO- 007 scFv and then stimulated 5 minutes with 100ng/ml FGFl. Pre-incubation with PRO- 007 scFv completely blocked ERKl and 2 activation by FGFl as judged by Western with anti-phospho MAPK antibodies (Fig. 9). Example 8: inhibition of Osteosarcoma cell proliferation
SA0A2 and SJSA are two Osteosarcoma cell lines. Osteosarcoma SAOA2 cells were cultured in Dulbecco Modified Eagle's Minimal Essential Medium (DMEM- Gibco, California, USA)+10% FCS and SJSA cells were cultured in Roswell Park Memorial Institute medium (RPMI Gibco, California, USA) +10% FCS. Cells were seeded in a concentration of 2000 cells per well in a 96 well plate and incubated with the antibodies for 4 days. Antibody containing medium was replaced once. Cell proliferation was measured using the XTT assay).
Relative to untreated cells, treatment with single chain Fv of PRO-007 significantly inhibited cell proliferation in SAOA2 cell line (fig. 10A). Figure 1OB demonstrates that PRO-007 scFv, PRO-001 scFv and PRO-001 Fab all inhibit cell proliferation in SASO2 cells. Figure HA again shows a marked inhibition of SASO2 cell proliferation by PRO-007 scFv and an even more pronounced inhibition by PRO- 001 scFv. In SJSAl cells there was significant inhibition of cell proliferation with a stronger inhibition by PRO-001 scFv (fig. 1 IB).
In different cell lines FGFR2 and FGFR3 have varying significance with respect to cell proliferation. In Bladder cancer cell lines FGFR2 is more important for cell proliferation while in Osteosarcoma cell lines FGFR3 is more important for cell proliferation. Example 9: Antibody binding to heparin binding domain
PRO-007, PRO-001 and PRO-008 were examined for interference with the interaction between FGFR2 or FGFR3 and heparin. Heparin binding to the fragment crystalline fusion of the extracellular domain of FGFR3 (FGFR3/Fc) or FGFR2 (FGFR2/Fc) was measured in the presence of PRO-007, PRO-001 and PRO-008 antibodies. Heparin Sepharose anchored FGFR3/Fc or FGFR2/Fc were incubated with the indicated antibodies. Bound (B) and unbound (U) fractions were then analyzed using Coomassie-stained SDS-PAGE. As shown in figure 12, FGFR3 was located entirely in the unbound fraction in the presence of either PRO-001 or PRO-007. The scFv and the Fab format of both antibodies demonstrated equal receptor dissociating activity. In contrast, FGFR3 remained heparin bound in the presence of PRO-008 Fab. These results demonstrate that contrary to PRO-001 and PRO-007 which compete with heparin, PRO- 008 does not bind the receptor in the heparin binding domain. Figure 12 further demonstrates that PRO-007 also competes with heparin binding to FGFR2.
Example 10: inhibition of ERK phosphorylation by PRO-007 in RCJ cells RCJ cell assay RCJ cells (fetal rat calvaria-derived mesenchymal cells, RCJ 3.1C5.18;
Grigoriadis, 1988) were generated to express various FGF receptors in an inducible manner, in the absence of tetracycline. RCJ cells that stably express either FGFRl, FGFR2 or FGFR3 were stimulated for 5 minutes with 20ng/ml of either FGF2 (stimulating FGFRl and FGFR2, ProChon Biotech Ltd., Rehovot, Israel) or FGF9 (stimulating FGFR3, ProChon Biotech Ltd., Rehovot, Israel). Parallel cultures were pre- incubated for 20 minutes with either 12.5 μg/ml, 25 μg/ml or 50 μg/ml of PRO-007. Cell lysates were analyzed by Western blot with anti-phospho-ERK (Sigma) followed by HRP-secondary antibody. ECL-exposed X-ray demonstrates that increasing amounts of PRO-007 inhibit ligand-dependent receptor phosphorylation in RCJ cells specifically expressing FGFR2 or FGFR3 (fig. 13).
Example 11 : Examining PRO-007 in additional bladder cancer cell lines
In addition to RTl 12, SW780, 5637, RT4 and Jon TCC cell lines MGHU3 and
97-7 bladder cancer cell lines are also examined for the effect of PRO-007. MGHU3 and the 97-7 cells are seeded at a concentration of about 2000 cells/well in a 96 well plate. The next day, PRO-007sc or a control scFv are added at increasing concentrations. Five days latter, cell proliferation is measured by XTT.
Example 12: Examining PRO-007 in bladder cancer cells from human patients
The aim in this study is to test the effects of neutralizing anti-FGFR antibodies on primary cultures of human bladder cancer and to compare them to normal bladder cells. Patients
Five to ten patients undergoing transurethral resection of bladder tumor (TUR- BT) form the experimental group and 5-10 patients undergoing open transvesical prostatectomy for benign prostate hyperplasia (BPH) serve as controls. Pathological characteristic of the tumors is obtained from histopathological analysis. Methods
A small part of the resected tumor (< 1% of the tumor) or normal bladder is removed for establishment of a primary cell culture. Samples are transported in sterile Hanks' balanced salt solution (HBSS) containing 10 mM Hepes and 20 KIU/ml of aprotonin. Samples are washed in transport medium to remove excess blood cells, and incubated at 4°C in 0.02% EDTA in phosphate-buffered saline (PBS) overnight; EDTA is replaced at least once during incubation. The following morning the urothelium is gently removed using fine forceps or a scalpel blade. Urothelial sheets are washed twice in HBSS and digested to a single-cell suspension with 200 U/ml of collagenase type IV for 10 min at 370C. Cells are washed in keratinocyte serum-free medium containing 30 ng/ml cholera toxin and 100 μg/ml of penicillin/streptomycin solution (KSFM). Cells are resuspended in fresh KSFM and placed in 25 cm2 culture flasks at a density of 4χlO4/cm2 and incubated at 37°C in 5% CO2.
Cells are passaged by incubating with 0.02% EDTA at 37°C for 10 min, followed by trypsin-EDTA (Sigma) for a further 60 s. Cells are disaggregated by gentle agitation, and then serum containing medium is added to the flask to neutralize residual trypsin.
Analysis of FGFR expression
All samples are analyzed for FGFRs expression using Western blotting. On the first splitting, part of the cells are grown for whole cell protein extraction and processed for immunoblotting using anti-FGFRs antibodies.
Viability assay
Cell viability is assessed by XTT dye absorbance. Cells are seeded in 96-well plates at a density of 3,000-10,000 cells per well in culture medium. Cells are incubated in the absence or presence of increasing concentrations of the PRO-007 antibody. Plates are incubated for 48 or 72 hours at 370C, 5% CO2. XTT assays are performed according to the manufacturer's instruction.
Data analysis
The results of cell viability from tumor and normal cells are correlated with FGFR expression levels, grade and pathological stage of the disease. While certain embodiments of the invention have been illustrated and described, it will be clear that the invention is not limited to the embodiments described herein. Numerous modifications, changes, variations, substitutions and equivalents will be apparent to those skilled in the art without departing from the spirit and scope of the present invention as described by the claims, which follow.
REFERENCES
Bange J, Prechtl D, Cheburkin Y5 Specht K, Harbeck N, Schmitt M, Knyazeva T, Muller S, Gartner S, Sures I, Wang H, Imyanitov E, Haring HU, Knayzev P, Iacobelli S, Hofier H, Ullrich A.1 Cancer progression and tumor cell motility are associated with the FGFR4 Arg(388) allele. Cancer Res. 62(3):840-7. 2002.
Bernard-Pierrot I, Ricol D, Cassidy A, Graham A, Elvin P, Caillault A, Lair S, Broet P, Thiery JP, Radvanyi F. Inhibition of human bladder tumour cell growth by fibroblast growth factor receptor 2b is independent of its kinase activity. Involvement of the carboxy-terminal region of the receptor. Oncogene. 23(57):9201-ll. 2004. Better M, Chang CP, Robinson RR, Horwitz AH. Escherichia coli secretion of an active chimeric antibody fragment. Science. 240(4855): 1041-3. 1988.
Billerey C, Chopin D, Aubriot-Lorton MH, Ricol D, Gil Diez de Medina S, Van Rhijn B, Bralet MP, Lefrere-Belda MA, Lahaye JB, Abbou CC, Bonaventure J, Zafrani ES, van der Kwast T, Thiery JP, Radvanyi F. Frequent FGFR3 mutations in papillary non-invasive bladder (pTa) tumors. Am J Pathol. 158(6):1955-9. 2001.
Cabilly S, Riggs AD, Pande H, Shively JE, Holmes WE, Rey M3 Perry LJ, Wetzel R, Heyneker HL. Generation of antibody activity from immunoglobulin polypeptide chains produced in Escherichia coli. Proc Natl Acad Sci U S A. 81(11):3273-7. 1984. Cappellen D, De Oliveira C, Ricol D, de Medina S, Bourdin J, Sastre-Garau X,
Chopin D, Thiery JP, Radvanyi F. Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas. Nat Genet. 23(1): 18-20. 1999.
Colligan et al (Eds.), Current Protocols in Immunology, Chapter 17, Section 17.1. 1992-2000, Easton DF et al. Genome-wide association study identifies novel breast cancer susceptibility loci. Nature. 2007.
Ezzat S, Zheng L, Zhu XF, Wu GE, Asa SL. Targeted expression of a human pituitary tumor-derived isoform of FGF receptor-4 recapitulates pituitary tumorigenesis. J Clin Invest. 109(1): 15-6. 2002.
Fortin D, Rom E, Sun H, Yayon A, Bansal R. Distinct fibroblast growth factor (FGF)/FGF receptor signaling pairs initiate diverse cellular responses in the oligodendrocyte lineage. JNeurosci. 25(32):7470-9. 2005.
Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press, 1990.
Hunter DJ et al. A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat Genet. 2007.
Ishiwata T, Friess H, Buchler MW, Lopez ME, Korc M. Characterization of keratinocyte growth factor and receptor expression in human pancreatic cancer. Am J Pathol. 153(l):213-22. 1998.
Johnston CL, Cox HC, Gomm JJ, Coombes RC Fibroblast growth factor receptors (FGFRs) localize in different cellular compartments. A splice variant of FGFR- 3 localizes to the nucleus J Biol Chem. 270(51):30643-50. 1995.
Kan M, Wang F, Xu J, Crabb JW, Hou J, McKeehan WL. An essential heparin- binding domain in the fibroblast growth factor receptor kinase. Science. 259(5103):1918- 21. 1993.
Khnykin D, Troen G, Berner JM, Delabie J. The expression of fibroblast growth factors and their receptors in Hodgkin's lymphoma. J Pathol. 208(3):431-8. 2006.
Knappik A, Ge L, Honegger A, Pack P, Fischer M, Wellnhofer G, Hoess A, Wolle J, Pluckthun A, Virnekas B. Fully synthetic human combinatorial antibody libraries (HuCAL) based on modular consensus frameworks and CDRs randomized with trinucleotides. J MoI Biol. 296(l):57-86. 2000.
Kohler G, Baumann B, Iglesias A, McCubrey J, Potash MJ, Traunecker A, Zhu D. Different ways to modify monoclonal antibodies. Med Oncol Tumor Pharmacother. l(4):227-33. 1984. Kurban G, Ishiwata T5 Kudo M, Yokoyama M, Sugisaki Y, Naito Z. Expression of keratinocyte growth factor receptor (KGFR/FGFR2 IHb) in human uterine cervical cancer. Oncol Rep. 11(5):987-91. 2004.
Lorenzi MV, Horii Y, Yamanaka R, Sakaguchi K, Miki T FRAGl, a gene that potently activates fibroblast growth factor receptor by C-terminal fusion through chromosomal rearrangement. Proc Natl Acad Sci U S A. 93(17):8956-61. 1996.
Martinez-Torrecuadrada J, Cifuentes J, Lόpez-Serra P, Saenz,P, Martinez A and Casal JI. Targeting the Extracellular Domain of Fibroblast Growth Factor Receptor 3 with Human Single-Chain Fv Antibodies Inhibits Bladder Carcinoma Cell Line Proliferation. Clinical Cancer Research 11: 6280-6290. 2005.
Meinkoth J, Wahl G. Hybridization of nucleic acids immobilized on solid supports. Anal Biochem. 138(2):267-84. 1984.
Mϋller KM, Arndt KM, Strittmatter W, Plϋckthun A. The first constant domain (C(H)I and C(L)) of an antibody used as heterodimerization domain for bispecific miniantibodies. FEBS Lett. 422(2):259-64. 1998.
Ornitz DM, ItohN. Fibroblast growth factors. Genome Biol. 2(3) reviews3005.1- reviews3005.12. 2001.
Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa., 1990. Ricol D, Cappellen D, El Marjou A5 Gil-Diez-de-Medina S5 Girault JM, Yoshida
T5 Ferry G5 Tucker G, Poupon MF5 Chopin D5 Thiery JP5 Radvanyi F. Tumour suppressive properties of fibroblast growth factor receptor 2-IIIb in human bladder cancer. Oncogene.l8(51):7234-43. 1999.
Saltzman WM5 Langer R. Transport rates of proteins in porous materials with known microgeometry. Biophys J. 55(1):163-71. 1989.
Sambrook et ah, Molecular Cloning: A Laboratory Manual, 2d edition, Cold Spring Harbor Press. 1989.
Sherwood JK5 Dause RB5 Saltzman WM. Controlled antibody delivery systems. Biotechnology (N Y). 10(ll):1446-9. 1992. van Rhijn BW, van Tilborg AA, Lurkin I5 Bonaventure J, de Vries A, Thiery JP, van der Kwast TH, Zwarthoff EC, Radvanyi F. Novel fibroblast growth factor receptor 3 (FGFR3) mutations in bladder cancer previously identified in non-lethal skeletal disorders. Eur J Hum Genet. 10(12):819-24. 2002. Yee CJ, Lin O, Boyd J. Analysis of fibroblast growth factor receptor 3 S249C mutation in cervical carcinoma. J Natl Cancer Inst. 92(22): 1848-9. 2000.
Zhang B, Xu G, Evans JS. Model peptide studies of sequence repeats derived from the intracrystalline biomineralization protein, SM50. II. Pro,Asn-rich tandem repeats. Biopolymers. 54(6):464-75. 2000. Zieger K, Dyrskjot L, Wiuf C, Jensen JL, Andersen CL, Jensen KM, Orntoft TF.
Role of activating fibroblast growth factor receptor 3 mutations in the development of bladder tumors. Clin Cancer Res. ll(21):7709-19. 2005.

Claims

1. A molecule comprising at least the antigen-binding portion of an antibody which has affinity for fibroblast growth factor receptor 2 (FGFR2) optionally with cross-reactivity to other FGF receptors, which blocks both ligand-dependent and constitutive ligand-independent receptor activation.
2. The molecule according to claim 1 wherein the antibody has affinity to FGFR2 and FGFR3.
3. The molecule according to claim 2 wherein the antibody binds both FGFR2 and FGFR3 with affinity of at least 5OnM, preferably at least 10 nM.
4. The molecule according to claim 1 substantially devoid of affinity to FGFRl .
5. The molecule according to claim 1 wherein the antibody is selected from a monoclonal antibody having binding affinity to FGFR2 with cross-reactivity to other FGF receptors, a proteolytic fragment of said monoclonal antibody and a single chain antibody.
6. The molecule according to claim 1 comprising a VH-CDR3 region having an amino acid sequence set forth in SEQ ID NO:1 and a VL-CDR3 region having an amino acid sequence set forth in SEQ ID NO:2.
7. The molecule according to claim 5 wherein said fragment of an antibody is a single chain Fv.
8. The molecule according to claim 7 wherein said single chain Fv has an amino acid sequence set forth in SEQ ID NO: 5.
9. The molecule according to claim 5 wherein said fragment of an antibody is an Fab fragment.
10. The molecule according to claim 5 wherein said fragment of an antibody is an F(ab')2 fragment.
11. The molecule according to claim 5 wherein the antibody or antibody fragment is modified.
12. The molecule according to claim 11 wherein the modification is PEGylation.
13. A pharmaceutical composition for preventing, attenuating or treating a disease or disorder associated with FGFR2, comprising at least one molecule comprising at least the antigen-binding portion of an antibody which has affinity for FGFR2 with cross-reactivity to other FGF receptors; and a pharmaceutically acceptable
5 carrier.
14. The pharmaceutical composition according to claim 13 wherein the molecule has affinity to FGFR2 and FGFR3.
15. The pharmaceutical composition according to claim 14 wherein the antibody binds both FGFR2 and FGFR3 with affinity of at least 5OnM, preferably at least
10 1O nM.
16. The pharmaceutical composition according to claim 13 wherein the molecule is substantially devoid of affinity to FGFRl.
17. The pharmaceutical composition according to claim 13 wherein the molecule is selected from a monoclonal antibody having binding affinity to FGFR2 with
15 cross-reactivity to other FGF receptors, a proteolytic fragment of said monoclonal antibody and a single chain antibody.
18. The pharmaceutical composition according to claim 17 wherein said molecule comprises a VR-CDR3 region having an amino acid sequence set forth in SEQ ID NO:1 and a VL-CDR3 region having an amino acid sequence set forth in SEQ ID
20 NO:2.
19. The pharmaceutical composition according to claim 17 wherein said molecule is a single chain Fv.
20. The pharmaceutical composition according to claim 19 wherein said single chain Fv has an amino acid sequence set forth in SEQ ID NO:5.
25 21. The pharmaceutical composition according to claim 17 wherein said fragment of an antibody is an F'ab fragment.
22. The pharmaceutical composition according to claim 17 wherein said fragment of an antibody is an F(ab')2 fragment.
23. The pharmaceutical composition according to claim 17 wherein the antibody or 30 antibody fragment is modified. 000732
24. The pharmaceutical composition according to claim 23 wherein the modification is PEGylation.
25. The pharmaceutical composition according to claim 13 wherein the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder.
5 26. The pharmaceutical composition according to claim 25 wherein the cell proliferative disease or disorder is of epithelial origin.
27. The pharmaceutical composition according to claim 26 wherein the cell proliferative disease or disorder is selected from bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
10 28. A method for preventing, attenuating or treating a disease or disorder associated with FGFR2, comprising administering to an individual in need thereof a therapeutically effective amount of the molecule according to claim 1 ; and a pharmaceutically acceptable carrier.
29. The method according to claim 28 wherein the molecule has affinity to FGFR2 15 and FGFR3.
30. The method according to claim 29 wherein the antibody binds both FGFR2 and FGFR3 with affinity of at least 5OnM, preferably at least 1OnM.
31. The method according to claim 28 wherein the molecule is substantially devoid of affinity to FGFRl.
20 32. The method according to claim 28 wherein said molecule is selected from a monoclonal antibody, a proteolytic fragment of a monoclonal antibody and a single chain antibody.
33. The method according to claim 32 wherein said molecule comprises a VH-CDR3 region having an amino acid sequence set forth in SEQ ID NO:1 and a VL-CDR3
25 region having an amino acid sequence set forth in SEQ ID NO:2.
34. The method according to claim 32 wherein said molecule is a single chain Fv.
35. The method according to claim 34 wherein said single chain Fv has an amino acid sequence set forth in SEQ ID NO:5.
36. The method according to claim 32 wherein said fragment of an antibody is a Fab 30 fragment.
37. The method according to claim 32 wherein said fragment of an antibody is a F(ab')2 fragment.
38. The method according to claim 32 wherein the antibody or antibody fragment is modified.
5 39. The method according to claim 38 wherein the modification is PEGylation.
40. The method according to claim 28 wherein the disease or disorder associated with FGFR2 is a cell proliferative disease or disorder.
41. The method according to claim 40 wherein the cell proliferative disease or disorder is of epithelial origin.
10 42. The method according to claim 41 wherein the cell proliferative disease or disorder is selected from bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
43. Use of a molecule according to claim 1 for the preparation of a pharmaceutical composition useful for the treatment of a cell proliferative disease or disorder.
15 44. Use of the pharmaceutical composition according to claim 13 for the treatment of a cell proliferative disease or disorder, wherein the cell proliferative disease or disorder is selected from bladder cancer, breast cancer, ovarian cancer, prostate cancer, osteosarcoma and chondrosarcoma.
20 .
PCT/IL2007/000732 2006-06-15 2007-06-17 Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof WO2007144893A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP07766788A EP2046384A4 (en) 2006-06-15 2007-06-17 Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof
CA002655504A CA2655504A1 (en) 2006-06-15 2007-06-17 Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof
US12/304,638 US8101721B2 (en) 2006-06-15 2007-06-17 Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81369706P 2006-06-15 2006-06-15
US60/813,697 2006-06-15

Publications (2)

Publication Number Publication Date
WO2007144893A2 true WO2007144893A2 (en) 2007-12-21
WO2007144893A3 WO2007144893A3 (en) 2009-04-23

Family

ID=38832213

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2007/000732 WO2007144893A2 (en) 2006-06-15 2007-06-17 Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof

Country Status (4)

Country Link
US (1) US8101721B2 (en)
EP (1) EP2046384A4 (en)
CA (1) CA2655504A1 (en)
WO (1) WO2007144893A2 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010054265A2 (en) 2008-11-07 2010-05-14 Galaxy Biotech, Llc. Monoclonal antibodies to fibroblast growth factor receptor 2
WO2010111367A1 (en) 2009-03-25 2010-09-30 Genentech, Inc. Anti-fgfr3 antibodies and methods using same
US8187601B2 (en) 2008-07-01 2012-05-29 Aveo Pharmaceuticals, Inc. Fibroblast growth factor receptor 3 (FGFR3) binding proteins
EP2569012A2 (en) * 2010-05-11 2013-03-20 Aveo Pharmaceuticals, Inc. Anti-fgfr2 antibodies
WO2013076186A1 (en) 2011-11-23 2013-05-30 Bayer Intellectual Property Gmbh Anti-fgfr2 antibodies and uses thereof
WO2013087725A1 (en) 2011-12-12 2013-06-20 Institut National De La Sante Et De La Recherche Medicale (Inserm) Antagonist of the fibroblast growth factor receptor 3 (fgfr3) for use in the treatment or the prevention of skeletal disorders linked with abnormal activation of fgfr3
WO2013087716A2 (en) 2011-12-14 2013-06-20 Bayer Pharma Aktiengesellschaft New antibody drug conjugates (adcs) and the use thereof
WO2014138364A2 (en) 2013-03-06 2014-09-12 Genentech, Inc. Methods of treating and preventing cancer drug resistance
WO2015191986A1 (en) 2014-06-13 2015-12-17 Genentech, Inc. Methods of treating and preventing cancer drug resistance
US9226949B2 (en) 2010-09-21 2016-01-05 Hepacore Ltd. FGF-18 truncated variants having increased receptor specificity and uses thereof
WO2016073789A3 (en) * 2014-11-05 2016-06-30 Genentech, Inc. Anti-fgfr2/3 antibodies and methods using same
AU2013205318B2 (en) * 2009-03-25 2016-07-07 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
WO2016139227A1 (en) 2015-03-03 2016-09-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Fgfr3 antagonists
WO2017091580A1 (en) * 2015-11-23 2017-06-01 Five Prime Therapeutics, Inc. Predicting response to cancer treatment with fgfr2 inhibitors
WO2018185050A1 (en) 2017-04-03 2018-10-11 Covagen Ag Fgfr3 binding molecules
US10172937B2 (en) 2013-08-01 2019-01-08 Five Prime Therapeutics, Inc. Method of treatment of malignant solid tumors with afucosylated anti-FGFR2IIIb antibodies
WO2019064304A1 (en) * 2017-09-28 2019-04-04 Avner Yayon Methods and compositions for treating multiple sclerosis
US10294289B2 (en) 2016-07-07 2019-05-21 Therachon Sas Soluble fibroblast growth factor receptor 3 (SFGFR3) polypeptides and uses thereof
US10689705B2 (en) 2012-09-27 2020-06-23 Chugai Seiyaku Kabushiki Kaisha FGFR3 fusion gene and pharmaceutical drug targeting same
US10724014B2 (en) 2013-01-16 2020-07-28 Institut National De La Sante Et De La Recherche Medicale Soluble fibroblast growth factor receptor 3 (FGR3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
US11091555B2 (en) 2017-05-16 2021-08-17 Five Prime Therapeutics, Inc. Method of treating gastric cancer with anti-FGFR2-IIIb antibodies and modified FOLFOX6 chemotherapy
US11505611B2 (en) 2020-08-21 2022-11-22 Genzyme Corporation FGFR3 antibodies and methods of use

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JOP20190083A1 (en) 2008-06-04 2017-06-16 Amgen Inc Fgf21 mutant fusion polypeptides and uses thereof
MX341149B (en) 2008-10-10 2016-08-09 Amgen Inc Fgf21 mutants and uses thereof.
AU2010246038A1 (en) 2009-05-05 2011-12-01 Amgen Inc. FGF21 mutants and uses thereof
SI3248610T1 (en) 2009-05-05 2024-03-29 Amgen Inc., Fgf21 mutants and uses thereof
WO2010148142A1 (en) * 2009-06-17 2010-12-23 Amgen Inc. Chimeric fgf19 polypeptides and uses thereof
WO2011000384A1 (en) * 2009-06-30 2011-01-06 Общество С Ограниченной Ответсвенностью "Онкомакс" Method for suppressing tumor growth by blocking fibroblast growth factor receptor, and method for diagnosing malignant neoplasms
AU2010326024A1 (en) * 2009-12-02 2012-07-05 Amgen Inc. Binding proteins that bind to human FGFR1c, human beta-Klotho and both human FGFR1c and human beta-Klotho
UA109888C2 (en) * 2009-12-07 2015-10-26 ANTIBODY OR ANTIBODILITY ANTIBODY OR ITS BINDING TO THE β-CLOTE, FGF RECEPTORS AND THEIR COMPLEXES
SG185832A1 (en) 2011-05-10 2012-12-28 Agency Science Tech & Res Fgfr1 antibodies and treatment of cancer
KR20220119143A (en) * 2019-12-24 2022-08-26 디잘 (지앙수) 파마슈티칼 씨오., 리미티드 Novel anti-FGFR2B antibody

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4376110A (en) * 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
JPS61134325A (en) 1984-12-04 1986-06-21 Teijin Ltd Expression of hybrid antibody gene
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5096815A (en) * 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
US6900053B2 (en) * 2001-09-14 2005-05-31 Isis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 2 expression
DE122004000008I1 (en) 1991-06-14 2005-06-09 Genentech Inc Humanized heregulin antibody.
AU676150B2 (en) 1992-01-23 1997-03-06 Merck Patent Gmbh Monomeric and dimeric antibody-fragment fusion proteins
US5783568A (en) * 1994-06-10 1998-07-21 Sugen, Inc. Methods for treating cancer and other cell proliferative diseases
WO1996013583A2 (en) 1994-10-20 1996-05-09 Morphosys Gesellschaft Für Proteinoptimierung Mbh Targeted hetero-association of recombinant proteins to multi-functional complexes
GB9421405D0 (en) * 1994-10-21 1994-12-07 Dow Chemical Co Low voc laminating formulations
JPH11508126A (en) 1995-05-23 1999-07-21 モルフォシス ゲゼルシャフト ファー プロテインオプティマイルング エムベーハー Multimeric protein
US5719867A (en) 1995-06-30 1998-02-17 Scientific-Atlanta, Inc. Plural telephony channel baseband signal demodulator for a broadband communications system
AU725609C (en) * 1995-08-18 2002-01-03 Morphosys Ag Protein/(poly)peptide libraries
AU3494397A (en) * 1996-06-18 1998-01-07 United States Of America, Represented By The Secretary, Department Of Health And Human Services, The Fibroblast growth factor receptor activating gene i and related compositions and methods
US6316429B1 (en) * 1997-05-07 2001-11-13 Sugen, Inc. Bicyclic protein kinase inhibitors
US6987113B2 (en) * 1997-06-11 2006-01-17 Sugen, Inc. Tyrosine kinase inhibitors
US20030087854A1 (en) 2001-09-10 2003-05-08 Isis Pharmaceuticals Inc. Antisense modulation of fibroblast growth factor receptor 3 expression
US6342221B1 (en) * 1999-04-28 2002-01-29 Board Of Regents, The University Of Texas System Antibody conjugate compositions for selectively inhibiting VEGF
WO2002102973A2 (en) 2001-06-20 2002-12-27 Prochon Biotech Ltd. Antibodies that block receptor protein tyrosine kinase activation, methods of screening for and uses thereof
IL156495A0 (en) 2003-06-17 2004-01-04 Prochon Biotech Ltd Use of fgfr3 antagonists for treating t cell mediated diseases
AU2004312376A1 (en) * 2003-12-19 2005-07-21 Five Prime Therapeutics, Inc. Fibroblast growth factor receptors 1, 2, 3, and 4 as targets for therapeutic intervention
US8497081B2 (en) * 2004-02-24 2013-07-30 Allergan, Inc. Botulinum toxin screening assays
WO2006048877A2 (en) 2004-11-04 2006-05-11 Fibron Limited Treatment of b-cell malignancies with fgfr3 inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2046384A4 *

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8187601B2 (en) 2008-07-01 2012-05-29 Aveo Pharmaceuticals, Inc. Fibroblast growth factor receptor 3 (FGFR3) binding proteins
US8603987B2 (en) 2008-11-07 2013-12-10 Galaxy Biotech, Llc Monoclonal antibodies to fibroblast growth factor receptor 2
EP3290052A1 (en) 2008-11-07 2018-03-07 Galaxy Biotech, LLC Monoclonal antibodies to fibroblast growth factor receptor 2
US8101723B2 (en) 2008-11-07 2012-01-24 Galaxy Biotech, Llc Monoclonal antibodies to fibroblast growth factor receptor 2
US9834609B2 (en) 2008-11-07 2017-12-05 Galaxy Biotech, Llc Methods of detecting a tumor expressing fibroblast growth factor receptor 2
US10689448B2 (en) 2008-11-07 2020-06-23 Galaxy Biotech, Llc Monoclonal antibodies to fibroblast growth factor receptor 2
WO2010054265A2 (en) 2008-11-07 2010-05-14 Galaxy Biotech, Llc. Monoclonal antibodies to fibroblast growth factor receptor 2
US10138301B2 (en) 2008-11-07 2018-11-27 Galaxy Biotech, Llc Monoclonal antibodies to fibroblast growth factor receptor 2
US9382324B2 (en) 2008-11-07 2016-07-05 Galaxy Biotech, Llc Monoclonal antibodies to fibroblast growth factor receptor 2
EP3783024A1 (en) 2008-11-07 2021-02-24 Galaxy Biotech, LLC Monoclonal antibodies to fibroblast growth factor receptor 2
EP2842573A1 (en) 2008-11-07 2015-03-04 Galaxy Biotech, LLC Monoclonal antibodies to Fibroblast Growth Factor receptor 2
AU2013205318B2 (en) * 2009-03-25 2016-07-07 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
CN104788564A (en) * 2009-03-25 2015-07-22 健泰科生物技术公司 Anti-FGFR3 antibodies and methods using same
EP2679600A1 (en) 2009-03-25 2014-01-01 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
US8710189B2 (en) 2009-03-25 2014-04-29 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
US11401333B2 (en) 2009-03-25 2022-08-02 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
CN102378767B (en) * 2009-03-25 2015-01-14 健泰科生物技术公司 Anti-fgfr3 antibodies and methods using same
US10287356B2 (en) 2009-03-25 2019-05-14 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
US8410250B2 (en) 2009-03-25 2013-04-02 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
US9161977B2 (en) 2009-03-25 2015-10-20 F. Hoffmann-La Roche Ag Anti-FGFR3 antibodies and methods using same
US10000571B2 (en) 2009-03-25 2018-06-19 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
WO2010111367A1 (en) 2009-03-25 2010-09-30 Genentech, Inc. Anti-fgfr3 antibodies and methods using same
EP3702371A1 (en) 2009-03-25 2020-09-02 Genentech, Inc. Anti-fgfr3 antibodies and methods using same
CN102378767A (en) * 2009-03-25 2012-03-14 霍夫曼-拉罗奇有限公司 Anti-fgfr3 antibodies and methods using same
US9499623B2 (en) 2009-03-25 2016-11-22 Genentech, Inc. Anti-FGFR3 antibodies and methods using same
JP2013529076A (en) * 2010-05-11 2013-07-18 アベオ ファーマシューティカルズ, インコーポレイテッド Anti-FGFR2 antibody
EP2569012A2 (en) * 2010-05-11 2013-03-20 Aveo Pharmaceuticals, Inc. Anti-fgfr2 antibodies
EP2569012A4 (en) * 2010-05-11 2013-10-30 Aveo Pharmaceuticals Inc Anti-fgfr2 antibodies
US9226949B2 (en) 2010-09-21 2016-01-05 Hepacore Ltd. FGF-18 truncated variants having increased receptor specificity and uses thereof
WO2013076186A1 (en) 2011-11-23 2013-05-30 Bayer Intellectual Property Gmbh Anti-fgfr2 antibodies and uses thereof
WO2013087725A1 (en) 2011-12-12 2013-06-20 Institut National De La Sante Et De La Recherche Medicale (Inserm) Antagonist of the fibroblast growth factor receptor 3 (fgfr3) for use in the treatment or the prevention of skeletal disorders linked with abnormal activation of fgfr3
WO2013087716A2 (en) 2011-12-14 2013-06-20 Bayer Pharma Aktiengesellschaft New antibody drug conjugates (adcs) and the use thereof
US10689705B2 (en) 2012-09-27 2020-06-23 Chugai Seiyaku Kabushiki Kaisha FGFR3 fusion gene and pharmaceutical drug targeting same
US10724014B2 (en) 2013-01-16 2020-07-28 Institut National De La Sante Et De La Recherche Medicale Soluble fibroblast growth factor receptor 3 (FGR3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
US11814654B2 (en) 2013-01-16 2023-11-14 Institut National De La Sante Et De La Recherche Medicale Soluble fibroblast growth factor receptor 3 (FGR3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
US11702642B2 (en) 2013-01-16 2023-07-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Soluble fibroblast growth factor receptor 3 (FGR3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
WO2014138364A2 (en) 2013-03-06 2014-09-12 Genentech, Inc. Methods of treating and preventing cancer drug resistance
US11235059B2 (en) 2013-08-01 2022-02-01 Five Prime Therapeutics, Inc. Afucosylated anti-FGFR2IIIB antibodies
US10172937B2 (en) 2013-08-01 2019-01-08 Five Prime Therapeutics, Inc. Method of treatment of malignant solid tumors with afucosylated anti-FGFR2IIIb antibodies
WO2015191986A1 (en) 2014-06-13 2015-12-17 Genentech, Inc. Methods of treating and preventing cancer drug resistance
CN107108740A (en) * 2014-11-05 2017-08-29 豪夫迈·罗氏有限公司 Anti- FGFR2/3 antibody and its application method
CN107108740B (en) * 2014-11-05 2021-09-17 豪夫迈·罗氏有限公司 anti-FGFR 2/3 antibodies and methods of use thereof
WO2016073789A3 (en) * 2014-11-05 2016-06-30 Genentech, Inc. Anti-fgfr2/3 antibodies and methods using same
US10208120B2 (en) 2014-11-05 2019-02-19 Genentech, Inc. Anti-FGFR2/3 antibodies and methods using same
WO2016139227A1 (en) 2015-03-03 2016-09-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Fgfr3 antagonists
WO2017091580A1 (en) * 2015-11-23 2017-06-01 Five Prime Therapeutics, Inc. Predicting response to cancer treatment with fgfr2 inhibitors
US11447553B2 (en) 2015-11-23 2022-09-20 Five Prime Therapeutics, Inc. FGFR2 inhibitors alone or in combination with immune stimulating agents in cancer treatment
US11021528B2 (en) 2016-07-07 2021-06-01 INSERM (Institut National de la Santé et de la Recherche Médicale Soluble fibroblast growth factor receptor 3 (SFGFR3) polypeptides and uses thereof
US10294289B2 (en) 2016-07-07 2019-05-21 Therachon Sas Soluble fibroblast growth factor receptor 3 (SFGFR3) polypeptides and uses thereof
US11697678B2 (en) 2016-07-07 2023-07-11 Pfizer Inc. Soluble fibroblast growth factor receptor 3 (SFGFR3) polypeptides and uses thereof
US11351267B2 (en) 2017-04-03 2022-06-07 Cilag Gmbh International FGFR3 binding molecules
WO2018185050A1 (en) 2017-04-03 2018-10-11 Covagen Ag Fgfr3 binding molecules
US10722589B2 (en) 2017-04-03 2020-07-28 Covagen Ag FGFR3 binding molecules
US11091555B2 (en) 2017-05-16 2021-08-17 Five Prime Therapeutics, Inc. Method of treating gastric cancer with anti-FGFR2-IIIb antibodies and modified FOLFOX6 chemotherapy
WO2019064304A1 (en) * 2017-09-28 2019-04-04 Avner Yayon Methods and compositions for treating multiple sclerosis
US11505611B2 (en) 2020-08-21 2022-11-22 Genzyme Corporation FGFR3 antibodies and methods of use

Also Published As

Publication number Publication date
WO2007144893A3 (en) 2009-04-23
EP2046384A2 (en) 2009-04-15
CA2655504A1 (en) 2007-12-21
US20100047251A1 (en) 2010-02-25
US8101721B2 (en) 2012-01-24
EP2046384A4 (en) 2009-12-02

Similar Documents

Publication Publication Date Title
US8101721B2 (en) Antibodies blocking fibroblast growth factor receptor activation and methods of use thereof
EP1423428B1 (en) Antibodies that block fgfr3 activation, methods of screening for and uses thereof
ES2753756T3 (en) Antibody polytherapy against human CSF-1R and uses thereof
KR101671039B1 (en) Anti CXCR4 antibodies and their use for the treatment of cancer
US8143025B2 (en) Antibodies against vascular endothelial growth factor receptor-1
EP3901175A1 (en) Anti-cd73 monoclonal antibody and application thereof
JP2009539870A (en) Compositions and methods for modulating angiogenesis
AU2005268505A1 (en) Novel anti-IGF-IR antibodies and uses thereof
MX2008000964A (en) Novel anti-igf-ir antibodies and uses thereof.
JP6786722B2 (en) New Antibodies Against Programmed Cell Death Protein (PD-1) and Their Uses
CN108350065B (en) Humanized anti-BAG 3 antibodies
JP2024504124A (en) Novel anti-gremlin 1 antibody
TW202208438A (en) Anti-lilrb1 antibody or antigen-binding fragment thereof, method of preparing the same, pharmaceutical composition, nucleic acid molecule, recombinant vector and recombinant cell
JP2019531337A (en) Cancer treatment using bifunctional molecules targeting growth factors
KR20150045286A (en) Polypeptide specifically binding to VEGF-C and use thereof
RU2813373C1 (en) Anti-lilrb1 antibody and uses thereof
WO2023103962A1 (en) Tnfr2 binding molecule and use thereof
KR102207221B1 (en) Methods of inhibiting pathological angiogenesis with doppel-targeting molecules
EP3440111B1 (en) Anti-vegfr-1 antibodies and uses thereof
CN117396507A (en) anti-BCAM antibodies or antigen-binding fragments thereof
CN117362433A (en) Isolated antigen binding proteins and uses thereof
JP2024508673A (en) Method for inhibiting tumor cell proliferation based on CCDC112
CN116940595A (en) anti-TIGIT antibodies and uses thereof

Legal Events

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

Ref document number: 07766788

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2007766788

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2655504

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12304638

Country of ref document: US