US20110091470A1 - Anti-flt3 antibodies - Google Patents

Anti-flt3 antibodies Download PDF

Info

Publication number
US20110091470A1
US20110091470A1 US12/994,956 US99495609A US2011091470A1 US 20110091470 A1 US20110091470 A1 US 20110091470A1 US 99495609 A US99495609 A US 99495609A US 2011091470 A1 US2011091470 A1 US 2011091470A1
Authority
US
United States
Prior art keywords
seq
antibody
flt3
sequence
antibodies
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/994,956
Other languages
English (en)
Inventor
Yiwen Li
Dan Lu
David Surguladze
James Robert Tonra
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ImClone LLC
Original Assignee
ImClone LLC
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=41092183&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110091470(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by ImClone LLC filed Critical ImClone LLC
Priority to US12/994,956 priority Critical patent/US20110091470A1/en
Assigned to IMCLONE LLC reassignment IMCLONE LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TONRA, JAMES ROBERT, SURGULADZE, DAVID, LU, DAN, LI, YIWEN
Publication of US20110091470A1 publication Critical patent/US20110091470A1/en
Abandoned legal-status Critical Current

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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • 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
    • 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
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention is directed to human antibodies, including fragments or portions thereof, that are specific to human Fms-like tyrosine kinase 3 receptor (FLT3).
  • the antibodies are used for treating growth of cancer cells and can be used alone or in combination with an anti-neoplastic agent, including but not limited to methotrexate (MXT), for treatment of leukemia.
  • MXT methotrexate
  • FLT3 Human Fms-like tyrosine kinase 3 receptor
  • FLK-2 fetal liver kinase 2
  • STK-1 stem cell tyrosine kinase 1
  • CD135 CD135
  • FLT3 is a member of the class III receptor tyrosine kinases.
  • FLT3 is expressed on immature myeloid-lymphocytic precursor cells and dendritic cell precursors, but rarely on mature adult cells.
  • FLT3 is overexpressed in approximately 90% of acute myeloid leukemia (AML), a majority of acute lymphocytic leukemia (ALL) and the blast-crisis phase of chronic myeloid leukemia (BC-CML).
  • Stimulation by FLT3 ligand (FL) enhances the proliferation and survival of leukemia cells. Inhibition of FLT3 signaling leads to apoptosis in dendritic cells and inhibition of immune responses.
  • Small-molecule inhibitors are not completely FLT3-specific and drug resistance can develop. Thus small-molecule FLT3 inhibitors have yet to provide effective targeted therapies for leukemia. New treatments for this unmet medical need are highly desirable.
  • An antibody approach may overcome some of the shortcomings associated with small molecule FLT3 inhibitors.
  • First, antibodies are specific to a defined antigen, thus avoiding potential side effects resulting from inhibition of multiple kinases.
  • FLT3 neutralizing antibodies target the extracellular domain, which is less prone to mutations than the kinase domain, reducing the possibility for drug resistance.
  • antibodies may recruit immune effector mechanisms, such as antibody-dependent cellular cytotoxicity (ADCC) and/or complement-mediated cytotoxicity (CMC), to kill target tumor cells, resulting in increased therapeutic efficacy.
  • ADCC antibody-dependent cellular cytotoxicity
  • CMC complement-mediated cytotoxicity
  • FLT3-specific antibodies can be active against both wild-type (especially in the case of a neutralizing antibody) and mutated FLT3 (
  • anti-FLT3 antibodies including EB10, NC7 and D4-3, inhibit both ligand-dependent (wild-type receptor) as well as ligand-independent (mutant receptor) activation of FLT3 (see Piloto, Cancer Res., supra.).
  • anti-FLT3 antibodies include EB10, NC7 and D4-3, inhibit both ligand-dependent (wild-type receptor) as well as ligand-independent (mutant receptor) activation of FLT3 (see Piloto, Cancer Res., supra.).
  • the precise CDR sequences and epitope binding domains of anti-FLT3 antibodies of the invention have not been known.
  • the present invention seeks to provide human anti-FLT3 antibodies which induce rapid and efficient internalization and down-modulation of cell surface FLT3 compared with those inhibitors known in the art.
  • the present invention seeks to provide human anti-FLT3 antibodies which inhibit FL-induced phosphorylation of wild-type FLT3 and downstream kinases of MPK, PI3K, and STAT5 pathways in leukemia compared with those inhibitors known in the art.
  • ADCC antibody dependent cellular cytotoxicity
  • the present invention provides a method of treating a pre-cancerous condition or cancer in a mammal comprising administering MTX in combination with a FLT3 inhibitor to the mammal in an amount effective to treat the pre-cancerous condition or cancer.
  • the present invention also provides a conjugate comprising a FLT3 inhibitor joined to MTX.
  • the present invention is directed to human antibodies, and fragments thereof, that bind to the human antigen FLT3 (SEQ ID NO:43) with an affinity no greater than 4.5 ⁇ 10 ⁇ 10 M for soluble FLT3-Fc fusion protein at 25° C. as determined by surface plasmon resonance.
  • the present invention is also directed to human monoclonal antagonist antibodies, and fragments thereof, that bind to the human antigen FLT3.
  • One aspect of the present invention is an antibody or fragment thereof that binds FLT3, comprising a CDRH1 having the sequence GYTFTSYYMH (SEQ ID NO:1) or SYYMH (SEQ ID NO:2), a CDRH2 having the sequence IINPSGGSTSYAQKFQG (SEQ ID NO:3), a CDRH3 having the sequence GVGAHDAFDI (SEQ ID NO:4) or VVAAAVADY (SEQ ID NO:5), a CDRL1 having the sequence RSSQSLLHSNGNNYLD (SEQ ID NO:6) or RSSQSLLHSNGYNYLD (SEQ ID NO:7), a CDRL2 having the sequence LGSNRAS (SEQ ID NO:8), and a CDRL3 having the sequence MQGTHPAIS (SEQ ID NO:9) or MQSLQTPFT (SEQ ID NO:11).
  • One aspect of the present invention is an antibody or fragment thereof that binds FLT3 comprising a CDRH1 having the sequence GYTFTSYYMH (SEQ ID NO:1) or SYYMH (SEQ ID NO:2), a CDRH2 having the sequence IINPSGGSTSYAQKFQG (SEQ ID NO:3), a CDRH3 having the sequence GVGAHDAFDI (SEQ ID NO:4), a CDRL1 having the sequence RSSQSLLHSNGNNYLD (SEQ ID NO:6), a CDRL2 having the sequence LGSNRAS (SEQ ID NO:8), and a CDRL3 having the sequence MQGTHPAIS (SEQ ID NO:9).
  • the antibody having the aforementioned CDRs specifically binds human FLT3 with an affinity no greater than 4.5 ⁇ 10 ⁇ 10 M at 25° C. as determined by surface plasmon resonance.
  • Another aspect of the present invention is an antibody or fragment thereof that binds FLT3, and comprises a VL having the sequence:
  • Another aspect of the present invention is an antibody or fragment thereof that binds FLT3, and comprises a VL having the sequence:
  • the present invention is also directed to isolated DNA encoding such antibodies and portions thereof.
  • Other aspects of the present invention include: an isolated polynucleic acid comprising a nucleotide sequence encoding the antibody, or a fragment thereof; an expression vector comprising the nucleotide sequence linked to an expression sequence or a recombinant host cell comprising the expression vector or a recombinant host cell or a progeny thereof, wherein the cell expresses the antibody, or fragment thereof.
  • Yet another aspect of the present invention is a method of producing or purifying an antibody, or fragment thereof, comprising culturing the cells under conditions permitting expression of the antibody or fragment thereof.
  • the light chain can comprise one variable domain (abbreviated herein as VL) and/or one constant domain (abbreviated herein as CL).
  • VL variable domain
  • CL constant domain
  • the light chains of antibodies are either kappa (K) light chains or lambda ( ⁇ ) light chains.
  • K kappa
  • lambda
  • VL as used herein, is intended to include both the variable regions from kappa-type light chains (VK) and from lambda-type light chains (V ⁇ ).
  • the light chain constant region is comprised of one domain, CL.
  • the heavy chain can also comprise one variable domain (abbreviated herein as VH) and/or, depending on the class or isotype of antibody, three or four constant domains (CH1, CH2, CH3 and CH4) (abbreviated herein collectively as CH).
  • VH variable domain
  • CH constant domains
  • the present invention includes antibodies of any of the aforementioned classes or subclasses. Human IgG 1 is the preferred isotype for the antibodies of the present invention.
  • variable domains show considerable amino acid sequence variability from one antibody to the next, particularly at the location of the antigen-binding site.
  • Amino acids are assigned to a particular CDR region or domain in accordance with Kabat convention (Kabat, et al., Ann. NY Acad. Sci. 190:382-93 (1971).; Kabat, et al., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No.
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4.
  • Fv Fram variable
  • Single chain Fv is an antibody fragment containing a VL domain and a VH domain on one polypeptide chain, wherein the N terminus of one domain and the C terminus of the other domain are joined by a flexible linker (see, e.g., U.S. Pat. No.
  • the peptide linkers used to produce the single chain antibodies can be flexible peptides selected to assure that the proper three-dimensional folding and association of the VL and VH domains occurs.
  • the linker is generally 10 to 50 amino acid residues.
  • the linker is 10 to 30 amino acid residues. More preferably the linker is 12 to 30 amino acid residues. Most preferably is a linker of 15 to 25 amino acid residues.
  • a non-limiting example of such a linker peptides is (Gly-Gly-Gly-Gly-Ser) 3 .
  • an “isolated antibody” is an antibody that (1) has been partially, substantially, or fully purified from a mixture of components; (2) has been identified and separated and/or recovered from a component of its natural environment; (3) is monoclonal; (4) is free of other proteins from the same species; (5) is expressed by a cell from a different species; or (6) does not occur in nature. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. Examples of isolated antibodies include an antibody that has been affinity purified, an antibody that has been made by a hybridoma or other cell line in vitro, and a human antibody derived from a transgenic mouse.
  • the term “monoclonal antibody,” as used herein, refers to an antibody obtained from a population of substantially homogeneous antibodies, e.g., the individual antibodies comprising the population are substantially identical except for possible naturally occurring mutations or minor post-translational variations that may be present. Monoclonal antibodies are highly specific, being directed against a single antigenic site (also known as determinant or epitope). Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants, each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • human antibody includes antibodies having variable and constant regions corresponding to human germline immunoglobulin sequences (as described in Kabat, et al., supra and Chothia et al., supra).
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs.
  • the human antibody can have at least one position replaced with an amino acid residue, e.g., an activity enhancing amino acid residue which is not encoded by the human germline immunoglobulin sequence.
  • the term “human antibody”, as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody includes human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal that is transgenic for human immunoglobulin genes, or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences.
  • recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences (See, Kabat, et al., supra and Chothia et al., supra).
  • Fc fragment, crystallizable region
  • IgG antibody for example, the Fc comprises CH2 and CH3 domains.
  • the Fc of an IgA or an IgM antibody further comprises a CH4 domain.
  • the Fc is associated with Fc receptor binding, activation of complement-mediated cytotoxicity (CMC) and ADCC.
  • CMC complement-mediated cytotoxicity
  • IgA and IgM which are complexes of multiple IgG like proteins, complex formation requires Fc constant domains.
  • antibodies of the invention include, but are not limited to, naturally occurring antibodies, antibodies, human antibodies, humanized antibodies, recombinant human antibodies, monoclonal antibodies, digestion fragments, specified portions and variants thereof, including antibody mimetics or comprising portions of antibodies that mimic the structure and/or function of an antibody or specified fragment or portion thereof; each containing at least one CDR.
  • Functional fragments include antigen binding fragments that bind to a FLT3 antigen.
  • antibody fragments capable of binding to FLT3 or a portion thereof, and which are embraced by the present invention include bivalent fragments such as (Fab′) 2 with inter-chain disulfide bonds intact, monovalent fragments such as Fab (Fragment, antigen binding) which refers to the fragments of the antibody consisting of VL-CL VL-CH1 domains and do not retain the heavy chain hinge region (e.g., by papain digestion), fabs which retain the heavy chain hinge region, facb (e.g., by plasmin digestion), F(ab′) 2 , Fab′ which lack disulfide bonds, pFc′ (e.g., by pepsin or plasmin digestion), Fd (e.g., by pepsin digestion, partial reduction and re-aggregation) and Fv or scFv (e.g., by molecular biology techniques).
  • Fab′ fragment, fragment, antigen binding
  • Fab fragment, antigen binding
  • Fab fragment, antigen binding
  • Antibody fragments are also intended to include, e.g., domain deleted antibodies, linear antibodies, single chain antibodies, scFv, single domain antibodies, multivalent single chain antibodies, multi-specific antibodies formed from antibody fragments including diabodies, triabodies, and the like that bind specifically with antigens.
  • the hinge region separates the Fab and Fc portions of the antibody, providing for mobility of Fabs relative to each other and relative to Fc, as well as including multiple disulfide bonds for covalent linkage of the two heavy chains.
  • Antibody formats have been developed which retain binding specificity, but have other characteristics that may be desirable, including for example, bispecificity, multivalence (more than two binding sites), and compact size (e.g., binding domains alone).
  • the antibodies of the present invention are specific for FLT3.
  • Antibody specificity refers to selective recognition of the antibody for a particular epitope of an antigen.
  • Antibodies of the present invention can be monospecific or bispecific.
  • Bispecific antibodies are antibodies that have two different antigen-binding specificities or sites. Where an antibody has more than one specificity, the recognized epitopes can be associated with a single antigen or with more than one antigen.
  • the present invention provides bispecific antibodies that bind to two different antigens, with at least one specificity for FLT3. As stated above, such antibodies include any fragments thereof.
  • Specificity of the present antibodies or fragments thereof, for FLT3 can be determined based on affinity and/or avidity. Affinity, represented by the equilibrium constant for the dissociation of an antigen with an antibody (K D ), measures the binding strength between an antigenic determinant and an antibody-binding site.
  • the antibodies, or fragments thereof, of the invention bind to an epitope of FLT3 which may comprise any one of FLT3's five extracellular domain segments (hereinafter referred simply to as “domains” or “ECD”), i.e, D1, D2, D3, D4 and D5.
  • domains FLT3's five extracellular domain segments
  • Antibodies EB10 and D4-3 bind to an epitope within domain 4 of FLT3, whereas NC7 binds to an epitope within domain 5 of FLT3.
  • epitope refers to discrete, three-dimensional sites on an antigen that are recognized by the antibodies of the invention. Epitopes are the immunologically active regions on a complex antigen, the regions that actually bind to a B-cell receptor, and that are actually bound by the resulting antibody molecules that are produced by the B cell. Antigens generally contain at least one epitope and usually more than one epitope. Epitopes on protein antigens can be linear or non-linear.
  • Linear epitopes are those comprised of contiguous amino acid residues in the amino acid sequence of a protein. Linear epitopes may or may not require conformational folding to form the native three-dimensional structure and elicit an immune response that produces antibodies with binding specificity to the antigen. Non-linear epitopes are comprised of non-contiguous amino acid residues. Thus, non-linear epitopes require some degree of protein folding to bring the requisite amino acid residues into the proximity of one another to form the native three-dimensional structure and elicit an immune response that produces antibodies with binding specificity to the antigen.
  • the antibodies, or fragments thereof, of the present invention bind to wild-type or mutant FLT3.
  • FLT3, either mutant or wild-type, for example, is frequently expressed in AML and ALL, as well as other leukemias. It is mutated in about one-third of acute AML patients, either by internal tandem duplications (ITD) of the juxtamembrane domain or by point mutations usually involving the kinase domain (KD). Both types of mutation constitutively activate FLT3.
  • ITD internal tandem duplications
  • KD point mutations usually involving the kinase domain
  • Both types of mutation constitutively activate FLT3.
  • anti-FLT3 antibodies can also induce ADCC as an additional mechanism for inducing cytotoxicity.
  • Antibodies of the present invention, or fragments thereof, also include those for which binding characteristics have been modified or improved by direct mutation, methods of affinity maturation, phage display, or chain shuffling. Affinity and specificity can be modified or improved by mutating CDR and/or FW residues and screening for antigen binding sites having the desired characteristics (see, e.g., Yang et al., J. Mol. Biol. 254: 392-403 (1995).). CDRs are mutated in a variety of ways. One way is to randomize individual residues or combinations of residues so that in a population of, otherwise identical antigen binding sites, subsets of from two to twenty amino acids are found at particular positions.
  • mutations can be induced over a range of residues by error prone PCR methods (see, e.g., Hawkins et al., J. Mol. Biol. 226: 889-96 (1992)).
  • phage display vectors containing heavy and light chain variable region genes can be propagated in mutator strains of E. coli (see, e.g., Low et al., J. Mol. Biol. 250: 359-68 (1996).).
  • phage display vectors containing heavy and light chain variable region genes can be propagated in mutator strains of E. coli (see, e.g., Low et al., J. Mol. Biol. 250: 359-368 (1996)).
  • a convenient way for generating substitutional variants is affinity maturation using phage display. Briefly, several CDR region sites are mutated to generate all possible amino acid substitutions at each site. The antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle. The phage-displayed variants are then screened for their biological activity (e.g., binding affinity, specificity, IC50, EC50, K D ) as herein disclosed. In order to identify candidate CDR region sites for modification, alanine scanning mutagenesis can be performed to identify CDR region residues contributing significantly to antigen binding.
  • biological activity e.g., binding affinity, specificity, IC50, EC50, K D
  • random mutagenesis may be performed on one or more CDR sequences at one or more residue positions, either while the CDR is operably linked to the variable region or while the CDR is independent of other variable region sequence and then the altered CDR returned to a variable region using recombinant DNA technology.
  • substantially homologous modified antibodies can be readily designed and manufactured utilizing various recombinant DNA techniques well known to those skilled in the art.
  • the framework regions can vary from the native sequences at the primary structure level by several amino acid substitutions, terminal and intermediate additions and deletions, and the like.
  • a variety of different human framework regions may be used singly or in combination as a basis for the humanized immunoglobulins of the present invention.
  • modifications of the genes may be readily accomplished by a variety of well-known techniques, such as site-directed mutagenesis.
  • the present invention includes FLT3-binding polypeptides with amino acid sequences substantially the same as the described amino acid sequence of the variable or hypervariable regions of the full-length anti-FLT3 antibodies.
  • substantially the same amino acid sequence is defined herein as a sequence with at least 70%, preferably at least about 80%, and more preferably at least about 90% homology to another amino acid sequence, as determined by the FASTA search method in accordance with Pearson and Lipman (Proc. Natl. Acad. Sci. USA 85: 2444-8 (1988).).
  • the present invention includes conservative amino acid substitutions that preserve the functional characteristics of the presently disclosed antibodies.
  • the present invention includes nucleic acid sequences that encode an anti-FLT3 antibody heavy chain, comprising any one of the VH regions or a portion thereof, or any one of the VH CDRs, including any variants thereof, as disclosed herein.
  • the invention also includes nucleic acid molecules that encode an anti-FLT3 antibody light chain comprising any one of the VL regions or a portion thereof or any one of the VL CDRs, including any variants thereof as disclosed herein.
  • Each domain of the antibodies of this invention can be a complete antibody with the heavy or light chain variable domain, or it can be a functional equivalent or a mutant or derivative of a naturally-occurring domain, or a synthetic domain constructed, for example, in vitro using a technique such as one described in WO 93/11236 (Griffiths, et al.). For instance, it is possible to join together domains corresponding to antibody variable domains, which are missing at least one amino acid. Also included is an antibody with one or more amino acid substitution, mutation or deletion within one of the CDR sequences. The important characterizing feature is the ability of each domain to associate with a complementary domain to form an antigen-binding site. Accordingly, the terms variable heavy and light chain fragment should not be construed to exclude variants, including variants to the CDRs that do not have a material effect on specificity.
  • the antibodies of the present invention may be produced by methods known in the art. These methods include the immunological method described by Kohler and Milstein, Nature 256: 495-497 (1975) and Campbell, Monoclonal Antibody Technology, The Production and Characterization of Rodent and Human Hybridomas, Burdon et al., Eds., Laboratory Techniques in Biochemistry and Molecular Biology, Volume 13, Elsevier Science Publishers, Amsterdam (1985); as well as by the recombinant DNA method described by Huse et al., Science 246: 1275-1281 (1989).
  • the antibodies can also be obtained from phage display libraries bearing combinations of VH and VL domains in the form of scFv or Fab.
  • VH and VL domains can be encoded by nucleotides that are synthetic, partially synthetic, or naturally derived.
  • phage display libraries bearing human antibody fragments can be preferred.
  • Other sources of human antibodies are transgenic mice engineered to express human immunoglobulin genes.
  • Antibody fragments can be produced by cleaving a whole antibody, or by expressing DNA that encodes the fragment. Fragments of antibodies may be prepared by methods described by Lamoyi et al., J. Immunol. Methods 56: 235-243 (1983) and by Parham, J. Immunol. 131: 2895-2902 (1983). Such fragments may contain one or both Fab fragments or the F(ab′) 2 fragment. Such fragments may also contain single-chain fragment variable region antibodies, i.e. scFv, diabodies, or other antibody fragments. Methods of producing such functional equivalents are disclosed in PCT Application WO 93/21319, European Patent Application No. 239,400; PCT Application WO 89/09622; European Patent Application 338,745; and European Patent Application EP 332,424. Throughout this specification, the term “antibodies” of the invention includes any fragments thereof, whether or not specifically stated.
  • Preferred host cells for transformation of vectors and expression of the antibodies of the present invention are mammalian cells, e.g., NSO cells (non-secreting (0) mouse myeloma cells), 293 and CHO cells and other cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells.
  • NSO cells non-secreting (0) mouse myeloma cells
  • 293 and CHO cells and other cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells.
  • Other eukaryotic hosts, such as yeasts can be alternatively used.
  • the present invention provides isolated antibodies or fragments thereof specific for FLT3.
  • the antibodies of the invention are capable of one or more of the following activities: 1) displaying high affinity binding towards FLT3; 2) blocking ligand binding to FLT3 receptor and therefore to inhibit the activation of FLT3 and its signaling pathway; 3) inducing rapid and efficient internalization and down-modulation of cell surface FLT3; 4) inhibiting FL-induced phosphorylation of wild-type FLT3 and downstream kinases of MPK, PI3K, and STAT5 pathways in leukemia; 5) displaying reduced immunogenicity in humans; 6) displaying improved ability to activate downstream immune effector functions such as antibody dependent cellular cytotoxicity (ADCC); 7) inducing FLT3 receptor internalization and 8) inhibiting tumor growth in vitro and in vivo.
  • the anti-FLT3 antibodies of the present invention are human antibodies that exhibit one or more of following properties, further elucidated throughout the specification, including the Examples:
  • the antibodies of the present invention bind to the external domain of FLT3 and inhibit binding of FL to FLT3 Inhibition can be determined, for example, by a direct binding assay using purified or membrane bound receptor.
  • the antibodies of the present invention, or fragments thereof preferably bind FLT3 at least as strongly as the natural ligands of FLT3.
  • the antibodies of the present invention neutralize FLT3. Neutralization occurs via a variety of mechanisms. One such mechanism is the binding of a FL to an extracellular domain of FLT3 that stimulates autophosphorylation of the beta subunit and phosphorylation of FLT3 substrates, including STAT5, Akt, PI3K and MAPK which are downstream pathways. Neutralization of FLT3 also includes inhibition, diminution, inactivation and/or disruption of one or more of these activities normally associated with signal transduction. Further, neutralization includes inhibition of FLT3 heterodimers as well as FLT3 homodimers.
  • neutralizing FLT3 has various effects, including inhibition, diminution, inactivation and/or disruption of growth (proliferation and differentiation), angiogenesis (blood vessel recruitment, invasion, and metastasis), and cell motility and metastasis (cell adhesion and invasiveness).
  • angiogenesis blood vessel recruitment, invasion, and metastasis
  • cell motility and metastasis cell adhesion and invasiveness
  • tyrosine kinase inhibition can be determined using well-known methods; for example, by measuring the autophosphorylation level of recombinant kinase receptor, and/or phosphorylation of natural or synthetic substrates.
  • phosphorylation assays are useful in determining neutralizing antibodies in the context of the present invention. Phosphorylation can be detected, for example, using an antibody specific for phosphotyrosine in an ELISA assay or on a Western blot.
  • Antibodies of the invention cause a significant decrease in tyrosine phosphorylation of FLT3 of at least about 60%, preferably at least about 75%, and more preferably at least about 85-90% in cells that respond to ligand.
  • FLT3 neutralization Another measure of FLT3 neutralization is inhibition of phosphorylation of downstream substrates of FLT3. Accordingly, the level of phosphorylation of STAT5, PI3K, Akt or MAPK can be measured.
  • methods for detection of protein expression can be utilized to determine FLT3 neutralization, wherein the proteins being measured are regulated by FLT3 tyrosine kinase activity.
  • methods for detection of protein expression include immunohistochemistry (IHC) for detection of protein expression, fluorescence in situ hybridization (FISH) for detection of gene amplification, competitive radioligand binding assays, solid matrix blotting techniques, such as Northern and Southern blots, reverse transcriptase polymerase chain reaction (RT-PCR) and ELISA.
  • IHC immunohistochemistry
  • FISH fluorescence in situ hybridization
  • RT-PCR reverse transcriptase polymerase chain reaction
  • ELISA solid matrix blotting techniques, such as Northern and Southern blots, reverse transcriptase polymerase chain reaction
  • In vivo assays can also be utilized to determine FLT3 neutralization.
  • receptor tyrosine kinase inhibition can be observed by mitogenic assays using cell lines stimulated with receptor ligand in the presence and absence of inhibitor.
  • One method involves testing for inhibition of growth of FLT3-expressing tumor cells or cells transfected to express FLT3. Inhibition can also be observed using tumor models, for example, human tumor cells injected into a mouse.
  • the present invention is not limited by any particular mechanism of FLT3 neutralization.
  • the anti-FLT3 antibodies of the present invention can (1) bind externally to the FLT3 cell surface receptor, (2) block binding to FL and subsequent signal transduction mediated via the receptor-associated tyrosine kinase, and (3) prevent phosphorylation of the FLT3 and other downstream proteins in the signal transduction cascade.
  • the antibodies of the present invention down-modulate FLT3.
  • the amount of FLT3 present on the surface of a cell depends on receptor protein production, internalization, and degradation.
  • the amount of FLT3 present on the surface of a cell can be measured indirectly, by detecting internalization of the receptor or a molecule bound to the receptor.
  • receptor internalization can be measured by contacting or coating cells that express FLT3 with a labeled antibody. The membrane-bound antibody is then stripped, collected and counted. Internalization of the antibody is determined by lysing the cells and detecting the labeled components.
  • the amount of FLT3 present on the surface of a cell can be measured directly by measuring the amount of the receptor present on the cell following treatment with an anti-FLT3 antibody or other substance, for example, by fluorescence-activated cell-sorting analysis of cells stained for surface expression of FLT3. Stained cells are incubated and fluorescence intensity measured over time. As a control, part of the stained population can be incubated at conditions under which receptor internalization is halted.
  • cell surface FLT3 can be detected and measured using a different antibody that is specific for FLT3 and that does not block or compete with binding of the antibody being tested. (Burtrum, et al., Cancer Res. 63:8912-21 (2003)). In one embodiment, treatment of an FLT3 expressing cell with an antibody of the present invention results in reduction of cell surface FLT3.
  • Another measure of down-modulation is reduction of the total receptor protein present in a cell, and reflects degradation of internal receptors. Accordingly, treatment of cells (particularly cancer cells) with antibodies of the invention results in a significant reduction in total cellular FLT3.
  • the antibodies of the present invention inhibit tumor growth.
  • subcutaneous xenograft tumors can be established by injection of cells of a cancer cell line into an immunodeficient mouse. The mice are then treated by intraperitoneal injection of antibodies and tumor size measured at regular intervals. Compared to control injections, antibodies of the invention inhibit tumor growth.
  • an antibody of the invention promotes tumor regression when combined with an anti-neoplastic agent.
  • antibodies of the invention promote tumor regression when used in a monotherapy. Promoting tumor regression means that administration of an effective amount of antibody, or an effective amount of a combination of an antibody and a neoplastic agent results in a reduction in size or necrosis of the tumor. Tumor regression may can be measured as an average across a group of subjects undergoing a particular treatment regimen, or can be measured by the number of subjects in a treatment group in which tumors regress.
  • the antibodies of the invention may be isolated or purified by any method known in the art, including precipitation by ammonium sulfate or sodium sulfate followed by dialysis against saline, ion exchange chromatography, affinity or immuno-affinity chromatography as well as gel filtration or zone electrophoresis.
  • a preferred method of purification for the antibodies of the current invention is Protein-A affinity chromatography.
  • DNA encoding human antibodies can be prepared by recombining DNA encoding human constant regions and variable regions, other than the CDRs, derived substantially or exclusively from the corresponding human antibody regions and DNA encoding CDRs derived from a human.
  • Suitable sources of DNA that encode fragments of antibodies include any cell, such as hybridomas and spleen cells that express the full-length antibody.
  • the fragments may be used by themselves as antibody equivalents, or may be recombined into equivalents, as described above.
  • the DNA deletion, recombination and other techniques described in this section may be carried out by known methods.
  • Another source of DNA is a phage display library of antibodies, as is known in the art.
  • the exemplified antibodies of the current invention were made via phage display technology.
  • the present invention provides expression vectors containing the polynucleotide sequences previously described operably linked to an expression sequence, a promoter and an enhancer sequence.
  • a variety of expression vectors for the efficient synthesis of antibody polypeptide in prokaryotic, such as bacteria and eukaryotic systems, including but not limited to yeast and mammalian cell culture systems have been developed.
  • the vectors of the present invention can comprise segments of chromosomal, non-chromosomal and synthetic DNA sequences.
  • prokaryotic cloning vectors include plasmids from E. coli , such as colE1, pCR1, pBR322, pMB9, pUC, pKSM, and RP4.
  • Prokaryotic vectors also include derivatives of phage DNA such as M13 and other filamentous single-stranded DNA phages.
  • An example of a vector useful in yeast is the 2 ⁇ , plasmid.
  • Suitable vectors for expression in mammalian cells include well-known derivatives of SV-40, adenovirus, retrovirus-derived DNA sequences and shuttle vectors derived from combination of functional mammalian vectors, such as those described above, and functional plasmids and phage DNA.
  • Additional eukaryotic expression vectors are known in the art (e.g., P. J. Southern and P. Berg, J. Mol. Appl. Genet. 1:327-41 (1982); Subramani et al., Mol. Cell. Biol. 1:854-64 (1981); Kaufmann and Sharp, J. Mol. Biol. 159:601-21 (1982); Kaufmann and Sharp, Mol. Cell. Biol. 159:601-64 (1982); Scahill et al., Proc. Nat'l Acad. Sci. 80:4654-59 (1983); Urlaub and Chasin, Proc. Nat'l Acad. Sci. 77:4216-20 (1980)).
  • the expression vectors useful in the present invention contain at least one expression control sequence that is operatively linked to the DNA sequence or fragment to be expressed.
  • the control sequence is inserted in the vector in order to control and to regulate the expression of the cloned DNA sequence.
  • useful expression control sequences are the lac system, the trp system, the tac system, the trc system, major operator and promoter regions of phage lambda, the control region of fd coat protein, the glycolytic promoters of yeast, e.g., the promoter for 3-phosphoglycerate kinase, the promoters of yeast acid phosphatase, e.g., Pho5, the promoters of the yeast alpha-mating factors, and promoters derived from polyoma, adenovirus, retrovirus, and simian virus, e.g., the early and late promoters or SV40, and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells and their
  • a suitable selection gene for use in yeast is the trp1 gene present in the yeast plasmid YRp7. Stinchcomb et al., Nature 282:39 (1979); Kingsman et al., Gene 7:141 (1979).
  • the trp1 gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1. Jones, Genetics 85:12 (1977).
  • the presence of the trp1 lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Leu2-deficient yeast strains (ATCC 20,622 or 38,626) are complemented by known plasmids bearing the Leu2 gene.
  • the present invention also provides recombinant host cells containing the expression vectors previously described. Antibodies of the present invention can be expressed in cell lines other than in hybridomas. Nucleic acids, which comprise a sequence encoding a polypeptide according to the invention, can be used for transformation of a suitable mammalian host cell.
  • Cell lines of particular preference are selected based on high level of expression, constitutive expression of protein of interest and minimal contamination from host proteins.
  • Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines, such as but not limited to, COS-7 cells, Chinese Hamster Ovary (CHO) cells, Baby Hamster Kidney (BHK) cells and many others including cell lines of lymphoid origin such as lymphoma, myeloma, or hybridoma cells.
  • Suitable additional eukaryotic cells include yeast and other fungi.
  • Useful prokaryotic hosts include, for example, E. coli , such as E. coli SG-936, E. coli HB 101, E. coli W3110, E. coli X1776, E. coli X2282, E. coli DHI, and E. coli MRC1, Pseudomonas, Bacillus , such as Bacillus subtilis , and Streptomyces.
  • These recombinant host cells can be used to produce an antibody, or fragment thereof, by culturing the cells under conditions permitting expression of the antibody or fragment thereof and purifying the antibody or fragment thereof from the host cell or medium surrounding the host cell.
  • Targeting of the expressed antibody or fragment for secretion in the recombinant host cells can be facilitated by inserting a signal or secretory leader peptide-encoding sequence (see, Shokri et al., Appl Microbiol Biotechnol. 60:654-64 (2003); Nielsen et al., Prot. Eng. 10:1-6 (1997); and von Heinje et al., Nucl. Acids Res. 14:4683-90 (1986)) at the 5′ end of the antibody-encoding gene of interest.
  • secretory leader peptide elements can be derived from either prokaryotic or eukaryotic sequences. Accordingly, suitable secretory leader peptides are used, being amino acids joined to the N-terminal end of a polypeptide to direct movement of the polypeptide out of the host cell cytosol and secretion into the medium.
  • the transformed host cells are cultured by methods known in the art in a liquid medium containing assimilable sources of carbon (carbohydrates such as glucose or lactose), nitrogen (amino acids, peptides, proteins or their degradation products such as peptones, ammonium salts or the like), and inorganic salts (sulfates, phosphates and/or carbonates of sodium, potassium, magnesium and calcium).
  • the medium furthermore contains, for example, growth-promoting substances, such as trace elements, for example iron, zinc, manganese and the like.
  • the antibodies of this invention can be fused to additional amino acid residues.
  • Such amino acid residues can be a peptide tag, perhaps to facilitate isolation.
  • Other amino acid residues for homing of the antibodies to specific organs or tissues are also contemplated.
  • Another embodiment for the preparation of antibodies in the present invention is the expression of the nucleic acid encoding the antibody according to the invention in a transgenic animal that has a substantial portion of the human antibody producing genome inserted and is rendered deficient in the production of endogenous antibodies.
  • Transgenic animals include but not limited to mice, goat, and rabbit.
  • One further embodiment of the invention includes expression of the antibody-coding gene in, for example, the mammary gland of the animal for secretion of the polypeptide during lactation.
  • high affinity anti-FLT3 antibodies according to the present invention can be isolated from a phage display library constructed from human heavy chain and light chain variable region genes.
  • a variable domain of the invention can be obtained from a peripheral blood lymphocyte that contains a rearranged variable region gene.
  • variable domain portions such as CDR and FW regions, can be derived from different human sequences. Over 90% of recovered clones after three rounds of selection are specific to FLT3.
  • the binding affinities for FLT3 of the screened Fabs can be in the nM range, which is as high as many bivalent anti-FLT3 monoclonal antibodies produced using hybridoma technology.
  • Antibodies of the present invention can be obtained, for example, from naturally occurring antibodies, or Fab or scFv phage display libraries.
  • Single domain antibodies can be obtained by selecting a VH or a VL domain from a naturally occurring antibody or hybridoma, or selected from a library of VH domains or a library of VL domains. It is understood that amino acid residues that are primary determinants of binding of single domain antibodies can be within Kabat or Chothia defined CDRs, but may include other residues as well, such as, for example, residues that would otherwise be buried in the VH-VL interface of a VH-VL heterodimer.
  • Antibodies of the present invention also include those for which binding characteristics have been improved by direct mutation, methods of affinity maturation, phage display, or chain shuffling. Affinity and specificity may be modified or improved by mutating CDRs and screening for antigen binding sites having the desired characteristics (see, e.g., Yang et al., J. Mol. Biol. 254:392-403 (1995)). CDRs are mutated in a variety of ways. One way is to randomize individual residues or combinations of residues so that in a population of otherwise identical antigen binding sites, all twenty amino acids are found at particular positions.
  • mutations are induced over a range of CDR residues by error-prone PCR methods (see, e.g., Hawkins et al., J. Mol. Biol. 226:889-896 (1992)).
  • phage display vectors containing heavy and light chain variable region genes may be propagated in mutator strains of E. coli (see, e.g., Low et al., J. Mol. Biol. 250: 359-368 (1996)).
  • the protein used to identify FLT3 binding antibodies of the invention is preferably FLT3 and, more preferably, is the extracellular domain of FLT3.
  • the FLT3 extracellular domain can be free or conjugated to another molecule.
  • the antibodies of this invention can be fused to additional amino acid residues.
  • Such amino acid residues can be a peptide tag, perhaps to facilitate isolation.
  • Other amino acid residues for homing of the antibodies to specific organs or tissues are also contemplated.
  • anti-FLT3 inhibitors including but not limited to antibodies of the invention
  • antibodies of the invention are efficiently internalized upon binding to cells bearing FLT3.
  • Anti-tumor agents administered in conjunction with, conjugated to, or linked to an antibody include any agents which destroy or damage a tumor to which the antibody has bound or in the environment of the cell to which the antibody has bound.
  • an anti-FLT3 inhibitor including the antibodies of the present invention, can be administered as a conjugate which binds specifically to the receptor and delivers a toxin following ligand-toxin internalization.
  • a FLT3 inhibitor-agent conjugate can be directly linked to each other or joined via a linker, peptide or non-peptide.
  • an anti-tumor agent is a toxic agent such as a chemotherapeutic agent or a radioisotope.
  • Suitable anti-neoplastic agents are known to those skilled in the art and include anthracyclines (e.g.
  • chemotherapeutic agents are conjugated to the inhibitor, antibody or small molecule using conventional methods (See, e.g., Hermentin and Seiler, Behring Inst. Mitt. 82:197-215 (1988)).
  • MTX is a preferred anti-neoplastic agent of the invention.
  • the invention further contemplates anti-FLT3 antibodies linked to target or reporter moieties, including by way of example only anti-neoplastic agents, other antibodies or reporters, such as radiolabled isotopes, in a diagnostic system where a detectable signal-producing agent is conjugated to the antibody.
  • Detectable signal-producing agents are useful in vivo and in vitro for diagnostic purposes.
  • the signal producing agent produces a measurable signal which is detectable by external means, usually the measurement of electromagnetic radiation.
  • the signal producing agent is an enzyme or chromophore, or emits light by fluorescence, phosphorescence or chemiluminescence.
  • Chromophores include dyes which absorb light in the ultraviolet or visible region, and can be substrates or degradation products of enzyme catalyzed reactions.
  • the invention further contemplates anti-FLT3 antibodies to which target or reporter moieties are linked.
  • Target moieties are first members of binding pairs.
  • Anti-neoplastic agents for example, are conjugated to second members of such pairs and are thereby directed to the site where the anti-FLT3 antibody is bound.
  • a common example of such a binding pair is avidin and biotin.
  • biotin is conjugated to an anti-FLT3 antibody, and thereby provides a target for an anti-neoplastic agent or other moiety, which is conjugated to avidin or streptavidin.
  • biotin or another such moiety is linked to an anti-FLT3 antibody of the invention and used as a reporter, for example in a diagnostic system where a detectable signal-producing agent is conjugated to avidin or streptavidin.
  • Suitable radioisotopes for use as anti-tumor agents are also known to those skilled in the art. For example, 131 I or 211 At is used. These isotopes are attached to the antibody using conventional techniques (See, e.g., Pedley et al., Br. J. Cancer 68:69-73 (1993)).
  • the anti-tumor agent which is attached to the antibody is an enzyme which activates a prodrug. In this way, a prodrug is administered which remains in its inactive form until it reaches the tumor site where it is converted to its cytotoxin form once the antibody complex is administered.
  • the antibody-enzyme conjugate is administered to the patient and allowed to localize in the region of the tissue to be treated.
  • the anti-tumor agent conjugated to the antibody is a cytokine such as interleukin-2 (IL-2), interleukin-4 (IL-4) or tumor necrosis factor alpha (TNF- ⁇ ).
  • IL-2 interleukin-2
  • IL-4 interleukin-4
  • TNF- ⁇ tumor necrosis factor alpha
  • the antibody targets the cytokine to the tumor so that the cytokine mediates damage to or destruction of the tumor without affecting other tissues.
  • the cytokine is fused to the antibody at the DNA level using conventional recombinant DNA techniques.
  • a method of treating tumor growth in a mammal by administering to the mammal an effective amount of an antibody as previously described is also provided by the present invention.
  • Suitable conditions to be treated according to the present invention involve cells preferably expressing FLT3. While not intended to be bound to any particular mechanism, the present methods provide for treatment of the growth of cancer cells including for example, those in which neoplastic growth, organ transplant rejection or an immune disorder such as an autoimmune disease which is stimulated by FLT3.
  • Treatment refers to therapeutic treatment including inhibiting, slowing, lessening or reversing the progress of the underlying condition or undesired physiological change associated with a disease or disorder, ameliorating clinical symptoms of a condition or preventing the appearance of clinical symptoms of the condition.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of the extent of a disease or disorder, stabilization of a disease or disorder (i.e., where the disease or disorder does not worsen), delay or slowing of the progression of a disease or disorder, amelioration or palliation of the disease or disorder, and remission (whether partial or total) of the disease or disorder, whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the disease.
  • the present invention can be used as a medicament.
  • One precancerous condition to be treated is myelodysplastic syndrome.
  • Other cancers to be treated include but are not limited to hematological malignancies such as leukemia, i.e., AML, ALL and CML in blast crisis, among others.
  • leukemias include those in Table 9, which lists the expression of FLT3 in selected human leukemia cell lines obtained from EB10 staining of selected leukemia cell lines.
  • the cancer may also be a solid tumor, such as a thyroid or brain tumor.
  • a therapeutically effective amount of an antibody of the invention is administered to a mammal in need thereof.
  • Effective doses of the compositions of the present invention, for treatment of disorders as described herein vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic.
  • the term administering as used herein means delivering the antibodies of the present invention to a mammal by any method that can achieve the result sought. They can be administered, for example, intravenously or intramuscularly.
  • human antibodies of the invention are particularly useful for administration to humans, they can be administered to other mammals as well.
  • therapeutically effective amount means an amount of antibody of the present invention that, when administered to a mammal, is effective in producing the desired therapeutic effect, such as inhibiting tumor growth. Treatment dosages may be titrated using routine methods known to those of skill in the art to optimize safety and efficacy.
  • compositions of the invention may include a “therapeutically effective amount” of an anti-FLT3 antibody of the invention.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the antibody may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects. Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response).
  • the present anti-FLT3 antibodies are administered for therapeutic treatments to a patient in need thereof in an amount sufficient to inhibit, or reduce the progression of the tumor or pathologic condition.
  • Progression includes, e.g., the growth, invasiveness, metastases and/or recurrence of the tumor or pathologic condition.
  • An amount adequate to accomplish this is defined as a therapeutically effective dose. Amounts effective for this use will depend upon the severity of the disease and the general state of the patient's own immune system. Dosing schedules will also vary with the disease state and status of the patient, and will typically range from a single bolus dosage or continuous infusion to multiple administrations per day (e.g., every 4-6 hours), or as indicated by the treating physician and the patient's condition.
  • An exemplary, non-limiting range for a therapeutically effective amount of an antibody of the invention is 0.1-50 mg/kg, more preferably 3-35 mg/kg, and more preferably 5-20 mg/kg.
  • Dosing amounts and frequencies will be determined by the physicians treating the patient and may include doses from less than 1 mg/kg to over 100 mg/kg given daily, three times per week, weekly, once every two weeks, or less often. Dose per administration may be in the range of 1-100, 2-75, or 5-60 mg/kg. It should be noted, however, that the present invention is not limited to any particular dose.
  • the invention also includes methods of inhibiting dendritic cell activation or maturation comprising contacting the dendritic cell with an antibody of the invention either alone or in combination with other agents.
  • Another method of the invention includes preventing organ transplant rejection or treating an autoimmune disease such as multiple sclerosis or encephalitis comprising administering the antibody of the invention alone or in combination with other agents.
  • anti-FLT3 antibodies can be administered in combination with one or more other anti-neoplastic agents.
  • combination therapies see, e.g., U.S. Pat. No. 6,217,866 (Schlessinger et al., Anti-EGFR antibodies in combination with anti-neoplastic agents); WO 99/60023 (Waksal et al., Anti-EGFR antibodies in combination with radiation).
  • Any suitable anti-neoplastic agent can be used, such as a chemotherapeutic agent, radiation or combinations thereof.
  • anti-neoplastic agents which are presently known in the art or being evaluated can be grouped into a variety of classes including, for example, mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, anti survival agents, biological response modifiers, anti-hormones, and anti-angiogenesis agents.
  • alkylating agents include, but are not limited to, cisplatin, cyclophosphamide, melphalan, and dacarbazine.
  • anti-metabolites include, but are not limited to, cytosine arabinoside, doxorubicin, daunorubicin, paclitaxel, gemcitabine, ALIMTA® and topoisomerase inhibitors irinotecan (CPT-11), aminocamptothecin, camptothecin, DX-8951f, topotecan (topoisomerase I), etoposide (VP-16), and teniposide (VM-26) (topoisomerase II).
  • the anti-neoplastic agent is radiation
  • the source of the radiation can be either external (external beam radiation therapy—EBRT) or internal (brachytherapy—BT) to the patient being treated.
  • EBRT internal beam radiation therapy
  • BT brachytherapy
  • the dose of anti-neoplastic agent administered depends on numerous factors, including, for example, the type of agent, the type and severity tumor being treated and the route of administration of the agent. It should be emphasized, however, that the present invention is not limited to
  • MTX is the preferred anti-neoplastic agent to be given in combination with an antibody of the invention.
  • Data provided herein demonstrate synergistic effects of the anti-FLT3 antibody, EB10, combined with MTX. This combination is particularly novel and unexpected given the in vitro work published by Furukawa et al. (Leukemia 21: 1005-1014 (2007)), which reported that simultaneous administration of a FLT3 inhibitor, PKC412, with other chemotherapeutic agents excluding MTX is clinically effective against FLT3 leukemia.
  • any suitable method or route can be used to administer anti-FLT3 antibodies of the invention, and optionally, to co-administer anti-neoplastic agents and/or antagonists of other receptors.
  • the anti-FLT3 antibody could be administered before, during, or after commencing therapy with another agent, including by of example only MTX, as well as any combination thereof, i.e., before and during, before and after, during and after, or before, during and after commencing the anti-neoplastic agent therapy.
  • the anti-FLT3 antibody can be administered between 1 and 30 days, preferably 3 and 20 days, more preferably between 5 and 12 days before commencing radiation therapy.
  • chemotherapy is administered concurrently with or, more preferably, subsequent to antibody therapy.
  • any FLT3 inhibitor can used in combination with MTX for the treatment of leukemia.
  • Anti-FLT3 antibodies of the invention can be administered with antibodies that neutralize other receptors involved in tumor growth or angiogenesis.
  • an anti-FLT3 antibody is used in combination with a receptor antagonist that binds specifically to EGFR.
  • Another example of such a receptor is VEGFR.
  • An anti-FLT3 antibody of the present invention can be used in combination with a VEGFR antagonist.
  • the FLT3 antibody can be administered in combination with one or more suitable adjuvants, such as, for example, cytokines (IL-10 and IL-13, for example) or other immune stimulators, such as, but not limited to, chemokine, tumor-associated antigens, and peptides.
  • these stimulators can be administered with MTX. It should be appreciated, however, that administration of an anti-FLT3 antibody alone, as a monotherapy, is sufficient to prevent, inhibit, or reduce the progression of the tumor in a therapeutically effective manner.
  • any suitable method or route can be used to administer anti-FLT3 antibodies of the invention, and optionally, to co-administer anti-neoplastic agents such as MTX and/or antagonists of other receptors.
  • the anti-neoplastic agent regimens utilized according to the invention include any regimen believed to be optimally suitable for the treatment of the patient's neoplastic condition. Different malignancies, including various forms of leukemia can require the use of specific anti-tumor antibodies and specific anti-neoplastic agents, which will be determined on a patient to patient basis.
  • Routes of administration include, for example, oral, intravenous, intraperitoneal, subcutaneous, intathecal, or intramuscular administration.
  • the dose of antagonist administered depends on numerous factors, including, for example, the type of antagonists, the type and severity tumor being treated and the route of administration of the antagonists. It should be emphasized, however, that the present invention is not limited to any particular method or route of administration.
  • the anti-FLT3 antibodies of the invention where used in a mammal for the purpose of prophylaxis or treatment, will be administered in the form of a composition additionally comprising a pharmaceutically acceptable carrier.
  • suitable pharmaceutically acceptable carriers include, for example, one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • Pharmaceutically acceptable carriers can further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the binding proteins.
  • the compositions of the injection can, as is well known in the art, be formulated so as to provide rapid, sustained or delayed release of the active ingredient after administration to the mammal.
  • kits for inhibiting tumor growth and/or angiogenesis comprising a therapeutically effective amount of a human anti-FLT3 antibody.
  • kits for inhibiting tumor growth and/or angiogenesis comprising a therapeutically effective amount of a human anti-FLT3 antibody with MTX.
  • the kits can further contain any suitable antagonist of, for example, another growth factor receptor involved in tumorigenesis or angiogenesis (e.g., EGFR, VEGFR-1/Flt-1, VEGFR-2, PDGFR, NGFR, and FGFR).
  • the kits of the present invention can further comprise an anti-neoplastic agent. Examples of suitable anti-neoplastic agents in the context of the present invention have been described herein.
  • the kits of the present invention can further comprise an adjuvant, examples of which have been described above.
  • kits which contain antibodies of the present invention.
  • the antibodies of this invention bind to FLT3 with a binding strength stronger than that of FLT3 ligand binding activity, i.e., with a K D of about 200 ⁇ 10 ⁇ 12 M to 500 ⁇ 10 ⁇ 12 M.
  • K D for antibody binding to a human FLT3-Fc fusion protein is from 0.5 ⁇ 10 ⁇ 10 M to 5 ⁇ 10 ⁇ 10 M determined at 25° C.; preferred antibodies bind to human FLT3-Fc fusion protein with a K D from, 1.0 ⁇ 10 ⁇ 10 M to 4.75 ⁇ 10 ⁇ 10 M, 1.5 ⁇ 10 ⁇ 10 M to 4.5 ⁇ 10 ⁇ 10 M, or no greater than 4.5 ⁇ 10 ⁇ 10 M, all determined at 25° C.
  • the antibody binds to FLT3 with a dissociation rate constant (K d or k off ) between 4.5 ⁇ 10 ⁇ 5 1/s (sec ⁇ 1 , 1/seconds) and 6 ⁇ 10 ⁇ 5 1/s, 5.0 ⁇ 10 ⁇ 5 1/s and 5.7 ⁇ 10 ⁇ 5 1/s, all as measured by surface plasmon resonance, described herein, at 25° C. and more preferably that antibody binds to FLT3 with a K d or k off between 5.1 ⁇ 10 ⁇ 5 1/s and 5.6 ⁇ 10 ⁇ 5 1/s, or within 10% of these rate constants.
  • K d or k off dissociation rate constant
  • the antibody binds to FLT3 with an association rate constant (K a or k on ) between 0.5 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 (1/Ms; 1/molar 1/seconds) and 5 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 , or 1 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 and 4 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 all as measured by surface plasmon resonance, described herein, at 25° C. and more preferably that antibody binds to FLT3 with a K a or k on between 1.2 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 and 3.6 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 , or within 10% of these rate constants.
  • K a or k on between 0.5 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 (1/Ms; 1/molar 1/seconds) and 5 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 , or 1 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 and 4 ⁇ 10 5 M ⁇ 1 sec ⁇ 1 all as
  • the antibody binds to FLT3 with a dissociation rate constant, as measured by surface plasmon resonance at 25° C., that is within 10% of the dissociation rate constant determined for EB10, NC7, or D4-3 under the same conditions.
  • the present invention comprises a monoclonal antibody, or fragment thereof, specific for FLT3 comprising one or more CDRs selected from the group consisting of the CDRs in Tables 1 and 2.
  • the invention is a monoclonal antibody, or fragment thereof, specific for FLT3 having a light chain CDR3 region with the sequence: MQGTHPAIS (SEQ ID NO:9).
  • the invention is a monoclonal antibody, or fragment thereof, specific for FLT3 having a heavy chain CDR3 with the sequence: GVGAHDAFDI (SEQ ID NO:4).
  • the invention is a monoclonal antibody, or fragment thereof, comprising (i) a light chain variable region selected from the group consisting of EB10, NC7, and D4-3 and (ii) a heavy chain variable region selected from the group consisting of EB10, NC7, and D4-3.
  • the invention is a monoclonal antibody, or fragment thereof, specific for FLT3 comprising (i) a light chain variable region of EB10, NC7, and D4-3 (ii) a heavy chain variable region of EB10, NC7, and D4-3, and (iii) human immunoglobulin G 1 (hIgG 1 ) constant regions.
  • One aspect of the invention is an antibody, or fragment thereof, which binds an epitope of FLT3, wherein the epitope comprises up to five extracellular domains (D1, D2, D3, D4 and D5), and at a minimum comprises either or both domains D4 or D5.
  • the epitope is D4.
  • the epitope is D5.
  • the antibody or antibody fragment inhibits phosphorylation of a downstream pathway of FLT3 including STAT5, Akt, PI3K and MAPK.
  • the presently disclosed antibodies are immunoconjugated to an antineoplastic agent including auristatin or methotrexate.
  • the immunoconjugate can be linked to a detectable label.
  • Another aspect is a method of detecting by contacting a target cell sample with the antibody or antibody fragment and determining if the sample contains FLT3 by detecting the labeled antibody.
  • the therapeutic composition is effective to inhibit growth of neoplastic cells that express FLT3 or promoting regression of human tumors that express FLT3.
  • the therapeutic compositions are the presently disclosed antibodies and a pharmaceutically acceptable carrier.
  • Another aspect of the present invention is a method of neutralizing the activation of FLT3 within a mammal by administering to the mammal an effective amount of the antibody or antibody fragment thereof.
  • Yet another aspect is a method of inhibiting dendritic cell activation or maturation comprising contacting the dendritic cell with the antibody or fragment presently disclosed in an amount effective to inhibit dendritic cell activation or maturation.
  • Another aspect of this invention is a method of treating cancer in a mammal comprising administering to the mammal an effective amount of an antibody, or fragment thereof, of any of the aspects already described.
  • the invention also provides a method to treat hematological malignancies including leukemia.
  • leukemia includes, but is not limited to: acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), chronic myeloid leukemia blast crisis (CML in blast crisis) and myelodysplastic syndrome.
  • AML acute myeloid leukemia
  • ALL acute lymphocytic leukemia
  • CML in blast crisis chronic myeloid leukemia blast crisis
  • myelodysplastic syndrome myelodysplastic syndrome.
  • Another treatment method provided by this invention combines using the antibodies or fragments thereof of this invention along with administering an additional anti-cancer agent or treatment.
  • the anti-cancer agent is methotrexate (MXT).
  • the present receptor antibodies thus can be used in vivo and in vitro for investigative, diagnostic, prophylactic, or treatment methods, which are well known in the art.
  • Variations in the principles of invention herein disclosed can be made by one skilled in the art and it is intended that such modifications are to be included within the scope of the present invention.
  • a suitable heavy chain nucleotide sequence for example SEQ ID NOs 37, 38, or 39 (for EB10, NC7 and D4-3 respectively) into a suitable expression plasmid, for example pGSHC
  • a suitable light chain nucleotide sequence for example SEQ ID No. 40, 41, or 42 (for EB10, NC7 and D4-3 respectively) into a suitable expression plasmid, such as pGSLC, by a suitable method such as PCR cloning.
  • a suitable host cell line such as NSO or CHO cells
  • linearized heavy and light chain plasmids by electroporation and culture in suitable media such as glutamine free Dulbecco's Modified Eagle Medium with dialyzed fetal calf serum and glutamine synthetase supplement.
  • suitable media such as glutamine free Dulbecco's Modified Eagle Medium with dialyzed fetal calf serum and glutamine synthetase supplement.
  • ELISA enzyme-linked immunosorbent assay
  • Purify antibodies by a suitable method such as protein-A affinity chromatography.
  • the present invention includes the recombinant human monoclonal antibody EB10, a full length IgG1 ⁇ targeting the human FLT3 receptor. It is comprised of a human gamma-1 heavy chain of subgroup I and a human kappa light chain of subgroup II.
  • the EB10 Fab was isolated from a human Fab phage display library by selection for high affinity binding to the human FLT3 receptor and its ability to block ligand binding to the receptor. EB10 was shown to selectively bind to human FLT3 with high affinity, block FL binding and mediated potent anti-tumor activity in xenograft models by a mechanism involving activation of immune effector function.
  • the present invention also includes the recombinant human monoclonal antibody NC7, a full length IgG1 ⁇ targeting the human FLT3 receptor. It is comprised of a human gamma-1 heavy chain of subgroup I and a human kappa light chain of subgroup II. NC7 was shown to selectively bind to human FLT3 with high affinity and block FL binding.
  • the present invention also includes the recombinant human monoclonal antibody D4-3, a full length IgG1 ⁇ targeting the human FLT3 receptor. It is comprised of a human gamma-1 heavy chain of subgroup I and a human kappa light chain of subgroup II. D4-3 was shown to selectively bind to human FLT3 with high affinity and block FL binding.
  • Tables 1 and 2 provide the amino acid sequences and SEQ ID NOs of the various CDRs of the present invention. All CDR sequences are determined using the Kabat convention except for SEQ ID NOs 1 and 12, which were determined using the Chothia convention.
  • Table 3 provides the SEQ ID NOs of the various sequences related to the present invention. Polynucleic acid sequences that encode the amino acid sequences disclosed below are also included within the scope of the present invention.
  • Antibodies used in experiments comprised full-length heavy and light chains without signals, as given in Table 4.
  • Generate a set of FLT3 ECD variants by serially deleting the Ig-like domains of the FLT3 ECD using a PCR-based strategy.
  • Fd1 amino acid 24-183
  • Fd1-2 amino acid 24-271
  • Fd1-3 amino acid 24-370
  • Fd1-4 amino acid 24-451
  • As the positive control produce the full-length FLT3 (Fd1-5) containing amino acid 24-541 in a similar manner.
  • ligand binding assay coat the plates with FL (100 ng/well) overnight at 4° C., followed by incubation with the FLT3 ECD Ig domain deletion variants for 1 hour with gentle shaking. After 3 washes, incubate the plates with 100 ⁇ l/well of HRP-conjugated anti-human Fc antibody (Jackson ImmunoResearch Labs Inc.). Develop the plates as described above. Examine binding of FL to the mutant receptors.
  • FLT3 binding assay coat a 96-well plate was coated with an anti-His antibody (Qiagen, Hilden, Germany) overnight at 4° C. Block wells for 1 hour with blocking buffer (PBS containing 0.1% TWEEN-20® and 5% fetal calf serum (FCS)) and then incubate with FLT3-Fc (1 ⁇ g/ml ⁇ 100 ⁇ l/well) for 1 hour at room temperature. Wash wells three times with PBST and then add EB10 or control IgG and incubated at room temperature for 1 hour. After washing, incubate the plate with 100 ⁇ l of anti-human kappa chain antibody-HRP conjugate at room temperature for 1 hour. Wash the plates and then incubate with 100 ⁇ l of 3,3′,5,5′-tetra-methylbenzidine. Use the microplate reader at the absorbance at 450 nm.
  • blocking buffer PBS containing 0.1% TWEEN-20® and 5% fetal calf serum (FCS)
  • the receptor-ligand blocking assay For the receptor-ligand blocking assay, mix varying amounts of EB10 or control IgG with a fixed amount of biotinated FLT3-Fc fusion protein (45 ng/well) and incubate at room temperature for 1 hour. Transfer the mixture to 96-well plates precoated with FL (25 ng/well) and then incubate at room temperature for an additional hour. After washing, add streptavidin HRP conjugate, and read the absorbance at 450 nm. Calculate the antibody concentration required for 50% inhibition of FLT3 binding to FL (IC 50 ).
  • EB10 binds FLT3 with an EC 50 within the range of 0.5-1 nM.
  • EB10 The ability of EB10 to block receptor-ligand binding was examined in a FL binding competition ELISA. As shown in Table 7, EB10 blocked the binding of FL to FLT3 with an IC 50 within the range of 0.5-1 nM while no blocking activity was seen for control IgG.
  • Binding Kinetics Analysis by Surface Plasmon Resonance/BiacoreTM Analysis Measure the binding kinetics of the antibody to FLT3 at 25° C. using the surface plasmon resonance, for example a Biacore 2000TM biosensor (Pharmacia Biosensor, Uppsala, Sweden) Immobilize approximately 1000 RU (reference units) of the antibody onto a sensor chip, and inject soluble FLT3-Fc fusion protein at concentrations ranging from 1.5-100 nM. Obtain sensorgrams at each concentration and evaluate using the instrument's program, for example BIA Evaluation 2.0 program, to determine the rate constants k on and k off . K d , also referred to as k off , is the rate constant of the dissociation reaction.
  • K a also referred to as k on
  • K D is the rate constant of the association reaction.
  • the affinity constant (K D ) is calculated from the ratio of rate constants k on :k off measured in Molar (M).
  • K a , the K d , and K D for the antibodies exemplified herein, EB10, NC7, and D4-3, are summarized in Table 8.
  • Table 9 lists the results of EB10 staining of selected leukemia cell lines. Flow cytometric analysis showed that EB10 bound to wild-type FLT3 expressed on EOL-1 cells, and also to ITD-mutant FLT3 expressed on BaF3-ITD cells. No binding for EB10 was observed on the FLT3-negative JM1 or BaF3-control cell lines.
  • EB10 Inhibits FL-Induced Phosphorylation of Wild-Type FLT3 and Ligand-Independent Constitutive Phosphorylation of ITD-Mutant FLT3: In EOL-1 and EM3 cells, FL addition strongly increased FLT3 receptor phosphorylation. Incubation with EB10 blocked FL-induced phosphorylation in a dose-dependent manner with an IC 50 of 0.4 ⁇ 4 nM. These results indicate that EB10 is a potent inhibitor of ligand-induced activation of wild-type FLT3.
  • the ITD mutation found with high frequency in AML is known to cause FL-independent receptor phosphorylation and activation of kinase signaling pathways.
  • EB10's inhibitory effect on constitutive activation of mutant FLT3 using BaF3-ITD and MV4; 11 cell lines is reported in Table 10.
  • the mutant FLT3 in both BaF3-ITD and MV4; 11 cell lines was constitutively phosphorylated.
  • EB10 inhibited FL-independent FLT3-ITD phosphorylation in BaF3-ITD cells.
  • EB10 also significantly inhibited FL-independent FLT3-ITD phosphorylation in MV4; 11 cells.
  • these results demonstrate that EB10 is also a potent inhibitor of FLT3-ITD kinase activity.
  • EB10 Inhibits FLT3-Mediated Activation of Downstream Kinases:
  • the MAPK, PI3K and Stat5 pathways have been identified to be involved in the downstream signaling of activated FLT3 (Stirewalt D L and J P, Radich, J P. Nat Rev Cancer 3:650-665 (2003)).
  • the antibodies of the present invention inhibit downstream kinases of MPK, PI3K, and STAT5 pathways in leukemia.
  • FL plays an important role in the proliferation of leukemia cells. Incubation with FL increased the [3H]-thymidine uptake of EOL-1 cells. Treatment with EB10 inhibited FL-induced proliferation of EOL-1 cells in a dose-dependent manner (Table 14).
  • EB10 Inhibits Proliferation of Leukemia Cells Expressing ITD-Mutant FLT3: FLT3-ITD transformed BaF3 cells proliferate in the absence of FL stimulation. FL stimulation did not increase the proliferation of BaF3-ITD cells. EB10 treatment inhibited FL-independent proliferation of these cells in a dose-dependent manner (Table 15).
  • Radio-iodinate antibodies with 125 I using IODO-beads (Pierce Biotechnology, Rockford, Ill., USA) according to the manufacturer's instructions. Aliquot EOL-1 cells into microfuge tubes at 5 ⁇ 10 5 cells/sample in 500 ml cold complete media. Add approximately 1 mg of EB10 and D4-3 125 I-labeled antibody to the cells and incubate for 1 hour at 4° C. Wash cells twice in cold PBS, resuspend in 500 ml complete media, then incubate at 4 or 37° C. for 0, 30, 60, 120, 240, or 360 minutes. At each time point, wash cells three times in PBS, then count the 4° C.
  • ADCC Antibody Dependent Cellular Cytotoxicity
  • ADCC assays by the standard 51 Cr release assay.
  • NK human natural killer cells
  • normal donor blood such as from RosettaSep NK Cell Enrichment Cocktail; StemCell Technologies, Inc., Vancouver, British Columbia, Canada, or NK Cell Isolation Kit II, Miltenyi Biotec.
  • NK Cell Isolation Kit II Miltenyi Biotec.
  • NK cells (0 to 400,000 cells), which were previously incubated with 10 ⁇ g/mL (67 nmol/L) control IgG, C225, EB10, NC7 or D4-3 mAb for 45 minutes, with ⁇ 4,000 51 Cr-labeled target cells, in triplicate in V-bottomed 96-well plates for 6 hours at 37° C., or with 5% SDS to measure total lysis.
  • incubate targets cells (4,000 cells in 100 ⁇ /well) with 100 ⁇ l medium only.
  • antibodies to FLT3 may initiate patient immune responses towards FLT3 expressing tumor cells.
  • antibodies can induce cell lysis if FLT3 expressing cells are coated with human IgG. This lysis can be the result of the activation of leukocytes, in particular Fc receptor expressing natural killer (NK) cells.
  • the antibodies of the present invention provide human anti-FLT3 antibodies which have an improved ability to activate downstream immune effector functions such as ADCC. EB10 induced a strong ADCC response; NC7 and D4-3 induced a less potent ADCC response (Table 17).
  • EOL-1 Xenograft Human AML Leukemia Model Intravenously (i.v.) inject NOD-SCID mice in groups of 10 with 5 ⁇ 10 6 leukemia cells. Start treatment one day after tumor injection. Treat mice three times weekly with an intraperitoneal (i.p.) injection of indicated amounts of 500 ⁇ g, 250 ⁇ g or 100 ⁇ g/dose of EB10 in 200 ⁇ l phosphate buffered saline (PBS). Treat the control group with purified human IgG (500 ⁇ g/dose). Monitor mice daily for survival. Compare survival in the treatment groups by Log Rank test.
  • PBS phosphate buffered saline
  • mice in the control group succumbed to extensive dissemination of disease within 40 days (mean survival time 36.0 ⁇ 3.1 days).
  • survival was significantly prolonged in groups of mice treated with 500 ⁇ g, 250 ⁇ g or 100 ⁇ g/dose of EB10 (mean survival time 62.3 ⁇ 18.7, 55.3 ⁇ 18.9, or 52.8 ⁇ 18.6, with a P value of ⁇ 0.001, ⁇ 0.005, or ⁇ 0.001, respectively).
  • No effect was observed in the group treated with 10 ⁇ g of EB10, indicating that the anti-leukemic effect of EB10 was dose-dependent.
  • Bone Marrow Engraftment of Human Leukemia Cells Harvest bone marrow from the femurs of EB10-treated mice (500 ⁇ g/dose) and the control-IgG treated group at day 20. Analyze cells for human CD45 by flow cytometry at 6 and 14 weeks post leukemia cell injection for the degree of tumor cell infiltration of bone marrow. Compare the degree of tumor cell infiltration of bone marrow using immunohistochemical staining with a fluorescence-labeled anti-human CD45 antibody. The number of tumor cells in bone marrow was decreased significantly in EB10-treated mice.
  • BaF3-ITD Leukemia Model Initiate i.v. injection of groups of 10 athymic (nu/nu) mice with 5 ⁇ 10 4 BaF3-ITD cells. For statistic analysis, use the non-parametric one-tailed Mann-Whitney Rank Sum test (SigmaStat 2.03, SPSS, Inc., Chicago, Ill.). Start treatment one day after tumor injection. Treat mice three times weekly with an i.p. injection of 500 ⁇ g/dose or 100 ⁇ g/dose of EB10 in 200 ⁇ l PBS. Treat the control group with purified human IgG (500 ⁇ g/dose). Monitor mice daily for survival.
  • MOLM14 Xenograft AML Leukemia Model Irradiate mice with 200 rads with a gamma irradiator and then i.v. inject with 10 ⁇ 10 6 MOLM-14 leukemia cells. Start treatment one day after tumor injection. Treat mice two times weekly with an i.p. injection of 10 mg/kg and 0.2 mg/kg doses of EB10 in 200 ⁇ l PBS. Treat the control group with 10 ⁇ g/ml USP saline. Monitor mice daily for survival. Compare survival in the treatment groups by Log Rank test.
  • EB10 significantly prolonged survival. Median survivals ranged from 63 days to 38.5 days for the 10 mg/kg and 0.2 mg/kg doses, respectively, compared to 36 days for the saline control. This effect was dose dependent, with each dose being significantly more efficacious than the next lower dose.
  • SEM-K2 Xenograft Human Leukemia Model Initiate i.v. injection of NOD-SCID mice in groups of 10 with 5 ⁇ 10 6 leukemia cells. Start treatment one day after tumor injection. Treat mice two times weekly with an i.p. injection of 20 mg/kg of EB10, NC7 and D4-3 in 200 ⁇ l PBS. Treat the control group with purified human IgG (500 ⁇ g/dose). Monitor mice daily for survival. Compare survival in the treatment groups by Log Rank test.
  • EB10 was shown to be efficacious at very low doses.
  • the threshold for maximum efficacy was found to be between 2 and 10 ⁇ g/ml.
  • EB10 achieves maximal efficacy in murine leukemia models between 41 and 401 ⁇ g/ml Cavg, and between 49 and 475 ⁇ g/ml Cmax.
  • a secondary range for Cmax is 375 to 475 ⁇ g/ml.
  • NOD/SCID mice One day prior to the start of treatment, i.v. inject 65, 7 weeks old, male NOD/SCID mice with 5 ⁇ 10 5 SEM-K2 cells suspended in PBS in a total volume of 200 ⁇ l. Drop from the study mice receiving less than 90% of cells intravenously. The following day, divide the NOD-SCID mice bearing SEM-K2 leukemia cells into 4 treatment groups of 12 mice/group:
  • Combination therapy with both EB10 and MTX had enhanced efficacy (P ⁇ 0.001) in comparison to monotherapies.
  • the combination of EB10 and MTX resulted in a significantly longer median survival than any other treatments in this model.
  • four mice in the combination group survived to the end of the study, day 168, whereas no mice receiving other treatments survived to that point.
  • the fact that combination treatment allowed some mice to survive beyond 150 days of treatment, whereas neither EB10 nor MTX as monotherapy were able to achieve this longevity demonstrates that the combination of EB10 and MTX is synergistic. This result is particularly unexpected given the in vitro work published by Furukawa et al. (Leukemia 21:1005-1014 (2007)), which reported that simultaneous administration of a FLT3 inhibitor, PKC412, with other chemotherapeutic agents excluding MTX is clinically effective against FLT3 leukemia.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US12/994,956 2008-05-30 2009-05-28 Anti-flt3 antibodies Abandoned US20110091470A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/994,956 US20110091470A1 (en) 2008-05-30 2009-05-28 Anti-flt3 antibodies

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US13039408P 2008-05-30 2008-05-30
US13039508P 2008-05-30 2008-05-30
US13053908P 2008-05-30 2008-05-30
US12/994,956 US20110091470A1 (en) 2008-05-30 2009-05-28 Anti-flt3 antibodies
PCT/US2009/045365 WO2009155015A1 (en) 2008-05-30 2009-05-28 Anti-flt3 antibodies

Publications (1)

Publication Number Publication Date
US20110091470A1 true US20110091470A1 (en) 2011-04-21

Family

ID=41092183

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/994,956 Abandoned US20110091470A1 (en) 2008-05-30 2009-05-28 Anti-flt3 antibodies
US12/473,295 Abandoned US20090297529A1 (en) 2008-05-30 2009-05-28 Anti-flt3 antibodies
US12/890,793 Expired - Fee Related US8071099B2 (en) 2008-05-30 2010-09-27 Anti-FLT3 antibodies

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/473,295 Abandoned US20090297529A1 (en) 2008-05-30 2009-05-28 Anti-flt3 antibodies
US12/890,793 Expired - Fee Related US8071099B2 (en) 2008-05-30 2010-09-27 Anti-FLT3 antibodies

Country Status (18)

Country Link
US (3) US20110091470A1 (es)
EP (1) EP2300500A1 (es)
JP (1) JP2011521647A (es)
KR (1) KR20110004455A (es)
CN (1) CN102046659A (es)
AR (1) AR071891A1 (es)
AU (1) AU2009260517B2 (es)
BR (1) BRPI0912173A2 (es)
CA (1) CA2726522A1 (es)
CL (1) CL2009001277A1 (es)
EA (1) EA201071421A1 (es)
IL (1) IL209142A0 (es)
MX (1) MX2010013144A (es)
NZ (1) NZ588791A (es)
PE (1) PE20091963A1 (es)
TW (1) TWI374032B (es)
WO (1) WO2009155015A1 (es)
ZA (1) ZA201008024B (es)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017053889A3 (en) * 2015-09-23 2017-05-26 Precision Immunotherapy, Inc. Flt3 directed car cells for immunotherapy
WO2021212069A1 (en) 2020-04-17 2021-10-21 City Of Hope Flt3-targeted chimeric antigen receptor modified cells for treatment of flt3-positive malignancies
WO2021209495A1 (en) 2020-04-14 2021-10-21 Symphogen A/S Anti-flt3 antibodies and compositions

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR071891A1 (es) * 2008-05-30 2010-07-21 Imclone Llc Anticuerpos humanos anti-flt3 (receptor tirosina cinasa 3 tipo fms humano)
HUE045270T2 (hu) 2010-01-05 2019-12-30 Inst Nat Sante Rech Med FLT3 receptor anatgonisták fájdalom rendellenességek kezelésére
US20130156764A1 (en) * 2010-03-12 2013-06-20 The Johns Hopkins University Neutralization of flt3 ligand as a leukemia therapy
EA201790757A1 (ru) * 2011-09-22 2017-07-31 Эмджен Инк. Связывающие антиген cd27l белки
US20150064203A1 (en) * 2012-03-08 2015-03-05 Japan Science And Technology Agency Anticancer agent
EP3119423B1 (en) 2014-03-15 2022-12-14 Novartis AG Treatment of cancer using chimeric antigen receptor
JP2017528433A (ja) 2014-07-21 2017-09-28 ノバルティス アーゲー 低い免疫増強用量のmTOR阻害剤とCARの組み合わせ
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
WO2016145099A1 (en) * 2015-03-09 2016-09-15 Agensys, Inc. Antibody drug conjugates (adc) that bind to flt3 proteins
US20180298068A1 (en) 2015-04-23 2018-10-18 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
CN108603200B (zh) 2015-11-23 2022-08-19 诺华股份有限公司 优化的慢病毒转移载体及其用途
MX2018008106A (es) 2015-12-30 2019-03-14 Novartis Ag Terapias con celulas inmunoefectoras de una eficacia mejorada.
WO2017165683A1 (en) 2016-03-23 2017-09-28 Novartis Ag Cell secreted minibodies and uses thereof
EP3464366A1 (en) * 2016-05-27 2019-04-10 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Flt3-specific chimeric antigen receptors and methods using same
KR20230066647A (ko) 2016-06-17 2023-05-16 마젠타 테라퓨틱스 인코포레이티드 Cd117+ 세포의 고갈을 위한 조성물 및 방법
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
CA3045902A1 (en) 2016-12-21 2018-06-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies specific for flt3 and uses thereof
CN110461876B (zh) 2017-01-20 2024-05-17 海德堡医药研究有限责任公司 用于耗尽cd137+细胞的组合物和方法
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
JP2020506700A (ja) 2017-01-31 2020-03-05 ノバルティス アーゲー 多重特異性を有するキメラt細胞受容体タンパク質を用いた癌の治療
EP3589647A1 (en) 2017-02-28 2020-01-08 Novartis AG Shp inhibitor compositions and uses for chimeric antigen receptor therapy
AU2018269678A1 (en) 2017-05-17 2019-12-12 Biodol Therapeutics FLT3 inhibitors for improving pain treatments by opioids
TWI757499B (zh) * 2017-06-02 2022-03-11 美商輝瑞大藥廠 對flt3具特異性之抗體及其用途
US20230181634A1 (en) * 2017-06-02 2023-06-15 Pfizer Inc. Chimeric antigen receptors targeting flt3
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
SG11202000632QA (en) * 2017-07-31 2020-02-27 Dragonfly Therapeutics Inc Proteins binding nkg2d, cd16 and flt3
EP3700926A1 (en) 2017-10-25 2020-09-02 Novartis AG Methods of making chimeric antigen receptor-expressing cells
EP3700933A1 (en) 2017-10-25 2020-09-02 Novartis AG Antibodies targeting cd32b and methods of use thereof
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
US20210396739A1 (en) 2018-05-01 2021-12-23 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
EP3801769A1 (en) 2018-05-25 2021-04-14 Novartis AG Combination therapy with chimeric antigen receptor (car) therapies
US20210123075A1 (en) 2018-06-08 2021-04-29 Novartis Ag Compositions and methods for immunooncology
AR116109A1 (es) 2018-07-10 2021-03-31 Novartis Ag Derivados de 3-(5-amino-1-oxoisoindolin-2-il)piperidina-2,6-diona y usos de los mismos
WO2020128638A1 (en) * 2018-12-18 2020-06-25 Northern Biologics Inc. Flt3 agonist antibodies and uses thereof
JP2022514315A (ja) 2018-12-20 2022-02-10 ノバルティス アーゲー 3-(1-オキソイソインドリン-2-イル)ピペリジン-2,6-ジオン誘導体を含む投与計画及び薬剤組み合わせ
JP7488826B2 (ja) 2019-02-15 2024-05-22 ノバルティス アーゲー 置換3-(1-オキソイソインドリン-2-イル)ピペリジン-2,6-ジオン誘導体及びその使用
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020210719A1 (en) * 2019-04-10 2020-10-15 Elevatebio Management, Inc. Flt3-specific chimeric antigen receptors and methods of using the same
SG11202111130SA (en) * 2019-04-30 2021-11-29 Senti Biosciences Inc Chimeric receptors and methods of use thereof
TW202112812A (zh) * 2019-05-24 2021-04-01 香港商安立璽榮生醫(香港)有限公司 Csf1r抗體、il10融合蛋白及其用途
AU2020366000A1 (en) * 2019-10-15 2022-05-12 Dragonfly Therapeutics, Inc. Antibodies targeting FLT3 and use thereof
US20230056470A1 (en) 2019-12-20 2023-02-23 Novartis Ag Uses of anti-tgf-beta antibodies and checkpoint inhibitors for the treatment of proliferative diseases
EP4142749A1 (en) 2020-04-30 2023-03-08 The Regents of the University of Colorado, A Body Corporate Multispecific anti-flt3 chimeric antigen receptors
JP2023529211A (ja) 2020-06-11 2023-07-07 ノバルティス アーゲー Zbtb32阻害剤及びその使用
CN115916199A (zh) 2020-06-23 2023-04-04 诺华股份有限公司 包含3-(1-氧代异吲哚啉-2-基)哌啶-2,6-二酮衍生物的给药方案
CN111808821B (zh) * 2020-06-24 2022-06-14 南方医科大学珠江医院 Flt3-nkg2d双靶点car-t的构建与制备
CN116134027A (zh) 2020-08-03 2023-05-16 诺华股份有限公司 杂芳基取代的3-(1-氧代异吲哚啉-2-基)哌啶-2,6-二酮衍生物及其用途
IL302700A (en) 2020-11-13 2023-07-01 Novartis Ag Combined treatments with cells expressing chimeric antigens (vehicle)
TW202304979A (zh) 2021-04-07 2023-02-01 瑞士商諾華公司 抗TGFβ抗體及其他治療劑用於治療增殖性疾病之用途
AR125468A1 (es) 2021-04-27 2023-07-19 Novartis Ag Sistema de producción de vectores virales
AU2022330106A1 (en) * 2021-08-16 2024-03-21 Hemogenyx Pharmaceuticals Llc Anti-flt3 antibodies, cars, car t cells and methods of use
WO2023105087A1 (en) * 2021-12-10 2023-06-15 Tubulis Gmbh Novel flt3 antibodies and antibody-drug-conjugates based thereon, therapeutic methods and uses thereof in combination with tyrosine kinase inhibitors
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2024089639A1 (en) 2022-10-26 2024-05-02 Novartis Ag Lentiviral formulations

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5548065A (en) * 1991-04-02 1996-08-20 The Trustees Of Princeton University Tyrosine kinase receptor human flk-2-specific antibodies
US5635388A (en) * 1994-04-04 1997-06-03 Genentech, Inc. Agonist antibodies against the flk2/flt3 receptor and uses thereof
US5777084A (en) * 1996-03-07 1998-07-07 Eberhard-Karls-Universitat Tubingen Antibody BV10A4H2 specific for human FLT3/FLK2 receptor and mybridoma
US6156882A (en) * 1997-06-30 2000-12-05 Eberhard-Karls-Universitat Tubingen Antibody 4G8B4B12
US6217866B1 (en) * 1988-09-15 2001-04-17 Rhone-Poulenc Rorer International (Holdings), Inc. Monoclonal antibodies specific to human epidermal growth factor receptor and therapeutic methods employing same
US7537932B1 (en) * 1993-05-19 2009-05-26 Schering Corporation Antibodies that bind purified mammalian FLT3 ligands
US20090297529A1 (en) * 2008-05-30 2009-12-03 Yiwen Li Anti-flt3 antibodies
US7679666B2 (en) * 1999-07-22 2010-03-16 Minolta Co., Ltd. Solid-state logarithmic image sensing device

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3869584D1 (de) 1987-04-03 1992-04-30 Hermann Hofmann Verfahren zum kompostieren von organischen stoffen und vorrichtung zur durchfuehrung dieses verfahrens.
AU612370B2 (en) 1987-05-21 1991-07-11 Micromet Ag Targeted multifunctional proteins
WO1989009622A1 (en) 1988-04-15 1989-10-19 Protein Design Labs, Inc. Il-2 receptor-specific chimeric antibodies
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5185438A (en) 1991-04-02 1993-02-09 The Trustees Of Princeton University Nucleic acids encoding hencatoporetic stem cell receptor flk-2
US5885793A (en) 1991-12-02 1999-03-23 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
WO1993021319A1 (en) 1992-04-08 1993-10-28 Cetus Oncology Corporation HUMANIZED C-erbB-2 SPECIFIC ANTIBODIES
US5554512A (en) 1993-05-24 1996-09-10 Immunex Corporation Ligands for flt3 receptors
JPH09502352A (ja) 1993-09-08 1997-03-11 イムクローン システムズ インコーポレイテッド Flk−2レセプターを認識するモノクローナル抗体および原始造血幹細胞集団の単離
US6084060A (en) 1996-12-09 2000-07-04 Imclone Systems Incorporated Composition and method for preserving progenitor cells
JP2002515511A (ja) 1998-05-15 2002-05-28 イムクローン システムズ インコーポレイティド 放射線及び成長因子レセプター・チロシン・キナーゼのインヒビターを使用するヒト腫瘍の治療
AU2002329540A1 (en) 2001-06-20 2003-01-02 Morphosys Ag Antibodies that block receptor protein tyrosine kinase activation, methods of screening for and uses thereof
WO2005094823A1 (ja) 2004-03-30 2005-10-13 Kyowa Hakko Kogyo Co., Ltd. Flt-3阻害剤
US8227455B2 (en) 2005-04-18 2012-07-24 Rigel Pharmaceuticals, Inc. Methods of treating cell proliferative disorders
WO2008153926A2 (en) 2007-06-05 2008-12-18 Yale University Inhibitors of receptor tyrosine kinases and methods of use thereof

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US6217866B1 (en) * 1988-09-15 2001-04-17 Rhone-Poulenc Rorer International (Holdings), Inc. Monoclonal antibodies specific to human epidermal growth factor receptor and therapeutic methods employing same
US5548065A (en) * 1991-04-02 1996-08-20 The Trustees Of Princeton University Tyrosine kinase receptor human flk-2-specific antibodies
US7537932B1 (en) * 1993-05-19 2009-05-26 Schering Corporation Antibodies that bind purified mammalian FLT3 ligands
US5635388A (en) * 1994-04-04 1997-06-03 Genentech, Inc. Agonist antibodies against the flk2/flt3 receptor and uses thereof
US5777084A (en) * 1996-03-07 1998-07-07 Eberhard-Karls-Universitat Tubingen Antibody BV10A4H2 specific for human FLT3/FLK2 receptor and mybridoma
US6156882A (en) * 1997-06-30 2000-12-05 Eberhard-Karls-Universitat Tubingen Antibody 4G8B4B12
US7679666B2 (en) * 1999-07-22 2010-03-16 Minolta Co., Ltd. Solid-state logarithmic image sensing device
US20090297529A1 (en) * 2008-05-30 2009-12-03 Yiwen Li Anti-flt3 antibodies
US20110008355A1 (en) * 2008-05-30 2011-01-13 Imclone Llc Anti-flt3 antibodies
US8071099B2 (en) * 2008-05-30 2011-12-06 ImClone, LLC Anti-FLT3 antibodies

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017053889A3 (en) * 2015-09-23 2017-05-26 Precision Immunotherapy, Inc. Flt3 directed car cells for immunotherapy
CN109310744A (zh) * 2015-09-23 2019-02-05 赛通免疫有限责任公司 用于免疫治疗的flt3定向car细胞
US10961312B2 (en) 2015-09-23 2021-03-30 Cytoimmune Therapeutics, Inc. FLT3 directed car cells for immunotherapy
WO2021209495A1 (en) 2020-04-14 2021-10-21 Symphogen A/S Anti-flt3 antibodies and compositions
WO2021212069A1 (en) 2020-04-17 2021-10-21 City Of Hope Flt3-targeted chimeric antigen receptor modified cells for treatment of flt3-positive malignancies

Also Published As

Publication number Publication date
AU2009260517B2 (en) 2012-12-06
US8071099B2 (en) 2011-12-06
US20090297529A1 (en) 2009-12-03
WO2009155015A1 (en) 2009-12-23
AU2009260517A1 (en) 2009-12-23
AR071891A1 (es) 2010-07-21
IL209142A0 (en) 2011-01-31
KR20110004455A (ko) 2011-01-13
ZA201008024B (en) 2012-04-25
PE20091963A1 (es) 2010-01-15
CA2726522A1 (en) 2009-12-23
JP2011521647A (ja) 2011-07-28
MX2010013144A (es) 2010-12-21
TW201010722A (en) 2010-03-16
EP2300500A1 (en) 2011-03-30
BRPI0912173A2 (pt) 2016-07-26
CN102046659A (zh) 2011-05-04
US20110008355A1 (en) 2011-01-13
NZ588791A (en) 2012-05-25
EA201071421A1 (ru) 2011-08-30
TWI374032B (en) 2012-10-11
CL2009001277A1 (es) 2010-10-08

Similar Documents

Publication Publication Date Title
US8071099B2 (en) Anti-FLT3 antibodies
JP7222024B2 (ja) 上皮増殖因子受容体変異体iiiおよびcd3の単一および二重特異性抗体およびそれらの使用
CA2795081C (en) Antibodies against csf-1r
KR102638104B1 (ko) 이특이적 her2 및 cd3 결합 분자
CA2524305C (en) Fully human antibodies directed against the human insulin-like growth factor-1 receptor
JP5587589B2 (ja) 併用療法
RU2733315C2 (ru) Комбинированная терапия для лечения злокачественной опухоли
US7951370B2 (en) Anti-TYRP1 antibodies
US8071730B2 (en) Anti-JAM-A antibodies
JP2018503365A (ja) 抗pd−1抗体およびその使用方法
CA2589374A1 (en) Antibodies directed to gpnmb and uses thereof
CN115697419A (zh) 多聚体抗dr5结合分子与癌症疗法组合治疗癌症的用途
WO2024074498A1 (en) Combination of a btn3a activating antibody, a bcl2 inhibitor and hypomethylating agent for use in treating cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: IMCLONE LLC, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, YIWEN;LU, DAN;SURGULADZE, DAVID;AND OTHERS;SIGNING DATES FROM 20101029 TO 20101106;REEL/FRAME:025430/0088

STCB Information on status: application discontinuation

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