WO2004039842A2 - Humanized tissue factor antibodies - Google Patents

Humanized tissue factor antibodies Download PDF

Info

Publication number
WO2004039842A2
WO2004039842A2 PCT/DK2003/000741 DK0300741W WO2004039842A2 WO 2004039842 A2 WO2004039842 A2 WO 2004039842A2 DK 0300741 W DK0300741 W DK 0300741W WO 2004039842 A2 WO2004039842 A2 WO 2004039842A2
Authority
WO
WIPO (PCT)
Prior art keywords
value
human
rfviia
ffr
antibodies
Prior art date
Application number
PCT/DK2003/000741
Other languages
French (fr)
Other versions
WO2004039842A3 (en
Inventor
Ivan Svendsen
Kristian Kjaergaard
Stefan Zahn
Original Assignee
Novo Nordisk A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novo Nordisk A/S filed Critical Novo Nordisk A/S
Priority to AU2003277832A priority Critical patent/AU2003277832A1/en
Publication of WO2004039842A2 publication Critical patent/WO2004039842A2/en
Publication of WO2004039842A3 publication Critical patent/WO2004039842A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/36Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1018Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1093Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody conjugates with carriers being antibodies
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/86Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving blood coagulating time or factors, or their receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/745Assays involving non-enzymic blood coagulation factors
    • G01N2333/7454Tissue factor (tissue thromboplastin, Factor III)

Definitions

  • the present invention relates to isolated antibodies that immunoreacts with tissue factor (TF) to inhibit the binding of coagulation factor Vila (FVIIa) and thus a novel immuno- therapeutic method using humanized antibodies against TF to inhibit thrombus formation associated with surgery, microsurgery, angioplasty or trauma or to inhibit thrombus formation and other functions of TF in abnormal haemostatic conditions associated with a disease (such as deep vein thrombosis, disseminated intravascular coagulation (DIC), coronary ar- tery disease, sepsis, inflammation, atherosclerosis, or cancer). Also disclosed are a method for preparation of antibodies as well as cell lines for preparation of the humanized antibodies (abs).
  • TF tissue factor
  • FVIIa coagulation factor Vila
  • Blood coagulation is a process consisting of a complex interaction of various blood components, or factors, which eventually gives rise to a fibrin clot.
  • the blood components which participate in what has been referred to as the coagulation "cascade” are pro- enzymes or zymogens, enzymatically inactive proteins which are converted to proteolytic enzymes by the action of an activator, itself an activated clotting factor.
  • Coagulation factors that have undergone such a conversion and generally referred to as “active factors,” and are des- ignated by the addition of a lower case “a” suffix (e.g., Factor Vila).
  • Activated Factor X (“Xa”) is required to convert prothrombin to thrombin, which then converts fibrinogen to fibrin as a final stage in forming a fibrin clot.
  • the "intrinsic pathway” refers to those reactions that lead to thrombin formation through utilization of factors present only in plasma.
  • a series of protease-mediated activations ultimately generates Factor IXa which, in conjunction with Factor Villa, cleaves Factor X into Xa.
  • An identical proteolysis is effected by FVIIa and its co-factor, TF, in the "extrinsic pathway” of blood coagulation.
  • TF is a membrane bound protein and does not normally circulate in an active form in plasma. Upon vessel disruption, however, TF can complex with FVIIa to catalyze Factor X activation or Factor IX activation in the presence of Ca 2+ and phospholipid. While the relative importance of the two coagulation pathways in hemostasis is unclear, Factor VII and TF have been found to play a pivotal role in the initiation of blood coagulation.
  • Anticoagulants such as heparin, coumarin, derivatives of coumarin, indandione derivatives, or other agents may be used, for example, during kidney dialysis, or to treat deep vein thrombosis, disseminated intravascular coagulation (DIC), and a host of other medical disorders.
  • heparin treatment or extracorporeal treatment with citrate ion may be used in dialysis to prevent coagulation during the course of treatment.
  • Heparin is also used in preventing deep vein thrombosis in patients undergoing surgery.
  • heparin and other anticoagulants may, however, have undesirable side effects.
  • Available anticoagulants generally act throughout the body, rather than acting specifically at site of injury. Heparin, for example, may cause heavy bleeding. Furthermore, with a half-life of approximately 80 minutes, heparin is rapidly cleared from the blood, necessitating frequent administration. Because heparin acts as a cofactor for antithrombin III (ATIII), and ATIII is rapidly depleted in DIC treatment, it is often difficult to maintain the proper heparin dosage, necessitating continuous monitoring of ATIII and heparin levels. Heparin is also ineffective if ATIII depletion is extreme.
  • ATIII antithrombin III
  • heparin may also increase platelet aggregation and reduce platelet count, and has been implicated in the development of heparin-induced thrombocytopenia. Indandione derivatives may also have toxic side effects.
  • anticoagulants briefly described above, several naturally occurring proteins have been found to have anticoagulant activity. Also, ATIII has been proposed as a therapeutic anticoagulant.
  • International Application No. WO 92/15686 relates to inactivated Factor Vila for inhibiting blood coagulation.
  • Antibodies are specific immunoglobulin (Ig) polypeptides produced by the vertebrate immune system in response to challenges by foreign proteins, glycoproteins, cells, or other antigenic foreign substances.
  • Ig immunoglobulin
  • the sequence of events which permits the organism to overcome invasion by foreign cells or to rid the system of foreign substances is at least partially understood.
  • An important part of this process is the manufacture of antibodies which bind specifically to a particular foreign substance.
  • the binding specificity of such polypeptides to a particular antigen is highly refined, and the multitude of specificities capable of being generated by the individual vertebrate is remarkable in its complexity and variability. Millions of antigens are capable of eliciting antibody responses, each antibody almost exclusively directed to the particular antigen which elicited it.
  • Antibodies are generated in situ as a result of the differentiation of immature B lymphocytes into plasma cells, which occurs in response to stimulation by specific antigens.
  • the undifferentiated B cells the portions of DNA coding for the various regions on the immunoglobulin chains are separated in the genomic DNA. The sequences are assembled sequentially prior to expression. The resulting rearranged gene is capable of expression in the mature B lymphocyte to produce the desired antibody.
  • a uniform population of antibodies does not result.
  • the in situ immune response to any particular antigen is defined by the mosaic of responses to the various determinants which are present on the antigen.
  • Each subset of homologous antibodies is contributed by a single population of B cells, hence in situ generation of antibodies is "polyclonal”.
  • the relatively short-lived, or mortal, splenocytes or lymphocytes from a mammal which has been injected with antigen are fused with an immortal tumor cell line, thus producing hybrid cells or "hybridomas" which are both immortal and capable of producing the genetically coded antibody of the B cell.
  • the hybrids thus formed are segregated into single genetic strains by selection, dilution, and regrowth, and each strain thus represents a single genetic line. They therefore, produce antibodies which are assured to be homogeneous against a desired antigen. These antibodies, referencing their pure genetic parentage, are called "monoclonal".
  • Monoclonal antibodies with mono-specificity have greatly influenced immunology, and their usefulness has already been demonstrated in such sciences as biology, pharmacology, biochemistry and others. Such monoclonal antibodies have found widespread use not only as diagnostics reagents, but also therapeutically (see, for example, Ritz and Schlossman, Blood, 59:1-11 , (1982)). Monoclonal antibodies produced by hybridomas, while theoretically effective as discussed above and clearly preferable to polyclonal antibodies because of their specificity, suffer from an important disadvantage. In many applications, the use of monoclonal antibodies produced in non-human animals is severely restricted where the monoclonal antibodies are to be used in humans. Repeated injections of a "foreign" antibody in humans, such as a mouse antibody, may lead to harmful hypersensitivity reactions. Such a non-human derived monoclonal antibody, when injected into humans, causes an anti-nonhuman antibody response.
  • European patent application No. 833911 relates to CDR-grafted antibodies against human TF.
  • Presta L. et al., Thrombosis and Haemostasis, Vol. 85 (3) pp. 379-389 (2001) relates to humanized antibody against TF inhibiting plasma clotting and TF/FVI la-mediated activation of factors IX and X.
  • the present invention fulfils this need by providing anticoagulants that do not have the side effects associated with the traditional antibodies with non-human sequences, they act specifically at sites of injury, and further provides other related advantages.
  • the present invention provides compounds, which acts to inhibit the cellular functions of TF, which is implicated in conditions like sepsis, inflammation, atherosclerosis, restenosis, or cancer.
  • the invention provides non-immunogenic high affinity humanized antibodies against human TF, which antibodies inhibit the binding of coagulation factor Vll Vlla.
  • the invention provides novel and useful methods for selection of therapeutically effective humanized antibodies against human TF.
  • the present invention relates to an isolated humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
  • the invention relates to methods of using these and other TF antibodies, methods of producing such antibodies, cells for producing such antibodies, and related compositions.
  • humanized antibodies and “humanized antibody”, as used herein, refer to recombinant antibodies in which complementarity determining region (CDR) sequences or variants thereof derived from the germline of another mammalian species, such as a mouse, have been grafted into human framework sequences, e.g. the so-called humanized antibodies.
  • CDR complementarity determining region
  • the isolated humanized monoclonal antibody comprises CDR amino acid sequences derived from a monoclonal antibody.
  • the isolated humanized monoclonal antibody has CDR amino acid sequences derived from a mouse monoclonal antibody.
  • Antibodies are normally synthesized by lymphoid cells derived from B lymphocytes of bone marrow. Lymphocytes derived from the same clone produce immunoglobulin of a single amino acid sequence. Lymphocytes can not be directly cultured over long periods of time to produce substantial amounts of their specific antibody.
  • lymphocyte tumor cells which, depending upon the cell strain, frequently produce an antibody themselves, although "non-producing" strains are known.
  • recombinant antibody is intended to include all 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 (described further in Section II, below), antibodies isolated from a recombinant combinatorial antibody library (described further in Section III, below), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain aspects of the invention, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies may be sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • the terms "human tissue factor” or "human TF” as used herein, refers to the full length polypeptide receptor comprising the amino acid sequence 1-263 of native human tissue factor.
  • antibody refers to immunoglobulin molecules and fragments thereof, that have the ability to specifically bind to an antigen (e.g., human TF).
  • Full-length antibodies comprises four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • 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, and FR4.
  • an antigen e.g., human TF
  • binding fragments encompassed within the term "antibody” include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH I domains; (ii) F(ab) 2 and F(ab') 2 fragments, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (see e.g.
  • VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426: and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • scFv single chain Fv
  • single chain antibodies are also intended to be encompassed within the term "antibody”.
  • Other forms of single chain antibodies, such as diabodies are also encompassed by the term “antibody.”.
  • Diabodies are bivalent and bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but are separated by a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain so as to create two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123).
  • human TF may have one or more antigenic determinants comprising (1) peptide antigenic determinants which consist of single peptide chains within human TF, (2) conformational antigenic determinants which consist of more than one spatially contiguous peptide chains whose respective amino acid sequences are located disjointedly along the human TF polypeptide sequence; and (3) post-translational antigenic determinants which consist, either in whole or part, of molecular structures covalently attached to human TF after translation, such as carbohydrate groups.
  • antigenic determinants comprising (1) peptide antigenic determinants which consist of single peptide chains within human TF, (2) conformational antigenic determinants which consist of more than one spatially contiguous peptide chains whose respective amino acid sequences are located disjointedly along the human TF polypeptide sequence; and (3) post-translational antigenic determinants which consist, either in whole or part, of molecular structures covalently attached to human TF after translation, such as carbohydrate groups.
  • an "isolated humanized antibody”, as used herein, refers to a humanized antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds human TF is substantially free of antibodies that specifically bind antigens other than human TF).
  • An isolated antibody that specifically binds human TF may, however, have cross-reactivity to other antigens, such as TF molecules from other species (discussed in further detail below).
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • epipe as used herein means any antigenic determinant on an antigen to which the antibody binds.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • immunosorbents or “immunoreacting”, as used herein, means any binding of an antibody to its epitope with a dissociation constant K d lower than 10 "4 M.
  • an antibody that "inhibits" the binding of human coagulation factor Vila to human TF means any antibody, that reduces the ability of human coagulation factor Vila to bind human TF compared to the ability of human coagulation factor Vila to bind human TF in the absense of the antibody.
  • affinity means the strength of the binding of an antibody to an epitope.
  • the affinity of an antibody is measured by the dissociation constant K d , defined as [Ab] x [Ag] / [Ab-Ag] where [Ab-Ag] is the molar concentration of the antibody-antigen complex, [Ab] is the molar concentration of the unbound antibody and [Ag] is the molar concentration of the unbound antigen.
  • K d dissociation constant
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
  • a therapheutically effective amount is the effective dose to be determined by a qualified practitioner, who may titrate dosages to achieve the desired response.
  • Factors for consideration of dose will include potency, bioavailability, desired pharmacokinetic/pharmacodynamic profiles, condition of treatment (e.g. trauma, inflammation, septic chock), patient-related factors (e.g. weight, health, age, etc.), presence of co-administered medications, time of administration, and/or other factors known to a medical practitioner.
  • the dosage of a humanized antibody against TF administered to a patient will vary with the type and severity of the condition to be treated, but is generally in the range of 0.1 -5.0 mg/kg body weight.
  • the term "subject” as used herein means any animal, in particular mammals, such as humans, and may, where appropriate, be used interchangeably with the term “patient”.
  • the invention relates to a composition comprising a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
  • the invention relates to a method for treatment of a FVlla/TF related disorder in a human, which method comprises administering to the human a therapeutically effective amount of a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
  • Treatment means the administration of an amount of antibodies of the invention and/or a compound of the invention with the effect of reducing the intensity of, delaying the onset of, and/or reducing the risk of, and preferably preventing, the occurrence of one or more physiological symptoms associated with a disease state or reducing and/or eliminating one or more detectable physiological conditions or symptoms associated with a disease state present in a subject (and, preferably, eliminating all conditions associated with the disease state).
  • Humanized TF antibodies and compositions provided by the invention can be used in any and all aspects of treating a subject.
  • FVlla/TF related disorder means a disease or disorder, where TF and FVIIa are involved.
  • FVlla/TF related disorders include thrombotic and/or coagulopathic related diseases, and disorders including inflammatory response and chronic thromboembolic diseases, and disorders associated with fibrin formation including vascular disorders such as deep venous thrombosis, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary an- gioplastry (PTCA), stroke, tumor growth, tumor metastasis, angiogenesis-related disease(s), thrombolysis, atherosclerosis, arteriosclerosis e.g.
  • arteriosclerosis following angioplastry and restenosis (such as restenosis following angioplastry), acute and chronic indications such as inflammation, septic chock, septicemia, hypotension, adult respiratory distress syndrome (ARDS), disseminated intravascular coagulopathy (DIC), pulmonary embolism, platelet deposition, myocardial infarction, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis, and other diseases or disorders.
  • the FVlla/TF related disorder is not limited to in vivo coagulopatic disorders such as those named above but includes ex vivo FVllaTF related processes such as coagulation that may result from the ex- tracorporeal circulation of blood, including blood removed in-line from a patient in such processes as dialysis procedures, blood filtration, or blood bypass during surgery.
  • the term "Factor Vila”, or “FVIIa” means "two chain” activated coagulation factor VII cleaved by specific cleavage at the Arg152-lle153 peptide bond. FVIIa, may be purified from blood or produced by recombinant means.
  • FVII means "single chain” coagulation factor VII.
  • the invention relates to a method for preparation of a humanized antibody, which method comprises preparation of humanized antibodies against human TF, testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC 50 value lower than the IC 50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC 50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC 50 value of FFR-rFVIIa, or testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding, or testing antibodies in a FVIIa signalling assay and selecting humanized antibodies which inhibits
  • FVIIa-induced intracellular signalling means the intracellular events mediated when FVIIa is added to the cell.
  • FVIIa-induced intracellular signalling may be measured by the MAPK signalling assay or a gene expression analysis assay which meas- ures the up-regulation of genes independently selected from the list comprising Fra-1 , Id2, and Cyr61.
  • TF-induced clot assay means any assay where clotting time is measured in sample comprising blood plasma and TF.
  • TF-induced clot assay means any assay where clotting time is measured in sample comprising blood plasma and TF.
  • An example of a TF-induced clot assay is described in Example 1 , assay 7.
  • FXa generation assay means any assay where activation of FX is measured in a sample comprising TF, FVIIa, FX, calcium and phospholipids.
  • FXa generation assay is described in Example 1 , assay 5.
  • FVlla/TF amidolytic assay means any assay where the amidolytic activity, i.e. cleavage of a small peptide substrate, of FVIIa is measured in the presence of TF.
  • An example of a FVlla/TF amidolytic assay is described in Example 1 , assay 4.
  • TF ELISA assay means any ELISA assay comprising TF and antibodies against TF. Examples of TF ELISA assays are the direct and indirect TF ELISA assays described in Example 1 , assay 1 and 2.
  • direct TF ELISA assay as used herein means any TF ELISA assay comprising immobilized TF. An example of a direct TF ELISA assays is described in Example 1 , assay 1.
  • the term "indirect TF ELISA assay” as used herein means any TF ELISA assay, where TF is in solution.
  • Example of direct TF ELISA assays is described in Example 1 , assay 2.
  • the invention relates to a humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF and are produced by a method that comprises : preparation of humanized antibodies against human TF, testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC 50 value lower than the IC 50 value of
  • FFR-rFVIIa + 100 nM (using 10 nM FVIIa), such as lower than the IC 50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC-50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC 50 value of FFR-rFVIIa, or testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding, or testing antibodies in a FVIIa signalling assay and selecting humanized antibodies which inhibits FVIIa-induced intracellular signalling.
  • the invention relates to a method for preparation of a humanized anti- body, which method comprises a) preparation of immortal cells to secrete humanized antibodies, b) isolation of culture medium from immortal cells comprising produced humanized antibodies, c) testing antibodies in an indirect TF ELISA assay comprising TF in solution and se- lecting humanized antibodies that immunoreacts with human TF in solution, d) testing antibodies in a FVIIa competition assay and selecting humanized antibodies which competes with FVIIa binding, e) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC 50 value lower than the IC 50 value of FFR-rFVIIa + 100 nM (using 10 nm FVIIa in the assay), such as lower than the IC 50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC 50 value of FFR-rFVIIa +
  • antibody-producing cell means any cell capable of producing an antibody. Included are hybridomas, transfected cell lines and the relatively short- lived, or mortal, splenocytes or lymphocytes from a mammal which has been injected with an antigen.
  • the invention relates to a humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF and are produced by a method that comprises : a) preparation of immortal cells to secrete humanized antibodies, b) isolation of culture medium from immortal cells comprising produced antibodies, c) testing antibodies in an indirect TF ELISA assay comprising TF in solution and selecting humanized antibodies that immunoreacts with human TF in solution, d) testing antibodies in a FVIIa competition assay and selecting humanized antibodies which competes with FVIIa binding, e) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC 50 value lower than the IC 5 o value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC 50 value of FFR-r
  • the invention relates to a cell producing humanized antibodies that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
  • the cell is a mammal cell.
  • the cell is isolated from a mouse.
  • the cell is a hybridoma cell.
  • the hybridoma cell is obtained by fusion of an anti- body-producing lymphoid cell with an immortal cell to provide an antibody-producing hybridoma cell.
  • the cell is a myeloma cell.
  • myeloma cell lines may be used for the production of humanized antibodies in accordance with the techniques described herein, including, e.g., Mouse myeloma NSO (ECACC 85110503), rat myeloma YB2/0 (ATCC CRL 1662), P3X63-Ag8, P3X63-AG8.653, P3/NS1-Ag4-1 (NS-1), Sp2/0-Agl4 and S194/5.XXO.Bu.1.
  • the P3X63-Ag8 and NS-1 cell lines have been described by Kohler and Milstein (Eur. J. Immunol. 6:511 (1976)). Shulman et al.
  • humanized or human antibodies are developed by phage display, ribo- some display, RNA display, surface display on a "living organism” or by using transgenic animals or in immune incompetent animals where human immune competent cells are introduced. Common for the mentioned display techniques are that the humanized antibodies produced by these techniques have affinities that are below what is normally needed for therapeutic antibodies. One has to affinity mature these humanized antibodies further.
  • the invention relates to an isolated humanized antibody, that immunoreacts with an epitope P present on a protein, wherein the human framework amino acid sequences of said humanized antibody are derived from a human antibody, wherein said human antibody immunoreacts with an epitope H present on said protein, said epitope H comprising an amino acid residue at a particular position of said protein, which amino acid residue at this particular position of said protein is also comprised within the epi- tope P with which said humanized antibody immunoreacts.
  • this protein may be any protein.
  • the protein may be a therapeutic target protein.
  • framework amino acid sequence and “framework sequence” individually refer to an amino acid sequence of an antibody that is not a CDR sequence.
  • a framework sequence that is derived from a parent antibody consists of the amino acids present in the corresponding non-CDR sequence of the parent antibody from which such sequences are considered to be derived. Unless otherwise stated or clearly contradicted by context, any framework sequence incorporated in any antibody described herein can be substituted with a suitable framework sequence variant.
  • a framework sequence variant can be any sequence of amino acids that (a) has similar biological property as an antibody produced by a mammal in response to an antigen of interest (e.g., permits immunogenic binding of the antibody of which it is a part to its target, such as TF, preferably with an affinity that is at least about as great, if not greater, than its mammalian-produced counterpart and desirably allows the antibody to retain a similar biological effect as its mammalian-produced counterpart - e.g., a similar level of inhibition of TF binding to FVIIa) and (b) has a high level of amino acid sequence identity with its mammalian-produced counterpart (e.g., has at least about 80%, at least about 85%, at least about 90%, or at least about 95% identity to).
  • an antigen of interest e.g., permits immunogenic binding of the antibody of which it is a part to its target, such as TF, preferably with an affinity that is at least about as great, if not greater, than its ma
  • a framework sequence variant will vary by only a few amino acids from its mammalian-produced counterpart (e.g., a variant may differ by 3, 2, or 1 substitutions, deletions, and/or additions) and/or will vary only in terms of conservative amino acid substitutions.
  • a framework sequence variant will consist essentially of the same sequence as the framework sequence of its mammalian-produced counterpart.
  • the CDR amino acid sequences of the isolated humanized antibody are derived from a parent monoclonal antibody.
  • the parent monoclonal antibody is a mouse monoclonal antibody.
  • the CDR amino acid sequences is variant CDR amino acid sequences relative to the CDR amino acid sequences of a parent monoclonal antibody.
  • the terms "variant”, as used herein with respect to CDR amino acid sequences, designates, designates CDR amino acid sequences, wherein one or more amino acids have been substituted by another amino acid and/or wherein one or more amino acids have been deleted and/or wherein one or more amino acids have been inserted into the CDR amino acid sequence.
  • the "variant" within this definition still have CDR functionality.
  • the variant CDR amino acid sequences is 70 % identical with the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences is 80 % identical with the CDR sequences of the parent mono- clonal antibody. In one embodiment the variant CDR amino acid sequences is 90 % identical with the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences is 95 % identical with the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences has one amino acid substitution relative to the CDR sequences of the parent monoclonal antibody.
  • the variant CDR amino acid sequences has two amino acid substitutions rela- tive to the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences has three amino acid substitutions relative to the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences has four amino acid substitutions relative to the CDR sequences of the parent monoclonal antibody. In one embodiment of the invention the isolated humanized antibody is a Fab fragment.
  • the isolated humanized antibody is a F(ab) 2 fragment.
  • the isolated humanized antibody is a F(ab') 2 fragment.
  • the isolated humanized antibody is a single chain Fv fragment.
  • the isolated humanized antibody has human framework amino acid sequences derived from a human antibody, that immunoreacts with a second epitope present on human TF.
  • this second epitope comprises an amino acid residue at a particular position of human TF, which amino acid residue at this particular position of human TF is also comprised within the first epitope with which said humanized antibody immunoreacts.
  • first epitope comprises an amino acid residue at a particular position of human TF selected from the list consisting of Trp45, Lys46 and Tyr94.
  • second epitope comprises an amino acid residue at a particular position of human TF selected from the list consisting of Trp45, Lys46 and Tyr94.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • 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 and in particular CDR3.
  • 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, e.g. the so-called humanized antibodies.
  • the isolated humanized antibody has a K d for binding to human TF within the range of 10 "15 - 10 "8 M. It is to be understood, that the K d for humanized antibody binding to human TF referred to is as determined in an assay, wherein the humanized antibody is immobilized (see assay 6).
  • the isolated humanized antibody has a K d for binding to human TF within the range of 10 "15 - 10 "10 M.
  • the isolated humanized antibody has a K d for binding to human TF within the range of 10 "15 - 10 "12 M.
  • the isolated humanized antibody has a K d for binding to human TF lower than 10 "8 M. In a further embodiment of the invention the isolated humanized antibody has a K for binding to human TF lower than 10 "9 M In a further embodiment of the invention the isolated humanized antibody has a K d for binding to human TF lower than 10 "10 M. In a further embodiment of the invention the isolated humanized antibody has a K d for binding to human TF lower than 10 "11 M. In a further embodiment of the invention the isolated humanized antibody has a K d for binding to human TF lower than 10 "12 M. In a further embodiment of the invention the isolated humanized antibody has a K d for binding to human TF lower than 10 "13 M.
  • the isolated humanized antibody has a K d for binding to human TF lower than 10 "14 M. In a further embodiment of the invention the isolated humanized antibody has a K d for binding to human TF lower than 10 "15 M.
  • the method for preparation of a humanized antibody comprises testing antibodies in a TF-induced clot assay and selecting humanized antibody which inhibit clot formation in this assay with an IC 50 value lower than 1 nM.
  • the IC 50 value is lower than 500 pM.
  • the IC 50 value is lower than 200 pM.
  • the IC 50 value is lower than 100 pM.
  • the IC 50 value is lower than 50 pM.
  • the IC 50 value is lower than 10 pM.
  • the IC 50 value is lower than 5 pM.
  • the method for preparation of a humanized antibody comprises testing antibodies in a TF-induced clot assay and selecting humanized antibody which inhibit clot formation in this assay with an IC 50 value lower than the IC 50 value of FFR-rFVIIa + 1 nM, such as lower than the IC 50 value of FFR-rFVIIa + 500 pM, preferably lower than the IC 50 value of FFR-rFVIIa + 200 pM, preferably lower than the IC 50 value of FFR-rFVIIa + 100 pM, such as lower than the IC 50 value of FFR-rFVIIa + 50 pM, preferably lower than the IC 50 value of FFR-rFVIIa + 10 pM, more preferably lower than the IC 50 value of FFR-rFVIIa + 5 pM, more preferably lower than the IC 50 value of FFR-rFVIIa.
  • the specific IC 50 values referred to in the TF-induced clot assay is when using normal human plasma.
  • the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC 50 value lower than 100 nM (in an assay with a FVIIa concentration of 0.1 nM)
  • the IC 50 value is lower than 10 nM.
  • the IC 50 value is lower than 5 nM.
  • the IC 50 value is lower than 1 nM .
  • the IC 50 value is lower than 0.1 nM.
  • the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC 50 value lower than the IC 50 value of FFR- rFVIIa + 100 nM (using 0.1 nM FVIIa in the assay), such as lower than the IC 50 value of FFR- rFVIIa + 10 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 5 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 1 nM, more preferably lower than the IC 50 value of FFR- rFVIIa + 0.1 nM, more preferably lower than the IC 50 value of FFR-rFVIIa.
  • the method for preparation of a humanized antibody comprises testing antibodies in a FVlla/TF amidolytic assay and selecting human- ized antibody which inhibit TF-induced FVIIa amidolytic activity with an IC 50 value lower than 100 nM (in an assay with a FVIIa concentration of 10 nM).
  • the IC 50 value is lower than 40 nM.
  • the IC 50 value is lower than 20 nM.
  • the IC 50 value is lower than 10 nM.
  • the IC 50 value is lower than 5 nM.
  • the method for preparation of a humanized antibody comprises testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC 50 value lower than the IC-50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC 50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 10 nM, more preferably lower than the ICso value of FFR-rFVIIa.
  • the method for preparation of a humanized antibody comprises testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding.
  • the method for preparation of a humanized antibody comprises testing antibodies in a TF ELISA assay comprising TF and selecting humanized antibody which bind human TF.
  • the method for preparation of a humanized antibody comprises testing antibodies in a direct TF ELISA assay comprising immobilized TF and selecting humanized antibodies which binds immobilized human TF.
  • the method for preparation of a humanized antibody comprises testing antibodies in an indirect TF ELISA assay comprising TF in solution and selecting humanized antibodies which binds human TF in solution
  • the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay on TF expressing cell and selecting humanized antibodies which inhibits FXa generation on TF expressing cell with an IC- 50 value lower than 500 nM (in an assay with a FVIIa concentration of 1 nM).
  • the IC 50 value is lower than 100 nM.
  • the IC 50 value is lower than 50 nM
  • the IC 50 value is lower than 10 nM
  • the IC 50 value is lower than 5 nM.
  • the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay on TF expressing cell and selecting humanized antibodies which inhibits FXa generation on TF expressing cell with an IC- 50 value lower than the IC 50 value of FFR-rFVIIa + 500 nM (using 1 nM FVIIa in the assay), such as lower than the IC 50 value of FFR-rFVIIa + 100 nM, preferably lower than the IC 50 value of FFR-rFVIIa + 50 nM preferably lower than the IC 50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC 50 value of FFR-rFVIIa + 5 nM, more preferably lower than the IC 50 value of FFR-rFVIIa.
  • the term "TF expressing cell” mean any mammalian cell, that expresses human TF.
  • the method for preparation of a humanized antibody comprises testing antibodies in a whole cell TF binding assay and selecting humanized antibodies which competes with FVIIa binding to human TF expressed on the cell surface of whole cells.
  • the method for preparation of a humanized antibody comprises testing antibodies in a biosensor assay and selecting humanized antibodies with a K d value for binding to human TF lower than 100 nM.
  • the K d value for binding to human TF is lower than 10 nM.
  • the K d value for binding to human TF is lower than 5 nM.
  • the K d value for binding to human TF is lower than 1 nM.
  • the K d value for binding to human TF is lower than 0.5 nM. In a further embodiment the K d value for binding to human TF is lower than 10 "10 M. In a further embodiment the K d value for binding to human TF is lower than 10 "11 M. In a further embodiment the K d value for binding to human TF is lower than 10 "12 M. In a further embodiment the K d value for binding to human TF is lower than 10 "13 M. In a further embodiment the K d value for binding to human TF is lower than 10 "14 M. In a further embodiment the K d value for binding to human TF is lower than 10 "15 M.
  • the method for preparation of a humanized antibody comprises testing antibodies in a MAPK signalling assay and selecting humanized antibodies which inhibits FVIIa-induced activation of the MAPK signalling.
  • the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 98 %. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 90 %. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 70 %. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 50 %.
  • the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 30 %.
  • MAPK signalling means a cascade of intracellular events that mediate activation of Mitogen-Activated-Protein-Kinase (MAPK) or homologues thereof in response to various extracellular stimuli.
  • MAPK Mitogen-Activated-Protein-Kinase
  • Three distinct groups of MAP kinases have been identified in mammalian cells: 1) extracellular-regulated kinase (Erk1/2 or p44/42), 2) c-Jun N-terminal kinase (JNK) and 3) p38 kinase.
  • the Erk1/2 pathway involves phosphorylation of Erk 1 (p 44) and/or Erk 2 (p 42).
  • Activated MAP kinases e.g. p44/42 MAPK can translocate to the nucleus where they can phosphorylate and activate transcription factors including (Elk 1 ) and signal transducers and activators of transcription (Stat). Erk1/2 can also phosphorylate the kinase p90RSK in the cytoplasm or in the nucleus, and p90RSK then can activate several transcription factors.
  • the term "protein kinase” is intended to indicate an enzyme that is capable of phosphorylating serine and/or threonine and/or tyrosine in peptides and/or proteins.
  • FVIIa-induced activation of the MAPK signalling is intended to indicate that FVIIa binds to TF in a mammalian cell and thereby induce MAPK signalling.
  • the method for preparation of a humanized antibody comprises testing antibodies in a gene expression analysis assay (assay 15) and selecting humanized antibodies which inhibits FVIIa induced up-regulation of genes independently selected from the list comprising Fra-1 , Id2, and Cyr61.
  • antibodies against TF which inhibits the activity of TF may bind different epitopes present on TF and may inhibit both the binding of FVIIa or the binding of FX or FXa to human TF. It is an object of the present invention to select antibod- ies, which inhibits the binding of FVIIa to human TF and thereby the FVIIa induced intracellular signalling.
  • the method for preparation of a humanized antibody comprises testing antibodies in a human cancer assay (assay 13) and selecting humanized antibodies which inhibits growth or metastasis of human cancers.
  • the isolated humanized antibody inhibits FVIIa induced up-regulation of genes independently selected from the list comprising Fra-1 , Id2, and Cyr61.
  • the isolated humanized antibody does not inhibit binding of FX or FXa to human TF.
  • the isolated humanized antibody inhibits the intracellular activity of TF.
  • the method for preparation of a humanized antibody comprises testing antibodies in an epitope mapping assay and selecting humanized antibodies which binds preferred epitopes on TF.
  • the preferred epitope comprises one or more of the residues Trp45, Lys46 and Tyr94.
  • the preferred epitope comprises the residue Trp45.
  • the preferred epitope comprises the residue Lys46.
  • the preferred epitope comprises the residue Tyr94.
  • the isolated humanized antibody binds to an epitope within the interface between TF and FVIIa.
  • the residues in TF that are responsible for the interaction between the protease domain of FVIIa and TF determined from the X-ray structure are; Ser39, Gly43, Trp45, Ser47, Phe50, Arg74, Phe76, Tyr94, Pro92.
  • This interface between the protease domain of FVIIa and TF is characterized as being a complex interface region containing many intermolecular hydrogen bonds allowing many fine contacts between TF and FVIIa to obtain high specificity in binding process of FVIIa.
  • the present invention also relates to high affinity humanized antibodies to TF.
  • the TF surface containing the contact interface for the protease domain of FVIIa holds a specific topology that are prone to react to create protein-protein interactions, wherein another type of protein-protein interaction is the complex formation between an antibody and a protein ligand.
  • One aspect of the present invention is high affinity humanized antibodies, that are immunoreacting with the contact interface for the protease domain of FVIIa.
  • Humanized TF antibodies of the present invention act as antagonists for
  • Humanized TF antibodies are particularly useful for administration to humans to treat a variety of conditions involving intravascular coagulation. As such, humanized TF antibodies may be useful for inhibiting TF activity resulting in, for example, the inhibition of blood coagulation, thrombosis or platelet deposition. Furthermore, humanized TF antibodies according to the present invention, which acts to inhibit the cellular functions of TF, the signalling function of TF, may be useful in conditions like sepsis, inflammation, atherosclerosis, restenosis, or cancer.
  • Humanized TF antibodies may be useful in a variety of diseases. Included are thrombotic or coagulopathic related diseases or disorders including inflammatory response and chronic thromboembolic diseases or disorders associated with fibrin formation including vascular disorders such as deep venous thrombosis, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, tumor growth, tumor metastasis, angiogenesis-related disease(s), thrombolysis, atherosclerosis, arteriosclerosis e.g. arteriosclerosis following angioplastry, and restenosis e.g.
  • thrombotic or coagulopathic related diseases or disorders including inflammatory response and chronic thromboembolic diseases or disorders associated with fibrin formation including vascular disorders such as deep venous thrombosis, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry
  • restenosis following angioplastry acute and chronic indications such as inflammation, septic chock, septicemia, hypotension, adult respiratory distress syndrome (ARDS), systemic inflammatory response syndrome (SIRS), disseminated intravascular coagulopathy (DIC), pulmonary embolism, pathological platelet deposition, myocardial infarction, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis, venoocclusive disease following peripheral blood progenitor cell (PBPC) transplantation, hemolytic uremic syndrome (HUS), thrombotic thrombocytopenic purpura (TTP) and other diseases or disorders.
  • PBPC peripheral blood progenitor cell
  • HUS hemolytic uremic syndrome
  • TTP thrombotic thrombocytopenic purpura
  • the humanized TF antibodies may be used to prevent the occurrence of thromboembolic complications in identified high risk patients, such as those undergoing surgery or those with congestive heart failure.
  • the humanized TF antibodies may be particularly useful in the treatment of intimal hyperplasia or restenosis due to acute vascular injury.
  • Acute vascular injuries are those which occur rapidly (i.e. over hours to months, in contrast to chronic vascular injuries (e.g. atherosclerosis) which develop over a lifetime.
  • Acute vascular injuries often result from surgical procedures such as vascular reconstruction, wherein the techniques of angioplasty, endarterectomy, atherectomy, vascular graft emplacement or the like are employed.
  • Hyperplasia may also occur as a delayed response in response to, e.g., graft emplacement or organ transplantation. Since humanized TF antibodies is more selective than heparin, binding only TF which has been exposed at sites of injury, and because humanized TF antibodies does not destroy or inhibit other coagulation proteins, it will be more effective and less likely to cause bleeding complications than heparin when used prophylactically for the prevention of deep vein thrombosis.
  • the humanized TF antibodies of the present invention is able to bind selectively to cell-surface TF and inhibit its functional activity by inhibiting the binding of coagulation FVIIa to TF.
  • Humanized TF antibodies which maintain binding to TF inhibit platelet accumulation at the site of vascular injury by blocking the production of thrombin and the subsequent deposition of fibrin.
  • humanized TF antibodies Due to the ability of humanized TF antibodies to block thrombin generation and limit platelet deposition at sites of acute vascular injury, humanized TF antibodies which maintain binding to TF thereby inhibiting FVIIa binding can be used to inhibit vascular restenosis.
  • Compositions comprising humanized TF antibodies are particularly useful in methods for treating patients when formulated into pharmaceutical compositions, where they may be given to individuals suffering from a variety of disease states to treat coagulation- related conditions.
  • Such humanized TF antibodies, capable of binding TF and inhibiting FVIIa binding to TF may possess a longer plasma half-life and thus a correspondingly longer period of anticoagulant activity when compared to other anticoagulants.
  • compositions are those commonly treated with anticoagulants, such as, for example, deep vein thrombosis, pulmonary embolism, stroke, disseminated intravascular coagulation (DIC), fibrin deposition in lungs and kidneys associated with sepsis, antiphospholipid syndrome (APS), atherosclerosis and myocardial infarction.
  • anticoagulants such as, for example, deep vein thrombosis, pulmonary embolism, stroke, disseminated intravascular coagulation (DIC), fibrin deposition in lungs and kidneys associated with sepsis, antiphospholipid syndrome (APS), atherosclerosis and myocardial infarction.
  • the compositions can be used to inhibit vascular restenosis as occurs following mechanical vascular injury, such as injury caused by surgery, microsurgery, balloon angioplasty, endarterectomy, reductive atherectomy, stent placement, laser therapy or rotablation, or as occurs secondary to vascular grafts, stents, bypass grafts or organ transplants.
  • a method of inhibiting coagulation, vascular restenosis or platelet deposition comprises administering to a patient a composition comprising humanized TF antibodies in an amount sufficient to effectively inhibit coagulation, vascular restenosis or platelet deposition.
  • the methods also find use in the treatment of acute closure of a coronary artery in an individual (e.g. acute myocardial infarction), which comprises administering the humanized TF antibodies, in conjunction with tissue plasminogen activator or streptokinase, and can accelerate tPA induced thrombolysis.
  • the humanized TF antibodies are given prior to, in conjunction with, or shortly following administration of a thrombolytic agent, such as tissue plasminogen activator.
  • a thrombolytic agent such as tissue plasminogen activator.
  • TF may be produced by immunizing transgenic mice (Obtained from Medarex) carrying parts of the human immune system rather than the mouse system with human TF. Splenocytes from these transgenic mice are used to produce hybridomas that secrete human monoclonal antibodies as described (see, e.g., Wood et al. International Application WO 91/00906, Kucherlapati et al. PCT publication WO 91/10741 ; Lonberg et al. International Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg, N. et al. 1994 Nature 368: 856-859; Green, L L. et al. 1994 Nature Genet. 7: 13-21; Morrison, S.
  • Human monoclonal antibodies directed against human TF may also be produced by phage display.
  • Human antibody libraries can be constructed from immunized persons and displayed on the surface of filamentous phage.
  • Phage display of large na ' ive libraries has also made it possible to isolate human antibodies directly without immunization (DeHaard H. J.et al J. Biol. Chem. 1999, 18218-18230).
  • Figure 1 Schematic presentation of an examplified screening assay for selection of human monoclonal high affinity antibodies against TF.
  • FIG. 1 Detailed schematic representation of screening assays 1-3 as described in example 1.
  • Figure 4 Detailed schematic representation of screening assays 8-10 as described in example 1.
  • Figure 5. An example of screening antibodies by assay no. 4. Inhibition of sTF/FVIIa amidolytic activity by FFR-rFVIIa (closed circles) and the human anti TF monoclonal antibody HuTF-31 F2 (open circles).
  • Figure 6 An example of screening antibodies by assay no. 5. Inhibition of factor Xa generation by FFR-rFVIIa (closed circles) and the human anti-TF monoclonal antibody HuTF-31 F2 (open circles).
  • Figure 7 An example of screening antibodies by assay no. 7. Inhibition of human TF-induced clotting by FFR-rFVIIa (closed circles) and the human anti TF monoclonal antibody HuTF- 31 F2 (open circles).
  • Figure 8 An example of screening antibodies by assay no. 10. Only anti-TF monoclonal antibodies preventing FVIIa binding inhibits TF/FVI la-mediated signaling.
  • FIG. 9 Human anti TF Mab inhibit FVIIa induced phosphorylation of p44/42 MAPK (assay no. 10). BHK cell transfected with TF were serum-starved for 2 hr to make cells quiescent. The antibodies HuMab 30F5 (500 nM) and HuMab 31 F2 (500 nM) were added to the cells 15 min prior addition of FVIIa (15 nM). Cells were lysed and proteins were separate on SDS- PAGE and transferred to nitrocellulose by electroblotting. Western blot analysis was performed using polyclonal phospho-specific antibodies to p44/42 MAPK. Secondary antibodies were anti-rabbit IgG conjugated to Horse Radish Peroxidase.
  • Figure 10 An example of screening antibodies by assay no. 16. The figure demonstrates the inhibition of TF intracellular activity in TF expressing cells by monoclonal antibodies against TF. Anti TF Mab B inhibits TF intracellular activity, while Anti-TF Mab A do not.
  • FIG. 11 An example of screening antibodies by assay no. 12. Velocity profile of thromboe- lastograms obtained with 0.5 nM of FFR-rFVIIa and the human anti-TF antibody HuTF-31 F2.
  • the present invention is further illustrated by the following examples which, however, are not to be construed as limiting the scope of protection.
  • the features disclosed in the forego- ing description and in the following examples may, both separately and in any combination thereof, be material for realizing the invention in diverse forms thereof.
  • Human TF can be isolated from human brain as described by Rao, LN.M., Thrombosis Research, 51:373-384 1988.
  • Lipidated recombinant human TF can also be used as human thromboplastin reagent.
  • Rat, rabbit, baboon, and pig thromboplastin are prepared from brain tissue. Two volumes of 45°C 0.9 % ⁇ aCI are added to the brain tissue, and the tissue is homogenized with a manual glass homogenisator. After 30 min incubation at 45°C with occasional shaking, the samples are centrifuged 20 min at 2000 x g. The precipitate are discarded, and the supernatant is aliquoted and stored at -80°C until use.
  • Relipidated TF may be obtained by reconstitution of recombinant human full length TF (American Diagnostica #4500) into phospholipid vesicles (PC/PS 75/25).
  • Biotinylated human TF is produced as follows: Biotin- ⁇ HS (n-succinimido biotin, Sigma H-1759) is dissolved in DMF (dimethylformamid) at a concentration of 1.7 mg/ml. 1 mg/ml of human TF in 0.1 M ⁇ aHCO 3 buffer is added 60 ⁇ l of the biotin-NHS solution and is allowed to react for 4 hours at room temperature. The reacting solution containing the bioti- nylated TF is dialysed against PBS-buffer over night.
  • FVIIa used is recombinant human FVIIa prepared as described by Thim, L. et al. Biochem 27: 7785-7793, 1988.
  • sTF Recombinant human soluble TF-
  • S2288 reconstituted in H 2 O to 17.24 mg/ml (Chromogenix)
  • FX Purified human plasma FX (HFX, Enzyme Research Laboratories Ltd.)
  • FXa Purified human plasma FX activated with Russel's Viper Venom (HFXa, Enzyme Research Laboratories Ltd.)
  • Chromozyme X Dissolved in H 2 O to 1.25 mg/ml. (Boehringer Mannheim) 125 l-FVIIa is obtained by standard radiolabelling procedures.
  • FFR-rFVIIa FVIIa blocked in the active site with D-Phe-L-Phe-L-Arg-chloromethyl ketone. Prepared as described by Sorensen B. B. et al. J.Biol.Chem. 272: 11863-11868, 1997.
  • Human TF is emulsified in Freunds Complete Adjuvant.
  • HuMab mice or hybrids therof (Medarex) are given 40 ⁇ g by a subcutaneous injection. After 14 and 28 days, and eventually more times with intervals of 14 days the mice are boosted with a similar injection of 20 ⁇ g of TF in Incomplete Freunds Adjuvant. Ten days after the last injection a blood sample is taken and sera are tested for human TF specific antibodies by TF ELISA (Assay 1 and 2).
  • mice with positive serum test from assay 1-3 are boosted with 20 ⁇ g of human TF by an intravenous injection and sacrificed after three days. The spleen is removed aseptically and dispersed to a single cell suspension.
  • Fox-myeloma cells are grown in CD Hybridoma medium (Gibco 11279-023 ). Fusion of spleen cells and myeloma cells (P3x63 Ag8.653, ATCC CRL-1580), and the Sp2/0 (ATCC CRL-1581 ) myeloma cell lines for our fusions are done by the PEG- methods (K ⁇ hler, G & Milstein C. (1976), European J. Immunology, 6:511-19). Cells are seeded in microtiter plates and incubated at 37 °C. Medium is changed three times over the next two weeks. 100 ⁇ l of supernatant from hybridoma cells is removed from each well and tested for TF specific antibodies in TF ELISA (Assay 1 and 2).
  • Nunc immunoplates are coated overnight at 4°C with 1 ⁇ g/ml of human sTF in PBS. Plates are blocked with blocking buffer (TBS with 5 mM CaCI 2 and 2% BSA) and washed in TBS+ 0.05 % Tween 20, and the supernatants from the host cells are added. After incubation at room temperature for 1 hour, plates are washed and anti-human IgG labelled with horseradish peroxidase (HRPO) is added. After another hour of incubation, plates are washed and developed with TMB-substate (Kem-EN-Tec) as described by the manufactures. Absorbance at 450 nm is measured on an ELISA-reader.
  • TBS blocking buffer
  • HRPO horseradish peroxidase
  • Nunc-immunoplates are coated with 0.5 ⁇ g/ml of goat anti-human IgG (Southern Biotechnology Associates, Cat-#2040-1) in PBS and incubated overnight at 4°C. Plates are blocked with blocking buffer (TBS with 5 mM CaCI 2 and 2% BSA) and washed in TBS+ 5 mM CaCI 2 + 0.05 % Tween 20. Culture supernatants from the host cells are added and the plates incubated for 1 hour at room temperature. After another wash, biotinylated human sTF are added at a concentration of 1 ⁇ g/ml, and incubated for 1 hour. After washing, 100 ⁇ l of a Streptavidin-HRPO solution is added and incubated for 1 hour. Plates are developed with TMB-substate as described for assay 1.
  • Nunc-immunoplates are incubated with human sTF (cone 5 ⁇ g/ml in PBS) over night, 4 °C. Plates are washed and blocked in TBS buffer with 5 mM CaCI 2 and 2 % BSA. Anti-human TF antibodies are added and plates are incubation for 2 hours. Plates are washed before bioti- nylated human FVIIa are added (1 ⁇ g/ml in TBS buffer with 5 mM CaCI 2 and 2 % BSA) and the plates incubated for 1 hour. Plates are washed before addition of HRPO-labeled Strepta- vidin and incubated for 45 min. Plates are washed again before development with TMB substrate (Kem-EN-Tec) as described by the manufactures.
  • TMB substrate Kerm-EN-Tec
  • Inhibition of FVIIa/sTF amidolytic activity (Assay 4): Inhibition of FVIIa-TF catalyzed amidolytic activity by anti-human TF antibodies is tested employing soluble human TF (10 nM), recombinant human FVIIa (10 nM) and increasing concentrations of antibodies (0.0122 - 50 nM).
  • Varying concentrations of anti-human TF antibodies or FFR-rFVIIa are preincubated with 10 nM sTF and 10 nM FVIIa in BSA buffer (50 mM Hepes, pH 7.4, 100 mM NaCI, 5 mM CaCI 2 and 1 mg/ml BSA) for 60 min at room temperature before addition of substrate S2288 (1.2 mM, Chromogenix). The colour development is measured continuously for 30 min at 405 nm. Amidolytic activity is presented as mOD/min. IC 50 values for inhibition of FVlla/TF amidolytic activity by the antibodies may be calculated. The IC 50 value for FFR-rFVIIa is 7 +/- 3 nM in this assay. Inhibition of FXa generation (Assay 5).
  • TF (10 pM), FVIIa (100 pM) and anti-TF antibodies or FFR-rFVIIa (0 - 50 nM) in BSA buffer are incubated 60 min at room temperature before FX (50 nM ) is added. The reaction is stopped after another 10 min by addition of A volume stopping buffer (50 mM Hepes, pH 7.4, 100 mM NaCI, 20 mM EDTA). The amount of FXa generated is determined by adding substrate S2765 (0.6 mM, Chromogenix, and measuring absorb- ance at 405 nm continuously for 10 min. IC 50 values for antibody inhibition of FVIIa/lipidated TF-mediated activation of FX may be calculated. The IC 50 value for FFR-rFVIIa is 51 +/- 26 pM in this assay.
  • Biosensor assay (Assay 6): antibodies are tested on the Biacore instrument by passing a standard solution of anti-human TF antibody over a chip with immobilized antibody to human IgG. This is followed by different concentrations of sTF in 10 mM hepes pH 7.4 containing 150 mM NaCI, 10 mM CaCI 2 and 0.0003 % polysorbate 20. K d values are calculated from the sensorgrams using the integrated Biacore evaluation software.
  • TF-dependent clotting assay (Assay 7): The assay is carried out on an ACL300 Research clotting apparatus (ILS Laboratories). Dilutions of anti-human TF antibodies in 50 mM imidazole, pH 7.4, 100 mM NaCI, 0.1 % BSA are mixed with 25 mM CaCI 2 in the ratio of 2 to 5 and added to sample cups in the clotting apparatus. Thromboplastin from human, rat, rabbit, baboon, or pig diluted with the imidazole buffer to give clotting time of approximately 30 sec in samples without antibody is placed in reagent reservoir 2, and human, rat, rabbit, baboon, or pig plasma, in reagent reservoir 3.
  • Monolayers of cells expressing human TF e.g. human lung fibroblasts WI-38 (ATTC No. CCL-75), human bladder carcinoma cell line J82 (ATTC No. HTB-1), human keratinocyte cell line CCD 1102KerTr (ATCC no. CRL-2310), human glioblastoma cell line U87, or human breast cancer cell line MDA-MB231, are employed as TF source in FVlla/TF catalyzed activation of FX.
  • human lung fibroblasts WI-38 (ATTC No. CCL-75)
  • human bladder carcinoma cell line J82 ATTC No. HTB-1
  • human keratinocyte cell line CCD 1102KerTr ATCC no. CRL-2310
  • human glioblastoma cell line U87 or human breast cancer cell line MDA-MB231
  • Confluent cell monolayers in a 24-, 48- or 96-well plate are washed one time in buffer A (10 mM Hepes, pH 7.45, 150 mM NaCI, 4 mM KCI, and 11 mM glucose) and one time in buffer B (buffer A supplemented with 1 mg/ml BSA and 5 mM Ca 2+ ).
  • buffer A 10 mM Hepes, pH 7.45, 150 mM NaCI, 4 mM KCI, and 11 mM glucose
  • buffer B buffer A supplemented with 1 mg/ml BSA and 5 mM Ca 2+
  • FVIIa (1 nM) FX (135 nM
  • varying concentrations of antibody (or FFR-rFVIIa) in buffer B are simultaneously added to the cells.
  • the cells are preincubated 15 min with anti-TF antibodies or FFR-rFVIIa before addition of rFVIIa and FX.
  • FXa formation is allowed for 15 min at 37°C
  • FXa 50- ⁇ l aliquots are removed from each well and added to 50 ⁇ l stopping buffer (Buffer A supplemented with 10 mM EDTA and 1 mg/ml BSA).
  • Buffer A supplemented with 10 mM EDTA and 1 mg/ml BSA.
  • the amount of FXa generated is de- termined by transferring 50 ⁇ l of the above mixture to a microtiter plate well and adding 25 ⁇ l Chromozym X (final concentration 0.6 mM) to the wells.
  • the absorbance at 405 nm is measured continuously and the initial rates of colour development are converted to FXa concentrations using a FXa standard curve.
  • the IC S0 value for FFR-rFVIIa is 1.5 nM in this assay.
  • Binding studies are employed using cells expressing human TF, e.g. human lung fibroblasts WI-38 (ATTC No. CCL-75), human bladder carcinoma cell line J82 (ATTC No. HTB-1), human keratinocyte cell line CCD 1102KerTr (ATCC no. CRL-2310), human glioblastoma cell line U87, or human breast cancer cell line MDA-MB231.
  • Confluent monolayers in 24-well tissue culture plates are washed once with buffer A (see assay 8) and once with buffer B (see assay 8). The monolayers are preincubated 2 min with 100 ⁇ l cold buffer B.
  • Varying concentrations of antibodies (or FFR-rFVIIa) and radiolabelled FVIIa are simultaneously added to the cells (final volume 200 ⁇ l). The plates are incubated for 2 hours at 4 °C. At the end of the incubation, the unbound material is removed, the cells are washed 4 times with ice-cold buffer B and lysed with 300 ⁇ l lysis buffer (200 mM NaOH, 1 % SDS and 10 mM EDTA). Radioactivity is measured in a gamma counter (Cobra, Packard Instruments). The binding data are analyzed and curve fitted using GraFit4 (Erithacus Software, Ltd., (U.K.). The IC 50 value for FFR-rFVIIa is 4 nM in this assay. Inhibition of FVIIa/TF-induced p44/42 MAPK activation by anti TF antibody (Assay 10):
  • the amount of phosphorylated p44/42 MAPK and/or Akt, and/or p90RSK is determined by quantitative detection of chemiluminescence (Fujifilm LAS-1000) from western blot analysis.
  • Cells expressing human TF e.g. CCD1102KerTr, NHEK P166, human glioblastoma cell line U87, or human breast cancer cell line MDA-MB231 , are cultured in medium with 0 - 0.1 % FCS for 24 or 48 hours prior to the experiment to make cells quiescent. At the day of the experiment the cells must be 70-80% confluent.
  • the experiment is performed by prein- cubating the cells with excess antibody or FFR-rFVIIa in medium without serum for 30 min at 37°C before addition of 10 - 100 nM FVIIa and incubating for 10 min.
  • As a positive control of cell signaling cells are treated with 10 % FCS for 10 minutes.
  • Cells are washed 2 times in ice-cold PBS before cells are lysed in lysis buffer (20 mM Tris, 0.1 % Triton X-100, 1 mM EDTA, 1 mM EGTA, 50 mM sodium-fluoride, 10 mM sodium ⁇ -glycerophosphate, 5 mM sodium pyrophosphate, 150 mM NaCI, pH 7.5 containing 0.1 mM 4-(2-aminoethyl)benzene- sulfonyl fluoride (AEBSF) and 1 mM benzamidine.
  • lysis buffer 20 mM Tris, 0.1 % Triton X-100, 1 mM EDTA, 1 mM EGTA, 50 mM sodium-fluoride, 10 mM sodium ⁇ -glycerophosphate, 5 mM sodium pyrophosphate, 150 mM NaCI, pH 7.5 containing 0.1 mM 4-(2-aminoe
  • lysates were mixed with SDS-sample buffer and loaded on a SDS-polyacrylamide gel. A standard biotinylated protein marker is loaded on each gel. Proteins separated on the SDS-polyacrylamide gel were transferred to nitrocellulose by electroblotting, and the kinases p44/42 MAPK, Akt and p90RSK were visualized by immunoblotting with phosphospecific antibodies, and chemiluminiscence is quaniti- ated by Fujifilm LAS1000.
  • sTF variants I22C, W45C, K46C, Y94C, F140C, W158C, K201C
  • sTF variants I22C, W45C, K46C, Y94C, F140C, W158C, K201C
  • Refolding is achieved after 1-hour incubation at room temperature by dropwise diluting the denatured protein into a 1 L solution containing 50 mM Tris-HCI, 0.25 M NaCI, pH 8.0 with gentle stirring for approximately 2 hours. Purification is performed using Q- Sepharose Fast Flow (Pharmacia, Uppsala, Sweden) and FVIIa affinity chromatography as described by Freskgard et al. (1996). The homogeneity of the protein is verified by SDS- PAGE. The concentration is measured at A 280nm and determined using a calculated extinction coefficient of 37440 M "1 cm "1 (Gill and von Hippel, 1989).
  • Human thromboplastin e.g. Innovin, Dade Behring, final dilution 50,000 x
  • CaCI 2 final concentration 16.7 mM
  • anti-TF antibodies e.g. 1 mM
  • Citrate-stabilized human whole blood 280 ⁇ l
  • RoTEG sample cups Pentapharm
  • 40 ⁇ l throm- boplastin/CaCI 2 /anti-TF antibody mixture 40 ⁇ l throm- boplastin/CaCI 2 /anti-TF antibody mixture.
  • Thromboelastography is followed for one hour in a RoTEG apparatus (Pentapharm). Velocity profiles are obtained from the thrombograms using CoagPro SoftwareTM (MedScience, Arhus, Denmark).
  • mice Healthy female athymic mice (nu/nu) aged 7-8 weeks are used. To destroy the residual immunoresistance of the nude mice to the human cell implantation, the mice are routinely irradiated at 5 Gy 2 days before human tumor grafting (Vogel et al., 1997). Mice are challenged by tumor grafting of LS174T humancolon carcinoma cells (ATCC CCL 188) cultured in RPMI 1640 with 15% fetal calf serum (FCS) as described (Li et al., Human Gene Therapy 10: 3045-3053, 1999).
  • FCS fetal calf serum
  • the cells are harvested with trypsin-EDTA, washed twice, and resuspended in serumfree RPMI supplemented with sodium-heparinate solution (1 U/ml).
  • a small left subcostal incision is then carried out in mice under anesthesia and 3 3 106 LS174T cells in 50 m lof phosphate-buffer ed saline (PBS) are injected into the spleen.
  • PBS phosphate-buffer ed saline
  • the spleen vessels are ligated and the spleen is surgically removed. This procedure will lead to a stable incidence of liver metastasis (more than 95%).
  • the treatment with anti-TF antibodies will be initiated immediately after implantation and will last for the remaining study period.
  • liver samples are fixed overnight in AFA (5% acetic acid, 75% ethyl alcohol, 2% formalin, 18% water), transferred to 100% ethanol, and embedded in paraffin, and 5- m m sections are prepared for histological quantification of metastatic nodules, for immuno- histochemistry and apoptosis quantification.
  • AFA 5% acetic acid, 75% ethyl alcohol, 2% formalin, 18% water
  • mice 60 homozygotous nu/nu 6 weeks old NMRI males.
  • mice are randomly allocated in four groups of 15 and treated with solutions A,
  • Termination At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated.
  • Parameters Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2-3 times per week. Postmortem determination of metastasis formation in the liver.
  • MDA-MB-231 Human breast carcinoma cells MDA-MB-231 (ATCC HTB26) are cultured in Dul- becco's modified Eagle's medium (DMEM) supplemented with 10% fetal calf serum (FCS). MDA-MB-231 cells (3 3 106) are injected subcutaneously in nude mice (7- to 8-week-old female mice). Primary tumor growth and metastasis is evaluated as described previously (Li et al., Human Gene Therapy 12: 515-526, 2001)
  • Aim To examine the effect on macroscopical growth and lung metastasis of MDA- MB-231 mammary tumors in nude mice of anti-TF antibodies given bolus injection i.v.; 10 mg/kg. Mice: 60 homozygotous nu/nu 6 weeks old NMRI males.
  • mice are randomly allocated in four groups of 15 and treated with solutions A,
  • Termination At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated.
  • Parameters Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2-3 times per week. Postmortem determination of metastasis formation in the lung.
  • the tumor cell line MG U373 is a human glioblastoma multiforme cell line, with high angiogenic activity, high vascular density and fast growth in nude mice. Tumors are inoculated in the flanks, following standard procedures (see enclosed protocols for experimental plan). The mice are observed twice daily for signs of toxicity and the tumors are measured daily in two perpendicular diameters.
  • mice are 7-week-old males obtained from M&B (Ry, Denmark). Animals are kept in a gnotobiotic environment and they receives sterile food pellets and drinking water ad libitum. Three different studies is conducted with the glioma tumor model:
  • Aim To examine the effect on macroscopical growth of U373 tumors in nude mice of anti-TF antibodies given bolus injection i.v.; 10 mg/kg. Mice: 60 homozygotous nu/nu 6 weeks old NMRI males.
  • mice are randomly allocated in four groups of 15 and treated with solutions A,
  • Termination At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated.
  • Parameters Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2-3 times per week.
  • Aim To examine the effect on macroscopic growth of U373 tumors in nude mice of anti-TF antibodies given bolus injection i.v.; 10 mg/kg. after pretherapeutic tumor growth has been established.
  • mice 60 homozygotous nu/nu 6 weeks old NMRI males.
  • mice are randomly allocated in four groups of 15 and treated with solutions A,
  • Termination Treatment lasts until the tumors have grown beyond the maximal size of approximately 1.0 cm 3 , i.e. no tumor diameter larger than 15 mm or until Gompertzian regrowth has been established by 6 consecutive measurements. At time of termination tumors from each group are excised for histological and immunochemical evaluation. At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated
  • Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2 times per week. Study 111-3:
  • Aim To examine the effect of anti-TF antibodies on growth of intracranial U373 tumors in nude mice.
  • mice 60 homozygotous nu/nu 6 weeks old NMRI males.
  • Tumor U373 implanted orthotopically in the right hemisphere following standard procedures.
  • mice are randomly allocated in four groups of 15 and treated with solutions A, B, or C.
  • a 0.5 ml matrigel plug will be located subcutaneously under the abdominal skin.
  • b-FGF 5 ng
  • haemoglobin angiogenesis
  • cDNA microarray analyses a specific up-regulation of three genes in BHK-TF cells treated with FVIIa has been observed. These include: Fra-1 , a gene coding for Fos related antigen 1 , Id2, a gene encoding a member of the helix-loop-helix class of proteins, and Cyr61 encoding an extracellular matrix signalling protein.
  • the following assay is designed to screen for anti-TF antibodies which prevents FVIIa induced up-regulation of Fra-1, Id2 or Cyr61. Cell culture.
  • Reagents are purchased from GIBCO-BRL Life Technologies unless otherwise noted.
  • BHK-TF cells created as described by Poulsen L.K. et al., J Biol. Chem. 273, 6228- 6232, 1998, are grown in Dulbecco's modified Eagle's medium containing 10% FCS, 100 lU/ml penicillin, and 100 ⁇ g/ml streptomycin to obtain 95-100% confluence, washed and grown for additional 16-18 hs in medium without FCS. The cells are again washed and exposed to FCS-free medium containing 100 nM FVIIa. For cloning of fragments for Northern blot analyses the cells are treated as follows.
  • BHK-TF cells are grown in Dulbecco's modified Eagle's medium containing 10% FCS, 100 IU/ml penicillin, and 100 ⁇ g/ml streptomycin to obtain 95-100% confluence, washed and grown for additional 16-18 hs in medium without FCS. The cells are again washed and exposed to FCS-free medium containing 100 nM FVIIa for 1 h.
  • CRL2091 cells (ATCC) are grown in Is- cove's modified Dulbecco's medium containing 10% FBS, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin to 95-100% confluence.
  • Murine 3T3-L1 cells are maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin. Cells are grown to confluence and induced to with media containing 1 ⁇ M dexamethasone (Sigma), 10 ⁇ g/ml human insulin (Novo Nordisk A/S), and 1 ⁇ M BRL49653 (Novo Nordisk A/S) for 1 h.
  • dexamethasone Sigma
  • 10 ⁇ g/ml human insulin Novo Nordisk A/S
  • 1 ⁇ M BRL49653 Novo Nordisk A/S
  • Fra-1 is cloned by reverse transcription PCR from RNA isolated from 3T3-L1 cells treated for
  • Id2 and Cyr61 are cloned by reverse transcription PCR from RNA isolated from BHK-TF cells treated for 1 h with FVIIa and from CRL2091 cells treated for 6 hours with FVIIa, respectively.
  • the upstream and downstream primers are: 5'-GCGGCCGCCATGTACCGAGACTACGGGGAACCG-3' and 5'- GCGGCCGCTCACAAAGCCAGGAGTGTAGG-3' for Fra-1 , 5'- CAGCATGAAAGCCTTCAGTC-3' and 5'-CTCTGGTGATGCAGGCTGAC-3' for Id2, 5'- CGTCACCCTTCTCCACTTGA-3' and 5'-CTTGGTCTTGCTGCATTTCT-3' for Cyr61.
  • Parameters for PCR are one cycle of denaturing at 94 °C for 10 s, annealing at 65 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, annealing at 64 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, an- nealing at 63 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, annealing at 62 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of dena- turing at 94 °C for 10 s, annealing at 61 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, annealing at 60 °C for 15
  • RNA are isolated from BHK-TF cells incubated with FVIIa, FX, ASIS, 1F44A1 or TF ⁇ -
  • RNA size-fractionated in a denaturing gel containing 1% agarose, 20 mM MOPS, 5 mM NaOAc, 6% formaldehyde, and 1 mM EDTA, transferred to a Hybond N + membrane (Amersham) by capillary blotting and immobilized by UV crosslinking.
  • cDNA encoding Fra-1 , Id2 or Cyr61 are labelled with the Prime It kit (Stratagene) using [ ⁇ - 32 P] dATP (Amersham) and hybridized using Express Hyb (Clontech) following the manufacturer's instructions and results are visualized by autoradio- graphy.
  • HeLa cells are seeded to 40 % confluence in a T-80 flask one day prior to transfec- tion.
  • Cells are transfected with 150 ng pFA-Elk1 (Stratagene), 3 ⁇ g pFR-Luc (Stratagene), 3 ⁇ g human TF/pcDNA3 and 3 ⁇ g mouse Protease Activated Receptor 2/pcDNA3,1+ using 36 ⁇ l FuGene (Roche) as described in the manual.
  • the following day the cells are detached by VerseneTM (Invitrogen) and seeded in black 96 well view plates (Packard) at a cell density of 20.000 cells per well. After the cells had reattached to the plate, the medium is replaced with 160 ⁇ l per well serum-free Dulbeccos Modified Eagle Medium (Invitrogen) and incubated for 16 hours.
  • Human monoclonal antibodies are made as described in example 1. These antibod- ies have relatively low affinity to tissue factor compared to murine monoclonal antibodies made against the same epitope.
  • the hyper variable regions of the murine monoclonal antibodies have been sequenced and transferred to a human monoclonal antibody that reacts with the same epitope on tissue factor as well as to a "na ' ive" human monoclonal antibody framework. Affinities have been compared between the two antibody frameworks. Antibody generation.
  • Transgenic mice expressing human antibodies and normal mice are three times immunized with the antigen, here human TF. 10 days after the last immunization the antibody titer were determined and the highest responder of the transgenic mice and the normal mice were selected for fusions. Spleen cells of these mice were isolated and fused to the mouse myeloma cell line AG8.X63 in the case of the transgenic mice and FOX in the case of the normal mice. After 14 days of selection clones were screened for binding towards human TF. Afterwards the affinity of positive clones is measured by BIAcore. mRNA extraction and cDNA synthesis. mRNA from clones of the highest affinity mouse and human antibodies is isolated and used as template for cDNA synthesis by RT-PCR amplification of the hypervariable regions according to standard procedures. Amplification of murine TF antibody variable regions.
  • TF antibody variable heavy chains are amplified from cDNA using a degenerated primer mix of: 5'- ACTAGTTTTGGCTGAGGAGACGGTGACCGTGG-3', 5'- AC- TAGTTTTGGCTGAGGAGACTGTGAGAGTGG-3', 5'- ACTAGTTTTGGCTGCAGA-
  • GACAGTGACCAGAG-3' 5'- ACTAGTTTTGGCTGAGGAGACGGTGACTGAGG-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1).
  • TF antibody variable kappa-light chains are amplified from cDNA using 5'- TCAT- CAACACTCATTCCTGTTGAAGCTCTTGA-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1).
  • HuTF antibody variable heavy chains are amplified from cDNA of using primer 5'- GTGCCAGGGGGAAGACCGATGGG-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1).
  • HuTF antibody variable kappa-light chains are amplified from cDNA using primer 5'- GCAGGCACACAACAGAGGCAGTTCCAGATTTC-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1).
  • Cloning of TF antibodies into E. coli The PCR product of the TF antibody variable chains are TOPO-cloned according to manufactures protocol (Invitrogen kat.#45-0030) and transformed in competent E. coli cells.
  • DNA is extracted from transformants and the variable regions are characterized by sequencing. Genetic modification of HuTF antibody hyper variable regions.
  • the CDR's are characterized by sequencing from which conserved consensus motifs can be derived.
  • new CDRs are engineered and introduced into the HuTF antibody framework.
  • the new CDRs are created in the variable regions by extension PCR and the engineered variable regions are introduced into IgG expression vectors plESRgl , plESRglf, and plESRg4 (provided by Medarex) in the following manner: Variable light chains introduced into Hind 1 '11/BsiWi site and variable heavy chains introduced into Notl/Nhel site of the vectors. Expression of recombinant TF antibodies.
  • the resultant plasmids are analyzed by sequencing and transfected into mammalian cells according to standard procedures.
  • Antibodies are purified from the supernatant by protein A and affinities between those recombinant generated antibodies are compared.

Abstract

In one aspect, the invention provides non-immunogenic high affinity humanized antibodies against human TF, which antibodies inhibit the binding of coagulation factor VII/Vila. In another aspect, the invention provides novel and useful methods for selection of therapeutically effective humanized antibodies against human TF. In a further aspect the invention provides an isolated humanized antibody, that immunoreacts with an epitope P present on a protein, wherein the human framework amino acid sequences of said humanized antibody are derived from a human antibody, wherein said human antibody immunoreacts with an epitope H present on said protein, said epitope H comprising an amino acid residue at a particular position of said protein, which amino acid residue at this particular position of said protein is also comprised within the epitope P with which said humanized antibody immunoreacts.

Description

HUMANIZED TISSUE FACTOR ANTIBODIES
FIELD OF THE INVENTION
The present invention relates to isolated antibodies that immunoreacts with tissue factor (TF) to inhibit the binding of coagulation factor Vila (FVIIa) and thus a novel immuno- therapeutic method using humanized antibodies against TF to inhibit thrombus formation associated with surgery, microsurgery, angioplasty or trauma or to inhibit thrombus formation and other functions of TF in abnormal haemostatic conditions associated with a disease (such as deep vein thrombosis, disseminated intravascular coagulation (DIC), coronary ar- tery disease, sepsis, inflammation, atherosclerosis, or cancer). Also disclosed are a method for preparation of antibodies as well as cell lines for preparation of the humanized antibodies (abs).
BACKGROUND OF THE INVENTION
Blood coagulation is a process consisting of a complex interaction of various blood components, or factors, which eventually gives rise to a fibrin clot. Generally, the blood components which participate in what has been referred to as the coagulation "cascade" are pro- enzymes or zymogens, enzymatically inactive proteins which are converted to proteolytic enzymes by the action of an activator, itself an activated clotting factor. Coagulation factors that have undergone such a conversion and generally referred to as "active factors," and are des- ignated by the addition of a lower case "a" suffix (e.g., Factor Vila).
Activated Factor X ("Xa") is required to convert prothrombin to thrombin, which then converts fibrinogen to fibrin as a final stage in forming a fibrin clot. There are two systems, or pathways, that promote the activation of Factor X. The "intrinsic pathway" refers to those reactions that lead to thrombin formation through utilization of factors present only in plasma. A series of protease-mediated activations ultimately generates Factor IXa which, in conjunction with Factor Villa, cleaves Factor X into Xa. An identical proteolysis is effected by FVIIa and its co-factor, TF, in the "extrinsic pathway" of blood coagulation. TF is a membrane bound protein and does not normally circulate in an active form in plasma. Upon vessel disruption, however, TF can complex with FVIIa to catalyze Factor X activation or Factor IX activation in the presence of Ca2+ and phospholipid. While the relative importance of the two coagulation pathways in hemostasis is unclear, Factor VII and TF have been found to play a pivotal role in the initiation of blood coagulation.
It is often necessary to selectively block the coagulation cascade in a patient. Anticoagulants such as heparin, coumarin, derivatives of coumarin, indandione derivatives, or other agents may be used, for example, during kidney dialysis, or to treat deep vein thrombosis, disseminated intravascular coagulation (DIC), and a host of other medical disorders. For example, heparin treatment or extracorporeal treatment with citrate ion may be used in dialysis to prevent coagulation during the course of treatment. Heparin is also used in preventing deep vein thrombosis in patients undergoing surgery.
Treatment with heparin and other anticoagulants may, however, have undesirable side effects. Available anticoagulants generally act throughout the body, rather than acting specifically at site of injury. Heparin, for example, may cause heavy bleeding. Furthermore, with a half-life of approximately 80 minutes, heparin is rapidly cleared from the blood, necessitating frequent administration. Because heparin acts as a cofactor for antithrombin III (ATIII), and ATIII is rapidly depleted in DIC treatment, it is often difficult to maintain the proper heparin dosage, necessitating continuous monitoring of ATIII and heparin levels. Heparin is also ineffective if ATIII depletion is extreme. Further, prolonged use of heparin may also increase platelet aggregation and reduce platelet count, and has been implicated in the development of heparin-induced thrombocytopenia. Indandione derivatives may also have toxic side effects. In addition to the anticoagulants briefly described above, several naturally occurring proteins have been found to have anticoagulant activity. Also, ATIII has been proposed as a therapeutic anticoagulant. International Application No. WO 92/15686 relates to inactivated Factor Vila for inhibiting blood coagulation.
Antibodies are specific immunoglobulin (Ig) polypeptides produced by the vertebrate immune system in response to challenges by foreign proteins, glycoproteins, cells, or other antigenic foreign substances. The sequence of events which permits the organism to overcome invasion by foreign cells or to rid the system of foreign substances is at least partially understood. An important part of this process is the manufacture of antibodies which bind specifically to a particular foreign substance. The binding specificity of such polypeptides to a particular antigen is highly refined, and the multitude of specificities capable of being generated by the individual vertebrate is remarkable in its complexity and variability. Millions of antigens are capable of eliciting antibody responses, each antibody almost exclusively directed to the particular antigen which elicited it.
Two major sources of vertebrate antibodies are presently utilized, generation in situ by the mammalian B lymphocytes, and generation in cell culture by B-cell hybrids. Antibodies are generated in situ as a result of the differentiation of immature B lymphocytes into plasma cells, which occurs in response to stimulation by specific antigens. In the undifferentiated B cells, the portions of DNA coding for the various regions on the immunoglobulin chains are separated in the genomic DNA. The sequences are assembled sequentially prior to expression. The resulting rearranged gene is capable of expression in the mature B lymphocyte to produce the desired antibody. However, even when a particular mammal is exposed to only a single antigen a uniform population of antibodies does not result. The in situ immune response to any particular antigen is defined by the mosaic of responses to the various determinants which are present on the antigen. Each subset of homologous antibodies is contributed by a single population of B cells, hence in situ generation of antibodies is "polyclonal".
This limited but inherent heterogeneity has been overcome in numerous particular cases by use of hybridoma technology to create "monoclonal" antibodies in cell cultures by B cell hybridomas.
In this process, the relatively short-lived, or mortal, splenocytes or lymphocytes from a mammal which has been injected with antigen are fused with an immortal tumor cell line, thus producing hybrid cells or "hybridomas" which are both immortal and capable of producing the genetically coded antibody of the B cell. The hybrids thus formed are segregated into single genetic strains by selection, dilution, and regrowth, and each strain thus represents a single genetic line. They therefore, produce antibodies which are assured to be homogeneous against a desired antigen. These antibodies, referencing their pure genetic parentage, are called "monoclonal". Monoclonal antibodies with mono-specificity have greatly influenced immunology, and their usefulness has already been demonstrated in such sciences as biology, pharmacology, biochemistry and others. Such monoclonal antibodies have found widespread use not only as diagnostics reagents, but also therapeutically (see, for example, Ritz and Schlossman, Blood, 59:1-11 , (1982)). Monoclonal antibodies produced by hybridomas, while theoretically effective as discussed above and clearly preferable to polyclonal antibodies because of their specificity, suffer from an important disadvantage. In many applications, the use of monoclonal antibodies produced in non-human animals is severely restricted where the monoclonal antibodies are to be used in humans. Repeated injections of a "foreign" antibody in humans, such as a mouse antibody, may lead to harmful hypersensitivity reactions. Such a non-human derived monoclonal antibody, when injected into humans, causes an anti-nonhuman antibody response.
Therapeutic use of mouse Mabs against TF is known from U.S. patent no. 6,001 ,978 and 5,223,427. International Application No. WO 99/51743 relates to human/mouse chimera monoclonal antibodies directed against human TF.
European patent application No. 833911 relates to CDR-grafted antibodies against human TF.
Presta L. et al., Thrombosis and Haemostasis, Vol. 85 (3) pp. 379-389 (2001) relates to humanized antibody against TF inhibiting plasma clotting and TF/FVI la-mediated activation of factors IX and X. There is still a need in the art for improved compositions having anticoagulant activity which can be administered at relatively low doses and do not produce the undesirable side effects associated with traditional anticoagulant compositions. The present invention fulfils this need by providing anticoagulants that do not have the side effects associated with the traditional antibodies with non-human sequences, they act specifically at sites of injury, and further provides other related advantages. Furthermore the present invention provides compounds, which acts to inhibit the cellular functions of TF, which is implicated in conditions like sepsis, inflammation, atherosclerosis, restenosis, or cancer.
These and other advantages of the invention, as well as additional inventive features, will be apparent from the description of the invention provided herein. SUMMARY OF THE INVENTION
In one aspect, the invention provides non-immunogenic high affinity humanized antibodies against human TF, which antibodies inhibit the binding of coagulation factor Vll Vlla. In another aspect, the invention provides novel and useful methods for selection of therapeutically effective humanized antibodies against human TF. In a more particular aspect, the present invention relates to an isolated humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF. In other aspects, the invention relates to methods of using these and other TF antibodies, methods of producing such antibodies, cells for producing such antibodies, and related compositions. Detailed DESCRIPTION OF THE INVENTION
The terms "humanized antibodies" and "humanized antibody", as used herein, refer to recombinant antibodies in which complementarity determining region (CDR) sequences or variants thereof derived from the germline of another mammalian species, such as a mouse, have been grafted into human framework sequences, e.g. the so-called humanized antibodies. In one aspect of the invention the isolated humanized monoclonal antibody comprises CDR amino acid sequences derived from a monoclonal antibody. In a further aspect of the invention the isolated humanized monoclonal antibody has CDR amino acid sequences derived from a mouse monoclonal antibody. The term "monoclonal antibody" as used herein, refers to a homogeneous population of immunoglobulins, i.e. the individual molecules of the antibody population are identical except for naturally occurring mutations. Antibodies are normally synthesized by lymphoid cells derived from B lymphocytes of bone marrow. Lymphocytes derived from the same clone produce immunoglobulin of a single amino acid sequence. Lymphocytes can not be directly cultured over long periods of time to produce substantial amounts of their specific antibody. However, Kohler et al., 1975, Nature, 256:495, demonstrated that a process of somatic cell fusion, specifically between a lymphocyte and a myeloma cell, could yield hybridoma cells which grow in culture and produce a specific antibody called a "monoclonal antibody". Myeloma cells are lymphocyte tumor cells which, depending upon the cell strain, frequently produce an antibody themselves, although "non-producing" strains are known. The term "recombinant antibody", as used herein, is intended to include all 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 (described further in Section II, below), antibodies isolated from a recombinant combinatorial antibody library (described further in Section III, below), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295) and/or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain aspects of the invention, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies may be sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. The terms "human tissue factor" or "human TF" as used herein, refers to the full length polypeptide receptor comprising the amino acid sequence 1-263 of native human tissue factor.
The term "antibody", as used herein, refers to immunoglobulin molecules and fragments thereof, that have the ability to specifically bind to an antigen (e.g., human TF). Full-length antibodies comprises four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). 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, and FR4. Thus, within the definition of an antibody is also one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., human TF). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antibody" include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH I domains; (ii) F(ab)2 and F(ab')2 fragments, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (see e.g. Ward et al., (1989) Nature 341 :544-546 ), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426: and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antibody". Other forms of single chain antibodies, such as diabodies are also encompassed by the term "antibody.". Diabodies are bivalent and bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but are separated by a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain so as to create two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). It is understood that human TF may have one or more antigenic determinants comprising (1) peptide antigenic determinants which consist of single peptide chains within human TF, (2) conformational antigenic determinants which consist of more than one spatially contiguous peptide chains whose respective amino acid sequences are located disjointedly along the human TF polypeptide sequence; and (3) post-translational antigenic determinants which consist, either in whole or part, of molecular structures covalently attached to human TF after translation, such as carbohydrate groups. An "isolated humanized antibody", as used herein, refers to a humanized antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds human TF is substantially free of antibodies that specifically bind antigens other than human TF). An isolated antibody that specifically binds human TF may, however, have cross-reactivity to other antigens, such as TF molecules from other species (discussed in further detail below). Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals. The term "epitope" as used herein means any antigenic determinant on an antigen to which the antibody binds. Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. The terms "immunoreacts" or "immunoreacting", as used herein, means any binding of an antibody to its epitope with a dissociation constant Kd lower than 10"4 M. The terms
"immunoreacts" or "immunoreacting" are used where appropriate interchangeably with the phrase " specifically bind".
The term "inhibits", as used herein, means any reduction compared to a reference. As an example, an antibody, that "inhibits" the binding of human coagulation factor Vila to human TF means any antibody, that reduces the ability of human coagulation factor Vila to bind human TF compared to the ability of human coagulation factor Vila to bind human TF in the absense of the antibody.
The term "affinity", as used herein, means the strength of the binding of an antibody to an epitope. The affinity of an antibody is measured by the dissociation constant Kd, defined as [Ab] x [Ag] / [Ab-Ag] where [Ab-Ag] is the molar concentration of the antibody-antigen complex, [Ab] is the molar concentration of the unbound antibody and [Ag] is the molar concentration of the unbound antigen. The affinity constant Ka is defined by 1/Kd. Preferred methods for determining antibody specificity and affinity by competitive inhibition can be found in Harlow, et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988), Colligan et al., eds., Current Protocols in
Immunology, Greene Publishing Assoc. and Wiley Interscience, N.Y., (1992, 1993), and Muller, Meth. Enzymol. 92:589-601 (1983), which references are entirely incorporated herein by reference. In another aspect, the invention relates to a pharmaceutical composition comprising a therapeutically effective amount of a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
The phrase "a therapheutically effective amount" is the effective dose to be determined by a qualified practitioner, who may titrate dosages to achieve the desired response. Factors for consideration of dose will include potency, bioavailability, desired pharmacokinetic/pharmacodynamic profiles, condition of treatment (e.g. trauma, inflammation, septic chock), patient-related factors (e.g. weight, health, age, etc.), presence of co-administered medications, time of administration, and/or other factors known to a medical practitioner. The dosage of a humanized antibody against TF administered to a patient will vary with the type and severity of the condition to be treated, but is generally in the range of 0.1 -5.0 mg/kg body weight.
The term "subject" as used herein means any animal, in particular mammals, such as humans, and may, where appropriate, be used interchangeably with the term "patient". In a another aspect, the invention relates to a composition comprising a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
In a further aspect, the invention relates to a method for treatment of a FVlla/TF related disorder in a human, which method comprises administering to the human a therapeutically effective amount of a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
"Treatment" means the administration of an amount of antibodies of the invention and/or a compound of the invention with the effect of reducing the intensity of, delaying the onset of, and/or reducing the risk of, and preferably preventing, the occurrence of one or more physiological symptoms associated with a disease state or reducing and/or eliminating one or more detectable physiological conditions or symptoms associated with a disease state present in a subject (and, preferably, eliminating all conditions associated with the disease state). Humanized TF antibodies and compositions provided by the invention can be used in any and all aspects of treating a subject.
The terms "FVlla/TF related disorder" as used herein means a disease or disorder, where TF and FVIIa are involved. Examples of FVlla/TF related disorders include thrombotic and/or coagulopathic related diseases, and disorders including inflammatory response and chronic thromboembolic diseases, and disorders associated with fibrin formation including vascular disorders such as deep venous thrombosis, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary an- gioplastry (PTCA), stroke, tumor growth, tumor metastasis, angiogenesis-related disease(s), thrombolysis, atherosclerosis, arteriosclerosis e.g. arteriosclerosis following angioplastry, and restenosis (such as restenosis following angioplastry), acute and chronic indications such as inflammation, septic chock, septicemia, hypotension, adult respiratory distress syndrome (ARDS), disseminated intravascular coagulopathy (DIC), pulmonary embolism, platelet deposition, myocardial infarction, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis, and other diseases or disorders. The FVlla/TF related disorder is not limited to in vivo coagulopatic disorders such as those named above but includes ex vivo FVllaTF related processes such as coagulation that may result from the ex- tracorporeal circulation of blood, including blood removed in-line from a patient in such processes as dialysis procedures, blood filtration, or blood bypass during surgery. The term "Factor Vila", or "FVIIa" means "two chain" activated coagulation factor VII cleaved by specific cleavage at the Arg152-lle153 peptide bond. FVIIa, may be purified from blood or produced by recombinant means. It is evident that the practice of the methods described herein is independent of how the purified factor Vila is derived and, therefore, the applicable aspects of the invention can include the use of any suitable Factor Vila preparation. In many aspects, hu- man Factor FVIIa is preferred.
The term "FVII" means "single chain" coagulation factor VII.
In a further aspect, the invention relates to a method for preparation of a humanized antibody, which method comprises preparation of humanized antibodies against human TF, testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, or testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding, or testing antibodies in a FVIIa signalling assay and selecting humanized antibodies which inhibits FVIIa-induced intracellular signalling. The term "FVIIa signalling assay" as used herein means any assay which measures the FVIIa-induced intracellular signalling.
The term "FVIIa-induced intracellular signalling" as used herein means the intracellular events mediated when FVIIa is added to the cell. "FVIIa-induced intracellular signalling" may be measured by the MAPK signalling assay or a gene expression analysis assay which meas- ures the up-regulation of genes independently selected from the list comprising Fra-1 , Id2, and Cyr61.
Unless otherwise indicated, the specific IC50 values referred to with respect to the TF-induced clot assays described herein are those obtained when using normal human plasma. The term "TF-induced clot assay" as used herein means any assay where clotting time is measured in sample comprising blood plasma and TF. An example of a TF-induced clot assay is described in Example 1 , assay 7. The term "FXa generation assay" as used herein means any assay where activation of FX is measured in a sample comprising TF, FVIIa, FX, calcium and phospholipids. An example of a FXa generation assay is described in Example 1 , assay 5.
The term "FVlla/TF amidolytic assay" as used herein means any assay where the amidolytic activity, i.e. cleavage of a small peptide substrate, of FVIIa is measured in the presence of TF. An example of a FVlla/TF amidolytic assay is described in Example 1 , assay 4. The term "TF ELISA assay" as used herein means any ELISA assay comprising TF and antibodies against TF. Examples of TF ELISA assays are the direct and indirect TF ELISA assays described in Example 1 , assay 1 and 2. The term "direct TF ELISA assay" as used herein means any TF ELISA assay comprising immobilized TF. An example of a direct TF ELISA assays is described in Example 1 , assay 1.
The term "indirect TF ELISA assay" as used herein means any TF ELISA assay, where TF is in solution. Example of direct TF ELISA assays is described in Example 1 , assay 2. In a further aspect, the invention relates to a humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF and are produced by a method that comprises : preparation of humanized antibodies against human TF, testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of
FFR-rFVIIa + 100 nM (using 10 nM FVIIa), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC-50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, or testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding, or testing antibodies in a FVIIa signalling assay and selecting humanized antibodies which inhibits FVIIa-induced intracellular signalling.
In a further aspect, the invention relates to a method for preparation of a humanized anti- body, which method comprises a) preparation of immortal cells to secrete humanized antibodies, b) isolation of culture medium from immortal cells comprising produced humanized antibodies, c) testing antibodies in an indirect TF ELISA assay comprising TF in solution and se- lecting humanized antibodies that immunoreacts with human TF in solution, d) testing antibodies in a FVIIa competition assay and selecting humanized antibodies which competes with FVIIa binding, e) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 10 nm FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, f) testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC50 value lower than the IC50 value of FFR- rFVIIa + 100 nM (using 0.1 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 10 nM preferably lower than the IC50 value of FFR-rFVIIa + 5 nM, preferably lower than the IC50 value of FFR-rFVIIa + 1 nM, more preferably lower than the IC50 value of FFR-rFVIIa + 0.1 nM, more preferably lower than the IC50 value of FFR-rFVIIa, g) testing antibodies in a TF-induced clot assay and selecting humanized antibodies which inhibits clot formation in this assay with an IC50 value lower than the IC50 value of FFR-rFVIIa + 1 nM, such as lower than the IC50 value of FFR-rFVIIa + 500 pM, preferably lower than the IC50 value of FFR-rFVIIa + 200 pM, preferably lower than the IC50 value of FFR-rFVIIa + 100 pM, such as lower than the IC50 value of FFR- rFVIIa + 50 pM, preferably lower than the IC50 value of FFR-rFVIIa + 10 pM, more preferably lower than the IC50 value of FFR-rFVIIa + 5 pM, more preferably lower than the IC50 value of FFR-rFVIIa, h) selection and cultivation in a suitable culture medium of the immortal cell producing the selected antibody following step c - g, i) isolation of selected antibody from culture medium of selected immortal cell.
The term "antibody-producing cell" as used herein means any cell capable of producing an antibody. Included are hybridomas, transfected cell lines and the relatively short- lived, or mortal, splenocytes or lymphocytes from a mammal which has been injected with an antigen.
In a further aspect, the invention relates to a humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF and are produced by a method that comprises : a) preparation of immortal cells to secrete humanized antibodies, b) isolation of culture medium from immortal cells comprising produced antibodies, c) testing antibodies in an indirect TF ELISA assay comprising TF in solution and selecting humanized antibodies that immunoreacts with human TF in solution, d) testing antibodies in a FVIIa competition assay and selecting humanized antibodies which competes with FVIIa binding, e) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC5o value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, f) testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC50 value lower than the IC50 value of FFR- rFVIIa + 100 nM (using 0.1 nM FVIIa in the assay), such as lower than the IC5o value of FFR-rFVIIa + 10 nM, preferably lower than the IC50 value of FFR-rFVIIa + 5 nM, preferably lower than the IC5o value of FFR-rFVIIa + 1 nM, more preferably lower than the IC50 value of FFR-rFVIIa + 0.1 nM, more preferably lower than the IC5o value of FFR-rFVIIa, g) testing antibodies in a TF-induced clot assay and selecting humanized antibodies which inhibits clot formation in this assay with an IC50 value lower than the IC50 value of FFR-rFVIIa + 1 nM, such as lower than the IC50 value of FFR-rFVIIa + 500 pM, preferably lower than the IC5o value of FFR-rFVIIa + 200 pM, preferably lower than the IC-50 value of FFR-rFVIIa + 100 pM, such as lower than the IC50 value of FFR- rFVIIa + 50 pM, preferably lower than the IC50 value of FFR-rFVIIa + 10 pM, more preferably lower than the IC50 value of FFR-rFVIIa + 5 pM, more preferably lower than the IC50 value of FFR-rFVIIa, h) selection and cultivation in a suitable culture medium of the immortal cell producing the selected antibody following step c - g, i) isolation of selected antibody from culture medium of selected immortal cell.
It is to be understood, that the specific IC50 values referred to in the TF-induced clot assay is when using normal human plasma.
In a further aspect, the invention relates to a cell producing humanized antibodies that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF. In one embodiment the cell is a mammal cell. In a further embodiment the cell is isolated from a mouse. In a further embodiment the cell is a hybridoma cell. In a further embodiment the hybridoma cell is obtained by fusion of an anti- body-producing lymphoid cell with an immortal cell to provide an antibody-producing hybridoma cell. In a further embodiment the cell is a myeloma cell.
Many myeloma cell lines may be used for the production of humanized antibodies in accordance with the techniques described herein, including, e.g., Mouse myeloma NSO (ECACC 85110503), rat myeloma YB2/0 (ATCC CRL 1662), P3X63-Ag8, P3X63-AG8.653, P3/NS1-Ag4-1 (NS-1), Sp2/0-Agl4 and S194/5.XXO.Bu.1. The P3X63-Ag8 and NS-1 cell lines have been described by Kohler and Milstein (Eur. J. Immunol. 6:511 (1976)). Shulman et al. (Nature 276:269 (1978)) developed the Sp2/0-Agl4 myeloma line. The S194/5.XXO.Bu.1 line was reported by Trowbridge (J. Exp. Med. 148:313 (1979)). Traditionally humanized or human antibodies are developed by phage display, ribo- some display, RNA display, surface display on a "living organism" or by using transgenic animals or in immune incompetent animals where human immune competent cells are introduced. Common for the mentioned display techniques are that the humanized antibodies produced by these techniques have affinities that are below what is normally needed for therapeutic antibodies. One has to affinity mature these humanized antibodies further. One reason is that CDR regions from large libraries are combined with a "naive" human framework that has never seen the epitope of interest before, which makes the conformation of this human framework sub optimal. Using transgenic animals where the human Ig repertoire has been introduced to produce human antibodies will not always result in high affinity antibodies probably due to a sub optimally functioning immune system of the transgenic animals. Immunising immune compromised animals that have received human immune competent cells is another way of developing human antibodies, however the length of the immunisation strategy that can be used in this set-up, will not guaranty high affinity antibodies.
It is therefore also an object of the present invention to provide humanized antibod- ies directed against a target protein, where fully human antibodies generated in one of the above mentioned animal systems are combined with CDR sequences from high affinity antibodies generated in another animal, wherein both the fully human antibody and the CDR sequences from high affinity antibodies generated in another animal are directed against the same target protein. Thus, in a further aspect, the invention relates to an isolated humanized antibody, that immunoreacts with an epitope P present on a protein, wherein the human framework amino acid sequences of said humanized antibody are derived from a human antibody, wherein said human antibody immunoreacts with an epitope H present on said protein, said epitope H comprising an amino acid residue at a particular position of said protein, which amino acid residue at this particular position of said protein is also comprised within the epi- tope P with which said humanized antibody immunoreacts. It is to be understood, that this protein may be any protein. The protein may be a therapeutic target protein.
The phrases "framework amino acid sequence" and "framework sequence" individually refer to an amino acid sequence of an antibody that is not a CDR sequence. A framework sequence that is derived from a parent antibody consists of the amino acids present in the corresponding non-CDR sequence of the parent antibody from which such sequences are considered to be derived. Unless otherwise stated or clearly contradicted by context, any framework sequence incorporated in any antibody described herein can be substituted with a suitable framework sequence variant. A framework sequence variant can be any sequence of amino acids that (a) has similar biological property as an antibody produced by a mammal in response to an antigen of interest (e.g., permits immunogenic binding of the antibody of which it is a part to its target, such as TF, preferably with an affinity that is at least about as great, if not greater, than its mammalian-produced counterpart and desirably allows the antibody to retain a similar biological effect as its mammalian-produced counterpart - e.g., a similar level of inhibition of TF binding to FVIIa) and (b) has a high level of amino acid sequence identity with its mammalian-produced counterpart (e.g., has at least about 80%, at least about 85%, at least about 90%, or at least about 95% identity to). Typically, a framework sequence variant will vary by only a few amino acids from its mammalian-produced counterpart (e.g., a variant may differ by 3, 2, or 1 substitutions, deletions, and/or additions) and/or will vary only in terms of conservative amino acid substitutions. Often, a framework sequence variant will consist essentially of the same sequence as the framework sequence of its mammalian-produced counterpart.
In one embodiment of the invention the CDR amino acid sequences of the isolated humanized antibody are derived from a parent monoclonal antibody. In one embodiment the parent monoclonal antibody is a mouse monoclonal antibody. In one embodiment, the CDR amino acid sequences is variant CDR amino acid sequences relative to the CDR amino acid sequences of a parent monoclonal antibody. The terms "variant", as used herein with respect to CDR amino acid sequences, designates, designates CDR amino acid sequences, wherein one or more amino acids have been substituted by another amino acid and/or wherein one or more amino acids have been deleted and/or wherein one or more amino acids have been inserted into the CDR amino acid sequence. The "variant" within this definition still have CDR functionality. In one embodiment the variant CDR amino acid sequences is 70 % identical with the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences is 80 % identical with the CDR sequences of the parent mono- clonal antibody. In one embodiment the variant CDR amino acid sequences is 90 % identical with the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences is 95 % identical with the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences has one amino acid substitution relative to the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences has two amino acid substitutions rela- tive to the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences has three amino acid substitutions relative to the CDR sequences of the parent monoclonal antibody. In one embodiment the variant CDR amino acid sequences has four amino acid substitutions relative to the CDR sequences of the parent monoclonal antibody. In one embodiment of the invention the isolated humanized antibody is a Fab fragment.
In a further embodiment of the invention the isolated humanized antibody is a F(ab)2 fragment.
In a further embodiment of the invention the isolated humanized antibody is a F(ab')2 fragment.
In a further embodiment of the invention the isolated humanized antibody is a single chain Fv fragment.
In a further embodiment of the invention the isolated humanized antibody has human framework amino acid sequences derived from a human antibody, that immunoreacts with a second epitope present on human TF. In one embodiment this second epitope comprises an amino acid residue at a particular position of human TF, which amino acid residue at this particular position of human TF is also comprised within the first epitope with which said humanized antibody immunoreacts. In one embodiment first epitope comprises an amino acid residue at a particular position of human TF selected from the list consisting of Trp45, Lys46 and Tyr94. In a further embodiment second epitope comprises an amino acid residue at a particular position of human TF selected from the list consisting of Trp45, Lys46 and Tyr94.
The terms "human antibody", "human antibodies", "human TF antibody", and "human TF antibodies", as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. 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 and in particular CDR3. However, 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, e.g. the so-called humanized antibodies.
In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF within the range of 10"15 - 10"8 M. It is to be understood, that the Kd for humanized antibody binding to human TF referred to is as determined in an assay, wherein the humanized antibody is immobilized (see assay 6).
In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF within the range of 10"15 - 10"10 M.
In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF within the range of 10"15 - 10"12 M.
In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF lower than 10"8 M. In a further embodiment of the invention the isolated humanized antibody has a K for binding to human TF lower than 10"9 M In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF lower than 10"10 M. In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF lower than 10"11 M. In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF lower than 10"12 M. In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF lower than 10"13 M. In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF lower than 10"14 M. In a further embodiment of the invention the isolated humanized antibody has a Kd for binding to human TF lower than 10"15 M.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a TF-induced clot assay and selecting humanized antibody which inhibit clot formation in this assay with an IC50 value lower than 1 nM. In a further embodiment the IC50 value is lower than 500 pM. In a further embodiment the IC50 value is lower than 200 pM. In a further embodiment the IC50 value is lower than 100 pM. In a further embodiment the IC50 value is lower than 50 pM. In a further embodiment the IC50 value is lower than 10 pM. In a further embodiment the IC50 value is lower than 5 pM. In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a TF-induced clot assay and selecting humanized antibody which inhibit clot formation in this assay with an IC50 value lower than the IC50 value of FFR-rFVIIa + 1 nM, such as lower than the IC50 value of FFR-rFVIIa + 500 pM, preferably lower than the IC50 value of FFR-rFVIIa + 200 pM, preferably lower than the IC50 value of FFR-rFVIIa + 100 pM, such as lower than the IC50 value of FFR-rFVIIa + 50 pM, preferably lower than the IC50 value of FFR-rFVIIa + 10 pM, more preferably lower than the IC50 value of FFR-rFVIIa + 5 pM, more preferably lower than the IC50 value of FFR-rFVIIa.
It is to be understood, that the specific IC50 values referred to in the TF-induced clot assay is when using normal human plasma. In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC50 value lower than 100 nM (in an assay with a FVIIa concentration of 0.1 nM) In a further embodiment the IC50 value is lower than 10 nM. In a further embodiment the IC50 value is lower than 5 nM. In a further embodiment the IC50 value is lower than 1 nM . In a further embodiment the IC50 value is lower than 0.1 nM.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC50 value lower than the IC50 value of FFR- rFVIIa + 100 nM (using 0.1 nM FVIIa in the assay), such as lower than the IC50 value of FFR- rFVIIa + 10 nM, preferably lower than the IC50 value of FFR-rFVIIa + 5 nM, preferably lower than the IC50 value of FFR-rFVIIa + 1 nM, more preferably lower than the IC50 value of FFR- rFVIIa + 0.1 nM, more preferably lower than the IC50 value of FFR-rFVIIa.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a FVlla/TF amidolytic assay and selecting human- ized antibody which inhibit TF-induced FVIIa amidolytic activity with an IC50 value lower than 100 nM (in an assay with a FVIIa concentration of 10 nM). In a further embodiment the IC50 value is lower than 40 nM. In a further embodiment the IC50 value is lower than 20 nM. In a further embodiment the IC50 value is lower than 10 nM. In a further embodiment the IC50 value is lower than 5 nM. In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC-50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the ICso value of FFR-rFVIIa.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding. In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a TF ELISA assay comprising TF and selecting humanized antibody which bind human TF.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a direct TF ELISA assay comprising immobilized TF and selecting humanized antibodies which binds immobilized human TF.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in an indirect TF ELISA assay comprising TF in solution and selecting humanized antibodies which binds human TF in solution In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay on TF expressing cell and selecting humanized antibodies which inhibits FXa generation on TF expressing cell with an IC-50 value lower than 500 nM (in an assay with a FVIIa concentration of 1 nM). In a further embodiment the IC50 value is lower than 100 nM. In a further embodiment the IC50 value is lower than 50 nM In a further embodiment the IC50 value is lower than 10 nM In a further embodiment the IC50 value is lower than 5 nM.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a FXa generation assay on TF expressing cell and selecting humanized antibodies which inhibits FXa generation on TF expressing cell with an IC-50 value lower than the IC50 value of FFR-rFVIIa + 500 nM (using 1 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 100 nM, preferably lower than the IC50 value of FFR-rFVIIa + 50 nM preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa + 5 nM, more preferably lower than the IC50 value of FFR-rFVIIa. The term "TF expressing cell" mean any mammalian cell, that expresses human TF.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a whole cell TF binding assay and selecting humanized antibodies which competes with FVIIa binding to human TF expressed on the cell surface of whole cells. In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a biosensor assay and selecting humanized antibodies with a Kd value for binding to human TF lower than 100 nM. In a further embodiment the Kd value for binding to human TF is lower than 10 nM. In a further embodiment the Kd value for binding to human TF is lower than 5 nM. In a further embodiment the Kd value for binding to human TF is lower than 1 nM. In a further embodiment the Kd value for binding to human TF is lower than 0.5 nM. In a further embodiment the Kd value for binding to human TF is lower than 10"10 M. In a further embodiment the Kd value for binding to human TF is lower than 10"11 M. In a further embodiment the Kd value for binding to human TF is lower than 10"12 M. In a further embodiment the Kd value for binding to human TF is lower than 10"13 M. In a further embodiment the Kd value for binding to human TF is lower than 10"14 M. In a further embodiment the Kd value for binding to human TF is lower than 10"15 M.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a MAPK signalling assay and selecting humanized antibodies which inhibits FVIIa-induced activation of the MAPK signalling. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 98 %. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 90 %. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 70 %. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 50 %. In one embodiment the humanized antibody inhibits FVIIa-induced activation of the MAPK signalling with 30 %. The term "MAPK signalling" means a cascade of intracellular events that mediate activation of Mitogen-Activated-Protein-Kinase (MAPK) or homologues thereof in response to various extracellular stimuli. Three distinct groups of MAP kinases have been identified in mammalian cells: 1) extracellular-regulated kinase (Erk1/2 or p44/42), 2) c-Jun N-terminal kinase (JNK) and 3) p38 kinase. The Erk1/2 pathway involves phosphorylation of Erk 1 (p 44) and/or Erk 2 (p 42). Activated MAP kinases e.g. p44/42 MAPK can translocate to the nucleus where they can phosphorylate and activate transcription factors including (Elk 1 ) and signal transducers and activators of transcription (Stat). Erk1/2 can also phosphorylate the kinase p90RSK in the cytoplasm or in the nucleus, and p90RSK then can activate several transcription factors. The term "protein kinase" is intended to indicate an enzyme that is capable of phosphorylating serine and/or threonine and/or tyrosine in peptides and/or proteins.
The term "FVIIa-induced activation of the MAPK signalling" is intended to indicate that FVIIa binds to TF in a mammalian cell and thereby induce MAPK signalling.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a gene expression analysis assay (assay 15) and selecting humanized antibodies which inhibits FVIIa induced up-regulation of genes independently selected from the list comprising Fra-1 , Id2, and Cyr61.
It is to be understood, that antibodies against TF, which inhibits the activity of TF may bind different epitopes present on TF and may inhibit both the binding of FVIIa or the binding of FX or FXa to human TF. It is an object of the present invention to select antibod- ies, which inhibits the binding of FVIIa to human TF and thereby the FVIIa induced intracellular signalling.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in a human cancer assay (assay 13) and selecting humanized antibodies which inhibits growth or metastasis of human cancers.
In a further embodiment of the invention the isolated humanized antibody inhibits FVIIa induced up-regulation of genes independently selected from the list comprising Fra-1 , Id2, and Cyr61.
In a further embodiment of the invention the isolated humanized antibody does not inhibit binding of FX or FXa to human TF.
In a further embodiment of the invention the isolated humanized antibody inhibits the intracellular activity of TF.
In a further embodiment of the invention the method for preparation of a humanized antibody comprises testing antibodies in an epitope mapping assay and selecting humanized antibodies which binds preferred epitopes on TF. In one embodiment the preferred epitope comprises one or more of the residues Trp45, Lys46 and Tyr94. In one embodiment the preferred epitope comprises the residue Trp45. In one embodiment the preferred epitope comprises the residue Lys46. In one embodiment the preferred epitope comprises the residue Tyr94. In a further embodiment of the invention the isolated humanized antibody binds to an epitope within the interface between TF and FVIIa.
The residues in TF that are responsible for the interaction between the protease domain of FVIIa and TF determined from the X-ray structure (Banner et al. 1996 Nature, 380: 41-46) are; Ser39, Gly43, Trp45, Ser47, Phe50, Arg74, Phe76, Tyr94, Pro92. This interface between the protease domain of FVIIa and TF is characterized as being a complex interface region containing many intermolecular hydrogen bonds allowing many fine contacts between TF and FVIIa to obtain high specificity in binding process of FVIIa.
The present invention also relates to high affinity humanized antibodies to TF. The TF surface containing the contact interface for the protease domain of FVIIa holds a specific topology that are prone to react to create protein-protein interactions, wherein another type of protein-protein interaction is the complex formation between an antibody and a protein ligand.
One aspect of the present invention is high affinity humanized antibodies, that are immunoreacting with the contact interface for the protease domain of FVIIa. Humanized TF antibodies of the present invention act as antagonists for
TF-mediated induction of coagulation, thus inhibiting the binding of coagulation FVIIa to TF and thereby blocking the production of thrombin and the subsequent deposition of fibrin. Humanized TF antibodies are particularly useful for administration to humans to treat a variety of conditions involving intravascular coagulation. As such, humanized TF antibodies may be useful for inhibiting TF activity resulting in, for example, the inhibition of blood coagulation, thrombosis or platelet deposition. Furthermore, humanized TF antibodies according to the present invention, which acts to inhibit the cellular functions of TF, the signalling function of TF, may be useful in conditions like sepsis, inflammation, atherosclerosis, restenosis, or cancer.
Humanized TF antibodies may be useful in a variety of diseases. Included are thrombotic or coagulopathic related diseases or disorders including inflammatory response and chronic thromboembolic diseases or disorders associated with fibrin formation including vascular disorders such as deep venous thrombosis, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, tumor growth, tumor metastasis, angiogenesis-related disease(s), thrombolysis, atherosclerosis, arteriosclerosis e.g. arteriosclerosis following angioplastry, and restenosis e.g. restenosis following angioplastry, acute and chronic indications such as inflammation, septic chock, septicemia, hypotension, adult respiratory distress syndrome (ARDS), systemic inflammatory response syndrome (SIRS), disseminated intravascular coagulopathy (DIC), pulmonary embolism, pathological platelet deposition, myocardial infarction, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis, venoocclusive disease following peripheral blood progenitor cell (PBPC) transplantation, hemolytic uremic syndrome (HUS), thrombotic thrombocytopenic purpura (TTP) and other diseases or disorders. The humanized TF antibodies may be used to prevent the occurrence of thromboembolic complications in identified high risk patients, such as those undergoing surgery or those with congestive heart failure. The humanized TF antibodies may be particularly useful in the treatment of intimal hyperplasia or restenosis due to acute vascular injury. Acute vascular injuries are those which occur rapidly (i.e. over hours to months, in contrast to chronic vascular injuries (e.g. atherosclerosis) which develop over a lifetime. Acute vascular injuries often result from surgical procedures such as vascular reconstruction, wherein the techniques of angioplasty, endarterectomy, atherectomy, vascular graft emplacement or the like are employed. Hyperplasia may also occur as a delayed response in response to, e.g., graft emplacement or organ transplantation. Since humanized TF antibodies is more selective than heparin, binding only TF which has been exposed at sites of injury, and because humanized TF antibodies does not destroy or inhibit other coagulation proteins, it will be more effective and less likely to cause bleeding complications than heparin when used prophylactically for the prevention of deep vein thrombosis.
As shown in the examples that follow, the humanized TF antibodies of the present invention is able to bind selectively to cell-surface TF and inhibit its functional activity by inhibiting the binding of coagulation FVIIa to TF. Humanized TF antibodies which maintain binding to TF inhibit platelet accumulation at the site of vascular injury by blocking the production of thrombin and the subsequent deposition of fibrin.
Due to the ability of humanized TF antibodies to block thrombin generation and limit platelet deposition at sites of acute vascular injury, humanized TF antibodies which maintain binding to TF thereby inhibiting FVIIa binding can be used to inhibit vascular restenosis. Compositions comprising humanized TF antibodies are particularly useful in methods for treating patients when formulated into pharmaceutical compositions, where they may be given to individuals suffering from a variety of disease states to treat coagulation- related conditions. Such humanized TF antibodies, capable of binding TF and inhibiting FVIIa binding to TF, may possess a longer plasma half-life and thus a correspondingly longer period of anticoagulant activity when compared to other anticoagulants. Among the medical indications for the subject compositions are those commonly treated with anticoagulants, such as, for example, deep vein thrombosis, pulmonary embolism, stroke, disseminated intravascular coagulation (DIC), fibrin deposition in lungs and kidneys associated with sepsis, antiphospholipid syndrome (APS), atherosclerosis and myocardial infarction. The compositions can be used to inhibit vascular restenosis as occurs following mechanical vascular injury, such as injury caused by surgery, microsurgery, balloon angioplasty, endarterectomy, reductive atherectomy, stent placement, laser therapy or rotablation, or as occurs secondary to vascular grafts, stents, bypass grafts or organ transplants. The compositions can thus be used to inhibit platelet deposition and associated disorders. Thus, a method of inhibiting coagulation, vascular restenosis or platelet deposition, for example, comprises administering to a patient a composition comprising humanized TF antibodies in an amount sufficient to effectively inhibit coagulation, vascular restenosis or platelet deposition. The methods also find use in the treatment of acute closure of a coronary artery in an individual (e.g. acute myocardial infarction), which comprises administering the humanized TF antibodies, in conjunction with tissue plasminogen activator or streptokinase, and can accelerate tPA induced thrombolysis. The humanized TF antibodies are given prior to, in conjunction with, or shortly following administration of a thrombolytic agent, such as tissue plasminogen activator. According to the invention, human monoclonal antibodies directed against human
TF may be produced by immunizing transgenic mice (Obtained from Medarex) carrying parts of the human immune system rather than the mouse system with human TF. Splenocytes from these transgenic mice are used to produce hybridomas that secrete human monoclonal antibodies as described (see, e.g., Wood et al. International Application WO 91/00906, Kucherlapati et al. PCT publication WO 91/10741 ; Lonberg et al. International Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg, N. et al. 1994 Nature 368: 856-859; Green, L L. et al. 1994 Nature Genet. 7: 13-21; Morrison, S. L. et al. 1994 Proc. Natl. Acad. Sci. USA 81 : 6851-6855; Bruggeman et al. 1993 Year Immunol 7: 33-40; Tuaillon et al. 1993 PNAS 90: 3720-3724; Bruggeman et al. 1991 Eur J Immunol 21 : 1323- 1326). Human monoclonal antibodies directed against human TF may also be produced by phage display. Human antibody libraries can be constructed from immunized persons and displayed on the surface of filamentous phage. High-affinity human single-chain Fv (ScFv) and Fab antibody fragments have in numerous of cases been isolated from such libraries using a panning technique in which the antigen of interest is immobilised on a solid surface i.e. microtiter plates or beads (Barbas OF., Ill and Burton, D.R. Trends. Biotechnol. 1996,
14:230-234; Aujame L. et al, Hum. Antibodies 1997, 8:155-68). Phage display of large na'ive libraries has also made it possible to isolate human antibodies directly without immunization (DeHaard H. J.et al J. Biol. Chem. 1999, 18218-18230).
BRIEF DESCRIPTION OF THE DRAWINGS
The present invention is described in further detail in the examples with reference to the appended drawings wherein
Figure 1. Schematic presentation of an examplified screening assay for selection of human monoclonal high affinity antibodies against TF.
Figure 2. Detailed schematic representation of screening assays 1-3 as described in example 1.
Figure 3. Detailed schematic representation of screening assays 4-7 as described in example !
Figure 4. Detailed schematic representation of screening assays 8-10 as described in example 1. Figure 5. An example of screening antibodies by assay no. 4. Inhibition of sTF/FVIIa amidolytic activity by FFR-rFVIIa (closed circles) and the human anti TF monoclonal antibody HuTF-31 F2 (open circles).
Figure 6. An example of screening antibodies by assay no. 5. Inhibition of factor Xa generation by FFR-rFVIIa (closed circles) and the human anti-TF monoclonal antibody HuTF-31 F2 (open circles).
Figure 7. An example of screening antibodies by assay no. 7. Inhibition of human TF-induced clotting by FFR-rFVIIa (closed circles) and the human anti TF monoclonal antibody HuTF- 31 F2 (open circles).
Figure 8. An example of screening antibodies by assay no. 10. Only anti-TF monoclonal antibodies preventing FVIIa binding inhibits TF/FVI la-mediated signaling.
Figure 9. Human anti TF Mab inhibit FVIIa induced phosphorylation of p44/42 MAPK (assay no. 10). BHK cell transfected with TF were serum-starved for 2 hr to make cells quiescent. The antibodies HuMab 30F5 (500 nM) and HuMab 31 F2 (500 nM) were added to the cells 15 min prior addition of FVIIa (15 nM). Cells were lysed and proteins were separate on SDS- PAGE and transferred to nitrocellulose by electroblotting. Western blot analysis was performed using polyclonal phospho-specific antibodies to p44/42 MAPK. Secondary antibodies were anti-rabbit IgG conjugated to Horse Radish Peroxidase. Detection of chemilumines- cence was performed using a cooled CCD-camera. The bands on the digitalized picture were quantified and the band obtained with FVIIa was set to 100%. When cells were pre-incubated with HuMab 30F5 (500 nM) a 50% reduction in the phosphorylated band was observed and when cells were pre-incubated with HuMab 31 F2 (500 nM) a 25 % reduction was observed. In conclusion, this experiment show that the human antibodies against TF (30F5 and 31 F2) partially inhibited the FVIIa induced phosphorylation of p44/42 MAPK. Similar results were obtained using 50 nM FVIIa.
Figure 10. An example of screening antibodies by assay no. 16. The figure demonstrates the inhibition of TF intracellular activity in TF expressing cells by monoclonal antibodies against TF. Anti TF Mab B inhibits TF intracellular activity, while Anti-TF Mab A do not.
Figure 11. An example of screening antibodies by assay no. 12. Velocity profile of thromboe- lastograms obtained with 0.5 nM of FFR-rFVIIa and the human anti-TF antibody HuTF-31 F2. The present invention is further illustrated by the following examples which, however, are not to be construed as limiting the scope of protection. The features disclosed in the forego- ing description and in the following examples may, both separately and in any combination thereof, be material for realizing the invention in diverse forms thereof.
EXAMPLES
Example 1
Preparation of human monoclonal antibodies immunospecific for human TF.
Reagents.
Human TF can be isolated from human brain as described by Rao, LN.M., Thrombosis Research, 51:373-384 1988.
Lipidated recombinant human TF (Dade Innovin, Baxter) can also be used as human thromboplastin reagent. Rat, rabbit, baboon, and pig thromboplastin are prepared from brain tissue. Two volumes of 45°C 0.9 % ΝaCI are added to the brain tissue, and the tissue is homogenized with a manual glass homogenisator. After 30 min incubation at 45°C with occasional shaking, the samples are centrifuged 20 min at 2000 x g. The precipitate are discarded, and the supernatant is aliquoted and stored at -80°C until use.
Relipidated TF may be obtained by reconstitution of recombinant human full length TF (American Diagnostica #4500) into phospholipid vesicles (PC/PS 75/25).
Biotinylated human TF is produced as follows: Biotin-ΝHS (n-succinimido biotin, Sigma H-1759) is dissolved in DMF (dimethylformamid) at a concentration of 1.7 mg/ml. 1 mg/ml of human TF in 0.1 M ΝaHCO3 buffer is added 60 μl of the biotin-NHS solution and is allowed to react for 4 hours at room temperature. The reacting solution containing the bioti- nylated TF is dialysed against PBS-buffer over night.
FVIIa used is recombinant human FVIIa prepared as described by Thim, L. et al. Biochem 27: 7785-7793, 1988. sTF: Recombinant human soluble TF-|.209 expressed in E. coli and purified essentially as de- scribed by Freskgard, P.-O. et al. Protein Sci. 5, 1531-1540 (1996). S2288: reconstituted in H2O to 17.24 mg/ml (Chromogenix) FX: Purified human plasma FX (HFX, Enzyme Research Laboratories Ltd.) FXa: Purified human plasma FX activated with Russel's Viper Venom (HFXa, Enzyme Research Laboratories Ltd.)
Chromozyme X: Dissolved in H2O to 1.25 mg/ml. (Boehringer Mannheim) 125l-FVIIa is obtained by standard radiolabelling procedures. FFR-rFVIIa: FVIIa blocked in the active site with D-Phe-L-Phe-L-Arg-chloromethyl ketone. Prepared as described by Sorensen B. B. et al. J.Biol.Chem. 272: 11863-11868, 1997.
Immunization.
Human TF is emulsified in Freunds Complete Adjuvant. HuMab mice or hybrids therof (Medarex) are given 40 μg by a subcutaneous injection. After 14 and 28 days, and eventually more times with intervals of 14 days the mice are boosted with a similar injection of 20 μg of TF in Incomplete Freunds Adjuvant. Ten days after the last injection a blood sample is taken and sera are tested for human TF specific antibodies by TF ELISA (Assay 1 and 2).
Fusion. Mice with positive serum test from assay 1-3 are boosted with 20 μg of human TF by an intravenous injection and sacrificed after three days. The spleen is removed aseptically and dispersed to a single cell suspension.
Fox-myeloma cells are grown in CD Hybridoma medium (Gibco 11279-023 ). Fusion of spleen cells and myeloma cells (P3x63 Ag8.653, ATCC CRL-1580), and the Sp2/0 (ATCC CRL-1581 ) myeloma cell lines for our fusions are done by the PEG- methods (Kόhler, G & Milstein C. (1976), European J. Immunology, 6:511-19). Cells are seeded in microtiter plates and incubated at 37 °C. Medium is changed three times over the next two weeks. 100 μl of supernatant from hybridoma cells is removed from each well and tested for TF specific antibodies in TF ELISA (Assay 1 and 2).
Example 2:
Screening.
The various assays used in the screening of serum and culture supernatants for se- lecting specific humanized or human antibodies are described in the following:
Direct TF ELISA assay (Assay 1):
Nunc immunoplates are coated overnight at 4°C with 1 μg/ml of human sTF in PBS. Plates are blocked with blocking buffer (TBS with 5 mM CaCI2and 2% BSA) and washed in TBS+ 0.05 % Tween 20, and the supernatants from the host cells are added. After incubation at room temperature for 1 hour, plates are washed and anti-human IgG labelled with horseradish peroxidase (HRPO) is added. After another hour of incubation, plates are washed and developed with TMB-substate (Kem-EN-Tec) as described by the manufactures. Absorbance at 450 nm is measured on an ELISA-reader.
Indirect TF ELISA assay (Assay 2):
Nunc-immunoplates are coated with 0.5 μg/ml of goat anti-human IgG (Southern Biotechnology Associates, Cat-#2040-1) in PBS and incubated overnight at 4°C. Plates are blocked with blocking buffer (TBS with 5 mM CaCI2and 2% BSA) and washed in TBS+ 5 mM CaCI2 + 0.05 % Tween 20. Culture supernatants from the host cells are added and the plates incubated for 1 hour at room temperature. After another wash, biotinylated human sTF are added at a concentration of 1 μg/ml, and incubated for 1 hour. After washing, 100 μl of a Streptavidin-HRPO solution is added and incubated for 1 hour. Plates are developed with TMB-substate as described for assay 1.
FVIIa competition assay (Assay 3):
Nunc-immunoplates are incubated with human sTF (cone 5 μg/ml in PBS) over night, 4 °C. Plates are washed and blocked in TBS buffer with 5 mM CaCI2 and 2 % BSA. Anti-human TF antibodies are added and plates are incubation for 2 hours. Plates are washed before bioti- nylated human FVIIa are added (1 μg/ml in TBS buffer with 5 mM CaCI2 and 2 % BSA) and the plates incubated for 1 hour. Plates are washed before addition of HRPO-labeled Strepta- vidin and incubated for 45 min. Plates are washed again before development with TMB substrate (Kem-EN-Tec) as described by the manufactures.
Inhibition of FVIIa/sTF amidolytic activity (Assay 4): Inhibition of FVIIa-TF catalyzed amidolytic activity by anti-human TF antibodies is tested employing soluble human TF (10 nM), recombinant human FVIIa (10 nM) and increasing concentrations of antibodies (0.0122 - 50 nM). Varying concentrations of anti-human TF antibodies or FFR-rFVIIa are preincubated with 10 nM sTF and 10 nM FVIIa in BSA buffer (50 mM Hepes, pH 7.4, 100 mM NaCI, 5 mM CaCI2 and 1 mg/ml BSA) for 60 min at room temperature before addition of substrate S2288 (1.2 mM, Chromogenix). The colour development is measured continuously for 30 min at 405 nm. Amidolytic activity is presented as mOD/min. IC50 values for inhibition of FVlla/TF amidolytic activity by the antibodies may be calculated. The IC50 value for FFR-rFVIIa is 7 +/- 3 nM in this assay. Inhibition of FXa generation (Assay 5).
Lipidated TF (10 pM), FVIIa (100 pM) and anti-TF antibodies or FFR-rFVIIa (0 - 50 nM) in BSA buffer (see assay 4) are incubated 60 min at room temperature before FX (50 nM ) is added. The reaction is stopped after another 10 min by addition of A volume stopping buffer (50 mM Hepes, pH 7.4, 100 mM NaCI, 20 mM EDTA). The amount of FXa generated is determined by adding substrate S2765 (0.6 mM, Chromogenix, and measuring absorb- ance at 405 nm continuously for 10 min. IC50 values for antibody inhibition of FVIIa/lipidated TF-mediated activation of FX may be calculated. The IC50 value for FFR-rFVIIa is 51 +/- 26 pM in this assay.
Biosensor assay (Assay 6): antibodies are tested on the Biacore instrument by passing a standard solution of anti-human TF antibody over a chip with immobilized antibody to human IgG. This is followed by different concentrations of sTF in 10 mM hepes pH 7.4 containing 150 mM NaCI, 10 mM CaCI2 and 0.0003 % polysorbate 20. Kd values are calculated from the sensorgrams using the integrated Biacore evaluation software.
TF-dependent clotting assay (Assay 7): The assay is carried out on an ACL300 Research clotting apparatus (ILS Laboratories). Dilutions of anti-human TF antibodies in 50 mM imidazole, pH 7.4, 100 mM NaCI, 0.1 % BSA are mixed with 25 mM CaCI2 in the ratio of 2 to 5 and added to sample cups in the clotting apparatus. Thromboplastin from human, rat, rabbit, baboon, or pig diluted with the imidazole buffer to give clotting time of approximately 30 sec in samples without antibody is placed in reagent reservoir 2, and human, rat, rabbit, baboon, or pig plasma, in reagent reservoir 3. During the analysis 70 μl of the antibody and CaCI2 mixture is transferred to 25 μl thromboplastin reagent and preincubated 900 sec before addition of 60 μl plasma and measuring of the clotting time. Maximal clotting time is set to 400 sec. A dilution of the thromboplastin is used as standard curve for converting clotting times into TF activity relative to the control without anti-TF antibodies or FFR-rFVIIa added. The IC50 value for FFR-rFVIIa is 4.4 +/- 0.4 pM in this assay. Inhibition of FVIIa/cell surface TF catalyzed activation of FX by anti TF antibodies (Assay 8):
Monolayers of cells expressing human TF, e.g. human lung fibroblasts WI-38 (ATTC No. CCL-75), human bladder carcinoma cell line J82 (ATTC No. HTB-1), human keratinocyte cell line CCD 1102KerTr (ATCC no. CRL-2310), human glioblastoma cell line U87, or human breast cancer cell line MDA-MB231, are employed as TF source in FVlla/TF catalyzed activation of FX. Confluent cell monolayers in a 24-, 48- or 96-well plate are washed one time in buffer A (10 mM Hepes, pH 7.45, 150 mM NaCI, 4 mM KCI, and 11 mM glucose) and one time in buffer B (buffer A supplemented with 1 mg/ml BSA and 5 mM Ca2+). FVIIa (1 nM), FX (135 nM) and varying concentrations of antibody (or FFR-rFVIIa) in buffer B are simultaneously added to the cells. Alternatively the cells are preincubated 15 min with anti-TF antibodies or FFR-rFVIIa before addition of rFVIIa and FX. FXa formation is allowed for 15 min at 37°C. 50-μl aliquots are removed from each well and added to 50 μl stopping buffer (Buffer A supplemented with 10 mM EDTA and 1 mg/ml BSA). The amount of FXa generated is de- termined by transferring 50 μl of the above mixture to a microtiter plate well and adding 25 μl Chromozym X (final concentration 0.6 mM) to the wells. The absorbance at 405 nm is measured continuously and the initial rates of colour development are converted to FXa concentrations using a FXa standard curve. The ICS0 value for FFR-rFVIIa is 1.5 nM in this assay.
Inhibition of 125l-FVIIa binding to cell surface TF by antibody (Assay 9):
Binding studies are employed using cells expressing human TF, e.g. human lung fibroblasts WI-38 (ATTC No. CCL-75), human bladder carcinoma cell line J82 (ATTC No. HTB-1), human keratinocyte cell line CCD 1102KerTr (ATCC no. CRL-2310), human glioblastoma cell line U87, or human breast cancer cell line MDA-MB231. Confluent monolayers in 24-well tissue culture plates are washed once with buffer A (see assay 8) and once with buffer B (see assay 8). The monolayers are preincubated 2 min with 100 μl cold buffer B. Varying concentrations of antibodies (or FFR-rFVIIa) and radiolabelled FVIIa (0.5 nM 125l-FVIIa) are simultaneously added to the cells (final volume 200 μl). The plates are incubated for 2 hours at 4 °C. At the end of the incubation, the unbound material is removed, the cells are washed 4 times with ice-cold buffer B and lysed with 300 μl lysis buffer (200 mM NaOH, 1 % SDS and 10 mM EDTA). Radioactivity is measured in a gamma counter (Cobra, Packard Instruments). The binding data are analyzed and curve fitted using GraFit4 (Erithacus Software, Ltd., (U.K.). The IC50 value for FFR-rFVIIa is 4 nM in this assay. Inhibition of FVIIa/TF-induced p44/42 MAPK activation by anti TF antibody (Assay 10):
The amount of phosphorylated p44/42 MAPK and/or Akt, and/or p90RSK is determined by quantitative detection of chemiluminescence (Fujifilm LAS-1000) from western blot analysis. Cells expressing human TF, e.g. CCD1102KerTr, NHEK P166, human glioblastoma cell line U87, or human breast cancer cell line MDA-MB231 , are cultured in medium with 0 - 0.1 % FCS for 24 or 48 hours prior to the experiment to make cells quiescent. At the day of the experiment the cells must be 70-80% confluent. The experiment is performed by prein- cubating the cells with excess antibody or FFR-rFVIIa in medium without serum for 30 min at 37°C before addition of 10 - 100 nM FVIIa and incubating for 10 min. As a positive control of cell signaling, cells are treated with 10 % FCS for 10 minutes. Cells are washed 2 times in ice-cold PBS before cells are lysed in lysis buffer (20 mM Tris, 0.1 % Triton X-100, 1 mM EDTA, 1 mM EGTA, 50 mM sodium-fluoride, 10 mM sodium β-glycerophosphate, 5 mM sodium pyrophosphate, 150 mM NaCI, pH 7.5 containing 0.1 mM 4-(2-aminoethyl)benzene- sulfonyl fluoride (AEBSF) and 1 mM benzamidine. Added just before use: 1 mM sodium or- thovanadate, 5 μg/ml leupeptin, 10 μg/ml aprotinin). Lysates were mixed with SDS-sample buffer and loaded on a SDS-polyacrylamide gel. A standard biotinylated protein marker is loaded on each gel. Proteins separated on the SDS-polyacrylamide gel were transferred to nitrocellulose by electroblotting, and the kinases p44/42 MAPK, Akt and p90RSK were visualized by immunoblotting with phosphospecific antibodies, and chemiluminiscence is quaniti- ated by Fujifilm LAS1000.
Epitope mapping assay (Assay 11):
Preparation of soluble TF (sTF) variants. sTF variants (I22C, W45C, K46C, Y94C, F140C, W158C, K201C) are constructed using in- verse PCR (QuikChange, Stratagene, La Jolla, CA, USA) using the wild type plasmid
(Freskgard et al. Protein Sci. 5, 1531-1540, 1996) as template. The wild type and variants are expressed and purified in E. coli as described elsewhere (Freskgard et al., Protein Sci. 5, 1531-1540, 1996) with some modifications. The cell lysis is performed by the X-press (Biox, Sweden) technique in 10 mM Tris-HCI buffer, pH 7.5 and thereafter resuspended in the same buffer with the addition of 1 mg of DNAse. The solution is centrifuged at 11000xg for 20 min at 4°C, and the inclusion bodies are denatured in 75 ml of 6 M GuHCI, 0.5 M NaCI, 20 mM Tris-HCI, pH 8.0. Refolding is achieved after 1-hour incubation at room temperature by dropwise diluting the denatured protein into a 1 L solution containing 50 mM Tris-HCI, 0.25 M NaCI, pH 8.0 with gentle stirring for approximately 2 hours. Purification is performed using Q- Sepharose Fast Flow (Pharmacia, Uppsala, Sweden) and FVIIa affinity chromatography as described by Freskgard et al. (1996). The homogeneity of the protein is verified by SDS- PAGE. The concentration is measured at A280nm and determined using a calculated extinction coefficient of 37440 M"1cm"1 (Gill and von Hippel, 1989).
MaxiSorp plates (Nunc-lmmuno) are coated with wild type sTF and the variants (10 μg/ml) in TBS and blocked with blocking buffer (TBS with 0.1% Tween 20 and 0.5% BSA). The plates are washed with washing buffer (TBS and 0.1% Tween 20). The anti-human TF antibodies are applied at a concentration of 1 ng/ml in blocking buffer and incubated for one hour. The plates are then washed (6x) using the washing buffer. The antibody binding is subsequently detected using an HRP-labelled anti-human IgG (Helica Biosystems, Inc) at a 1 :2000 dilution in blocking buffer using the TMBp|US-substrate (Kem Tech Cat. 4390A). The final ELISA signal (OD 50.62o) is used as a measure of the binding of each antibody to all sTF variants.
Thromboelastography (Assay 12)
Human thromboplastin (e.g. Innovin, Dade Behring, final dilution 50,000 x) is mixed with CaCI2 (final concentration 16.7 mM) and anti-TF antibodies and incubated 15 min at room temperature. Citrate-stabilized human whole blood (280 μl) is added to RoTEG sample cups (Pentapharm) and preheated 5 min at 37°C, before addition of 40 μl throm- boplastin/CaCI2/anti-TF antibody mixture. Thromboelastography is followed for one hour in a RoTEG apparatus (Pentapharm). Velocity profiles are obtained from the thrombograms using CoagPro Software™ (MedScience, Arhus, Denmark).
Example 3.
Human cancer assay. Investigating the effects of treatment with human anti-TF antibodies on growth and metastasis of human cancers in mouse models (Assay 13)
Treatment:
Human anti-TF antibodies given by bolus injection i.v.; 10 mg/kg = 0.1mg/10g; Injection- volume is 0.1 ml per 10 g mouse of either of three treatment solutions:
A. Vehicle control B. 1 mg/ml Human FFR-rFVIIa
C. 1 mg/ml anti-TF antibodies
Description of models:
/. Primary growth and liver metastasis of colon cancer
Healthy female athymic mice (nu/nu) aged 7-8 weeks are used. To destroy the residual immunoresistance of the nude mice to the human cell implantation, the mice are routinely irradiated at 5 Gy 2 days before human tumor grafting (Vogel et al., 1997). Mice are challenged by tumor grafting of LS174T humancolon carcinoma cells (ATCC CCL 188) cultured in RPMI 1640 with 15% fetal calf serum (FCS) as described (Li et al., Human Gene Therapy 10: 3045-3053, 1999). In brief, the cells are harvested with trypsin-EDTA, washed twice, and resuspended in serumfree RPMI supplemented with sodium-heparinate solution (1 U/ml). A small left subcostal incision is then carried out in mice under anesthesia and 3 3 106 LS174T cells in 50 m lof phosphate-buffer ed saline (PBS) are injected into the spleen. After 3 to 5 min, the spleen vessels are ligated and the spleen is surgically removed. This procedure will lead to a stable incidence of liver metastasis (more than 95%). The treatment with anti-TF antibodies will be initiated immediately after implantation and will last for the remaining study period. On days 15 and 30 after tumor cell inoculation mice are sacrificed, the livers ae removed and weighed, and the number of visible tumor nodules on the liver surfaces are counted. Liver samples are fixed overnight in AFA (5% acetic acid, 75% ethyl alcohol, 2% formalin, 18% water), transferred to 100% ethanol, and embedded in paraffin, and 5- m m sections are prepared for histological quantification of metastatic nodules, for immuno- histochemistry and apoptosis quantification.
Study 1 -1 :
Aim: To examine the effect on macroscopical growth and liver metastasis of
LS174T colon tumors in nude mice of anti-TF antibodies given bolus injection i.v.; 10 mg/kg.
Mice: 60 homozygotous nu/nu 6 weeks old NMRI males.
Groups Mice are randomly allocated in four groups of 15 and treated with solutions A,
B, or C. Termination: At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated. Parameters: Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2-3 times per week. Postmortem determination of metastasis formation in the liver.
//. Primary growth and lung metastasis of mammary cancer
Human breast carcinoma cells MDA-MB-231 (ATCC HTB26) are cultured in Dul- becco's modified Eagle's medium (DMEM) supplemented with 10% fetal calf serum (FCS). MDA-MB-231 cells (3 3 106) are injected subcutaneously in nude mice (7- to 8-week-old female mice). Primary tumor growth and metastasis is evaluated as described previously (Li et al., Human Gene Therapy 12: 515-526, 2001)
Study 11 -1:
Aim: To examine the effect on macroscopical growth and lung metastasis of MDA- MB-231 mammary tumors in nude mice of anti-TF antibodies given bolus injection i.v.; 10 mg/kg. Mice: 60 homozygotous nu/nu 6 weeks old NMRI males.
Groups: Mice are randomly allocated in four groups of 15 and treated with solutions A,
B, or C. Termination: At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated. Parameters: Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2-3 times per week. Postmortem determination of metastasis formation in the lung.
///. Primary growth ofglioma tumor xenografts
The tumor cell line MG U373 is a human glioblastoma multiforme cell line, with high angiogenic activity, high vascular density and fast growth in nude mice. Tumors are inoculated in the flanks, following standard procedures (see enclosed protocols for experimental plan). The mice are observed twice daily for signs of toxicity and the tumors are measured daily in two perpendicular diameters.
Tumors are transplanted to the flanks of nu/nu homozygotous nude mice of NMRI background. The mice are 7-week-old males obtained from M&B (Ry, Denmark). Animals are kept in a gnotobiotic environment and they receives sterile food pellets and drinking water ad libitum. Three different studies is conducted with the glioma tumor model:
Study 111-1:
Aim: To examine the effect on macroscopical growth of U373 tumors in nude mice of anti-TF antibodies given bolus injection i.v.; 10 mg/kg. Mice: 60 homozygotous nu/nu 6 weeks old NMRI males.
Groups: Mice are randomly allocated in four groups of 15 and treated with solutions A,
B, or C. Termination: At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated. Parameters: Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2-3 times per week.
Study III-2:
Aim: To examine the effect on macroscopic growth of U373 tumors in nude mice of anti-TF antibodies given bolus injection i.v.; 10 mg/kg. after pretherapeutic tumor growth has been established.
Mice: 60 homozygotous nu/nu 6 weeks old NMRI males.
Groups: Mice are randomly allocated in four groups of 15 and treated with solutions A,
B, or C. Treatment starts when 6 consecutive (daily) measurements show Gompertzian growth. This corresponds to 100-200 mm3
Termination: Treatment lasts until the tumors have grown beyond the maximal size of approximately 1.0 cm3, i.e. no tumor diameter larger than 15 mm or until Gompertzian regrowth has been established by 6 consecutive measurements. At time of termination tumors from each group are excised for histological and immunochemical evaluation. At a weight loss of > 20% or other objective signs of severe toxicity the animal is terminated
Parameters: Tumor size in two orthogonal diameters is recorded daily during the growth phase. The body weight is recorded initially and 2 times per week. Study 111-3:
Aim: To examine the effect of anti-TF antibodies on growth of intracranial U373 tumors in nude mice.
Mice: 60 homozygotous nu/nu 6 weeks old NMRI males.
Tumor: U373 implanted orthotopically in the right hemisphere following standard procedures.
Groups: Mice are randomly allocated in four groups of 15 and treated with solutions A, B, or C.
Termination: Mice with signs of chronic neurological impairment are euthanized.
Data: Survival (i.e. time to neurological impairment) is quantified by Kaplan-Meyer statistics.
Example 4 (Assay 14).
In mouse wherein the TF gene is knocked out and human TF gene is inserted (mTF- KO/hTF-KI mice) a 0.5 ml matrigel plug will be located subcutaneously under the abdominal skin. In the matrigel b-FGF (5 ng) will be incorporated and one week later the formation of new patent vessels in the gel will be quantitated by measuring the content of haemoglobin (angiogenesis). The inhibitory capacity (% inhibition of the haemoglobin content) of the human anti-TF antibodies can be evaluated after single or repeated parenteral administrations of the proteins.
Example 5 (Assay 15).
Gene expression analysis assay for discriminating antibodies, that prevents FVIIa binding to TF and antibodies, that prevents FX binding to TF.
In cDNA microarray analyses a specific up-regulation of three genes in BHK-TF cells treated with FVIIa has been observed. These include: Fra-1 , a gene coding for Fos related antigen 1 , Id2, a gene encoding a member of the helix-loop-helix class of proteins, and Cyr61 encoding an extracellular matrix signalling protein. The following assay is designed to screen for anti-TF antibodies which prevents FVIIa induced up-regulation of Fra-1, Id2 or Cyr61. Cell culture.
Reagents are purchased from GIBCO-BRL Life Technologies unless otherwise noted.
BHK-TF cells, created as described by Poulsen L.K. et al., J Biol. Chem. 273, 6228- 6232, 1998, are grown in Dulbecco's modified Eagle's medium containing 10% FCS, 100 lU/ml penicillin, and 100 μg/ml streptomycin to obtain 95-100% confluence, washed and grown for additional 16-18 hs in medium without FCS. The cells are again washed and exposed to FCS-free medium containing 100 nM FVIIa. For cloning of fragments for Northern blot analyses the cells are treated as follows. BHK-TF cells are grown in Dulbecco's modified Eagle's medium containing 10% FCS, 100 IU/ml penicillin, and 100 μg/ml streptomycin to obtain 95-100% confluence, washed and grown for additional 16-18 hs in medium without FCS. The cells are again washed and exposed to FCS-free medium containing 100 nM FVIIa for 1 h. CRL2091 cells (ATCC) are grown in Is- cove's modified Dulbecco's medium containing 10% FBS, 100 U/ml penicillin, and 100 μg/ml streptomycin to 95-100% confluence. Subsequently, the cells are serum-starved for 16-18 hs and treated with FBS-free medium containing 100 nM FVIIa for 6 hs. Murine 3T3-L1 cells (ATCC) are maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 100 U/ml penicillin, and 100 μg/ml streptomycin. Cells are grown to confluence and induced to with media containing 1 μM dexamethasone (Sigma), 10 μg/ml human insulin (Novo Nordisk A/S), and 1 μM BRL49653 (Novo Nordisk A/S) for 1 h.
Cloning of fragments for Northern blot analyses.
Fra-1 is cloned by reverse transcription PCR from RNA isolated from 3T3-L1 cells treated for
1 h with dexamethasone, insulin, and BRL49653 using the superscript II kit (Life Technolo- gies) according to the manufacturer's instructions. Id2 and Cyr61 are cloned by reverse transcription PCR from RNA isolated from BHK-TF cells treated for 1 h with FVIIa and from CRL2091 cells treated for 6 hours with FVIIa, respectively. The upstream and downstream primers are: 5'-GCGGCCGCCATGTACCGAGACTACGGGGAACCG-3' and 5'- GCGGCCGCTCACAAAGCCAGGAGTGTAGG-3' for Fra-1 , 5'- CAGCATGAAAGCCTTCAGTC-3' and 5'-CTCTGGTGATGCAGGCTGAC-3' for Id2, 5'- CGTCACCCTTCTCCACTTGA-3' and 5'-CTTGGTCTTGCTGCATTTCT-3' for Cyr61. Parameters for PCR are one cycle of denaturing at 94 °C for 10 s, annealing at 65 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, annealing at 64 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, an- nealing at 63 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, annealing at 62 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of dena- turing at 94 °C for 10 s, annealing at 61 °C for 15 s, and extension at 72 °C for 1.5 min, one cycle of denaturing at 94 °C for 10 s, annealing at 60 °C for 15 s, and extension at 72 °C for 1.5 min, 40 cycles of denaturing at 94 °C for 10 s, annealing at 55 °C for 15 s, and extension at 72 °C for 1.5 min. All fragments are cloned into into TOPO 2.1 (Invitrogen) and sequenced using a Megabase sequencer.
Northern blot analysis.
Total RNA are isolated from BHK-TF cells incubated with FVIIa, FX, ASIS, 1F44A1 or TFδ-
5G9 using TriZol following the instructions of the vendor. 20 μg of RNA are size-fractionated in a denaturing gel containing 1% agarose, 20 mM MOPS, 5 mM NaOAc, 6% formaldehyde, and 1 mM EDTA, transferred to a Hybond N+ membrane (Amersham) by capillary blotting and immobilized by UV crosslinking. cDNA encoding Fra-1 , Id2 or Cyr61 are labelled with the Prime It kit (Stratagene) using [α-32P] dATP (Amersham) and hybridized using Express Hyb (Clontech) following the manufacturer's instructions and results are visualized by autoradio- graphy.
Example 6 (Assay 16).
MAPK assay via the Elk1 transcription factor/Luciferase reporter (PathDetect)
HeLa cells are seeded to 40 % confluence in a T-80 flask one day prior to transfec- tion. Cells are transfected with 150 ng pFA-Elk1 (Stratagene), 3 μg pFR-Luc (Stratagene), 3 μg human TF/pcDNA3 and 3 μg mouse Protease Activated Receptor 2/pcDNA3,1+ using 36 μl FuGene (Roche) as described in the manual. The following day the cells are detached by Versene™ (Invitrogen) and seeded in black 96 well view plates (Packard) at a cell density of 20.000 cells per well. After the cells had reattached to the plate, the medium is replaced with 160 μl per well serum-free Dulbeccos Modified Eagle Medium (Invitrogen) and incubated for 16 hours.
Cells are preincubated for 1 hour with either 20 μl serum-free medium (control), 20 μl 2,5 μM FFR-rFVIIa (control), 20 μl 2,5 μM anti TF antibody B or 20 μl 2,5 μM anti TF antibody A. 20 μl 0,5 μM FVIIa is added to half of the wells and medium to the other half. Following 4 hours of incubation the cells are subjected to the Luciferase gene assay. LucLite (Packard) reagent is added to the cells as described by the manufacturer. Luciferase expression levels are read on a TopCount Microplate Scintillation (Packard). Example 7:
Preparation of humanized antibodies immunospecific for human TF.
Human monoclonal antibodies are made as described in example 1. These antibod- ies have relatively low affinity to tissue factor compared to murine monoclonal antibodies made against the same epitope. The hyper variable regions of the murine monoclonal antibodies have been sequenced and transferred to a human monoclonal antibody that reacts with the same epitope on tissue factor as well as to a "na'ive" human monoclonal antibody framework. Affinities have been compared between the two antibody frameworks. Antibody generation.
Transgenic mice expressing human antibodies and normal mice (e.g. Balb/C or RBF) are three times immunized with the antigen, here human TF. 10 days after the last immunization the antibody titer were determined and the highest responder of the transgenic mice and the normal mice were selected for fusions. Spleen cells of these mice were isolated and fused to the mouse myeloma cell line AG8.X63 in the case of the transgenic mice and FOX in the case of the normal mice. After 14 days of selection clones were screened for binding towards human TF. Afterwards the affinity of positive clones is measured by BIAcore. mRNA extraction and cDNA synthesis. mRNA from clones of the highest affinity mouse and human antibodies is isolated and used as template for cDNA synthesis by RT-PCR amplification of the hypervariable regions according to standard procedures. Amplification of murine TF antibody variable regions.
TF antibody variable heavy chains are amplified from cDNA using a degenerated primer mix of: 5'- ACTAGTTTTGGCTGAGGAGACGGTGACCGTGG-3', 5'- AC- TAGTTTTGGCTGAGGAGACTGTGAGAGTGG-3', 5'- ACTAGTTTTGGCTGCAGA-
GACAGTGACCAGAG-3', 5'- ACTAGTTTTGGCTGAGGAGACGGTGACTGAGG-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1).
TF antibody variable kappa-light chains are amplified from cDNA using 5'- TCAT- CAACACTCATTCCTGTTGAAGCTCTTGA-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1).
Amplification of human TF antibody variable regions.
HuTF antibody variable heavy chains are amplified from cDNA of using primer 5'- GTGCCAGGGGGAAGACCGATGGG-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1). HuTF antibody variable kappa-light chains are amplified from cDNA using primer 5'- GCAGGCACACAACAGAGGCAGTTCCAGATTTC-3' and a universal primer mix present in SMART RACE kit supplied by Clontech (kat.# K1811-1). Cloning of TF antibodies into E. coli The PCR product of the TF antibody variable chains are TOPO-cloned according to manufactures protocol (Invitrogen kat.#45-0030) and transformed in competent E. coli cells. DNA is extracted from transformants and the variable regions are characterized by sequencing. Genetic modification of HuTF antibody hyper variable regions. The CDR's are characterized by sequencing from which conserved consensus motifs can be derived. Based on the sequence data obtained from HuTF and murine TF antibody variable regions new CDRs are engineered and introduced into the HuTF antibody framework. The new CDRs are created in the variable regions by extension PCR and the engineered variable regions are introduced into IgG expression vectors plESRgl , plESRglf, and plESRg4 (provided by Medarex) in the following manner: Variable light chains introduced into Hind 1 '11/BsiWi site and variable heavy chains introduced into Notl/Nhel site of the vectors. Expression of recombinant TF antibodies.
The resultant plasmids are analyzed by sequencing and transfected into mammalian cells according to standard procedures. Antibodies are purified from the supernatant by protein A and affinities between those recombinant generated antibodies are compared.

Claims

1. An isolated humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
2. The isolated humanized antibody according to claim 1 , wherein the CDR amino acid sequences of said humanized antibody are derived from a parent monoclonal antibody.
3. The isolated humanized antibody according to claim 2, wherein said parent monoclonal antibody is a mouse monoclonal antibody.
4. The isolated humanized antibody according to any one of the claims 1-3, wherein said humanized antibody is a Fab fragment.
5. The isolated humanized antibody according to any one of the claims 1-3, wherein said humanized antibody is a F(ab)2 fragment.
6. The isolated humanized antibody according to any one of the claims 1-3, wherein said humanized antibody is a F(ab')2 fragment.
7. The isolated humanized antibody according to any one of the claims 1-3, wherein said humanized antibody is a single chain Fv fragment.
8. The isolated humanized antibody according to any one of the claim 1-7, wherein said hu- manized antibody has a Kd for binding to human TF within the range of 10"15- 10"8 M.
9. The isolated humanized antibody according to any one of the claim 1-8, wherein said humanized antibody has a Kd for binding to human TF within the range of 10"15- 10"10 M.
10. The isolated humanized antibody according to any one of the claim 1-8, wherein said humanized antibody has a Kd for binding to human TF within the range of 10"15- 10"12 M.
11. The isolated humanized antibody according to any one of the claim 1-10, wherein the human framework amino acid sequences of said humanized antibody are derived from a human antibody, that immunoreacts with a second epitope present on human TF.
12. The isolated humanized antibody according to claim 11 , wherein said second epitope comprises an amino acid residue at a particular position of human TF, which amino acid residue at this particular position of human TF is also comprised within the first epitope with which said humanized antibody immunoreacts.
13. The isolated humanized antibody according to claim 12, wherein said amino acid residue at a particular position of human TF is selected from the list consisting of Trp45, Lys46 and Tyr94.
14. The isolated humanized antibody according to any one of the claim 1-11 , wherein said first epitope or second epitope comprises an amino acid residue selected from the list consisting of Trp45, Lys46 and Tyr94.
15. A pharmaceutical composition comprising a therapeutically effective amount of a human- ized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
16. The pharmaceutical composition comprising a therapeutically effective amount of a humanized antibody, that immunoreacts with an epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF, wherein said humanized antibody is according to any one of the claims 1-14.
17. A composition comprising a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
18. The composition comprising a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF, wherein said humanized antibody is according to any one of the claims 1-14.
19. A method for treatment of a FVlla/TF related disorder in a human, which method comprises administering to said human a therapeutically effective amount of a humanized antibody, that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
20. A method for treatment of a FVlla/TF related disorder in a human, which comprises administering to said human a therapeutically effective amount of the humanized antibody according to any one of the claims 1-14.
21. A method for preparation of a humanized antibody, which method comprises a) preparation of humanized antibodies against human TF, b) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of
FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, or testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding, or testing antibodies in a FVIIa signalling assay and selecting humanized antibodies which inhibits FVIIa-induced intracellular signalling.
22. The method according to claim 21 , which comprises testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR- rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa.
23. The method according to any one of the claims 21-22, which comprises testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding.
24. The method according to any one of the claims 21-23, which comprises testing antibod- ies in a FVIIa signalling assay and selecting humanized antibodies which inhibits FVIIa- induced intracellular signalling.
25. A humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF and are produced by a method that comprises : a) preparation of humanized antibodies against human TF, b) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, or testing antibodies in a FVIIa competition assay and selecting humanized antibody which compete with FVIIa binding, or testing antibodies in a FVIIa signalling assay and selecting humanized antibodies which inhibits FVIIa-induced intracellular signalling.
26. A method for preparation of a humanized antibody, which method comprises a) preparation of immortal cells to secrete humanized antibodies, b) isolation of culture medium from immortal cells comprising produced humanized antibodies, c) testing antibodies in an indirect TF ELISA assay comprising TF in solution and selecting humanized antibodies that immunoreacts with human TF in solution, d) testing antibodies in a FVIIa competition assay and selecting humanized antibodies which competes with FVIIa binding, e) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 10 nm FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR- rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, f) testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC50 value lower than the IC50 value of FFR- rFVIIa + 100 nM (using 0.1 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 10 nM preferably lower than the IC50 value of FFR-rFVIIa + 5 nM, preferably lower than the IC50 value of FFR-rFVIIa + 1 nM, more preferably lower than the IC50 value of FFR-rFVIIa + 0.1 nM, more preferably lower than the ICso value of FFR-rFVIIa, g) testing antibodies in a TF-induced clot assay and selecting humanized antibodies which inhibits clot formation in this assay with an IC50 value lower than the IC50 value of FFR-rFVIIa + 1 nM, such as lower than the IC50 value of FFR-rFVIIa +
500 pM, preferably lower than the IC50 value of FFR-rFVIIa + 200 pM, preferably lower than the IC50 value of FFR-rFVIIa + 100 pM, such as lower than the IC50 value of FFR-rFVIIa + 50 pM, preferably lower than the IC50 value of FFR-rFVIIa + 10 pM, more preferably lower than the IC50 value of FFR-rFVIIa + 5 pM, more preferably lower than the IC50 value of FFR-rFVIIa, h) selection and cultivation in a suitable culture medium of the immortal cell producing the selected antibody following step c - g, i) isolation of selected antibody from culture medium of selected immortal cell.
27. The method according to claim 26, wherein said method further comprises testing anti- bodies in a direct TF ELISA assay comprising immobilized TF and selecting humanized antibodies that immunoreacts with immobilized human TF.
28. The method according to any one of the claims 26-27, wherein said method further comprises testing antibodies in a FXa generation assay on TF expressing cell and selecting hu- manized antibodies which inhibits FXa generation on TF expressing cell with an IC50 value lower than the IC50 value of FFR-rFVIIa + 500 nM (using 1 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 100 nM, preferably lower than the IC50 value of FFR-rFVIIa + 50 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa + 5 nM, more preferably lower than the IC50 value of FFR-rFVIIa.
29. The method according to any one of the claims 26-28, wherein said method further comprises testing antibodies in a whole cell TF binding assay and selecting humanized antibodies which competes with FVIIa binding to human TF expressed on the cell surface of whole cells.
30. The method according to any one of the claims 26-29, wherein said method further comprises testing antibodies in a biosensor assay and selecting humanized antibodies with Kd for binding to human TF lower than 100 nM, such as lower than 10 nM, preferably lower than 5 nM preferably lower than 1 nM, more preferably lower than 0.5 nM.
31. The method according to any one of the claims 26-30, wherein said method further comprises testing antibodies in a FVIIa signalling assay and selecting humanized antibodies which inhibits FVIIa-induced intracellular signalling.
32. The method according to any one of the claims 26-31, wherein said method further comprises testing antibodies in an epitope mapping assay and selecting humanized antibodies that immunoreacts with preferred epitopes on TF.
33. The method according according to claim 32, wherein said preferred epitope comprises an amino acid residue selected from the list consisting of Trp45, Lys46 and Tyr94.
34. The method according to any one of the claims 26-33, wherein said immortal cell is a mammalian cell.
35. A humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF and are produced by a method that comprises : a) preparation of immortal cells to secrete humanized antibodies, b) isolation of culture medium from immortal cells comprising produced antibodies, c) testing antibodies in an indirect TF ELISA assay comprising TF in solution and selecting humanized antibodies that immunoreacts with human TF in solution, d) testing antibodies in a FVIIa competition assay and selecting humanized antibodies which competes with FVIIa binding, e) testing antibodies in a FVlla/TF amidolytic assay and selecting humanized antibody which inhibit TF-induced FVIIa amidolytic activity, with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 10 nM FVIIa in the assay), such as lower than the IC50 value of FFR-rFVIIa + 40 nM, preferably lower than the IC50 value of FFR-rFVIIa + 20 nM, preferably lower than the IC50 value of FFR-rFVIIa + 10 nM, more preferably lower than the IC50 value of FFR-rFVIIa, f) testing antibodies in a FXa generation assay and selecting humanized antibody which inhibit FXa generation with an IC50 value lower than the IC50 value of FFR-rFVIIa + 100 nM (using 0.1 nM FVIIa in the assay), such as lower than the IC50 value of FFR- rFVIIa + 10 nM, preferably lower than the IC50 value of FFR-rFVIIa + 5 nM, preferably lower than the IC50 value of FFR-rFVIIa + 1 nM, more preferably lower than the IC50 value of FFR-rFVIIa + 0.1 nM, more preferably lower than the IC50 value of FFR- rFVIIa, g) testing antibodies in a TF-induced clot assay and selecting humanized antibodies which inhibits clot formation in this assay with an IC50 value lower than the IC50 value of FFR-rFVIIa + 1 nM, such as lower than the IC50 value of FFR-rFVIIa + 500 pM, preferably lower than the IC50 value of FFR-rFVIIa + 200 pM, preferably lower than the IC50 value of FFR-rFVIIa + 100 pM, such as lower than the IC50 value of FFR- rFVIIa + 50 pM, preferably lower than the IC50 value of FFR-rFVIIa + 10 pM, more preferably lower than the IC50 value of FFR-rFVIIa + 5 pM, more preferably lower than the IC50 value of FFR-rFVIIa, h) selection and cultivation in a suitable culture medium of the immortal cell producing the selected antibody following step c - g, i) isolation of selected antibody from culture medium of selected immortal cell.
36. A humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF produced by a method according to any one of the claims 26-33.
37. A cell producing humanized antibody that immunoreacts with a first epitope present on human TF and inhibits the binding of human coagulation factor Vila to human TF.
38. The cell according to claim 37, wherein said cell is a mammalian cell.
39. The cell according to any one of the claims 37-38, wherein said cell is a selected from the list consisting of CHO, BHK, HEK293, P3X63-Ag8, P3X63-AG8.653, PERC6, NS0, YB2/0, P3/NS1-Ag4-1 (NS-1), Sp2/0-Agl4 and S194/5.XXO.Bu.1,
40. The cell according to any one of the claims 37-39, wherein said humanized antibody is according to any one of the claims 1-14.
41. An isolated humanized antibody, that immunoreacts with an epitope P present on a protein, wherein the human framework amino acid sequences of said humanized antibody are derived from a human antibody, wherein said human antibody immunoreacts with an epitope H present on said protein, said epitope H comprising an amino acid residue at a particular position of said protein, which amino acid residue at this particular position of said protein is also comprised within the epitope P with which said humanized antibody immunoreacts.
PCT/DK2003/000741 2002-10-31 2003-10-31 Humanized tissue factor antibodies WO2004039842A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003277832A AU2003277832A1 (en) 2002-10-31 2003-10-31 Humanized tissue factor antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA200201661 2002-10-31
DKPA200201661 2002-10-31

Publications (2)

Publication Number Publication Date
WO2004039842A2 true WO2004039842A2 (en) 2004-05-13
WO2004039842A3 WO2004039842A3 (en) 2004-09-23

Family

ID=32187133

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2003/000741 WO2004039842A2 (en) 2002-10-31 2003-10-31 Humanized tissue factor antibodies

Country Status (2)

Country Link
AU (1) AU2003277832A1 (en)
WO (1) WO2004039842A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004064870A2 (en) * 2003-01-22 2004-08-05 Novo Nordisk A/S Radiolabelled tissue factor binding agent and the use thereof
EP1633309A2 (en) * 2003-05-30 2006-03-15 Centocor, Inc. Method of inhibiting tumor growth with anti-tissue factor antibodies
WO2007014772A2 (en) * 2005-08-03 2007-02-08 Thrombotargets Europe, S.L. Direct and indirect effector cell protease receptor-1 (epr-1) inhibitors as antiplatelet agents
US7605235B2 (en) 2003-05-30 2009-10-20 Centocor, Inc. Anti-tissue factor antibodies and compositions
EP2376537A2 (en) * 2008-12-09 2011-10-19 Genmab A/S Human antibodies against human tissue factor.
US8722044B2 (en) 2011-03-15 2014-05-13 Janssen Biotech, Inc. Human tissue factor antibody and uses thereof
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
US9708410B2 (en) 2003-05-30 2017-07-18 Janssen Biotech, Inc. Anti-tissue factor antibodies and compositions
CN111727075A (en) * 2017-11-27 2020-09-29 普渡制药公司 Humanized antibodies targeting human tissue factor

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996040921A1 (en) * 1995-06-07 1996-12-19 Ortho Farmaceutical Corporation Cdr-grafted anti-tissue factor antibodies and methods of use thereof
WO1997018838A1 (en) * 1995-11-21 1997-05-29 Athena Neurosciences, Inc. Therapeutic uses of humanized antibodies against alpha-4 integrin
WO1998001757A1 (en) * 1996-07-08 1998-01-15 Cambridge Antibody Technology Limited Labelling and selection of molecules
US6001978A (en) * 1987-03-31 1999-12-14 The Scripps Research Institute Human tissue factor related DNA segments polypeptides and antibodies
EP1069185A1 (en) * 1998-04-03 2001-01-17 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (tf) and process for constructing humanized antibody
WO2001070984A2 (en) * 2000-03-16 2001-09-27 Genentech, Inc. Anti-tissue factor antibodies with enhanced anticoagulant potency
US6461610B1 (en) * 1997-07-18 2002-10-08 Novo Nordisk A/S Methods for modifying cell motility using factor VIIa or inactivated factor VIIa
WO2003029295A1 (en) * 2001-10-02 2003-04-10 Novo Nordisk A/S Human tissue factor antibodies
WO2003037911A2 (en) * 2001-10-29 2003-05-08 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6001978A (en) * 1987-03-31 1999-12-14 The Scripps Research Institute Human tissue factor related DNA segments polypeptides and antibodies
WO1996040921A1 (en) * 1995-06-07 1996-12-19 Ortho Farmaceutical Corporation Cdr-grafted anti-tissue factor antibodies and methods of use thereof
WO1997018838A1 (en) * 1995-11-21 1997-05-29 Athena Neurosciences, Inc. Therapeutic uses of humanized antibodies against alpha-4 integrin
WO1998001757A1 (en) * 1996-07-08 1998-01-15 Cambridge Antibody Technology Limited Labelling and selection of molecules
US6461610B1 (en) * 1997-07-18 2002-10-08 Novo Nordisk A/S Methods for modifying cell motility using factor VIIa or inactivated factor VIIa
EP1069185A1 (en) * 1998-04-03 2001-01-17 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (tf) and process for constructing humanized antibody
WO2001070984A2 (en) * 2000-03-16 2001-09-27 Genentech, Inc. Anti-tissue factor antibodies with enhanced anticoagulant potency
WO2003029295A1 (en) * 2001-10-02 2003-04-10 Novo Nordisk A/S Human tissue factor antibodies
WO2003037911A2 (en) * 2001-10-29 2003-05-08 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FRANK KOLBINGER ET AL: "Humanization of a mouse anti-human IgE antibody: a potential therapeutic for IgE-mediated allergies" PROTEIN ENGINEERING, vol. 6, no. 8, 1993, pages 971-980, XP002271768 *
LEONARD PRESTA ET AL: "Generation of a Humanized, High Affinity Anti-tissue Factor Antibody for Use as a Novel Antithrombotic Therapeutic" THROMB HAEMOST, vol. 85, 2001, pages 379-389, XP002271767 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004064870A3 (en) * 2003-01-22 2005-04-28 Novo Nordisk As Radiolabelled tissue factor binding agent and the use thereof
WO2004064870A2 (en) * 2003-01-22 2004-08-05 Novo Nordisk A/S Radiolabelled tissue factor binding agent and the use thereof
US8293882B2 (en) 2003-05-30 2012-10-23 Centocor, Inc. Anti-tissue factor antibodies and compositions
EP1633309A2 (en) * 2003-05-30 2006-03-15 Centocor, Inc. Method of inhibiting tumor growth with anti-tissue factor antibodies
EP1633309A4 (en) * 2003-05-30 2007-06-13 Centocor Inc Method of inhibiting tumor growth with anti-tissue factor antibodies
US7605235B2 (en) 2003-05-30 2009-10-20 Centocor, Inc. Anti-tissue factor antibodies and compositions
US9708410B2 (en) 2003-05-30 2017-07-18 Janssen Biotech, Inc. Anti-tissue factor antibodies and compositions
WO2007014772A2 (en) * 2005-08-03 2007-02-08 Thrombotargets Europe, S.L. Direct and indirect effector cell protease receptor-1 (epr-1) inhibitors as antiplatelet agents
WO2007014772A3 (en) * 2005-08-03 2007-04-12 Thrombotargets Europe Sl Direct and indirect effector cell protease receptor-1 (epr-1) inhibitors as antiplatelet agents
CN106632678A (en) * 2008-12-09 2017-05-10 根马布股份公司 Human antibodies against tissue factor
EP2376537B1 (en) * 2008-12-09 2016-09-28 Genmab A/S Human antibodies against human tissue factor.
CN106632678B (en) * 2008-12-09 2022-05-31 健玛保 Human antibodies to tissue factor
US9714297B2 (en) 2008-12-09 2017-07-25 Genmab A/S Human antibodies against tissue factor and methods of use thereof
US9150658B2 (en) 2008-12-09 2015-10-06 Genmab A/S Human antibodies against tissue factor and methods of use thereof
EP2376537A2 (en) * 2008-12-09 2011-10-19 Genmab A/S Human antibodies against human tissue factor.
CN102317319B (en) * 2008-12-09 2016-06-29 根马布股份公司 People's antibody for tissue factor
CN102317319A (en) * 2008-12-09 2012-01-11 根马布股份公司 People's antibody to tissue factor
US9492565B2 (en) 2010-06-15 2016-11-15 Genmab A/S Human antibody drug conjugates against tissue factor
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
US8722044B2 (en) 2011-03-15 2014-05-13 Janssen Biotech, Inc. Human tissue factor antibody and uses thereof
US8999333B2 (en) 2011-03-15 2015-04-07 Janssen Biotech, Inc. Human tissue factor antibody and uses thereof
US8951525B2 (en) 2011-03-15 2015-02-10 Janssen Biotech, Inc. Human tissue factor antibody and uses thereof
CN111727075A (en) * 2017-11-27 2020-09-29 普渡制药公司 Humanized antibodies targeting human tissue factor
CN111727075B (en) * 2017-11-27 2024-04-05 普渡制药公司 Humanized antibodies targeting human tissue factor

Also Published As

Publication number Publication date
WO2004039842A3 (en) 2004-09-23
AU2003277832A8 (en) 2004-05-25
AU2003277832A1 (en) 2004-05-25

Similar Documents

Publication Publication Date Title
WO2003029295A1 (en) Human tissue factor antibodies
ES2702365T3 (en) Use of anti-factor XI antibodies for the prevention or treatment of thrombus formation
AU780775B2 (en) Factor IX/factor IXa antibodies and antibody derivatives
KR100553629B1 (en) Anticoagulant Agents Useful in Treatment of Thrombosis
JP4423680B2 (en) CDR-grafted anti-tissue factor antibody and use thereof
EP1135477B1 (en) Humanized monoclonal integrin antibodies
JP2017158564A (en) Human tissue factor antibodies and uses thereof
JP2021121615A (en) MONOCLONAL ANTIBODY INHIBITORS OF FACTOR XIIa
WO2004039842A2 (en) Humanized tissue factor antibodies
US20060057140A1 (en) Anticoagulant agents useful in treatment of thrombosis
JP2004516236A (en) Antithrombotic agent
US20050169927A1 (en) Human tissue factor antibodies
US20040001830A1 (en) Human tissue factor antibodies
US20050106139A1 (en) Humanized tissue factor antibodies
AU2002333213A1 (en) Human tissue factor antibodies
US7514539B2 (en) Epitope directed selection of antibodies to murine tissue factor
JP6848016B2 (en) Monoclonal antibody against tissue factor pathway inhibitor (TFPI)
TW200407427A (en) Human tissue factor antibodies

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP