WO2005079766A2 - Therapeutic combination comprising a tissue factor antagonist and anti-cancer compounds - Google Patents

Therapeutic combination comprising a tissue factor antagonist and anti-cancer compounds Download PDF

Info

Publication number
WO2005079766A2
WO2005079766A2 PCT/DK2005/000098 DK2005000098W WO2005079766A2 WO 2005079766 A2 WO2005079766 A2 WO 2005079766A2 DK 2005000098 W DK2005000098 W DK 2005000098W WO 2005079766 A2 WO2005079766 A2 WO 2005079766A2
Authority
WO
WIPO (PCT)
Prior art keywords
agent
phe
pharmaceutical composition
fviia
antibody
Prior art date
Application number
PCT/DK2005/000098
Other languages
French (fr)
Other versions
WO2005079766A3 (en
Inventor
Jørn Roland MÜLLER
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 EP05700648A priority Critical patent/EP1744734A2/en
Priority to JP2006553435A priority patent/JP2007523099A/en
Publication of WO2005079766A2 publication Critical patent/WO2005079766A2/en
Publication of WO2005079766A3 publication Critical patent/WO2005079766A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones
    • A61P5/20Drugs for disorders of the endocrine system of the parathyroid hormones for decreasing, blocking or antagonising the activity of PTH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to novel compositions comprising a combination of a compounds which binds to and inhibits the activity of tissue factor and another compound, which is an anti-cancer compound.
  • the invention also relates to the novel pharmaceutical compositions as well as their use in the prophylaxis or treatment of diseases or disorders related to pathophysiological tissue factor (TF) functions including cancer, inflammation, atherosclerosis and ischemia/reperfusion.
  • TF pathophysiological tissue factor
  • Tissue Factor is a cellular transmembrane receptor for plasma coagulation factor Vila (FVIIa) and formation of TF/FVIIa complexes on the cell surface triggers the coagulation cascade in vivo.
  • the TF/FVIIa complex efficiently activates coagulation factors IX and X.
  • the resultant protease factor Xa (FXa) activates prothrombin to thrombin, which in turn converts fibrinogen into a fibrin matrix.
  • TF is constitutively expressed on the surface of many extravascuiar cell types that are not in contact with the blood, such as fibroblasts, pericytes, smooth muscle cells and epithelial cells, but not on the surface of cells that come in contact with blood, such as en- dothelial cells and monocytes.
  • TF is also expressed in various pathophysiological conditions where it is believed to be involved in progression of disease states within cancer, inflammation, atherosclerosis and ischemia/reperfusion.
  • TF is now recognised as a target for therapeutic intervention in conditions associated with increased expression.
  • FVIIa is a two-chain, 50 kilodalton (kDa) vitamin-K dependent, plasma serine protease which participates in the complex regulation of in vivo haemostasis.
  • FVIIa is generated from proteolysis of a single peptide bond from its single chain zymogen, Factor VII (FVII), which is present at approximately 0.5 ⁇ g/ml in plasma.
  • the zymogen is catalytically inactive. The conversion of zymogen FVII into the activated two-chain molecule occurs by cleavage of an internal peptide bond.
  • FVIIa binds with high affinity to exposed TF, which acts as a cofactor for FVIIa, enhancing the proteolytic activation of its substrates FVII, Factor IX and FX.
  • TF was recently shown to function as a mediator of intracellular activities either by interactions of the cytoplasmic domain of TF with the cytoskeleton or by supporting the FVI la-protease dependent signaling. Such activities may be responsible, at least partly, for the implicated role of TF in tumor development, metastasis and angiogenesis.
  • Cellular exposure of TF activity is advantageous in a crisis of vascular damage but may be fatal when exposure is sustained as it is in these various diseased states.
  • FVIIai Inactivated FVII
  • FVIIai is FVIIa modified in such a way that it is catalytically inactive.
  • FVIIai is not able to catalyze the conversion of FX to FXa, but still able to bind tightly to TF in competition with active endogenous FVIIa and thereby inhibit the TF function.
  • 97/47651 relates to FVIIai and the uses thereof.
  • International patent applications WO 90/03390, WO 95/00541 , WO 96/18653, and European Patent EP 500800 describe peptides derived from FVIIa having TF/FVIIa antagonist activity.
  • International patent application WO 01/21661 relates to bivalent inhibitor of FVII and FXa. Hu Z and Garen A (2001) Proc. Natl. Acad. Sci. USA 98; 12180-12185, Hu Z and
  • the present invention relates in a broad aspect to combination treatment with TF antagonists.
  • the present invention provides a pharmaceutical composition which comprises of a TF antagonist and an anti-cancer compound.
  • the present invention also relates to a use of the pharmaceutical composition in the treatment of diseases and disorders related to TF antagonist.
  • the present invention relates to a pharmaceutical composition useful for preventing or treating a disease or disorder associated with pathophysiological TF function, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from the first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , analogues or derivatives thereof.
  • the present invention relates to the use of a first agent, which is a TF antagonist in combination with a second agent, which is an anti-cancer compound for the manufacture of a medicament for treating a disease or disorder associated with pathophysiological TF function, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , analogues or derivatives thereof.
  • the present invention relates to a method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising administering to a mammal in need of such a treatment a therapeutically effective amount of a pharmaceutical composition, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , analogues or derivatives thereof.
  • a pharmaceutical composition comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different
  • the present invention relates to method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising (i) administering to a mammal in need of such a treatment a therapeutically effective amount of a first agent, which is a TF antagonist, and (ii) administering to a mammal in need of such a treatment a therapeutically effective amount of a second agent different from the first agent, which is an anti-cancer compound, and optionally iii) administering to a mammal in need of such a treatment a therapeutically effective amount of one or more further agents different for said first and second agent, which is an anti-cancer compound, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , ana- logues or derivatives thereof.
  • the present invention relates to a pharmaceutical kit comprising a first agent, which is a TF antagonist, and a second agent, which is an anti-cancer compound, and optionally a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutical composition useful for preventing or treating a disease or disorder associated with pathophysiological TF function comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from the first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient.
  • the present invention relates to the use of a first agent, which is a TF antagonist in combination with a second agent, which is an anti-cancer compound for the manufacture of a medicament for treating a disease or disorder associated with pathophysiological TF function.
  • the present invention relates to a method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising administering to a mammal in need of such a treatment a therapeutically effective amount of a pharmaceutical composition, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutical composition comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient.
  • the present invention relates to method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising (i) administering to a mammal in need of such a treatment a therapeutically effective amount of a first agent, which is a TF antagonist, and (ii) administering to a mammal in need of such a treatment a therapeutically effective amount of a second agent different from the first agent, which is an anti-cancer compound, and optionally iii) administering to a mammal in need of such a treatment a therapeutically effective amount of one or more further agents different for said first and second agent, which is an anti-cancer compound.
  • the present invention relates to TF antagonists in combination with another anti- cancer drug.
  • the TF antagonists bind TF with high affinity and specificity but do not initiate blood coagulation.
  • the TF antagonist is factor FVIIa polypeptides chemically inactivated in the active site.
  • the TF antagonist is an antibody against TF.
  • the TF antagonist is a fully human antibody against TF.
  • the human antibody immunoreacts with an epitope present on human 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. In one embodiment the preferred epitope comprises the residue Tyr94.
  • the isolated human antibody binds to an epitope within the interface between TF and FVIIa. In one embodiment the antibody is a monoclonal antibody. In one embodiment the antibody is a human monoclonal antibody. In one embodiment the antibody is an antibody against human TF.
  • TF antagonist or “TF antagonists”, as used herein is intended to mean any compound which binds directly to TF and inhibits TF-mediated FVIIa activity.
  • TF-mediated FVIIa activity means any TF-dependent activity.
  • TF-mediated coagulation activity e.g. TF-mediated MAPK signaling.
  • TF-mediated FVIIa activity is MAPK signaling.
  • TF-mediated MAPK signaling is intended to mean a cascade of intracellular events that mediate activation of Mitogen-Activated-Protein-Kinase (MAPK) or homo- logues thereof in response to the binding of a FVII polypeptide to TF.
  • MAP kinases 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. MAPK signaling may be measured as described in assay 5.
  • 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.
  • TF antagonists include but are not limited to factor FVIIa polypeptides chemically inactivated in the active site, antibodies against TF, e.g. a monoclonal antibody, such as a human monoclonal antibody against human TF.
  • Methods of preparing human antibodies against human TF is described in International patent application 03/029295 the content of which is hereby incorporated be reference in its entirety.
  • International patent applications 03/076461 , DK03/00481 , and DK03/00480 describe the preparation of different TF antagonist that may be used according to the invention the content of which is hereby incorporated by reference in its entirety.
  • 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 as used herein, is intended to refer to immunoglobulin molecules and fragments thereof that have the ability to specifically bind to an antigen (e.g., hu- man TF).
  • Full-length antibodies comprise 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 hypervariabil- ity, 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 or- der: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • 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.
  • 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 (Ward et al., (1989) Nature 341 :544-546 ), which consists of a VH domain; and (vi) an isolated complementarity de- termining region (CDR).
  • a Fab fragment a monovalent fragment , consisting of the VL, VH, CL and CH I domains
  • F(ab) 2 and F(ab') 2 fragments a
  • 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).
  • single chain Fv single chain Fv
  • 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.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using 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 com- plementary domains of another chain and creating two antigen binding sites (see e.g., Hol- liger, 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, or the like.
  • the terms "human antibody”, “human antibodies”, “human TF antibody”, and “hu- man TF antibodies”, as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • 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 mutagene- sis 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 or human/mouse chimera antibodies.
  • an "isolated human antibody”, as used herein, is intended to refer to a human anti- body 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.
  • the terms “immunoreacts” or “immunoreacting” are used where appropriate interchangeably with the term “ specifically bind”.
  • inhibitors means any reduction compared to a refer- ence.
  • an antibody, which inhibits the binding of human coagulation factor Vila to human TF means any antibody, which 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.
  • the affinity constant K a is defined by 1/K d .
  • Preferred methods for determining Mabs 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.
  • the TF antagonist is a human antibody that inhibits FVIIa- induced activation of the MAPK signalling with 98 %.
  • the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 90 %. In one embodiment the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 70 %. In one embodiment the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 50 %. In one embodiment the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 30 %.
  • the term "FVIIa-induced activation of the MAPK signaling" is intended to indicate that FVIIa binds to TF in a mammalian cell and thereby induce MAPK signaling.
  • MAPK signalling is intended to mean 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.
  • TF-mediated coagulation activity means coagulation initiated by TF through the formation of the TF/FVIIa complex and its activation of FIX and Factor X to FlXa and FXa, respectively.
  • TF-mediated coagulation activity is measured in a FXa generation assay.
  • FXa generation assay as used herein is intended to mean any assay where activation of FX is measured in a sample comprising TF, FVIIa, FX, calcium and phospholip- ids.
  • An example of a FXa generation assay is described in assay 1.
  • Examples of TF antagonist includes, but at not limited to FVIIai and inhibitory antibodies against TF.
  • the inactivation of the FVIIa proteolytic activity in the FVIIai molecule is obtained in vitro by covalent active site inhibitors e.g. chloromethyl ketones.
  • the FVIIai molecule has increased affinity for TF as compared to the binding of native FVII.
  • TF presenting cell or "TF presenting cells” as used herein refers to the presence of TF protein on a cell surface plasma membrane.
  • the cell membrane, where TF is located may be the cell, where TF was synthezised by protein synthesis or it may be a cell, that contain shedded TF protein synthezised by another cell.
  • the term "disease or disorder associated with pathophysiological TF function" as used herein means any disease or disorder, where TF is involved.
  • Thrombotic and Coagulopathic related diseases or disorders, Respiratory diseases or disorders, and Inflammatory diseases or disorders include deep venous thrombosis, chronic thromboembolic diseases or disorders associated with fibrin formation, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, cancer, tumour metastasis, pathological angiogenesis, thrombolysis, arteriosclerosis and restenosis following angioplastry, acute and chronic indications such as inflammation, septic chock, septice- mia, hypotension, acute lung injury (ALI), Acute Respiratory Distress Syndrome (ARDS), pulmonary embolism, disseminated intravascular coagulation (DIC),
  • ALI acute lung injury
  • ARDS Acute Respiratory Distress Syndrome
  • DIC disseminated intravascular coagulation
  • the diseases or disorders are Respiratory disease and Inflammatory disease.
  • Respiratory disease and Inflammatory disease include lower respiratory diseases such as systemic inflammatory response syndrome, asthma, bronchitis, acute lung injury, acute resporatory distress syndrome, idiopathic pulmonary fibrosis, pneumonia, pulmonary edema, pulmonary obstructive disease, endotoxin induced lung damage, non cell lung cancer; inflammatory bowel disease, sepsis, septic shock, acute respiratory distress syndrome, pancreatitis, trauma-induced shock, bronchial asthma, allergic rhinitis, rheumatoid arthritis, cystic fibrosis, stroke, acute bronchitis, chronic bronchitis, acute bronchiolitis, chronic bronchiolitis, osteoarthritis, gout, spondylarthropathris, ankylosing spondylitis, Reiter's syndrome, psoriatic arthropathy, enterapathric spond
  • lower respiratory diseases
  • the diseases or disorders are Thrombotic or Coagulopatic related diseases or disorders.
  • Thrombotic or Coagulopatic related dis- ease include vascular diseases and inflammatory responses such as deep venous thrombo- sis, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, tumour metastasis, inflammation, septic chock, hypotension, acute lung injury (ALI), Acute Respiratory Distress Syndromr (ARDS), pulmonary embolism, disseminated intravascular coagulation (DIC), sepsis, sys- temic inflammatory response syndrome (SIRS), vascular restenosis, platelet deposition, myocardial infarction, angiogenesis, or the treatment of mammals with atherosclerotic vessels at risk for thrombosis, and multiple organ failure resulting from any of the preceding pathologic processes.
  • the disease or disorder associated with patho include vascular
  • TF/FVIIa related processes such as coagulation that may result from the extracorporeal 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 disease or disorder associated with pathophysiological TF function is selected from the group consisting of cancer, tumor growth, tumor metastasis, and pathological angiogenesis.
  • cancer or "tumor growth” are to be understood as referring to all forms of neoplastic cell growth, including both cystic and solid tumors, bone and soft tissue tumors, including both benign and malignant tumors, including tumors in anal tissue, bile duct, blad- der, blood cells, bone, bone (secondary), bowel (colon & rectum), brain, brain (secondary), breast, breast (secondary), carcinoid, cervix, children's cancers, eye, gullet (oesophagus), head & neck, kaposi's sarcoma, kidney, larynx, leukaemia (acute lymphoblastic), leukaemia (acute myeloid), leukaemia (chronic lymphocytic), leukaemia (chronic myeloid), leukaemia (other), liver, liver (secondary), lung, lung (secondary), lymph nodes (secondary), lymphoma (hodgkin's), lymphom
  • Soft tissue tumors include Benign schwannoma Monosomy, Desmoid tumor, Lipo- blastoma, Lipoma, Uterine leiomyoma, Clear cell sarcoma, Dermatofibrosarcoma, Ewing sarcoma, Extraskeletal myxoid chondrosarcoma, Liposarcoma myxoid, Liposarcoma, well differentiated, Alveolar rhabdomyosarcoma, and Synovial sarcoma.
  • Specific bone tumor include Nonossifying Fibroma, Unicameral bone cyst, Enchon- droma, Aneurysmal bone cyst, Osteoblastoma, Chondroblastoma, Chondromyxofibroma, Ossifying fibroma and Adamantinoma, Giant cell tumor, Fibrous dysplasia, Ewing's Sarcoma, Eosinophilic Granuloma, Osteosarcoma, Chondroma, Chondrosarcoma, Malignant Fibrous Histiocytoma, and Metastatic Carcinoma.
  • Leukaemias referes to cancers of the white blood cells which are produced by the bone marrow.
  • the medicament is formulated for intravenous administration, preferably injection or infusion, in particular injection.
  • the medicament is formulated in single-unit dosage form; in an- other it is formulated in the form of a first unit dosage form comprising a preparation of a TF antagonist and a second unit dosage form comprising an anti-cancer compound.
  • the TF antagonist and the anti-cancer compound are present in the pharmaceutical formulation in a ratio by mass of between about 1000:1 and about 1:1000 (w/w).
  • treatment means the administration of an effective amount of a therapeutically active compound of the invention with the purpose of preventing any symptoms or disease state to develop or with the purpose of curing or easing such symptoms or disease states already developed.
  • treatment is thus meant to include prophylactic treatment.
  • the first agent is an inactive FVIIa polypeptide.
  • the first agent is an inactive FVIIa polypeptide is native human FVIIa or a fragment thereof catalytically inactivated in the active site.
  • the inactive FVIIa polypeptide is native human FVIIa catalytically inactivated in the active site.
  • the FVIIa polypeptide is catalytically inactivated in the active site with a chloromethyl ketone inhibitor independently selected from the group consisting of Phe-Phe-Arg chloromethyl ketone, Phe-Phe-Arg chloromethylketone, D-Phe- Phe-Arg chloromethyl ketone, D-Phe-Phe-Arg chloro-methylketone Phe-Pro-Arg chloromethylketone, D-Phe-Pro-Arg chloromethylketone, Phe-Pro-Arg chloromethylketone, D-Phe- Pro-Arg chloromethylketone, L-Glu-Gly-Arg chloro-methylketone and D-Glu-Gly-Arg chloro- methylketone, Dansyl-Phe-Phe-Arg chloromethyl ke-tone, dansyl-Phe-Phe-Arg chloromethylketone, Dansyl-D-Phe-Phe-Arg chloromethyl ketone, Dansyl-D-Phe-Phe-Arg chloromethylketone,
  • the first agent is an antibody which immunoreacts with an epitope present on human TF.
  • the antibody inhibits the binding of human coagulation factor Vila to human TF.
  • the epitope comprises one or more of the residues Trp45, Lys46 and Tyr94.
  • the antibody is a monoclonal antibody.
  • the antibody is a recombinant antibody.
  • the antibody is a Fab fragment.
  • the antibody is a F(ab)2 fragment.
  • the antibody is a F(ab')2 fragment.
  • the an- tibody is a single chain Fv fragment.
  • the antibody has a Kd for binding to human TF within the range of 10 "15 - 10 "8 M. In one embodiment the antibody has a Kd for binding to human TF within the range of 10 "15 - 10 "10 M.
  • the second agent is selected from the group consisting of protein ionophores, cytostatica, che-motherapeutic compound, compounds which induce apoptosis, compound containing ra-dionuclides, antisense nucleotide molecules independent selected from the group consisting of PNAs, DNAs, RNAs and LNAs. In one embodiment of the invention, the second agent comprises a cytotoxic protein or peptide.
  • the disease or disorder associated with patho- physiological TF function are deep venous thrombosis, chronic thromboembolic diseases or disorders associated with fibrin formation, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, cancer, tumour metastasis, pathological angiogenesis, thrombolysis, arteriosclerosis and restenosis following angioplastry, acute and chronic indications such as in- flammation, septic chock, septicemia, hypotension, acute lung injury (ALI), Acute Respiratory Distress Syndrome (ARDS), pulmonary embolism, disseminated intravascular coagulation (DIC), sepsis, systemic inflammatory response syndrome (SIRS), vascular restenosis, platelet deposition, myocardial infarction, angiogenesis, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis; asthma,
  • the second agent is a cytostatica. In one embodiment of the invention, the second agent is a chemotherapeutic compound. In one embodiment of the invention, the second agent is a compound containing radionuclides. In one embodiment of the invention, the second agent is an antisense nucleotide molecule. In one embodiment of the invention, the second agent is melphalan. In one embodiment of the invention, the second agent is a compound containing I 125 . In a further embodiment of the invention, the second agent is a cytotoxic protein or peptide.
  • the second agent comprises the amino acid sequence (KLAKLAK) n , wherein n is selected from the group consisting of 1 , 2, 3, 4, 5, 6, 7, and 8. In one embodiment n is 2. In a further embodiment of the invention, the second agent has the amino acid se- quence (KLAKLAK) 2
  • anti-cancer compound refers to a compound effective in the treatment of cancer. This includes compounds, which kill the tumor cells and/or reduce the size of the tumor and/or reduce the growth and/or spreading of the tumor. The term also encompasses traditional chemotherapeutic drugs and cytotoxic drugs.
  • Other exemplary anti-cancer compounds include, e.g., neomycin, podophyl- lotoxin(s), TNF-alpha, calcium ionophores, calcium-flux inducing compounds, anti-tubulin drugs, colchicine, taxol, vinblastine, vincristine, vindescine, and combretastatin.
  • the anti-cancer compound is an antibody, such as monoclonal antibodies.
  • MAbs have been developed for the treatment of leukaemia and lymphoma as well as solid tumor, and this principle is gaining increasing interest. These antibodies work either by inhib-iting functions that are vital for survival of the tumor cells, by delivering a toxic pay- load, by interrupting key signalling events, or by induction of ADCC or CDC against the tumor cells. Death of the tumor cells might then lead to the release of tumor antigens that "vacci- nates" the immune system and stimulates it to produce a secondary response that then targets the tumor cell (i.e. 'internal vaccination' as described below).
  • the anti-cancer compound is an antibody, such as Rituximab, Alemtuzumab, Trastuzumab, HuMax-CD20, HuMax-EGFr, Zamyl, Pertuzu- mab, antibodies against tissue factor, killer Ig-like receptors (KIR) and laminin-5.
  • the anti-cancer compound is further combined with additional antibodies, such as Rituximab, Alemtuzumab, Trastuzumab, HuMax-CD20, HuMax-EGFr, Zamyl, Pertuzu- mab, antibodies against tissue factor, killer Ig-like receptors (KIR) and laminin-5.
  • the anti-cancer compound is further combined with additional
  • ADCC-enhancing compounds e.g. blocking anti-KIR antibodies or activating NKG2A antibodies or IL-2.
  • the anti-cancer compound is an antibody against viral antigens.
  • the anti-cancer compound is a regulator of cell cycle control and/or apoptosis. A series of regulators are involved in the maintenance of normal cell-cycle.
  • drugs that interfere with apoptotic pathways are within the present invention.
  • TNF-related apoptosis-inducing ligand (TRAIL)/apoptosis-2 ligand (Apo-2L) antibodies that activate TRAIL receptors, IFN ⁇ and anti-sense Bcl-2.
  • the compounds are selected from the group comprising cdc-25, NSC 663284, flavopiridol, 7-hydroxystaurosporine, roscovitine , BIBR1532 SOT-095, TNF-related apoptosis-inducing ligand (TRAIL)/apoptosis-2 ligand (Apo-2L), antibodies that activate TRAIL receptors, IFN ⁇ and anti-sense Bcl-2.
  • the anti-cancer compound is a growth factor inhibitor, such as an antibody against a growth factor. A number of mAbs against growth factors and growth factor receptors are being developed for the treatment of cancer.
  • members of the epi- dermal growth factor receptor (EGF-R) family are abnormally activated in many epithelial tumors, which often correlate with more aggressive clinical course.
  • Antibodies directed against the extracellular ligand binding domain of these receptors and low molecular weight molecules that inhibit their tyrosine kinase domains are in late-stage clinical development or approved for treatment of cancer either as single compounds or in combination with other can- cer drugs.
  • Non-limiting examples are Herceptin (monoclonal antibody), cetuximab (monoclonal antibody), Tarceva (low molecular weight inhibitor), and Iressa (low molecular weight inhibitor).
  • the ligand can be neutralised before binding to the receptor.
  • the growth factor inhibitors are selected from the group comprising Herceptin (monoclonal antibody), cetuximab (monoclonal antibody), Tarceva (low molecular weight inhibitor), and Iressa (low molecular weight inhibitor).
  • the anti-cancer compound is an anti- angiogenesis drug.
  • the anti-cancer compound is an anti-metastatic compound. Tumor growth is dependent on sufficient blood supply and hence development of new blood vessels. This general feature of solid tumors seems attractive from a therapeutic point of view, i.e. reduced tumor growth and tumor regression is expected when treating patients wth cancer with anti-angionesis drugs.
  • the anti-angiogenic compounds is selected from the group consisting of endostatin, angiostatin, antibodies that block factors that initiate angiogenesis (e.g. anti-VEGF - Avastin), low mo- lecular compounds that inhibit angiogenesis by inhibiting key elements in relevant signal transduction pathways. Attacking the vasculature of the tumor and the extracellular matrix has attracted increasing awareness.
  • the anti-angiogenesis drug is selected from the group consisting of avastin, neovastat, thalidomide, PTK787, ZK222584, ZD-6474 , SU6668, PD547.632, VEGF-Trap, CEP-7055, NM-3, SU11248. (Nature Biotech 20, 1067-1068).
  • the anti-cancer compound is a viral targeting compound.
  • the anti-cancer compound is a hormonal compounds. Hormonal compounds are primarily know in the treatment of hormonal dependent cancers such as ovarian cancer, breast cancer and prostate cancer such as anti-androgen and anti-oestrogen therapy.
  • a) Adjuvants Immunotherapy consist of specific and non-specific modalities. As examples of nonspecific immunotherapy are adjuvants acting primarily as catalyst for the initiation of an immune response. Non-limiting examples of such vaccine adjuvants are QS21 , GM-CSF and CpG oligodeoxynucleotides, lipopolysaccharide and polyinosinic:polycytidylic acid.
  • the TF antagonist is combined with one or more adjuvants.
  • the adjuvants are selected from the group consisting of QS21, GM-CSF and CpG oligodeoxynucleotides, lipopolysaccharide and polyinos- inic:polycytidylic acid, b) Cytokines
  • cytokines are IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , IL-2, PEG-IL-2, IL-4; IL-
  • cytokine 6
  • IL-7 IL-12, IL-13, IL-15, IL-18, IL-21 , IL-23, IL-27, IL-28a, IL-28b, IL-29, GM-CSF, Flt3 ligand or stem cell factor.
  • Other compounds that may be combined with a cytokine include autologous TILs, Cis-platin, tamoxifen, DTIC, Carmustine, carboplatin, Vinblastine, temo- zolomide, Vindesine, 5-fluorouracil, Fotemustine, autologous LAK cells, and Gemcitabine.
  • Cell adoptive therapy may include isolation of cells that can stimulate or exert an anti-cancer response from patients and expand these into higher number and reintroduce into the patient.
  • this may be CD4 + or CD8 + T cells recognizing tumor specific antigens or tumor-associated antigens.
  • this may be B cell expressing antibodies specific for tumor specific antigens or tumor-associated antigens.
  • this may be NK cells that are able to kill the tumor cells.
  • this may be dendritic cells (DC) that are cultured in vivo with a DC expanding compound (e.g. GM-CSF or Flt3-L), loaded with tumor specific antigens or tumor-associated antigens and reintroduced in vivo.
  • a DC expanding compound e.g. GM-CSF or Flt3-L
  • the cell adoptive therapy comprises CD4 + or CD8 + T cells recognizing tumor specific antigens or tumor-associated antigens.
  • the cell adoptive therapy comprises B cell expressing antibodies specific for tumor specific antigens or tumor-associated antigens.
  • cell adoptive therapy comprises NK cells that are able to kill the tumor cells.
  • cell adoptive therapy comprises dendritic cells (DC).
  • the dendritic cells are cultured in vivo with a DC expanding compound (e.g. GM-CSF or Flt3-L), loaded with tumor specific antigens or tumor-associated antigens and reintroduced in vivo.
  • a DC expanding compound e.g. GM-CSF or Flt3-L
  • Tumor specific antigens or tumor-associated antigens e.g. GM-CSF or Flt3-L
  • Intracellular signalling inhibitors e.g. GM-CSF or Flt3-L
  • the Intracellular signalling inhibitor is selected from the group consisting of tyrosine kinase inhibitors, serine/threonine kinase inhibitors, protein-tyrosine phosphatases inhibitors, dual-specificity phosphatases inhibitors, or serine/threonine phosphatases inhibitors.
  • Anti-anergic compounds are small compounds, proteins, glycoproteins or antibodies that can break tolerance to tumor and cancer antigens.
  • TILs tumor infiltrating lymphocytes
  • the anergic condition may in a substantial number of cases be counteracted by monoclonal antibodies that prevent CTLA-4 -induced anergy or tolerance.
  • Blockade of CTLA-4 has been shown in animal models to improve the effectiveness of cancer therapy suggesting that CTLA-4 blockade can be used to break the tolerance to cancer and tumor antigens.
  • a non-limiting example of a monoclonal antibody that may be used for induction of the activity of TILs is MDX-010 (Phan , et al.
  • Tumor-specific antigens are a relatively small group of antigens exemplified by the cancer-testis antigens. These genes are silent in normal tissue but are expressed by cancerous cells. They are highly specific mark- ers of disease and include MAGE (melanoma antigen gene) found in melanoma. Tumor-associated antigens. Tumor-associated antigens (TAAs) are usually differentiation antigens expressed by normal cells but massively over-expressed in cancerous tissue. Targets initially thought to be specific for a particular cancer are actually quite common in many tumors, such as the gangliosides and mucin antigens.
  • Classical differentiation antigens include MART-1 (melanoma antigen recognized by T cells) and gp 100, both from melanoma, tyrosinase and gp75. Mutational antigens. Point mutations are common in many cancers, and often occur in a similar location, such as the common mutation of the P53 oncogene. In vitro induction of human cytotoxic T-lymphocyte (CTL) responses against peptides of mutant and wild-type p53 has been reported. In a mouse model, mutant p53-pulsed dendritic cells were able to induce p53 specific CTL and inhibit the growth of established tumors. Viral antigens.
  • CTL cytotoxic T-lymphocyte
  • viruses are oncogenic and gene products encoded by these viruses can elicit immune responses and thus serve as cancer antigens.
  • An example is the E6 and E7 proteins from human papilloma virus type 16, which have been shown to induce cytotoxic T-lymphocyte responses in vitro.
  • Tumor-specific antigens, tumor-associated antigens and/or mutational antigens and ⁇ viral antigens may be used either as peptides, recombinant purified single-agent antigens, combination of recombinant purified antigens and/or purified or pools of antigens isolated from cancer cells or tumor cells as a vaccine to elicit an anti-tumor immune response.
  • peptides, recombinant purified single-agent antigens, combinations of recombinant antigens and/or purified or pools of antigens isolated from virus-infected cells may be used in a vaccine to elicit a response against virus-infected cells.
  • Therapeutic vaccines can also be in the form of a DNA vaccine to elicit immune response against cancer and virus-infected cells. Said vaccine-mediated elicitation of an anti-tumor response or a response against virus- infected cells may be enhanced by administering adjuvants, cytokines, CpG oligodeoxynucleotides, dendritic cells, GM-CSF, or heat-shock proteins.
  • combination therapy is performed by administering a TF antagonist with one or more therapeutic vaccines with or without adjuvants, cytokines, CpG oligodeoxynucleotides, dendritic cells, GM-CSF, or heat-shock proteins.
  • metalloproteinase inhibitors Metastatic cancer cells penetrate the extracellular matrix (ECM) and the basement membrane of the blood vessels to metastasise to a target organ (ectopic site). EMC consists of proteins embedded in a carbohydrate complex (heparan sulfate peptidoglycans), and proteases surrounding the tumour are active in this breaking down the host tissue.
  • Internal vaccination and “internal vaccination therapy” refer to drug- or radition- induced cell death of tumor cells that leads to elicitation of an immune response directed towards (i) said tumor cells as a whole or (ii) parts of said tumor cells including (a) secreted proteins, glycoproteins or other products, (b) membrane-associated proteins or glycoproteins or other components associated with or inserted in membranes and (c) intracellular proteins or other intracellular components.
  • the immune response may be humoral (i.e.
  • Internal vaccination bears many similarities to other vaccination procedures and involves many or all of the same cellular components of the hematopoietic and immune system with the advantage 'that the immunogens or antigenic components are endogenous and thus representative for the antigenic repertoire of said tumor cells. Internal vaccination may thus be considered personalized vaccination, which is elicited by use of general procedures for cancer treatment leading to tumor cell death.
  • cytotoxic domain or "cytotoxic compound” as used herein refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells.
  • cytotoxic domain and “cytotoxic compound” may be used interchangebly.
  • the term is intended to include radioactive isotopes or radionuclides (e.g. 1131 , 1125, Y90 and Re186), chemotherapeutic agents, cytostatica and toxins such as enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof.
  • Cytotoxic compounds also includes peptides, which directly mediate mitochondrial cytochrome C release and apoptosis, e.g., (KLAKLAK) 2 including both enantiomers (Ellerby et al. Nature medicine 5, (9) 1032- 1038, 1999).
  • chemotherapeutic compound is a chemical compound useful in the treatment of cancer.
  • chemotherapeutic compounds include adriamycin, doxorubicin, epirubi- cin, 5-fluorouracil, cytosine arabinoside ("Ara-C”), cyclophosphamide, thiotepa, busulfan, cy- toxin, taxoids, e.g.
  • paclitaxel TAXOLTM, Bristol-Myers Squibb Oncology, Princeton, N.J.
  • doxetaxel toxotere, methotrexate, cisplatin, vinblastine, bleomycin, etoposide, ifos- famide, mitomycin C, mitoxantrone, vincristine, vinorelbine, carboplatin, teniposide, dauno- mycin, carminomycin, aminopterin, dactinomycin, mitomycins, esperamicins (see U.S. Pat. No. 4,675,187), melphalan and other related nitrogen mustards.
  • hormonal compounds that act to regulate or inhibit hormone action on tumors, such as tamoxifen and onapristone.
  • aminoglutethimide aminoglutethimide, asparaginase, bleomy- cin, L-buthiamine sulphoxide, busulfan, camptothecin, carboplatin, carmustine, chlorambucil, cisplatin, cyclophosphamide, cytarabine HCI, dacarbazine, dactinomycin, daunorubicin HCI, doxorubicin HCI, edatrexate, estramustine phosphate sodium, etopside (V16-213), floxu- ridine, fluorouracil, flutamide, gallium nitrite, hydroxyurea, idarubicin, ifosfamide, interferon alpha -2a, alpha -2b, leuprolide acetate (LHRH-releasing factor analogue), lo
  • Cytotoxic compounds may include, but are not limited to, a therapeutically effective amount of: toxins; drugs; enzymes; cytokines; radionuclides; photodynamic compounds; and molecules which induce apoptosis (e.g., Fas ligand or 2-methoxyestradioI).
  • Toxins may include a therapeutically effective amount of ricin A chain, mutant Pseudomonas exotoxins, diphtheria toxoid, streptonigrin, boamycin, saporin, gelonin, and pokeweed antiviral protein.
  • Drugs may include a therapeutically effective amount of cytotoxic drugs including, but not limited to, fludarabine, chlorambucil, daunorubicin, doxorubicin (e.g., in liposomes), cisplatin, bleomycin, melphalan, mitomycin-C, and methotrexate.
  • cytotoxic drugs including, but not limited to, fludarabine, chlorambucil, daunorubicin, doxorubicin (e.g., in liposomes), cisplatin, bleomycin, melphalan, mitomycin-C, and methotrexate.
  • radionuclides may include, but are not limited to, proteins labeled with radiometals such as yttrium which emits a high energy beta particle, and I 125 that emits Auger electrons, that may be absorbed by adjacent TF presenting cells.
  • Photodynamic compounds may include therapeutically effective amounts of porphyrins and their derivatives.
  • Cytotoxic compounds suitable for use herein include conventional chemotherapeu- tics, such as vinblastine, anthracycline antitumor antibiotics including doxorubicin, 2- pyrrolino- doxorubicin, Doxorubicin hydrochloride (Adriamycin) (Schally VA and Nagy A (1999) Eur J Endocrinol 141 , 1-14, Vasey PA et al (1999) Clin Cancer Res 5, 83-94), bleo- mycin, methotrexate, 5-fluorouracil, 6-thioguanine, cytarabine, cyclophosphamide (N,N-bis- (beta-Chlorethyl)-amino-1-oxa-3-aza-2-phosphocyclohexane-2-oxide) and cisplatinum, as well as other conventional chemotherapeutics as described in Cancer: Principles and Prac- tice of Oncology, 2d
  • Trichothecenes are drugs produced by soil fungi of the class Fungi imperfecti or isolated from Baccharus megapotamica (Bamburg, J. R. Proc. Molec. Subcell. Biol. 8:41-110, 1983; Jarvis & Mazzola, Ace. Chem. Res. 15:338-395, 1982). They appear to be the most toxic molecules that contain only carbon, hydrogen and oxygen
  • trichothecenes those that have only a central ses- quiterpenoid structure and those that have an additional macrocyclic ring (simple and macro- cyclic trichothecenes, respectively).
  • the simple trichothecenes may be subdivided into three groups (i.e., Group A, B, and C) as described in U.S. Pat. Nos. 4,744,981 and 4,906,452 (incorporated herein by reference).
  • Group A simple trichothecenes include: Scirpene, Roridin C, dihydrotrichothecene, Scirpen-4, 8-diol, Verrucarol, Scirpentriol, T-2 tetraol, pentahydroxyscirpene, 4-deacetylneosolaniol, trichodermin, deacetylcalonectrin, calonectrin, diacetylverrucarol, 4-monoacetoxyscirpenol, 4,15-diacetoxyscirpenol, 7- hydroxydiacetoxyscirpenol, 8-hydroxydiacetoxy-scirpenol (Neosolaniol), 7,8- dihydroxydiacetoxyscirpenol, 7-hydroxy-8-acetyldiacetoxyscirpenol, 8-acetylneosolaniol, NT- 1 , NT-2, HT-2, T-2, and
  • Group B simple tricho- thecenes include: Trichothecolone, Trichothecin, deoxynivalenol, 3-acetyldeoxynivalenol, 5- acetyldeoxynivalenol, 3,15-diacetyldeoxynivalenol, Nivalenol, 4-acetylnivalenol (Fusarenon- X) 4,15-idacetylnivalenol, 4,7,15-triacetylnivalenol, and tetra-acetylnivalenol.
  • Representative examples of Group C simple trichothecenes include: Crotocol and Crotocin.
  • Representative macrocyclic trichothecenes include Verrucarin A, Verrucarin B, Verrucarin J (Satratoxin C), Roridin A, Roridin D, Roridin E (Satratoxin D), Roridin H, Satratoxin F, Satratoxin G, Satratoxin H, Vertisporin, Mytoxin A, Mytoxin C, Mytoxin B, Myrotoxin A, Myrotoxin B, Myrotoxin C, Myrotoxin D, Roritoxin A, Roritoxin B, and Roritoxin D.
  • trichothecene sesquiterpenoid ring structure
  • baccharins isolated from the higher plant Baccharis megapotamica, and these are described in the litera- ture, for instance as disclosed by Jarvis et al. (Chemistry of Alleopathy, ACS Symposium Series No. 268: ed. A. C. Thompson, 1984, pp. 149-159).
  • cytotoxic compound within the present invention include N,N-cis(2- chloroethyI)N-nitroso-urea (BCNU), D-myo-inositoI-1,2,6-trisphosphate, Melphalan (p-Di-(2- chloroethyl)-amino-L-phenylalanine), Procarbazine (p-(N'-Methyl-hydrazinomethyl)-N- isopropyl-benzamide), Dactinomycin (Actinomycin D), Polyestradiolphosphate, thalidomid, temozolomide, mitozolomide, mercaptopurine, N-methylformamide, 2-amino-1 ,3,4- thiadiazole, hexamethylmelamine, gallium nitrate, 3% thymidine, dichloromethotrexate, mi- toguazone, suramin, bromodeoxyuridine, iododeoxyuridine, semustine, 1-
  • the cytotoxic compound stimulates the production of free radicals NO*, O2*.
  • the cytotoxic compound stimulates apoptosis by regulation of p53, superoxiddismutase, phospholipase C, cyclooxygenase 2, caspase- associated recruitment domains (CARD).
  • the cytotoxic compound is selected from the group consisting of an cyclooxygenase 2 inhibitor, apoptin, chicken Anemia Virus (CAV), (e.g.
  • Radionuclides useful within the present invention include gamma-emitters, positron- emitters, Auger electron-emitters, X-ray emitters and fluorescence-emitters, with beta- or alpha-emitters preferred for therapeutic use.
  • Radionuclides are well-known in the art and include 123-1, 125-1, 130-1, 131-1, 133-1, 135-1 47-Sc, 72-As, 72-Se, 90-Y, 88-Y, 97-Ru, 100-Pd, 101m-Rh, 119-Sb, 128-Ba, 197-Hg, 211 -At, 212-Bi, 153-Sm, 169-Eu, 212-Pb, 109-Pd, 111- ln, 67-Ga, 68-Ga, 64-Cu, 67-Cu, 75-Br, 76-Br, 77-Br, 99m-Tc, 11-C, 13-N, 15-0, 166-Ho and 18-F.
  • Preferred therapeutic radionuclides include 188-Re, 186-Re, 203-Pb, 212-Pb, 212-Bi, 109-Pd, 64-Cu, 67-Cu, 90-Y, 125-1, 131-1, 77-Br, 211-At, 97-Ru, 105-Rh, 198-Au and 199- Ag, 166-Ho or 177-Lu.
  • the TF antagonist comprises gadophrin e.g. gadophrin-2 (previously referred as bis-gadolinium-mesoporhyrin) (Pislaru SV. et al., Circula- tion, 99 (5) pp.
  • Nucleic acid sequence or “nucleotide sequence” as used herein refers to an oligonucleotide, nucleotide, or polynucleotide, and fragments or portions thereof, and to DNA or RNA of genomic or synthetic origin which may be single- or double-stranded, and represent the sense or antisense strand.
  • amino acid sequence refers to an oligopeptide, peptide, polypeptide, or protein sequence, and fragments or portions thereof, and to naturally occurring or synthetic molecules.
  • amino acid sequence is recited herein to refer to an amino acid sequence of a naturally occurring protein molecule
  • amino acid sequence and like terms, such as “polypeptide” or “protein” are not meant to limit the amino acid sequence to the complete, native amino acid sequence associated with the recited protein molecule.
  • PNA or “Peptide nucleic acid”, as used herein, refers to a molecule which comprises an oligomer to which an amino acid residue, such as lysine, and an amino group have been added.
  • anti-gene agents stop transcript elongation by binding to their complementary strand of nucleic acid (Nielsen, P. E. et al. (1993) Anticancer Drug Des.
  • antisense refers to nucleotide sequences which are complementary to a specific DNA or RNA sequence.
  • antisense strand is used in reference to a nucleic acid strand that is complementary to the "sense” strand.
  • Antisense molecules may be produced by any method, including synthesis by ligating the gene(s) of interest in a reverse orientation to a viral promoter which permits the synthesis of a complementary strand. Once introduced into a cell, this transcribed strand combines with natural sequences produced by the cell to form duplexes. These duplexes then block either the further transcription or translation. In this manner, mutant phenotypes may be generated.
  • a further prefered antisense molecule includes Locked Nucleic Acids (LNAs) in which the 2'-hydroxyl group is linked to the 3' or 4' carbon atom of the sugar ring thereby forming a bicyclic sugar moiety.
  • the linkage is preferably a methelyne (-CH 2 -).sub.n group bridging the 2' oxygen atom and the 4' carbon atom wherein n is 1 or 2.
  • LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226.
  • FVIIa polypeptide or “FVIIa polypeptides” as used herein means native Factor Vila, as well as equivalents of Factor Vila that contain one or more amino acid sequence alterations relative to native Factor Vila (i.e., Factor VII variants), and/or contain truncated amino acid sequences relative to native Factor Vila (i.e., Factor Vila fragments). Such equivalents may exhibit different properties relative to native Factor Vila, including stability, phospholipid binding, altered specific proteolytic activity, and the like.
  • Factor VII equivalent encompasses, without limitation, equivalents of Factor Vila exhibiting TF binding activity.
  • TF binding activity means the ability of a FVIIa polypeptide or TF antagonist to inhibit the binding of recombinant human 125 l-FVIIa to cell surface human TF.
  • the TF binding activity may be measured as described in Assay 3.
  • Factor VII equivalents also includes proteolytically inactive variants of FVIIa.
  • the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the lysine corresponding to position 341 of wild type human FVIIa.
  • the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the serine corresponding to position 344 of wild type human FVIIa.
  • the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the aspartic acid corresponding to position 242 of wild type human FVIIa. In one embodiment of the invention the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the histidine corresponding to position 193 of wild type human FVIIa.
  • the FVIIa polypeptide is FVII-(K341A) In one embodiment the FVIIa polypeptide is FVII-(S344A) In one embodiment the FVIIa polypeptide is FVII-(D242A) In one embodiment the FVIIa polypeptide is FVII-(H193A)
  • the terminology for specific amino acid substitutions used herein are as follows. The first letter represent the amino acid naturally present at a position of wild type human FVIIa. The following number represent the position in wild type human FVIIa. The second letter represent the different amino acid substituting for the natural amino acid.
  • FVII- (K341A) where a lysine at position 341 of wild type human FVIIa is replaced by an alanine.
  • FVII-(K341A/S344A) the lysine at position 341 of wild type human FVIIa is replaced by an alanine and the serine in position 344 of wild type human FVIIa is replaced by an alanine in the same Factor VII polypeptide.
  • the terms "Factor VII” or “FVII” are intended to mean Factor VII polypeptides in their uncleaved (zymogen) form.
  • the terms "Factor Vila” or “FVIIa” are intended to mean native bioactive forms of FVII.
  • FVII is cleaved between residues 152 and 153 to yield FVIIa.
  • the term "Fac- tor Vila” is also intended to encompass, without limitation, polypeptides having the amino acid sequence 1-406 of wild-type human Factor Vila (as disclosed in U.S. Patent No. 4,784,950), as well as wild-type Factor Vila derived from other species, such as, e.g., bovine, porcine, canine, murine, and salmon Factor Vila. It further encompasses natural allelic variations of Factor Vila that may exist and occur from one individual to another. Also, degree and location of glycosylation or other post-translation modifications may vary depending on the chosen host cells and the nature of the host cellular environment.
  • variant or “variants”, as used herein, is intended to designate human Factor VII having the sequence of SEQ ID NO: 1 , wherein one or more amino acids of the parent protein have been substituted by another amino acid and/or wherein one or more amino acids of the parent protein have been deleted and/or wherein one or more amino acids have been inserted in protein and/or wherein one or more amino acids have been added to the parent protein. Such addition can take place either at the N-terminal end or at the C-terminal end of the parent protein or both.
  • the variant has a total amont of amino acid substitutions and/or additions and/or deletions independently selected from the group consisting of 1 , 2, 3, 4, 5, 6, 7, 8, 9, and 10.
  • the ability of any particular cytotoxic compound to mediate lysis of the tumor cell target can be assayed.
  • the tumor cells of interest are grown and labeled in vivo or in vitro; the cytotoxic compound is added to the tumor cell culture. Cytolysis of the target tumor cells is detected by the release of label from the lysed cells.
  • the cytotoxic compound that is capable of mediating cell ablation, lysis or apoptosis in the in vitro test can then be used therapeutically in that particular patient.
  • active site and the like when used herein with reference to FVIIa refer to the catalytic and zymogen substrate binding site, including the "Si" site of FVIIa as that term is defined by Schecter, I.
  • TF-mediated coagulation activity means coagulation initiated by TF through the formation of the TF/FVIIa complex and its activation of FIX and Factor X to FlXa and FXa, respectively. TF-mediated coagulation activity is measured in a FXa generation as- say.
  • FXa generation assay as used herein is intended to mean any assay where activation of FX is measured in a sample comprising TF, FVIIa, FX, calcium and phospholip- ids. An example of a FXa generation assay is described in assay 1.
  • a TF/FVIIa mediated or associated process or event, or a process or event associated with TF-mediated coagulation activity is any event, which requires the presence of TF/FVIIa.
  • Such processes or events include, but are not limited to, formation of fibrin which leads to thrombus formation; platelet deposition; proliferation of smooth muscle cells (SMCs) in the vessel wall, such as, for example, in intimal hyperplasia or restenosis, which is thought to result from a complex interaction of biological processes including platelet deposition and thrombus formation, release of chemotactic and mitogenic factors, and the migration and proliferation of vascular smooth muscle cells into the intima of an arterial segment; and deleterious events associated with post-ischemic reperfusion, such as, for example, in patients with acute myocardial infarction undergoing coronary thrombolysis.
  • Fibrinogen is converted to fibrin by the action of thrombin.
  • Thrombin is formed by the proteolytic cleavage of prothrombin.
  • This proteolysis is effected by FXa which binds to the surface of activated plate- lets and in the presence of FVa and calcium, cleaves prothrombin.
  • TF/FVIIa is required for the proteolytic activation of FX by the extrinsic pathway of coagulation.
  • a process mediated by or associated with TF/FVIIa, or an TF-mediated coagulation activity includes any step in the coagulation cascade from the formation of the TF/FVIIa complex to the formation of a fibrin platelet clot and which initially requires the presence of TF/FVIIa.
  • the TF/FVIIa complex initiates the extrinsic pathway by activation of FX to FXa, FIX to FlXa, and additional FVII to FVIIa.
  • TF/FVIIa mediated or associated process, or TF-mediated coagulation activity can be conveniently measured employing standard assays such as those described in Roy, S., (1991) J. Biol. Chem.
  • peptides, proteins and amino acids as used herein can comprise or refer to "natural", ⁇ _e_, naturally occurring amino acids as well as “non.classical” D- amino acids including, but not limited to, the D-isomers of the common amino acids, ⁇ - isobutyric acid, 4-aminobutyric acid, hydroxyproline, sarcosine, citrulline, cysteic acid, t- butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyl amino acids, N ⁇ -methyl amino acids, and amino acid analogues in general.
  • amino acids can include Abu, 2-amino butyric acid; ⁇ -Abu, 4-aminobutyric acid; ⁇ -Ahx, 6-aminohexanoic acid; Aib, 2-amino-isobutyric acid; ⁇ -Ala, 3-aminopropionic acid; Orn, ornithine; Hyp, trans-hydroxyproline; Nle, norleucine; Nva, norvaline.
  • GLA as used herein means 4-carboxyglutamic acid ( ⁇ - carboxyglutamate).
  • a FVIIa inhibitor is bound to the FVIIa polypeptide and decreases or prevents the FVIIa- catalysed conversion of FX to FXa.
  • a FVIIa inhibitor may be identified as a substance, which reduces the amidolytic activity by at least 50% at a concentration of the substance at 400 ⁇ M in the FVIIa amidolytic assay described by Persson et al. (Persson et al., J. Biol. Chem. 272: 19919-19924 (1997)).
  • FVIIa inhibitor may be selected from any one of several groups of FVIIa directed inhibitors. Such inhibitors are broadly categorised for the purpose of the present invention into i) inhibitors which reversibly bind to FVIIa and are cleavable by FVIIa, ii) inhibitors which reversibly bind to FVIIa but cannot be cleaved, and iii) inhibitors which irreversibly bind to FVIIa.
  • the FVIIa inhibitor moiety may also be an irreversible FVIIa serine protease inhibitor.
  • Such irreversible active site inhibitors generally form covalent bonds with the protease active site.
  • Such irreversible inhibitors include, but are not limited to, general serine protease inhibitors such as peptide chloromethylketones (see, Williams et al., J. Biol. Chem.
  • peptidyl cloromethanes or peptidyl cloromethanes; azapeptides; acylating agents such as various guanidinobenzoate derivatives and the 3-alkoxy-4-chloroisocoumarins; sulphonyl fluorides such as phenylmethylsulphonylfluoride (PMSF); diisopropylfluorophosphate (DFP); tosylpropylchloromethyl ketone (TPCK); tosyllysylchloromethyl ketone (TLCK); nitrophenyl- sulphonates and related compounds; heterocyclic protease inhibitors such as isocoumarines, and coumarins.
  • peptidic irreversible FVIIa inhibitors include, but are not limited to, Phe-Phe-Arg chloromethyl ketone, Phe-Phe-Arg chloromethylketone, D-Phe-Phe-
  • FVIIa inhibitors also include benzoxazinones or heterocyclic analogues thereof such as described in PCT/DK99/00138.
  • small peptides such as for example Phe-Phe-Arg, D-Phe-Phe-Arg, Phe-Phe-Arg, D-Phe-Phe-Arg, Phe-Pro- Arg, D-Phe-Pro-Arg, L- and D-Glu-G
  • the TF antagonist and anti-cancer compound as defined in the present specification may be administered simultaneously or sequentially.
  • the factors may be supplied in single- dosage form wherein the single-dosage form contains both compounds, or in the form of a kit-of-parts comprising a preparation of a TF antagonist as a first unit dosage form and a preparation of an anti-cancer compound as a second unit dosage form.
  • a preparation of a TF antagonist and a preparation of anti-cancer compound administration of the compounds in single-dosage form, or administration of a first agent followed by administration of a second agent with a time separation of no more than 15 minutes, preferably 10, more preferred 5, more preferred 2 min- utes. Either factor may be administered first.
  • sequential dosing is meant administration of a first agent followed by administration of a second agent with a time separation of more than 15 minutes.
  • Either of the two unit dosage form, or coagulation factor proteins may be administered first.
  • both products are injected through the same intravenous access.
  • amino acids are represented using abbreviations, as indicated in table 1 , approved by IUPAC-IUB Commission on Biochemical Nomenclature (CBN).
  • Amino acid and the like having isomers represented by name or the following abbreviations are in natural L-form unless otherwise indicated.
  • the left and right ends of an amino acid sequence of a peptide are, respectively, the N- and C-termini unless otherwise specified.
  • the invention also relates to TF antagonists as mentioned above.
  • the TF antagonist may be produced by recombinant DNA techniques.
  • DNA sequences encoding human FVIIa may be isolated by preparing a genomic or cDNA library and screening for DNA sequences coding for all or part of the protein by hybridization using synthetic oligonucleotide probes in accordance with standard techniques (cf. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989).
  • the DNA sequence encoding the protein is preferably of human origin, i.e. derived from a human genomic DNA or cDNA library.
  • the DNA sequences encoding the human FVIIa polypeptides may also be prepared synthetically by established standard methods, e.g. the phosphoamidite method described by Beaucage and Caruthers, Tetrahedron Letters 22 (1981 ), 1859 - 1869, or the method described by Matthes et al., EMBO Journal 3 (1984), 801 - 805.
  • phosphoamidite method oligonucleotides are synthesized, e.g. in an automatic DNA synthesizer, purified, annealed, ligated and cloned in suitable vectors.
  • the DNA sequences may also be prepared by polymerase chain reaction using specific primers, for instance as described in US 4,683,202, Saiki et al., Science 239 (1988), 487 - 491 , or Sambrook et al., supra.
  • the DNA sequences encoding the human FVIIa polypeptides are usually inserted into a recombinant vector which may be any vector, which may conveniently be subjected to recombinant DNA procedures, and the choice of vector will often depend on the host cell into which it is to be introduced.
  • the vector may be an autonomously replicating vector, i.e. a vector, which exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, e.g.
  • the vector may be one which, when introduced into a host cell, is integrated into the host cell genome and replicated together with the chromosome(s) into which it has been integrated.
  • the vector is preferably an expression vector in which the DNA sequence encoding the human FVIIa polypeptides is operably linked to additional segments required for transcription of the DNA.
  • the expression vector is derived from plasmid or viral DNA, or may contain elements of both.
  • operably linked indicates that the segments are arranged so that they function in concert for their intended purposes, e.g. transcription initiates in a promoter and proceeds through the DNA sequence coding for the polypeptide.
  • the promoter may be any DNA sequence, which shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell.
  • suitable promoters for directing the transcription of the DNA encoding the human FVIIa polypeptide in mammalian cells are the SV40 promoter (Subramani et al., Mol. Cell Biol. 1 (1981 ), 854 -864), the MT-1 (metallothionein gene) promoter (Palmiter et al.,
  • a suitable promoter for use in insect cells is the polyhedrin promoter (US 4,745,051 ; Vasuvedan et al., FEBS Lett. 311 , (1992) 7 - 11 ), the P10 promoter (J.M. Vlak et al., J. Gen. Virology 69, 1988, pp.
  • yeast host cells include promoters from yeast glycolytic genes (Hitzeman et al., J. Biol. Chem. 255 (1980), 12073 - 12080; Alber and Kawasaki, J. Mol. Appl. Gen.
  • Suitable promoters for use in filamentous fungus host cells are, for instance, the ADH3 promoter (McKnight et al., The EMBO J. 4 (1985), 2093 - 2099) or the tpiA promoter.
  • promoters examples include those derived from the gene encoding A oryzae TAKA amylase, Rhizomucor miehei aspartic proteinase, A niger neutral ⁇ -amylase, A. niger acid stable ⁇ -amylase, A. niger or A. awamo ⁇ ' glucoamylase (gluA), Rhizomucor miehei lipase, A oryzae alkaline protease, A. oryzae triose phosphate isomerase or A. nidulans acetamidase.
  • gluA glucoamylase
  • Rhizomucor miehei lipase Rhizomucor miehei lipase
  • a oryzae alkaline protease A. oryzae triose phosphate isomerase or A. nidulans acetamidase.
  • Preferred are the TAKA-amylase and gluA promote
  • the DNA sequences encoding the human FVIIa polypeptides may also, if necessary, be operably connected to a suitable terminator, such as the human growth hormone terminator (Palmiter et al., Science 222, 1983, pp. 809-814) or the TPI1 (Alber and Kawasaki, J. Mol. Appl. Ge 1 , 1982, pp. 419-434) or ADH3 (McKnight et al., The EMBO J. 4, 1985, pp. 2093-2099) terminators.
  • the vector may also contain a set of RNA splice sites located downstream from the promoter and upstream from the insertion site for the FVIIa sequence itself.
  • RNA splice sites may be obtained from adenovirus and/or immunoglobulin genes.
  • a polyadenylation signal located downstream of the insertion site.
  • Particularly preferred polyadenylation signals include the early or late polyadenylation signal from SV40 (Kaufman and Sharp, ibid.), the polyadenylation signal from the adenovirus 5 Elb region, the human growth hormone gene terminator (DeNoto et al. Nuc. Acids Res. 9:3719-3730, 1981) or the polyadenylation signal from the human FVII gene or the bovine FVII gene.
  • the expression vectors may also include a noncoding viral leader sequence, such as the adenovirus 2 tripartite leader, located between the promoter and the RNA splice sites; and enhancer sequences, such as the SV40 enhancer.
  • the recombinant vector may further comprise a DNA sequence enabling the vector to replicate in the host cell in question.
  • An example of such a sequence (when the host cell is a mammalian cell) is the SV40 origin of replication.
  • suitable sequences enabling the vector to replicate are the yeast plasmid 2 ⁇ replication genes REP 1-3 and origin of replication.
  • the vector may also comprise a selectable marker, e.g.
  • DHFR dihydrofolate reductase
  • Schizosaccharomyces pombe TP gene (described by P.R. Russell, Gene 40, 1985, pp. 125-130), or one which confers resistance to a drug, e.g. ampicillin, kanamycin, tetracyclin, chloramphenicol, neomycin, hygromycin or methotrexate.
  • selectable markers include amdS, pyrG, argB, niaD or sC.
  • a secretory signal sequence (also known as a leader sequence, prepro sequence or pre sequence) may be provided in the recombinant vector.
  • the secretory signal sequence is joined to the DNA sequences encoding the human FVIIa polypeptides in the correct reading frame.
  • Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the peptide.
  • the secretory signal sequence may be that, normally associated with the protein or may be from a gene encoding another secreted protein.
  • the secretory signal sequence may encode any signal peptide, which ensures efficient direction of the expressed human FVIIa polypeptides into the secretory pathway of the cell.
  • the signal peptide may be naturally occurring signal peptide, or a functional part thereof, or it may be a synthetic peptide. Suitable signal peptides have been found to be the ⁇ -factor signal peptide (cf. US 4,870,008), the signal peptide of mouse salivary amylase (cf. O. Hagenbuchle et al., Nature 289, 1981 , pp. 643-646), a modified carboxypeptidase signal peptide (cf. L.A. Vails et al., Cell 48, 1987, pp. 887-897), the yeast
  • BAR1 signal peptide (cf. WO 87/02670), or the yeast aspartic protease 3 (YAP3) signal peptide (cf. M. Egel-Mitani et al., Yeas.6, 1990, pp. 127-137).
  • a sequence encoding a leader peptide may also be inserted downstream of the signal sequence and upstream of the DNA sequence encoding the human FVIIa polypeptides.
  • the function of the leader peptide is to allow the expressed peptide to be directed from the endoplasmic reticulum to the Golgi apparatus and further to a secretory vesicle for secretion into the culture medium (i.e.
  • the leader peptide may be the yeast alpha-factor leader (the use of which is described in e.g. US 4,546,082, US 4,870,008, EP 16 201 , EP 123 294, EP 123 544 and EP 163 529).
  • the leader peptide may be a synthetic leader peptide, which is to say a leader peptide not found in nature. Synthetic leader peptides may, for instance, be constructed as described in WO 89/02463 or WO 92/11378.
  • the signal peptide may conveniently be derived from a gene encoding an Aspergillus sp. amylase or glucoamylase, a gene encoding a Rhizomucor miehei lipase or protease or a Humicola lanuginosa lipase.
  • the signal peptide is preferably derived from a gene encoding A. oryzae TAKA amylase, A. niger neutral ⁇ -amylase, A. niger acid-stable amylase, or A. niger glucoamylase.
  • Suitable signal peptides are disclosed in, e.g. EP 238023 and EP 215594.
  • the signal peptide may conveniently be derived from an insect gene (cf. WO 90/05783), such as the lepidopteran Manduca sexta adipokinetic hormone precursor signal peptide (cf. US 5,023,328).
  • insect gene cf. WO 90/05783
  • the procedures used to ligate the DNA sequences coding for the human FVIIa polypeptides, the promoter and optionally the terminator and/or secretory signal sequence, respectively, and to insert them into suitable vectors containing the information necessary for replication, are well known to persons skilled in the art (cf., for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, New York, 1989).
  • Selectable markers may be introduced into the cell on a separate plasmid at the same time as the gene of interest, or they may be introduced on the same plasmid. If on the same plasmid, the selectable marker and the gene of interest may be under the control of different promoters or the same promoter, the latter arrangement producing a dicistronic message. Constructs of this type are known in the art (for example, Levinson and Simonsen, U.S. Pat. No. 4,713,339). It may also be advantageous to add additional DNA, known as "carrier DNA,” to the mixture that is introduced into the cells. After the cells have taken up the DNA, they are grown in an appropriate growth medium, typically 1-2 days, to begin expressing the gene of interest.
  • carrier DNA additional DNA
  • appropriate growth medium means a medium containing nutrients and other components required for the growth of cells and the expression of the human FVIIa polypeptides of inter- est.
  • Media generally include a carbon source, a nitrogen source, essential amino acids, essential sugars, vitamins, salts, phospholipids, protein and growth factors.
  • the medium will contain vitamin K, preferably at a concentration of about 0.1 ⁇ g/ml to about 5 ⁇ g/ml.
  • Drug selection is then applied to select for the growth of cells that are expressing the selectable marker in a stable fashion.
  • the drug concentration may be increased to select for an increased copy number of the cloned sequences, thereby increasing expression levels.
  • Clones of stably transfected cells are then screened for expression of the human FVIIa polypeptide of interest.
  • the host cell into which the DNA sequences encoding the human FVIIa polypeptides is introduced may be any cell, which is capable of producing the posttranslational modified human FVIIa polypeptides and includes yeast, fungi and higher eucaryotic cells.
  • Examples of mammalian cell lines for use in the present invention are the COS-1 (ATCC CRL 1650), baby hamster kidney (BHK) and 293 (ATCC CRL 1573; Graham et al., J. Gen. Virol. 36:59-72, 1977) cell lines.
  • a preferred BHK cell line is the tk " ts13 BHK cell line (Waechter and Baserga, Proc. Natl. Acad. Sci. USA 79: 1106-1110, 1982, incorporated • herein by reference), hereinafter referred to as BHK 570 cells.
  • the BHK 570 cell line has been deposited with the American Type Culture Collection, 12301 Parklawn Dr., Rockville, Md.
  • a tk " ts13 BHK cell line is also available from the ATCC under accession number CRL 1632.
  • a number of other cell lines may be used within the present invention, including Rat Hep I (Rat hepatoma; ATCC CRL 1600), Rat Hep II (Rat hepatoma; ATCC CRL 1548), TCMK (ATCC CCL 139), Human lung (ATCC HB 8065), NCTC 1469 (ATCC CCL 9.1), CHO (ATCC CCL 61) and DUKX cells (Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 77:4216-4220, 1980).
  • yeasts cells include cells of Saccharomyces spp. or Schizosac- charomyces spp., in particular strains of Saccharomyces cerevisiae or Saccharomyces kluyveri.
  • Methods for transforming yeast cells with heterologous DNA and producing heterologous polypeptides there from are described, e.g. in US 4,599,311 , US 4,931 ,373, US 4,870,008, 5,037,743, and US 4,845,075, all of which are hereby incorporated by reference.
  • Transformed cells are selected by a phenotype determined by a selectable marker, commonly drug resis- tance or the ability to grow in the absence of a particular nutrient, e.g.
  • a preferred vector for use in yeast is the POT1 vector disclosed in US 4,931 ,373.
  • the DNA sequences encoding the human FVIIa polypeptides may be preceded by a signal sequence and optionally a leader sequence, e.g. as described above.
  • suitable yeast cells are strains of Kluyveromyces, such as K. lactis, Hansenula, e.g. H. polymorpha, or Pichia, e.g. P. pastoris (cf. Gleeson et al., J. Gen. Microbiol. 132, 1986, pp. 3459-3465; US 4,882,279).
  • Examples of other fungal cells are cells of filamentous fungi, e.g.
  • Aspergillus spp. Neurospora spp., Fusarium spp. or Trichoderma spp., in particular strains of A. oryzae, A. nidulans or A. niger.
  • the use of Aspergillus spp. for the expression of proteins is described in, e.g., EP 272 277, EP 238 023, EP 184438
  • the transformation of F. oxysporum may, for instance, be carried out as described by Malardier et al., 1989, Gene 78: 147-156.
  • the transformation of Trichoderma spp. may be performed for instance as described in EP 244 234.
  • a filamentous fungus When a filamentous fungus is used as the host cell, it may be transformed with the DNA construct of the invention, conveniently by integrating the DNA construct in the host chromosome to obtain a recombinant host cell.
  • This integration is generally considered to be an advantage as the DNA sequence is more likely to be stably maintained in the cell.
  • Integration of the DNA constructs into the host chromosome may be performed according to conventional methods, e.g. by homologous or heterologous recombination. Transformation of insect cells and production of heterologous polypeptides therein may be performed as described in US 4,745,051 ; US 4,879,236; US 5,155,037; 5,162,222; EP
  • the insect cell line used as the host may suitably be a Lepidoptera cell line, such as Spodoptera frugiperda cells or Trichoplusia ni cells (cf. US 5,077,214). Culture conditions may suitably be as described in, for instance, WO 89/01029 or WO 89/01028, or any of the aforementioned references.
  • the transformed or transfected host cell described above is then cultured in a suitable nutrient medium under conditions permitting expression of the human FVIIa polypeptide after which all or part of the resulting peptide may be recovered from the culture.
  • the medium used to culture the cells may be any conventional medium suitable for growing the host cells, such as minimal or complex media containing appropriate supplements. Suitable media are available from commercial suppliers or may be prepared according to published recipes (e.g. in catalogues of the American Type Culture Collection).
  • the human FVIIa polypeptide produced by the cells may then be recovered from the culture medium by conventional procedures including separating the host cells from the medium by centrifugation or filtration, precipitating the proteinaqueous components of the supernatant or filtrate by means of a salt, e.g. ammonium sulphate, purification by a variety of chromatographic procedures, e.g. ion exchange chromatography, gelfiltration chromatography, affinity chromatography, or the like, dependent on the type of polypeptide in question.
  • a salt e.g. ammonium sulphate
  • a cloned wild-type FVIIa DNA sequence is used for the preparation of recombinant human FVIIa polypeptides.
  • This sequence may be modified to encode a desired FVIIa variant.
  • the complete nucleotide and amino acid sequences for human FVIIa are known. See U.S. Pat. No. 4,784,950, which is incorporated herein by reference, where the cloning and expression of recombinant human FVIIa is described.
  • the bovine FVIIa sequence is described in Takeya et al., J. Biol. Chem, 263:14868-14872 (1988), which is incorporated by reference herein.
  • the amino acid sequence alterations may be accomplished by a variety of tech- niques.
  • Modification of the DNA sequence may be by site-specific mutagenesis. Techniques for site-specific mutagenesis are well known in the art and are described by, for example, Zoller and Smith (DNA 3:479-488, 1984). Thus, using the nucleotide and amino acid sequences of FVII, one may introduce the alterations of choice.
  • DNA sequences for use within the present invention will typically encode a pre-pro peptide at the amino-terminus of the FVIIa protein to obtain proper post-translational processing (e.g. gamma-carboxylation of glutamic acid residues) and secretion from the host cell.
  • the pre-pro peptide may be that of FVIIa or another vitamin K-dependent plasma protein, such as factor IX, factor X, prothrombin, protein C or protein S.
  • additional modifications can be made in the amino acid sequence of FVIIa where those modifications do not significantly impair the ability of the protein to act as a coagulation factor.
  • FVIIa in the catalytic triad can also be modified in the activation cleavage site to inhibit the conversion of zymogen FVII into its activated two-chain form, as generally described in U.S. Pat. No. 5,288,629, incorporated herein by reference.
  • transgenic animal technology may be employed to produce the human FVIIa polypeptide.
  • the proteins within the ⁇ mammary glands of a host female mammal are preferred. Expression in the mammary gland and subsequent secretion of the protein of interest into the milk overcomes many difficulties encountered in isolating proteins from other sources. Milk is readily collected, available in large quantities, and well characterized biochemically. Furthermore, the major milk 1 proteins are present in milk at high concentrations (typically from about 1 to 15 g/l). From a commercial point of view, it is clearly preferable to use as the host a species that has a large milk yield. While smaller animals such as mice and rats can be used (and are preferred at the proof of principle stage), within the present invention it is preferred to use livestock mammals includ- ing, but not limited to, pigs, goats, sheep and cattle.
  • Sheep are particularly preferred due to such factors as the previous history of transgenesis in this species, milk yield, cost and the ready availability of equipment for collecting sheep milk. See WIPO Publication WO 88/00239 for a comparison of factors influencing the choice of host species. It is generally desirable to select a breed of host animal that has been bred for dairy use, such as East Friesland sheep, or to introduce dairy stock by breeding of the transgenic line at a later date. In any event, animals of known, good health status should be used. To obtain expression in the mammary gland, a transcription promoter from a milk protein gene is used. Milk protein genes include those genes encoding caseins (see U.S. Pat. No.
  • beta-lactoglobulin a region of at least the proximal 406 bp of 5' flanking sequence of the gene will generally be used, although larger portions of the 5' flanking sequence, up to about 5 kbp, are preferred, such as about 4.25 kbp DNA segment encompassing the 5' flanking promoter and non-coding portion of the beta-lactoglobulin gene. See Whitelaw et al., Biochem J. 286: 31-39 (1992).
  • beta-lactoglobulin gene may also be incorporated in constructs, as may genomic regions of the gene to be expressed. It is generally accepted in the art that constructs lacking introns, for example, express poorly in comparison with those that contain such DNA sequences (see Brinster et al., Proc. Natl. Acad. Sci. USA 85: 836-840 (1988); Palmiter et al., Proc. Natl. Acad. Sci. USA 88: 478-482 (1991); Whitelaw et al., Transgenic Res. 1: 3-13 (1991); WO 89/01343; and WO 91/02318, each of which is incorporated herein by reference).
  • genomic sequences containing all or some of the native introns of a gene encoding the protein or poly- peptide of interest thus the further inclusion of at least some introns from, e.g, the beta- lactoglobulin gene, is preferred.
  • One such region is a DNA segment which provides for intron splicing and RNA polyadenylation from the 3' non-coding region of the ovine beta- ⁇ lactoglobulin gene. When substituted for the natural 3' non-coding sequences of a gene, this ovine beta-lactoglobulin segment can both enhance and stabilize expression levels of the protein or polypeptide of interest.
  • the region surrounding the ini- ' tiation ATG of the sequence encoding the human FVIIa polypeptide is replaced with corresponding sequences from a milk specific protein gene.
  • Such replacement provides a putative tissue-specific initiation environment to enhance expression. It is convenient to replace the entire pre-pro sequence of the human FVIIa polypeptide and 5' non-coding sequences with those of, for example, the BLG gene, although smaller regions may be replaced.
  • a DNA segment encoding the human FVIIa polypeptide is operably linked to additional DNA segments required for its expression to produce expression units.
  • the expression units will further include a DNA segment encoding a secretory signal sequence operably linked to the segment encoding the human FVIIa polypeptide.
  • the secretory signal sequence may be a native secretory signal sequence of the human FVIIa polypeptide or may be that of another protein, such as a milk protein. See, for example, von Heinje, Nuc. Acids Res. 14: 4683-4690 (1986); and Meade et al., U.S. Pat. No. 4,873,316, which are incorporated herein by reference.
  • Construction of expression units for use in transgenic animals is conveniently carried out by inserting a sequence encoding the human FVIIa polypeptide into a plasmid or phage vector containing the additional DNA segments, although the expression unit may be constructed by essentially any sequence of ligations. It is particularly convenient to provide a vector containing a DNA segment encoding a milk protein and to replace the coding sequence for the milk protein with that of the human FVIIa polypeptide, thereby creating a gene fusion that includes the expression control sequences of the milk protein gene. In any event, cloning of the expression units in plasmids or other vectors facilitates the amplification of the human FVIIa polypeptide. Amplification is conveniently carried out in bacterial (e.g. E.
  • the vectors will typically include an origin of replication and a selectable marker functional in bacterial host cells.
  • the expression unit is then introduced into fertilized eggs (including early-stage embryos) of the chosen host species.
  • Introduction of heterologous DNA can be accomplished by one of several routes, including microinjection (e.g. U.S. Pat. No. 4,873,191 ), refroviral infection (Jaenisch, Science 240: 1468-1474 (1988)) or site-directed integration using embryonic stem (ES) cells (reviewed by Bradley et al., Bio/Technology ⁇ 0: 534-539 (1992)).
  • the eggs are then implanted into the oviducts or uteri of pseudopregnant females and allowed to develop.
  • FVIIa produced according to the present invention may be purified by affinity chromatography on an anti-FVII antibody column. It is preferred that the immunoadsorption col- umn comprise a high-specificity monoclonal antibody.
  • FVII may be autoactivated by passing it through an ion-exchange chromatography column, such as mono Q.RTM. (Pharmacia Fire Chemicals) or the like (Bjo- ern et al., 1986, Research Disclosures 269:564-565).
  • the FVIIa molecules of the present invention and pharmaceutical compositions thereof are particularly useful for administration to humans to treat a variety of conditions involving intravascular coagulation.
  • the compounds of the present invention may have one or more asymmetric centres and it is intended that stereoisomers (optical isomers), as separated, pure or partially purified stereoisomers or racemic mixtures thereof are included in the scope of the invention.
  • the TF antagonist may be prepared in the form of pharmaceutically acceptable salts, especially acid-addition salts, including salts of organic acids and mineral acids.
  • salts include salts of organic acids such as formic acid, fumaric acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, succinic acid, malic acid, tartaric acid, citric acid, benzoic acid, salicylic acid and the like.
  • Suitable inorganic acid-addition salts include salts of hydrochloric, hydrobromic, sulphuric and phosphoric acids and the like.
  • compositions include the pharmaceutically acceptable salts listed in Journal of Pharmaceutical Science. 66, 2 (1977) which are known to the skilled artisan.
  • pharmaceutically acceptable acid addition salts are the hydrates which the present compounds are able to form.
  • the acid addition salts may be obtained as the direct products of compound synthe- sis.
  • the free base may be dissolved in a suitable solvent containing the appropriate acid, and the salt isolated by evaporating the solvent or otherwise separating the salt and solvent.
  • the compounds of this invention may form solvates with standard low molecularweight solvents using methods known to the skilled artisan.
  • the TF antagonist may be administered in pharmaceutically acceptable acid addition salt form or, where appropriate, as a alkali metal or alkaline earth metal or lower alkylammonium salt. Such salt forms are believed to exhibit approximately the same order of activity as the free base forms.
  • the present invention includes within its scope pharmaceutical compositions comprising a TF antagonist and an anti-cancer compound, as the active ingredients, or pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier or diluent.
  • the pharmaceutical composition of the invention may further comprise one or more other compounds exhibiting anticoagulant activity, e.g., platelet aggregation inhibitor.
  • the compounds of the invention may be formulated into pharmaceutical composition comprising the compounds and a pharmaceutically acceptable carrier or diluent.
  • Such carriers include water, physiological saline, ethanol, polyols, e.g., glycerol or propylene glycol, or vegetable oils.
  • compositions may be prepared by conventional techniques and appear in conventional forms, for example, capsules, tablets, solutions or suspensions.
  • the pharmaceutical carrier employed may be a conventional solid or liquid carrier. Examples of solid carriers are lactose, terra alba, sucrose, talc, gelatine, agar, pectin, acacia, magnesium stearate and stearic acid.
  • liquid carriers examples include syrup, peanut oil, olive oil and water.
  • the carrier or diluent may include any time delay material known to the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax.
  • the formulations may also include wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavouring agents.
  • the formulations of the invention may be formulated so as to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art.
  • the pharmaceutical compositions can be sterilised and mixed, if desired, with auxiliary agents, emulsifiers, salt for influencing osmotic pressure, buffers and/or colouring substances and the like, which do not deleteriously react with the active compounds.
  • the route of administration may be any route, which effectively transports the active compound to the appropriate or desired site of action, such as oral or parenteral, e.g., rectal, transdermal, subcutaneous, intranasal, intramuscular, topical, intravenous, intraurethral, ophthalmic solution or an ointment, the oral route being preferred.
  • the preparation can be tabletted, placed in a hard gelatine capsule in powder or pellet form or it can be in the form of a troche or lozenge.
  • the amount of solid carrier may vary widely but will usually be from about 25 mg to about 1 g.
  • the preparation may be in the form of a syrup, emulsion, soft gelatine capsule or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution.
  • the preparation may contain a compound of formula (I) dissolved or suspended in a liquid carrier, in particular an aqueous carrier, for aerosol application.
  • the carrier may contain additives such as solubilizing agents, e.g.
  • propylene glycol propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabenes.
  • injectable solutions or suspensions preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil.
  • Tablets, dragees, or capsules having talc and/or a carbohydrate carrier or binder or the like are particularly suitable for oral application.
  • Preferable carriers for tablets, dragees, or capsules include lactose, corn starch, and/or potato starch.
  • a syrup or elixir can be used in cases where a sweetened vehicle can be employed.
  • a typical tablet which may be prepared by conventional tabletting techniques, contains Core: Active compound (as free compound 10 mg or salt thereof) Colloidal silicon dioxide (Areosil ® ) 1.5 mg '- Cellulose, microcryst. (Avicel ® ) 70 mg Modified cellulose gum (Ac-Di-Sol ® ) 7.5 mg Magnesium stearate
  • the compounds of the invention may be administered to a mammal, especially a human in need of such treatment, prevention, elimination, alleviation or amelioration of various thrombolytic or coagulophatic diseases or disorders as mentioned above.
  • mammals also include animals, both domestic animals, e.g. household pets, and non- domestic animals such as wildlife.
  • the essential ingredients (a) a TF antagonist and (b) anti-cancer compound are present in the formulation in such proportion that a dose of the formulation pro-vides an amount of each ingredient that together is a pharmaceutically effective amount to the patient being treated.
  • the dose of composition of the invention to be administered is determined depending upon age, body weight, symptom, the desired therapeutic effect, the route of administration, and the duration of the treatment etc.
  • the weight ratio of TF antagonist and the amount of anti-cancer compound may vary from a ratio of between about 1 :100 to about 100:1 (w/w).
  • the ratio of TF antagonist to anti-cancer compound may thus be, e.g., about 1 :100, or 1 :90, or 1 :80, or 1:70 or 1:60, or 1:50, or 1 :40, or 1:30, or 1:20, or 1:10, or 1:5, or 1:2, or 1:1 , or 2:1, or 5:1 , or 10:1 , or 20:1, or 30.1 , or 40:1 , or 50:1 , or 60:1, or 70:1, or 80:1 , or 90:1, or 100:1; or between about 1 :90 to about 1 :1 , or between about 1 :80 to about 1 :2, or between about 1 :70 to about 1 :5, or between about 1 :60 to about 1 :10, or between about 1 :50 to about 1 :25, or
  • the ratio by mass of TF antagonist and anti- cancer compound is between about 100: 1 and about 1 : 100 (w/w). In one embodiment of the invention the ratio by mass of TF antagonist and anti-cancer compound is between about 1 : 90 to about 1 : 1(w/w).
  • the dose of the TF antagonist suitable for oral, nasal, pulmonal or transdermal administration ranges from about 0.05 mg to about 500 mg/day, e.g., from about 1 mg to about 200 mg/day, or, e.g., from about 5 mg to about 175 mg/day for a 70-kg subject as loading and maintenance doses, depending on the weight of the subject, the condition and the severity of the condition.
  • the compounds may be administered concurrently, simultaneously, or together with a pharmaceutically acceptable carrier or diluent, whether by oral, rectal, or parenteral (including subcutaneous) route.
  • a pharmaceutically acceptable carrier or diluent whether by oral, rectal, or parenteral (including subcutaneous) route.
  • the compounds are often, and preferably, in the form of an alkali metal or earth alkali metal salt thereof. Suitable dosage ranges varies as indicated above depending upon the exact mode of administration, form in which administered, the indication towards which the administration is directed, the subject involved and the body weight of the subject involved, and the preference and experience of the physician or veterinarian in charge.
  • FVIIa generation assay (assay 1): In the following example all concentrations are final. Lipidated TF (10 pM), FVIIa
  • TF antagonist or FFR-rFVIIa (0 - 50 nM) in HBS/BSA (50 mM hepes, pH 7.4, 150 mM NaCI, 5 mM CaCI 2 ,1 mg/ml BSA) are incubated 60 min at room temperature before FX (50 nM) is added. The reaction is stopped after another 10 min by addition of 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 absorbance at 405 nm continuously for 10 min.
  • IC 50 values for TF antagonist 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.
  • CRL-2310) constitutively expressing TF are employed as TF source in FVIIa/TF catalyzed activation of FX.
  • Confluent cell monolayers in a 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 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 with 1 mg/ml BSA and 5 mM Ca 2+
  • FX (135 nM) and varying con- centrations of TF antagonist or FFR-rFVIIa in buffer B are simultaneously added to the cells.
  • 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).
  • the amount of FXa generated is determined 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 IC50 value for FFR-rFVIIa is 1.5 nM in this assay.
  • Binding studies are employed using the human bladder carcinoma cell line J82 (ATTC No. HTB-1 ) or the human keratinocyte cell line (CCD1102KerTr ATCC No CRL-2310) or NHEK P166 (Clonetics No. CC-2507) all constitutively expressing TF.
  • Confluent monolayers in 24-well tissue culture plates are washed once with buffer A (10 mM Hepes, pH 7.45, 150 mM NaCI, 4 mM KCI, and 11 mM glucose) supplemented with 5 mM EDTA and then once with buffer A and once with buffer B (buffer A supplemented with with 1 mg/ml BSA and 5 mM Ca 2+ ).
  • the monolayers are prein- cubated 2 min with 100 ⁇ l cold buffer B.
  • Varying concentrations of Mabs (or FFR-FVIIa) and radiolabelled FVIIa are simultaneously added to the cells (final volume 200 ⁇ l). The plates are incubated for 2 hours at 4 °C.
  • Kd's are calculated from the sensorgrams using the integrated Biacore evaluation software.
  • Inhibition of FVIIa/TF-induced p44/42 MAPK activation by TF antagonists with effector domain (Assay 5): The amount of phosphorylated p44/42 MAPK and/or Akt, and/or p90RSK is deter- mined 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.
  • the cells must be 70-80% confluent.
  • the experiment is performed by prein- cubating the cells with excess TF antagonist 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-aminoethyl)
  • 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.
  • Figure 1 shows the full amino acid sequence of native (wild type) human coagulation Factor VII (SEQ ID NO:1).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Diabetes (AREA)
  • Pain & Pain Management (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Endocrinology (AREA)
  • Mycology (AREA)
  • Urology & Nephrology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Vascular Medicine (AREA)
  • Otolaryngology (AREA)
  • Transplantation (AREA)
  • Communicable Diseases (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a novel pharmaceutical compositions comprising a combination of a compound, which binds to and inhibits the activity of tissue factor and a chemotherapeutic compound. The invention also relates to their use in the prophylaxis or treatment of diseases or disorders related to patho-physiological tissue factor (TF) functions.

Description

COMBINATION TREATMENT
FIELD OF THE INVENTION This invention relates to novel compositions comprising a combination of a compounds which binds to and inhibits the activity of tissue factor and another compound, which is an anti-cancer compound. The invention also relates to the novel pharmaceutical compositions as well as their use in the prophylaxis or treatment of diseases or disorders related to pathophysiological tissue factor (TF) functions including cancer, inflammation, atherosclerosis and ischemia/reperfusion.
BACKGROUND OF THE INVENTION Tissue Factor (TF) is a cellular transmembrane receptor for plasma coagulation factor Vila (FVIIa) and formation of TF/FVIIa complexes on the cell surface triggers the coagulation cascade in vivo. The TF/FVIIa complex efficiently activates coagulation factors IX and X. The resultant protease factor Xa (FXa), activates prothrombin to thrombin, which in turn converts fibrinogen into a fibrin matrix. Normally, TF is constitutively expressed on the surface of many extravascuiar cell types that are not in contact with the blood, such as fibroblasts, pericytes, smooth muscle cells and epithelial cells, but not on the surface of cells that come in contact with blood, such as en- dothelial cells and monocytes. However, TF is also expressed in various pathophysiological conditions where it is believed to be involved in progression of disease states within cancer, inflammation, atherosclerosis and ischemia/reperfusion. Thus, TF is now recognised as a target for therapeutic intervention in conditions associated with increased expression. FVIIa is a two-chain, 50 kilodalton (kDa) vitamin-K dependent, plasma serine protease which participates in the complex regulation of in vivo haemostasis. FVIIa is generated from proteolysis of a single peptide bond from its single chain zymogen, Factor VII (FVII), which is present at approximately 0.5 μg/ml in plasma. The zymogen is catalytically inactive. The conversion of zymogen FVII into the activated two-chain molecule occurs by cleavage of an internal peptide bond. In the presence of calcium ions, FVIIa binds with high affinity to exposed TF, which acts as a cofactor for FVIIa, enhancing the proteolytic activation of its substrates FVII, Factor IX and FX. In addition to its established role as an initiator of the coagulation process, TF was recently shown to function as a mediator of intracellular activities either by interactions of the cytoplasmic domain of TF with the cytoskeleton or by supporting the FVI la-protease dependent signaling. Such activities may be responsible, at least partly, for the implicated role of TF in tumor development, metastasis and angiogenesis. Cellular exposure of TF activity is advantageous in a crisis of vascular damage but may be fatal when exposure is sustained as it is in these various diseased states. Thus, it is critical to regulate the expression of TF function in maintaining the health. Inactivated FVII (FVIIai) is FVIIa modified in such a way that it is catalytically inactive. Thus, FVIIai is not able to catalyze the conversion of FX to FXa, but still able to bind tightly to TF in competition with active endogenous FVIIa and thereby inhibit the TF function. International patent applications WO 92/15686, WO 94/27631 , WO 96/12800, WO
97/47651 relates to FVIIai and the uses thereof. International patent applications WO 90/03390, WO 95/00541 , WO 96/18653, and European Patent EP 500800 describe peptides derived from FVIIa having TF/FVIIa antagonist activity. International patent application WO 01/21661 relates to bivalent inhibitor of FVII and FXa. Hu Z and Garen A (2001) Proc. Natl. Acad. Sci. USA 98; 12180-12185, Hu Z and
Garen A (2000) Proc. Natl. Acad. Sci. USA 97; 9221-9225, Hu Z and Garen A (1999) Proc. Natl. Acad. Sci. USA 96; 8161-8166, and International patent application WO 0102439 relates to immunoconjugates which comprises the Fc region of a human lgG1 immunoglobulin and a mutant FVII polypeptide, that binds to TF but do not initiate blood clotting. Furthermore, International patent application WO 98/03632 describes bivalent agonists having affinity for one or more G-coupled receptors, and Burgess, L.E. et al., Proc. Natl. Acad. Sci. USA 96, 8348-8352 (July 1999) describes "Potent selective non-peptidic inhibitors of human lung tryptase".
SUMMARY OF THE INVENTION There is still a need in the art for improved combination treatments, which efficiently inhibit pathophysiological TF function at relatively low doses and which does not produce undesirable side effects. It is therefore the object of the present invention to provide pharmaceutical combinations that act specifically on pathophysiological TF function and at the same time elicits an anti-cancer response in a patient. The present invention relates in a broad aspect to combination treatment with TF antagonists. To achieve the aforementioned object, the present invention provides a pharmaceutical composition which comprises of a TF antagonist and an anti-cancer compound. The present invention also relates to a use of the pharmaceutical composition in the treatment of diseases and disorders related to TF antagonist. In a first aspect, the present invention relates to a pharmaceutical composition useful for preventing or treating a disease or disorder associated with pathophysiological TF function, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from the first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , analogues or derivatives thereof. In a second aspect, the present invention relates to the use of a first agent, which is a TF antagonist in combination with a second agent, which is an anti-cancer compound for the manufacture of a medicament for treating a disease or disorder associated with pathophysiological TF function, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , analogues or derivatives thereof. In a third aspect, the present invention relates to a method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising administering to a mammal in need of such a treatment a therapeutically effective amount of a pharmaceutical composition, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , analogues or derivatives thereof. In a further aspect, the present invention relates to method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising (i) administering to a mammal in need of such a treatment a therapeutically effective amount of a first agent, which is a TF antagonist, and (ii) administering to a mammal in need of such a treatment a therapeutically effective amount of a second agent different from the first agent, which is an anti-cancer compound, and optionally iii) administering to a mammal in need of such a treatment a therapeutically effective amount of one or more further agents different for said first and second agent, which is an anti-cancer compound, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , ana- logues or derivatives thereof. In a further aspect, the present invention relates to a pharmaceutical kit comprising a first agent, which is a TF antagonist, and a second agent, which is an anti-cancer compound, and optionally a pharmaceutically acceptable carrier or excipient. In a futher aspect, the present invention relates to a pharmaceutical composition useful for preventing or treating a disease or disorder associated with pathophysiological TF function, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from the first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient. In a further aspect, the present invention relates to the use of a first agent, which is a TF antagonist in combination with a second agent, which is an anti-cancer compound for the manufacture of a medicament for treating a disease or disorder associated with pathophysiological TF function. In a further aspect, the present invention relates to a method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising administering to a mammal in need of such a treatment a therapeutically effective amount of a pharmaceutical composition, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient. In a further aspect, the present invention relates to method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising (i) administering to a mammal in need of such a treatment a therapeutically effective amount of a first agent, which is a TF antagonist, and (ii) administering to a mammal in need of such a treatment a therapeutically effective amount of a second agent different from the first agent, which is an anti-cancer compound, and optionally iii) administering to a mammal in need of such a treatment a therapeutically effective amount of one or more further agents different for said first and second agent, which is an anti-cancer compound.
DETAILED DESCRIPTION OF THE INVENTION The present invention relates to TF antagonists in combination with another anti- cancer drug. The TF antagonists bind TF with high affinity and specificity but do not initiate blood coagulation. In one embodiment of the present invention the TF antagonist is factor FVIIa polypeptides chemically inactivated in the active site. In another embodiment of the present invention the TF antagonist is an antibody against TF. In one embodiment of the present invention the TF antagonist is a fully human antibody against TF. In one embodiment the human antibody immunoreacts with an epitope present on human 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 human antibody binds to an epitope within the interface between TF and FVIIa. In one embodiment the antibody is a monoclonal antibody. In one embodiment the antibody is a human monoclonal antibody. In one embodiment the antibody is an antibody against human TF. The terms "TF antagonist" or "TF antagonists", as used herein is intended to mean any compound which binds directly to TF and inhibits TF-mediated FVIIa activity. The term "TF-mediated FVIIa activity", as used herein means any TF-dependent activity. The term is intended to include both a TF-mediated coagulation activity and a signaling activity mediated by TF, e.g. TF-mediated MAPK signaling. In one embodiment the TF- mediated FVIIa activity is MAPK signaling. The term " TF-mediated MAPK signaling" is intended to mean a cascade of intracellular events that mediate activation of Mitogen-Activated-Protein-Kinase (MAPK) or homo- logues thereof in response to the binding of a FVII polypeptide to TF. 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. MAPK signaling may be measured as described in assay 5. 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.
TF antagonists include but are not limited to factor FVIIa polypeptides chemically inactivated in the active site, antibodies against TF, e.g. a monoclonal antibody, such as a human monoclonal antibody against human TF. Methods of preparing human antibodies against human TF is described in International patent application 03/029295 the content of which is hereby incorporated be reference in its entirety. International patent applications 03/076461 , DK03/00481 , and DK03/00480 describe the preparation of different TF antagonist that may be used according to the invention the content of which is hereby incorporated by reference in its entirety. 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, is intended to refer to immunoglobulin molecules and fragments thereof that have the ability to specifically bind to an antigen (e.g., hu- man TF). Full-length antibodies comprise 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 hypervariabil- ity, 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 or- der: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, 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 (Ward et al., (1989) Nature 341 :544-546 ), which consists of a VH domain; and (vi) an isolated complementarity de- termining 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. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using 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 com- plementary domains of another chain and creating two antigen binding sites (see e.g., Hol- liger, 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, or the like. The terms "human antibody", "human antibodies", "human TF antibody", and "hu- man 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 mutagene- sis 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 or human/mouse chimera antibodies. An "isolated human antibody", as used herein, is intended to refer to a human anti- body 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 term " specifically bind". The term "inhibits", as used herein, means any reduction compared to a refer- ence. As an example, an antibody, which inhibits the binding of human coagulation factor Vila to human TF means any antibody, which 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 Mabs 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 one embodiment the TF antagonist is a human antibody that inhibits FVIIa- induced activation of the MAPK signalling with 98 %. In one embodiment the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 90 %. In one embodiment the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 70 %. In one embodiment the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 50 %. In one embodiment the human antibody inhibits FVIIa-induced activation of the MAPK signalling with 30 %. The term "FVIIa-induced activation of the MAPK signaling" is intended to indicate that FVIIa binds to TF in a mammalian cell and thereby induce MAPK signaling. The term "MAPK signalling" is intended to mean 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.
The term "TF-mediated coagulation activity" means coagulation initiated by TF through the formation of the TF/FVIIa complex and its activation of FIX and Factor X to FlXa and FXa, respectively. TF-mediated coagulation activity is measured in a FXa generation assay. The term "FXa generation assay" as used herein is intended to mean any assay where activation of FX is measured in a sample comprising TF, FVIIa, FX, calcium and phospholip- ids. An example of a FXa generation assay is described in assay 1. Examples of TF antagonist includes, but at not limited to FVIIai and inhibitory antibodies against TF. The inactivation of the FVIIa proteolytic activity in the FVIIai molecule is obtained in vitro by covalent active site inhibitors e.g. chloromethyl ketones. The FVIIai molecule has increased affinity for TF as compared to the binding of native FVII. The terms "TF presenting cell" or "TF presenting cells" as used herein refers to the presence of TF protein on a cell surface plasma membrane. The cell membrane, where TF is located may be the cell, where TF was synthezised by protein synthesis or it may be a cell, that contain shedded TF protein synthezised by another cell. The term "disease or disorder associated with pathophysiological TF function" as used herein means any disease or disorder, where TF is involved. This includes, but is not limited to diseases or disorders related to TF-mediated coagulation activity, thrombotic or co- agulopathic related diseases or disorders, respiratory diseases or disorders, and Inflammatory diseases or disorders.or diseases or disorders. In one embodiment thereof, the Thrombotic and Coagulopathic related diseases or disorders, Respiratory diseases or disorders, and Inflammatory diseases or disorders include deep venous thrombosis, chronic thromboembolic diseases or disorders associated with fibrin formation, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, cancer, tumour metastasis, pathological angiogenesis, thrombolysis, arteriosclerosis and restenosis following angioplastry, acute and chronic indications such as inflammation, septic chock, septice- mia, hypotension, acute lung injury (ALI), Acute Respiratory Distress Syndrome (ARDS), pulmonary embolism, disseminated intravascular coagulation (DIC), sepsis, systemic inflammatory response syndrome (SIRS), vascular restenosis, platelet deposition, myocardial infarction, angiogenesis, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis; asthma, bronchitis, idiopathic pulmonary fibrosis, pneumonia, pulmonary edema, pulmonary obstructive disease, endotoxin induced lung damage, non cell lung cancer; inflammatory bowel disease, pancreatitis, trauma-induced shock, bronchial asthma, allergic rhinitis, rheumatoid arthritis, cystic fibrosis, stroke, acute bronchitis, chronic bronchitis, acute bronchiolitis, chronic bronchiolitis, osteoarthritis, gout, spondylarthropathris, ankylosing spondylitis, Reiter's syndrome, psoriatic arthropathy, enterapathric spondylitis, juvenile arthropathy or juvenile ankylosing spondylitis, reactive arthropathy, infectious or post-infectious arthritis, gonoccocal arthritis, tuberculous arthritis, viral arthritis, fungal arthritis, syphilitic arthritis, Lyme disease, arthritis associated with "vasculitic syndromes," polyar- teritis nodosa, hypersensitivity vasculitis, Luegenec's granulomatosis, polymyalgin rheu- matica, joint cell arteritis, calcium crystal deposition arthropathris, pseudo gout, non-articular rheumatism, bursitis, tenosynomitis, epicondylitis (tennis elbow), carpal tunnel syndrome, repetitive use injury (typing), miscellaneous forms of ar-thritis, neuropathic joint disease (charco and joint), hemarthrosis (hemarthrosic), Henoch-Schonlein Purpura, hypertrophic osteoarthropathy, multicentric reticulohistiocytosis, arthritis associated with certain diseases, surcoilosis, hemochromatosis, sickle cell disease and other hemo-globinopathries, hyperli- poproteineimia, hypogammaglobulinemia, hyperparathyroidism, acromegaly, familial Mediterranean fever, Behat's Disease, systemic lupus erythrematosis, relapsing, and multiple organ failure resulting from any of the preceding pathologic processes. In one embodiment, the diseases or disorders are Respiratory disease and Inflammatory disease. In one embodiment, Respiratory disease and Inflammatory disease include lower respiratory diseases such as systemic inflammatory response syndrome, asthma, bronchitis, acute lung injury, acute resporatory distress syndrome, idiopathic pulmonary fibrosis, pneumonia, pulmonary edema, pulmonary obstructive disease, endotoxin induced lung damage, non cell lung cancer; inflammatory bowel disease, sepsis, septic shock, acute respiratory distress syndrome, pancreatitis, trauma-induced shock, bronchial asthma, allergic rhinitis, rheumatoid arthritis, cystic fibrosis, stroke, acute bronchitis, chronic bronchitis, acute bronchiolitis, chronic bronchiolitis, osteoarthritis, gout, spondylarthropathris, ankylosing spondylitis, Reiter's syndrome, psoriatic arthropathy, enterapathric spondylitis, juvenile ar- thropathy or juvenile ankylosing spondylitis, reactive arthropathy, infectious or post-infectious arthritis, gonoccocal arthritis, tuberculous arthritis, viral arthritis, fungal arthritis, syphilitic arthritis, Lyme disease, arthritis associated with "vasculitic syndromes," polyarthritis nodosa, hypersensitivity vasculitis, Luegenec's granulomato-sis, polymyalgin rheumatica, joint cell arteritis, calcium crystal deposition arthropathris, pseudo gout, non-articular rheumatism, bursitis, tenosynomitis, epicondylitis (tennis elbow), carpal tunnel syndrome, repetitive use injury (typing), miscellaneous forms of arthritis, neuropathic joint disease (charco and joint), hemarthrosis (hemarthrosic), Henoch-Schonlein Purpura, hypertrophic osteoarthropathy, multicentric reticulohistiocytosis, arthritis associated with certain diseases, surcoilosis, hemochromatosis, sickle cell disease and other hemoglobinopathries, hyperlipopro-teineimia, hy- pogammaglobulinemia, hyperparathyroidism, acromegaly, familial Mediterranean fever, Behat's Disease, systemic lupus erythrematosis, relapsing, and multiple organ failure result-ing from any of the preceding pathologic processes. In another embodiment, the diseases or disorders are Thrombotic or Coagulopatic related diseases or disorders. In one embodiment, Thrombotic or Coagulopatic related dis- ease include vascular diseases and inflammatory responses such as deep venous thrombo- sis, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, tumour metastasis, inflammation, septic chock, hypotension, acute lung injury (ALI), Acute Respiratory Distress Syndromr (ARDS), pulmonary embolism, disseminated intravascular coagulation (DIC), sepsis, sys- temic inflammatory response syndrome (SIRS), vascular restenosis, platelet deposition, myocardial infarction, angiogenesis, or the treatment of mammals with atherosclerotic vessels at risk for thrombosis, and multiple organ failure resulting from any of the preceding pathologic processes. The disease or disorder associated with pathophysiological TF function are not lim- ited to in vivo coagulopatic disorders such as those named above, but includes ex vivo
TF/FVIIa related processes such as coagulation that may result from the extracorporeal circulation of blood, including blood removed in-line from a patient in such processes as dialysis procedures, blood filtration, or blood bypass during surgery. ■ In one embodiment the disease or disorder associated with pathophysiological TF function is selected from the group consisting of cancer, tumor growth, tumor metastasis, and pathological angiogenesis. The terms "cancer" or "tumor growth" are to be understood as referring to all forms of neoplastic cell growth, including both cystic and solid tumors, bone and soft tissue tumors, including both benign and malignant tumors, including tumors in anal tissue, bile duct, blad- der, blood cells, bone, bone (secondary), bowel (colon & rectum), brain, brain (secondary), breast, breast (secondary), carcinoid, cervix, children's cancers, eye, gullet (oesophagus), head & neck, kaposi's sarcoma, kidney, larynx, leukaemia (acute lymphoblastic), leukaemia (acute myeloid), leukaemia (chronic lymphocytic), leukaemia (chronic myeloid), leukaemia (other), liver, liver (secondary), lung, lung (secondary), lymph nodes (secondary), lymphoma (hodgkin's), lymphoma (non-hodgkin's), melanoma, mesothelioma, myeloma, ovary, pancreas, penis, prostate, skin, soft tissue sarcomas, stomach, testes, thyroid, unknown primary tumor, vagina, vulva, womb (uterus). Soft tissue tumors include Benign schwannoma Monosomy, Desmoid tumor, Lipo- blastoma, Lipoma, Uterine leiomyoma, Clear cell sarcoma, Dermatofibrosarcoma, Ewing sarcoma, Extraskeletal myxoid chondrosarcoma, Liposarcoma myxoid, Liposarcoma, well differentiated, Alveolar rhabdomyosarcoma, and Synovial sarcoma. Specific bone tumor include Nonossifying Fibroma, Unicameral bone cyst, Enchon- droma, Aneurysmal bone cyst, Osteoblastoma, Chondroblastoma, Chondromyxofibroma, Ossifying fibroma and Adamantinoma, Giant cell tumor, Fibrous dysplasia, Ewing's Sarcoma, Eosinophilic Granuloma, Osteosarcoma, Chondroma, Chondrosarcoma, Malignant Fibrous Histiocytoma, and Metastatic Carcinoma. Leukaemias referes to cancers of the white blood cells which are produced by the bone marrow. This includes but are not limited to the four main types of leukaemia; acute lymphoblastic (ALL), acute myeloblastic (AML), chronic lymphocytic (CLL) and chronic myeloid (CML). In another embodiment, the medicament is formulated for intravenous administration, preferably injection or infusion, in particular injection. In one embodiment, the medicament is formulated in single-unit dosage form; in an- other it is formulated in the form of a first unit dosage form comprising a preparation of a TF antagonist and a second unit dosage form comprising an anti-cancer compound. In one embodiment the TF antagonist and the anti-cancer compound are present in the pharmaceutical formulation in a ratio by mass of between about 1000:1 and about 1:1000 (w/w). The terms "treatment" and "treating" means the administration of an effective amount of a therapeutically active compound of the invention with the purpose of preventing any symptoms or disease state to develop or with the purpose of curing or easing such symptoms or disease states already developed. The term "treatment" is thus meant to include prophylactic treatment. In one embodiment of the invention the first agent is an inactive FVIIa polypeptide. In a further embodiment of the invention, the first agent is an inactive FVIIa polypeptide is native human FVIIa or a fragment thereof catalytically inactivated in the active site. In one embodiment the inactive FVIIa polypeptide is native human FVIIa catalytically inactivated in the active site. In one embodiment the FVIIa polypeptide is catalytically inactivated in the active site with a chloromethyl ketone inhibitor independently selected from the group consisting of Phe-Phe-Arg chloromethyl ketone, Phe-Phe-Arg chloromethylketone, D-Phe- Phe-Arg chloromethyl ketone, D-Phe-Phe-Arg chloro-methylketone Phe-Pro-Arg chloromethylketone, D-Phe-Pro-Arg chloromethylketone, Phe-Pro-Arg chloromethylketone, D-Phe- Pro-Arg chloromethylketone, L-Glu-Gly-Arg chloro-methylketone and D-Glu-Gly-Arg chloro- methylketone, Dansyl-Phe-Phe-Arg chloromethyl ke-tone, Dansyl-Phe-Phe-Arg chloromethylketone, Dansyl-D-Phe-Phe-Arg chloromethyl ketone, Dansyl-D-Phe-Phe-Arg chloromethylketone, Dansyl-Phe-Pro-Arg chloromethylketone, Dan-syl-D-Phe-Pro-Arg chloromethylketone, Dansyl-Phe-Pro-Arg chloromethylketone, Dansyl-D-Phe-Pro-Arg chloromethylketone, Dansyl-L-Glu-Gly-Arg chloromethylketone and Dansyl-D-Glu-Gly-Arg chloro- methylketone, Tosyl-Lys- chloromethylketone, 2,4-dichloroisocoumarin, di- isopropylfluorophosphate, phenylmethylsulphonyl fluoride. In a further embodiment of the invention, the first agent is an antibody which immunoreacts with an epitope present on human TF. In one embodiment the antibody inhibits the binding of human coagulation factor Vila to human TF. In one embodiment the epitope comprises one or more of the residues Trp45, Lys46 and Tyr94. In one embodiment the antibody is a monoclonal antibody. In one embodiment the antibody is a recombinant antibody. In one embodiment the antibody is a Fab fragment. In one embodiment the antibody is a F(ab)2 fragment. In one embodiment the antibody is a F(ab')2 fragment. In one embodiment the an- tibody is a single chain Fv fragment. In one embodiment the antibody has a Kd for binding to human TF within the range of 10"15- 10"8 M. In one embodiment the antibody has a Kd for binding to human TF within the range of 10"15- 10"10 M. In one embodiment of the invention, the second agent is selected from the group consisting of protein ionophores, cytostatica, che-motherapeutic compound, compounds which induce apoptosis, compound containing ra-dionuclides, antisense nucleotide molecules independent selected from the group consisting of PNAs, DNAs, RNAs and LNAs. In one embodiment of the invention, the second agent comprises a cytotoxic protein or peptide. In one embodiment of the invention, the disease or disorder associated with patho- physiological TF function are deep venous thrombosis, chronic thromboembolic diseases or disorders associated with fibrin formation, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, cancer, tumour metastasis, pathological angiogenesis, thrombolysis, arteriosclerosis and restenosis following angioplastry, acute and chronic indications such as in- flammation, septic chock, septicemia, hypotension, acute lung injury (ALI), Acute Respiratory Distress Syndrome (ARDS), pulmonary embolism, disseminated intravascular coagulation (DIC), sepsis, systemic inflammatory response syndrome (SIRS), vascular restenosis, platelet deposition, myocardial infarction, angiogenesis, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis; asthma, bronchitis, idiopathic pulmonary fibrosis, pneumonia, pulmonary edema, pulmonary obstructive disease, endotoxin induced lung damage, non cell lung cancer; inflammatory bowel disease, pancreatitis, trauma- induced shock, bronchial asthma, allergic rhinitis, rheumatoid arthritis, cystic fibrosis, stroke, acute bronchitis, chronic bronchitis, acute bronchiolitis, chronic bronchiolitis, osteoarthritis, gout, spondylarthropathris, ankylosing spondylitis, Reiter's syndrome, psoriatic arthropathy, enterapathric spondylitis, juvenile arthropathy or juvenile ankylosing spondylitis, reactive ar- thropathy, infectious or post-infectious arthritis, gonoccocal arthritis, tuberculous arthritis, viral arthritis, fungal arthritis, syphilitic arthritis, Lyme disease, arthritis associated with "vas- culitic syndromes," polyarteritis nodosa, hypersensitivity vasculitis, Luegenec's granulomato- sis, polymyalgin rheumatica, joint cell arteritis, calcium crystal deposition ar-thropathris, pseudo gout, non-articular rheumatism, bursitis, tenosynomitis, epicondylitis (tennis elbow), carpal tunnel syndrome, repetitive use injury (typing), miscellaneous forms of arthritis, neuropathic joint disease (charco and joint), hemarthrosis (hemarthrosic), Henoch-Schonlein Purpura, hypertrophic osteoarthropathy, multicentric reticulohistiocytosis, arthritis associated with certain diseases, surcoilosis, hemochromatosis, sickle cell disease and other hemo- globinopathries, hyperiipoproteineimia, hypogammaglobulinemia, hyperparathyroidism, acromegaly, familial Mediterranean fever, Behat's Disease, systemic lupus erythrematosis, relapsing, and multiple organ failure resulting from any of the preceding pathologic processes. In one embodiment of the invention, the second agent is a cytostatica. In one embodiment of the invention, the second agent is a chemotherapeutic compound. In one embodiment of the invention, the second agent is a compound containing radionuclides. In one embodiment of the invention, the second agent is an antisense nucleotide molecule. In one embodiment of the invention, the second agent is melphalan. In one embodiment of the invention, the second agent is a compound containing I125. In a further embodiment of the invention, the second agent is a cytotoxic protein or peptide. In a further embodiment of the invention, the second agent comprises the amino acid sequence (KLAKLAK)n, wherein n is selected from the group consisting of 1 , 2, 3, 4, 5, 6, 7, and 8. In one embodiment n is 2. In a further embodiment of the invention, the second agent has the amino acid se- quence (KLAKLAK)2
The term "anti-cancer compound" as used herein refers to a compound effective in the treatment of cancer. This includes compounds, which kill the tumor cells and/or reduce the size of the tumor and/or reduce the growth and/or spreading of the tumor. The term also encompasses traditional chemotherapeutic drugs and cytotoxic drugs. Other exemplary anti-cancer compounds include, e.g., neomycin, podophyl- lotoxin(s), TNF-alpha, calcium ionophores, calcium-flux inducing compounds, anti-tubulin drugs, colchicine, taxol, vinblastine, vincristine, vindescine, and combretastatin. In one embodiment of the invention, the anti-cancer compound is an antibody, such as monoclonal antibodies. MAbs have been developed for the treatment of leukaemia and lymphoma as well as solid tumor, and this principle is gaining increasing interest. These antibodies work either by inhib-iting functions that are vital for survival of the tumor cells, by delivering a toxic pay- load, by interrupting key signalling events, or by induction of ADCC or CDC against the tumor cells. Death of the tumor cells might then lead to the release of tumor antigens that "vacci- nates" the immune system and stimulates it to produce a secondary response that then targets the tumor cell (i.e. 'internal vaccination' as described below). Over-expressed onco- genes and tumor-specific antigens are key targets for many mAbs under development. Tumor antigens are described for example in Stauss H, Kawakami Y and Parmiani G: Tumor antigens recognized by T cells and antibodies. Taylor and Frances (2003). The invention covers antibodies raised against these targets. The invention also covers antibodies raised against viral antigens. In one embodiment of the invention, the anti-cancer compound is an antibody, such as Rituximab, Alemtuzumab, Trastuzumab, HuMax-CD20, HuMax-EGFr, Zamyl, Pertuzu- mab, antibodies against tissue factor, killer Ig-like receptors (KIR) and laminin-5. In one embodiment the anti-cancer compound is further combined with additional
ADCC-enhancing compounds, e.g. blocking anti-KIR antibodies or activating NKG2A antibodies or IL-2. In one embodiment of the invention, the anti-cancer compound is an antibody against viral antigens. In one embodiment of the invention, the anti-cancer compound is a regulator of cell cycle control and/or apoptosis. A series of regulators are involved in the maintenance of normal cell-cycle. Compounds, which target regulators such as (i) cdc-25 (with NSC 663284 as a non-limiting example (Pu et al (2003) J Biol Chem 278, 46877)), (ii) cyclin-dependent kinases that over- stimulate the cell cycle (with the following non-limiting examples: flavopiridol (L868275, HMR1275; Aventis), 7-hydroxystaurosporine (UCN-01 , KW-2401; Kyowa Hakko Kogyo) and roscovitine (R-roscovitine, CYC202; Cyclacel) - as reviewed by Fischer & Gianella-Borradori (2003) Exp Op Invest Drugs 12, 955-970), and (iii) telomerase, the enzyme that helps cancer cells rebuild its telomeres are within the present invention such as the following non-limiting examples BIBR1532 (Damm et al (2001 ) EMBO J 20, 6958-6968) and SOT-095 (Tauchi et al (2003) Oncogene 22, 5338-5347) . Furthermore, drugs that interfere with apoptotic pathways are within the present invention.such as the following non-limiting examples: TNF-related apoptosis-inducing ligand (TRAIL)/apoptosis-2 ligand (Apo-2L), antibodies that activate TRAIL receptors, IFNα and anti-sense Bcl-2.(see Igney and Krammer (2002) Nature Rev. Cancer 2, 277-288; Makin and Dive (2003) Trends Mol Med 9, 2519; Smyth et al (2003) Immunity 18, 1-6; Panaretakis et al (2003) Oncogene 22, 4543-4556 and references therein). . In an embodiment of the invention above the compounds are selected from the group comprising cdc-25, NSC 663284, flavopiridol, 7-hydroxystaurosporine, roscovitine , BIBR1532 SOT-095, TNF-related apoptosis-inducing ligand (TRAIL)/apoptosis-2 ligand (Apo-2L), antibodies that activate TRAIL receptors, IFNα and anti-sense Bcl-2. In one embodiment the anti-cancer compound is a growth factor inhibitor, such as an antibody against a growth factor. A number of mAbs against growth factors and growth factor receptors are being developed for the treatment of cancer. Thus, as a non-limiting example, members of the epi- dermal growth factor receptor (EGF-R) family are abnormally activated in many epithelial tumors, which often correlate with more aggressive clinical course. Antibodies directed against the extracellular ligand binding domain of these receptors and low molecular weight molecules that inhibit their tyrosine kinase domains are in late-stage clinical development or approved for treatment of cancer either as single compounds or in combination with other can- cer drugs. Non-limiting examples are Herceptin (monoclonal antibody), cetuximab (monoclonal antibody), Tarceva (low molecular weight inhibitor), and Iressa (low molecular weight inhibitor). In addition, the ligand can be neutralised before binding to the receptor. In an embodiment of the invention the growth factor inhibitors are selected from the group comprising Herceptin (monoclonal antibody), cetuximab (monoclonal antibody), Tarceva (low molecular weight inhibitor), and Iressa (low molecular weight inhibitor). In one embodiment of the invention, the anti-cancer compound is an anti- angiogenesis drug. In one embodiment of the invention, the anti-cancer compound is an anti-metastatic compound. Tumor growth is dependent on sufficient blood supply and hence development of new blood vessels. This general feature of solid tumors seems attractive from a therapeutic point of view, i.e. reduced tumor growth and tumor regression is expected when treating patients wth cancer with anti-angionesis drugs. Currently more than 60 anti-angionesis drugs are in clinical trials including the natural occurring endostatin and angiostatin (reviewed in Marx (2003) Science 301, 452-454). But also older chemotherapy drugs, other medicines and radiation therapy have anti-angiogenic effects. In one embodiment of the invention, the anti-angiogenic compounds is selected from the group consisting of endostatin, angiostatin, antibodies that block factors that initiate angiogenesis (e.g. anti-VEGF - Avastin), low mo- lecular compounds that inhibit angiogenesis by inhibiting key elements in relevant signal transduction pathways. Attacking the vasculature of the tumor and the extracellular matrix has attracted increasing awareness. The following principles have so far been developed: Blockage of the endothelial cell, administration of angiostatin and endostatin, VEGF targeting and extracellu- lar matrix. In an embodiment of the invention the anti-angiogenesis drug is selected from the group consisting of avastin, neovastat, thalidomide, PTK787, ZK222584, ZD-6474 , SU6668, PD547.632, VEGF-Trap, CEP-7055, NM-3, SU11248. (Nature Biotech 20, 1067-1068). In one embodiment of the invention, the anti-cancer compound is a viral targeting compound. Viral targeting uses a recombinant virus - usually replication incompetent - to destroy a tumor directly. In practice, at least one round of replication occurs before the virus is incapacitated. Hence, the tumor is lysed, which often leads to systemic immunization with resulting protection. This approach has been refined further using genetic modification to enhance the immune response. For example, the genetic insertion of a human GM-CSF gene into a herpes simplex virus type 2 vector has been used improve the efficacy of the vaccine. In one embodiment of the invention, the anti-cancer compound is a hormonal compounds. Hormonal compounds are primarily know in the treatment of hormonal dependent cancers such as ovarian cancer, breast cancer and prostate cancer such as anti-androgen and anti-oestrogen therapy.
The following list of components or compounds that can be used together with a TF antagonist in combination therapy of cancer by enhancing the efficacy of the treatment is not intended in any way to limit the scope of the invention: a) Adjuvants: Immunotherapy consist of specific and non-specific modalities. As examples of nonspecific immunotherapy are adjuvants acting primarily as catalyst for the initiation of an immune response. Non-limiting examples of such vaccine adjuvants are QS21 , GM-CSF and CpG oligodeoxynucleotides, lipopolysaccharide and polyinosinic:polycytidylic acid. In one embodiment of the invention the TF antagonist is combined with one or more adjuvants. In an embodiment of the invention the adjuvants are selected from the group consisting of QS21, GM-CSF and CpG oligodeoxynucleotides, lipopolysaccharide and polyinos- inic:polycytidylic acid, b) Cytokines Non-limiting examples of cytokines are IFN-α, IFN-β, IFN-γ, IL-2, PEG-IL-2, IL-4; IL-
6, IL-7, IL-12, IL-13, IL-15, IL-18, IL-21 , IL-23, IL-27, IL-28a, IL-28b, IL-29, GM-CSF, Flt3 ligand or stem cell factor. Other compounds that may be combined with a cytokine include autologous TILs, Cis-platin, tamoxifen, DTIC, Carmustine, carboplatin, Vinblastine, temo- zolomide, Vindesine, 5-fluorouracil, Fotemustine, autologous LAK cells, and Gemcitabine. c) Passive immunotherapy Examples of passive immunotherapy are re-infusion of tumor infiltrating T cells expansion of tumor-specific antigens , genetic enhancement of T-cells and blocking of inhibitory receptors. d) Cell adoptive therapy Cell adoptive therapy may include isolation of cells that can stimulate or exert an anti-cancer response from patients and expand these into higher number and reintroduce into the patient. In one aspect this may be CD4+ or CD8+ T cells recognizing tumor specific antigens or tumor-associated antigens. In another aspect this may be B cell expressing antibodies specific for tumor specific antigens or tumor-associated antigens. In another aspect this may be NK cells that are able to kill the tumor cells. In a preferred aspect this may be dendritic cells (DC) that are cultured in vivo with a DC expanding compound (e.g. GM-CSF or Flt3-L), loaded with tumor specific antigens or tumor-associated antigens and reintroduced in vivo. In an embodiment of the invention the cell adoptive therapy comprises CD4+ or CD8+ T cells recognizing tumor specific antigens or tumor-associated antigens. In an embodiment of the invention the cell adoptive therapy comprises B cell expressing antibodies specific for tumor specific antigens or tumor-associated antigens. In an embodiment of the invention cell adoptive therapy comprises NK cells that are able to kill the tumor cells. In an embodiment of the invention cell adoptive therapy comprises dendritic cells (DC). In an embodiment of the above the dendritic cells are cultured in vivo with a DC expanding compound (e.g. GM-CSF or Flt3-L), loaded with tumor specific antigens or tumor-associated antigens and reintroduced in vivo. e) Intracellular signalling inhibitors A number of different principles has been explored including interference with tyrosine kinases (Gleevec®, imatinib mesylate), the ras signalling pathway, and regulators of protein trafficking. In one embodiment of the invention, the Intracellular signalling inhibitor is selected from the group consisting of tyrosine kinase inhibitors, serine/threonine kinase inhibitors, protein-tyrosine phosphatases inhibitors, dual-specificity phosphatases inhibitors, or serine/threonine phosphatases inhibitors. f) Anti-anergic compounds Anti-anergic compounds are small compounds, proteins, glycoproteins or antibodies that can break tolerance to tumor and cancer antigens. , Although the presence of tumor infiltrating lymphocytes (TILs) correlates with improved clini- cal outcome in a number of different cancer forms, there is clearly a need to improve the activity of these TILs due to anergy or tolerance to tumor antigens. The anergic condition may in a substantial number of cases be counteracted by monoclonal antibodies that prevent CTLA-4 -induced anergy or tolerance. Blockade of CTLA-4 has been shown in animal models to improve the effectiveness of cancer therapy suggesting that CTLA-4 blockade can be used to break the tolerance to cancer and tumor antigens. A non-limiting example of a monoclonal antibody that may be used for induction of the activity of TILs is MDX-010 (Phan , et al. (2003) Proc. Natl. Acad. Sci. U.S.A. 100: 8372). g) Therapeutic vaccines The development of almost all human cancers involves genetic alterations, and this may lead to expression of altered molecules in tumor cells and over-expression of normal molecules, respectively. In principle, these changes should lead to an immune response from the host (immune surveillance). Obviously, this theoretical activation of the immune system only leads to spontaneous regression of the tumor in very few, exceptional cases. This may, among other factors, be due to lack of "warning signals", a phenomenon that has attracted increasing interest. Tumour specific antigens have been identified, and vaccination with such antigens may stimulate the immune system to eradicate the Tumor-specific antigens (TSAs) are a relatively small group of antigens exemplified by the cancer-testis antigens. These genes are silent in normal tissue but are expressed by cancerous cells. They are highly specific mark- ers of disease and include MAGE (melanoma antigen gene) found in melanoma. Tumor-associated antigens. Tumor-associated antigens (TAAs) are usually differentiation antigens expressed by normal cells but massively over-expressed in cancerous tissue. Targets initially thought to be specific for a particular cancer are actually quite common in many tumors, such as the gangliosides and mucin antigens. Classical differentiation antigens include MART-1 (melanoma antigen recognized by T cells) and gp 100, both from melanoma, tyrosinase and gp75. Mutational antigens. Point mutations are common in many cancers, and often occur in a similar location, such as the common mutation of the P53 oncogene. In vitro induction of human cytotoxic T-lymphocyte (CTL) responses against peptides of mutant and wild-type p53 has been reported. In a mouse model, mutant p53-pulsed dendritic cells were able to induce p53 specific CTL and inhibit the growth of established tumors. Viral antigens. Certain viruses are oncogenic and gene products encoded by these viruses can elicit immune responses and thus serve as cancer antigens. An example is the E6 and E7 proteins from human papilloma virus type 16, which have been shown to induce cytotoxic T-lymphocyte responses in vitro. Tumor-specific antigens, tumor-associated antigens and/or mutational antigens and viral antigens may be used either as peptides, recombinant purified single-agent antigens, combination of recombinant purified antigens and/or purified or pools of antigens isolated from cancer cells or tumor cells as a vaccine to elicit an anti-tumor immune response. Simi- larly, peptides, recombinant purified single-agent antigens, combinations of recombinant antigens and/or purified or pools of antigens isolated from virus-infected cells may be used in a vaccine to elicit a response against virus-infected cells. Therapeutic vaccines can also be in the form of a DNA vaccine to elicit immune response against cancer and virus-infected cells. Said vaccine-mediated elicitation of an anti-tumor response or a response against virus- infected cells may be enhanced by administering adjuvants, cytokines, CpG oligodeoxynucleotides, dendritic cells, GM-CSF, or heat-shock proteins. In one embodiment of the invention, combination therapy is performed by administering a TF antagonist with one or more therapeutic vaccines with or without adjuvants, cytokines, CpG oligodeoxynucleotides, dendritic cells, GM-CSF, or heat-shock proteins. h) metalloproteinase inhibitors Metastatic cancer cells penetrate the extracellular matrix (ECM) and the basement membrane of the blood vessels to metastasise to a target organ (ectopic site). EMC consists of proteins embedded in a carbohydrate complex (heparan sulfate peptidoglycans), and proteases surrounding the tumour are active in this breaking down the host tissue. Anti- metastatic compounds antagonise the effect of such proteases (e.g. metalloproteinase inhibitors) (Coussens et al. Science 2002;295:2387-2392). i) internal vaccination therapy "Internal vaccination" and "internal vaccination therapy" refer to drug- or radition- induced cell death of tumor cells that leads to elicitation of an immune response directed towards (i) said tumor cells as a whole or (ii) parts of said tumor cells including (a) secreted proteins, glycoproteins or other products, (b) membrane-associated proteins or glycoproteins or other components associated with or inserted in membranes and (c) intracellular proteins or other intracellular components. The immune response may be humoral (i.e. antibody - complement-mediated) or cell-mediated including but not limited to development of cytotoxic T lymhocytes that recognized said tumor cells or parts thereof. Internal vaccination bears many similarities to other vaccination procedures and involves many or all of the same cellular components of the hematopoietic and immune system with the advantage 'that the immunogens or antigenic components are endogenous and thus representative for the antigenic repertoire of said tumor cells. Internal vaccination may thus be considered personalized vaccination, which is elicited by use of general procedures for cancer treatment leading to tumor cell death. In addition to radiotherapy, non-limiting examples of drugs and compounds that can be used to induce said tumor cell-death and internal vaccination are conventional chemotherapeutic compounds, cell-cycle inhibitors, anti-angiogenesis drugs, monoclonal antibodies, apoptosis-inducing compounds and signal transduction inhibitors. The terms "cytotoxic domain" or "cytotoxic compound" as used herein refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells.
The terms "cytotoxic domain" and "cytotoxic compound" may be used interchangebly. The term is intended to include radioactive isotopes or radionuclides (e.g. 1131 , 1125, Y90 and Re186), chemotherapeutic agents, cytostatica and toxins such as enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof. Cytotoxic compounds also includes peptides, which directly mediate mitochondrial cytochrome C release and apoptosis, e.g., (KLAKLAK)2 including both enantiomers (Ellerby et al. Nature medicine 5, (9) 1032- 1038, 1999). A "chemotherapeutic compound" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic compounds include adriamycin, doxorubicin, epirubi- cin, 5-fluorouracil, cytosine arabinoside ("Ara-C"), cyclophosphamide, thiotepa, busulfan, cy- toxin, taxoids, e.g. paclitaxel (TAXOL™, Bristol-Myers Squibb Oncology, Princeton, N.J.), and doxetaxel, toxotere, methotrexate, cisplatin, vinblastine, bleomycin, etoposide, ifos- famide, mitomycin C, mitoxantrone, vincristine, vinorelbine, carboplatin, teniposide, dauno- mycin, carminomycin, aminopterin, dactinomycin, mitomycins, esperamicins (see U.S. Pat. No. 4,675,187), melphalan and other related nitrogen mustards. Also included in this definition are hormonal compounds that act to regulate or inhibit hormone action on tumors, such as tamoxifen and onapristone. Also included are aminoglutethimide, asparaginase, bleomy- cin, L-buthiamine sulphoxide, busulfan, camptothecin, carboplatin, carmustine, chlorambucil, cisplatin, cyclophosphamide, cytarabine HCI, dacarbazine, dactinomycin, daunorubicin HCI, doxorubicin HCI, edatrexate, estramustine phosphate sodium, etopside (V16-213), floxu- ridine, fluorouracil, flutamide, gallium nitrite, hydroxyurea, idarubicin, ifosfamide, interferon alpha -2a, alpha -2b, leuprolide acetate (LHRH-releasing factor analogue), lomustine, mechlorethamine HCI, megestrol, melphalan, mercaptopurine, methotrexate, mitomycin, mi- totane, mitoxantrone HCI, octreotide, plicamycin, prednisone, procarbazine HCI, streptozo- cin, tamoxifen citrate, taxanes, alkylating agents, antibiotics, topisomerase inhibitors, anti- metabolites, vinca alkaloids, taxoids, thioguanine, thiotepa, tiasofuran, topotecan, vinblastine sulphate, amsacrine, azacitidine,hexamethylmelamine, interleukin 2, mitoguazone, methyl glyoxal bis-guanylhydrazone, pentostatin, semustine and teniposide. Cytotoxic compounds may include, but are not limited to, a therapeutically effective amount of: toxins; drugs; enzymes; cytokines; radionuclides; photodynamic compounds; and molecules which induce apoptosis (e.g., Fas ligand or 2-methoxyestradioI). Toxins may include a therapeutically effective amount of ricin A chain, mutant Pseudomonas exotoxins, diphtheria toxoid, streptonigrin, boamycin, saporin, gelonin, and pokeweed antiviral protein. Drugs may include a therapeutically effective amount of cytotoxic drugs including, but not limited to, fludarabine, chlorambucil, daunorubicin, doxorubicin (e.g., in liposomes), cisplatin, bleomycin, melphalan, mitomycin-C, and methotrexate. Due to the sensitivity of B cells to radiation, radionuclides may include, but are not limited to, proteins labeled with radiometals such as yttrium which emits a high energy beta particle, and I125 that emits Auger electrons, that may be absorbed by adjacent TF presenting cells. Photodynamic compounds may include therapeutically effective amounts of porphyrins and their derivatives. The methods for coupling ligands or targeting molecules with therapeutic compounds are well known to those skilled in the art (See, for example, conjugates as reviewed by Ghetie et al., 1994, Pharma- col. Ther. 63:209-34; U.S. Pat. No. 5,789,554, the disclosure of which is herein incorporated by reference). Often such methods utilize one of several available hetero-bifunctional reagents used for coupling or linking molecules. Cytotoxic compounds suitable for use herein include conventional chemotherapeu- tics, such as vinblastine, anthracycline antitumor antibiotics including doxorubicin, 2- pyrrolino- doxorubicin, Doxorubicin hydrochloride (Adriamycin) (Schally VA and Nagy A (1999) Eur J Endocrinol 141 , 1-14, Vasey PA et al (1999) Clin Cancer Res 5, 83-94), bleo- mycin, methotrexate, 5-fluorouracil, 6-thioguanine, cytarabine, cyclophosphamide (N,N-bis- (beta-Chlorethyl)-amino-1-oxa-3-aza-2-phosphocyclohexane-2-oxide) and cisplatinum, as well as other conventional chemotherapeutics as described in Cancer: Principles and Prac- tice of Oncology, 2d ed., V. T. DeVita, Jr., S. Hellman, S. A. Rosenberg, J.B. Lippincott Co., Philadelphia, Pa., 1985, Chapter 14. Another suitable cytotoxic compound within the present invention is a trichothecene. Trichothecenes are drugs produced by soil fungi of the class Fungi imperfecti or isolated from Baccharus megapotamica (Bamburg, J. R. Proc. Molec. Subcell. Biol. 8:41-110, 1983; Jarvis & Mazzola, Ace. Chem. Res. 15:338-395, 1982). They appear to be the most toxic molecules that contain only carbon, hydrogen and oxygen
(Tamm, C. Fortschr. Chem. Org. Naturst. 31 :61-117, 1974). They are all reported to act at the level of the ribosome as inhibitors of protein synthesis at the initiation, elongation, or termination phases. There are two broad classes of trichothecenes: those that have only a central ses- quiterpenoid structure and those that have an additional macrocyclic ring (simple and macro- cyclic trichothecenes, respectively). The simple trichothecenes may be subdivided into three groups (i.e., Group A, B, and C) as described in U.S. Pat. Nos. 4,744,981 and 4,906,452 (incorporated herein by reference). Representative examples of Group A simple trichothecenes include: Scirpene, Roridin C, dihydrotrichothecene, Scirpen-4, 8-diol, Verrucarol, Scirpentriol, T-2 tetraol, pentahydroxyscirpene, 4-deacetylneosolaniol, trichodermin, deacetylcalonectrin, calonectrin, diacetylverrucarol, 4-monoacetoxyscirpenol, 4,15-diacetoxyscirpenol, 7- hydroxydiacetoxyscirpenol, 8-hydroxydiacetoxy-scirpenol (Neosolaniol), 7,8- dihydroxydiacetoxyscirpenol, 7-hydroxy-8-acetyldiacetoxyscirpenol, 8-acetylneosolaniol, NT- 1 , NT-2, HT-2, T-2, and acetyl T-2 toxin. Representative examples of Group B simple tricho- thecenes include: Trichothecolone, Trichothecin, deoxynivalenol, 3-acetyldeoxynivalenol, 5- acetyldeoxynivalenol, 3,15-diacetyldeoxynivalenol, Nivalenol, 4-acetylnivalenol (Fusarenon- X) 4,15-idacetylnivalenol, 4,7,15-triacetylnivalenol, and tetra-acetylnivalenol. Representative examples of Group C simple trichothecenes include: Crotocol and Crotocin. Representative macrocyclic trichothecenes include Verrucarin A, Verrucarin B, Verrucarin J (Satratoxin C), Roridin A, Roridin D, Roridin E (Satratoxin D), Roridin H, Satratoxin F, Satratoxin G, Satratoxin H, Vertisporin, Mytoxin A, Mytoxin C, Mytoxin B, Myrotoxin A, Myrotoxin B, Myrotoxin C, Myrotoxin D, Roritoxin A, Roritoxin B, and Roritoxin D. In addition, the general "trichothecene" sesquiterpenoid ring structure is also present in compounds termed "baccharins" isolated from the higher plant Baccharis megapotamica, and these are described in the litera- ture, for instance as disclosed by Jarvis et al. (Chemistry of Alleopathy, ACS Symposium Series No. 268: ed. A. C. Thompson, 1984, pp. 149-159). Other suitable cytotoxic compound within the present invention include N,N-cis(2- chloroethyI)N-nitroso-urea (BCNU), D-myo-inositoI-1,2,6-trisphosphate, Melphalan (p-Di-(2- chloroethyl)-amino-L-phenylalanine), Procarbazine (p-(N'-Methyl-hydrazinomethyl)-N- isopropyl-benzamide), Dactinomycin (Actinomycin D), Polyestradiolphosphate, thalidomid, temozolomide, mitozolomide, mercaptopurine, N-methylformamide, 2-amino-1 ,3,4- thiadiazole, hexamethylmelamine, gallium nitrate, 3% thymidine, dichloromethotrexate, mi- toguazone, suramin, bromodeoxyuridine, iododeoxyuridine, semustine, 1-(2-chloroethyl)-3- (2,6-dioxo-3-piperidyl)-1-nitrosourea, N,N'-hexamethyIene-bis-acetamide, azacitidine, dibro- modulcitol, Erwinia asparaginase, ifosfamide, 2-mercaptoethane sulfonate, teniposide, taxol, 3-deazauridine, soluble Baker's antifol, homoharringtonine, cyclocytidine, acivicin, ICRF-187, spiromustine, levamisole, chlorozotocin, aziridinyl benzoquinone, spirogermanium, aclarubi- cin, pentostatin, PALA, carboplatin, amsacrine, caracemide, iproplatin, misonidazole, dihy- dro-5-azacytidine, 4'-deoxy-doxorubicin, menogaril, triciribine phosphate, fazarabine, tiazo- , furin, teroxirone, ethiofos, N-(2-hydroxyethyl)-2-nitro-1H-imidazole-1-acetamide, mitoxantrone, acodazole, amonafide, fludarabine phosphate, pibenzimol, didemnin B, merbarone, dihydrolenperone, flavone-8-acetic acid, oxantrazole, ipomeanol, trimetrexate, deoxysper- gualin, echinomycin, and dideoxycytidine (see NCI Investigational Drugs, Pharmaceutical Data 1987, NIH Publication No. 88-2141, Revised November 1987) are also preferred. In one embodiment of the invention, the cytotoxic compound stimulates the production of free radicals NO*, O2*. In one embodiment of the invention, the cytotoxic compound stimulates apoptosis by regulation of p53, superoxiddismutase, phospholipase C, cyclooxygenase 2, caspase- associated recruitment domains (CARD). In one embodiment of the invention, the cytotoxic compound is selected from the group consisting of an cyclooxygenase 2 inhibitor, apoptin, chicken Anemia Virus (CAV), (e.g. CAV protein VP1, VP2 and VP3 (apoptin) US patent 5,981 ,502), Sulforaphane (SUL). Radionuclides useful within the present invention include gamma-emitters, positron- emitters, Auger electron-emitters, X-ray emitters and fluorescence-emitters, with beta- or alpha-emitters preferred for therapeutic use. Radionuclides are well-known in the art and include 123-1, 125-1, 130-1, 131-1, 133-1, 135-1 47-Sc, 72-As, 72-Se, 90-Y, 88-Y, 97-Ru, 100-Pd, 101m-Rh, 119-Sb, 128-Ba, 197-Hg, 211 -At, 212-Bi, 153-Sm, 169-Eu, 212-Pb, 109-Pd, 111- ln, 67-Ga, 68-Ga, 64-Cu, 67-Cu, 75-Br, 76-Br, 77-Br, 99m-Tc, 11-C, 13-N, 15-0, 166-Ho and 18-F. Preferred therapeutic radionuclides include 188-Re, 186-Re, 203-Pb, 212-Pb, 212-Bi, 109-Pd, 64-Cu, 67-Cu, 90-Y, 125-1, 131-1, 77-Br, 211-At, 97-Ru, 105-Rh, 198-Au and 199- Ag, 166-Ho or 177-Lu. In one embodiment of the invention the TF antagonist comprises gadophrin e.g. gadophrin-2 (previously referred as bis-gadolinium-mesoporhyrin) (Pislaru SV. et al., Circula- tion, 99 (5) pp. 690-696, 1999). The terms "Nucleic acid sequence" or "nucleotide sequence" as used herein refers to an oligonucleotide, nucleotide, or polynucleotide, and fragments or portions thereof, and to DNA or RNA of genomic or synthetic origin which may be single- or double-stranded, and represent the sense or antisense strand. Similarly, "amino acid sequence" as used herein refers to an oligopeptide, peptide, polypeptide, or protein sequence, and fragments or portions thereof, and to naturally occurring or synthetic molecules. Where "amino acid sequence" is recited herein to refer to an amino acid sequence of a naturally occurring protein molecule, "amino acid sequence" and like terms, such as "polypeptide" or "protein" are not meant to limit the amino acid sequence to the complete, native amino acid sequence associated with the recited protein molecule. The terms "PNA" or "Peptide nucleic acid", as used herein, refers to a molecule which comprises an oligomer to which an amino acid residue, such as lysine, and an amino group have been added. These small molecules, also designated anti-gene agents, stop transcript elongation by binding to their complementary strand of nucleic acid (Nielsen, P. E. et al. (1993) Anticancer Drug Des. 8:53-63). The term "antisense", as used herein, refers to nucleotide sequences which are complementary to a specific DNA or RNA sequence. The term "antisense strand" is used in reference to a nucleic acid strand that is complementary to the "sense" strand. Antisense molecules may be produced by any method, including synthesis by ligating the gene(s) of interest in a reverse orientation to a viral promoter which permits the synthesis of a complementary strand. Once introduced into a cell, this transcribed strand combines with natural sequences produced by the cell to form duplexes. These duplexes then block either the further transcription or translation. In this manner, mutant phenotypes may be generated. The designation "negative" is sometimes used in reference to the antisense strand, and "positive" is some- times used in reference to the sense strand. A further prefered antisense molecule includes Locked Nucleic Acids (LNAs) in which the 2'-hydroxyl group is linked to the 3' or 4' carbon atom of the sugar ring thereby forming a bicyclic sugar moiety. The linkage is preferably a methelyne (-CH2 -).sub.n group bridging the 2' oxygen atom and the 4' carbon atom wherein n is 1 or 2. LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226. The terms "FVIIa polypeptide" or "FVIIa polypeptides" as used herein means native Factor Vila, as well as equivalents of Factor Vila that contain one or more amino acid sequence alterations relative to native Factor Vila (i.e., Factor VII variants), and/or contain truncated amino acid sequences relative to native Factor Vila (i.e., Factor Vila fragments). Such equivalents may exhibit different properties relative to native Factor Vila, including stability, phospholipid binding, altered specific proteolytic activity, and the like. As used herein, "Factor VII equivalent" encompasses, without limitation, equivalents of Factor Vila exhibiting TF binding activity. The term "TF binding activity" as used herein means the ability of a FVIIa polypeptide or TF antagonist to inhibit the binding of recombinant human 125l-FVIIa to cell surface human TF. The TF binding activity may be measured as described in Assay 3. Factor VII equivalents also includes proteolytically inactive variants of FVIIa. In one embodiment of the invention the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the lysine corresponding to position 341 of wild type human FVIIa. In one embodiment of the invention the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the serine corresponding to position 344 of wild type human FVIIa. In one embodiment of the invention the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the aspartic acid corresponding to position 242 of wild type human FVIIa. In one embodiment of the invention the FVIIa polypeptide is human FVIIa, which has an amino acid substitution of the histidine corresponding to position 193 of wild type human FVIIa. In one embodiment the FVIIa polypeptide is FVII-(K341A) In one embodiment the FVIIa polypeptide is FVII-(S344A) In one embodiment the FVIIa polypeptide is FVII-(D242A) In one embodiment the FVIIa polypeptide is FVII-(H193A) The terminology for specific amino acid substitutions used herein are as follows. The first letter represent the amino acid naturally present at a position of wild type human FVIIa. The following number represent the position in wild type human FVIIa. The second letter represent the different amino acid substituting for the natural amino acid. An example is FVII- (K341A), where a lysine at position 341 of wild type human FVIIa is replaced by an alanine. In another example, FVII-(K341A/S344A), the lysine at position 341 of wild type human FVIIa is replaced by an alanine and the serine in position 344 of wild type human FVIIa is replaced by an alanine in the same Factor VII polypeptide. The terms "Factor VII" or "FVII" are intended to mean Factor VII polypeptides in their uncleaved (zymogen) form. The terms "Factor Vila" or "FVIIa" are intended to mean native bioactive forms of FVII. Typically, FVII is cleaved between residues 152 and 153 to yield FVIIa. The term "Fac- tor Vila" is also intended to encompass, without limitation, polypeptides having the amino acid sequence 1-406 of wild-type human Factor Vila (as disclosed in U.S. Patent No. 4,784,950), as well as wild-type Factor Vila derived from other species, such as, e.g., bovine, porcine, canine, murine, and salmon Factor Vila. It further encompasses natural allelic variations of Factor Vila that may exist and occur from one individual to another. Also, degree and location of glycosylation or other post-translation modifications may vary depending on the chosen host cells and the nature of the host cellular environment. The terms "variant" or "variants", as used herein, is intended to designate human Factor VII having the sequence of SEQ ID NO: 1 , wherein one or more amino acids of the parent protein have been substituted by another amino acid and/or wherein one or more amino acids of the parent protein have been deleted and/or wherein one or more amino acids have been inserted in protein and/or wherein one or more amino acids have been added to the parent protein. Such addition can take place either at the N-terminal end or at the C-terminal end of the parent protein or both. In one embodiment of the invention the variant has a total amont of amino acid substitutions and/or additions and/or deletions independently selected from the group consisting of 1 , 2, 3, 4, 5, 6, 7, 8, 9, and 10.
The ability of any particular cytotoxic compound to mediate lysis of the tumor cell target can be assayed. The tumor cells of interest are grown and labeled in vivo or in vitro; the cytotoxic compound is added to the tumor cell culture. Cytolysis of the target tumor cells is detected by the release of label from the lysed cells. The cytotoxic compound that is capable of mediating cell ablation, lysis or apoptosis in the in vitro test can then be used therapeutically in that particular patient. The term "active site" and the like when used herein with reference to FVIIa refer to the catalytic and zymogen substrate binding site, including the "Si" site of FVIIa as that term is defined by Schecter, I. and Berger, A., (1967) Biochem. Biophys. Res. Commun. 7:157- 162. The term "TF-mediated coagulation activity" means coagulation initiated by TF through the formation of the TF/FVIIa complex and its activation of FIX and Factor X to FlXa and FXa, respectively. TF-mediated coagulation activity is measured in a FXa generation as- say. The term "FXa generation assay" as used herein is intended to mean any assay where activation of FX is measured in a sample comprising TF, FVIIa, FX, calcium and phospholip- ids. An example of a FXa generation assay is described in assay 1. A TF/FVIIa mediated or associated process or event, or a process or event associated with TF-mediated coagulation activity, is any event, which requires the presence of TF/FVIIa. Such processes or events include, but are not limited to, formation of fibrin which leads to thrombus formation; platelet deposition; proliferation of smooth muscle cells (SMCs) in the vessel wall, such as, for example, in intimal hyperplasia or restenosis, which is thought to result from a complex interaction of biological processes including platelet deposition and thrombus formation, release of chemotactic and mitogenic factors, and the migration and proliferation of vascular smooth muscle cells into the intima of an arterial segment; and deleterious events associated with post-ischemic reperfusion, such as, for example, in patients with acute myocardial infarction undergoing coronary thrombolysis. The no-reflow phenomenon, that is, lack of uniform perfusion to the microvascula- ture of a previously ischemic tissue has been described for the first time by Krug et al.j (Circ. Res. 1966; 19:57-62). The general mechanism of blood clot formation is reviewed by Ganong, in Review of Medical Physiology, 13th ed., Lange, Los Altos Calif., pp 411-414 (1987). Coagulation requires the confluence of two processes, the production of thrombin which induces platelet aggregation and the formation of fribrin which renders the platelet plug stable. The process comprises several stages each requiring the presence of discrete proenzymes and profac- tors. The process ends in fibrin crosslinking and thrombus formation. Fibrinogen is converted to fibrin by the action of thrombin. Thrombin, in turn, is formed by the proteolytic cleavage of prothrombin. This proteolysis is effected by FXa which binds to the surface of activated plate- lets and in the presence of FVa and calcium, cleaves prothrombin. TF/FVIIa is required for the proteolytic activation of FX by the extrinsic pathway of coagulation. Therefore, a process mediated by or associated with TF/FVIIa, or an TF-mediated coagulation activity includes any step in the coagulation cascade from the formation of the TF/FVIIa complex to the formation of a fibrin platelet clot and which initially requires the presence of TF/FVIIa. For example, the TF/FVIIa complex initiates the extrinsic pathway by activation of FX to FXa, FIX to FlXa, and additional FVII to FVIIa. TF/FVIIa mediated or associated process, or TF-mediated coagulation activity can be conveniently measured employing standard assays such as those described in Roy, S., (1991) J. Biol. Chem. 266:4665-4668, and O'Brien, D. et al., (1988) J. Clin. Invest. 82:206-212 for the conversion of FX to FXa in the presence of TF/FVIIa and other necessary reagents. It should be noted that peptides, proteins and amino acids as used herein can comprise or refer to "natural", \_e_, naturally occurring amino acids as well as "non.classical" D- amino acids including, but not limited to, the D-isomers of the common amino acids, α- isobutyric acid, 4-aminobutyric acid, hydroxyproline, sarcosine, citrulline, cysteic acid, t- butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, designer amino acids such as β-methyl amino acids, Cα-methyl amino acids, Nα-methyl amino acids, and amino acid analogues in general. In addition, the amino acids can include Abu, 2-amino butyric acid; γ-Abu, 4-aminobutyric acid; ε-Ahx, 6-aminohexanoic acid; Aib, 2-amino-isobutyric acid; β-Ala, 3-aminopropionic acid; Orn, ornithine; Hyp, trans-hydroxyproline; Nle, norleucine; Nva, norvaline. The three-letter indication "GLA" as used herein means 4-carboxyglutamic acid (γ- carboxyglutamate). By "catalytically inactivated in the active site of the FVIIa polypeptide" is meant that a FVIIa inhibitor is bound to the FVIIa polypeptide and decreases or prevents the FVIIa- catalysed conversion of FX to FXa. A FVIIa inhibitor may be identified as a substance, which reduces the amidolytic activity by at least 50% at a concentration of the substance at 400 μM in the FVIIa amidolytic assay described by Persson et al. (Persson et al., J. Biol. Chem. 272: 19919-19924 (1997)). Preferred are substances reducing the amidolytic activity by at least 50% at a concentration of the substance at 300 μM; more preferred are substances reducing the amidolytic activity by at least 50% at a concentration of the substance at 200 μM. The "FVIIa inhibitor" may be selected from any one of several groups of FVIIa directed inhibitors. Such inhibitors are broadly categorised for the purpose of the present invention into i) inhibitors which reversibly bind to FVIIa and are cleavable by FVIIa, ii) inhibitors which reversibly bind to FVIIa but cannot be cleaved, and iii) inhibitors which irreversibly bind to FVIIa. For a review of inhibitors of serine proteases see Proteinase Inhibitors (Research Monographs in cell and Tissue Physiology; v. 12) Elsevier Science Publishing Co., Inc., New York (1990). The FVIIa inhibitor moiety may also be an irreversible FVIIa serine protease inhibitor. Such irreversible active site inhibitors generally form covalent bonds with the protease active site. Such irreversible inhibitors include, but are not limited to, general serine protease inhibitors such as peptide chloromethylketones (see, Williams et al., J. Biol. Chem. 264:7536-7540 (1989)) or peptidyl cloromethanes; azapeptides; acylating agents such as various guanidinobenzoate derivatives and the 3-alkoxy-4-chloroisocoumarins; sulphonyl fluorides such as phenylmethylsulphonylfluoride (PMSF); diisopropylfluorophosphate (DFP); tosylpropylchloromethyl ketone (TPCK); tosyllysylchloromethyl ketone (TLCK); nitrophenyl- sulphonates and related compounds; heterocyclic protease inhibitors such as isocoumarines, and coumarins. Examples of peptidic irreversible FVIIa inhibitors include, but are not limited to, Phe-Phe-Arg chloromethyl ketone, Phe-Phe-Arg chloromethylketone, D-Phe-Phe-
Arg chloromethyl ketone, D-Phe-Phe-Arg chloromethylketone Phe-Pro-Arg chloromethylketone, D-Phe-Pro-Arg chloromethylketone, Phe-Pro-Arg chloromethylketone, D-Phe-Pro-Arg chloromethylketone, L-Glu-Gly-Arg chloromethylketone and D-Glu-Gly-Arg chloromethylketone. Examples of FVIIa inhibitors also include benzoxazinones or heterocyclic analogues thereof such as described in PCT/DK99/00138. Examples of other FVIIa inhibitors include, but are not limited to, small peptides such as for example Phe-Phe-Arg, D-Phe-Phe-Arg, Phe-Phe-Arg, D-Phe-Phe-Arg, Phe-Pro- Arg, D-Phe-Pro-Arg, Phe-Pro-Arg, D-Phe-Pro-Arg, L- and D-Glu-Gly-Arg; peptidomimetics; benzamidine systems; heterocyclic structures substituted with one or more amidino groups; aromatic or heteroaromatic systems substituted with one or more C(=NH)NHR groups in which R is H, C1-3alkyl, OH or a group which is easily split of in vivo.
The TF antagonist and anti-cancer compound as defined in the present specification may be administered simultaneously or sequentially. The factors may be supplied in single- dosage form wherein the single-dosage form contains both compounds, or in the form of a kit-of-parts comprising a preparation of a TF antagonist as a first unit dosage form and a preparation of an anti-cancer compound as a second unit dosage form. Whenever a first or second or third, etc., unit dose is mentioned throughout this specification this does not indi- cate the preferred order of administration, but is merely done for convenience purposes By "simultaneous" dosing of a preparation of a TF antagonist and a preparation of anti-cancer compound is meant administration of the compounds in single-dosage form, or administration of a first agent followed by administration of a second agent with a time separation of no more than 15 minutes, preferably 10, more preferred 5, more preferred 2 min- utes. Either factor may be administered first. By "sequential" dosing is meant administration of a first agent followed by administration of a second agent with a time separation of more than 15 minutes. Either of the two unit dosage form, or coagulation factor proteins, may be administered first. Preferably, both products are injected through the same intravenous access. In the present specification, amino acids are represented using abbreviations, as indicated in table 1 , approved by IUPAC-IUB Commission on Biochemical Nomenclature (CBN). Amino acid and the like having isomers represented by name or the following abbreviations are in natural L-form unless otherwise indicated. Further, the left and right ends of an amino acid sequence of a peptide are, respectively, the N- and C-termini unless otherwise specified.
Table 1 : Abbreviations for amino acids:
Figure imgf000033_0001
The invention also relates to TF antagonists as mentioned above. The TF antagonist may be produced by recombinant DNA techniques. To this end, DNA sequences encoding human FVIIa may be isolated by preparing a genomic or cDNA library and screening for DNA sequences coding for all or part of the protein by hybridization using synthetic oligonucleotide probes in accordance with standard techniques (cf. Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1989). For the present purpose, the DNA sequence encoding the protein is preferably of human origin, i.e. derived from a human genomic DNA or cDNA library. The DNA sequences encoding the human FVIIa polypeptides may also be prepared synthetically by established standard methods, e.g. the phosphoamidite method described by Beaucage and Caruthers, Tetrahedron Letters 22 (1981 ), 1859 - 1869, or the method described by Matthes et al., EMBO Journal 3 (1984), 801 - 805. According to the phosphoamidite method, oligonucleotides are synthesized, e.g. in an automatic DNA synthesizer, purified, annealed, ligated and cloned in suitable vectors. The DNA sequences may also be prepared by polymerase chain reaction using specific primers, for instance as described in US 4,683,202, Saiki et al., Science 239 (1988), 487 - 491 , or Sambrook et al., supra. The DNA sequences encoding the human FVIIa polypeptides are usually inserted into a recombinant vector which may be any vector, which may conveniently be subjected to recombinant DNA procedures, and the choice of vector will often depend on the host cell into which it is to be introduced. Thus, the vector may be an autonomously replicating vector, i.e. a vector, which exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, e.g. a plasmid. Alternatively, the vector may be one which, when introduced into a host cell, is integrated into the host cell genome and replicated together with the chromosome(s) into which it has been integrated. The vector is preferably an expression vector in which the DNA sequence encoding the human FVIIa polypeptides is operably linked to additional segments required for transcription of the DNA. In general, the expression vector is derived from plasmid or viral DNA, or may contain elements of both. The term, "operably linked" indicates that the segments are arranged so that they function in concert for their intended purposes, e.g. transcription initiates in a promoter and proceeds through the DNA sequence coding for the polypeptide. The promoter may be any DNA sequence, which shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell. Examples of suitable promoters for directing the transcription of the DNA encoding the human FVIIa polypeptide in mammalian cells are the SV40 promoter (Subramani et al., Mol. Cell Biol. 1 (1981 ), 854 -864), the MT-1 (metallothionein gene) promoter (Palmiter et al.,
Science 222 (1983), 809 - 814), the CMV promoter (Boshart et al., Cell 41 :521-530, 1985) or the adenovirus 2 major late promoter (Kaufman and Sharp, Mol. Cell. Biol, 2:1304-1319, 1982). An example of a suitable promoter for use in insect cells is the polyhedrin promoter (US 4,745,051 ; Vasuvedan et al., FEBS Lett. 311 , (1992) 7 - 11 ), the P10 promoter (J.M. Vlak et al., J. Gen. Virology 69, 1988, pp. 765-776), the Autographa californica polyhedrosis virus basic protein promoter (EP 397485), the baculovirus immediate early gene 1 promoter (US 5,155,037; US 5,162,222), or the baculovirus 39K delayed-early gene promoter (US 5,155,037; US 5,162,222). Examples of suitable promoters for use in yeast host cells include promoters from yeast glycolytic genes (Hitzeman et al., J. Biol. Chem. 255 (1980), 12073 - 12080; Alber and Kawasaki, J. Mol. Appl. Gen. 1 (1982), 419 - 434) or alcohol dehydrogenase genes (Young et al., in Genetic Engineering of Microorganisms for Chemicals (Hollaender et al, eds.), Plenum Press, New York, 1982), or the TPI1 (US 4,599,311 ) or ADH2-4c (Russell et al., Nature 304 (1983), 652 - 654) promoters. Examples of suitable promoters for use in filamentous fungus host cells are, for instance, the ADH3 promoter (McKnight et al., The EMBO J. 4 (1985), 2093 - 2099) or the tpiA promoter. Examples of other useful promoters are those derived from the gene encoding A oryzae TAKA amylase, Rhizomucor miehei aspartic proteinase, A niger neutral α-amylase, A. niger acid stable α-amylase, A. niger or A. awamoπ'glucoamylase (gluA), Rhizomucor miehei lipase, A oryzae alkaline protease, A. oryzae triose phosphate isomerase or A. nidulans acetamidase. Preferred are the TAKA-amylase and gluA promoters. Suitable promoters are mentioned in, e.g. EP 238 023 and EP 383 779. The DNA sequences encoding the human FVIIa polypeptides may also, if necessary, be operably connected to a suitable terminator, such as the human growth hormone terminator (Palmiter et al., Science 222, 1983, pp. 809-814) or the TPI1 (Alber and Kawasaki, J. Mol. Appl. Ge 1 , 1982, pp. 419-434) or ADH3 (McKnight et al., The EMBO J. 4, 1985, pp. 2093-2099) terminators. The vector may also contain a set of RNA splice sites located downstream from the promoter and upstream from the insertion site for the FVIIa sequence itself. Preferred RNA splice sites may be obtained from adenovirus and/or immunoglobulin genes. Also contained in the expression vectors is a polyadenylation signal located downstream of the insertion site. Particularly preferred polyadenylation signals include the early or late polyadenylation signal from SV40 (Kaufman and Sharp, ibid.), the polyadenylation signal from the adenovirus 5 Elb region, the human growth hormone gene terminator (DeNoto et al. Nuc. Acids Res. 9:3719-3730, 1981) or the polyadenylation signal from the human FVII gene or the bovine FVII gene. The expression vectors may also include a noncoding viral leader sequence, such as the adenovirus 2 tripartite leader, located between the promoter and the RNA splice sites; and enhancer sequences, such as the SV40 enhancer. The recombinant vector may further comprise a DNA sequence enabling the vector to replicate in the host cell in question. An example of such a sequence (when the host cell is a mammalian cell) is the SV40 origin of replication. When the host cell is a yeast cell, suitable sequences enabling the vector to replicate are the yeast plasmid 2μ replication genes REP 1-3 and origin of replication. The vector may also comprise a selectable marker, e.g. a gene the product of which complements a defect in the host cell, such as the gene coding for dihydrofolate reductase (DHFR) or the Schizosaccharomyces pombe TP gene (described by P.R. Russell, Gene 40, 1985, pp. 125-130), or one which confers resistance to a drug, e.g. ampicillin, kanamycin, tetracyclin, chloramphenicol, neomycin, hygromycin or methotrexate. For filamentous fungi, selectable markers include amdS, pyrG, argB, niaD or sC. To direct the human FVIIa polypeptides of the present invention into the secretory pathway of the host cells, a secretory signal sequence (also known as a leader sequence, prepro sequence or pre sequence) may be provided in the recombinant vector. The secretory signal sequence is joined to the DNA sequences encoding the human FVIIa polypeptides in the correct reading frame. Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the peptide. The secretory signal sequence may be that, normally associated with the protein or may be from a gene encoding another secreted protein. For secretion from yeast cells, the secretory signal sequence may encode any signal peptide, which ensures efficient direction of the expressed human FVIIa polypeptides into the secretory pathway of the cell. The signal peptide may be naturally occurring signal peptide, or a functional part thereof, or it may be a synthetic peptide. Suitable signal peptides have been found to be the α-factor signal peptide (cf. US 4,870,008), the signal peptide of mouse salivary amylase (cf. O. Hagenbuchle et al., Nature 289, 1981 , pp. 643-646), a modified carboxypeptidase signal peptide (cf. L.A. Vails et al., Cell 48, 1987, pp. 887-897), the yeast
BAR1 signal peptide (cf. WO 87/02670), or the yeast aspartic protease 3 (YAP3) signal peptide (cf. M. Egel-Mitani et al., Yeas.6, 1990, pp. 127-137). For efficient secretion in yeast, a sequence encoding a leader peptide may also be inserted downstream of the signal sequence and upstream of the DNA sequence encoding the human FVIIa polypeptides. The function of the leader peptide is to allow the expressed peptide to be directed from the endoplasmic reticulum to the Golgi apparatus and further to a secretory vesicle for secretion into the culture medium (i.e. exportation of the human FVIIa polypeptides across the cell wall or at least through the cellular membrane into the periplasmic space of the yeast cell). The leader peptide may be the yeast alpha-factor leader (the use of which is described in e.g. US 4,546,082, US 4,870,008, EP 16 201 , EP 123 294, EP 123 544 and EP 163 529). Alternatively, the leader peptide may be a synthetic leader peptide, which is to say a leader peptide not found in nature. Synthetic leader peptides may, for instance, be constructed as described in WO 89/02463 or WO 92/11378. For use in filamentous fungi, the signal peptide may conveniently be derived from a gene encoding an Aspergillus sp. amylase or glucoamylase, a gene encoding a Rhizomucor miehei lipase or protease or a Humicola lanuginosa lipase. The signal peptide is preferably derived from a gene encoding A. oryzae TAKA amylase, A. niger neutral α-amylase, A. niger acid-stable amylase, or A. niger glucoamylase. Suitable signal peptides are disclosed in, e.g. EP 238023 and EP 215594. For use in insect cells, the signal peptide may conveniently be derived from an insect gene (cf. WO 90/05783), such as the lepidopteran Manduca sexta adipokinetic hormone precursor signal peptide (cf. US 5,023,328). The procedures used to ligate the DNA sequences coding for the human FVIIa polypeptides, the promoter and optionally the terminator and/or secretory signal sequence, respectively, and to insert them into suitable vectors containing the information necessary for replication, are well known to persons skilled in the art (cf., for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, New York, 1989). Methods of transfecting mammalian cells and expressing DNA sequences introduced in the cells are described in e.g. Kaufman and Sharp, J. Mol. Biol. 159 (1982), 601 - 621; Southern and Berg, J. Mol. Appl. Genet. 1 (1982), 327 - 341 ; Loyter et al., Proc. Natl. Acad. Sci. USA 79 (1982), 422 - 426; Wigler et al., Cell 14 (1978), 725; Corsaro and Pearson, Somatic Cell Genetics 7 (1981), 603, Graham and van der Eb, Virology 52 (1973), 456; and Neumann et al., EMBO J. 1 (1982), 841 - 845. Selectable markers may be introduced into the cell on a separate plasmid at the same time as the gene of interest, or they may be introduced on the same plasmid. If on the same plasmid, the selectable marker and the gene of interest may be under the control of different promoters or the same promoter, the latter arrangement producing a dicistronic message. Constructs of this type are known in the art (for example, Levinson and Simonsen, U.S. Pat. No. 4,713,339). It may also be advantageous to add additional DNA, known as "carrier DNA," to the mixture that is introduced into the cells. After the cells have taken up the DNA, they are grown in an appropriate growth medium, typically 1-2 days, to begin expressing the gene of interest. As used herein the term "appropriate growth medium" means a medium containing nutrients and other components required for the growth of cells and the expression of the human FVIIa polypeptides of inter- est. Media generally include a carbon source, a nitrogen source, essential amino acids, essential sugars, vitamins, salts, phospholipids, protein and growth factors. For production of gamma-carboxylated proteins, the medium will contain vitamin K, preferably at a concentration of about 0.1 μg/ml to about 5 μg/ml. Drug selection is then applied to select for the growth of cells that are expressing the selectable marker in a stable fashion. For cells that have been transfected with an amplifiable selectable marker the drug concentration may be increased to select for an increased copy number of the cloned sequences, thereby increasing expression levels. Clones of stably transfected cells are then screened for expression of the human FVIIa polypeptide of interest. The host cell into which the DNA sequences encoding the human FVIIa polypeptides is introduced may be any cell, which is capable of producing the posttranslational modified human FVIIa polypeptides and includes yeast, fungi and higher eucaryotic cells. Examples of mammalian cell lines for use in the present invention are the COS-1 (ATCC CRL 1650), baby hamster kidney (BHK) and 293 (ATCC CRL 1573; Graham et al., J. Gen. Virol. 36:59-72, 1977) cell lines. A preferred BHK cell line is the tk" ts13 BHK cell line (Waechter and Baserga, Proc. Natl. Acad. Sci. USA 79: 1106-1110, 1982, incorporated herein by reference), hereinafter referred to as BHK 570 cells. The BHK 570 cell line has been deposited with the American Type Culture Collection, 12301 Parklawn Dr., Rockville, Md. 20852, under ATCC accession number CRL 10314. A tk" ts13 BHK cell line is also available from the ATCC under accession number CRL 1632. In addition, a number of other cell lines may be used within the present invention, including Rat Hep I (Rat hepatoma; ATCC CRL 1600), Rat Hep II (Rat hepatoma; ATCC CRL 1548), TCMK (ATCC CCL 139), Human lung (ATCC HB 8065), NCTC 1469 (ATCC CCL 9.1), CHO (ATCC CCL 61) and DUKX cells (Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 77:4216-4220, 1980). Examples of suitable yeasts cells include cells of Saccharomyces spp. or Schizosac- charomyces spp., in particular strains of Saccharomyces cerevisiae or Saccharomyces kluyveri. Methods for transforming yeast cells with heterologous DNA and producing heterologous polypeptides there from are described, e.g. in US 4,599,311 , US 4,931 ,373, US 4,870,008, 5,037,743, and US 4,845,075, all of which are hereby incorporated by reference. Transformed cells are selected by a phenotype determined by a selectable marker, commonly drug resis- tance or the ability to grow in the absence of a particular nutrient, e.g. leucine. A preferred vector for use in yeast is the POT1 vector disclosed in US 4,931 ,373. The DNA sequences encoding the human FVIIa polypeptides may be preceded by a signal sequence and optionally a leader sequence, e.g. as described above. Further examples of suitable yeast cells are strains of Kluyveromyces, such as K. lactis, Hansenula, e.g. H. polymorpha, or Pichia, e.g. P. pastoris (cf. Gleeson et al., J. Gen. Microbiol. 132, 1986, pp. 3459-3465; US 4,882,279). Examples of other fungal cells are cells of filamentous fungi, e.g. Aspergillus spp., Neurospora spp., Fusarium spp. or Trichoderma spp., in particular strains of A. oryzae, A. nidulans or A. niger. The use of Aspergillus spp. for the expression of proteins is described in, e.g., EP 272 277, EP 238 023, EP 184438 The transformation of F. oxysporum may, for instance, be carried out as described by Malardier et al., 1989, Gene 78: 147-156. The transformation of Trichoderma spp. may be performed for instance as described in EP 244 234. When a filamentous fungus is used as the host cell, it may be transformed with the DNA construct of the invention, conveniently by integrating the DNA construct in the host chromosome to obtain a recombinant host cell. This integration is generally considered to be an advantage as the DNA sequence is more likely to be stably maintained in the cell. Integration of the DNA constructs into the host chromosome may be performed according to conventional methods, e.g. by homologous or heterologous recombination. Transformation of insect cells and production of heterologous polypeptides therein may be performed as described in US 4,745,051 ; US 4,879,236; US 5,155,037; 5,162,222; EP
397,485) all of which are incorporated herein by reference. The insect cell line used as the host may suitably be a Lepidoptera cell line, such as Spodoptera frugiperda cells or Trichoplusia ni cells (cf. US 5,077,214). Culture conditions may suitably be as described in, for instance, WO 89/01029 or WO 89/01028, or any of the aforementioned references. The transformed or transfected host cell described above is then cultured in a suitable nutrient medium under conditions permitting expression of the human FVIIa polypeptide after which all or part of the resulting peptide may be recovered from the culture. The medium used to culture the cells may be any conventional medium suitable for growing the host cells, such as minimal or complex media containing appropriate supplements. Suitable media are available from commercial suppliers or may be prepared according to published recipes (e.g. in catalogues of the American Type Culture Collection). The human FVIIa polypeptide produced by the cells may then be recovered from the culture medium by conventional procedures including separating the host cells from the medium by centrifugation or filtration, precipitating the proteinaqueous components of the supernatant or filtrate by means of a salt, e.g. ammonium sulphate, purification by a variety of chromatographic procedures, e.g. ion exchange chromatography, gelfiltration chromatography, affinity chromatography, or the like, dependent on the type of polypeptide in question. For the preparation of recombinant human FVIIa polypeptides, a cloned wild-type FVIIa DNA sequence is used. This sequence may be modified to encode a desired FVIIa variant. The complete nucleotide and amino acid sequences for human FVIIa are known. See U.S. Pat. No. 4,784,950, which is incorporated herein by reference, where the cloning and expression of recombinant human FVIIa is described. The bovine FVIIa sequence is described in Takeya et al., J. Biol. Chem, 263:14868-14872 (1988), which is incorporated by reference herein. The amino acid sequence alterations may be accomplished by a variety of tech- niques. Modification of the DNA sequence may be by site-specific mutagenesis. Techniques for site-specific mutagenesis are well known in the art and are described by, for example, Zoller and Smith (DNA 3:479-488, 1984). Thus, using the nucleotide and amino acid sequences of FVII, one may introduce the alterations of choice. DNA sequences for use within the present invention will typically encode a pre-pro peptide at the amino-terminus of the FVIIa protein to obtain proper post-translational processing (e.g. gamma-carboxylation of glutamic acid residues) and secretion from the host cell. The pre-pro peptide may be that of FVIIa or another vitamin K-dependent plasma protein, such as factor IX, factor X, prothrombin, protein C or protein S. As will be appreciated by those skilled in the art, additional modifications can be made in the amino acid sequence of FVIIa where those modifications do not significantly impair the ability of the protein to act as a coagulation factor. For example, FVIIa in the catalytic triad can also be modified in the activation cleavage site to inhibit the conversion of zymogen FVII into its activated two-chain form, as generally described in U.S. Pat. No. 5,288,629, incorporated herein by reference. Within the present invention, transgenic animal technology may be employed to produce the human FVIIa polypeptide. It is preferred to produce the proteins within the < mammary glands of a host female mammal. Expression in the mammary gland and subsequent secretion of the protein of interest into the milk overcomes many difficulties encountered in isolating proteins from other sources. Milk is readily collected, available in large quantities, and well characterized biochemically. Furthermore, the major milk1 proteins are present in milk at high concentrations (typically from about 1 to 15 g/l). From a commercial point of view, it is clearly preferable to use as the host a species that has a large milk yield. While smaller animals such as mice and rats can be used (and are preferred at the proof of principle stage), within the present invention it is preferred to use livestock mammals includ- ing, but not limited to, pigs, goats, sheep and cattle. Sheep are particularly preferred due to such factors as the previous history of transgenesis in this species, milk yield, cost and the ready availability of equipment for collecting sheep milk. See WIPO Publication WO 88/00239 for a comparison of factors influencing the choice of host species. It is generally desirable to select a breed of host animal that has been bred for dairy use, such as East Friesland sheep, or to introduce dairy stock by breeding of the transgenic line at a later date. In any event, animals of known, good health status should be used. To obtain expression in the mammary gland, a transcription promoter from a milk protein gene is used. Milk protein genes include those genes encoding caseins (see U.S. Pat. No. 5,304,489, incorporated herein by reference), beta-lactoglobulin, alpha-lactalbumin, and whey acidic protein. The beta-lactoglobulin (BLG) promoter is preferred. In the case of the ovine beta-lactoglobulin gene, a region of at least the proximal 406 bp of 5' flanking sequence of the gene will generally be used, although larger portions of the 5' flanking sequence, up to about 5 kbp, are preferred, such as about 4.25 kbp DNA segment encompassing the 5' flanking promoter and non-coding portion of the beta-lactoglobulin gene. See Whitelaw et al., Biochem J. 286: 31-39 (1992). Similar fragments of promoter DNA from other species are also suitable. Other regions of the beta-lactoglobulin gene may also be incorporated in constructs, as may genomic regions of the gene to be expressed. It is generally accepted in the art that constructs lacking introns, for example, express poorly in comparison with those that contain such DNA sequences (see Brinster et al., Proc. Natl. Acad. Sci. USA 85: 836-840 (1988); Palmiter et al., Proc. Natl. Acad. Sci. USA 88: 478-482 (1991); Whitelaw et al., Transgenic Res. 1: 3-13 (1991); WO 89/01343; and WO 91/02318, each of which is incorporated herein by reference). In this regard, it is generally preferred, where possible, to use genomic sequences containing all or some of the native introns of a gene encoding the protein or poly- peptide of interest, thus the further inclusion of at least some introns from, e.g, the beta- lactoglobulin gene, is preferred. One such region is a DNA segment which provides for intron splicing and RNA polyadenylation from the 3' non-coding region of the ovine beta- lactoglobulin gene. When substituted for the natural 3' non-coding sequences of a gene, this ovine beta-lactoglobulin segment can both enhance and stabilize expression levels of the protein or polypeptide of interest. Within other embodiments, the region surrounding the ini- ' tiation ATG of the sequence encoding the human FVIIa polypeptide is replaced with corresponding sequences from a milk specific protein gene. Such replacement provides a putative tissue-specific initiation environment to enhance expression. It is convenient to replace the entire pre-pro sequence of the human FVIIa polypeptide and 5' non-coding sequences with those of, for example, the BLG gene, although smaller regions may be replaced. For expression of a human FVIIa polypeptide in transgenic animals, a DNA segment encoding the human FVIIa polypeptide is operably linked to additional DNA segments required for its expression to produce expression units. Such additional segments include the above-mentioned promoter, as well as sequences which provide for termination of transcrip- tion and polyadenylation of mRNA. The expression units will further include a DNA segment encoding a secretory signal sequence operably linked to the segment encoding the human FVIIa polypeptide. The secretory signal sequence may be a native secretory signal sequence of the human FVIIa polypeptide or may be that of another protein, such as a milk protein. See, for example, von Heinje, Nuc. Acids Res. 14: 4683-4690 (1986); and Meade et al., U.S. Pat. No. 4,873,316, which are incorporated herein by reference. Construction of expression units for use in transgenic animals is conveniently carried out by inserting a sequence encoding the human FVIIa polypeptide into a plasmid or phage vector containing the additional DNA segments, although the expression unit may be constructed by essentially any sequence of ligations. It is particularly convenient to provide a vector containing a DNA segment encoding a milk protein and to replace the coding sequence for the milk protein with that of the human FVIIa polypeptide, thereby creating a gene fusion that includes the expression control sequences of the milk protein gene. In any event, cloning of the expression units in plasmids or other vectors facilitates the amplification of the human FVIIa polypeptide. Amplification is conveniently carried out in bacterial (e.g. E. coli) host cells, thus the vectors will typically include an origin of replication and a selectable marker functional in bacterial host cells. The expression unit is then introduced into fertilized eggs (including early-stage embryos) of the chosen host species. Introduction of heterologous DNA can be accomplished by one of several routes, including microinjection (e.g. U.S. Pat. No. 4,873,191 ), refroviral infection (Jaenisch, Science 240: 1468-1474 (1988)) or site-directed integration using embryonic stem (ES) cells (reviewed by Bradley et al., Bio/Technology λ0: 534-539 (1992)). The eggs are then implanted into the oviducts or uteri of pseudopregnant females and allowed to develop. Offspring carrying the introduced DNA in their germ line can pass the DNA on to their progeny in the normal, Mendelian fashion, allowing the development of transgenic herds. General procedures for producing transgenic animals are known in the art. See, for example, Hogan et al., Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory, 1986; Simons et al., Bio/Technology 6: 179-183 (1988); Wall et al., Biol. Reprod. 32: 645-651 (1985); Buhler et al., Bio/Technology 8: 140-143 (1990); Ebert et al., Bio/Technology 9: 835-838 (1991 ); Krimpenfort et al. , Bio/Technology 9: 844-847 (1991 ); Wall et al., J. Cell. Biochem. 49: 113-120 (1992); U.S. Pat. Nos. 4,873,191 and 4,873,316; WIPO publications WO 88/00239, WO 90/05188, WO 92/11757; and GB 87/00458, which are incorporated herein by reference. Techniques for introducing foreign DNA sequences into mammals and their germ cells were originally developed in the mouse. See, e.g., Gor- don et al., Proc. Natl. Acad. Sci. USA 77: 7380-7384 (1980); Gordon and Ruddle, Science 214: 1244-1246 (1981); Palmiter and Brinster, Ce//41 : 343-345 (1985); and Brinster et al., Proc. Natl. Acad. Sci. USA 82: 4438-4442 (1985). These techniques were subsequently adapted for use with larger animals, including livestock species (see e.g., WIPO publications WO 88/00239, WO 90/05188, and WO 92/11757; and Simons et al., Bio/Technology 6: 179- 183 (1988). To summarize, in the most efficient route used to date in the generation of trans- genie mice or livestock, several hundred linear molecules of the DNA of interest are injected into one of the pro-nuclei of a fertilized egg according to established techniques. Injection of DNA into the cytoplasm of a zygote can also be employed. Production in transgenic plants may also be employed. Expression may be generalized or directed to a particular organ, such as a tuber. See, Hiatt, Nature 344:469-479 (1990); Edelbaum et al., J. Interferon Res. 12:449-453 (1992); Sijmons et al., Bio/Technology 8:217-221 (1990); and European Patent Office Publication EP 255,378. FVIIa produced according to the present invention may be purified by affinity chromatography on an anti-FVII antibody column. It is preferred that the immunoadsorption col- umn comprise a high-specificity monoclonal antibody. The use of calcium-dependent monoclonal antibodies, as described by Wakabayashi et al., J. Biol. Chem, 261 :11097-11108, (1986) and Thim et al., Biochem. 27: 7785-7793, (1988), incorporated by reference herein, is particularly preferred. Additional purification may be achieved by conventional chemical purification means, such as high performance liquid chromatography. Other methods of purifica- tion, including barium citrate precipitation, are known in the art, and may be applied to the purification of the FVIIa described herein (see, generally, Scopes, R., Protein Purification, Springer-Verlag, N.Y., 1982). Substantially pure FVIIa of at least about 90 to 95% homogeneity is preferred, and 98 to 99% or more homogeneity most preferred, for pharmaceutical uses. Once purified, partially or to homogeneity as desired, the FVIIa may then be used therapeutically. Conversion of single-chain FVII to active two-chain FVIIa may be achieved using factor Xlla as described by Hedner and Kisiel (1983, J. Clin. Invest. 71 : 1836-1841 ), or with other proteases having trypsin-like specificity (Kisiel and Fujikawa, Behring Inst. Mitt. 73: 29- 42, 1983). Alternatively FVII may be autoactivated by passing it through an ion-exchange chromatography column, such as mono Q.RTM. (Pharmacia Fire Chemicals) or the like (Bjo- ern et al., 1986, Research Disclosures 269:564-565). The FVIIa molecules of the present invention and pharmaceutical compositions thereof are particularly useful for administration to humans to treat a variety of conditions involving intravascular coagulation. The compounds of the present invention may have one or more asymmetric centres and it is intended that stereoisomers (optical isomers), as separated, pure or partially purified stereoisomers or racemic mixtures thereof are included in the scope of the invention.
Within the present invention, the TF antagonist may be prepared in the form of pharmaceutically acceptable salts, especially acid-addition salts, including salts of organic acids and mineral acids. Examples of such salts include salts of organic acids such as formic acid, fumaric acid, acetic acid, propionic acid, glycolic acid, lactic acid, pyruvic acid, oxalic acid, succinic acid, malic acid, tartaric acid, citric acid, benzoic acid, salicylic acid and the like. Suitable inorganic acid-addition salts include salts of hydrochloric, hydrobromic, sulphuric and phosphoric acids and the like. Further examples of pharmaceutically acceptable inorganic or organic acid addition salts include the pharmaceutically acceptable salts listed in Journal of Pharmaceutical Science. 66, 2 (1977) which are known to the skilled artisan. Also intended as pharmaceutically acceptable acid addition salts are the hydrates which the present compounds are able to form. The acid addition salts may be obtained as the direct products of compound synthe- sis. In the alternative, the free base may be dissolved in a suitable solvent containing the appropriate acid, and the salt isolated by evaporating the solvent or otherwise separating the salt and solvent. The compounds of this invention may form solvates with standard low molecularweight solvents using methods known to the skilled artisan. The TF antagonist may be administered in pharmaceutically acceptable acid addition salt form or, where appropriate, as a alkali metal or alkaline earth metal or lower alkylammonium salt. Such salt forms are believed to exhibit approximately the same order of activity as the free base forms.
PHARMACEUTICAL COMPOSITIONS In another aspect, the present invention includes within its scope pharmaceutical compositions comprising a TF antagonist and an anti-cancer compound, as the active ingredients, or pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier or diluent. Optionally, the pharmaceutical composition of the invention may further comprise one or more other compounds exhibiting anticoagulant activity, e.g., platelet aggregation inhibitor. The compounds of the invention may be formulated into pharmaceutical composition comprising the compounds and a pharmaceutically acceptable carrier or diluent. Such carriers include water, physiological saline, ethanol, polyols, e.g., glycerol or propylene glycol, or vegetable oils. As used herein, "pharmaceutically acceptable carriers" also encompasses any and all solvents, dispersion media, coatings, antifungal agents, preservatives, isotonic agents and the like. Except insofar as any conventional medium is incompatible with the active ingredient and its intended use, its use in the compositions of the present invention is contemplated. The compositions may be prepared by conventional techniques and appear in conventional forms, for example, capsules, tablets, solutions or suspensions. The pharmaceutical carrier employed may be a conventional solid or liquid carrier. Examples of solid carriers are lactose, terra alba, sucrose, talc, gelatine, agar, pectin, acacia, magnesium stearate and stearic acid. Examples of liquid carriers are syrup, peanut oil, olive oil and water. Similarly, the carrier or diluent may include any time delay material known to the art, such as glyceryl monostearate or glyceryl distearate, alone or mixed with a wax. The formulations may also include wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavouring agents. The formulations of the invention may be formulated so as to provide quick, sustained, or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art. The pharmaceutical compositions can be sterilised and mixed, if desired, with auxiliary agents, emulsifiers, salt for influencing osmotic pressure, buffers and/or colouring substances and the like, which do not deleteriously react with the active compounds. The route of administration may be any route, which effectively transports the active compound to the appropriate or desired site of action, such as oral or parenteral, e.g., rectal, transdermal, subcutaneous, intranasal, intramuscular, topical, intravenous, intraurethral, ophthalmic solution or an ointment, the oral route being preferred. If a solid carrier for oral administration is used, the preparation can be tabletted, placed in a hard gelatine capsule in powder or pellet form or it can be in the form of a troche or lozenge. The amount of solid carrier may vary widely but will usually be from about 25 mg to about 1 g. If a liquid carrier is used, the preparation may be in the form of a syrup, emulsion, soft gelatine capsule or sterile injectable liquid such as an aqueous or non-aqueous liquid suspension or solution. For nasal administration, the preparation may contain a compound of formula (I) dissolved or suspended in a liquid carrier, in particular an aqueous carrier, for aerosol application. The carrier may contain additives such as solubilizing agents, e.g. propylene glycol, surfactants, absorption enhancers such as lecithin (phosphatidylcholine) or cyclodextrin, or preservatives such as parabenes. For parenteral application, particularly suitable are injectable solutions or suspensions, preferably aqueous solutions with the active compound dissolved in polyhydroxylated castor oil. Tablets, dragees, or capsules having talc and/or a carbohydrate carrier or binder or the like are particularly suitable for oral application. Preferable carriers for tablets, dragees, or capsules include lactose, corn starch, and/or potato starch. A syrup or elixir can be used in cases where a sweetened vehicle can be employed. A typical tablet, which may be prepared by conventional tabletting techniques, contains Core: Active compound (as free compound 10 mg or salt thereof) Colloidal silicon dioxide (Areosil®) 1.5 mg '- Cellulose, microcryst. (Avicel®) 70 mg Modified cellulose gum (Ac-Di-Sol®) 7.5 mg Magnesium stearate
Coating: HPMC approx. 9 mg *Mywacett® 9-40 T approx. 0.9 mg Αcylated monoglyceride used as plasticizer for film coating.
The compounds of the invention may be administered to a mammal, especially a human in need of such treatment, prevention, elimination, alleviation or amelioration of various thrombolytic or coagulophatic diseases or disorders as mentioned above. Such mammals also include animals, both domestic animals, e.g. household pets, and non- domestic animals such as wildlife. The essential ingredients (a) a TF antagonist and (b) anti-cancer compound are present in the formulation in such proportion that a dose of the formulation pro-vides an amount of each ingredient that together is a pharmaceutically effective amount to the patient being treated. The dose of composition of the invention to be administered is determined depending upon age, body weight, symptom, the desired therapeutic effect, the route of administration, and the duration of the treatment etc. Typically, the weight ratio of TF antagonist and the amount of anti-cancer compound may vary from a ratio of between about 1 :100 to about 100:1 (w/w). The ratio of TF antagonist to anti-cancer compound may thus be, e.g., about 1 :100, or 1 :90, or 1 :80, or 1:70 or 1:60, or 1:50, or 1 :40, or 1:30, or 1:20, or 1:10, or 1:5, or 1:2, or 1:1 , or 2:1, or 5:1 , or 10:1 , or 20:1, or 30.1 , or 40:1 , or 50:1 , or 60:1, or 70:1, or 80:1 , or 90:1, or 100:1; or between about 1 :90 to about 1 :1 , or between about 1 :80 to about 1 :2, or between about 1 :70 to about 1 :5, or between about 1 :60 to about 1 :10, or between about 1 :50 to about 1 :25, or between about 1 :40 to about 1 :30, or between about 90:1 to about 1 :1 , or between about 80:1 to about 2:1, or between about 70:1 to about 5:1, or between about 60:1 to about 10:1, or between about 50:1 to about 25:1, or between about 40:1 to about 30:1; or between about 10:1 to about 1:10, or between about 5:1 to about 1 :5. In one embodiment of the invention the ratio by mass of TF antagonist and anti- cancer compound is between about 100: 1 and about 1 : 100 (w/w). In one embodiment of the invention the ratio by mass of TF antagonist and anti-cancer compound is between about 1 : 90 to about 1 : 1(w/w). The dose of the TF antagonist suitable for oral, nasal, pulmonal or transdermal administration ranges from about 0.05 mg to about 500 mg/day, e.g., from about 1 mg to about 200 mg/day, or, e.g., from about 5 mg to about 175 mg/day for a 70-kg subject as loading and maintenance doses, depending on the weight of the subject, the condition and the severity of the condition. The compounds may be administered concurrently, simultaneously, or together with a pharmaceutically acceptable carrier or diluent, whether by oral, rectal, or parenteral (including subcutaneous) route. The compounds are often, and preferably, in the form of an alkali metal or earth alkali metal salt thereof. Suitable dosage ranges varies as indicated above depending upon the exact mode of administration, form in which administered, the indication towards which the administration is directed, the subject involved and the body weight of the subject involved, and the preference and experience of the physician or veterinarian in charge.
Assays
Inhibition of FVIIa/phospholipids-embedded TF-catalyzed activation of FX by TF antagonists FXa generation assay (assay 1): In the following example all concentrations are final. Lipidated TF (10 pM), FVIIa
(100 pM) and TF antagonist or FFR-rFVIIa (0 - 50 nM) in HBS/BSA (50 mM hepes, pH 7.4, 150 mM NaCI, 5 mM CaCI2,1 mg/ml BSA) are incubated 60 min at room temperature before FX (50 nM) is added. The reaction is stopped after another 10 min by addition of 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 absorbance at 405 nm continuously for 10 min. IC50 values for TF antagonist 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. Inhibition of FVIIa/cell surface TF-catalyzed activation of FX by TF antagonists (Assay 2): In the following example all concentrations are final. Monolayers of human lung fi- broblasts WI-38 (ATTC No. CCL-75) or human bladder carcinoma cell line J82 (ATTC No. HTB-1 ) or human keratinocyte cell line CCD 1102KerTr (ATCC no. CRL-2310) constitutively expressing TF are employed as TF source in FVIIa/TF catalyzed activation of FX. Confluent cell monolayers in a 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 with 1 mg/ml BSA and 5 mM Ca2+). FVIIa (1 nM), FX (135 nM) and varying con- centrations of TF antagonist or FFR-rFVIIa in buffer B are simultaneously added to the cells. 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 determined 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 IC50 value for FFR-rFVIIa is 1.5 nM in this assay.
Inhibition of 125l-FVIIa binding to cell surface TF by TF antagonists (Assay 3): In the following example all concentrations are final. Binding studies are employed using the human bladder carcinoma cell line J82 (ATTC No. HTB-1 ) or the human keratinocyte cell line (CCD1102KerTr ATCC No CRL-2310) or NHEK P166 (Clonetics No. CC-2507) all constitutively expressing TF. Confluent monolayers in 24-well tissue culture plates are washed once with buffer A (10 mM Hepes, pH 7.45, 150 mM NaCI, 4 mM KCI, and 11 mM glucose) supplemented with 5 mM EDTA and then once with buffer A and once with buffer B (buffer A supplemented with with 1 mg/ml BSA and 5 mM Ca2+). The monolayers are prein- cubated 2 min with 100 μl cold buffer B. Varying concentrations of Mabs (or FFR-FVIIa) 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. Biosensor assay (Assay 4): TF antagonists are tested on the Biacore instrument by passing a standard solution of the TF antagonist over a chip with immobilized TF. 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's are calculated from the sensorgrams using the integrated Biacore evaluation software.
Inhibition of FVIIa/TF-induced p44/42 MAPK activation by TF antagonists with effector domain (Assay 5): The amount of phosphorylated p44/42 MAPK and/or Akt, and/or p90RSK is deter- mined 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 TF antagonist 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.
The present invention is not to be limited in scope by the specific embodiments dis- closed in the examples which are intended as illustrations of a number of aspects of the invention and any embodiments which are functionally equivalent are within the scope of this invention. Those skilled in the art will know, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention de- scribed herein. These and all other equivalents are intended to be encompassed by the following claims.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the full amino acid sequence of native (wild type) human coagulation Factor VII (SEQ ID NO:1).

Claims

1. A pharmaceutical composition useful for preventing or treating a disease or disorder associated with pathophysiological TF function, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient, with the proviso that if said first agent is an antibody against TF, said second agent and optionally further agents is not IL-21 , analogues or derivatives thereof.
2. The pharmaceutical pharmaceutical composition according to claim 1 , wherein said sec- ond compound is a cytotoxic compound.
3. The pharmaceutical pharmaceutical composition according to claim 1 , wherein said second compound is a compound selected from the group consisting of Rituximab, Alemtuzu- mab, Trastuzumab, HuMax-CD20, HuMax-EGFr, Zamyl, Pertuzumab, antibodies against tis- sue factor, killer Ig-like receptors (KIR) and laminin-5, cdc-25, NSC 663284, flavopiridol, 7- hydroxystaurosporine, roscovitine , BIBR1532 SOT-095, TNF-related apoptosis-inducing ligand (TRAIL)/apoptosis-2 ligand (Apo-2L), antibodies that activate TRAIL receptors, IFNα and anti-sense Bcl-2, avastin, neovastat, thalidomide, PTK787, ZK222584, ZD-6474 , SU6668, PD547.632, VEGF-Trap, CEP-7055, NM-3, SU11248, QS21 , GM-CSF and CpG oligodeoxynucleotides, lipopolysaccharide and polyinosinic:polycytidylic acid, IFN-α, IFN-β, IFN-γ, IL-2, PEG-IL-2, IL-4, IL-6, IL-7, IL-12, IL-13, IL-15, IL-18, IL-21 , IL-23, IL-27, IL-28a, IL- 28b, IL-29, GM-CSF, Flt3 ligand or stem cell factor, autologous TILs, Cis-platin, tamoxifen, DTIC, Carmustine, carboplatin, Vinblastine, temozolomide, Vindesine, 5-fluorouracil, Fote- mustine, autologous LAK cells, and Gemcitabine.
4. The pharmaceutical pharmaceutical composition according to any one of the claims 1-3, wherein the ratio by mass of TF antagonist and anti-cancer compound is between about 100: 1 and about 1 : 100 (w/w)
5. The pharmaceutical pharmaceutical composition according to claim 4, wherein the weight ratio of TF antagonist and anti-cancer compound is between about 1 : 90 to about 1 : 1 (w/w).
6. The pharmaceutical composition according to any one of the claims 1-5, wherein said first agent is an inactive FVIIa polypeptide.
7. The pharmaceutical composition according to claim 6, wherein said inactive FVIIa polypeptide is native human FVIIa or a fragment thereof catalytically inactivated in the active site.
8. The pharmaceutical composition according to claim 7, wherein said inactive FVIIa polypeptide is native human FVIIa catalytically inactivated in the active site.
9. The pharmaceutical composition according to any one of the claims 1-8, wherein the FVIIa polypeptide is catalytically inactivated in the active site with a chloromethyl ketone inhibitor independently selected from the group consisting of Phe-Phe-Arg chloromethyl ketone, Phe- Phe-Arg chloromethylketone, D-Phe-Phe-Arg chloromethyl ketone, D-Phe-Phe-Arg chloromethylketone Phe-Pro-Arg chloromethylketone, D-Phe-Pro-Arg chloromethylketone, Phe- Pro-Arg chloromethylketone, D-Phe-Pro-Arg chloromethylketone, L-Glu-Gly-Arg chloromethylketone and D-Glu-Gly-Arg chloromethylketone, Dansyl-Phe-Phe-Arg chloromethyl ke- tone, Dansyl-Phe-Phe-Arg chloromethylketone, Dansyl-D-Phe-Phe-Arg chloromethyl ketone, Dansyl-D-Phe-Phe-Arg chloromethylketone, Dansyl-Phe-Pro-Arg chloromethylketone, Dan- syl-D-Phe-Pro-Arg chloromethylketone, Dansyl-Phe-Pro-Arg chloromethylketone, Dansyl-D- Phe-Pro-Arg chloromethylketone, Dansyl-L-Glu-Gly-Arg chloromethylketone and Dansyl-D- Glu-Gly-Arg chloromethylketone, Tosyl-Lys- chloromethylketone, 2,4-dichloroisocoumarin, di-isopropylfluorophosphate, phenylmethylsulphonyl fluoride.
10. The pharmaceutical composition according to any one of the claims 1-5, wherein said first agent is an antibody which immunoreacts with an epitope present on human TF.
11. The pharmaceutical composition according to claim 10, wherein said antibody inhibits the binding of human coagulation factor Vila to human TF.
12. The pharmaceutical composition according to claim 11, wherein said epitope comprises one or more of the residues Trp45, Lys46 and Tyr94.
13. The pharmaceutical composition according to any one of the claims 10-12, wherein said antibody is a monoclonal antibody.
14. The pharmaceutical composition according to any one of the claims 10-13, wherein said antibody is a recombinant antibody.
15. The pharmaceutical composition according to any one of the claims 10-14, wherein said antibody is a Fab fragment.
16. The pharmaceutical composition according to any one of the claims 10-14; wherein said antibody is a F(ab)2 fragment.
17. The pharmaceutical composition according to any one of the claims 10-14' wherein said antibody is a F(ab')2 fragment.
18. The pharmaceutical composition according to any one of the claims 10-14, wherein said antibody is a single chain Fv fragment.
19. The pharmaceutical composition according to any one of the claims 10-118, wherein said antibody has a Kd for binding to human TF within the range of 10"15- 10"8 M.
20. The pharmaceutical composition according to claim 19, wherein said antibody has a Kd for binding to human TF within the range of 10"15- 10"10 M.
21. The pharmaceutical composition according to any one of the claims 1-20, wherein said second agent is selected from the group consisting of protein ionophores, cytostatica, chemotherapeutic compound, compounds which induce apoptosis, compound containing radionuclides, antisense nucleotide molecules independent selected from the group consisting of PNAs, DNAs, RNAs and LNAs.
22. The pharmaceutical composition according to claim any one of the claims 1-20, wherein said second agent comprises a cytotoxic protein or peptide.
23. Use of a first agent, which is a TF antagonist in combination with a second agent, which is an anti-cancer compound for the manufacture of a medicament for treating a disease or disorder associated with pathophysiological TF function.
24. Use according to claim 23, wherein said first agent is according to any one of the claims 6-20.
25. Use according to any one of claims 23-24, wherein said second agent is according to any one of the claims 21-22.
26. A method for preventing or treating a disease or disorder associated with pathophysiological TF function, said method comprising administering to a mammal in need of such a treatment a therapeutically effective amount of a pharmaceutical composition, comprising (i) a first agent, which is a TF antagonist, and (ii) a second agent different from said first agent, which is an anti-cancer compound, and (iii) optionally one or more further agents different for. said first and second agent, which is an anti-cancer compound, and (iiii) a pharmaceutically acceptable carrier or excipient.
27. The method according to claim 26, wherein the pharmaceutical composition is according to any one of the claims 1 -22.
28. A method for preventing or treating a disease or disorder associated with pathophysi- ological TF function, said method comprising (i) administering to a mammal in need of such a treatment a therapeutically effective amount of a first agent, which is a TF antagonist, and (ii) administering to a mammal in need of such a treatment a therapeutically effective amount of a second agent different from the first agent, which is an anti-cancer compound, and optionally iii) administering to a mammal in need of such a treatment a therapeutically effective amount of one or more further agents different for said first and second agent, which is an anti-cancer compound.
29. The method according to claim 28, wherein said first agent is according to any one of the claims 4-18.
30. The method according to any of claim 28 or 29, wherein said second agent is according to any one of the claims 21-22.
31 The method according to any one of claims 26-30, wherein the disease or disorder associated with pathophysiological TF function are deep venous thrombosis, chronic thromboembolic diseases or disorders associated with fibrin formation, arterial thrombosis, post surgical thrombosis, coronary artery bypass graft (CABG), percutaneous transdermal coronary angioplastry (PTCA), stroke, cancer, tumour metastasis, pathological angiogenesis, thrombolysis, arteriosclerosis and restenosis following angioplastry, acute and chronic indications such as inflammation, septic chock, septicemia, hypotension, acute lung injury (ALI), Acute Respiratory Distress Syndrome (ARDS), pulmonary embolism, disseminated intravascular coagu- lation (DIC), sepsis, systemic inflammatory response syndrome (SIRS), vascular restenosis, platelet deposition, myocardial infarction, angiogenesis, or the prophylactic treatment of mammals with atherosclerotic vessels at risk for thrombosis; asthma, bronchitis, idiopathic pulmonary fibrosis, pneumonia, pulmonary edema, pulmonary obstructive disease, endotoxin induced lung damage, non cell lung cancer; inflammatory bowel disease, pancreatitis, trauma-induced shock, bronchial asthma, allergic rhinitis, rheumatoid arthritis, cystic fibrosis, stroke, acute bronchitis, chronic bronchitis, acute bronchiolitis, chronic bronchiolitis, osteoarthritis, gout, spondylarthropathris, ankylosing spondylitis, Reiter's syndrome, psoriatic arthropathy, enterapathric spondylitis, juvenile arthropathy or juvenile ankylosing spondylitis, reactive arthropathy, infectious or post-infectious arthritis, gonoccocal arthritis, tuberculous arthritis, viral arthritis, fungal arthritis, syphilitic arthritis, Lyme disease, arthritis associated with "vasculitic syndromes," polyarteritis nodosa, hypersensitivity vasculitis, Luegenec's granulomatosis, polymyalgin rheumatica, joint cell arteritis, calcium crystal deposition arthropathris, pseudo gout, non-articular rheumatism, bursitis, tenosynomitis, epicondylitis (tennis elbow), carpal tunnel syndrome, repetitive use injury (typing), miscellaneous forms of ar-thritis, neuropathic joint disease (charco and joint), hemarthrosis (hemarthrosic), Henoch- Schonlein Purpura, hypertrophic osteoarthropathy, multicentric reticulohistiocytosis, arthritis associated with certain diseases, surcoilosis, hemochromatosis, sickle cell disease and other hemo-globinopathries, hyperiipoproteineimia, hypogammaglobulinemia, hyperparathyroidism, acromegaly, familial Mediterranean fever, Behat's Disease, systemic lupus erythremato- sis, relapsing, and multiple organ failure resulting from any of the preceding pathologic processes.
32. A pharmaceutical kit comprising a first agent, which is a TF antagonist, and a second agent, which is an anti-cancer compound, and optionally a pharmaceutically acceptable carrier or excipient.
33. The kit according to claim 32, wherein said first agent is according to any one of the claims 6-20.
34. The kit according to claim 11 , wherein said second agent is according to any one of the claims 21-22.
PCT/DK2005/000098 2004-02-20 2005-02-14 Therapeutic combination comprising a tissue factor antagonist and anti-cancer compounds WO2005079766A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP05700648A EP1744734A2 (en) 2004-02-20 2005-02-14 Therapeutic combination comprising a tissue factor antagonist and anti-cancer compounds
JP2006553435A JP2007523099A (en) 2004-02-20 2005-02-14 Combination therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DKPA200400264 2004-02-20
DKPA200400264 2004-02-20

Publications (2)

Publication Number Publication Date
WO2005079766A2 true WO2005079766A2 (en) 2005-09-01
WO2005079766A3 WO2005079766A3 (en) 2005-10-13

Family

ID=34878005

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2005/000098 WO2005079766A2 (en) 2004-02-20 2005-02-14 Therapeutic combination comprising a tissue factor antagonist and anti-cancer compounds

Country Status (3)

Country Link
EP (1) EP1744734A2 (en)
JP (1) JP2007523099A (en)
WO (1) WO2005079766A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1980571A1 (en) * 2007-04-11 2008-10-15 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
WO2008125635A1 (en) * 2007-04-11 2008-10-23 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
EP2446897A1 (en) 2005-01-06 2012-05-02 Novo Nordisk A/S Anti-KIR combination treatments and methods
WO2012160448A2 (en) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
US8372810B2 (en) 2007-04-11 2013-02-12 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
WO2014151535A1 (en) * 2013-03-15 2014-09-25 Bayer Healthcare Llc Gla domains as targeting agents
US9150658B2 (en) 2008-12-09 2015-10-06 Genmab A/S Human antibodies against tissue factor and methods of use thereof
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
US9376489B2 (en) 2012-09-07 2016-06-28 Novartis Ag IL-18 binding molecules
WO2017009842A2 (en) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
CN114184792A (en) * 2021-11-17 2022-03-15 中国人民解放军总医院第二医学中心 Coronary heart disease marker IL-29 and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000004148A1 (en) * 1998-07-15 2000-01-27 Genentech, Inc. Tissue factor protein variants with increased affinity for coagulation factor fvii/fviia
WO2001001749A2 (en) * 1999-07-02 2001-01-11 Genentech, Inc. FVIIa ANTAGONISTS
WO2003029295A1 (en) * 2001-10-02 2003-04-10 Novo Nordisk A/S Human tissue factor antibodies

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000004148A1 (en) * 1998-07-15 2000-01-27 Genentech, Inc. Tissue factor protein variants with increased affinity for coagulation factor fvii/fviia
WO2001001749A2 (en) * 1999-07-02 2001-01-11 Genentech, Inc. FVIIa ANTAGONISTS
WO2003029295A1 (en) * 2001-10-02 2003-04-10 Novo Nordisk A/S Human tissue factor antibodies

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DATABASE BIOSIS [Online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 16 November 2003 (2003-11-16), NIMEIRI HALLA ET AL: "Severe retro pharyngeal bleeding in a patient with acquired Factor FVIII inhibitor." XP002339029 Database accession no. PREV200400161757 & BLOOD, vol. 102, no. 11, 16 November 2003 (2003-11-16), page 101b, 45TH ANNUAL MEETING OF THE AMERICAN SOCIETY OF HEMATOLOGY; SAN DIEGO, CA, USA; DECEMBER 06-09, 2003 ISSN: 0006-4971 *
KIRCHHOFER D ET AL: "EPITOPE LOCATION ON TISSUE FACTOR DETERMINES THE ANTICOAGULANT POTENCY OF MONOCLONAL ANTI-TISSUE FACTOR ANTIBODIES" THROMBOSIS AND HAEMOSTASIS, STUTTGART, DE, vol. 84, no. 6, December 2000 (2000-12), pages 1072-1081, XP001041490 ISSN: 0340-6245 *
MILLER DEBRA L ET AL: "Extrinsic coagulation blockade attenuates lung injury and proinflammatory cytokine release after intratracheal lipopolysaccharide" AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY, vol. 26, no. 6, June 2002 (2002-06), pages 650-658, XP002337200 ISSN: 1044-1549 *
PERSSON E ET AL: "Substitution of aspartic acid for methionine-306 in factor VIIa abolishes the allosteric linkage between the active site and the binding interface with tissue factor" BIOCHEMISTRY, AMERICAN CHEMICAL SOCIETY. EASTON, PA, US, vol. 40, no. 11, 20 March 2001 (2001-03-20), pages 3251-3256, XP002311730 ISSN: 0006-2960 *
ZHIWEI HU ET AL: "Targeting tissue factor on tumor vascular endothelial cells and tumor cells for immunotherapy in mouse models of prostatic cancer" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, vol. 98, no. 21, 2 October 2002 (2002-10-02), pages 12180-12185, XP002262739 ISSN: 0027-8424 cited in the application *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2446897A1 (en) 2005-01-06 2012-05-02 Novo Nordisk A/S Anti-KIR combination treatments and methods
EP3072522A1 (en) 2005-01-06 2016-09-28 Novo Nordisk A/S Anti-kir combination treatments and methods
WO2008125635A1 (en) * 2007-04-11 2008-10-23 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
JP2010523628A (en) * 2007-04-11 2010-07-15 ジェネ シグナル インターナショナル エスエー Antitumor agents, pharmaceuticals, compositions and uses thereof
AU2008237952B2 (en) * 2007-04-11 2013-02-07 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
US8372810B2 (en) 2007-04-11 2013-02-12 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
EP1980571A1 (en) * 2007-04-11 2008-10-15 Gene Signal International Sa Anti-tumor drug, medicament, composition, and use thereof
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
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
WO2012160448A2 (en) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
US9376489B2 (en) 2012-09-07 2016-06-28 Novartis Ag IL-18 binding molecules
US10081677B2 (en) 2012-09-07 2018-09-25 Novartis Ag IL-18 binding molecules
US11111293B2 (en) 2012-09-07 2021-09-07 Novartis Ag IL-18 binding molecules
WO2014151535A1 (en) * 2013-03-15 2014-09-25 Bayer Healthcare Llc Gla domains as targeting agents
RU2711091C2 (en) * 2013-03-15 2020-01-15 Гладиатор Байосайенсиз, Инк. Gla domains as targeting agents
US10894075B2 (en) 2013-03-15 2021-01-19 GLAdiator Biosciences, Inc. Gla domains as targeting agents
WO2017009842A2 (en) 2015-07-16 2017-01-19 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
EP3744340A2 (en) 2015-07-16 2020-12-02 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
EP3943098A2 (en) 2015-07-16 2022-01-26 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
CN114184792A (en) * 2021-11-17 2022-03-15 中国人民解放军总医院第二医学中心 Coronary heart disease marker IL-29 and application thereof

Also Published As

Publication number Publication date
JP2007523099A (en) 2007-08-16
WO2005079766A3 (en) 2005-10-13
EP1744734A2 (en) 2007-01-24

Similar Documents

Publication Publication Date Title
EP1744734A2 (en) Therapeutic combination comprising a tissue factor antagonist and anti-cancer compounds
AU2006332713B2 (en) Metalloproteinase binding proteins
JP4361728B2 (en) Human coagulation factor VII mutant
JP2018076333A (en) Targeting of chondroitin sulfate glycans
RU2412199C2 (en) Versions of il-21
JP2000513720A (en) Modified factor VII
US20210246212A1 (en) Anti-gpiib/iiia antibodies and uses thereof
CZ2004454A3 (en) Human tissue factor antibodies
US20060018831A1 (en) TF binding agent and use thereof
WO2018156649A1 (en) Compositions of t cell modulator (tcm) molecules and uses thereof
US20060024730A1 (en) TF binding compounds
US20040072755A1 (en) TF antagonist
WO2004007557A2 (en) Tf antagonist
WO2004006962A2 (en) A tissue factor binding immunoconjugate comprising factor viia
TWI673063B (en) Treatment of bleeding disorders caused by anti-protein C antibodies
US20040198660A1 (en) Tissue factor antagonist and protein C polypeptide compositions
WO2008117179A2 (en) Polynucleotides and polypeptides of human factor vii gene, snps
US20060034846A1 (en) Use of TF antagonists
US20230322922A1 (en) Her-2 targeted bispecific compositions and methods for making and using the same
US20020107361A1 (en) Novel metalloproteases having thrombospondin domains and nucleic acid compositions encoding the same
WO2003076461A2 (en) Dimeric tf antagonist comprising two factor vii polypeptides
US20060160741A1 (en) Dimeric TF antagonists
Nopmongkol Identification of fibulin-1 as a human bone marrow stromal (HS-5) derived factor that induces prostate cancer cell death
KR20100087532A (en) Anti-inflammatory pharmaceutical composition

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 BW 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 NA 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 US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA 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 IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005700648

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006553435

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

WWP Wipo information: published in national office

Ref document number: 2005700648

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2005700648

Country of ref document: EP