US20110159006A1 - Use of anti-factor xi antibodies for prevention of thrombus formation - Google Patents

Use of anti-factor xi antibodies for prevention of thrombus formation Download PDF

Info

Publication number
US20110159006A1
US20110159006A1 US13/000,284 US200913000284A US2011159006A1 US 20110159006 A1 US20110159006 A1 US 20110159006A1 US 200913000284 A US200913000284 A US 200913000284A US 2011159006 A1 US2011159006 A1 US 2011159006A1
Authority
US
United States
Prior art keywords
factor
binding
binding molecule
antibody
antibodies
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/000,284
Other languages
English (en)
Inventor
Erik Hack
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40933725&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110159006(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US13/000,284 priority Critical patent/US20110159006A1/en
Publication of US20110159006A1 publication Critical patent/US20110159006A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/36Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention is in the field of haematology, in particular the field of coagulation.
  • the invention relates to methods for inhibiting the formation of blood clots using binding molecules that specifically bind and inhibit the activation and/or activity of factor XI.
  • the antigen binding molecule may or may not be specific for the activated conformation of factor XI and is preferably used in methods for reducing or preventing thrombus formation on synthetic grafts, atherosclerotic plaques or in other pathological thrombotic and thrombo-embolic processes.
  • Coagulation consists of a humoral and a cellular response.
  • the former leads to the conversion of soluble fibrinogen into insoluble fibrin, the latter consists of activation of platelets leading to a platelet plug.
  • Platelet plugs, fibrin threads and included red blood cells together constitute a blood clot.
  • a key molecule in clot formation is thrombin. This protease converts soluble fibrinogen into the insoluble fibrin, it activates platelets, and converts a number of other factors including factors XI, VIII and V, into active species.
  • Pathological thrombosis refers to clot formation that is not part of a normal hemostatic process and that may result in disease symptoms.
  • thrombosis on an atherosclerotic plaque in a coronary artery may result in acute myocardial infarction and can be considered as a type of pathological thrombosis.
  • Deep venous thrombosis and thrombosis on vascular grafts are other examples of pathological thrombosis.
  • Factor XI is not a target for current anticoagulants. This is mainly due to the fact that factor XI deficiency, in contrast to a deficiency of factor VIII or IX, does not result in a severe bleeding tendency. As a matter of fact many factor XI deficient persons never experience a severe bleeding episode. Only two studies have been performed with antibodies, in both cases polyclonal, against factor XI that inhibit the function of factor XI in vivo, either by inhibiting the activity of the molecule or by preventing its activation. In a first study (Minnema et al., 1998, J Clin Invest.
  • polyclonal antibodies against rabbit factor XI were used to evaluate the effect of factor XI blockade on clot formation in vivo in an experimental thrombosis model in rabbits. Incorporation of anti-factor XI antibodies in jugular vein thrombi resulted in an almost twofold increase in endogenous thrombolysis compared with a control antibody. A similar effect was observed when the anti-factor XI antibody was administered systemically.
  • antithrombotic effect of the polyclonal antibodies against factor XI was found to be comparable with that of heparin at doses that significantly prolonged the partial thromboplastin time, prothrombin time, and bleeding time, whereas anti-FXI antibodies only affected the partial thromboplastin time but not prothrombin time and bleeding time.
  • the present invention relates to means and methods for anti-coagulant therapy that are based on the inhibition of factor XI.
  • factor XI is a preferred target for inhibition as this leaves coagulation via the “basal” pathway and the FIX/VIII amplification loop intact, which is sufficient for most hemostatic conditions.
  • factor XI inhibitors have less risk for bleeding side effects.
  • factor XI deficiency or factor XI inhibition had no effect on the bleeding time, in contrast to high dose heparin which strongly prolonged bleeding time.
  • factor XI is an attractive target for anti-coagulant therapy.
  • the present invention relates to a binding molecule that specifically binds to factor XI and that inhibits the functional activity of factor XI.
  • Factor XI is herein understood as the mammalian plasma coagulation factor XI.
  • a binding molecule of the invention specifically binds to and inhibits the functional activity of at least human factor XI.
  • Plasma coagulation factor XI is a glycoprotein present in human plasma at a concentration of 25-30 nM as a zymogen that when converted by limited proteolysis to an active serine protease, participates in the contact phase of blood coagulation.
  • the sequence of the human factor XI gene has been identified and the deduced amino acid sequence is depicted in SEQ ID NO: 1.
  • the gene for human factor XI is 23 kilobases (kb) in length and consists of 15 exons (I-XV) and 14 introns. Exon I encodes the 5′-untranslated region, and exon II encodes a signal peptide. The next eight exons (III-X) encode four tandem repeat sequences of 90 or 91 amino acids (Apple domains) that are present in the amino-terminal region of the mature protein.
  • the carboxyl-terminal region of the protein which contains the catalytic serine protease domain, is encoded by five exons (XI-XV) that are interrupted by four introns.
  • Factor XI is a zymogen, which is unique among the other clotting factors in that it exists as a homo-dimer consisting of two identical subunits of 80 kDa connected by a single disulfide bond.
  • factor XIa Conversion of zymogen factor XI to the active form, factor XIa, is accomplished by a single cleavage at Arg 369 -Ile 370 (SEQ ID NO: 1), upon which cleavage each subunit is converted into a 35 kDa light chain and a 50 kDa heavy chain linked by disulfide bonds.
  • the light chain contains a catalytic site.
  • Each heavy chain is composed of four 90-91 amino acid repeats called apple domains (A1-A4) with A1 located at the aminoterminal end of the heavy chain.
  • apple domains A1 located at the aminoterminal end of the heavy chain.
  • the A1 domain contains the binding site for high molecular weight kininogen and thrombin.
  • the function of the A2 domain is not clear, initially it was reported to contain an interaction site with factor IX, but later it was shown that this site was located in the A3 domain.
  • the A3 domain also contains a binding site for heparin, and for platelets.
  • the binding sites for factor IX and platelets are close to each other and it has been postulated by Gailani et al. (D Gailani et al., Blood 2001; 97:3117-3122) that this is the reason for the dimeric structure: due to its dimeric structure factor XI can bind via the A3 domain of one subunit to platelets and use its other A3 domain to interact with factor IX.
  • factor XI can target activation of coagulation to the platelet surface.
  • the A4 domain of factor XI is involved in dimer formation, contains a free cystein of unknown significance, and an interaction site with factor XIIa.
  • Factor XI can be activated by at least three coagulation proteases, factor XIIa, factor XIa (autoactivation) and thrombin.
  • a binding molecule of the invention that can bind to, that has affinity for and/or that has specificity for a factor XI target molecule (or an epitope on the factor XI target molecule) may be said to be “against” or “directed against” said target molecule or antigen.
  • the term “specificity” refers to the number of different types of epitopes or antigenic targets on factor XI to which a particular (antigen-) binding molecule can bind. The specificity of an antigen-binding molecule can be determined based on affinity and/or avidity.
  • the affinity represented by the equilibrium constant for the dissociation of an antigen with an antigen-binding protein (K D ), is a measure for the binding strength between an antigenic determinant and an antigen-binding site on the antigen-binding protein.
  • the affinity can also be expressed as the affinity constant (K A ), which is 1/K D .
  • Affinity can be determined in a manner known per se, depending on the specific combination of antigen-binding protein and antigen of interest.
  • Avidity is herein understood to refer to the strength of binding of a target molecule with multiple binding sites by a larger complex of binding agents, i.e. the strength of binding of multivalent binding. Avidity is related to both the affinity between an antigenic determinant and its antigen-binding site on the antigen-binding molecule and the number of binding sites present on the antigen-binding molecule. Affinity, on the other hand refers to simple monovalent receptor ligand systems.
  • factor XI-binding molecules of the invention will bind the target molecule with a dissociation constant (K 0 ) of about 10 ⁇ 7 to 10 ⁇ 12 M or less, and preferably 10 ⁇ 8 to 10 ⁇ 12 M or less, and/or with a binding affinity of at least 10 ⁇ 7 M, preferably at least 10 ⁇ 8 M, more preferably at least 10 ⁇ 9 M, such as at least 10 ⁇ 10 , 10 ⁇ 11 , 10 ⁇ 12 M or more.
  • K D value greater than 10 ⁇ 4 M i.e. less than 100 ⁇ M is generally considered to indicate non-specific binding.
  • a binding molecule of the invention will bind to factor XI with an affinity less than 50, 10 or 5 nM, more preferably less than 1 nM, such as less than 500, 200, 100, 50, 10 or 5 pM.
  • Specific binding of a binding molecule to factor XI can be determined in any suitable manner known per se, including, for example, Scatchard analysis and/or competitive binding assays, such as radioimmunoassays (RIA), enzyme immunoassays (EIA) and sandwich competition assays, and the different variants thereof known per se in the art.
  • binding molecules of the invention inhibit at least one of the functional activities of factor XI and the activation of factor XI.
  • binding molecules of the invention inhibit the functional activity of factor XI and/or inhibit the activation of factor XI, independently of how factor XI is activated.
  • a preferred binding molecule of the invention e.g. an antibody or antibody fragment, has only one binding site that binds and inhibits factor XI activation or factor XI activity.
  • the binding molecule inhibits the functional activity of factor XI by preventing it to become activated.
  • the binding molecule inhibits the activation of factor XI by binding to or near to the peptidyl bond Arg 369 -Ile 370 , and preventing the cleavage of this bond by thrombin, factor XIIa, factor XIa or any other protease.
  • the binding molecule upon binding of the binding molecule to factor XI, there is a reduction in the activation of factor XI by factor XIIa, thrombin or factor XIa of at least 60, 70, 80, 90, 95, 99%. More preferably no activation of factor XI by factor XIIa, thrombin or factor XIa, is detectable upon binding of the binding molecule to factor XI.
  • the amount of reduction in the activation of factor XI may be determined in the various assays as described in the Examples herein by comparing activation of factor XI in the presence of the binding molecule with activation in the absence of the molecule.
  • the binding molecule inhibits the activation of factor XI by binding to the A1 domain of factor XI thereby preventing the interaction of factor XI with high molecular weight kininogen and/or thrombin (BN Bouma et al., Curr Opin Hematol 2000, 7:266-272).
  • factor XIIa a reduction in the activation of factor XI by factor XIIa, thrombin or factor XIa of at least 60, 70, 80, 90, 95, 99%. More preferably no activation of factor XI by thrombin is detectable upon binding of the binding molecule to factor XI.
  • the amount of reduction in the activation of factor XI may be determined in the various assays as described in the Examples herein by comparing activation of factor XI in the presence of the binding molecule with activation in the absence of the molecule.
  • the binding molecule inhibits the activity of factor XI by binding to or near to the active site located in the light chain region of the molecule.
  • the binding molecule inhibits the activity of factor XI by binding to the A2 or A3 domain of factor XI, that are involved in the interaction with factor IX, heparin and or platelets (BN Bouma et al., Curr Opin Hematol 2000, 7:266-272).
  • the binding molecule inhibits the activation of factor XI by binding to or near to sites that are involved in the interaction with its substrate factor IX.
  • the binding molecule preferentially binds to the activated form of factor XI and inhibits the activation of factor XI by binding to or near to sites that are involved in the interaction with its substrate factor IX.
  • the binding molecule preferentially binds to the activated form of factor XI and inhibits the activation of factor XI by binding to the A1 domain of factor XI thereby preventing the interaction of factor XI with high molecular weight kininogen and/or thrombin.
  • the binding molecule preferentially binds to the activated form of factor XI and inhibits the activity of factor XI by binding to or near to sites that are involved in the interaction with its substrate factor IX.
  • the binding molecule preferentially binds to the activated form of factor XI and inhibits the activity of factor XI by binding to or near to the active site located in the light chain region of the molecule.
  • the binding molecule preferentially binds to the activated form of factor XI and inhibits the activity of factor XI by binding to the A2 or A3 domain of factor XI, that are involved in the interaction with factor IX, heparin and or platelets.
  • the binding molecules of the present invention are thus characterised by their ability to prevent activation and/or inhibit the activity of factor XI and/or to inhibit activation of factor IX.
  • Said inhibitors can be selected by the assessment of their effect in various assays as described in the Examples herein.
  • factor XI-binding molecules can be selected by the assessment of their effect on the clotting activity of the coagulation system as determined with an activated partial thromboplastin time (APTT) in human plasma.
  • APTT activated partial thromboplastin time
  • the functional properties of factor XI-binding molecules of the invention may be tested by adding these to fresh human plasma, followed by measurement of the APTT in a regular clotting assay. In case of an inhibiting antibody, a prolongation of the APTT will be observed.
  • normal plasma normal APTT
  • factor XI-deficient plasma prolonged APTT
  • factor XI-binding molecules on the function of the factor XI molecule can also be tested using chromogenic substrates.
  • Chromogenic substrates consist of small peptides coupled to p-nitroanilide (pNA). Hydrolysis of the substrate releases pNA which can be measured with a spectrophotometer. Specificity of the substrate for certain proteases is dependent on the precise sequence of the peptide linked to pNA.
  • the substrate S2366 Pyr-Glu-Pro-Arg-pNA-2H2O; Chromogenix, Molndal, Sweden
  • Measurement of factor XIa activity with this substrate can be done using the method described by Minnema et al.
  • Factor XI-binding molecules that inhibit the catalytic center of factor XIa can be identified by their inhibiting effect on the chromogenic activity of factor XIa in such a chromogenic activity.
  • purified factor XIIa can be used to activate factor XI, which then can be monitored by measuring its chromogenic activity.
  • Factor XI-binding molecules that inhibit the activation of factor XI by factor XIIa will manifest themselves by decreasing the amount of factor XIa generated in this system.
  • a factor XI-binding molecule of the invention that inhibits the functional activity or activation of factor XI preferably is a molecule that produces at least 90% inhibition of factor XI activity at a concentration of about 50-80 nM in an activated partial thromboplastin time (APTT) assay. More preferably the molecule produces at least 95% inhibition of factor XI activity at a concentration of about 50-80 nM in an activated partial thromboplastin time (APTT) assay. Most preferably the molecule produces at least 99% inhibition of factor XI activity at a concentration of about 50-80 nM in an activated partial thromboplastin time (APTT) assay.
  • APTT activated partial thromboplastin time
  • a factor XI-binding molecule of the invention that inhibits the functional activity or activation of factor XI is a molecule that produces at least 90% inhibition of factor XI activity at a concentration of about 20-50 nM in an activated partial thromboplastin time (APTT) assay. More preferably the molecule produces at least 95% inhibition of factor XI activity at a concentration of about 20-50 nM in an activated partial thromboplastin time (APTT) assay. Most preferably the molecule produces at least 99% inhibition of factor XI activity at a concentration of about 20-50 nM in an activated partial thromboplastin time (APTT) assay.
  • APTT activated partial thromboplastin time
  • activator refers to “a molecule capable of activating factor XI in such a way that this subsequently activates factor IX, which in presence of its cofactor FVIII in its turn activates factor X and the rest of the coagulation cascade”.
  • Activator molecules include thrombin, factor XIIIa and factor XIa.
  • a factor XI-binding molecule of the invention is an antibody or a factor XI-binding fragment of an antibody.
  • binding molecule thus encompasses, but is not limited to, an antibody and fragments thereof, a unibody, a diabody, a triabody, a tetravalent or other multivalent antibody specifically binding factor XI and inhibiting the functional activity of factor XI.
  • antibody refers to polyclonal antibodies, monoclonal antibodies, antibodies which are derived from a phage library, humanised antibodies, human antibodies, synthetic antibodies, chimeric antibodies, single domain antigen binding proteins and antibody fragments such as, but not limited to single-chain Fv's.
  • antibodies made in other animal species such as camelid antibodies or fragments thereof (“Nanobodies”) fall within the scope of this application.
  • molecules with antibody-like binding properties such as Designed Repeat Proteins like DARPins (Designed Ankyrin Repeat Proteins) are within the scope of this application.
  • the factor XI-binding molecule of the invention is a component that specifically binds to the target molecule with a desired binding affinity (as herein defined).
  • the factor XI-binding protein of the invention preferably is a mono-specific antigen-binding protein.
  • a composition comprising a mono-specific antigen-binding protein is understood to mean a composition having a homogeneous population of the factor XI-binding protein. It follows that the mono-specific factor XI-binding protein is specific for a single epitope on a factor XI monomer. It is however expressly included in the invention that the compositions of the invention may comprise more than one type of mono-specific factor XI-binding protein, each consisting of a homogeneous population.
  • a composition of the invention will not comprise more than 4, 6, 8, 10 or 20 different mono-specific factor XI-binding proteins.
  • the factor XI-binding protein will usually be an antibody or fragment thereof, in which case the mono-specific factor XI-binding protein will thus be a monoclonal antibody or a fragment thereof, which may be obtained from a cloned cell-line (e.g. hybridoma) or expressed from a cloned coding sequence.
  • the term “monoclonal antibody” is thus not intended to be limited by the manner in which it is made.
  • the term mono-specific factor XI-binding protein as used herein thus excludes polyclonal antibodies and antisera.
  • any construct of an antibody or a fragment is also a subject of current invention.
  • the term “construct” relates to diabodies, triabodies, tetravalent antibodies, pepta- or hexabodies, and the like, that are derived from an anti-human factor XI antibody according to the present invention.
  • Said multivalent antibodies comprising at least one hypervariable domain from an anti-factor XI antibody according to the present invention can be mono-, bi- or multispecific.
  • a preferred antibody or antibody fragment has only one binding site that binds and inhibits factor XI activation or factor XI activity.
  • human antibody as used herein is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • the human antibodies may include amino acids residues not encoded by human germline immunoglobulin sequences, e.g. mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo.
  • humanised antibody means that at least a portion of the framework regions of an immunoglobulin or engineered antibody construct is derived from human immunoglobulin sequences. It should be clear that any method to humanise antibodies or antibody constructs, as for example by variable domain resurfacing (Roguska et al., 1994) or CDR grafting or reshaping (Hurle et al., 1994), can be used.
  • chimeric antibody refers to an engineered antibody construct comprising of variable domains of one species (such as mouse, rat, goat, sheep, cow, lama or camel variable domains), which may be humanised or not, and constant domains of another species (such as non-human primate or human constant domains) (for review see Hurle et al., 1994, supra). It should be clear that any method known in the art to develop chimeric antibodies or antibody constructs can be used.
  • the antibody is an intact murine IgG1, an intact human IgG1 mutated in the constant region to reduce or prevent complement activation or Fc receptor interactions, or an intact human IgG4.
  • the antibody or antibody fragment is a monomeric IgM antibody subunit.
  • the antibody or antibody fragment does not activate the classical or lectin pathways of complement, and/or does not interact with Fc-receptors.
  • the binding molecule is a binding-domain immunoglobulin fusion protein comprising (i) a binding domain polypeptide in the form of a heavy chain variable region or a light chain variable region that is fused to an immunoglobulin hinge region polypeptide, (ii) an immunoglobulin heavy chain CH2 constant region fused to the hinge region, and (iii) an immunoglobulin heavy chain CH3 constant region fused to the CH2 constant region.
  • fragment refers to Fab, F(ab′) 2 , Fv, scFv, Fd (consisting of the V H and C H1 domains), dAb (consisting of a V H domain), a complementarity determining region (CDR), e.g. V H CDR3, and other fragments which retain the antigen binding function and specificity of the parent antibody.
  • CDR complementarity determining region
  • single chain Fv also termed scFv
  • scFv refers to engineered antibodies prepared by isolating the binding domains (both heavy and light chains) of a binding antibody, and supplying a linking moiety which permits preservation of the binding function. This forms, in essence, a radically abbreviated antibody, having only that part of the hyper-variable domain necessary for binding the antigen. Determination and construction of single chain antibodies are described in e.g. U.S. Pat. No. 4,946,778 to Ladner et al.
  • single domain antigen binding protein refers to antibodies or fragments thereof that are derived from antibodies naturally devoid of light chains.
  • Antibodies naturally devoid of light chains may be obtained e.g. by immunisation of camelids (e.g. llama's) or sharks (see further below). These antibodies comprise heavy chains only and are devoid of light chains.
  • the advantage of use of such single domain heavy chain antibodies is that they are exceptionally stable even at higher temperatures, small and are easily produced in microbial host organisms such as Saccharomyces cerevisiae .
  • a factor XI-binding protein of the invention preferably comprises an immunoglobulin-derived variable domain that comprises a complete antigen-binding site for an epitope on a target molecule in a single polypeptide chain.
  • Such factor XI-binding proteins specifically include but are not limited to:
  • variable domains of the antibodies defined in 1) usually referred to as VHH domains (see WO2006/040153);
  • Monoclonal antibodies of the present invention can be obtained by isolating immune cells from an animal immunised with human factor XI or with activated human factor XI (factor XIa), or with parts of these molecules, and immortalisation of these cells to yield antibody secreting cell lines such as hybridomas.
  • Cell lines that produce the desired antibodies can be identified by screening culture supernatants for the presence of antibody activity, and by establishment of the effect of the selected antibody on the functional activity of factor XI.
  • Human factor XI, factor XIa, fragments thereof and/or synthetic peptides comprising factor XI amino acid sequences isolated according to a variety of purification methods may be used to immunise an appropriate host animal.
  • immunization protocols may be employed, and may consist of intravenous, subcutaneous, or intraperitoneal immunization, followed by one or more boosts.
  • a suitable adjuvant is Freund's adjuvant.
  • the precise schedule of administration of the human factor XI, factor XIa, or fragments thereof to the host animal in general is not well defined.
  • the choice of the immunization procedure is more dependent on host animal antibody responses to the administered factor XI, as measured by a suitable assay (vide supra).
  • a suitable immunization procedure is hyperimmunization with human factor XI, or factor XIa, or fragments thereof at a concentration that, dependent on the host animal, may be in the range of 10 to 500 microgram, mixed with Freund's complete adjuvant. The mix is injected subcutaneously. Injections are repeated 2 to 5 times using the same factor XI preparation mixed with Freund's incomplete adjuvant. Blood samples are taken from the animal 1 week after the 3 rd , 4 th , 5 th injection, and screened for the presence of antibodies against factor XI. In a scheme as indicated above, the length of the interval between the injections can be varied. Alternative adjuvants can also be used.
  • lymphocytes human or murine or other
  • human lymphocytes those obtained from patients with antibodies against human factor XI can be used, for example from persons deficient for factor XI who have developed antibody responses against administered exogenous factor XI (0 Salomon et al., Blood 2003; 101:4783-4788).
  • An alternative approach for immunization comprises the use of synthetic peptides that mimic the sequence of functional sites of factor XI, such as individual apple domains.
  • the methods of making antibodies against peptides are well-known in the art and generally require coupling of the peptides to a suitable carrier molecule, for example bovine serum albumin or keyhole limpet hemocyanin.
  • a suitable carrier molecule for example bovine serum albumin or keyhole limpet hemocyanin.
  • the peptides can be made according to procedures well known in the art. The procedure also may use commercially available peptide synthesiser machines.
  • hybridoma cell line that produces the antibody may be used as a source of DNA or mRNA encoding the desired antibody, which may be isolated and transferred to cells by known genetic techniques to produce genetically engineered antibody.
  • the initial screening step of culture supernatants of hybridomas obtained by fusion of lymphocytes of mice immunised with factor XI, factor XIa, parts thereof, or with factor XI peptides, with an appropriate fusion partner is preferably done by an enzyme-linked immunosorbent assay (ELISA) or a radioimmunoassay (RIA). Both assays are known to those skilled in the art, and consist of coupling of human factor XI or factor XIa to a solid-phase matrix, and assaying for antibody binding to factor XI or factor XIa by a second, labelled antibody. In case peptides are used for immunization, peptides coupled to a solid-phase matrix, also can be used in these assays.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • the preferred assay is an enzyme-linked sorbent assay in which purified human factor XI or factor XIa (JF Tait et al., J Biol Chem 1987; 262:11651-11656) is used for coating, and which is further carried out according to the procedure described by (Smeenk R T J, et al. 1987, Arthr Rheum 30: 607), which comprises an incubation step with the hybridoma supernatants and an incubation step with the labeled anti-mouse immunoglobulin reagent.
  • an alternative screening procedure may be used to assess whether the selected antibody may bind factor XI or factor XIa in solution. This is achieved by a method in which an anti-immunoglobulin agent is coupled to a solid-phase matrix, and bound antibodies against factor XI or factor XIa are specifically detected using labelled purified factor XI or factor XIa.
  • a suitable radioimmunoassay procedure for screening anti-factor XI or anti-factor XIa antibodies may be that described for detection of anti-C3 antibodies by (Hack et al., J Immunol. 1988; 141:1602-9).
  • solutions containing human factor XI or factor XIa may be incubated with the antibody coupled to a solid-phase matrix via an anti-immunoglobulin reagent. The matrix is then washed, and bound factor XI or factor XIa is dissociated from it. The eluted factor XI or factor XIa is then detected by SDS-PAGE followed by Western blotting.
  • Monoclonal antibodies can also be produced in various other ways with techniques well understood by those having ordinary skill in the art. Details of these techniques are described in (Antibodies: A Laboratory Manual, Harlow et al., Cold Spring Harbor Publications, p. 726, 1988), or are described by (Campbell, A. M. “Monoclonal Antibody Technology Techniques in Biochemistry and Molecular Biology,” Elsevier Science Publishers, Amsterdam, The Netherlands, 1984) or by (St. Groth et al., J. Immunol. Methods 35:1-21, 1980). These other techniques include, but are not limited to techniques for recombinant production of monoclonal antibodies.
  • Monoclonal antibodies of any mammalian species, including humans, can be used in this invention. Accordingly, the antibodies according to this embodiment may be human monoclonal antibodies. Such human monoclonal antibodies may be prepared, for instance, by the generation of hybridomas, derived from immunised transgenic animals, containing large sections of the human immunoglobulin (Ig) gene loci in the germline, integrated by the yeast artificial chromosomal (YAC) technology (Mendez et al., 1997).
  • YAC yeast artificial chromosomal
  • WO 04/035607 Genmab
  • WO 04/043989 Medarex
  • Further similar methods have been described in WO 03/017935 (Genmab), WO 02/100348 (Genmab), WO 02/064634 (Medarex) and WO 03/040169 (Medarex).
  • Fully human antibodies which recognise a selected epitope can also be generated using a technique referred to as “guided selection”.
  • a selected non-human monoclonal antibody e.g. a mouse antibody, is used to guide the selection of a completely human antibody recognising the same epitope (Jespers et al., 1994).
  • recombinant antibodies such as chimeric and humanised monoclonal antibodies comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region, see e.g. U.S. Pat. No. 4,816,567; and U.S. Pat. No. 4,816,397.
  • Humanised antibodies are antibody molecules from non-human species having one or more complementarily determining regions from the non-human species and a framework region from a human immunoglobulin molecule, see e.g.
  • Such chimeric and humanised monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in WO 87/02671; EP 184 187; EP 171 496; EP 173,494; WO 86/01533; U.S. Pat. No.
  • the present invention also comprises antibodies in which one or more alterations have been made in the Fc region in order to change functional or pharma-cokinetic properties of the antibodies.
  • Such alterations may result in a decrease or increase of C1q binding and CDC (complement dependent cytotoxicity) or of Fc ⁇ R binding and antibody-dependent cellular cytotoxicity (ADCC).
  • Substitutions can for example be made in one or more of the amino acid positions 234, 235, 236, 237, 297, 318, 320, and 322 of the heavy chain constant region, thereby causing an alteration in an effector function while retaining binding to antigen as compared with the unmodified antibody, cf.
  • the antibodies of the present invention can be prepared by using a technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Kohler and Milstein (Kohler et al., 1975).
  • Human antibodies of the invention also can be produced in a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as is well known in the art, see e.g. (Morrison, 1985).
  • DNAs encoding partial or full-length light and heavy chains can be obtained by standard molecular biology techniques (e.g., PCR amplification, site directed mutagenesis) and can be inserted into expression vectors such that the genes are operatively linked to transcriptional and translational control sequences.
  • operatively linked is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene.
  • the expression vector and expression control sequences are chosen to be compatible with the expression host cell used.
  • the antibody light chain gene and the antibody heavy chain gene can be inserted into separate vectors or, more typically, both genes are inserted into the same expression vector.
  • the antibody genes are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • the light and heavy chain variable regions of the antibodies described herein can be used to create full-length antibody genes of any antibody isotype by inserting them into expression vectors already encoding heavy chain constant and light chain constant regions of the desired isotype such that the V H segment is operatively linked to the C H segment(s) within the vector and the V L segment is operatively linked to the C L segment within the vector.
  • the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell.
  • the antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene.
  • the signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).
  • the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the antibody chain genes in a host cell.
  • the term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes.
  • Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology. Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • Preferred regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • nonviral regulatory sequences may be used, such as the ubiquitin promoter or beta-globin promoter.
  • the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes.
  • the selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. No. 4,399,216, U.S. Pat. No. 4,634,665 and U.S. Pat. No. 5,179,017, all by Axel et al.).
  • the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced.
  • Preferred selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • DHFR dihydrofolate reductase
  • the expression vector(s) encoding the heavy and light chains is transfected into a host cell by standard techniques.
  • the various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection, lipofectin transfection and the like.
  • the antibodies are expressed in eukaryotic cells, such as mammalian host cells.
  • eukaryotic cells such as mammalian host cells.
  • Preferred mammalian host cells for expressing the recombinant antibodies of the invention include CHO cells (including dhfr-CHO cells, described in (Urlaub et al., 1980), used with a DHFR selectable marker, e.g., as described in (R. J. Kaufman et al., 1982), NS/0 myeloma cells, COS cells, HEK293 cells and SP2.0 cells.
  • GS glucose synthetase gene expression system
  • WO 87/04462 WO 89/01036
  • EP 338 841 a preferred expression system
  • the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown.
  • Antibodies can be recovered from the culture medium using standard protein purification methods.
  • the cloned antibody genes can be expressed in other expression systems, including prokaryotic cells, such as microorganisms, e.g. E. coli for the production of scFv antibodies, algi, as well as insect cells.
  • prokaryotic cells such as microorganisms, e.g. E. coli for the production of scFv antibodies, algi, as well as insect cells.
  • the antibodies can be produced in transgenic non-human animals, such as in milk from sheep and rabbits or eggs from hens, or in transgenic plants. See e.g. (Verma, et al., 1998); (Pollock et al., 1999); and (Fischer et al., 1999).
  • polyclonal, monoclonal, or recombinant it may be purified by standard techniques well known in the art. Most of these techniques use affinity chromatography, often in combination with a precipitation step.
  • Cell lines that secrete antibody against human factor XI or factor XIa can be identified by assaying culture supernatants, ascitic fluid, etc., for the presence of antibody.
  • the preferred screening procedure comprises two sequential steps, the first being identification of hybridomas that secrete mAb against human factor XI or factor XIa, the second being determination of the ability of the mAb to inhibit the activation or the functional activity of factor XI and to prevent or reduce activation of factor IX and the rest of the coagulation system.
  • Factor XI-binding diabodies, triabodies, tetravalent antibodies of the invention can be produced by the following methods: 1) chemical linkage of anti-factor XI antibodies of the present invention or univalent fragments thereof following a method as described by e.g. (Fanger M W, Morganelli P M, Guyre P M. Bispecific antibodies. Crit Rev Immunol. 1992; 12:101-24); 2) genetically engineering of non-covalently-linked diabodies as described by e.g. (Holliger P, Prospero T, Winter G. “Diabodies”: small bivalent and bispecific antibody fragments. Proc Natl Acad Sci USA. 1993; 90:6444-8) and tetravalent antibodies as described by e.g.
  • the invention in a second aspect, relates to methods of preventing or treating a disease, disorder and/or condition that is mediated by factor XI activation and/or wherein inhibition of factor XI has a beneficial effect.
  • the methods preferably comprise the step of administering to a subject a binding molecule as disclosed herein above, in an amount effective to treat or prevent the disease, disorder and/or condition.
  • the invention also relates to the use of a binding molecule as disclosed herein above for the preparation of a medicament for preventing or treating a disease, disorder and/or condition that is mediated by factor XI activation and/or wherein inhibition of factor XI has a beneficial effect.
  • the invention alternatively relates to a binding molecule as disclosed herein above, for use in preventing or treating a disease, disorder and/or condition that is mediated by factor XI activation and/or wherein inhibition of factor XI has a beneficial effect.
  • a disease, disorder and/or condition that is mediated by factor XI activation and/or wherein inhibition of factor XI has a beneficial effect is disease, disorder and/or condition in which coagulation is involved, such as e.g. a thromboembolic or inflammatory disease mediated by coagulation activation via factor XI.
  • the binding molecules of the invention may be used in a treatment for reducing or preventing thrombus formation and/or its complications.
  • the binding molecules of the present invention can be used for inhibiting coagulation in various human diseases.
  • the inhibitors of the present invention can be used for the preparation of a medicament for attenuating thrombo-embolic disorders by inhibiting coagulation in vivo.
  • the binding molecules can be used alone or in combination with other drugs.
  • binding molecules of the present invention can be used alone or in combination with other drugs in any suitable ratios, for the preparation of a medicament to treat a subject suffering of a disease or disease symptoms resulting from pathologic thrombosis and or embolism, or at risk with respect to such a disease.
  • the present binding molecules inhibit factor XI-dependent amplification of coagulation or factor XI-dependent coagulation by inhibiting the activation and or activity of factor XI.
  • disorders, diseases and conditions include e.g. (acute) myocardial infarction, ischemic stroke, cardio-embolism due to atrial fibrillation, vascular access thrombosis, deep venous thrombosis, arterial thrombosis, coronary artery thrombosis, atherosclerosis, arthritis, vasculitis, respiratory distress syndrome, pulmonary embolism, thrombo-embolism resulting from surgery such as prostate surgery, orthopaedic surgery, such as e.g.
  • thrombo-embolism resulting from immobilization, thrombosis and occlusion of synthetic grafts, stents, or AV-fistula
  • diffuse intravascular coagulation DIC
  • hemodialysis hemodialysis
  • atrial fibrillation sepsis
  • septic shock organ failure
  • kidney failure toxicity induced by the in vivo administration of therapeutic proteins (e.g. cytokines or mAbs)
  • multiple trauma ischemia-reperfusion injuries and local undesired fibrin deposition such as e.g. fibrin deposition in the lung alveoli during adult respiratory distress.
  • patients suffering from a disease involving coagulation-mediated damage can be administered an effective amount of a monoclonal antibody against factor XI, or fragments a monoclonal antibody against factor XI as described so that activation of factor XI is inhibited.
  • effective amount is meant a concentration of the binding molecule, which is capable of inhibiting coagulation activation.
  • Treatment will generally consist of administering a binding molecule of the invention parenterally, preferably intravenously, intraarterially, intramuscularly or subcutaneously.
  • a binding molecule of the invention preferably is administered goat anti-factor XI antibodies at a dose of 16-50 mg per kg to achieve sufficient inhibition of factor XI in baboons.
  • the dose and administration regimen of a binding molecule of the invention preferably is in the range of a dosage that is equivalent to a dosage of 0.5-20 mg of IgG per kg body weight per week.
  • a binding molecule of the invention is administered at a dosage that is equivalent to a dosage of less than 18, 16, 14, 12, 10 8, 6 or 4 mg of IgG per kg body weight per week and/or at a dosage that is equivalent to a dosage of at least 0.6, 0.8, 1.0, 1.2, 1.5, 2, or 4 mg of IgG per kg body weight per week. It is understood that in case of e.g. antibody fragments the dosage to be used will be the molar equivalent of the corresponding amount of mg of an IgG molecule per kg body weight as indicated.
  • the dosage regimes for the binding molecules of the invention are based on the average serum half-life of a human antibody of about 7 days. The skilled person will know how to adjust the dosage regime of binding molecules with a half-life that is shorter or longer than 7 days.
  • a further factor that influences the dosage regime for a given binding molecule of the invention is its affinity for factor XI.
  • a binding molecule having a Kd of about 1 nM requires a serum level of about 100 nM to produce 99% factor XI inhibition and a binding molecule having a Kd of about 0.1 nM requires a serum level of about 35 nM to produce 99% factor XI inhibition.
  • 95% factor XI inhibition may be achieved with a binding molecule having a Kd of about 1 nM at a serum level of about 50 nM or at 27 nM with a binding molecule having a Kd of about 0.1 nM.
  • Table 1 shows dosage regimes for anti-factor XI-binding molecule per kg of body weight as function of Kd and interval between administrations.
  • the binding molecule is administered at a dosage wherein the inhibition of factor XI activation in a human body fluid is measurable as at least one of (a) substantially complete blockade of factor IX activation in the body fluid; and (b) a substantially complete blockade of thrombin generation in the body fluid.
  • a substantially complete blockade of factor IX activation and/or of thrombin generation is herein understood as factor IX activation and/or thrombin generation being reduced to less than 1% of the amount of factor IX activation and of thrombin generation in the absence of the binding molecule in the body fluid.
  • An administered dosage that effects substantially complete blockade of factor IX activation and/or of thrombin generation preferably is a dosage that yields a molar ratio in the body fluid that is equal to or less than 35, 4, or 1 moles of factor XI binding sites of the binding molecule to 1 mole of factor XI, wherein the factor XI binding sites of the binding molecule have a Kd that is equal to or less than 10 nM, 1 nM, or 0.1 nM, respectively.
  • the body fluid is herein understood to be blood, plasma or serum.
  • the skilled person will know how to adjust the dosage regimes in Table 1 for binding molecules with a half-life that is shorter or longer than 7 days, with a M w different from human IgG, with a Kd different from 10 nM, 1 nM or 0.1 nM and/or to achieve another percentage of inhibition of circulating factor XI.
  • a binding molecule of the invention when administered to a human patient via intravenous infusion, provides complete inhibition of factor XI at dosages below 0.005 or 0.003 g/kg.
  • a binding molecule of the invention when administered to a human patient via intravenous infusion, provides therapeutic benefits at dosages below 0.005 or 0.003 g/kg.
  • a binding molecule of the invention is modified to achieve a desired in vivo serum half life.
  • a polyalkyleneglycol group e.g. polyethyleneglycol (PEG) group, polypropylene glycol, polybutylene glycol
  • a serum protein such as e.g. serum albumin or transferrin
  • the amino acid sequence of the binding molecule can be modified.
  • the amino acid sequence of the constant domains of a binding molecule that is an antibody can be modified (e.g. introducing amino acid substitutions, deletions and/or insertions).
  • the equivalent of the above-indicated weekly dosages of the binding molecules according to the invention can be administered by infusion. Such administration can be repeated as many times as desired.
  • the administration may be performed by bolus injection or infusion or continuous infusion over a period of from less than 2 hours to 24 hours, such as from 2 to 12 hours.
  • the binding molecules of the invention can be administered by slow continuous infusion over a long period, such as more than 24 hours.
  • Such regimen may be continued or repeated one or more times as necessary, for example, after 6 months or 12 months.
  • the dosage can be determined or adjusted by measuring the amount of circulating factor XI-binding molecules of the invention antibodies upon administration in a biological sample by using anti-idiotypic antibodies which target the factor XI-binding molecules of the invention.
  • the antibodies can be administered by maintenance therapy, such as, e.g., once a week for a period of 6 months or more.
  • the binding molecule of the invention is used in the prevention or reduction of thrombosis (and occlusion) of synthetic grafts, stents, or AV-fistula in e.g. kidney-patients undergoing regular dialysis.
  • the binding molecule may be administered at least weekly or several times (e.g. 2, 3 or 4) per week, preferably when the patients are undergoing dialysis.
  • the binding molecule is administered to the patient through the dialysis apparatus, e.g. in the dialysed body fluid that is returned to the patient.
  • the binding molecule of the invention is administered in patient with no regular parenteral access that require nonetheless continuous anticoagulant therapy, such as e.g. in patients with atrial fibrillation, unstable angina pectoris, deep venous thrombosis, diffuse intravascular coagulation, prostate surgery, orthopaedic surgery, particularly of the hip, and other thrombo-embolic disorders.
  • a certain number of administrations of the binding molecule per time period is applied, e.g. once per 2, 3, 4 or 6 weeks.
  • the binding molecule preferably has a half life of at least 6, 7, 8, 10, 12 or 14 days and a Kd that is less than 1 nM, preferably less than 0.5 nM, and most preferably less than 0.1 nM.
  • the binding molecule will be formulated in an injectable form combined with a pharmaceutically acceptable parenteral vehicle.
  • a pharmaceutically acceptable parenteral vehicle include saline, dextrose solution, Ringer's solution and solutions containing small amounts of human serum albumin.
  • the monoclonal antibody or fragments thereof will be formulated in such vehicles at a concentration of from about 20 mg to about 100 mg per ml.
  • the binding molecule is administered by intravenous injection.
  • the invention in a third aspect, relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a binding molecule as disclosed herein above and a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions may be formulated with pharmaceutically acceptable carriers or diluents as well as any other known adjuvants and excipients in accordance with conventional techniques such as those disclosed in (Remington: The Science and Practice of Pharmacy, 19 th Edition, Gennaro, Ed., Mack Publishing Co., Easton, Pa., 1995).
  • compositions according to the invention may be formulated in accordance with routine procedures for administration by any route, such as oral, topical, parenteral, sublingual, transdermal or by inhalation.
  • the compositions may be in the form of tablets, capsules, powders, granules, lozenges, creams or liquid preparations, such as oral or sterile parenteral solutions or suspensions or in the form of a spray, aerosol or other conventional method for inhalation.
  • pharmaceutically acceptable carrier relates to carriers or excipients, which are inherently nontoxic and nontherapeutic.
  • excipients are, but are not limited to, saline, Ringer's solution, dextrose solution and Hank's solution.
  • Nonaqueous excipients such as fixed oils and ethyl oleate may also be used.
  • a preferred excipient is 5% dextrose in saline.
  • the excipient may contain minor amounts of additives such as substances that enhance isotonicity and chemical stability, including buffers and preservatives.
  • the pharmaceutical composition may be administered by any suitable route and mode. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
  • the pharmaceutical compositions of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration.
  • Formulations of the present invention which are suitable for vaginal administration include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • compositions of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • composition is administered by intravenous or subcutaneous injection or infusion.
  • the antibody of the invention are administered in crystalline form by subcutaneous injection, cf. (Yang et al., 2003 PNAS, 100(12):6934-6939).
  • the compounds of the present invention which may be used in the form of a pharmaceutically acceptable salt or in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonicity agents, antioxidants and absorption delaying agents, and the like that are physiologically compatible.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated.
  • the carrier is suitable for parenteral administration, e.g. intravenous or subcutaneous injection or infusion.
  • compositions typically must be sterile, non-pyrogenic and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • suitable aqueous and non-aqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • the pharmaceutical compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonicity agents, such as sugars, polyalcohols such as mannitol, sorbitol, glycerol or sodium chloride in the compositions.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonicity agents, such as sugars, polyalcohol
  • antioxidants may also be included, for example (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients e.g. as enumerated above, as required, followed by sterilization microfiltration.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients e.g. from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the binding molecule may be used in a suitable hydrated form or in the form of a pharmaceutically acceptable salt.
  • a “pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (Berge, S. M., et al., (1977) J. Pharm. Sci. 66:1-19). Examples of such salts include acid addition salts and base addition salts.
  • Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like.
  • nontoxic inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like
  • nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like.
  • Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N,N′-dibenzylethylenediamine, N-methylglucamine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.
  • the active compound i.e., binding molecule
  • the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • the compound may be administered to a subject in an appropriate carrier, for example, liposomes.
  • Liposomes include water-in-oil-in-water CGF emulsions as well as conventional liposomes (Strejan et al., J. Neuroimmunol. 1984; 7:27).
  • the active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a controlled release formulation including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for the preparation of such formulations are generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • compositions can be administered with medical devices known in the art.
  • a therapeutic composition of the invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Pat. No. 5,399,163, U.S. Pat. No. 5,383,851, U.S. Pat. No. 5,312,335, U.S. Pat. No. 5,064,413, U.S. Pat. No. 4,941,880, U.S. Pat. No. 4,790,824, or U.S. Pat. No. 4,596,556.
  • Examples of well-known implants and modules useful in the present invention include: U.S. Pat. No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Pat. No.
  • the binding molecules of the invention can be formulated to prevent or reduce their transport across the placenta. This can be done by methods known in the art, e.g., by PEGylation of the antibodies or by use of F(ab′) 2 fragments. Further references can be made to (Cunningham-Rundles C et al, 1992); and to (Landor, 1995).
  • Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a composition of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect.
  • Such an effective dose will generally depend upon the factors described above. It is preferred that administration be intravenous, intramuscular, intraperitoneal, or subcutaneous.
  • S299 Metal-sulfonyl-D-cyclo-hexylglycyl-glycyl-arginine-p-nitroanilide
  • S2366 L-pyroglutamyl-L-prolyl-L-arginine-p-nitroanilide
  • pAB p-aminobenzamidine
  • the effect of monoclonal antibodies on the chromogenic activity is achieved by pre-incubation 5 ⁇ L of factor XIa at a final concentration of 1 nmol/L with 20 ⁇ L PBS containing various concentrations of monoclonal antibody to be tested for 15 minutes at room temperature. Seventy-five ⁇ L of PBS containing S2366 is added and the conversion of the substrate is measured as described above. A reference consisting of various dilutions of factor XIa (0.1-1 nmol/L) is tested as control. In case of antibodies that inhibit the active site of factor XIa, a decrease in the rate of conversion by the standard amount of factor XIa is observed. This assay is assay 1 described in example 8.
  • PBS phosphate-buffered saline
  • pH 7.4 containing the chromogenic substrate S-2366 1 mmol/L (final concentration)
  • 15 ⁇ L of PBS are added to wells of microtiter plates (Dynatech, Plochingen, Germany). Conversion of the substrate is measured by using a spectrophotometer. Microtiter plates are read on a Multiskan plate reader (Labsystems, Helsinki, Finland) after incubation for various time intervals at room temperature at 405 nm.
  • the effect of monoclonal antibodies on the activation is tested by adding 15 ⁇ L of PBS containing the antibody to be tested to the mixture of factor XI and factor XIIa, in a way that first the antibody is added to factor XI, and after 15 minutes incubation factor XIIa is added. Dilutions of factor XIIa are tested as control. Seventy-five ⁇ L of PBS containing S2366 are then added and the conversion of the substrate is measured as described above. A reference consisting of various dilutions of factor XIa (0.1-1 nmol/L) is also tested as control. In case of antibodies that inhibit the activation of factor XI, a decrease in the rate of conversion by the amount of factor XIa generated by factor XIIa is observed. This assay is referred to assay 2 in Table 2.
  • This assay is referred to as assay 3 in Table 2.
  • Factor XIa clotting activity Effect of the monoclonal antibodies on factor XI or factor XIa clotting activity is tested by incubating factor XI or factor XIa with an equal volume of PBS containing 1 to 10 molar excess of antibody versus factor XI. Factor XI clotting activity is then tested with a one-stage clotting assay.
  • assays are well known to those skilled in the art. These assays are referred to as assay 4 in Table 2 when performed with factor XI, and as assay 5 when performed with factor XIa.
  • mice with purified human factor XI with the aim to isolate lymphocytes from the immunised mice and producing murine hybridomas. It will be further appreciated that the procedure can be employed to produce antibodies against factor XI, factor XIa or fragments thereof.
  • the first factor XI or XIa gift is mixed with complete Freund's adjuvant, the subsequent with incomplete Freund's adjuvant.
  • spleens are removed from the immunised mice and the splenocytes are fused with the murine myeloma cell-line SP2/0-Ag14, according to the procedure first described by (Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975; 256:495-7), except that feeder cells are replaced by IL-6.
  • Immunised mice are sacrificed and splenocytes teased from the spleens, and washed in serum free Dulbecco's Modified Eagles medium.
  • SP2/0-Ag14 myeloma cells are washed, and added to the splenocytes yielding a 5:1 ratio of splenocytes to myeloma cells.
  • the cells are then pelleted, and the supernatant is removed.
  • One ml of a 40% (v/v) solution of polyethylene glycol 1500 is then added dropwise over a 60 sec period, after which the cells are incubated for another 60 sec at 37° C.
  • Nine ml of Dulbecco's Modified Eagles medium is then added with gentle agitation.
  • the cells are pelleted, washed to remove residual polyethylene glycol, and finally plated at a concentration of 10 5 cellsper well in Dulbecco's Modified Eagles medium containing 10% (v/v) fetal calf serum (100 ⁇ l per well). After 24 hours, 100 ⁇ l of a 2 ⁇ solution of hypoxanthine/azaserine selection medium is added to each well. At day 4 hypoxanthine/azaserine selection medium is replenished, at day 7 it is replaced by Dulbecco's Modified Eagles medium containing 10% (v/v) fetal calf serum. During all incubations monocyte derived or recombinant human IL-6 is present in the culture at concentrations of approximately 10 pg/ml. About 80% of the wells exhibit cell growth at day 10.
  • the wells are screened for the presence of antibody to factor XI or XIa using an enzyme-linked immunosorbent assay, in which purified human factor XI or XIa is used for coating (2 ⁇ g/ml in phosphate buffered saline, pH 7.4 [PBS]; 100 ⁇ l/well). Residual non-specific binding sites are then blocked by 30 minutes incubation at room temperature with PBS/0.1% (w/v) Tween 20 (PBS-T) containing 0.2% (w/v) gelatin (PBS-TG). Then, after a washing procedure (5 times with PBS-T), the plates are incubated for 60 minutes at 37° C.
  • PBS-T PBS/0.1% (w/v) Tween 20
  • PBS-TG 0.2% gelatin
  • This assay is referred to as assay 6 in Table 2, the denotation is FXI/FXIa when a mAb binds to factor XI and factor XIa when tested in these ELISAs; FXI when only binding to factor XI and not to factor XIa; and FXIa when the monoclonal antibody only binds to factor XIa and not to factor XI.
  • Antibody is produced in vitro from the hybridoma anti-factor XI or XIa by culturing the cells in 1 liter roller-bottles in Iscove's Modified Dulbecco medium supplemented with 2% (v/v) fetal calf serum, 10 pg/ml IL-6, 50 ⁇ M 2-mercaptoethanol, and penicillin and streptomycin. The cells are grown to a density of >10 6 cells per ml, and one to two weeks later the supernatants are collected. Solid ammonium sulphate is added to yield 50% saturation (i.e., approximately 2M), and an antibody-enriched fraction is obtained by centrifugation for 30 minutes at 1,300 g.
  • Iscove's Modified Dulbecco medium supplemented with 2% (v/v) fetal calf serum, 10 pg/ml IL-6, 50 ⁇ M 2-mercaptoethanol, and penicillin and streptomycin.
  • the cells are grown to a
  • the precipitate is dissolved in 1.5 M NaCl/0.75 M glycine, pH 8.9, and put onto a protein A-Sepharose column (Pharmacia). The column is washed with PBS, and then anti-factor XI monoclonal antibody is eluted off with glycine-HCl, pH 2.5. Fractions are neutralised instantaneously with 2M TRIS, pH 8.0, and those containing protein are pooled and dialysed against PBS.
  • Antibodies against factor XI are characterized by testing them in the assays described above. Outcome of these experiments is as shown in Table 2.
  • inhibitor For application as inhibitor several types of antibody are selected, referred to in the table as “inhibitor”. Note that this table is not complete, other functional patterns may be encountered.
  • Efficacy of anti-factor XI monoclonal antibody to attenuate or prevent thrombosis in vivo is studied in factor XI knock-out mice (D Gailani et al., Blood Coagul. Fibrinolysis. 1997; 8:134-144), which are supplemented with human factor XI.
  • the animals are then subjected to experimental thrombus formation induced by FeCl 3 -induced injury, for example as described by T Renné et al., J Exp Med 2005; 202:271-281. Briefly, 4-5-wk-old mice are anesthetized by i.p.
  • Human factor XI for example 0.2 mg/kg
  • 10e8 CFSE-labeled platelets per mouse are injected via the tail vein as a second injection.
  • Monoclonal antibody against human factor XI is administered either together with, before or after the injection of human factor XI at a dose of 0.5-10 mg/kg.
  • the mesentery is externalized through a midline abdominal incision.
  • 35-60- ⁇ m-diameter arterioles are visualized at 10 ⁇ with an inverted microscope (Axiovert 200; Carl Zeiss MicroImaging, Inc.) equipped with a 100-W fluorescent lamp source (HBO) and a CCD camera (CV-M300; Visitron Systems GmbH) connected to an S-VHS video recorder (AG-7355; Panasonic).
  • HBO 100-W fluorescent lamp source
  • CV-M300 Visitron Systems GmbH
  • S-VHS video recorder AG-7355; Panasonic
  • a thrombus was defined as a platelet aggregate >20 ⁇ m in diameter. Efficacy of the monoclonal antibody to inhibit thrombus formation is shown by comparing results of mice supplemented with human factor XI only (thrombus formation; occlusion), human factor XI and anti-factor XI monoclonal antibody (no thrombus formation; no occlusion), or not supplemented at all (no thrombus formation; no occlusion), and wild-type mice (thrombus formation; occlusion).
  • Efficacy of anti-factor XI monoclonal antibody to attenuate or prevent thrombosis in vivo is also studied in non-human primate models for thrombosis, for example the models as described by A Gruber et al. (Blood. 2003; 102:953-955). Briefly, baboons are appropriately anaestesized and injected intravenously with intravenous porcine heparin (1000 U/mL; Wyeth-Ayerst, Pearl River, N.Y.) as an anti-coagulant control or affinity-purified monoclonal antibody against factor XI at a dose of 0.5-10 mg per kg.
  • heparin or anti-factor XI monoclonal antibody do not receive heparin or anti-factor XI monoclonal antibody.
  • the antithrombotic effects of heparin or anti-factor XI monoclonal antibody are studied using thrombogenic devices deployed for 60 minutes into surgically placed high flow chronic arteriovenous (AV) shunts.
  • the device to be used can be a 20-mm long dacron graft segment (0.25 mL) and a silicone extension chamber (1.3 mL), or a 20-mm long (0.25 mL) ringed expanded polytetrafluoroethylene (ePTFE, teflon) graft segments (WL Gore, Newark, Del.) in the shunt.
  • hypothrombogenic ePTFE graft is converted into an acutely thrombogenic tissue factor-dependent device as described by Gruber et al.
  • the graft Prior to deployment, the graft is rinsed by passing 50 mL saline through the lumen. Blood flow is kept at 100 mL/min by proximal clamping.
  • Thrombogenesis is assessed by measuring radiolabeled fibrin and platelet contents of the thrombogenic devices. In brief, the terminal fibrin content of graft/chamber thrombi (fibrin deposition) was determined by direct 125 Iodine-labeled fibrin counting.
  • NPAR Net platelet accumulation rate
US13/000,284 2008-06-19 2009-06-19 Use of anti-factor xi antibodies for prevention of thrombus formation Abandoned US20110159006A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/000,284 US20110159006A1 (en) 2008-06-19 2009-06-19 Use of anti-factor xi antibodies for prevention of thrombus formation

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US7388208P 2008-06-19 2008-06-19
US61073882 2008-06-19
US13/000,284 US20110159006A1 (en) 2008-06-19 2009-06-19 Use of anti-factor xi antibodies for prevention of thrombus formation
PCT/NL2009/050361 WO2009154461A1 (en) 2008-06-19 2009-06-19 Use of anti-factors xi antibodies for prevention of thrombus formation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/NL2009/050361 A-371-Of-International WO2009154461A1 (en) 2008-06-19 2009-06-19 Use of anti-factors xi antibodies for prevention of thrombus formation

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/527,261 Continuation US8568724B2 (en) 2008-06-19 2012-06-19 Use of anti-factor XI antibodies for prevention of thrombus formation

Publications (1)

Publication Number Publication Date
US20110159006A1 true US20110159006A1 (en) 2011-06-30

Family

ID=40933725

Family Applications (4)

Application Number Title Priority Date Filing Date
US13/000,284 Abandoned US20110159006A1 (en) 2008-06-19 2009-06-19 Use of anti-factor xi antibodies for prevention of thrombus formation
US13/527,261 Active US8568724B2 (en) 2008-06-19 2012-06-19 Use of anti-factor XI antibodies for prevention of thrombus formation
US14/034,738 Abandoned US20140194600A1 (en) 2008-06-19 2013-09-24 Use of anti-factor xi antibodies for prevention of thrombus formation
US15/845,630 Abandoned US20180118850A1 (en) 2008-06-19 2017-12-18 Use of anti-factor xi antibodies for prevention of thrombus formation

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/527,261 Active US8568724B2 (en) 2008-06-19 2012-06-19 Use of anti-factor XI antibodies for prevention of thrombus formation
US14/034,738 Abandoned US20140194600A1 (en) 2008-06-19 2013-09-24 Use of anti-factor xi antibodies for prevention of thrombus formation
US15/845,630 Abandoned US20180118850A1 (en) 2008-06-19 2017-12-18 Use of anti-factor xi antibodies for prevention of thrombus formation

Country Status (9)

Country Link
US (4) US20110159006A1 (de)
EP (1) EP2297207B1 (de)
DK (1) DK2297207T3 (de)
ES (1) ES2702365T3 (de)
HR (1) HRP20181727T1 (de)
HU (1) HUE041556T2 (de)
PL (1) PL2297207T3 (de)
TR (1) TR201820051T4 (de)
WO (1) WO2009154461A1 (de)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9771417B2 (en) 2014-08-07 2017-09-26 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
US9988443B2 (en) 2014-08-07 2018-06-05 Novartis Ag Angiopoetin-like 4 (ANGPTL4) antibodies and methods of use
US20180305461A1 (en) * 2011-02-01 2018-10-25 Bac Ip B.V. Antigen-Binding Protein Directed Against Epitope in the CH1 Domain of Human IgG Antibodies
CN110325211A (zh) * 2016-12-23 2019-10-11 诺华股份有限公司 采用抗因子XI/XIa抗体治疗的方法
US10465011B2 (en) 2015-06-26 2019-11-05 Novartis Ag Factor XI antibodies and methods of use
US10647780B2 (en) 2016-05-25 2020-05-12 Novartis Ag Reversal binding agents for anti-factor XI/XIa antibodies and uses thereof
JP2020513757A (ja) * 2016-12-23 2020-05-21 ノバルティス アーゲー 第xi因子抗体および使用方法
JP2022519807A (ja) * 2019-08-08 2022-03-25 上海博槿生物科技有限公司 抗凝固因子XI活性型因子XIa抗体並びにその調製方法及び使用
CN114478782A (zh) * 2018-08-09 2022-05-13 上海仁会生物制药股份有限公司 抗凝血因子xi抗体
US11958911B2 (en) 2017-02-10 2024-04-16 Shanghai Benemae Pharmaceutical Anti-coagulation factor XI antibody

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160046728A1 (en) * 2007-10-31 2016-02-18 Bioxodes Sa Identification and molecular characterisation of proteins, expressed in the ixodes ricinus salivary glands
EP2222707B1 (de) 2007-11-21 2016-01-06 Oregon Health & Science University Monoklonale anti-faktor-xi-antikörper und verfahren zu deren anwendung
EP2373691B1 (de) 2008-12-18 2019-01-23 Oregon Health&Science University Anti-fxi-antikörper und verfahren zu ihrer verwendung
US9518128B2 (en) 2011-12-14 2016-12-13 Janssen Pharmaceuticals, Inc. Thrombin-binding antibody molecules
GB201121513D0 (en) 2011-12-14 2012-01-25 Cambridge Entpr Ltd Thrombin-binding antibody molecules and uses thereof
PL2847228T3 (pl) * 2012-05-10 2019-03-29 Bayer Pharma Aktiengesellschaft Przeciwciała zdolne do wiązania się z czynnikiem krzepnięcia XI i/lub jego aktywowaną postacią, czynnikiem Xla oraz ich zastosowanie
CA3025896A1 (en) * 2015-07-23 2017-01-26 The Regents Of The University Of California Antibodies to coagulation factor xia and uses thereof
CA3026032A1 (en) * 2015-07-23 2017-01-26 The Regents Of The University Of California Reversal agents for fxia inhibitors
AR107505A1 (es) 2016-01-22 2018-05-09 Merck Sharp & Dohme Anticuerpos anti-factor de la coagulación xi
BR112018069283A2 (pt) * 2016-03-23 2019-01-22 Prothix Bv anticorpos monoclonais contra o sítio ativo de fator xi e usos dos mesmos
TN2018000417A1 (en) 2016-06-14 2020-06-15 Adimab Llc Anti-coagulation factor xi antibodies.
WO2019030706A1 (en) 2017-08-10 2019-02-14 Janssen Pharmaceutica Nv ANTI-THROMBIN ANTIBODY MOLECULES AND METHODS OF USE IN ORTHOPEDIC SURGERY
WO2019035055A1 (en) 2017-08-16 2019-02-21 Janssen Pharmaceutica Nv ANTI-THROMBIN ANTIBODY MOLECULES AND METHODS OF USE WITH PLATELET ANTI-AGGREGATE AGENTS
WO2019102353A1 (en) 2017-11-22 2019-05-31 Novartis Ag Reversal binding agents for anti-factor xi/xia antibodies and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6005091A (en) * 1996-01-17 1999-12-21 Smithkline Beecham Corporation Nucleic acids encoding immunoglobulin domains
WO2003013423A2 (en) * 2001-08-05 2003-02-20 Gvg, Inc. Antithrombotic agent

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5437864A (en) * 1987-03-31 1995-08-01 The Scripps Research Institute Method of inhibiting blood coagulation in extracorporeal circulation by inhibiting human tissue factor
US4963657A (en) 1988-06-09 1990-10-16 Temple University Of The Commonwealth System Of Higher Education Monoclonal antibodies to the light chain region of human factor XII and methods of preparing and using the same
US5506134A (en) * 1990-10-22 1996-04-09 Corvas International, Inc. Hypridoma and monoclonal antibody which inhibits blood coagulation tissue factor/factor VIIa complex
WO1993009803A1 (en) * 1991-11-22 1993-05-27 The Scripps Research Institute Factor x-derived polypeptides and anti-peptide antibodies, systems and therapeutic methods for inhibiting inflammation
GB9224888D0 (en) * 1992-11-27 1993-01-13 Univ Singapore Monoclonal antibody
US6500660B1 (en) * 1996-11-27 2002-12-31 Université Catholique de Louvain Chimeric target molecules having a regulatable activity
WO1999001476A1 (en) * 1997-07-03 1999-01-14 Smithkline Beecham Corporation CRYSTAL STRUCTURES OF ANTI-FACTOR IX Fab FRAGMENTS AND METHODS OF USE FOR PEPTIDOMIMETIC DESIGN
WO2005016455A2 (en) * 2003-08-14 2005-02-24 D. Collen Research Foundation Vzw Antibodies against factor viii with modified glycosylation in the variable region
US20030143225A1 (en) * 2001-03-08 2003-07-31 Genentech, Inc. Combinations of anti-tissue factor antibodies and anticoagulant and/or antiplatelet agents
EP1433793A4 (de) * 2001-09-13 2006-01-25 Inst Antibodies Co Ltd Verfahren zum erstellen einer kamel-antikörperbibliothek
AU2002354047B2 (en) * 2001-11-05 2007-11-22 Board Of Regents, The University Of Texas System Recombinant antibodies for the detection and neutralization of anthrax toxin
US7579000B2 (en) * 2002-05-01 2009-08-25 Bayer Schering Pharma Ag Tissue factor targeted antibodies as anticoagulants
US20040180855A1 (en) * 2003-02-19 2004-09-16 Schumacher William A. Methods of treating thrombosis with reduced risk of increased bleeding times
CA2531796A1 (en) * 2003-08-08 2005-02-17 Transtech Pharma, Inc. Aryl and heteroaryl compounds, compositions, and methods of use
EP2222707B1 (de) * 2007-11-21 2016-01-06 Oregon Health & Science University Monoklonale anti-faktor-xi-antikörper und verfahren zu deren anwendung
JOP20200312A1 (ar) * 2015-06-26 2017-06-16 Novartis Ag الأجسام المضادة للعامل xi وطرق الاستخدام
BR112018069283A2 (pt) * 2016-03-23 2019-01-22 Prothix Bv anticorpos monoclonais contra o sítio ativo de fator xi e usos dos mesmos

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6005091A (en) * 1996-01-17 1999-12-21 Smithkline Beecham Corporation Nucleic acids encoding immunoglobulin domains
WO2003013423A2 (en) * 2001-08-05 2003-02-20 Gvg, Inc. Antithrombotic agent

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Baglia et al., Blood, 1989, 74:244-251. *
Minnema et al., J. Clin. Invest., 1998, 101:10-14. *
Sinha et al., Biochemistry, 1987, 26:3768-3775 *
Sriskandan et al. Cancer Chemother Pharmacol. 1986;18(1):63-8. abstract only *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180305461A1 (en) * 2011-02-01 2018-10-25 Bac Ip B.V. Antigen-Binding Protein Directed Against Epitope in the CH1 Domain of Human IgG Antibodies
US9988443B2 (en) 2014-08-07 2018-06-05 Novartis Ag Angiopoetin-like 4 (ANGPTL4) antibodies and methods of use
US9771417B2 (en) 2014-08-07 2017-09-26 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
US10577411B2 (en) 2014-08-07 2020-03-03 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
US10465011B2 (en) 2015-06-26 2019-11-05 Novartis Ag Factor XI antibodies and methods of use
US10647780B2 (en) 2016-05-25 2020-05-12 Novartis Ag Reversal binding agents for anti-factor XI/XIa antibodies and uses thereof
CN110325211A (zh) * 2016-12-23 2019-10-11 诺华股份有限公司 采用抗因子XI/XIa抗体治疗的方法
JP2020513757A (ja) * 2016-12-23 2020-05-21 ノバルティス アーゲー 第xi因子抗体および使用方法
US11168147B2 (en) 2016-12-23 2021-11-09 Novartis Ag Factor XI antibodies and methods of use
JP7139332B2 (ja) 2016-12-23 2022-09-20 ノバルティス アーゲー 第xi因子抗体および使用方法
US11958911B2 (en) 2017-02-10 2024-04-16 Shanghai Benemae Pharmaceutical Anti-coagulation factor XI antibody
CN114478782A (zh) * 2018-08-09 2022-05-13 上海仁会生物制药股份有限公司 抗凝血因子xi抗体
JP2022519807A (ja) * 2019-08-08 2022-03-25 上海博槿生物科技有限公司 抗凝固因子XI活性型因子XIa抗体並びにその調製方法及び使用
JP7319721B2 (ja) 2019-08-08 2023-08-02 上海博槿生物科技有限公司 抗凝固因子XI活性型因子XIa抗体並びにその調製方法及び使用

Also Published As

Publication number Publication date
US20180118850A1 (en) 2018-05-03
US20120259097A1 (en) 2012-10-11
US8568724B2 (en) 2013-10-29
HRP20181727T1 (hr) 2019-02-08
EP2297207A1 (de) 2011-03-23
EP2297207B1 (de) 2018-10-03
TR201820051T4 (tr) 2019-01-21
HUE041556T2 (hu) 2019-05-28
ES2702365T3 (es) 2019-02-28
PL2297207T3 (pl) 2019-03-29
US20140194600A1 (en) 2014-07-10
DK2297207T3 (en) 2018-12-03
WO2009154461A1 (en) 2009-12-23

Similar Documents

Publication Publication Date Title
US8568724B2 (en) Use of anti-factor XI antibodies for prevention of thrombus formation
US6391299B1 (en) Anti-factor IX/IXa antibodies
CA2402596A1 (en) Anti-tissue factor antibodies with enhanced anticoagulant potency
AU2001250814A1 (en) Anti-tissue factor antibodies with enhanced anticoagulant potency
US6703494B2 (en) Anti-tissue factor antibodies with enhanced anticoagulant potency
CN109153726A (zh) 抗因子xi的活性位点的单克隆抗体及其用途
WO2003029295A1 (en) Human tissue factor antibodies
US20060057140A1 (en) Anticoagulant agents useful in treatment of thrombosis
US20050037006A1 (en) Antithrombotic agents
US20020136725A1 (en) Antithrombotic agents
US20030124117A1 (en) Combinations of anti-tissue factor antibodies and anticoagulant and/or antiplatelet agents
WO2004039842A2 (en) Humanized tissue factor antibodies
US20030143225A1 (en) Combinations of anti-tissue factor antibodies and anticoagulant and/or antiplatelet agents
US20070190059A1 (en) Antithrombotic agents
US20050106139A1 (en) Humanized tissue factor antibodies
US20040001830A1 (en) Human tissue factor antibodies
AU2002333213A1 (en) Human tissue factor antibodies

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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