WO2002067857A3 - Modified annexin proteins and methods for preventing thrombosis - Google Patents

Modified annexin proteins and methods for preventing thrombosis Download PDF

Info

Publication number
WO2002067857A3
WO2002067857A3 PCT/US2002/005079 US0205079W WO02067857A3 WO 2002067857 A3 WO2002067857 A3 WO 2002067857A3 US 0205079 W US0205079 W US 0205079W WO 02067857 A3 WO02067857 A3 WO 02067857A3
Authority
WO
WIPO (PCT)
Prior art keywords
protein
annexin
modified annexin
nucleic acid
thrombosis
Prior art date
Application number
PCT/US2002/005079
Other languages
French (fr)
Other versions
WO2002067857A2 (en
Inventor
Anthony Allison
Original Assignee
Surromed Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Surromed Inc filed Critical Surromed Inc
Priority to DE60219611T priority Critical patent/DE60219611T2/en
Priority to JP2002567229A priority patent/JP2004528025A/en
Priority to EP02721083A priority patent/EP1379266B1/en
Priority to AU2002252035A priority patent/AU2002252035A1/en
Publication of WO2002067857A2 publication Critical patent/WO2002067857A2/en
Publication of WO2002067857A3 publication Critical patent/WO2002067857A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • 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

Abstract

A modified annexin protein, preferably annexin V, is used to prevent thrombosis without increasing hemorrhage. Annexin binds to phosphatidylserine on the outer surface of cell membranes, thereby preventing binding of the prothrombinase complex necessary for thrombus formation. It does not, however, affect platelet aggregation necessary for hemostasis. The modified annexin molecule can be a homodimer of annexin, an annexin molecule coupled to one or more polyethylene glycol chains, or an annexin molecule coupled to another protein. By increasing the molecular weight of annexin, the modified annexin is made to remain in circulation for sufficient time to provide a sustained therapeutic effect.

Description


  



   MODIFIED   ANNEXIN    PROTEINS AND METHODS FOR PREVENTING
THROMBOSIS
FIELD OF THE INVENTION
The present invention relates generally to methods and compositions for treating thrombosis. More particularly, it relates to modified annexin proteins and methods for their use.



  BACKGROUND OF THE INVENTION
Thrombosis-the formation, development, or presence of a blood clot (thrombus) in a blood vessel-is the most common severe medical disorder. The most frequent example of arterial thrombosis is coronary thrombosis, which leads to occlusion of the coronary arteries and often to myocardial infarction (heart attack).



  More than 1.3 million patients are admitted to the hospital for myocardial infarction each year in North America. The standard therapy is administration of a thrombolytic protein by infusion. Thrombolytic treatment of acute myocardial infarction is estimated to save 30 lives per 1000 patients treated; nevertheless the 30-day mortality for this disorder remains substantial (Mehta et   al.,    Lancet 356: 449-454 (2000), incorporated herein by reference). It would be convenient to administer antithrombotic and thrombolytic agents by bolus injection, since they might be used before admission to hospital with additional benefit (Rawles, J. Am. Coll. Cardiol.



  30: 1181-1186 (1997), incorporated herein by reference). However, bolus injection (as opposed to a more gradual intravenous infusion) significantly increases the risk of cerebral hemorrhage (Mehta et al., 2000). The development of an agent able to prevent thrombosis and/or increase thrombolysis, without augmenting the risk of bleeding, would be desirable.



   Unstable angina, caused by inadequate oxygen delivery to the heart due to coronary occlusion, is the most common cause of admission to hospital, with 1.5 million cases a year in the United States alone. When patients with occlusion of coronary arteries are treated with angioplasty and stenting, the use of an antibody against platelet gp IIb/IIIa decreases the likelihood of restenosis. However, the same antibody has shown no benefit in unstable angina without angioplasty, and a better method for preventing coronary occlusion in these patients is needed. 



   Another important example of arterial thrombosis is cerebral thrombosis.



  Intravenous recombinant tissue plasminogen activator (rtPA) is the only treatment for acute ischemic stroke that is approved by the Food and Drug Administration. The earlier it is administered the better (Ernst et   al.,    Stroke 31: 2552-2557 (2000), incorporated herein by reference). However, intravenous   rtPA    administration is associated with increased risk of intracerebral hemorrhage. Full-blown strokes are often preceded by transient ischemic attacks (TIA), and it is estimated that about 300,000 persons suffer TIA every year in the United States. It would be desirable to have a safe and effective agent that could be administered as a bolus and would for several days prevent recurrence of cerebral thrombosis without increasing the risk of cerebral hemorrhage.

   Thrombosis also contributes to peripheral arterial occlusion in diabetics and other patients, and an efficacious and safe antithrombotic agent for use in such patients is needed.



   Venous thrombosis is a frequent complication of surgical procedures such as hip and knee arthroplasties. It would be desirable to prevent thrombosis without increasing hemorrhage into the field of operation. Similar considerations apply to venous thrombosis associated with pregnancy and parturition. Some persons are prone to repeated venous thrombotic events and are currently treated by antithrombotic agents such as coumarin-type drugs. The dose of such drugs must be titrated in each patient, and the margin between effective antithrombotic doses and those increasing hemorrhage is small. Having a treatment with better separation of antithrombotic activity from increased risk of bleeding is desirable.

   All of the recently introduced antithrombotic therapies, including ligands of platelet gp   IIb/IIIa,    low molecular weight heparin, and a pentasaccharide inhibitor of factor Xa, carry an increased risk of bleeding (Levine et   al.,    Chest 119: 108S-121S (2001), incorporated herein by reference). Hence there is a need to explore alternative strategies for preventing arterial and venous thrombosis without augmenting the risk of hemorrhage. 



   To inhibit the extension of arterial or venous thrombi without increasing hemorrhage, it is necessary to exploit potential differences between mechanisms involved in hemostasis and those involved in thrombosis in large blood vessels.



  Primary hemostatic mechanisms include the formation of platelet microaggregates, which plug capillaries and accumulate over damaged or activated endothelial cells in small blood vessels. Inhibitors of platelet aggregation, including agents suppressing the formation or action of thromboxane   A2,    ligands of gp   IIa/IIIb,    and drugs acting on ADP receptors such as clopidogrel (Hallopeter, Nature 409: 202-207 (2001), incorporated herein by reference), interfere with this process and therefore increase the risk of bleeding (Levine et al., 2001). In contrast to microaggregate formation, occlusion by an arterial or venous thrombus requires the continued recruitment and incorporation of platelets into the thrombus.

   To overcome detachment by shear forces in large blood vessels, platelets must be bound tightly to one another and to the fibrin network deposited around them.



   Evidence has accumulated that the formation of tight macroaggregates of platelets is facilitated by a cellular and a humoral amplification mechanism, which reinforce each other. In the cellular mechanism, the formation of relatively loose microaggregates of platelets, induced by moderate concentrations of agonists such as
ADP, thromboxane A2, or collagen, is accompanied by the release from platelet a-granules of the 85-kD protein Gas6 (Angelillo-Scherrer et   al.,    Nature Medicine 7: 215-221 (2001), incorporated herein by reference). Binding of released Gas6 to receptor tyrosine kinases (Axl, Sky, Mer) expressed on the surface of platelets induces complete degranulation and the formation of tight macroaggregates of these cells. In the humoral amplification mechanism, a prothrombinase complex is formed on the surface of activated platelets and microvesicles.

   This generates thrombin and fibrin.



  Thrombin is itself a potent platelet activator and inducer of the release of Gas6 (Ishimoto and Nakano, FEBS Lett. 446: 197-199 (2000), incorporated herein by reference). Fully activated platelets bind tightly to the fibrin network deposited around them. Histological observations show that both platelets and fibrin are necessary for the formation of a stable coronary thrombus in humans (Falk et al.



  Interrelationship between atherosclerosis and thrombosis. In Vanstraete et al.



  (editors), Cardiovascular Thrombosis:   Thrombocardiology    and   Thromboneurology.    



  Philadelphia: Lipincott-Raven Publishers (1998), pp. 45-58, incorporated herein by reference). Another platelet adhesion molecule, amphoterin, is translocated to the platelet surface during activation, and binds anionic phospholipid (Rouhainen et al.,
Thromb. Hemost. 84: 1087-1094 (2000), incorporated herein by reference). Like
Gas6, amphoterin could form a bridge during platelet aggregation.



   The question arises whether it is possible to inhibit these amplification mechanisms but not the initial platelet aggregation step, thereby preventing thrombosis without increasing hemorrhage. The importance of cellular amplification has recently been established by studies of mice with targeted inactivation of Gas6   (Angelillo-Scherrer etal.,    2001). The Gas6-/-mice were found to be protected against thrombosis and embolism induced by collagen and epinephrine. However, the
Gas6-/-mice did not suffer from spontaneous hemorrhage and had normal bleeding after tail clipping. Furthermore, antibodies against Gas6 inhibited platelet aggregation in vitro as well as thrombosis induced in vivo by collagen and epinephrine. In principle, such antibodies, or ligands competing for Gas6 binding to receptor tyrosine kinases, might be used to inhibit thrombosis.

   However, in view of the potency of humoral amplification, it might be preferable to inhibit that step. Ideally such an inhibitor would also have additional suppressive activity on the Gas6-mediated cellular amplification mechanism.



   A strategy for preventing both cellular and humoral amplification of platelet aggregation is provided by the annexins, a family of highly homologous antithrombotic proteins of which ten are expressed in several human tissues (Benz and
Hofmann, Biol. Chem. 378: 177-183 (1997), incorporated herein be reference).



  Annexins share the property of binding calcium and negatively charged phospholipids, both of which are required for blood coagulation. Under physiological conditions, negatively charged phospholipid is mainly supplied by phosphatidylserine (PS) in activated or damaged cell membranes. In intact cells, PS is confined to the inner leaflet of the plasma membrane bilayer and is not accessible on the surface.



  When platelets are activated, the amounts of PS accessible on their surface, and therefore the extent of annexin binding, are greatly increased (Sun et al., Thrombosis
Res. 69: 289-296 (1993), incorporated herein by reference). During activation of platelets, microvesicles are released from their surfaces, greatly increasing the surface area expressing anionic phospholipids with procoagulant activity (Merten et al.,
Circulation 99: 2577-2582 (1999); Chow et al., J. Lab. Clin. Med. 135: 66-72 (2000), both incorporated herein by reference). These may play an important role in the propagation of platelet-mediated arterial thrombi.



   Proteins involved in the blood coagulation cascade (factors X, Xa, and Va) bind to membranes bearing PS on their surfaces, and to one another, forming a stable, tightly bound prothrombinase complex. Several annexins, including II, V, and VIII, bind PS with high affinity, thereby preventing the formation of a prothrombinase complex and exerting antithrombotic activity. Annexin V binds PS with very high affinity (Kd = 1. 7 nmol/L), greater than the affinity of factors X, Xa, and Va for negatively charged phospholipids (Thiagarajan and Tait, J. Biol. Chem. 265: 1742017423 (1990), incorporated herein by reference). Tissue factor-dependent blood coagulation on the surface of activated or damaged endothelial cells also requires surface expression of PS, and annexin V can inhibit this process (van Heerde et al.,
Arterioscl.

   Thromb. 14: 824-830 (1994), incorporated herein by reference), although annexin is less effective in this activity than in inhibition of prothrombinase generation (Rao et al., Thromb. Res. 62: 517-531 (1992), incorporated herein by reference).



   The binding of annexin V to activated platelets and to damaged cells probably explains the selective retention of the protein in thrombi. This has been shown in experimental animal models of venous and arterial thrombosis (Stratton et al.,
Circulation 92: 3113-3121 (1995); Thiagarajan and Benedict, Circulation 96: 23392347 (1997), both incorporated herein by reference), and labeled annexin has been proposed for medical imaging of vascular thrombi in humans, with reduced noise and increased safety (Reno and Kasina, International Patent Application   PCT/US95/07599    (WO 95/34315) (published December 21,1995), incorporated herein by reference).



  The binding to thrombi of a potent antithrombotic agent such as annexin V provides a strategy for preventing the extension or recurrence of thrombosis. Transient myocardial ischemia also increases annexin V binding (Dumont et   al.,    Circulation 102: 1564-1568 (2000), incorporated herein by reference). Annexin V imaging in humans has shown increased binding of the protein in transplanted hearts when endomyocardial biopsy has demonstrated vascular rejection (Acio et al., J. Nuclear
Med. 41 (5 Suppl.): 127P (2000), incorporated herein by reference). This binding is presumably due to PS exteriorized on the surface of damaged endothelial cells, as well as of apoptotic myocytes in hearts that are being rejected.

   It follows that administration of annexin after myocardial infarction should prevent the formation of pro-thrombotic complexes on both platelets and endothelial cells, thereby preventing the extension or recurrence of thrombosis. Annexin V binding is also augmented following cerebral hypoxia in humans (D'Arceuil et   al.,    Stroke 2000: 2692-2700 (2000), incorporated herein by reference), which supports the hypothesis that administration of annexin following TIA may decrease the likelihood of developing a full-blown stroke.



   Annexins have shown anticoagulant activity in several in vitro thrombindependent assays, as well as in experimental animal models of venous thrombosis (Romisch et al., Thrombosis Res. 61: 93-104 (1991); Van Ryn-McKenna et al.,
Thrombosis Hemostasis 69: 227-230 (1993), both incorporated herein by reference) and arterial thrombosis (Thiagarajan and Benedict, 1997). Remarkably, annexin in antithrombotic doses had no demonstrable effect on traditional ex vivo clotting tests in treated rabbits (Thiagarajan and Benedict, 1997) and did not significantly prolong bleeding times of treated rats (Van Ryn-McKenna et al., 1993). In treated rabbits annexin did not increase bleeding into a surgical incision (Thiagarajan and Benedict, 1997). Thus, uniquely among all the agents so far investigated, annexins exert antithrombotic activity without increasing hemorrhage.

   Annexins do not inhibit platelet aggregation triggered by agonists other than thrombin (van Heerde et al., 1994), and platelet aggregation is the primary hemostatic mechanism. In the walls of damaged blood vessels and in extravascular tissues, the tissue   factor/VIIa    complex also exerts hemostatic effects, and this system is less susceptible to inhibition by annexin V than is the prothrombinase complex (Rao   etal.,    1992). This is one argument for confining administered annexin V to the vascular compartment as far as possible; the risk of hemorrhage is likely to be reduced. 



   Despite such promising results for preventing thrombosis, a major problem associated with the therapeutic use of annexins is their short half-life in the circulation, estimated in experimental animals to be 5 to 15 minutes (Romisch et al.,
1991; Stratton   etal.,    1995; Thiagarajan and Benedict, 1997); annexin V also has a short half-life in the circulation of humans (Strauss et al., J. Nuclear Med. 41 (5
Suppl.): 149P (2000), incorporated herein by reference). Most of the annexin is lost into the urine, as expected of a   36 kDa    protein (Thiagarajan and Benedict, 1997).



  There is a need, therefore, for a method of preventing annexin loss from the vascular compartment into the extravascular compartment and urine, thereby prolonging antithrombotic activity following a single injection.



  SUMMARY OF THE INVENTION
The present invention provides compounds and methods for preventing arterial or venous thrombosis without increasing hemorrhage. A recombinant human annexin, preferably annexin V, is modified in such a way that its half-life in the vascular compartment is prolonged. This can be achieved in a variety of ways; three preferred embodiments are an annexin coupled to polyethylene glycol, a homopolymer or heteropolymer of annexin, and a fusion protein of annexin with another protein (e. g., the Fc portion of immunoglobulin). The modified annexin binds with high affinity to phosphatidylserine on the surface of activated platelets or injured cells, thereby preventing the binding of Gas6 as well as procoagulant proteins and the formation of a prothrombinase complex.

   Modified annexin therefore inhibits both the cellular and humoral mechanisms by which platelet aggregation is amplified, thereby preventing thrombosis.



   In one embodiment, the present invention provides an isolated modified annexin protein containing an annexin protein, preferably annexin V, coupled to polyethylene glycol (PEG). Preferably, at least two PEG chains are coupled to a single annexin molecule, with each PEG having a molecular weight of at least 5 kDa, more preferably at least 10 kDa, and most preferably at least 15 kDa. In an alternative embodiment, an isolated modified annexin protein contains an annexin protein coupled to at least one additional protein, such as an additional annexin protein (forming a homodimer) or the Fc portion of immunoglobulin. The additional protein preferably has a molecular weight of at least 30 kDa. Also provided by the present invention are pharmaceutical compositions containing an antithrombotically effective amount of any of the modified annexin proteins of the invention.



   In methods of the invention, the modified annexin is administered to a subject at risk of thrombosis in a pharmaceutical composition having an antithrombotically effective amount of any one of the modified annexin proteins of the present invention.



  For example, the pharmaceutical composition can be administered after an arterial thrombosis such as coronary thrombosis, cerebral thrombosis, or a transient cerebral ischemic attack. It can also be administered after a surgical operation associated with venous thrombosis. Additionally, it can be administered to subjects having conditions subject to arterial or venous thrombosis, such as diabetes, pregnancy, or parturition.



   Also provided by the present invention are an isolated nucleic acid molecule encoding a homodimer of annexin, a recombinant molecule containing at least a portion of the nucleic acid molecule, and a recombinant cell containing at least a portion of the nucleic acid molecule. The recombinant cell is cultured under suitable conditions in a method of the invention to produce a homodimer of annexin.



   The present invention also provides a method for screening for a modified annexin protein that modulates thrombosis using a thrombosis test system. The test system is contacted with a test modified annexin protein, after which the thrombolytic activity is assessed and compared with the activity of the system in the absence of the test modified annexin protein. Preferably, the activated partial thromboplastin time is measured. Also provided is a method for identifying a modified annexin protein by contacting activated platelets with a test modified annexin protein and assessing the platelet-binding and protein S-binding activity.



   Also provided by the present invention is a method for in vivo screening for a modified annexin protein. In this method, a thrombosis animal model is contacted with a test modified annexin protein, after which the in vivo anticoagulation activity and increase in hemorrhage of the test modified annexin protein is assessed. The anticoagulation activity and time are compared with the anticoagulation activity and time of annexin, and the amount of hemorrhage is compared with hemorrhage in the animal model in the absence of the test modified annexin protein.



  BRIEF DESCRIPTION OF THE FIGURES
FIG.   1    shows a DNA construct for making a homodimer of annexin V.



   FIG. 2 is a plot of clotting time of an in vitro clotting assay comparing the anticoagulant potency of recombinant human annexin V and pegylated recombinant human annexin V.



  DETAILED DESCRIPTION OF THE INVENTION
The present invention provides compounds and methods for preventing thrombosis in mammals without increasing hemorrhage. The invention relies in part on the recognition that the primary mechanisms of platelet aggregation are different from the mechanisms of amplifying platelet aggregation, which are required for the formation of an arterial or venous thrombus. By inhibiting thrombus formation but not primary platelet aggregation, thrombosis can be prevented without increasing hemorrhage.



   Compounds of the invention include any product containing annexin amino acid sequences that have been modified to increase the half-life of the product in humans or other mammals.   Where"amino    acid sequence"is recited herein to refer to an amino acid sequence of a naturally-occurring protein molecule,"amino acid sequence"and like terms, such as"polypeptide"or"protein,"are not meant to limit the amino acid sequence to the complete, native amino acid sequence associated with the recited proteins. The annexins are a family of homologous phospholipid-binding membrane proteins, of which ten represent distinct gene products expressed in mammals (Benz and Hofmann, 1997).

   Crystallographic analysis has revealed a common tertiary structure for all the family members so far studied, exemplified by annexin V (Huber et al., EMBO Journal 9: 3867 (1990), incorporated herein by reference). The core domain is a concave discoid structure that can be closely apposed to phospholipid membranes. It contains four subdomains, each consisting of a 70-amino-acid annexin repeat made up of five a-helices. The annexins also have a more hydrophilic tail domain that varies in length and amino acid sequence among the different annexins. The sequences of genes encoding annexins are well known (e. g.,
Funakoshi et   al.,    Biochemistry 26: 8087-8092 (1987) (annexin V), incorporated herein by reference).



   In the present invention, annexin proteins are modified to increase their halflife in humans or other mammals. In a preferred embodiment, the annexin protein is annexin V. One suitable modification of annexin is an increase in its effective size, which prevents loss from the vascular compartment into the extravascular compartment and urine, thereby prolonging antithrombotic activity following a single injection. Any increase in effective size that maintains a sufficient binding affinity with phosphatidylserine is within the scope of the present invention.



   In one embodiment of the invention, a modified annexin contains a recombinant human annexin protein coupled to polyethylene glycol (PEG) in such a way that the modified annexin is capable of performing the function of annexin in a phosphatidylserine (PS)-binding assay. The antithrombotic action of the intravenously administered annexin-PEG conjugate is prolonged as compared with that of the free annexin. The recombinant annexin protein coupled to PEG can be annexin V protein or another annexin protein.



   PEG consists of repeating units of ethylene oxide that terminate in hydroxyl groups on either end of a linear or, in some cases, branched chain. The size and molecular weight of the coupled PEG chain depend upon the number of ethylene oxide units it contains, which can be selected. For the present invention, any size of
PEG and number of PEG chains per annexin molecule can be used such that the halflife of the modified annexin is increased, relative to annexin, while preserving the function of binding of the modified molecule to PS. As stated above, sufficient binding includes binding that is diminished from that of the unmodified annexin, but still competitive with the binding of Gas6 and factors of the prothrombinase complex and therefore able to prevent thrombosis. The optimal molecular weight of the conjugated PEG varies with the number of PEG chains.

   In a most preferred embodiment, two PEG molecules of molecular weight of at least about 15 kDa each are coupled to each annexin molecule. The PEG molecules can be linear or branched. 



  The calcium-dependent binding of annexins to PS is affected not only by the size of the coupled PEG molecules, but also the sites on the protein to which PEG is bound.



  Optimal selection ensures that desirable properties are retained. Selection of PEG attachment sites is facilitated by knowledge of the three-dimensional structure of the molecule and by mutational and crystallographic analyses of the interaction of the molecule with phospholipid membranes (Campos et al., Biochemistry 37: 8004-8008 (1998), incorporated herein by reference).



   In the area of drug delivery, PEG derivatives have been widely used in covalent attachment (referred to as pegylation) to proteins to enhance solubility, as well as to reduce immunogenicity, proteolysis, and kidney clearance. The superior clinical efficacy of recombinant products coupled to PEG is well established. For example, PEG-interferon alpha-2a administered once weekly is significantly more effective against hepatitis C virus than three weekly doses of the free interferon (Heathcote et al., N. Engl. J. Med. 343: 1673-1680 (2000), incorporated herein by reference). Coupling to PEG has been used to prolong the half-life of recombinant proteins in vivo (Knauf et al., J. Biol.

   Chem. 266: 2796-2804 (1988), incorporated herein by reference), as well as to prevent the enzymatic degradation of recombinant proteins and to decrease the immunogenicity sometimes observed with homologous products (references in Hermanson, Bioconjugate techniques. New York, Academic
Press (1996), pp. 173-176, incorporated herein by reference).



   In another embodiment of the invention, the modified annexin protein is a polymer of annexin proteins that has an increased effective size. It is believed that the increase in effective size results in prolonged half-life in the vascular compartment and prolonged antithrombotic activity. One such modified annexin is a dimer of annexin V. Another such polymer is the heterotetramer of annexin II with   pll,    a member of the   S100    family of calcium-binding proteins. The binding of an S100 protein to an annexin increases the affinity of the annexin for   Ca2+.    The annexin homopolymer or heterotetramer can be produced by bioconjugate methods or recombinant methods, and be administered by itself or in a PEG-conjugated form. 



   In another embodiment of the invention, recombinant annexin is expressed with, or chemically coupled to, another protein such as the Fc portion of immunoglobulin. Such expression or coupling increases the effective size of the molecule, preventing the loss of annexin from the vascular compartment and prolonging its anticoagulant action.



   Preferably, a modified annexin protein of the invention is an isolated modified annexin protein. The modified annexin protein can contain annexin II, annexin V, or annexin VIII. In preferred embodiments, the protein is modified human annexin. In particularly preferred embodiments, the modified annexin contains recombinant human annexin. According to the present invention, an isolated or biologically pure protein is a protein that has been removed from its natural environment. As such, "isolated"and"biologically pure"do not necessarily reflect the extent to which the protein has been purified. An isolated modified annexin protein of the present invention can be obtained from its natural source, can be produced using recombinant
DNA technology, or can be produced by chemical synthesis.

   As used herein, an isolated modified annexin protein can be a full-length modified protein or any homologue of such a protein. It can also be (e. g., for a pegylated protein) a modified full-length protein or a modified homologue of such a protein.



   The minimal size of a protein homologue of the present invention is a size sufficient to be encoded by a nucleic acid molecule capable of forming a stable hybrid with the complementary sequence of a nucleic acid molecule encoding the corresponding natural protein. As such, the size of the nucleic acid molecule encoding such a protein homologue is dependent on nucleic acid composition and percent homology between the nucleic acid molecule and complementary sequence as well as upon hybridization conditions per se (e. g., temperature, salt concentration, and formamide concentration). The minimal size of such nucleic acid molecules is typically at least about 12 to about 15 nucleotides in length if the nucleic acid molecules are GC-rich and at least about 15 to about 17 bases in length if they are
AT-rich.

   As such, the minimal size of a nucleic acid molecule used to encode a protein homologue of the present invention is from about 12 to about 18 nucleotides in length. There is no limit on the maximal size of such a nucleic acid molecule in that the nucleic acid molecule can include a portion of a gene, an entire gene, or multiple genes or portions thereof. Similarly, the minimal size of an annexin protein homologue or a modified annexin protein homologue of the present invention is from about 4 to about 6 amino acids in length, with preferred sizes depending on whether a full-length, multivalent (i. e., fusion protein having more than one domain, each of which has a function) protein, or functional portions of such proteins are desired.



  Annexin and modified annexin homologues of the present invention preferably have activity corresponding to the natural protein, such as being able to perform the activity of the annexin protein in preventing thrombus formation.



   Annexin protein and modified annexin homologues can be the result of natural allelic variation or natural mutation. The protein homologues of the present invention can also be produced using techniques known in the art, including, but not limited to, direct modifications to the protein or modifications to the gene encoding the protein using, for example, classic or recombinant DNA techniques to effect random or targeted mutagenesis.



   Also preferred is a modified annexin protein containing an amino acid sequence that is at least about 75%, more preferably at least about 80%, more preferably at least about 85%, more preferably at least about 90%, more preferably at least about 95%, and most preferably at least about 98% identical to amino acid sequence SEQ ID NO : 3, SEQ   ID    NO : 6, or a protein encoded by an allelic variant of a nucleic acid molecule encoding a protein containing any of these sequences. Methods to determine percent identities between amino acid sequences and between nucleic acid sequences are known to those skilled in the art.

   Preferred methods to determine percent identities between sequences include computer programs such as the GCG
Wisconsin package (available from Accelrys Corporation), the   DNAsisTM    program (available from Hitachi Software, San Bruno, CA), the Vector NTI Suite (available from Informax, Inc., North Bethesda, MD), or the BLAST software available on the
NCBI website.



   In one embodiment, a preferred modified annexin protein includes an amino acid sequence of at least about 5 amino acids, preferably at least about 50 amino acids, more preferably at least about 100 amino acids, more preferably at least about 200 amino acids, more preferably at least about 250 amino acids, more preferably at least about 275 amino acids, more preferably at least about 300 amino acids, and most preferably at least about 319 amino acids or the full-length annexin protein, whichever is shorter. In another embodiment, preferred annexin proteins contain fulllength proteins, i. e., proteins encoded by full-length coding regions, or posttranslationally modified proteins thereof, such as mature proteins from which initiating methionine and/or signal sequences or"pro"sequences have been removed.



   A fragment of a modified annexin protein of the present invention preferably contains at least about 5 amino acids, more preferably at least about 10 amino acids, more preferably at least about 15 amino acids, more preferably at least about 20 amino acids, more preferably at least about 25 amino acids, more preferably at least about 30 amino acids, more preferably at least about 35 amino acids, more preferably at least about 40 amino acids, more preferably at least about 45 amino acids, more preferably at least about 50 amino acids, more preferably at least about 55 amino acids, more preferably at least about 60 amino acids, more preferably at least about 65 amino acids, more preferably at least about 70 amino acids, more preferably at least about 75 amino acids, more preferably at least about 80 amino acids, more preferably at least about 85 amino acids,

   more preferably at least about 90 amino acids, more preferably at least about 95 amino acids, and even more preferably at least about 100 amino acids in length.



   In one embodiment, a preferred isolated modified annexin protein of the present invention is a modified protein encoded by a nucleic acid molecule having the nucleic acid sequence SEQ   ID    NO : 4 or by an allelic variant of a nucleic acid molecule having this sequence. Alternatively, the modified annexin protein contains a protein encoded by a nucleic acid molecule having the nucleic acid sequence SEQ
ID NO : 1 or by an allelic variant of a nucleic acid molecule having this sequence.



   One embodiment of the present invention includes a non-native modified annexin protein encoded by a nucleic acid molecule that hybridizes under stringent hybridization conditions with an annexin gene. As used herein, stringent hybridization conditions refer to standard hybridization conditions under which nucleic acid molecules, including oligonucleotides, are used to identify molecules having similar nucleic acid sequences. Such standard conditions are disclosed, for example, in Sambrook et al., Molecular Cloning : A Laboratory Manual, Cold Spring
Harbor Labs Press (1989), incorporated herein by reference. Stringent hybridization conditions typically permit isolation of nucleic acid molecules having at least about 70% nucleic acid sequence identity with the nucleic acid molecule being used to probe in the hybridization reaction.

   Formulae to calculate the appropriate hybridization and wash conditions to achieve hybridization permitting 30% or less mismatch of nucleotides are disclosed, for example, in Meinkoth et   al.,    Anal.



  Biochem. 138: 267-284 (1984), incorporated herein by reference. In preferred embodiments, hybridization conditions will permit isolation of nucleic acid molecules having at least about 80% nucleic acid sequence identity with the nucleic acid molecule being used to probe. In more preferred embodiments, hybridization conditions will permit isolation of nucleic acid molecules having at least about 90% nucleic acid sequence identity with the nucleic acid molecule being used to probe. In more preferred embodiments, hybridization conditions will permit isolation of nucleic acid molecules having at least about 95% nucleic acid sequence identity with the nucleic acid molecule being used to probe.



   A preferred modified annexin protein includes a protein encoded by a nucleic acid molecule that is at least about 50 nucleotides and that hybridizes under conditions that preferably allow about 20% base pair mismatch, more preferably under conditions that allow about 15% base pair mismatch, more preferably under conditions that allow about 10% base pair mismatch, more preferably under conditions that allow about 5% base pair mismatch, and even more preferably under conditions that allow about 2% base pair mismatch with a nucleic acid molecule selected from the group consisting of SEQ ID NO :   1,    SEQ ID NO : 4, or a complement of either of these nucleic acid molecules.



   As used herein, an annexin gene includes all nucleic acid sequences related to a natural annexin gene such as regulatory regions that control production of the annexin protein encoded by that gene (such as, but not limited to, transcription, translation or post-translation control regions) as well as the coding region itself. In one embodiment, an annexin gene includes the nucleic acid sequence SEQ ID NO :   1.   



  It should be noted that since nucleic acid sequencing technology is not entirely errorfree, SEQ ID NO :   1    (as well as other sequences presented herein), at best, represents an apparent nucleic acid sequence of the nucleic acid molecule encoding an annexin protein of the present invention.



   In another embodiment, an annexin gene can be an allelic variant that includes a similar but not identical sequence to SEQ ID NO :   1.    An allelic variant of an annexin gene including SEQ ID NO :   1    is a gene that occurs at essentially the same locus (or loci) in the genome as the gene including SEQ ID NO :   1,    but which, due to natural variations caused by, for example, mutation or recombination, has a similar but not identical sequence. Allelic variants typically encode proteins having similar activity to that of the protein encoded by the gene to which they are being compared. Allelic variants can also comprise alterations in the 5'or 3'untranslated regions of the gene (e. g., in regulatory control regions).

   Allelic variants are well known to those skilled in the art and would be expected to be found within a given human since the genome is diploid and/or among a population comprising two or more humans.



   An isolated modified annexin protein of the present invention can be obtained from its natural source, can be produced using recombinant DNA technology, or can be produced by chemical synthesis. As used herein, an isolated modified annexin protein can contain a full-length protein or any homologue of such a protein.



  Examples of annexin and modified annexin homologues include annexin and modified annexin proteins in which amino acids have been deleted (e. g., a truncated version of the protein, such as a peptide or by a protein splicing reaction when an intein has been removed or two exteins are joined), inserted, inverted, substituted and/or derivatized (e. g., by glycosylation, phosphorylation, acetylation, methylation, myristylation, prenylation, palmitoylation, amidation and/or addition of glycerophosphatidyl inositol) such that the homologue includes at least one epitope capable of eliciting an immune response against an annexin protein. That is, when the homologue is administered to an animal as an immunogen, using techniques known to those skilled in the art, the animal will produce a humoral and/or cellular immune response against at least one epitope of an annexin protein.

   Annexin and modified annexin homologues can also be selected by their ability to selectively bind to immune serum. Methods to measure such activities are disclosed herein. Annexin and modified annexin homologues also include those proteins that are capable of performing the function of native annexin in a functional assay; that is, are capable of binding to phosphatidylserine or to activated platelets or exhibiting antithrombotic activity. Methods for such assays are described in the Examples section and elsewhere herein.



   A modified annexin protein of the present invention may be identified by its ability to perform the function of an annexin protein in a functional assay. The phrase "capable of performing the function of that protein in a functional assay"means that the protein or modified protein has at least about 10% of the activity of the natural protein in the functional assay. In other preferred embodiments, it has at least about 20% of the activity of the natural protein in the functional assay. In other preferred embodiments, it has at least about 30% of the activity of the natural protein in the functional assay. In other preferred embodiments, it has at least about 40% of the activity of the natural protein in the functional assay. In other preferred embodiments, it has at least about 50% of the activity of the natural protein in the functional assay.



  In other preferred embodiments, the protein or modified protein has at least about 60% of the activity of the natural protein in the functional assay. In more preferred embodiments, the protein or modified protein has at least about 70% of the activity of the natural protein in the functional assay. In more preferred embodiments, the protein or modified protein has at least about 80% of the activity of the natural protein in the functional assay. In more preferred embodiments, the protein or modified protein has at least about 90% of the activity of the natural protein in the functional assay. Examples of functional assays are described herein.



   An isolated protein of the present invention can be produced in a variety of ways, including recovering such a protein from a bacterium and producing such a protein recombinantly. One embodiment of the present invention is a method to produce an isolated modified annexin protein of the present invention using recombinant DNA technology. Such a method includes the steps of (a) culturing a recombinant cell containing a nucleic acid molecule encoding a modified annexin protein of the present invention to produce the protein and (b) recovering the protein therefrom. Details on producing recombinant cells and culturing thereof are presented below. The phrase"recovering the protein"refers simply to collecting the whole fermentation medium containing the protein and need not imply additional steps of separation or purification.

   Proteins of the present invention can be purified using a variety of standard protein purification techniques.



   Isolated proteins of the present invention are preferably retrieved in "substantially pure"form. As used herein,"substantially pure"refers to a purity that allows for the effective use of the protein in a functional assay.



  Modified Annexin Nucleic Acid Molecules or Genes
Another embodiment of the present invention is an isolated nucleic acid molecule capable of hybridizing under stringent conditions with a gene encoding a modified annexin protein such as a homodimer of annexin V. Such a nucleic acid molecule is also referred to herein as a modified annexin nucleic acid molecule.



  Particularly preferred is an isolated nucleic acid molecule that hybridizes under stringent conditions with a modified annexin gene. The characteristics of such genes are disclosed herein. In accordance with the present invention, an isolated nucleic acid molecule is a nucleic acid molecule that has been removed from its natural milieu (i. e., that has been subject to human manipulation). As such,"isolated"does not reflect the extent to which the nucleic acid molecule has been purified. An isolated nucleic acid molecule can include DNA, RNA, or derivatives of either DNA or RNA.



   As stated above, a modified annexin gene includes all nucleic acid sequences related to a natural annexin gene, such as regulatory regions that control production of an annexin protein encoded by that gene (such as, but not limited to, transcriptional, translational, or post-translational control regions) as well as the coding region itself.



  A nucleic acid molecule of the present invention can be an isolated modified annexin nucleic acid molecule or a homologue thereof. A nucleic acid molecule of the present invention can include one or more regulatory regions, full-length or partial coding regions, or combinations thereof. The minimal size of a modified annexin nucleic acid molecule of the present invention is the minimal size capable of forming a stable hybrid under stringent hybridization conditions with a corresponding natural gene.



  Annexin nucleic acid molecules can also include a nucleic acid molecule encoding a hybrid protein, a fusion protein, a multivalent protein or a truncation fragment.



   An isolated nucleic acid molecule of the present invention can be obtained from its natural source either as an entire (i. e., complete) gene or a portion thereof capable of forming a stable hybrid with that gene. As used herein, the phrase"at least a portion of'an entity refers to an amount of the entity that is at least sufficient to have the functional aspects of that entity. For example, at least a portion of a nucleic acid sequence, as used herein, is an amount of a nucleic acid sequence capable of forming a stable hybrid with the corresponding gene under stringent hybridization conditions.



   An isolated nucleic acid molecule of the present invention can also be produced using recombinant DNA technology (e. g., polymerase chain reaction (PCR) amplification, cloning, etc.) or chemical synthesis. Isolated modified annexin nucleic acid molecules include natural nucleic acid molecules and homologues thereof, including, but not limited to, natural allelic variants and modified nucleic acid molecules in which nucleotides have been inserted, deleted, substituted,   and/or    inverted in such a manner that such modifications do not substantially interfere with the ability of the nucleic acid molecule to encode a modified annexin protein of the present invention or to form stable hybrids under stringent conditions with natural nucleic acid molecule isolates.



   A modified annexin nucleic acid molecule homologue can be produced using a number of methods known to those skilled in the art (see, e. g., Sambrook et al., 1989). For example, nucleic acid molecules can be modified using a variety of techniques including, but not limited to, classic mutagenesis techniques and recombinant DNA techniques, such as site-directed mutagenesis, chemical treatment of a nucleic acid molecule to induce mutations, restriction enzyme cleavage of a nucleic acid fragment, ligation of nucleic acid fragments, polymerase chain reaction (PCR) amplification   and/or    mutagenesis of selected regions of a nucleic acid sequence, synthesis of oligonucleotide mixtures, and ligation of mixture groups to "build"a mixture of nucleic acid molecules and combinations thereof.

   Nucleic acid molecule homologues can be selected from a mixture of modified nucleic acids by screening for the function of the protein encoded by the nucleic acid (e. g., the ability of a homologue to elicit an immune response against an annexin protein and/or to function in a clotting assay, or other functional assay), and/or by hybridization with isolated annexin-encoding nucleic acids under stringent conditions.



   An isolated modified annexin nucleic acid molecule of the present invention can include a nucleic acid sequence that encodes at least one modified annexin protein of the present invention, examples of such proteins being disclosed herein. Although the phrase"nucleic acid molecule"primarily refers to the physical nucleic acid molecule and the phrase"nucleic acid sequence"primarily refers to the sequence of nucleotides on the nucleic acid molecule, the two phrases can be used interchangeably, especially with respect to a nucleic acid molecule, or a nucleic acid sequence, being capable of encoding a modified annexin protein.



   One embodiment of the present invention is a modified annexin nucleic acid molecule that is capable of hybridizing under stringent conditions to a nucleic acid strand that encodes at least a portion of a modified annexin protein or a homologue thereof or to the complement of such a nucleic acid strand. A nucleic acid sequence complement of any nucleic acid sequence of the present invention refers to the nucleic acid sequence of the nucleic acid strand that is complementary to (i. e., can form a complete double helix with) the strand for which the sequence is cited. It is to be noted that a double-stranded nucleic acid molecule of the present invention for which a nucleic acid sequence has been determined for one strand, that is represented by a
SEQ ID NO, also comprises a complementary strand having a sequence that is a complement of that SEQ   ID    NO.

   As such, nucleic acid molecules of the present invention, which can be either double-stranded or single-stranded, include those nucleic acid molecules that form stable hybrids under stringent hybridization conditions with either a given SEQ ID NO denoted herein and/or with the complement of that SEQ ID NO, which may or may not be denoted herein. Methods to deduce a complementary sequence are known to those skilled in the art.

   Preferred is a modified annexin nucleic acid molecule that includes a nucleic acid sequence having at least about 65 percent, preferably at least about 70 percent, more preferably at least about 75 percent, more preferably at least about 80 percent, more preferably at least about 85 percent, more preferably at least about 90 percent and even more preferably at least about 95 percent homology with the corresponding region (s) of the nucleic acid sequence encoding at least a portion of a modified annexin protein.



  Particularly preferred is a modified annexin nucleic acid molecule capable of encoding a homodimer of an annexin protein or homologue thereof.



   Preferred annexin nucleic acid molecules include SEQ   ID    NO : 4 and an allelic variants of SEQ ID NO : 4.



   Knowing a nucleic acid molecule of a modified annexin protein of the present invention allows one skilled in the art to make copies of that nucleic acid molecule as well as to obtain a nucleic acid molecule including additional portions of annexin protein-encoding genes (e. g., nucleic acid molecules that include the translation start site and/or transcription   and/or    translation control regions), and/or annexin nucleic acid molecule homologues. Knowing a portion of an amino acid sequence of an annexin protein of the present invention allows one skilled in the art to clone nucleic acid sequences encoding such an annexin protein.

   In addition, a desired modified annexin nucleic acid molecule can be obtained in a variety of ways including screening appropriate expression libraries with antibodies that bind to annexin proteins of the present invention; traditional cloning techniques using oligonucleotide probes of the present invention to screen appropriate libraries or DNA; and PCR amplification of appropriate libraries, or RNA or DNA using oligonucleotide primers of the present invention (genomic and/or   cDNA    libraries can be used).



   The present invention also includes nucleic acid molecules that are oligonucleotides capable of hybridizing, under stringent conditions, with complementary regions of other, preferably longer, nucleic acid molecules of the present invention that encode at least a portion of a modified annexin protein.



  Oligonucleotides of the present invention can be RNA, DNA, or derivatives of either.



  The minimal size of such oligonucleotides is the size required to form a stable hybrid between a given oligonucleotide and the complementary sequence on another nucleic acid molecule of the present invention. Minimal size characteristics are disclosed herein. The size of the oligonucleotide must also be sufficient for the use of the oligonucleotide in accordance with the present invention. Oligonucleotides of the present invention can be used in a variety of applications including, but not limited to, as probes to identify additional nucleic acid molecules, as primers to amplify or extend nucleic acid molecules or in therapeutic applications to modulate modified annexin production. Such therapeutic applications include the use of such oligonucleotides in, for example, antisense-, triplex formation-, ribozyme-and/or
RNA drug-based technologies.

   The present invention, therefore, includes such oligonucleotides and methods to modulate the production of modified annexin proteins by use of one or more of such technologies.



  Natural, Wild-Type Bacterial Cells and Recombinant Molecules and Cells
The present invention also includes a recombinant vector, which includes a modified annexin nucleic acid molecule of the present invention inserted into any vector capable of delivering the nucleic acid molecule into a host cell. Such a vector contains heterologous nucleic acid sequences, that is, nucleic acid sequences that are not naturally found adjacent to modified annexin nucleic acid molecules of the present invention. The vector can be either RNA or DNA, either prokaryotic or eukaryotic, and typically is a virus or a plasmid. Recombinant vectors can be used in the cloning, sequencing, and/or otherwise manipulating of modified annexin nucleic acid molecules of the present invention.

   One type of recombinant vector, herein referred to as a recombinant molecule and described in more detail below, can be used in the expression of nucleic acid molecules of the present invention. Preferred recombinant vectors are capable of replicating in the transformed cell. Preferred nucleic acid molecules to include in recombinant vectors of the present invention are disclosed herein.



   As heretofore disclosed, one embodiment of the present invention is a method to produce a modified annexin protein of the present invention by culturing a cell capable of expressing the protein under conditions effective to produce the protein, and recovering the protein. In an alternative embodiment, the method includes producing an annexin protein by culturing a cell capable of expressing the protein under conditions effective to produce the annexin protein, recovering the protein, and modifying the protein by coupling it to an agent that increases its effective size.



   In a preferred embodiment, the cell to culture is a natural bacterial cell, and modified annexin is isolated from these cells. In another embodiment, a preferred cell to culture is a recombinant cell that is capable of expressing the modified annexin protein, the recombinant cell being produced by transforming a host cell with one or more nucleic acid molecules of the present invention. Transformation of a nucleic acid molecule into a cell can be accomplished by any method by which a nucleic acid molecule can be inserted into the cell. Transformation techniques include, but are not limited to, transfection, electroporation, microinjection, lipofection, adsorption, and protoplast fusion. A recombinant cell may remain unicellular or may grow into a tissue, organ or a multicellular organism.

   Transformed nucleic acid molecules of the present invention can remain extrachromosomal or can integrate into one or more sites within a chromosome of the transformed (i. e., recombinant) cell in such a manner that their ability to be expressed is retained. Preferred nucleic acid molecules with which to transform a host cell are disclosed herein.



   Suitable host cells to transform include any cell that can be transformed and that can express the introduced modified annexin protein. Such cells are, therefore, capable of producing modified annexin proteins of the present invention after being transformed with at least one nucleic acid molecule of the present invention. Host cells can be either   untransformed    cells or cells that are already transformed with at least one nucleic acid molecule. Suitable host cells of the present invention can include bacterial, fungal (including yeast), insect, animal, and plant cells. Preferred host cells include bacterial cells, with E. coli cells being particularly preferred.



  Alternative preferred host cells are   untransformed    (wild-type) bacterial cells producing cognate modified annexin proteins, including attenuated strains with reduced pathogenicity, as appropriate.



   A recombinant cell is preferably produced by transforming a host cell with one or more recombinant molecules, each comprising one or more nucleic acid molecules of the present invention operatively linked to an expression vector containing one or more transcription control sequences. The phrase"operatively linked"refers to insertion of a nucleic acid molecule into an expression vector in a manner such that the molecule is able to be expressed when transformed into a host cell. As used herein, an expression vector is a DNA or RNA vector that is capable of transforming a host cell and of effecting expression of a specified nucleic acid molecule. Preferably, the expression vector is also capable of replicating within the host cell. Expression vectors can be either prokaryotic or eukaryotic, and are typically viruses or plasmids.



  Expression vectors of the present invention include any vectors that function (i. e., direct gene expression) in recombinant cells of the present invention, including in bacterial, fungal, insect, animal, and/or plant cells. As such, nucleic acid molecules of the present invention can be operatively linked to expression vectors containing regulatory sequences such as promoters, operators, repressors, enhancers, termination sequences, origins of replication, and other regulatory sequences that are compatible with the recombinant cell and that control the expression of nucleic acid molecules of the present invention. As used herein, a transcription control sequence includes a sequence that is capable of controlling the initiation, elongation, and termination of transcription.

   Particularly important transcription control sequences are those that control transcription initiation, such as promoter, enhancer, operator and repressor sequences. Suitable transcription control sequences include any transcription control sequence that can function in at least one of the recombinant cells of the present invention. A variety of such transcription control sequences are known to the art.



  Preferred transcription control sequences include those which function in bacterial, yeast, insect and mammalian cells, such as, but not limited to, tac, lac, tzp, trc, oxypro, omp/lpp, rrnB, bacteriophage lambda   (X)    (such as   RPL and XPR    and fusions that include such promoters), bacteriophage T7, T71ac, bacteriophage T3, bacteriophage
SP6, bacteriophage   SP01,    metallothionein, alpha mating factor, Pichia alcohol oxidase, alphavirus subgenomic promoters (such as Sindbis virus subgenomic promoters), baculovirus, Heliothis zea insect virus, vaccinia virus, herpesvirus, poxvirus, adenovirus, simian virus 40, retrovirus actin, retroviral long terminal repeat,
Rous sarcoma virus, heat shock,

   phosphate and nitrate transcription control sequences as well as other sequences capable of controlling gene expression in prokaryotic or eukaryotic cells. Additional suitable transcription control sequences include tissue specific promoters and enhancers as well as lymphokine-inducible promoters (e. g., promoters inducible by interferons or interleukins). Transcription control sequences of the present invention can also include naturally occurring transcription control sequences naturally associated with a DNA sequence encoding an annexin protein. A preferred transcription control sequence is the Kozak strong promotor and initiation sequence.



   Expression vectors of the present invention may also contain secretory signals (i. e., signal segment nucleic acid sequences) to enable an expressed annexin protein to be secreted from the cell that produces the protein. Suitable signal segments include an annexin protein signal segment or any heterologous signal segment capable of directing the secretion of an annexin protein, including fusion proteins, of the present invention. Preferred signal segments include, but are not limited to, tissue plasminogen activator (t-PA), interferon, interleukin, growth hormone, histocompatibility and viral envelope glycoprotein signal segments.



   Expression vectors of the present invention may also contain fusion sequences which lead to the expression of inserted nucleic acid molecules of the present invention as fusion proteins. Inclusion of a fusion sequence as part of a modified annexin nucleic acid molecule of the present invention can enhance the stability during production, storage and/or use of the protein encoded by the nucleic acid molecule. Furthermore, a fusion segment can function as a tool to simplify purification of a modified annexin protein, such as to enable purification of the resultant fusion protein using affinity chromatography. A preferred fusion segment that can be used for protein purification is the 8-amino acid peptide sequence asp-tyrlys-asp-asp-asp-asp-lys (SEQ   ID    NO : 9).



   A suitable fusion segment can be a domain of any size that has the desired function (e. g., increased stability and/or purification tool). It is within the scope of the present invention to use one or more fusion segments. Fusion segments can be joined to amino and/or carboxyl termini of an annexin protein. Another type of preferred fusion protein is a fusion protein wherein the fusion segment connects two or more annexin proteins or modified annexin proteins. Linkages between fusion segments and annexin proteins can be constructed to be susceptible to cleavage to enable straightforward recovery of the annexin or modified annexin proteins.

   Fusion proteins are preferably produced by culturing a recombinant cell transformed with a fusion nucleic acid sequence that encodes a protein including the fusion segment attached to either the carboxyl and/or amino terminal end of an annexin protein.



   A recombinant molecule of the present invention is a molecule that can include at least one of any nucleic acid molecule heretofore described operatively linked to at least one of any transcription control sequence capable of effectively regulating expression of the nucleic acid molecules in the cell to be transformed. A preferred recombinant molecule includes one or more nucleic acid molecules of the present invention, with those that encode one or more modified annexin proteins being particularly preferred. Preferred recombinant molecules of the present invention and their production are described in the Examples section. Similarly, a preferred recombinant cell includes one or more nucleic acid molecules of the present invention, with those that encode one or more modified annexin proteins.

   Preferred recombinant cells of the present invention include those disclosed in the Examples section.



   It may be appreciated by one skilled in the art that use of recombinant DNA technologies can improve expression of transformed nucleic acid molecules by manipulating, for example, the number of copies of the nucleic acid molecules within a host cell, the efficiency with which those nucleic acid molecules are transcribed, the efficiency with which the resultant transcripts are translated, and the efficiency of post-translational modifications.

   Recombinant techniques useful for increasing the expression of nucleic acid molecules of the present invention include, but are not limited to, operatively linking nucleic acid molecules to high-copy number plasmids, integration of the nucleic acid molecules into one or more host cell chromosomes, addition of vector stability sequences to plasmids, substitutions or modifications of transcription control signals (e. g., promoters, operators, enhancers), substitutions or modifications of translational control signals (e. g., ribosome binding sites, Shine
Dalgarno sequences), modification of nucleic acid molecules of the present invention to correspond to the codon usage of the host cell, deletion of sequences that destabilize transcripts, and use of control signals that temporally separate recombinant cell growth from recombinant protein production during fermentation.

   The activity of an expressed recombinant protein of the present invention may be improved by fragmenting, modifying, or derivatizing the resultant protein.



   In accordance with the present invention, recombinant cells can be used to produce annexin or modified annexin proteins of the present invention by culturing such cells under conditions effective to produce such a protein, and recovering the protein. Effective conditions to produce a protein include, but are not limited to, appropriate media, bioreactor, temperature, pH and oxygen conditions that permit protein production. An appropriate, or effective, medium refers to any medium in which a cell of the present invention, when cultured, is capable of producing an annexin or modified annexin protein. Such a medium is typically an aqueous medium comprising assimilable carbohydrate, nitrogen and phosphate sources, as well as appropriate salts, minerals, metals and other nutrients, such as vitamins. The medium may comprise complex, nutrients or may be a defined minimal medium.



   Cells of the present invention can be cultured in conventional fermentation   bioreactors,    which include, but are not limited to, batch, fed-batch, cell recycle, and continuous fermentors. Culturing can also be conducted in shake flasks, test tubes, microtiter dishes, and petri plates. Culturing is carried out at a temperature, pH and oxygen content appropriate for the recombinant cell. Such culturing conditions are well within the expertise of one of ordinary skill in the art.



   Depending on the vector and host system used for production, resultant modified annexin proteins may either remain within the recombinant cell; be secreted into the fermentation medium; be secreted into a space between two cellular membranes, such as the periplasmic space in E. coli ; or be retained on the outer surface of a cell or viral membrane. Methods to purify such proteins are disclosed in the Examples section. 



  Antibodies
The present invention also includes isolated anti-modified annexin antibodies and their use. An anti-modified annexin antibody is an antibody capable of selectively binding to a modified annexin protein. Isolated antibodies are antibodies that have been removed from their natural milieu. The term"isolated"does not refer to the state of purity of such antibodies. As such, isolated antibodies can include antisera containing such antibodies, or antibodies that have been purified to varying degrees. As used herein, the   term"selectively    binds to"refers to the ability of such antibodies to preferentially bind to the protein against which the antibody was raised (i. e., to be able to distinguish that protein from unrelated components in a mixture).



  Binding affinities, commonly expressed as equilibrium association constants, typically range from about   103      M-1    to about   10l2 M-l.    Binding can be measured using a variety of methods known to those skilled in the art including immunoblot assays, immunoprecipitation assays, radioimmunoassays, enzyme immunoassays (e. g.,
ELISA), immunofluorescent antibody assays and immunoelectron microscopy; see, e. g., Sambrook et al., 1989.



   Antibodies of the present invention can be either polyclonal or monoclonal antibodies. Antibodies of the present invention include functional equivalents such as antibody fragments and genetically-engineered antibodies, including single chain antibodies, that are capable of selectively binding to at least one of the epitopes of the protein used to obtain the antibodies. Antibodies of the present invention also include chimeric antibodies that can bind to more than one epitope. Preferred antibodies are raised in response to proteins that are encoded, at least in part, by a modified annexin nucleic acid molecule of the present invention.



   Anti-modified annexin antibodies of the present invention include antibodies raised in an animal administered a modified annexin. Anti-modified annexin antibodies of the present invention also include antibodies raised in an animal against one or more modified annexin proteins of the present invention that are then recovered from the cell using techniques known to those skilled in the art. Yet additional antibodies of the present invention are produced recombinantly using techniques as heretofore disclosed for modified annexin proteins of the present invention. Antibodies produced against defined proteins can be advantageous because such antibodies are not substantially contaminated with antibodies against other substances that might otherwise cause interference in a diagnostic assay or side effects if used in a therapeutic composition.



   Anti-modified annexin antibodies of the present invention have a variety of uses that are within the scope of the present invention. Anti-modified annexin antibodies can be used as tools to screen expression libraries and/or to recover desired proteins of the present invention from a mixture of proteins and other contaminants.



   A preferred anti-modified annexin antibody of the present invention can selectively bind to a modified annexin protein.



  Therapeutic Methods
Any of the above-described modified annexin proteins is used in methods of the invention to treat arterial or venous thrombosis caused by any medical procedure or condition. Generally, the therapeutic agents used in the invention are administered to an animal in an effective amount. Generally, an effective amount is an amount effective either (1) to reduce the symptoms of the disease sought to be treated or (2) to induce a pharmacological change relevant to treating the disease sought to be treated.



   For thrombosis, an effective amount includes an amount effective to exert prolonged antithrombotic activity without substantially increasing the risk of hemorrhage or to increase the life expectancy of the affected animal. As used herein, prolonged antithrombotic activity refers to the time of activity of the modified annexin protein with respect to the time of activity of the same amount (molar) of an unmodified annexin protein. Preferably, antithrombotic activity is prolonged by at least about a factor of two, more preferably by at least about a factor of five, and most preferably by at least about a factor of ten. Preferably, the effective amount does not substantially increase the risk of hemorrhage compared with the hemorrhage risk of the same subject to whom the modified annexin has not been administered.



  Preferably, the hemorrhage risk is very small and, at most, below that provided by alternative antithrombotic treatments available in the prior art. Therapeutically effective amounts of the therapeutic agents can be any amount or dose sufficient to bring about the desired antithrombotic effect and depends, in part, on the condition, type, and location of the thrombus, the size and condition of the patient, as well as other factors known to those skilled in the art. The dosages can be given as a single dose, or as several doses, for example, divided over the course of several weeks.



   Administration preferably occurs by bolus injection or by intravenous infusion, either after thrombosis to prevent further thrombosis or under conditions in which the subject is susceptible to or at risk of thrombosis.



   The therapeutic agents of the present invention can be administered by any suitable means, including, for example, parenteral or local administration, such as intravenous or subcutaneous injection, or by aerosol. A therapeutic composition can be administered in a variety of unit dosage forms depending upon the method of administration. Preferred delivery methods for a therapeutic composition of the present invention include intravenous administration and local administration by, for example, injection. For particular modes of delivery, a therapeutic composition of the present invention can be formulated in an excipient of the present invention. A therapeutic reagent of the present invention can be administered to any animal, preferably to mammals, and more preferably to humans.



   One suitable administration time occurs following coronary thrombosis, thereby preventing the recurrence of thrombosis without substantially increasing the risk of hemorrhage. Bolus injection of the modified annexin is preferably performed soon after thrombosis, e. g., before admission to hospital. The modified annexin can be administered in conjunction with a thrombolytic therapeutic such as tissue plasminogen activator, urokinase, or a bacterial enzyme.



   Methods of use of modified annexin proteins of the present invention include methods to treat cerebral thrombosis, including overt cerebral thrombosis or transient cerebral ischemic attacks, by administering an effective amount of modified annexin protein to a patient in need thereof. Transient cerebral ischemic attacks frequently precede full-blown strokes. The modified annexin can also be administered to diabetic and other patients who are at increased risk for thrombosis in peripheral arteries. Accordingly, the present invention provides a method for reducing the risk of thrombosis in a patient having an increased risk for thrombosis including administering an effective amount of a modified   annexirrprotein    to a patient in need thereof.

   For an adult patient, the modified annexin can be administered intravenously or as a bolus in the dosage range of about 1 to about 100 mg.



   The present invention also provides a method for decreasing the risk of venous thrombosis associated with some surgical procedures, such as hip and knee arthroplasties, by administering an effective amount of a modified annexin protein of the present invention to a patient in need thereof. The modified annexin treatment can prevent thrombosis without increasing hemorrhage into the operating field. In another embodiment, the present invention provides a method for preventing thrombosis associated with pregnancy and parturition without increasing hemorrhage, by administering an effective amount of a modified annexin protein of the present invention to a patient in need thereof.

   In a further embodiment, the present invention provides a method for the treatment of recurrent venous thrombosis, by administering an effective amount of a modified annexin protein of the present invention to a patient in need thereof. For an adult patient, the modified annexin can be administered intravenously as a bolus in the dosage range of about 1 to about 100 mg.



   The present invention also provides a method of screening for a modified annexin protein that modulates thrombosis, by contacting a thrombosis test system with at least one test modified annexin protein under conditions permissive for thrombosis, and comparing the antithrombotic activity in the presence of the test modified annexin protein with the antithrombotic activity in the absence of the test modified annexin protein, wherein a change in the antithrombotic activity in the presence of the test modified annexin protein is indicative of a modified annexin protein that modulates thrombotic activity. In a preferred embodiment, the thrombosis test system is a system for measuring activated partial thromboplastin time. Also included within the scope of the present invention are modified annexin proteins that modulate thrombosis as identified by this method. 



   The present invention also provides a method for identifying a modified annexin protein for annexin activity, including contacting activated platelets with at least one test modified annexin protein under conditions permissive for binding, and comparing the test modified annexin-binding activity and protein S-binding activity of the platelets in the presence of the test modified annexin protein with the annexinbinding activity and protein S-binding activity in the presence of unmodified annexin protein, whereby a modified annexin protein with annexin activity may be identified.



  Also included within the scope of the invention are modified annexin proteins identified by the method.



   In an additional embodiment, the present invention provides a method of screening for a modified annexin protein that modulates thrombosis, by contacting an in vivo thrombosis test system with at least one test modified annexin protein under conditions permissive for thrombosis, and comparing the antithrombotic activity in the presence of the test modified annexin protein with the antithrombotic activity in the absence of the test modified annexin protein. A change in the antithrombotic activity in the presence of the test modified annexin protein is indicative of a modified annexin protein that modulates thrombotic activity.

   Additionally, the time over which antithrombotic activity is sustained in the presence of the test modified annexin protein is compared with a time of antithrombotic activity in the presence of unmodified annexin to determine the prolongation of antithrombotic activity associated with the test modified annexin protein. The extent of hemorrhage in the presence of the test modified annexin protein is assessed, e. g., by measuring tail bleeding time, and compared with the extent of hemorrhage in the absence of the test modified annexin protein. In a preferred embodiment, the in vivo thrombosis test system is a mouse model of photochemically-induced thrombus in cremaster muscles.



  Also included within the scope of the present invention are modified annexin proteins that modulate thrombosis as identified by this method.



   In a further embodiment, the therapeutic agents of the present invention are useful for gene therapy. As used herein, the phrase"gene therapy"refers to the transfer of genetic material (e. g., DNA or RNA) of interest into a host to treat or prevent a genetic or acquired disease or condition. The genetic material of interest encodes a product (e. g., a protein polypeptide, peptide or functional RNA) whose production in vivo is desired. For example, the genetic material of interest can encode a hormone, receptor, enzyme or (poly) peptide of therapeutic value. In a specific embodiment, the subject invention utilizes a class of lipid molecules for use in nonviral gene therapy which can complex with nucleic acids as described in Hughes et al., U. S.

   Patent No. 6,169,078, incorporated herein by reference, in which a disulfide linker is provided between a polar head group and a lipophilic tail group of a lipid.



   These therapeutic compounds of the present invention effectively complex with DNA and facilitate the transfer of DNA through a cell membrane into the intracellular space of a cell to be transformed with heterologous DNA. Furthermore, these lipid molecules facilitate the release of heterologous DNA in the cell cytoplasm thereby increasing gene transfection during gene therapy in a human or animal.



   Cationic lipid-polyanionic macromolecule aggregates may be formed by a variety of methods known in the art. Representative methods are disclosed by Felgner et al., Proc. Natl. Acad. Sci. USA 86: 7413-7417 (1987); Eppstein et al., U. S. Patent
No. 4,897,355; Behr et al., Proc. Natl. Acad. Sci. USA 86: 6982-6986 (1989);
Bangham et al., J. Mol. Biol. 23: 238-252 (1965); Olson et al., Biochim. Biophys.



  Acta 557: 9 (1979); Szoka, et al., Proc. Natl. Acad. Sci. 75: 4194 (1978); Mayhew et al., Biochim. Biophys. Acta 775: 169 (1984); Kim et al., Biochim.   Biophys.    Acta 728: 339 (1983); and Fukunaga et al., Endocrinol. 115: 757 (1984), all incorporated herein by reference. In general, aggregates may be formed by preparing lipid particles consisting of either (1) a cationic lipid or (2) a cationic lipid mixed with a colipid, followed by adding a polyanionic macromolecule to the lipid particles at about room temperature (about 18 to   26  C.).    In general, conditions are chosen that are not conducive to deprotection of protected groups. In one embodiment, the mixture is then allowed to form an aggregate over a period of about 10 minutes to about 20 hours, with about 15 to 60 minutes most conveniently used.

   Other time periods may be appropriate for specific lipid types. The complexes may be formed over a longer period, but additional enhancement of transfection efficiency will not usually be gained by a longer period of complexing. 



   The compounds and methods of the subject invention can be used to   intracellularly    deliver a desired molecule, such as, for example, a polynucleotide, to a target cell. The desired polynucleotide can be composed of DNA or RNA or analogs thereof. The desired polynucleotides delivered using the present invention can be composed of nucleotide sequences that provide different functions or activities, such as nucleotides that have a regulatory function, e. g., promoter sequences, or that encode a polypeptide. The desired polynucleotide can also provide nucleotide sequences that are antisense to other nucleotide sequences in the cell. For example, the desired polynucleotide when transcribed in the cell can provide a polynucleotide that has a sequence that is antisense to other nucleotide sequences in the cell.

   The antisense sequences can hybridize to the sense strand sequences in the cell.



  Polynucleotides that provide antisense sequences can be readily prepared by the ordinarily skilled artisan. The desired polynucleotide delivered into the cell can also comprise a nucleotide sequence that is capable of forming a triplex complex with double-stranded DNA in the cell.



   The following examples illustrate the preparation of modified annexin proteins of the invention and in vitro and in vivo assays for anticoagulant activity of modified annexin proteins. It is to be understood that the invention is not limited to the exemplary work described or to the specific details set forth in the examples.



  EXAMPLES
Example 1: Modified Annexin Preparation
Annexins can be purified from human tissues or produced by recombinant technology. For instance, annexin V can be purified from human placentas as described by Funakoshi et al. (1987). Examples of recombinant products are the expression of annexin II and annexin V in   Escherichia    coli (Kang, H.-M., Trends
Cardiovasc. Med. 9: 92-102 (1999); Thiagarajan and Benedict, 1997,2000). A rapid and efficient purification method for recombinant annexin V, based on   Ca2+-enhanced    binding to phosphatidylserine-containing liposomes and subsequent elution by EDTA, has been described by Berger, FEBS Lett. 329: 25-28 (1993). This procedure can be improved by the use of phosphatidylserine coupled to a solid phase support. 



   Annexins can be coupled to polyethylene glycol (PEG) by any of several wellestablished procedures (reviewed by Hermanson, 1996) in a process referred to as pegylation. The present invention includes chemically-derivatized annexin molecules having mono-or   poly- (e.    g., 2-4) PEG moieties. Methods for preparing a pegylated annexin generally include the steps of (a) reacting the annexin with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the annexin becomes attached to one or more PEG groups and (b) obtaining the reaction product or products. In general, the optimal reaction conditions for the reactions must be determined case by case based on known parameters and the desired result.

   Furthermore, the reaction may produce different products having a different number of PEG chains, and further purification may be needed to obtain the desired product.



   Conjugation of PEG to annexin V can be performed using the EDC plus sulfo
NHS procedure. EDC   (1-ethyl-3- (3-dimethylaminopropyl)-carbodiimide    hydrochloride) is used to form active ester groups with carboxylate groups using   sulfo-NHS    (N-hydroxysulfosuccinamide). This increases the stability of the active intermediate, which reacts with an amine to give a stable amide linkage. The conjugation can be carried out as described in Hermanson, 1996.



   Bioconjugate methods can be used to produce homopolymers or heteropolymers of annexin; methods are reviewed by Hermanson, 1996.



  Recombinant methods can also be used to produce fusion proteins, e. g., annexin expressed with the Fc portion of immunoglobulin or another protein. The heterotetramer of annexin II with   P11    has also been produced in E. coli (Kang et al., 1999). All of these procedures increase the molecular weight of annexin and have the potential to increase the half-life of annexin in the circulation and prolong its anticoagulant effect.



   A homodimer of annexin V can be produced using a DNA construct shown schematically in FIG. 1 (5'-3'sense strand) (SEQ ID NO : 4) and coding for an amino acid sequence represented by SEQ ID NO : 6. In this example, the annexin V gene is cloned into the expression vector   pCMV    FLAG 2 (available from Sigma-Aldrich) at   EcoRI    and   BglII    sites. The exact sequences prior to and after the annexin V sequence are unknown and denoted   as"x".    It is therefore necessary to sequence the construct prior to modification to assure proper codon alignment. The   pCMV    FLAG 2 vector comes with a strong promotor and initiation sequence (Kozak) and start site (ATG) built in.

   The start codon before each annexin V gene must therefore be removed, and a strong stop for tight expression should be added at the terminus of the second annexin V gene. The vector also comes with an 8-amino acid peptide sequence that can be used for protein purification (asp-tyr-lys-asp-asp-asp-asp-lys) (SEQ ID NO : 9).



  A   14-amino    acid spacer with glycine-serine swivel ends allows optimal rotation between tandem genes. Addition of restriction sites   PvuII    and ScaI allow removal of the linker if necessary. Addition of a protease site allows cleavage of tandem proteins following expression.   PreScissionTM    protease is available from Amersham Pharmacia
Biotech and can be used to cleave tandem proteins.



  Annexin V has the following amino acid sequence:
MALRGTVTDFSGFDGRADAEVLRKAMKGLGTDEDSILNLLTARSNAQRQQI
AEEFKTLFGRDLVNDMKSELTGKFEKLIVALMKPSRLYDAYELKHAKLGAGT
DEKVLTEIIASRTPEELRAIKQAYEEEYGSNLEDDVVGDTSGYYQRMLVVLLQ
ANRDPDTAIDDAQVELDAQALFQAGELKWGTDEEKFITILGTRSVSHLRRVF
DKYMTISGFQIEETIDRETSGNLENLLLAVVKSIRSIPAYLAETLYYAMKGAGT   
DDHTLIRVIVSRSEIDLFNIRKEFRKNFATSLYSMIKGDTSGDYKKALLLLCGG   
EDD (*stop) (SEQ ID NO :

   3)
The nucleotide sequence of annexin V, inserted as indicated in the DNA construct illustrated in FIG.   1,    is as follows:
ATGGCCCTGCGCGGCACCGTGACCGACTTCTCCGGCTTCGACGGCCGCGC
CGACGCCGAGGTGCTGCGCAAGGCCATGAAGGGCCTGGGCACCGACGAG
GACTCCATCCTGAACCTGCTGACCGCCCGCTCCAACGCCCAGCGCCAGCA
GATCGCCGAGGAGTTCAAGACCCTGTTCGGCCGCGACCTGGTGAACGACA
TGAAGTCCGAGCTGACCGGCAAGTTCGAGAAGCTGATCGTGGCCCTGATG   AAGCCCTCCCGCCTGTACGACGCCTACGAGCTGAAGCACGCCAAGCTGGG   
CGCCGGCACCGACGAGAAGGTGCTGACCGAGATCATCGCCTCCCGCACCC
CCGAGGAGCTGCGCGCCATCAAGCAGGCCTACGAGGAGGAGTACGGCTC 
CAACCTGGAGGACGACGTGGTGGGCGACACCTCCGGCTACTACCAGCGCA   TGCTGGTGGTGCTGCTGCAGGCCAACCGCGACCCCGACACCGCCATCGAC   
GACGCCCAGGTGGAGCTGGACGCCCAGGCCCTGTTCCAGGCCGGCGAGCT   GAAGTGGGGCACCGACGAGGAGAAGTTCATCACCATCCTGGGCACCCGCT   

  
CCGTGTCCCACCTGCGCCGCGTGTTCGACAAGTACATGACCATCTCCGGCT
TCCAGATCGAGGAGACCATCGACCGCGAGACCTCCGGCAACCTGGAGAAC
CTGCTGCTGGCCGTGGTGAAGTCCATCCGCTCCATCCCCGCCTACCTGGCC
GAGACCCTGTACTACGCCATGAAGGGCGCCGGCACCGACGACCACACCCT
GATCCGCGTGATCGTGTCCCGCTCCGAGATCGACCTGTTCAACATCCGCAA
GGAGTTCCGCAAGAACTTCGCCACCTCCCTGTACTCCATGATCAAGGGCG
ACACCTCCGGCGACTACAAGAAGGCCCTGCTGCTGCTGTGCGGCGGCGAG
GACGACTGA (SEQ ID NO : 1)
Example 2 : In Vitro and In Vivo Assays
In vitro assays determine the ability of modified annexin proteins to bind to activated platelets. Annexin V binds to platelets, and this binding is markedly increased in vitro by activation of the platelets with thrombin (Thiagarajan and Tait, 1990; Sun et al., 1993).

   Preferably, the modified annexin proteins of the present invention are prepared in such a way that perform the function of annexin in that they bind to platelets and prevent protein S from binding to platelets (Sun et al., 1993).



  The modified annexin proteins also perform the function of exhibiting the same anticoagulant activity in vitro that unmodified annexin proteins exhibit. A method for measuring the clotting time is the activated partial thromboplastin time (Fritsma, in
Hemostasis and thrombosis in the clinical laboratory (Corriveau, D. M. and Fritsma,
G. A., eds) J. P. Lipincott Co., Philadelphia (1989), pp. 92-124, incorporated herein by reference).



   In vivo assays determine the antithrombotic activity of annexin proteins.



  Annexin V has been shown to decrease venous thrombosis induced by a laser or   photochemically    in rats (Romisch et   al.,    1991). The maximal anticoagulant effect was observed between 15 and 30 minutes after intravenous administration of annexin V, as determined functionally by thromboelastography. The modified annexin proteins of the present invention preferably show more prolonged activity in such a model than unmodified annexin. Annexin V was also found to decrease fibrin accretion in a rabbit model of jugular vein thrombosis (Van Ryn-McKenna   etal.,    1993). Air injection was used to remove the endothelium, and annexin V was shown to bind to the treated vein but not to the control contralateral vein. Decreased fibrin accumulation in the injured vein was not associated with systemic anticoagulation.



   Heparin did not inhibit fibrin accumulation in the injured vein. The modified annexin proteins of the present invention preferably perform the function of annexin in this model of venous thrombosis. A rabbit model of arterial thrombosis was used by
 Thiagarajan and Benedict, 1997. A partially occlusive thrombus was formed in the left carotid artery by application of an electric current. Annexin V infusion strongly inhibited thrombosis as manifested by measurements of blood flow, thrombus weight, labeled fibrin deposition and labeled platelet accumulation. Recently, a mouse model of photochemically-induced thrombus in cremaster muscles was introduced (Vollmar et al. Thromb. Haemost. 85: 160-164 (2001), incorporated herein by reference). Using this technique, thrombosis can be induced in any desired artery or vein.

   The modified annexin proteins of the present invention preferably perform the function of annexin in such models, even when administered by bolus injection.



  Example 3
The anticoagulant ability of human recombinant annexin V and pegylated human recombinant annexin V were compared in vitro.



   Annexin V production. The polymerase chain reaction was used to amplify the
   cDNA    from the initiator methionine to the stop codon with specific oligonucleotide primers from a human placental   cDNA    library. The forward primer was 5'
 ACCTGAGTAGTCGCCATGGCACAGGTTCTC-3' (SEQ ID NO : 7) and the reverse primer was 5'-CCCGAATTCACGTTAGTCATCTTCTCCACAGAGCAG-3' (SEQ
ID NO : 8). The amplified   1.      1-kb    fragment was digested with Nco I and Eco RI and ligated into the prokaryotic expression vector   pTRC    99A. The ligation product was used to transform competent Escherichia coli strain JM 105 and sequenced.



   Recombinant annexin V was isolated from the bacterial lysates as described by Berger et al., 1993, with some modification. An overnight culture of E. coli JM 
105 transformed with   pTRC    99A-annexin V was expanded 50-fold in fresh Luria
Bertrani medium containing 100 mg/L ampicillin. After 2 hours, isopropyl thiogalactopyranoside was added to a final concentration of 1 mmol/L. After 16 hours of induction, the bacteria were pelleted at 3500g for 15 minutes at   4 C.    The bacterial pellet was suspended in TBS, pH 7.5, containing 1 mmol/L PMSF, 5 mmol/L EDTA, and 6 mol/L urea. The bacterial suspension was sonicated with an ultrasonic probe at a setting of 6 on ice for 3 minutes.

   The lysate was centrifuged at
10,000g for 15 minutes, and the supernatant was dialyzed twice against 50 vol TBS containing 1 mmol/L EDTA and once against 50 vol TBS.



     Multilamellar    liposomes were prepared by dissolving phosphatidylserine, lyophilized bovine brain extract, cholesterol, and dicetylphosphate in chloroform in a molar ration of 10: 15: 1 and dried in a stream of nitrogen in a conical flask. TBS (5 mL) was added to the flask and agitated vigorously in a vortex mixer for 1 minute.



  The liposomes were washed by centrifugation at 3500g for 15 minutes, then incubated with the bacterial extract, and calcium chloride was added to a final concentration of 5 mmol/L. After 15 minutes of incubation at   37 C,    the liposomes were sedimented by centrifugation at 10,000g for 10 minutes, and the bound annexin V was eluted with 10 mmol/L EDTA. The eluted annexin V was concentrated by Amicon ultrafiltration and loaded onto a Sephacryl S 200 column. The annexin V was recovered in the included volume, whereas most of the liposomes were in the void volume. Fractions containing annexin V were pooled and dialyzed in 10 mmol/L Tris and 2 mmol/L
EDTA, pH 8.1, loaded onto an anion exchange column, and eluted with a linear gradient of 0 to 200 mmol/L   NaCI    in the same buffer.

   The purified preparation showed a single band in   SDS-PAGE    under reducing conditions.



   The annexin V produced as above was pegylated using the method of
Hermanson, 1996, as described above.



  Anti-coagulation assays. Prolongation of the clotting time (activated partial thromboplastin time) induced by annexin V and pegylated annexin V were compared.



  Activated partial thromboplastin times were assayed with citrated normal pooled plasma as described in Fritsma, 1989. Using different concentrations of annexin V and pegylated annexin V, produced as described above, dose-response curves for prolongation of clotting times were obtained. Results are shown in FIG. 2, a plot of clotting time versus annexin V and pegylated annexin V dose. As shown in the figure, the anticoagulant potency of the recombinant human annexin V and the pegylated recombinant human annexin V are substantially equivalent. The small difference observed is attributable to the change in molecular weight after pegylation.



  This experiment validates the assertion made herein that pegylation of annexin V can be achieved without significantly reducing its antithrombotic effects.



   It should be noted that the foregoing description is only illustrative of the invention. Various alternatives and modifications can be devised by those skilled in the art without departing from the invention. Accordingly, the present invention is intended to embrace all such alternatives, modifications and variances which fall within the scope of the disclosed invention.

Claims

CLAIMS What is claimed is: 1. An isolated modified annexin protein comprising an annexin protein coupled to polyethylene glycol.
2. The isolated modified annexin protein of claim 1, wherein said polyethylene glycol has a molecular weight of at least 10 kDa.
3. The isolated modified annexin protein of claim 2, wherein said polyethylene glycol has a molecular weight of at least 30 kDa.
4. The isolated modified annexin protein of claim 1, wherein each modified annexin protein comprises at least two polyethylene glycol chains.
5. The isolated modified annexin protein of claim 1, wherein said annexin protein is an annexin V protein.
6. The isolated modified annexin protein of claim 5, wherein said annexin V protein comprises an amino acid sequence selected from the group consisting of : a) an amino acid sequence selected from the group consisting of SEQ ID NO : 3 and SEQ ID NO : 6; and b) an amino acid sequence selected from the group consisting of an amino acid sequence having at least 95% sequence identity to an amino acid sequence of a).
7. A pharmaceutical composition comprising an antithrombotically effective amount of the modified annexin protein of claim 1.
8. A method of treating a subject at risk of thrombosis, comprising administering to said subject a pharmaceutical composition comprising an antithrombotically effective amount of the modified annexin protein of claim 1.
9. The method of claim 8, wherein said pharmaceutical composition is administered after coronary thrombosis.
10. The method of claim 8, wherein said pharmaceutical composition is administered after a condition selected from the group consisting of overt cerebral thrombosis and transient cerebral ischemic attack.
11. The method of claim 8, wherein said pharmaceutical composition is administered after a surgical operation associated with venous thrombosis.
12. The method of claim 8, wherein said subject is diabetic and said thrombosis is arterial thrombosis.
13. The method of claim 8, wherein said pharmaceutical composition is administered during a condition selected from the group consisting of pregnancy and parturition.
14. An isolated modified annexin protein comprising an annexin protein coupled to an additional protein.
15. The isolated modified annexin protein of claim 14, wherein said additional protein is the Fc portion of immunoglobulin.
16. The isolated modified annexin protein of claim 14, wherein said additional protein has a molecular weight of at least 30 kDa.
17. The isolated modified annexin protein of claim 14, wherein said isolated modified annexin protein is a dimer.
18. The isolated modified annexin protein of claim 14, wherein said isolated modified annexin protein is a heterotetramer of an annexin II protein and a pll protein.
19. The isolated modified annexin protein of claim 14, wherein said additional protein is an additional annexin protein.
20. The isolated modified annexin protein of claim 14, wherein said annexin protein is an annexin V protein.
21. The isolated modified annexin protein of claim 20, wherein said annexin V protein comprises an amino acid sequence selected from the group consisting of : a) SEQ ID NO : 3; and b) an amino acid sequence having at least 95% sequence identity to SEQ ID NO : 3.
22. The isolated modified annexin protein of claim 20, wherein said additional protein is an annexin V protein.
23. The isolated modified annexin protein of claim 22, wherein said additional protein comprises an amino acid sequence selected from the group consisting of : a) SEQ ID NO : 3; and b) an amino acid sequence having at least 95% sequence identity to SEQ ID NO : 3.
24. The isolated modified annexin protein of claim 22, wherein said isolated modified annexin protein comprises an amino acid sequence selected from the group consisting of : a) SEQ ID NO : 6; and b) an amino acid sequence having at least 95% sequence identity to SEQ ID NO : 6.
25. A pharmaceutical composition comprising an antithrombotically effective amount of the modified annexin protein of claim 14.
26. A method of treating a subject at risk of thrombosis comprising administering to said subject a pharmaceutical composition comprising an antithrombotically effective amount of the modified annexin protein of claim 14.
27. The method of claim 26, wherein said pharmaceutical composition is administered after coronary thrombosis.
28. The method of claim 26, wherein said pharmaceutical composition is administered after a condition selected from the group consisting of overt cerebral thrombosis and transient cerebral ischemic attack.
29. The method of claim 26, wherein said pharmaceutical composition is administered after a surgical operation associated with venous thrombosis.
30. The method of claim 26, wherein said subject is diabetic and said thrombosis is arterial thrombosis.
31. The method of claim 26, wherein said pharmaceutical composition is administered during a condition selected from the group consisting of pregnancy and parturition.
32. An isolated nucleic acid molecule selected from the group consisting of : a) an isolated bacterial nucleic acid molecule comprising a nucleic acid sequence encoding a protein selected from the group consisting of : i) SEQ ID NO : 6; and ii) a protein having at least 95% sequence identity to SEQ ID NO : 6; and b) a nucleic acid molecule which is fully complementary to any said nucleic acid molecule recited in a).
33. The nucleic acid molecule of claim 32, wherein said nucleic acid molecule comprises SEQ ID NO : 4.
34. A recombinant molecule comprising at least a portion of a nucleic acid molecule according to claim 32.
35. A recombinant cell comprising at least a portion of a nucleic acid molecule according to claim 32.
36. A method of producing a modified annexin protein, comprising culturing the cell of claim 35 under conditions permitting the production of said modified annexin protein, and recovering said modified annexin protein from said culture.
37. A method of screening for a modified annexin protein that modulates thrombosis, said method comprising: a) contacting a thrombosis test system with at least one test modified annexin protein under conditions permissive for thrombosis; b) assessing the antithrombotic activity in the presence of said test modified annexin protein; and c) comparing the antithrombotic activity in the presence of said test modified annexin protein with the antithrombotic activity in the absence of said test modified annexin protein, wherein a change in the antithrombotic activity in the presence of said test modified annexin protein is indicative of a modified annexin protein that modulates thrombotic activity.
38. The method of claim 37 wherein said thrombosis test system is a system for measuring activated partial thromboplastin time.
39. A modified annexin protein that modulates thrombosis identified by the method of claim 37.
40. A method for identifying a modified annexin protein for annexin activity, said method comprising: a) contacting activated platelets with at least one test modified annexin protein under conditions permissive for binding; b) assessing the test modified annexin-binding activity of said platelets; c) assessing the protein S-binding activity in the presence of said test modified annexin protein; and d) comparing the test modified annexin-binding activity and protein S binding activity in the presence of said test modified annexin protein with the unmodified annexin-binding activity and protein S-binding activity in the presence of unmodified annexin protein, whereby a modified annexin protein with annexin activity may be identified.
41. A modified annexin protein identified by the method of claim 40.
PCT/US2002/005079 2001-02-21 2002-02-21 Modified annexin proteins and methods for preventing thrombosis WO2002067857A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
DE60219611T DE60219611T2 (en) 2001-02-21 2002-02-21 MODIFIED ANNEXIN PROTEINS AND PREVENTION AND TREATMENT OF THROMBOSE
JP2002567229A JP2004528025A (en) 2001-02-21 2002-02-21 Modified annexin proteins and methods for preventing thrombosis
EP02721083A EP1379266B1 (en) 2001-02-21 2002-02-21 Modified annexin proteins and prevention and treatment of thrombosis
AU2002252035A AU2002252035A1 (en) 2001-02-21 2002-02-21 Modified annexin proteins and methods for preventing thrombosis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US27040201P 2001-02-21 2001-02-21
US60/270,402 2001-02-21
US33258201P 2001-11-21 2001-11-21
US60/332,582 2001-11-21

Publications (2)

Publication Number Publication Date
WO2002067857A2 WO2002067857A2 (en) 2002-09-06
WO2002067857A3 true WO2002067857A3 (en) 2002-11-07

Family

ID=26954268

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/005079 WO2002067857A2 (en) 2001-02-21 2002-02-21 Modified annexin proteins and methods for preventing thrombosis

Country Status (8)

Country Link
US (2) US6962903B2 (en)
EP (2) EP1839670A3 (en)
JP (3) JP2004528025A (en)
AT (1) ATE359807T1 (en)
AU (1) AU2002252035A1 (en)
DE (1) DE60219611T2 (en)
ES (1) ES2286247T3 (en)
WO (1) WO2002067857A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101015686B (en) * 2007-02-06 2010-05-19 中国人民解放军军事医学科学院基础医学研究所 Thrombolytic medicine synergist and its preparing process

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050222030A1 (en) * 2001-02-21 2005-10-06 Anthony Allison Modified annexin proteins and methods for preventing thrombosis
US7645739B2 (en) * 2001-02-21 2010-01-12 Alavita Pharmaceuticals, Inc. Modified annexin compositions and methods of using same
US20090291086A1 (en) * 2001-02-21 2009-11-26 Alavita Pharmaceuticals, Inc. Compositions and Methods for Treating Cerebral Thrombosis and Global Cerebral Ischemia
US7635676B2 (en) * 2001-02-21 2009-12-22 Alavita Pharmaccuticals, Inc. Modified annexin proteins and methods for their use in organ transplantation
DE10162434A1 (en) * 2001-12-18 2003-09-25 November Ag Molekulare Medizin Expression vector and method for producing an expression vector
EP1673108A4 (en) * 2003-09-09 2009-06-03 Point Biomedical Corp Methods and compositions for ultrasound imaging of apoptosis
AU2005221195B2 (en) * 2004-03-11 2011-05-19 Alavita Pharmaceuticals, Inc. Modified annexin proteins and methods for preventing thrombosis
WO2005099744A1 (en) * 2004-04-15 2005-10-27 Athera Biotechnologies Ab Annexin v for preventing atherothrombois and plaque rupture
US7393833B2 (en) * 2005-03-09 2008-07-01 The Board Of Regents Of The University Of Oklahoma Chimeric proteins with phosphatidylserine binding domains
WO2007149567A2 (en) 2006-06-21 2007-12-27 Musc Foundation For Research Development Targeting complement factor h for treatment of diseases
KR100835879B1 (en) 2006-10-10 2008-06-09 학교법인 한림대학교 Annexin fusion protein
WO2009105231A1 (en) 2008-02-20 2009-08-27 The General Hospital Corporation Compositions and methods for treating vascular disorders
EP3216457B1 (en) 2008-02-22 2019-04-10 Annexin Pharmaceuticals AB Compounds and methods for the prevention or treatment of restenosis
EP2123258A1 (en) * 2008-05-23 2009-11-25 Liplasome Pharma A/S Liposomes for drug delivery
WO2010040147A2 (en) 2008-10-03 2010-04-08 Advanced Proteome Therapeutics, Inc. Site-specific n-terminal modifications of proteins and conjugate formation
EP2380023A2 (en) 2008-12-19 2011-10-26 Medirista Biotechnologies AB Oxidized cardiolipin as a novel pro-inflammatory factor
WO2011003098A1 (en) 2009-07-02 2011-01-06 Musc Foundation For Research Development Methods of stimulating liver regeneration
WO2011057158A1 (en) 2009-11-05 2011-05-12 Taligen Therapeutics, Inc. Treatment of paroxysmal nocturnal hemoglobinuria, hemolytic anemias and disease states involving intravascular and extravascular hemolysis
US9650447B2 (en) 2010-05-14 2017-05-16 The Regents Of The University Of Colorado, A Body Corporate Complement receptor 2 (CR2) targeting groups
JP2013533243A (en) 2010-06-22 2013-08-22 ザ リージェンツ オブ ザ ユニヴァーシティ オブ コロラド,ア ボディ コーポレート Antibody to C3d fragment of complement component 3
JP5584026B2 (en) * 2010-07-02 2014-09-03 株式会社ダイヘン Resistance welding control method
CN102690345B (en) * 2011-03-24 2014-03-19 江苏靶标生物医药研究所有限公司 Human annexin V variant and its expression, preparation method and use
US20140037614A1 (en) 2011-04-05 2014-02-06 Knut Pettersson Therapeutic and prophylactic methods, uses and compositions comprising anexin a5
JP2015535212A (en) 2012-08-17 2015-12-10 ザ・リージエンツ・オブ・ザ・ユニバーシテイ・オブ・コロラド、ア・ボデイー・コーポレイト Compositions and methods for detecting complement activation
US10413620B2 (en) 2012-08-17 2019-09-17 The Regents Of The University Of Colorado, A Body Corporate Light-emitting versions of the monoclonal antibody to C3D (MAB 3D29) for imaging
WO2016205062A1 (en) * 2015-06-15 2016-12-22 Shifa Biomedical Corporation Antithrombotic therapies
GB2542391A (en) 2015-09-17 2017-03-22 Annexin Pharmaceuticals Ab Process of manufacture
CN105906800A (en) * 2016-05-06 2016-08-31 中国科学院长春应用化学研究所 Gene transmission system and preparation method thereof
JP7051826B2 (en) 2016-09-23 2022-04-11 シーエスエル、リミテッド Coagulation factor binding protein and its use
GB201702144D0 (en) 2017-02-09 2017-03-29 Annexin Pharmaceuticals Ab Therapeutic compositions
CN114672483B (en) * 2022-05-31 2022-09-02 上海百力格生物技术有限公司 Method for preparing nucleic acid probe by ultrasonic method

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5612460A (en) * 1989-04-19 1997-03-18 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5632986A (en) * 1991-05-09 1997-05-27 The University Of Washington Phospholipid-targeted thrombolytic agents
WO1999019470A2 (en) * 1997-10-09 1999-04-22 The Regents Of The University Of California Gfp-annexin fusion proteins
US5968477A (en) * 1994-01-24 1999-10-19 Neorx Corporation Radiolabeled annexin conjugates with hexose and a chelator
US6358508B1 (en) * 1997-06-11 2002-03-19 Human Genome Sciences, Inc. Antibodies to human tumor necrosis factor receptor TR9

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4002531A (en) * 1976-01-22 1977-01-11 Pierce Chemical Company Modifying enzymes with polyethylene glycol and product produced thereby
DE3315000A1 (en) * 1983-04-26 1984-10-31 Behringwerke Ag, 3550 Marburg TISSUE PROTEIN PP (DOWN ARROW) 4 (DOWN ARROW), METHOD FOR ITS RECOVERY AND USE
US5066788A (en) * 1984-09-21 1991-11-19 Boehringer Ingelheim International Gmbh Blood coagulation inhibiting proteins, processes for preparing them and their uses
DE3572179D1 (en) * 1984-09-21 1989-09-14 Boehringer Ingelheim Int Blood coagulation inhibiting proteins, process for preparing them and their use
US4897355A (en) * 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
CA1265446A (en) * 1985-09-30 1990-02-06 Masahiro Maki Anticoagulating substance, process for preparing same and anticoagulant comprising same as an effective component
JPH08840B2 (en) * 1986-11-14 1996-01-10 興和株式会社 Anti-PCI monoclonal antibody, anti-PCI purification method and immunoassay using the same
DE3643182A1 (en) * 1986-12-18 1988-06-30 Behringwerke Ag MEDICINAL PRODUCTS CONTAINING THE TISSUE PROTEIN PP4, METHOD FOR THE PRODUCTION OF PP4 AND ITS PASTEURIZATION AND THE USE OF PP4
US4937324A (en) * 1987-02-06 1990-06-26 Zymogenetics, Inc. Chromatographic purification of human proteins having anticoagulant and anti-inflammatory activity
JP2660514B2 (en) * 1987-02-20 1997-10-08 興和株式会社 Polypeptide having anticoagulant action
PT87083B (en) 1987-03-28 1992-07-31 Boehringer Ingelheim Int A PROCESS FOR THE PREPARATION OF A VASCULAR ANTICOAGULANT PROTEIN, OF DNA THAT CODIFIES FOR THIS PROTEIN AND OF PHARMACEUTICAL COMPOSITIONS CONTAINING IT
US4965251A (en) 1987-04-03 1990-10-23 The Board Of Regents Of The University Of Washington Pulse treatment of hemoglobinopathies with erythropoietin
DE3737239A1 (en) 1987-11-03 1989-10-12 Behringwerke Ag GENETIC PRODUCTION OF ANTICOAGULATORY PROTEIN PP4
ATE124087T1 (en) 1988-02-26 1995-07-15 Biogen Inc DNA SEQUENCES, RECOMBINANT DNA MOLECULES AND METHODS FOR PRODUCING LIPOCORTIN III, IV, V, AND VI.
DE4022010A1 (en) * 1989-07-15 1991-01-17 Boehringer Ingelheim Int Anticoagulant compsn. for treatment and prophylaxis of thrombosis - contains vascular anti-coagulant protein (annexine) and divalent salt of calcium, cadmium, zinc, manganese or cobalt
ES2045669T3 (en) 1989-07-15 1994-01-16 Boehringer Ingelheim Int PROCEDURE FOR THE PURIFICATION OF ANEXINS.
DE3937607A1 (en) 1989-11-11 1991-05-16 Boehringer Ingelheim Int USE OF AN ANTI-COAGULAR TREATMENT FOR TUMOR DISEASES
DE3942081A1 (en) 1989-12-20 1991-06-27 Behringwerke Ag MEANS FOR IMPROVING THE REINFORCEMENT OF ANNExINES
US5627036A (en) 1989-12-27 1997-05-06 Boehringer Ingelheim International Gmbh Use of an anticoagulant as a diagnostic agent
US5225537A (en) * 1989-12-29 1993-07-06 Zymogenetics, Inc. Methods for producing hybrid phospholipid-binding proteins
EP0527940A1 (en) 1990-05-08 1993-02-24 Liposome Technology, Inc. Direct spray-dried drug/lipid powder composition
CA2092315A1 (en) 1990-11-02 1992-05-03 John P. Burnier Platelet aggregation inhibitors
CA2096520C (en) 1990-11-20 2000-12-19 Hiroshi Nakao Therapeutic agent for skin or corneal disease
JP2916947B2 (en) * 1990-11-28 1999-07-05 興和株式会社 Method for stabilizing CPB-I and pharmaceutical composition
US5229367A (en) 1991-01-21 1993-07-20 Sclavo S.P.A. Antiinflammatory peptide derived from human lipocortin V
EP0538459A4 (en) 1991-05-09 1993-06-16 The Board Of Regents Of The University Of Washington Phospholipid-targeted thrombolytic agents
JPH051096A (en) 1991-06-26 1993-01-08 Kowa Co Oligopeptide having anticoagulating activity
EP0646019B9 (en) * 1992-06-09 2002-12-18 Neorx Corporation Biotin-DOTA conjugates and their use in pretargeting methods
SG46493A1 (en) 1992-07-08 1998-02-20 Innogenetics Nv Polypepides derived form endonexin 2, having hepatitis b virus receptor activity and their use in diagnostic and pharmaceutical compositions
JP3416912B2 (en) 1992-10-08 2003-06-16 興和株式会社 Oligopeptides with anticoagulant activity
JPH0772149A (en) 1993-06-30 1995-03-17 Nippon Shinyaku Co Ltd Chemical and method for diagnosis of liver cancer or cirrhosis
JPH0772147A (en) 1993-07-02 1995-03-17 Noboru Kaneko Antianexin v antibody as well as preparation and utilization of same antibody
US5849600A (en) 1993-11-10 1998-12-15 The Mclean Hospital Corporation Diagnostic assays for Alzheimer's disease
US5747446A (en) * 1994-03-22 1998-05-05 Beth Israel Deaconess Medical Center Modified polypeptides with increased biological activity
JPH10504534A (en) 1994-06-16 1998-05-06 ネオルクス コーポレイション Radiolabeled annexin-galactose conjugate
JP3574659B2 (en) 1994-07-23 2004-10-06 ジャン‐マリー フレイッシネ Method for determining prethrombotic condition
DE19541284C2 (en) 1995-11-06 1998-09-24 Kalden Joachim Robert Prof Dr Immunomodulation method
RU2178307C2 (en) * 1995-12-27 2002-01-20 Джинентех Инк. Derivatives of protein ob
ATE253117T1 (en) 1996-07-05 2003-11-15 Philip E Branton ADENOVIRUS E4 PROTEINS FOR CELL DEATH INDUCTION
EP0948533A1 (en) 1996-12-30 1999-10-13 Innogenetics N.V. ANNEXIN V-BINDING POLYPEPTIDES DERIVED FROM HBsAg AND THEIR USES
CA2277757C (en) * 1997-01-15 2008-09-16 Phoenix Pharmacologics, Inc. Modified tumor necrosis factor
US6242570B1 (en) * 1997-07-10 2001-06-05 Beth Israel Deaconess Medical Center Production and use of recombinant protein multimers with increased biological activity
WO1999002992A1 (en) 1997-07-11 1999-01-21 The Mount Sinai School Of Medicine Of The City University Of New York Assays for diagnosis of thrombophilic disease
JP2001522809A (en) 1997-11-06 2001-11-20 イノジェネティックス・ナムローゼ・フェンノートシャップ Host-derived protein-bound HCV: pharmaceutical, diagnostic and purification applications
WO1999048916A2 (en) 1998-03-27 1999-09-30 The Board Of Trustees Of The Leland Stanford Jr. University Hypoxia-inducible human genes, proteins, and uses thereof
US20040002056A1 (en) * 1998-05-12 2004-01-01 Lorens James B. Methods of screening for bioactive agents using cells transformed with self-inactivating viral vectors
US6169078B1 (en) * 1998-05-12 2001-01-02 University Of Florida Materials and methods for the intracellular delivery of substances
AU750414B2 (en) * 1998-07-13 2002-07-18 Board Of Regents, The University Of Texas System Cancer treatment methods using therapeutic conjugates that bind to aminophospholipids
WO2000010673A1 (en) 1998-08-24 2000-03-02 Nst Neurosurvival Technologies Ltd. Apparatus and method for capturing particles with surface exposure of anionic phospholipids from biological fluids
IL125908A (en) 1998-08-24 2005-05-17 Nst Neurosurvival Technologies Peptides and pharmaceutical compositions comprising same
EP1107983A2 (en) 1998-09-01 2001-06-20 Innogenetics N.V. Benzodiazepine and benzothiazepine derivatives and hbsag peptides binding to annexins, their compositions and use
US6387366B1 (en) 1998-12-31 2002-05-14 Alg Company Methods for reducing adverse side effects associated with cellular transplantation
CN1125084C (en) * 1999-03-12 2003-10-22 中国科学院上海生物化学研究所 Thrombolytic fusion protein targeting thrombus
US6323313B1 (en) 1999-06-01 2001-11-27 The University Of Washington Annexin derivative with endogenous chelation sites
US6645465B2 (en) 1999-08-06 2003-11-11 Michigan, University Of The Regents Annexin proteins and autoantibodies as serum markers for cancer
US20020197261A1 (en) 2001-04-26 2002-12-26 Chun Li Therapeutic agent/ligand conjugate compositions, their methods of synthesis and use

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5612460A (en) * 1989-04-19 1997-03-18 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5632986A (en) * 1991-05-09 1997-05-27 The University Of Washington Phospholipid-targeted thrombolytic agents
US5968477A (en) * 1994-01-24 1999-10-19 Neorx Corporation Radiolabeled annexin conjugates with hexose and a chelator
US6358508B1 (en) * 1997-06-11 2002-03-19 Human Genome Sciences, Inc. Antibodies to human tumor necrosis factor receptor TR9
WO1999019470A2 (en) * 1997-10-09 1999-04-22 The Regents Of The University Of California Gfp-annexin fusion proteins

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101015686B (en) * 2007-02-06 2010-05-19 中国人民解放军军事医学科学院基础医学研究所 Thrombolytic medicine synergist and its preparing process

Also Published As

Publication number Publication date
JP2010189405A (en) 2010-09-02
EP1839670A2 (en) 2007-10-03
US20050197295A1 (en) 2005-09-08
US7407475B2 (en) 2008-08-05
EP1379266A4 (en) 2004-05-06
ATE359807T1 (en) 2007-05-15
ES2286247T3 (en) 2007-12-01
US6962903B2 (en) 2005-11-08
JP2004528025A (en) 2004-09-16
EP1839670A3 (en) 2009-11-11
US20030166532A1 (en) 2003-09-04
AU2002252035A1 (en) 2002-09-12
DE60219611T2 (en) 2007-12-27
DE60219611D1 (en) 2007-05-31
WO2002067857A2 (en) 2002-09-06
JP2008263991A (en) 2008-11-06
EP1379266B1 (en) 2007-04-18
EP1379266A2 (en) 2004-01-14

Similar Documents

Publication Publication Date Title
US7407475B2 (en) Modified annexin proteins, and methods and compositions for using them
US20050222030A1 (en) Modified annexin proteins and methods for preventing thrombosis
US7635676B2 (en) Modified annexin proteins and methods for their use in organ transplantation
US7645739B2 (en) Modified annexin compositions and methods of using same
US7635680B2 (en) Attenuation of reperfusion injury
JP2012254992A (en) Modified annexin protein and method for preventing thrombosis
US11897924B2 (en) Anticoagulant fusion proteins and uses thereof
US20090291086A1 (en) Compositions and Methods for Treating Cerebral Thrombosis and Global Cerebral Ischemia
WO2004013303A2 (en) Modified annexin proteins and methods for treating vaso-occlusive sickle-cell disease
US6982154B2 (en) Modified annexin proteins and methods for treating vaso-occlusive sickle-cell disease

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A3

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

AL Designated countries for regional patents

Kind code of ref document: A3

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2002567229

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2002721083

Country of ref document: EP

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 2002721083

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2002721083

Country of ref document: EP