Anticoagulants and Thrombolytic Agents

Embed Size (px)

Citation preview

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    1/12

    Coagulation cascade

    After injury to a vessel wall, tissue factor is exposed on

    the surface of the damaged endothelium.

    The interaction of tissue factor with plasma factor VII

    activates the coagulation cascade, producing thrombin by

    stepwise activation of a series of proenzymes

    The coagulation cascade is regulated by natural

    anticoagulants, such as tissue factor pathway inhibitor

    TFPI, the protein C and protein S system, and

    antithrombin, all of which help to restrict the formation

    of the hemostatic plug to the site of injury.

    Effects of thrombin

    converts soluble fibrinogen to fibrinactivates factors V, VIII, and XI, which generates more

    thrombin

    stimulates platelets

    by activating factor XIII, thrombin favours the

    formation of cross-linked bonds among the fibrin

    molecules, stabilizing the clot

    Activated platelets

    An activated platelet exposes surface

    receptors for specific clotting factors,

    such as factor Va, and anionic

    phospholipids that function as binding

    sites for factor Xa.

    An analogous system exists for

    binding factor IXa.

    Endogenous inhibitors of coagulation

    antithrombin III

    from a family of serine protease

    inhibitors (serpins)

    glycose aminoglycans (GAGs)

    highly sulphated sugars bind to

    antithrombin by ionic interaction

    associated with surfaces of endothelial

    cells and subendothelial structures

    smaller heparin molecules inhibit Xa

    more effectively

    tissue factor pathway inhibitor

    (TFPI)

    inhibits Xa by forming a complex that

    can also inhibit VIIa bound to TF, but

    not free VII

    important for blocking the effect of TF

    where it is expressed on endothelial

    cells and subendothelial structures

    proteins C and Sthrombin in conjunction withthrombomodulin, activates protein CProtein C proteolytically cleaves VIIIa

    and Va (major cofactors that helpproduce Xa and IIa), with protein Sacting as cofactorin septic patients, activated protein C(available as a recombinant product)inhibit DIVC that occurs in smallvesselsprotein C may down-regulateinflammatory cytokines

    thrombomodulin

    a constituent of the endothelial cell

    membrane, very small amounts arepresent in blood

    binds thrombin and begins the

    sequence of protein C activation

    functions as cell-based

    inhibition of coagulation

    probably facilitates thrombin

    catabolism

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    2/12

    Thrombus formation at the site of damaged vessels

    Platelet factors

    platelet factor 1: coagulation factor V

    platelet factor 2: thromboplastic material

    platelet factor 3: platelet thromboplastin

    platelet factor 4: antiheparin factor

    platelet factor 5: fibrinogen coagulation factor

    platelet factor 6: antifibrinolytic factor

    platelet factor 7: platelet cothromboplastin

    Platelet membrane glycoproteins

    GP Ia: receptor for subendothelium

    GP Ib: receptor for von Willebrand

    GP IIb: receptor for von Willebrand, fibrinogen

    GP IIIa: receptor for von Willebrand, fibrinogen

    Prothrombin time

    monitoring of extrinsic coagulation pathway

    normal range between 9 to 15 seconds

    "normal" varies according to batch of thromboplastin in

    test reagent in different laboratories, hence the use ofInternational Normalised Ratio (INR)

    for a person on full anticoagulant therapy, the PT should

    be 2 to 3 times the laboratory "control" value (INR of 2-

    3)

    prolonged PT (more than 3 times control value)

    bile duct obstruction

    cirrhosis

    disseminated intravascular coagulation

    hepatitis

    malabsorption

    warfarin therapy

    > 10% deficiency in any of the following

    Vitamin K, VII, X, II, V, I

    Partial thromboplastin time

    monitoring of intrinsic and common coagulation

    pathways

    partial thromboplastin time (PTT):3045 s

    activated partial thromboplastin time (APTT):2539 s

    value will vary between laboratories

    patients receiving anticoagulant therapy usually will

    have value within 1.5 to 2.5 times control values

    not valid for patients on low molecular weight heparin

    therapy (anti Xa heparin assay)

    prolonged PTT may indicate

    cirrhosis

    disseminated intravascular coagulation (DIC)

    factor XII deficiency

    hemophilia A (factor VIII deficiency)

    hemophilia B (factor IX deficiency)

    hypofibrinogenemia

    malabsorption (inadequate absorption of

    nutrients from the intestinal tract)

    von Willebrand's disease

    lupus anticoagulant

    decreased aPTT can occur due to:digitalis

    tetracyclines

    antihistamines

    nicotine

    elevated factor VIII

    tissue inflammation or trauma

    Fibrinolytic system

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    3/12

    DRUGS USED IN COAGULATION DISORDERS

    ANTICOAGULANTS

    Heparina heterogeneous groupa member of the heparan sulphate family of complexsugars classified under glycosaminoglycans

    glycosaminoglycans or mucopolysaccharides

    are polymers of repeating disaccharideswithin the disaccharides, the sugars

    tend to be modified, with acidic

    groups, amino groups, sulfated

    hydroxyl and amino groups

    tend to be negatively charged, because

    of the prevalence of acidic groups

    heparan sulphate family of complex sugars is

    composed of long chains of alternating

    disaccharide units of uronic acid (glucuronic

    and iduronic acid) and glucosamine residues

    the backbone structure is thendecorated by with complex patterns of

    sulphate and carboxyl groups at various

    positions, giving rise to a very strongly

    negative charged molecule.

    strongly acidic because of its content of covalently

    linked sulfate and carboxylic acid groups (heparin

    sodium)

    has an extended helical conformation due to

    charge repulsion by the many negatively

    charged groups

    naturally occurring in the secretory granules of mastcells

    has no anticoagulation effect by itself

    biologic activity is dependent upon the plasma protease

    inhibitor, anti-thrombin III (AT III) (heparin cofactor)

    a specific pentasaccharide sequence containing a 3-O-

    sulphated glucosamine residue forms a high-affinity

    binding site for AT III

    AT III is one of the many naturally occurring

    inhibitors in coagulation but its action is slow

    small amounts of heparin (with AT III) inhibit

    thrombosis by inactivating activated Factor X

    (Xa) and inhibiting the conversion of

    prothrombin to thrombin (II to IIa)

    tight binding of heparin molecule to AT III

    causes a conformational change in this inhibitor,

    which exposes the active binding site of

    antithrombin III for more rapid interaction with

    the proteases (activated clotting factors) to

    inhibit the enzymes

    in the absence of heparin, formation of heparin-

    antithrombin-protease complexes is slow, in the presence

    of heparin, they are accelerated 1000-fold

    heparin catalyses the antithrombin-protease reaction

    without being consumed

    once the antithrombin-protease complex is

    formed, heparin is released intact for renewed

    binding to more AT III

    heparin-antithrombin III complex inactivates

    serine esterases

    factors XIIa, XIa, Xa, IXa, IIa

    plasmin

    kallilrein

    only unbound Xa is sensitive to heparin activity

    Xa bound to platelets in the prothrombinase

    complex is protected from the heparin actiononce active thrombosis has developed, larger amounts of

    heparin inhibit further coagulation by inactivating

    thrombin (IIa) and preventing the conversion of

    fibrinogen to fibrin (I to Ia)

    decreased platelet aggregation

    reduction of platelet membrane receptors for

    von Willebrand factor and fibrinogen (Ia)

    inhibition of VIIIa, Va

    facilitating the release of tissue plasminogen

    activator (tPA), resulting in an increase in

    plasmin and D-dimer concentrations, both ofwhich interfere with platelet aggregation

    increased platelet aggregation

    binding of antiplatelet IgG antibodies to

    platelet-bound heparin, activates platelets and

    induces platelet clumping (heparin-induced

    thrombocytopaenia, HIT)

    inhibition of VIIa-TF complex

    by releasing TFPI from endothelial cells, renal

    cells, carcinoma cells and lipoprotein fraction of

    plasma

    TFPI also inhibits Xa on TF bearing cellreleases lipoprotein lipase from capillary endothelial

    surfaces

    the enzyme hydrolyzes triacylglycerols in

    chylomicrons, very-low-density lipoproteins,

    low-density lipoproteins, and diacylglycerols

    inhibits growth of capillary endothelial cells but

    potentiates the activity of acid fibroblasts growth factors

    on these cells

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    4/12

    commercial preparations

    consists of repeated sulphated mucopolysaccharides

    D-glucosamine-L-iduronic acid, and

    D-glucosamine-D-glucuronic acid

    source

    porcine intestinal mucosa (higher potency)

    bovine lung

    partial substitution of acidic protons of the sulfate units

    by

    Na+ (as heparin sodium)

    Ca++ (as calcium heparin)

    Li+ (as lithium heparin)

    used in vitro as an anticoagulant for blood

    samples

    pH adjustment (between 5.0 and 7.5)

    titrated with HCl or NaOH

    standardization of activity

    regular heparin consists of a family of

    molecules of different molecular weights, the

    correlation between the concentration of a given

    heparin preparation and its effect on coagulationoften is low

    therefore standardized as units of activity by

    bioassay

    heparin sodium must contain at least 120 USP

    units per milligram

    1 U stops 1 ml of citrated sheep plasma from

    clotting for 1 hour after the addition of 0.2 ml of

    1% calcium chloride

    high molecular weight (HMW) fractions

    with high affinity for AT III, markedly inhibit

    blood coagulationfractions have a MW range of 5 000-30 000

    low molecular weight (LMW) fractions

    MW 1 000-10 000 (1-10kDa, mean 4.5 kDa)

    less effect on antithrombin III

    activity dependent on number of

    monosaccharide units per molecule

    < 8, no significant antithrombotic

    activity

    8-18, potentiate inhibition of factor Xa

    >18, potentiate inhibition of both

    factor Xa and thrombinisolated from standard heparin

    by gel filtration chromatography

    by differential precipitation with

    ethanol

    by partial depolymerization with

    nitrous acid

    by alkaline degradation of heparin

    benzyl ester / beta elimination

    degradation (enoxaparin Na)

    by enzymatic degradation

    compared with regular heparinincreased bioavailability after

    subcutaneous administration

    less frequent dosing requirements

    units of reference

    milligrams for enoxaparin

    anti-factor Xa units for dalteparin and

    danaproid

    indications

    prevention of deep venous thrombosis

    after hip replacement surgery

    treatment of acute deep vein

    thrombosis

    prophylaxis for ischaemic

    complications in unstable angina and

    non-Q-wave myocardial infarction

    pharmacokinetics

    absorption

    not absorbed from the intestinal mucosa,

    therefore administered parenterally (continuous

    infusion, intermittent intravenous injection, deep

    subcutaneous injection)

    onset of action

    after intravenous administration, immediate

    after subcutaneous administration, delay of 1-2

    hours

    clearance

    degraded primarily by reticuloendothelial

    system, and heparinase

    small amount of undegraded heparin appears in

    urine

    t

    dose-dependent

    100 U/kg, 1 hour

    400 U/kg, 2.5 hours

    800 U/kg, 5 hours

    shortened in patients with pulmonary embolism

    prolonged in patients with hepatic cirrhosis or

    renal failure

    LMW heparins have longer biological half-lives than do standard heparin

    adverse effects

    bleeding

    monitor partial thromboplastin time (PTT)

    elderly women and patients with renal failure

    are more prone to haemorrhage

    platelet dysfunction

    thrombocytopenia (HIT)

    platelet count of less than 50% of pretreatment

    value or

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    5/12

    contraindications

    hypersensitivity

    active bleeding

    haemophilia

    thrombocytopaenia and history of HIT

    purpura

    severe hypertension

    intracranial haemorrhage

    infective endocarditis

    active tuberculosis

    ulcerative lesions of gastrointestinal tract

    threatened abortion

    visceral carcinoma

    advance hepatic or renal disease

    during or after surgery of brain, spinal cord, or eye

    undergoing lumbar puncture or regional anaesthesia

    blocks

    administration and dosages

    used in pregnant women only when clearly indicated

    established venous thrombosis

    maintain plasma concentration of 0.2 U/ml toprolong the PTT to INR 2-2.5

    initial bolus injection of 5000-10000 U,

    followed by infusion of 10-15 U/kg/h

    with acute pulmonary embolism,

    larger doses require during the first few

    days because of increased heparin

    clearance

    intermittent administration, 75-100 U/kg every

    4 hours

    heparin resistance - causes

    increased serum concentration of other (acute reactant)proteins that have affinity for heparin

    fibroblast growth factors (FGFs)

    vascular endothelial growth factor (VEGF)

    heparin-binding EGF-like growth factor

    hepatocyte growth factor (HGF)

    transforming growth factor-beta (TGF-beta)

    interferon-gamma (IFN-gamma)

    platelet-derived growth factor (PDGF)

    platelet factor-4 (PF-4)

    interleukin-8 (IL-8)

    macrophage inflammatory protein-1 (MIP-1)interferon-gamma inducible protein-10 (IP-10)

    insulin-like growth factors I or II

    fibronectin

    laminin

    histidine-rich glycoprotein vitronectin

    increased concentration of factor VIII, a cofactor that

    increases the proteolytic activity of IXa

    congenital deficiency of AT III (concentration less than

    50% of normal), may be precipitated by pregnancy,

    infection or surgery

    acquired deficiency of AT III (concentration less than25% of normal), may occur in patients with hepatic

    cirrhosis, nephrotic syndrome, disseminated intravascular

    coagulation

    accelerated clearance of heparin, as may occur with

    massive pulmonary embolism

    reversal of heparinisation

    discontinuation of the drug

    protamine sulphate

    highly basic peptide that combines with heparin

    as an ion pair to form a stable complex devoid

    of anticoagulant activity

    for every 100 U of heparin remaining in the

    patient, 1 mg of protamine sulphate is

    administered intravenously

    excess protamine has an anticoagulant effect

    binds to platelets and fibrinogen

    metabolized by N-carboxypeptidase

    Enoxaparin

    obtained by alkaline degradation of heparin benzyl ester

    approximately one-third molecular size of standard

    heparin

    Fondaparinux sodium

    synthetic and specific inhibitor of activated factor X

    (Xa)

    molecular weight is 1728

    supplied as a clear and colorless liquid with a pH

    between 5.0 and 8.0

    mechanism of action

    antithrombin III-mediated selective inhibition of factor

    Xa, and potentiates (about 300 times) the innate

    neutralization of factor Xa by AT III

    does not inactivate thrombin and has no known effect on

    platelet function

    does not bind significantly to other plasma proteins

    (including platelet factor 4) or red blood cellsabsorption

    rapidly and completely absorbed after administration by

    subcutaneous injection, bioavailability is 100%

    distribution

    in healthy adults, volume of distribution of 7-11 L

    clearance

    in healthy individuals up to 75 years of age, up to 77%

    of a single subcutaneous or intravenous fondaparinux

    dose is eliminated in urine as unchanged drug in 72 hours

    25% lower in patients over 75 years of age

    elimination half-life is 17-21 hourstotal clearance is approximately lower in patients with

    renal impairment

    drug interactions

    concomitant use of oral anticoagulants (warfarin),

    platelet inhibitors (acetylsalicylic acid), NSAIDs

    (piroxicam) and digoxin did not significantly affect the

    pharmacokinetics/pharmacodynamics of fondaparinux

    sodium

    does not influence the pharmacodynamics of warfarin,

    acetylsalicylic acid, piroxicam, and digoxin, nor the

    pharmacokinetics of digoxin at steady statedoes not bind significantly to plasma proteins other than

    AT III, no drug interactions by protein-binding

    displacement are expected

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    6/12

    Warfarin

    oral anticoagulant introduced as rodenticide in 1948

    Wisconsin Alumni Research Foundation - arin

    discovery of anticoagulant substance formed in spoiled

    sweet clover silage which produced a deficiency of

    plasma prothrombin and haemorrhagic disease in cattle

    toxic agent identified as bishydroxycoumarin

    and synthesized as dicoumarol

    structure

    4-hydroxycoumarin residue, with a non-polar carbon

    substituent at the 3-position, is the minimal structural

    requirement for activity

    this carbon is asymmetrical in warfarin

    commercial preparations are racemic

    mixture of 2 enantiomorphs

    levorotatory S-warfarin

    dextrorotatory R-warfarin

    potency: S-warfarin > R-warfarin, 4:1

    mechanism of action

    warfarin prevents the -carboxylation of glutamate

    residues in factors II, VII, IX, and Xby blocking the reduction of inactive vitamin K

    epoxide (by vitamin K epoxide reductase) back

    to its active hydroquinone form

    results in incomplete molecules that are

    biologically inactive in coagulation

    role of vitamin K

    post-ribosomal modification of factors II, VII, IX, and

    X, and the endogenous anticoagulant protein C and S in

    the liver

    involves -carboxylation of glutamate residuesin them

    the carboxyl-glutamyl residues are responsible for the

    binding of Ca++ which are necessary for the binding of

    the activated factors to phospholipid vesicles

    this process is coupled with the oxidative deactivation of

    vitamin K

    in vitamin K deficiency, inactive precursors are

    liberated

    anticoagulant effects

    8-12 hour delay in the action of warfarin, duration of

    action 2-5 daysanticoagulant effect of warfarin results from a balance

    between partially inhibited synthesis and degradation rate

    of the 4 vitamin K-dependent clotting factors

    t: 6 h (VII), 24 h (IX), 36 h (X), 50 h (II)

    large initial doses of warfarin (0.75/kg) hasten the onset

    of anticoagulation effect, beyond this dosage, the speed

    of onset is independent of the dose size; only effect of a

    large loading dose is the prolongation of t of the drug

    pharmacokinetics

    absorption

    acidic, available as a sodium salt

    rapid oral absorption, 100% bioavailability

    decreased in malabsorption

    detectable in plasma within 1 hour of oral

    administration

    peak plasma concentration in 2-8 hours

    crosses placenta

    distribution

    99% of racemic warfarin bound to plasma

    albumin, which may contribute to its

    small Vd (the albumin space) 0.14L/kg

    clearance

    0.045 ml/kg/min

    transformed by CYP 1A2, CYP2C9 into

    inactive metabolite by the liver and kidney

    long t of 25-60 hours in plasma,

    prolonged with liver disease

    excreted via urine and stool

    adverse effects

    crosses placenta readily

    causing haemorrhagic disorder in the foetus

    affecting -carboxyglutamate residues in foetal

    bone and blood proteins and causing birth defect

    characterised by bone malformation

    cutaneous necrosis

    sometimes occur during the first week of

    therapy resulting from venous thrombosis

    due to reduced activity of protein C

    (endogenous anticoagulant)

    rarely same process causes haemorrhagicinfarction of the breast, fatty tissues, intestine,

    and extremities

    administration and dosages

    start with small daily dose of 5-10 mg, may be up to 40

    mg; initial adjustment of prothrombin time takes about 1

    week

    maintenance dose of 5-7 mg/day, may be up to 15

    mg/day

    INR of 2.5-3.5 for patients with prosthetic heart valves

    drug interactions

    pharmacokinetic mechanismsincreasing or decreasing anticoagulant effect

    and the risk of bleeding by

    enzyme induction or inhibition

    variation in plasma protein binding

    increasing activity of warfarin

    stereoselective inhibition of oxidative

    metabolism of S-warfarin, resulting in

    hypoprothrombinaemia

    pyrazolones, phenylbutazone,

    metronidazole, fluconazole,

    trimethoprim-sulpha methoxazoleinhibition of metabolism of warfarin

    amiodarone, disulfiram, cimetidine,

    chloramphenicol

    displacement of albumin-bound warfarin,

    increasing the free fraction

    pyrazolones, phenylbutazone,

    sulfinpyrazone, NSAID, chloral

    hydrate, mefenamic acid

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    7/12

    decreasing activity of warfarin

    induction of hepatic enzymes that metabolise

    warfarin

    barbiturates, rifampicin, alcohol

    reduction in absorption and bioavailability

    cholestyramine binds warfarin in the

    intestine

    increased albumin binding

    pharmacodynamic mechanisms

    synergism(impaired haemostasis, reduced

    clotting factor synthesis in hepatic disease,

    heparin, NSAIDs)

    competitive antagonism (vitamin K)

    altered physiologic control loop for vitamin K

    (hereditary resistance to oral anticoagulant

    augmentation of anticoagulant effect

    inhibition of platelet function

    pyrazolones, phenylbutazone, aspirin

    increasing turnover of clotting factors

    liver disease, hyperthyroidism

    inhibition of activity of VIIa, IXa, Xa, IIaheparin, (prolonging prothombin time)

    vitamin K production

    elimination of vitamin K producing

    bacteria in gastrointestinal tract by

    third generation cephalosporins

    direct inhibition of vitamin K epoxide

    reductase

    third generation cephalosporins

    reduction of anticoagulant effect

    increase synthesis of clotting factors with

    vitamin Kincrease supply of clotting factors with

    transfusion of fresh frozen plasma

    clotting factor concentration through

    haemoconcentration with diuretics

    hereditary resistance to warfarin via mutation

    of vitamin K epoxide reductase

    decreasing turnover rate of clotting factors

    hypothyroidism

    reversal of antocoagulant effects

    disappearance of anticoagulant effect is due toreestablishment of normal activity of the

    clotting factors

    does not correlate with plasma concentration of

    warfarin

    depends on the degree of correction required

    stopping warfarin alone with or without

    large doses of vitamin K (phytonadione)

    50 mg infusion

    fresh frozen plasma

    factor IX concentrates

    whole blood transfusion

    Direct thrombin inhibitor

    bind directly to thrombin and block its interaction with

    its substrates

    recombinant hirudins, bivalirudin, and

    ximelagatran

    parenteral DTIs:

    hirudin and argatroban for the treatment of

    heparin-induced thrombocytopenia

    bivalirudin as an alternative to heparin in

    percutaneous coronary intervention, and

    desirudin as prophylaxis against venous

    thromboembolism in hip replacement

    Hirudin

    source

    Hirudo medicinalis leeches,

    powerful and specific thrombin inhibitor

    recombinant DNA

    mechanism of action

    binds to active site of thrombin

    can reach and inactivate fibrin-bound thrombin

    little effects on platelets or bleeding time

    administration

    parenterally

    monitored by partial thromboplastin time

    Ximelagatran

    first oral direct thrombin inhibitor to be introduced

    a prodrug, its active metabolite is melagatran which

    directly inhibits thrombin

    ximelagatran developed to enhance bioavailability of

    melagatran, the molecules of latter are charged and

    become highly hydrophilic at intestinal pH, resulting in

    low absorption

    melagatran can also be given by injection

    melagatran resembles a peptide sequence on

    fibrinogens A- chain, where thrombin-inducedcleavage occurs

    it binds reversibly to the active site of thrombin and

    inhibits the normal function of thrombin, including both

    free and clot-bound thrombin

    the ability to bind clot-bound thrombin is an

    advantage since clot-bound thrombin may retain

    its enzymatic activity and continue to stimulate

    the coagulation cascade

    regulatory approval for ximelagatran in France for the

    prevention of venous thromboembolic events in major

    orthopaedic (hip or knee replacement) surgerydoes not have the same difficulties with dose adjustment

    or drug interaction as warfarin does

    no known antidote

    pharmacokinetics

    rapidly absorbed after oral administration, peak

    concentration Cmax achieved 1 h after administration

    has a rapid onset and offset of action and shows low

    potential for food and drug interactions

    oral bioavailability approximately 20%, compared with

    3%-7% for melagatran

    ximelagatran Vd 2.53L/kgpharmaokinetics of melagatran described as linear, first-

    order, one compartment model

    melagatran excreted unchanged via kidneys, t 3h

    renal clearance rate

    young 7.7L/h, elderly 5 L/h

    affected in patients with severe renal

    impairment, defined as creatinine

    clearance rate of

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    8/12

    adverse effects

    nausea, diarrhoea, headache

    bleeding, but not worse than with enoxaparin or

    dalteparin

    raised hepatic ALT and bilirubin concentrations

    contraindications

    renal, and hepatic impairment

    pregnancy

    infant and children

    precautions

    lactating women

    main properties and pharmacokinetic characteristics

    FIBRINOLYTIC DRUGS

    mode of action

    rapid lysis of haemostatic thrombus and target

    thromboemboli by catalysing the formation of serine

    protease plasmin from its precursor zymogen

    plasminogencan cause a generalized lytic state after

    intravenous administration

    (zymogen = inactive precursor of proteolytic

    enzyme)

    activation of free circulating plasminogen

    streptokinase

    urokinase

    activation of fibrin bound plasminogen

    anistreplase

    alteplase

    reteplasetissue plasminogen activator (t-PA)

    indications

    multiple pulmonary emboli that are massive enough to

    require surgical intervention

    central deep vein thrombosis of the superior venous

    cava, iliofemoral veins

    coronary thrombolysis after acute myocardial infarction

    peripheral arterial disease

    Streptokinase

    exotoxin of-haemolytic streptococci

    antigenic

    forms a stable, noncovalent 1:1 complex with free

    circulating plasminogen

    produces a conformational change that exposes the

    active site on plasminogen that cleaves arginine 560 on

    free plasminogen molecules to form free plasmin

    complex is not inhibited by 2-antiplasmin

    not fibrin specific, readily induces a systemic lytic state

    t 40-80 minutes

    adverse effects

    bleeding, allergic reactions, fever, anaphylaxis

    Anistreplase

    anisolyated plasminogen streptokinase

    activator complex, APSAC

    a complex of purified human plasminogen and

    bacterial streptokinase

    lys-plasminogen has been acylated at its

    catalytic site to protect the enzymes active site

    when administered, the acyl group

    spontaneously hydrolyses, allows the

    plasminogen-streptokinase complex to bind to

    fibrin prior to activation,

    this modification confers clot selectivity

    advantages

    allows for rapid intravenous injection

    greater clot selectivity

    Tissue plasminogen activator

    preferentially activates plasminogen that is bound tofibrin, several hundredfold more rapidly than free

    plasminogen in the circulation

    theoretically confines fibrinolysis to the formed

    thrombus and avoids systemic activation

    binds to fibrin via lysine binding sites at its amino

    terminus

    metabolised by liver, t 5-10 minutes

    produced by recombinant DNA technology

    alteplase is unmodified t-PA

    reteplase is t-PA from which several amino

    acids have been deleted

    Urokinase

    human enzyme synthesized by kidney, therefore not

    antigenic

    directly converts free plasminogen to plasmin

    lacks fibrin specificity, readily induces a systemic lytic

    state

    metabolised by liver, t of 15-20 minutes

    Prourokinase

    a zymogenic plasminogen activator, a precursor ofurokinase

    binds to fibrin before activation

    has selectivity for clots

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    9/12

    administration and dosages

    streptokinase (US$_00)

    intravenous infusion, loading dose 250 000 U

    to overcome plasma antibodies directed against

    the protein (from prior streptococcal infection),

    followed by maintenance dose of 100 000

    U/hour for 24-72 hours

    follow with full heparinisation as plasminogen

    is exhausted

    can act as antigen, patients with antibodies to

    streptokinase can develop fever, allergic

    reactions, therapeutic resistance

    urokinase (US$_000)

    intravenous infusion, loading dose 1000 to

    4500 U/kg over 10 minutes, followed by

    maintenance dose of 4400 U/kg/hour for 12

    hours

    alteplase (t-PA) ($_000)

    accelerated regime for coronary thrombolysis

    intravenous infusion, loading dose 15 mg

    followed by 0.75mg/kg over 30 minutes(maximum 50mg), followed by 0.5mg/kg

    (maximum 35mg) over the following hour

    reteplase ($_000)

    administered as 2 intravenous bolus injections

    of 10 U each separated by 30 minutes

    anistreplase ($_000)

    single intravenous bolus injection of 30 U over

    3-5 minutes

    ANTITHROMBOTIC DRUGS

    prevention of vascular events among patients withtransient ischaemic attacks

    complete strokes

    angina pectoris

    4 main groups

    cyclo-oxygenase inhibitors: e.g. aspirin

    increasing platelet cAMP:

    by stimulation of adenylyl cyclase:

    adenosine

    by inhibition of phosphodiesterase:

    dipyrimadole

    ADP receptor antagonists: e.g. clopidogrelGP IIb,IIIa blockers: tirofiban, abciximab

    target sites of antithrombotic drugs on the platelet

    Cyclooxygenase inhibition

    aspirin

    inhibition of the synthesis of thromboxane A2

    by irreversible, covalent acetylation of a serine

    residue near the active site of cyclooxygenase

    the anuclear platelet cannot synthesize new

    proteins or enzymes during its 7-10-day life-

    span

    repeated doses produce a cumulative effect on

    platelet function

    maximally effective as an antithrombotic agent

    at doses of 160mg to 320 mg/day

    higher doses inhibit the production of

    prostacyclin, an antithrombotic eicosanoid

    produced by the endothelium

    prolongs bleeding time

    other NSAIDS

    other salicylates and other nonsteroidal anti-

    inflammatory drugs also inhibit cyclooxygenase

    but have a shorter duration of inhibitory action

    because they cannot acetylate cyclooxygenase,

    therefore their action is reversible

    Increasing platelet cAMP

    following platelet activation, Ca++ is released from its

    storage sites (platelet dense tubular systems) to the

    platelet cytoplasm resulting in an increase of cytosolic

    free Ca++

    Ca++ mobilization is directly involved in

    platelet activation and Ca++ is an important

    second messenger for signal transduction in

    plateletscAMP is another second messenger which opposes the

    effect of Ca++by causing sequestration of cytosolic Ca++

    to the Ca++ storage sites

    agents which increase cAMP will suppress platelet

    activation

    stimulation of platelet adenylyl cyclase: action

    of adenosine on platelet A2 receptor

    inhibition of platelet phosphodiesterase:

    dipyridamole

    does not prolong bleeding time

    only current recommendation is for

    primary prophylaxis of thromboemboli

    in patients with prosthetic heart valves,

    and is given in combination with

    warfarin

    dipyridamole is metabolized in the

    liver and has a terminal half-life of 10h

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    10/12

    ADP receptor antagonism

    Ticlopidine

    a thienopyridine derivative, interferes selectively with

    ADP-induced transformation of GPIIb/IIIa complex

    expression in activated platelets

    it also inhibits platelet aggregation induced by thrombin,

    collagen, arachidonic acid, platelet-activating factor,

    prostaglandin endoperoxides, thromboxane A2-like

    substance, serotonin, and epinephrine

    pharmacokinetics

    absorption

    well absorbed after oral administration

    effect within 48 hours

    maximal effect after approximately 3 to 5 days

    activity still present 72 hours after last dose

    absorption decreased by concurrent antacid

    therapy

    metabolism

    rapidly and extensively metabolised in liver

    and excreted in urineone of its metabolite is active

    half-life at steady state is 4-5 days

    effects

    prolongs bleeding time, maximum effect after several

    days of treatment

    antiplatelet activity persists for a week or longer after

    treatment is discontinued

    possibly due to action of active metabolite

    act independent of aspirin with no effect on eicosanoid

    metabolism

    adverse effects

    gastrointestinal (20%): nausea, dyspepsia, diarrhoea

    haemorrhage (5%)

    bone marrow suppression

    leucopenia (1%): detected by regular

    monitoring of white cell count during the first 3

    months of therapy

    thrombocytopenia

    agranulocytosis

    aplastic anaemiacholestatic jaundice

    elevated serum cholesterol concentration

    rashes

    indications

    prevention of thrombosis in cerebral vascular and

    coronary artery disease

    for patients who are unable to tolerate aspirin

    Clopidogrel

    analogue of ticlopidine, another ADP antagonist from

    the thienopyridine group that inhibits ADP and thrombin-

    induced platelet aggregation

    a prodrug, activated by cytochrome P450 predominantly

    by CYP3A4 (less by CYP3A5) to a metabolite that

    inhibits ADP-induced platelet aggregation

    by binding the ADP receptor the drug selectively

    reduces the number of functional ADP receptors

    mediating the inhibition of stimulated adenylate cyclase

    inhibits the binding of fibrinogen to its platelet

    receptor, the GPIIb/IIIa integrin

    it does not modify the GPIIb/IIIa complex

    after oral administration, clopidogrel is rapidly absorbed

    and undergoes metabolic activation by CYP3A4 in the

    liver

    antiplatelet activity of clopidogrel can be inhibited by

    the CYP3A4 substrates erythromycin, troleadomycin,

    and HMG-CoA reductase inhibitors (atorvastatin,

    cerivastatin, lovastatin, and simvastatin) and enhanced by

    the CYP3A4 inducer rifampin

    co-administration of HMG-CoA reductase

    inhibitors will diminish the activation of

    clopidogrel

    the principal circulating metabolite is an inactive

    carboxylic acid derivative

    has an 8 hour elimination half-life, but the

    pharmacologic half-life is relatively long and it takes 4 to

    7 days of administration to reach a steady state effect on

    platelets

    side effects include thrombocytopenia, neutropenia

    clopidogrel-induced platelet inhibition persists severaldays after withdrawal of the drug and diminishes in

    proportion to platelet renewal

    in comparison with ticlopidine, clopidogrel is more

    potent, with less degree of neutropenia

    clopidogrel is significantly more active than aspirin.

    compared with aspirin, clopidogrel has less

    severe gastrointestinal bleeding but more severe

    rash incidence

    Platelet GP IIb/IIIa receptor antagonism

    mechanism of actionblocks platelet receptors for integrin and fibrinogen

    adverse effects

    bleeding

    immunogenicity

    thrombocytopenia

    in approximately 0.1% to 0.5% of patients,

    platelet count of

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    11/12

    free unbound abciximab undergoes rapid proteolytic

    degradation, resulting in a 10- to 15-minute plasma half-

    life

    once bound to platelet receptors, resolution of

    abciximab blockade is prolonged

    50-60% residual inhibition remains 24 hours after

    terminating the infusion

    effective or biologic half-life for abciximab is estimated

    to be > 12 to 24 hours

    indications

    used together with aspirin and heparin as adjuvant

    therapy in patients undergoing high-risk angioplasty and

    atherectomy

    Integrelin

    synthetic peptide with high affinity for the GP IIb/IIIa

    integrin receptor protein

    for prevention of thrombosis in percutaneous coronary

    angioplasty

    Eptifibatide and tirofiban

    binds selectively to GP IIb/IIIa receptor

    renally excreted, no active metabolites

    half-life of 1.5 to 2.5 hours

    rapidly dissociate from glycoprotein receptors, with

    platelet aggregation returning to normal within 4 hours

    after discontinuation of the drug

    Anticoagulants and thrombolytic agentsNC Hwang 2008

  • 8/2/2019 Anticoagulants and Thrombolytic Agents

    12/12

    Drugs used in bleeding disorders

    correction of prothrombin activity: vitamin K

    plasma fractions: factors VIII, IX, and I

    fibrinolytic inhibitors: aminocaproic acid, tranexamic

    acid

    serine protease inhibitor: aprotinin

    CORRECTION OF PROTHROMBIN ACTIVITY

    Vitamin K

    being lipid soluble, vitamin K1 and K2 require bile salts

    for absorption from the intestinal tract

    available as 5 mg tablet, or 50 mg ampoule

    effect delayed for 6 hours, completed by 24 hours when

    treating depression of prothrombin activity after warfarin

    therapy or vitamin K deficiency

    intravenous infusion must be slow, rapid infusion can

    cause dyspnoea, chest and back pain, even death

    indications

    correction of prothrombin activity

    after warfarin therapy

    vitamin K deficiency in premature infants, inhospitalised patients in intensive care units

    because of poor diet, parenteral nutrition, recent

    surgery, multiple antibiotic therapy, uraemia

    severe hepatic failure results in loss of protein synthesis

    and a haemorrhagic diathesis that is unresponsive to

    vitamin K

    FIBRINOLYTIC INHIBITORS

    target sites

    Aminocaproic acid

    chemically similar to lysine, is a synthetic inhibitor of

    fibrinolysis

    mechanism of action

    binds to lysine residues on plasminogen and plasmin

    competitively inhibits plasminogen activation

    blocks binding of plasmin to fibrin

    pharmacokinetics

    rapidly absorbed orally

    cleared from the body via the kidney, 50% excretedunchanged in the urine within 12 hours

    indications

    haemophilia

    bleeding from fibrinolytic therapy

    prophylaxis for rebleeding from intracranial aneurysms

    postsurgical gastrointestinal bleeding

    postprostatectomy bleeeding

    adverse effects

    intravascular thrombosis from inhibition of plasminogen

    activator

    hypotension

    ureteral obstruction by clot formation in patients with

    haematuria

    myopathy and muscle necrosis

    abdominal discomfort

    diarrhoea

    nasal stuffiness

    Tranexamic acid

    trans-amino-methyl-cyclo-hexanoic acid (AMCHA)

    analog of aminocaproic acid and has the same properties

    potency increased by factor of 6-10 due to the distance

    between the 2 function groups in the AMCHA molecule

    is fixed by a cyclic structure

    mechanism of action

    occupies the lysine binding site of the plasminogen

    molecule producing a conformational change in the

    molecule, resulting in a fibrin polymer with greater

    resistance to natural fibrinolysis

    plasminogen activators when released, find less

    substrate that can be converted to plasmin

    dosing

    intravenously, 10-15 mg/kg 2-3 times a day

    oral dose 1-1.5g up to 4 times a day

    SERINE PROTEINASE INHIBITORS

    Aprotinin

    originally found to be a kallikrein inhibitor (1930) and

    trypsin inhibitor (1936)serine protease inhibitor (serpin) that

    inhibits fibrinolysis by free plasmin

    inhibits plasmin-streptokinase complex in

    patients who received the thrombolytic agent

    mechanism of action

    fits into enzyme where the contact region for the normal

    enzyme substrate is located

    forms 1:1 complexes with the enzymes which include

    trypsin, kallikreins from organs, tissues and

    plasma, and plasmin

    aprotinin acts as pseudo-substrate that prevents furtherproteolytic activity while it remains tightly bound to the

    enzyme

    administration and dosage

    aqueous solution is stable at room temperature without

    loss of activity

    historical reasons, quantities and concentrations

    expressed as kallikrein inactivator units KIU

    administered by infusion

    4 M results in 100% plasmin inhibition

    15M results in only 90% kallikrein inhibition

    t is 5-8 hoursindications

    patients at high risk of excessive bleeding

    cardiac reoperations

    adverse effects

    anaphylaxis

    first exposure, 6 months

    Drugs used in Bleeding DisordersNC Hwang 2008