Anda di halaman 1dari 12

NC Hwang 2008

Anticoagulants and thrombolytic agents


Coagulation cascade 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)
After injury to a vessel wall, tissue factor is exposed on inhibits Xa by forming a complex that
the surface of the damaged endothelium. can also inhibit VIIa bound to TF, but
The interaction of tissue factor with plasma factor VII not free VII
activates the coagulation cascade, producing thrombin by important for blocking the effect of TF
stepwise activation of a series of proenzymes where it is expressed on endothelial
The coagulation cascade is regulated by natural cells and subendothelial structures
anticoagulants, such as tissue factor pathway inhibitor proteins C and S
TFPI, the protein C and protein S system, and thrombin in conjunction with
antithrombin, all of which help to restrict the formation thrombomodulin, activates protein C
of the hemostatic plug to the site of injury. Protein C proteolytically cleaves VIIIa
and Va (major cofactors that help
produce Xa and IIa), with protein S
acting as cofactor
in septic patients, activated protein C
(available as a recombinant product)
inhibit DIVC that occurs in small
vessels
protein C may down-regulate
inflammatory cytokines
thrombomodulin
a constituent of the endothelial cell
membrane, very small amounts are
present in blood
binds thrombin and begins the
sequence of protein C activation
functions as cell-based
inhibition of coagulation
probably facilitates thrombin
catabolism

Effects of thrombin
converts soluble fibrinogen to fibrin
activates 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
NC Hwang 2008
Anticoagulants and thrombolytic agents
Thrombus formation at the site of damaged vessels 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
Platelet factors antihistamines
platelet factor 1: coagulation factor V nicotine
platelet factor 2: thromboplastic material elevated factor VIII
platelet factor 3: platelet thromboplastin tissue inflammation or trauma
platelet factor 4: antiheparin factor
platelet factor 5: fibrinogen coagulation factor Fibrinolytic system
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 of
International 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)
NC Hwang 2008
Anticoagulants and thrombolytic agents
DRUGS USED IN COAGULATION DISORDERS

ANTICOAGULANTS
Heparin
a heterogeneous group in the absence of heparin, formation of heparin-
a member of the heparan sulphate family of complex antithrombin-protease complexes is slow, in the presence
sugars classified under glycosaminoglycans of heparin, they are accelerated 1000-fold
glycosaminoglycans or mucopolysaccharides heparin catalyses the antithrombin-protease reaction
are polymers of repeating disaccharides without being consumed
within the disaccharides, the sugars once the antithrombin-protease complex is
tend to be modified, with acidic formed, heparin is released intact for renewed
groups, amino groups, sulfated binding to more AT III
hydroxyl and amino groups heparin-antithrombin III complex inactivates
tend to be negatively charged, because serine esterases
of the prevalence of acidic groups factors XIIa, XIa, Xa, IXa, IIa
heparan sulphate family of complex sugars is plasmin
composed of long chains of alternating kallilrein
disaccharide units of uronic acid (glucuronic only unbound Xa is sensitive to heparin activity
and iduronic acid) and glucosamine residues Xa bound to platelets in the prothrombinase
the backbone structure is then complex is protected from the heparin action
decorated by with complex patterns of once active thrombosis has developed, larger amounts of
sulphate and carboxyl groups at various heparin inhibit further coagulation by inactivating
positions, giving rise to a very strongly thrombin (IIa) and preventing the conversion of
negative charged molecule. fibrinogen to fibrin (I to Ia)
strongly acidic because of its content of covalently decreased platelet aggregation
linked sulfate and carboxylic acid groups (heparin reduction of platelet membrane receptors for
sodium) von Willebrand factor and fibrinogen (Ia)
has an extended helical conformation due to inhibition of VIIIa, Va
charge repulsion by the many negatively facilitating the release of tissue plasminogen
charged groups activator (tPA), resulting in an increase in
naturally occurring in the secretory granules of mast plasmin and D-dimer concentrations, both of
cells which interfere with platelet aggregation
has no anticoagulation effect by itself increased platelet aggregation
biologic activity is dependent upon the plasma protease binding of antiplatelet IgG antibodies to
inhibitor, anti-thrombin III (AT III) (heparin cofactor) platelet-bound heparin, activates platelets and
a specific pentasaccharide sequence containing a 3-O- induces platelet clumping (heparin-induced
sulphated glucosamine residue forms a high-affinity thrombocytopaenia, HIT)
binding site for AT III inhibition of VIIa-TF complex
AT III is one of the many naturally occurring by releasing TFPI from endothelial cells, renal
inhibitors in coagulation but its action is slow cells, carcinoma cells and lipoprotein fraction of
small amounts of heparin (with AT III) inhibit plasma
thrombosis by inactivating activated Factor X TFPI also inhibits Xa on TF bearing cell
(Xa) and inhibiting the conversion of releases lipoprotein lipase from capillary endothelial
prothrombin to thrombin (II to IIa) surfaces
tight binding of heparin molecule to AT III the enzyme hydrolyzes triacylglycerols in
causes a conformational change in this inhibitor, chylomicrons, very-low-density lipoproteins,
which exposes the active binding site of low-density lipoproteins, and diacylglycerols
antithrombin III for more rapid interaction with inhibits growth of capillary endothelial cells but
the proteases (activated clotting factors) to potentiates the activity of acid fibroblasts growth factors
inhibit the enzymes on these cells
NC Hwang 2008
Anticoagulants and thrombolytic agents
commercial preparations indications
consists of repeated sulphated mucopolysaccharides prevention of deep venous thrombosis
D-glucosamine-L-iduronic acid, and after hip replacement surgery
D-glucosamine-D-glucuronic acid treatment of acute deep vein
source thrombosis
porcine intestinal mucosa (higher potency) prophylaxis for ischaemic
bovine lung complications in unstable angina and
partial substitution of acidic protons of the sulfate units non-Q-wave myocardial infarction
by pharmacokinetics
Na+ (as heparin sodium) absorption
Ca++ (as calcium heparin) not absorbed from the intestinal mucosa,
Li+ (as lithium heparin) therefore administered parenterally (continuous
used in vitro as an anticoagulant for blood infusion, intermittent intravenous injection, deep
samples subcutaneous injection)
pH adjustment (between 5.0 and 7.5) onset of action
titrated with HCl or NaOH after intravenous administration, immediate
standardization of activity after subcutaneous administration, delay of 1-2
regular heparin consists of a family of hours
molecules of different molecular weights, the clearance
correlation between the concentration of a given degraded primarily by reticuloendothelial
heparin preparation and its effect on coagulation system, and heparinase
often is low small amount of undegraded heparin appears in
therefore standardized as units of activity by urine
bioassay t
heparin sodium must contain at least 120 USP dose-dependent
units per milligram 100 U/kg, 1 hour
1 U stops 1 ml of citrated sheep plasma from 400 U/kg, 2.5 hours
clotting for 1 hour after the addition of 0.2 ml of 800 U/kg, 5 hours
1% calcium chloride shortened in patients with pulmonary embolism
high molecular weight (HMW) fractions prolonged in patients with hepatic cirrhosis or
with high affinity for AT III, markedly inhibit renal failure
blood coagulation LMW heparins have longer biological half-
fractions have a MW range of 5 000-30 000 lives than do standard heparin
low molecular weight (LMW) fractions adverse effects
MW 1 000-10 000 (1-10kDa, mean 4.5 kDa) bleeding
less effect on antithrombin III monitor partial thromboplastin time (PTT)
activity dependent on number of elderly women and patients with renal failure
monosaccharide units per molecule are more prone to haemorrhage
< 8, no significant antithrombotic platelet dysfunction
activity thrombocytopenia (HIT)
8-18, potentiate inhibition of factor Xa platelet count of less than 50% of pretreatment
>18, potentiate inhibition of both value or <100,000/l during heparin therapy
factor Xa and thrombin may be due to binding of antiplatelet IgG
isolated from standard heparin antibodies to platelet-bound heparin, which
by gel filtration chromatography activates platelets and induces platelet clumping
by differential precipitation with may also lead to paradoxical thromboembolism
ethanol transient reversible allopecia
by partial depolymerization with telogen effluvium type alopecia
nitrous acid premature transformation of growing hairs into
by alkaline degradation of heparin the resting phase
benzyl ester / beta elimination osteoporosis and spontaneous fractures with long term
degradation (enoxaparin Na) heparin therapy (greater effect with regular heparin)
by enzymatic degradation activation of fibroblast growth factor-mediated
compared with regular heparin bone resorption
increased bioavailability after inhibition of lipoprotein-mediated carriage of
subcutaneous administration vitamin K
less frequent dosing requirements vitamin K is essential for bone density
units of reference via the gamma-carboxylation of Gla-
milligrams for enoxaparin containing bone proteins such as
anti-factor Xa units for dalteparin and osteocalcin
danaproid also evidence of reduction in bone zinc content,
increased in serum transaminase concentration
NC Hwang 2008
Anticoagulants and thrombolytic agents
contraindications reversal of heparinisation
hypersensitivity discontinuation of the drug
active bleeding protamine sulphate
haemophilia highly basic peptide that combines with heparin
thrombocytopaenia and history of HIT as an ion pair to form a stable complex devoid
purpura of anticoagulant activity
severe hypertension for every 100 U of heparin remaining in the
intracranial haemorrhage patient, 1 mg of protamine sulphate is
infective endocarditis administered intravenously
active tuberculosis excess protamine has an anticoagulant effect
ulcerative lesions of gastrointestinal tract binds to platelets and fibrinogen
threatened abortion metabolized by N-carboxypeptidase
visceral carcinoma
advance hepatic or renal disease Enoxaparin
during or after surgery of brain, spinal cord, or eye obtained by alkaline degradation of heparin benzyl ester
undergoing lumbar puncture or regional anaesthesia approximately one-third molecular size of standard
blocks heparin
administration and dosages
used in pregnant women only when clearly indicated Fondaparinux sodium
established venous thrombosis synthetic and specific inhibitor of activated factor X
maintain plasma concentration of 0.2 U/ml to (Xa)
prolong the PTT to INR 2-2.5 molecular weight is 1728
initial bolus injection of 5000-10000 U, supplied as a clear and colorless liquid with a pH
followed by infusion of 10-15 U/kg/h between 5.0 and 8.0
with acute pulmonary embolism, mechanism of action
larger doses require during the first few antithrombin III-mediated selective inhibition of factor
days because of increased heparin Xa, and potentiates (about 300 times) the innate
clearance neutralization of factor Xa by AT III
intermittent administration, 75-100 U/kg every does not inactivate thrombin and has no known effect on
4 hours platelet function
heparin resistance - causes does not bind significantly to other plasma proteins
increased serum concentration of other (acute reactant) (including platelet factor 4) or red blood cells
proteins that have affinity for heparin absorption
fibroblast growth factors (FGFs) rapidly and completely absorbed after administration by
vascular endothelial growth factor (VEGF) subcutaneous injection, bioavailability is 100%
heparin-binding EGF-like growth factor distribution
hepatocyte growth factor (HGF) in healthy adults, volume of distribution of 7-11 L
transforming growth factor-beta (TGF-beta) clearance
interferon-gamma (IFN-gamma) in healthy individuals up to 75 years of age, up to 77%
platelet-derived growth factor (PDGF) of a single subcutaneous or intravenous fondaparinux
platelet factor-4 (PF-4) dose is eliminated in urine as unchanged drug in 72 hours
interleukin-8 (IL-8) 25% lower in patients over 75 years of age
macrophage inflammatory protein-1 (MIP-1) elimination half-life is 17-21 hours
interferon-gamma inducible protein-10 (IP-10) total clearance is approximately lower in patients with
insulin-like growth factors I or II renal impairment
fibronectin drug interactions
laminin concomitant use of oral anticoagulants (warfarin),
histidine-rich glycoprotein vitronectin platelet inhibitors (acetylsalicylic acid), NSAIDs
increased concentration of factor VIII, a cofactor that (piroxicam) and digoxin did not significantly affect the
increases the proteolytic activity of IXa pharmacokinetics/pharmacodynamics of fondaparinux
congenital deficiency of AT III (concentration less than sodium
50% of normal), may be precipitated by pregnancy, does not influence the pharmacodynamics of warfarin,
infection or surgery acetylsalicylic acid, piroxicam, and digoxin, nor the
acquired deficiency of AT III (concentration less than pharmacokinetics of digoxin at steady state
25% of normal), may occur in patients with hepatic does not bind significantly to plasma proteins other than
cirrhosis, nephrotic syndrome, disseminated intravascular AT III, no drug interactions by protein-binding
coagulation displacement are expected
accelerated clearance of heparin, as may occur with
massive pulmonary embolism
NC Hwang 2008
Anticoagulants and thrombolytic agents
Warfarin pharmacokinetics
oral anticoagulant introduced as rodenticide in 1948 absorption
Wisconsin Alumni Research Foundation - arin acidic, available as a sodium salt
discovery of anticoagulant substance formed in spoiled rapid oral absorption, 100% bioavailability
sweet clover silage which produced a deficiency of decreased in malabsorption
plasma prothrombin and haemorrhagic disease in cattle detectable in plasma within 1 hour of oral
toxic agent identified as bishydroxycoumarin administration
and synthesized as dicoumarol peak plasma concentration in 2-8 hours
structure crosses placenta
4-hydroxycoumarin residue, with a non-polar carbon distribution
substituent at the 3-position, is the minimal structural 99% of racemic warfarin bound to plasma
requirement for activity albumin, which may contribute to its
this carbon is asymmetrical in warfarin small Vd (the albumin space) 0.14L/kg
commercial preparations are racemic clearance
mixture of 2 enantiomorphs 0.045 ml/kg/min
levorotatory S-warfarin transformed by CYP 1A2, CYP2C9 into
dextrorotatory R-warfarin inactive metabolite by the liver and kidney
potency: S-warfarin > R-warfarin, 4:1 long t of 25-60 hours in plasma,
mechanism of action prolonged with liver disease
warfarin prevents the -carboxylation of glutamate excreted via urine and stool
residues in factors II, VII, IX, and X adverse effects
by blocking the reduction of inactive vitamin K crosses placenta readily
epoxide (by vitamin K epoxide reductase) back causing haemorrhagic disorder in the foetus
to its active hydroquinone form affecting -carboxyglutamate residues in foetal
results in incomplete molecules that are bone and blood proteins and causing birth defect
biologically inactive in coagulation 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 haemorrhagic
infarction 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
role of vitamin K maintenance dose of 5-7 mg/day, may be up to 15
post-ribosomal modification of factors II, VII, IX, and mg/day
X, and the endogenous anticoagulant protein C and S in INR of 2.5-3.5 for patients with prosthetic heart valves
the liver drug interactions
involves -carboxylation of glutamate residues pharmacokinetic mechanisms
in them increasing or decreasing anticoagulant effect
the carboxyl-glutamyl residues are responsible for the and the risk of bleeding by
binding of Ca++ which are necessary for the binding of enzyme induction or inhibition
the activated factors to phospholipid vesicles variation in plasma protein binding
this process is coupled with the oxidative deactivation of increasing activity of warfarin
vitamin K stereoselective inhibition of oxidative
in vitamin K deficiency, inactive precursors are metabolism of S-warfarin, resulting in
liberated hypoprothrombinaemia
anticoagulant effects pyrazolones, phenylbutazone,
8-12 hour delay in the action of warfarin, duration of metronidazole, fluconazole,
action 2-5 days trimethoprim-sulpha methoxazole
anticoagulant effect of warfarin results from a balance inhibition of metabolism of warfarin
between partially inhibited synthesis and degradation rate amiodarone, disulfiram, cimetidine,
of the 4 vitamin K-dependent clotting factors chloramphenicol
t: 6 h (VII), 24 h (IX), 36 h (X), 50 h (II) displacement of albumin-bound warfarin,
large initial doses of warfarin (0.75/kg) hasten the onset increasing the free fraction
of anticoagulation effect, beyond this dosage, the speed pyrazolones, phenylbutazone,
of onset is independent of the dose size; only effect of a sulfinpyrazone, NSAID, chloral
large loading dose is the prolongation of t of the drug hydrate, mefenamic acid
NC Hwang 2008
Anticoagulants and thrombolytic agents
decreasing activity of warfarin parenteral DTIs:
induction of hepatic enzymes that metabolise hirudin and argatroban for the treatment of
warfarin heparin-induced thrombocytopenia
barbiturates, rifampicin, alcohol bivalirudin as an alternative to heparin in
reduction in absorption and bioavailability percutaneous coronary intervention, and
cholestyramine binds warfarin in the desirudin as prophylaxis against venous
intestine thromboembolism in hip replacement
increased albumin binding
pharmacodynamic mechanisms Hirudin
synergism(impaired haemostasis, reduced source
clotting factor synthesis in hepatic disease, Hirudo medicinalis leeches,
heparin, NSAIDs) powerful and specific thrombin inhibitor
competitive antagonism (vitamin K) recombinant DNA
altered physiologic control loop for vitamin K mechanism of action
(hereditary resistance to oral anticoagulant binds to active site of thrombin
augmentation of anticoagulant effect can reach and inactivate fibrin-bound thrombin
inhibition of platelet function little effects on platelets or bleeding time
pyrazolones, phenylbutazone, aspirin administration
increasing turnover of clotting factors parenterally
liver disease, hyperthyroidism monitored by partial thromboplastin time
inhibition of activity of VIIa, IXa, Xa, IIa
heparin, (prolonging prothombin time) Ximelagatran
vitamin K production first oral direct thrombin inhibitor to be introduced
elimination of vitamin K producing a prodrug, its active metabolite is melagatran which
bacteria in gastrointestinal tract by directly inhibits thrombin
third generation cephalosporins ximelagatran developed to enhance bioavailability of
direct inhibition of vitamin K epoxide melagatran, the molecules of latter are charged and
reductase become highly hydrophilic at intestinal pH, resulting in
third generation cephalosporins low absorption
reduction of anticoagulant effect melagatran can also be given by injection
increase synthesis of clotting factors with melagatran resembles a peptide sequence on
vitamin K fibrinogens A- chain, where thrombin-induced
increase supply of clotting factors with cleavage occurs
transfusion of fresh frozen plasma it binds reversibly to the active site of thrombin and
clotting factor concentration through inhibits the normal function of thrombin, including both
haemoconcentration with diuretics free and clot-bound thrombin
hereditary resistance to warfarin via mutation the ability to bind clot-bound thrombin is an
of vitamin K epoxide reductase advantage since clot-bound thrombin may retain
decreasing turnover rate of clotting factors its enzymatic activity and continue to stimulate
hypothyroidism the coagulation cascade
regulatory approval for ximelagatran in France for the
reversal of antocoagulant effects prevention of venous thromboembolic events in major
disappearance of anticoagulant effect is due to orthopaedic (hip or knee replacement) surgery
reestablishment of normal activity of the does not have the same difficulties with dose adjustment
clotting factors or drug interaction as warfarin does
does not correlate with plasma concentration of no known antidote
warfarin pharmacokinetics
depends on the degree of correction required rapidly absorbed after oral administration, peak
stopping warfarin alone with or without concentration Cmax achieved 1 h after administration
large doses of vitamin K (phytonadione) has a rapid onset and offset of action and shows low
50 mg infusion potential for food and drug interactions
fresh frozen plasma oral bioavailability approximately 20%, compared with
factor IX concentrates 3%-7% for melagatran
whole blood transfusion ximelagatran Vd 2.53L/kg
pharmaokinetics of melagatran described as linear, first-
Direct thrombin inhibitor order, one compartment model
bind directly to thrombin and block its interaction with melagatran excreted unchanged via kidneys, t 3h
its substrates renal clearance rate
recombinant hirudins, bivalirudin, and young 7.7L/h, elderly 5 L/h
ximelagatran affected in patients with severe renal
impairment, defined as creatinine
clearance rate of <30ml/min
NC Hwang 2008
Anticoagulants and thrombolytic agents
adverse effects Streptokinase
nausea, diarrhoea, headache exotoxin of -haemolytic streptococci
bleeding, but not worse than with enoxaparin or antigenic
dalteparin forms a stable, noncovalent 1:1 complex with free
raised hepatic ALT and bilirubin concentrations circulating plasminogen
contraindications produces a conformational change that exposes the
renal, and hepatic impairment active site on plasminogen that cleaves arginine 560 on
pregnancy free plasminogen molecules to form free plasmin
infant and children complex is not inhibited by 2-antiplasmin
precautions not fibrin specific, readily induces a systemic lytic state
lactating women t 40-80 minutes
adverse effects
main properties and pharmacokinetic characteristics 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,
FIBRINOLYTIC DRUGS this modification confers clot selectivity
advantages
allows for rapid intravenous injection
greater clot selectivity

Tissue plasminogen activator


preferentially activates plasminogen that is bound to
fibrin, 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
mode of action metabolised by liver, t 5-10 minutes
rapid lysis of haemostatic thrombus and target produced by recombinant DNA technology
thromboemboli by catalysing the formation of serine alteplase is unmodified t-PA
protease plasmin from its precursor zymogen reteplase is t-PA from which several amino
plasminogen acids have been deleted
can cause a generalized lytic state after
intravenous administration Urokinase
(zymogen = inactive precursor of proteolytic human enzyme synthesized by kidney, therefore not
enzyme) antigenic
activation of free circulating plasminogen directly converts free plasminogen to plasmin
streptokinase lacks fibrin specificity, readily induces a systemic lytic
urokinase state
activation of fibrin bound plasminogen metabolised by liver, t of 15-20 minutes
anistreplase
alteplase Prourokinase
reteplase a zymogenic plasminogen activator, a precursor of
tissue plasminogen activator (t-PA) urokinase
indications binds to fibrin before activation
multiple pulmonary emboli that are massive enough to has selectivity for clots
require surgical intervention
central deep vein thrombosis of the superior venous
cava, iliofemoral veins
coronary thrombolysis after acute myocardial infarction
peripheral arterial disease
NC Hwang 2008
Anticoagulants and thrombolytic agents
administration and dosages Cyclooxygenase inhibition
streptokinase (US$_00) aspirin
intravenous infusion, loading dose 250 000 U inhibition of the synthesis of thromboxane A2
to overcome plasma antibodies directed against by irreversible, covalent acetylation of a serine
the protein (from prior streptococcal infection), residue near the active site of cyclooxygenase
followed by maintenance dose of 100 000 the anuclear platelet cannot synthesize new
U/hour for 24-72 hours proteins or enzymes during its 7-10-day life-
follow with full heparinisation as plasminogen span
is exhausted repeated doses produce a cumulative effect on
can act as antigen, patients with antibodies to platelet function
streptokinase can develop fever, allergic maximally effective as an antithrombotic agent
reactions, therapeutic resistance at doses of 160mg to 320 mg/day
urokinase (US$_000) higher doses inhibit the production of
intravenous infusion, loading dose 1000 to prostacyclin, an antithrombotic eicosanoid
4500 U/kg over 10 minutes, followed by produced by the endothelium
maintenance dose of 4400 U/kg/hour for 12 prolongs bleeding time
hours other NSAIDS
alteplase (t-PA) ($_000) other salicylates and other nonsteroidal anti-
accelerated regime for coronary thrombolysis inflammatory drugs also inhibit cyclooxygenase
intravenous infusion, loading dose 15 mg but have a shorter duration of inhibitory action
followed by 0.75mg/kg over 30 minutes because they cannot acetylate cyclooxygenase,
(maximum 50mg), followed by 0.5mg/kg therefore their action is reversible
(maximum 35mg) over the following hour
reteplase ($_000) Increasing platelet cAMP
administered as 2 intravenous bolus injections following platelet activation, Ca++ is released from its
of 10 U each separated by 30 minutes storage sites (platelet dense tubular systems) to the
anistreplase ($_000) platelet cytoplasm resulting in an increase of cytosolic
single intravenous bolus injection of 30 U over free Ca++
3-5 minutes Ca++ mobilization is directly involved in
platelet activation and Ca++ is an important
ANTITHROMBOTIC DRUGS second messenger for signal transduction in
prevention of vascular events among patients with platelets
transient ischaemic attacks cAMP is another second messenger which opposes the
complete strokes effect of Ca++ by causing sequestration of cytosolic Ca++
angina pectoris to the Ca++ storage sites
4 main groups agents which increase cAMP will suppress platelet
cyclo-oxygenase inhibitors: e.g. aspirin activation
increasing platelet cAMP: stimulation of platelet adenylyl cyclase: action
by stimulation of adenylyl cyclase: of adenosine on platelet A2 receptor
adenosine inhibition of platelet phosphodiesterase:
by inhibition of phosphodiesterase: dipyridamole
dipyrimadole does not prolong bleeding time
ADP receptor antagonists: e.g. clopidogrel only current recommendation is for
GP IIb,IIIa blockers: tirofiban, abciximab primary prophylaxis of thromboemboli
in patients with prosthetic heart valves,
target sites of antithrombotic drugs on the platelet and is given in combination with
warfarin
dipyridamole is metabolized in the
liver and has a terminal half-life of 10h
NC Hwang 2008
Anticoagulants and thrombolytic agents
ADP receptor antagonism Clopidogrel
Ticlopidine analogue of ticlopidine, another ADP antagonist from
a thienopyridine derivative, interferes selectively with the thienopyridine group that inhibits ADP and thrombin-
ADP-induced transformation of GPIIb/IIIa complex induced platelet aggregation
expression in activated platelets a prodrug, activated by cytochrome P450 predominantly
it also inhibits platelet aggregation induced by thrombin, by CYP3A4 (less by CYP3A5) to a metabolite that
collagen, arachidonic acid, platelet-activating factor, inhibits ADP-induced platelet aggregation
prostaglandin endoperoxides, thromboxane A2-like by binding the ADP receptor the drug selectively
substance, serotonin, and epinephrine 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
pharmacokinetics co-administration of HMG-CoA reductase
absorption inhibitors will diminish the activation of
well absorbed after oral administration clopidogrel
effect within 48 hours the principal circulating metabolite is an inactive
maximal effect after approximately 3 to 5 days carboxylic acid derivative
activity still present 72 hours after last dose has an 8 hour elimination half-life, but the
absorption decreased by concurrent antacid pharmacologic half-life is relatively long and it takes 4 to
therapy 7 days of administration to reach a steady state effect on
metabolism platelets
rapidly and extensively metabolised in liver side effects include thrombocytopenia, neutropenia
and excreted in urine clopidogrel-induced platelet inhibition persists several
one of its metabolite is active days after withdrawal of the drug and diminishes in
half-life at steady state is 4-5 days proportion to platelet renewal
in comparison with ticlopidine, clopidogrel is more
effects potent, with less degree of neutropenia
prolongs bleeding time, maximum effect after several clopidogrel is significantly more active than aspirin.
days of treatment compared with aspirin, clopidogrel has less
antiplatelet activity persists for a week or longer after severe gastrointestinal bleeding but more severe
treatment is discontinued rash incidence
possibly due to action of active metabolite
act independent of aspirin with no effect on eicosanoid Platelet GP IIb/IIIa receptor antagonism
metabolism mechanism of action
blocks platelet receptors for integrin and fibrinogen
adverse effects adverse effects
gastrointestinal (20%): nausea, dyspepsia, diarrhoea bleeding
haemorrhage (5%) immunogenicity
bone marrow suppression thrombocytopenia
leucopenia (1%): detected by regular in approximately 0.1% to 0.5% of patients,
monitoring of white cell count during the first 3 platelet count of <20000/L occurs after
months of therapy intravenous administration
thrombocytopenia due to GPIIb/IIIa antagonist-dependent
agranulocytosis antibody binding to the platelet fibrinogen
aplastic anaemia leading to platelet activation and accelerated
cholestatic jaundice platelet clearance, leading to thrombocytopenia
elevated serum cholesterol concentration
rashes Abciximab
chimeric human / mouse monoclonal antibody
indications MW approximately 50000 daltons
prevention of thrombosis in cerebral vascular and binds nonselectively to GP IIb/IIIa receptors and
coronary artery disease vitronectin receptors on endothelial and smooth muscle
for patients who are unable to tolerate aspirin cells
NC Hwang 2008
Anticoagulants and thrombolytic agents
free unbound abciximab undergoes rapid proteolytic Integrelin
degradation, resulting in a 10- to 15-minute plasma half- synthetic peptide with high affinity for the GP IIb/IIIa
life integrin receptor protein
once bound to platelet receptors, resolution of for prevention of thrombosis in percutaneous coronary
abciximab blockade is prolonged angioplasty
50-60% residual inhibition remains 24 hours after
terminating the infusion Eptifibatide and tirofiban
effective or biologic half-life for abciximab is estimated binds selectively to GP IIb/IIIa receptor
to be > 12 to 24 hours renally excreted, no active metabolites
half-life of 1.5 to 2.5 hours
indications rapidly dissociate from glycoprotein receptors, with
used together with aspirin and heparin as adjuvant platelet aggregation returning to normal within 4 hours
therapy in patients undergoing high-risk angioplasty and after discontinuation of the drug
atherectomy
NC Hwang 2008
Drugs used in Bleeding Disorders
Drugs used in bleeding disorders adverse effects
correction of prothrombin activity: vitamin K intravascular thrombosis from inhibition of plasminogen
plasma fractions: factors VIII, IX, and I activator
fibrinolytic inhibitors: aminocaproic acid, tranexamic hypotension
acid ureteral obstruction by clot formation in patients with
serine protease inhibitor: aprotinin haematuria
myopathy and muscle necrosis
CORRECTION OF PROTHROMBIN ACTIVITY abdominal discomfort
Vitamin K diarrhoea
being lipid soluble, vitamin K1 and K2 require bile salts nasal stuffiness
for absorption from the intestinal tract
available as 5 mg tablet, or 50 mg ampoule Tranexamic acid
effect delayed for 6 hours, completed by 24 hours when trans-amino-methyl-cyclo-hexanoic acid (AMCHA)
treating depression of prothrombin activity after warfarin analog of aminocaproic acid and has the same properties
therapy or vitamin K deficiency potency increased by factor of 6-10 due to the distance
intravenous infusion must be slow, rapid infusion can between the 2 function groups in the AMCHA molecule
cause dyspnoea, chest and back pain, even death is fixed by a cyclic structure
indications mechanism of action
correction of prothrombin activity occupies the lysine binding site of the plasminogen
after warfarin therapy molecule producing a conformational change in the
vitamin K deficiency in premature infants, in molecule, resulting in a fibrin polymer with greater
hospitalised patients in intensive care units resistance to natural fibrinolysis
because of poor diet, parenteral nutrition, recent plasminogen activators when released, find less
surgery, multiple antibiotic therapy, uraemia substrate that can be converted to plasmin
severe hepatic failure results in loss of protein synthesis dosing
and a haemorrhagic diathesis that is unresponsive to intravenously, 10-15 mg/kg 2-3 times a day
vitamin K oral dose 1-1.5g up to 4 times a day

FIBRINOLYTIC INHIBITORS SERINE PROTEINASE INHIBITORS


target sites 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 further
proteolytic activity while it remains tightly bound to the
Aminocaproic acid enzyme
chemically similar to lysine, is a synthetic inhibitor of administration and dosage
fibrinolysis aqueous solution is stable at room temperature without
mechanism of action loss of activity
binds to lysine residues on plasminogen and plasmin historical reasons, quantities and concentrations
competitively inhibits plasminogen activation expressed as kallikrein inactivator units KIU
blocks binding of plasmin to fibrin administered by infusion
pharmacokinetics 4 M results in 100% plasmin inhibition
rapidly absorbed orally 15M results in only 90% kallikrein inhibition
cleared from the body via the kidney, 50% excreted t is 5-8 hours
unchanged in the urine within 12 hours indications
indications patients at high risk of excessive bleeding
haemophilia cardiac reoperations
bleeding from fibrinolytic therapy adverse effects
prophylaxis for rebleeding from intracranial aneurysms anaphylaxis
postsurgical gastrointestinal bleeding first exposure, <0.1%
postprostatectomy bleeeding re-exposure: 5% < 6 months, 0.9% > 6 months
radiation- and drug-induced haemorrhagic cystitis renal dysfunction 4.4%

Anda mungkin juga menyukai