Download presentation
Presentation is loading. Please wait.
1
Clinical Use of Coagulation Inhibitors
Aleksandra Milosevski Shi Yi Chen Ya Ping Guo Vanessa Hoi PHM Fall 2019 Instructor: Chesa Dojo Soeandy Coordinator: Jeffrey Henderson
2
Coagulation Cascade Overview
- When a blood vessel is damaged a series of enzymatic reactions known as the coagulation cascade are initiated. The end goal of this cascade is to create a polymer of fibrin proteins in order to form a thrombus, or a blood clot at the site of damage. This cascade is divided into 2 pathways, the extrinsic and intrinsic pathways. These pathways will merge to form the common pathway at which point inactive prothrombin will be cleaved to form active thrombin, the enzyme that is responsible for converting soluble fibrinogen into insoluble fibrin. Silverthorn, Fig. 16.4
3
Extrinsic Pathway Tissue Factor (III) → found in tissue underlying blood vessels Tissue Factor (III) + Active VII Form Complex End result → X activated Positive feedback observed The extrinsic pathway will start when damage to the tissues exposes tissue factor 3, a protein found in the tissue underlying the blood vessel. The tissue factor 3 protein will then activate clotting factor 7 and together will form a complex that will in turn convert inactive clotting factor 10 into active factor 10. The complex will also activate factor 9, which is needed in the intrinsic pathway. In addition, active factor 10 will activate more factor 7 in a positive feedback fashion. Silverthorn, Fig. 16.4
4
Intrinsic Pathway Blood vessel damage exposes collagen
Factors XII, XI, IX, VII activated Factor X also activated The intrinsic pathway will begin when blood vessel damage exposes collagen. This will set off a series of steps the first being collagen will convert inactive clotting factor 12 to active factor 12. Activated factor 12 will then activate factor 11. Factor 11 will activate factor 9 and factor 8. Factor 9 will then activate factor 10. Silverthorn, Fig. 16.4
5
Common Pathway Silverthorn, Fig. 16.4
Active Factor X cleaves prothrombin → thrombin Fibrinogen → Fibrin Factor XIII also activated Both the intrinsic and extrinsic pathways lead to factor 10 activation and at this point merge to form the common pathway. Factor 10 will then cleave prothrombin to form thrombin which in turn will cleave fibrinogen to fibrin. Thrombin is also responsible for activating factor 13, which crosslinks the fibrin strands in different places to form a strong mesh like network and ultimately help form the clot. To understand why and when certain anticoagulants are used clinically, it is important to know the mechanism of action and the risks and benefits associated with each, which is what we’ll talk about next.
6
Vitamin K in Coagulation Cascade
Protein carboxylation reaction activates the clotting factors Coupled to the oxidation of vitamin K Vitamin K is reused Must be reduced to reactivate it via VKOR Warfarin: Vitamin K antagonist There are many cofactors needed in the coagulation cascade and one of them is vitamin K. It is needed in the synthesis of prothrombin, clotting factor 7, 9, 10 present in both intrinsic and extrinsic pathways. Silverthorn, Fig. 16.4
7
Vitamin K in Coagulation Cascade
Protein carboxylation reaction activates the clotting factors Coupled to the oxidation of vitamin K Vitamin K is reused Must be reduced to reactivate it via VKOR Warfarin: Vitamin K antagonist Inhibit VKOR Inhibition of VKOR → inhibits reactivation of vitamin K → decreases the synthesis of clotting factors VKOR In order to activate the clotting factors, it needs to undergo a carboxylation rxn which is coupled to the oxidation of vitamin K With limited vitamin K storage capacity in the body, vitamin K is reused through an oxidation-reduction cycle where the vitamin must be reduced to reactivate it by vitamin K epoxide reductase And warfarin, one of the commonly used drug to prevent blood clot formation is a vitamin K antagonist. It reduces the regeneration of vit K by inhibiting vitamin K epoxide reductase (VKOR) in the liver Inhibition of VKOR → inhibits reactivation of vitamin K → decreases the synthesis of these clotting factors Katzung, Fig. 34.6
8
Warfarin: Vitamin K antagonist
Clinical Use: Prophylaxis and/or treatment of venous thrombosis Atrial fibrillation Prosthetic heart valves Pharmacokinetics: Completely absorbed after oral administration Biotransformed in liver and excreted by the kidney Very little Warfarin is excreted unchanged in the urine → elimination by metabolism 8-12h delay in the action of warfarin Peak anticoagulant effect may be delayed 3-4 days Half-life: 20-60h Over 99% of the drug is bound to plasma albumin The primary medical use for warfarin is for prevention and treatment of venous thrombosis and it is a long-term therapy for all patients with mechanical heart valves. Since it is metabolized by the liver, patients with renal impairment can take this medication if their liver is healthy. Warfarin is completely absorbed after oral administration but there is an 8 to 12 hour delay in the action of warfarin and peak anticoagulant effect may be delayed 3-4 days. The effective half-life of warfarin ranges from 20-60h b/c over 99% of the drug is bound to plasma albumin.
9
Adverse Effects & Warnings/Precautions
Excessive bleeding Reversed by stopping the drug and administering vitamin K, fresh-frozen plasma Narrow therapeutic index Requires frequent monitoring Dosing is affected by polymorphism of the CYP2C9 enzyme and VKOR Dose is individualized Can cross placenta Cause birth malformations and fatal hemorrhage in fetus Up to 160 drugs would have potential drug interaction with warfarin such as acetaminophen and amoxicillin (CYP2C9, VKOR) 46 reported to have interactions such as atorvastatin and ranitidine Warfarin can have dangerous side effects and interactions. One of the side effects is excessive bleeding. But for warfarin, excessive anticoagulant effect and bleeding can be reversed by administering vitamin K or fresh-frozen plasma that contains all coagulation factors. Warfarin has a very narrow therapeutic index and inherited polymorphisms in CYP2C9 and VKORC1 can have significant effects on warfarin dosing therefore dosing must be individualized and requires routine monitoring, pregnant women should avoid taking this medication since warfarin can cross the placenta readily and cause hemorrhagic disorders in fetus. Since up to 160 drugs would have potential drug interactions with warfarin and vitamin K intake will interfere with the effectiveness of warfarin, patients on this medication will have dietary restrictions and drug interaction monitoring.
10
Direct Factor Xa Inhibitors (Rivaroxaban, Apixaban, Edoxaban)
Activation of Factor-X to Factor-Xa (FXa) FXa and FVa form prothrombinase complex Converts prothrombin to thrombin Ultimately increases clot formation Silverthorn, Fig. 16.4 Perzborn, Fig. 1 Coagulation cascade exhibits amplification FXa causes an explosive burst of thrombin and clot formation The next drug is a novel oral anticoagulant Activation of Factor-X is the start of the common pathway. FXa and FVa form the prothrombinase complex, converting prothrombin to thrombin and increasing clot formation. Factor Xa plays a pivotal role and causes an explosive burst in thrombin due to amplification of the coagulation cascade. Wheeless, 2015
11
Direct Factor Xa Inhibitors (Rivaroxaban, Apixaban, Edoxaban)
Activation of Factor-X to Factor-Xa (FXa) FXa and FVa form prothrombinase complex Converts prothrombin to thrombin Ultimately increases clot formation Silverthorn, Fig. 16.4 Perzborn, Fig. 1 Coagulation cascade exhibits amplification FXa causes an explosive burst of thrombin and clot formation Inhibition of FXa terminates thrombin generation and decreases coagulation. Inhibition of FXa terminates thrombin generation, thus decreasing coagulation. Wheeless, 2015
12
Binds free FXa and FXa bound in Prothombinase.
Rivaroxaban is a highly selective, competitive, direct, antithrombin-independent Factor Xa inhibitor. Binds free FXa and FXa bound in Prothombinase. This is exactly what rivaroxaban does; it is a highly selective, competitive, and direct antithrombin-independent Factor-Xa inhibitor. It binds free and prothrombinase-bound FXa. Perzborn, Fig. 2
13
Indications and Clinical Use
Rivaroxaban 10 mg, 15 mg, and 20 mg is indicated for: prevention of venous thromboembolic events (VTE) in patients after total hip or knee replacement surgery treatment of VTE like deep vein thrombosis (DVT) and pulmonary embolism (PE), and prevention of recurrence prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation (AF) No data to support safety and efficacy in patients with prosthetic heart valves Rivaroxaban 2.5 mg in combination with 75 mg-100 mg acetylsalicylic acid is indicated for: prevention of stroke, myocardial infarction, and cardiovascular death prevention of acute limb ischemia and mortality in coronary artery disease (CAD) Indications and Clinical uses for 10, 15, and 20 mg doses are similar to warfarin which include the prevention and treatment of VTE events like DVTs and PEs, especially after total knee or hip replacement surgeries; as well as prevention of stroke and systemic embolism in patients with non-valvular AF. Unlike warfarin, there have been no studies to support safety and efficacy in patients with prosthetic heart valves. 2.5 mg doses in combination with aspirin was recently indicated for: prevention of stroke, myocardial infarction, and cardiovascular death in patients with coronary artery disease.
14
Pharmacokinetics of Rivaroxaban
Elimination Approximately 1/3 of dose eliminated as unchanged active drug by kidneys 30% via active renal secretion, transporters involved are P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) 6% via glomerular filtration Approximately 2/3 of dose metabolically deactivated CYP 3A4/5 and CYP2J2 via hepatic oxidative biotransformation CYP-independent hydrolysis of amide bonds Resulting metabolites eliminated renally and via the hepatobiliary route Half-life: 5–9 hours (shorter than warfarin) Absorption, Bioavailability, Protein Binding, Distribution High Oral Bioavailability (Foral): 2.5 mg, 5 mg, 10 mg (with or without food) or 20 mg (with food): 80% - 100% 20 mg (without food): 66% Peak plasma level (t max): 2 – 4 hours High reversible plasma protein binding approximately 92–95 % in vitro mainly bound to serum albumin Volume of distribution at steady state = 50 L (0.62 L/kg) low-to-moderate affinity to peripheral tissues Here are some pharmacokinetic properties: Main things I wanted to point out were that rivaroxaban has high oral bioavailability and plasma protein binding. It has a rapid onset of action and shorter half-life than warfarin. It’s elimination contrasts warfarin’s, in that 1/3 of a dose is excreted as unchanged active drug by the kidneys, via P-glycoprotein and breast cancer resistance protein. The rest is metabolically deactivated by CYP 3A4/5 and CYP2J2 via hepatic oxidative biotransformation and CYP- independent hydrolysis
15
Pros & Cons CONS PROS no standardized test should monitoring be needed
wide therapeutic window and predictable pharmacokinetics fixed-dose oral administration regimens that do not require monitoring limited known drug interactions and no known dietary restrictions Andexanet, a FXa decoy, recently approved as antidote CONS no standardized test should monitoring be needed expensive PROS of using Rivaroxaban are that: It has a wide therapeutic window and predictable pharmacokinetics It follows fixed-dose oral administration regimens that do not require monitoring It has limited known drug interactions and no known dietary restrictions and Andexanet, a FXa decoy has also been recently approved as an antidote Some CONS are that: There exists no standardized test should monitoring be needed and it is expensive
16
Adverse Reactions/Contraindications/Warnings
increased risk of bleeding which can result in weakness, paleness, dizziness, or swelling and secondary complications like renal failure Contraindications no data to support use in children; pregnant or nursing women; patients with prosthetic heart valves; patients with clinically significant bleeding (ulcers, cerebral infarction etc.) Warnings and Precautions concomitant use with strong inhibitors of CYP3A4 and P-glycoprotein (ketoconazole, itraconazole, posaconazole, ritonavir) concomitant use with any other hemostasis-affecting drugs (non-steroidal anti-inflammatory drugs, acetylsalicylic acid, platelet aggregation inhibitors or selective serotonin reuptake inhibitors, and serotonin norepinephrine reuptake inhibitors) hepatic disease and renal impairment; must first determine creatinine clearance in all patients Furthermore, Adverse Reactions include increased risk of bleeding which can result in weakness, paleness, dizziness, or swelling and secondary complications like renal failure There is no data to support use in children, and pregnant, or nursing women; and those with clinically significant bleeding Lastly, warning and precautions should be taken for patients who concomitantly use strong inhibitors of CYP3A4 and P-glycoprotein or any other hemostasis-affecting drugs; AND in those with hepatic disease and renal impairment: you must first determine creatinine clearance in all patients
17
Heparin Mixture of mucopolysaccharides Co-factor of anti-thrombin
Target: Thrombin The last coagulation inhibitor is heparin. It is a mixture of mucopolysaccharides that are naturally produced in our body to stop blood clot. It works with anti-thrombin to inactivate thrombin within the common pathway. To understand heparin, we must first understand anti-thrombin. Silverthorn, Fig. 16.4
18
Heparin: Mechanism of Action
Enhance Anti-Thrombin Activity by 1000 times Inhibit thrombin Inhibit factor X Anti-Thrombin Slow Anti-thrombin itself can bind to thrombin and inactivate it. Therefore, stop the blood from clotting. Without heparin, this reaction only happens slowly Silverthorn, Fig. 16.4
19
Heparin: Mechanism of Action
Enhance Anti-Thrombin Activity by 1000 times Inhibit thrombin Inhibit factor X Heparin Anti-Thrombin with heparin, the reaction is accelerated by 1000 times. Because heparin can bind to anti-thrombin, induce a conformational change, and expose its active site for a rapid reaction Silverthorn, Fig. 16.4
20
Heparin: Mechanism of Action
Enhance Anti-Thrombin by 1000 times Inhibit thrombin Inhibit factor X Heparin Anti-Thrombin Besides, heparin can also work with anti-thrombin to inactivate factor ten. Silverthorn, Fig. 16.4
21
Heparin: Clinical Use Administration and onset Half-life: 90 mins
IV: immediate Deep subcutaneous: might be delayed mins No oral: Negatively charged and large Half-life: 90 mins Commonly used for Cardiac surgery Prevention of venous thromboembolism The onset of action is immediate after IV injection but can be delayed following subcutaneous injection. Due to the negative charge and large molecular size, it cannot pass through the oral route. Heparin can be used in some cardiac surgery to prevent venous thromboembolism
22
Heparin: pros and cons Advantages Disadvantages Safe for pregnancy
Antidote available: Protamine Protamine: positively charged Heparin: negatively charged Adverse effect Bleeding Platelet deficiency Monitoring required Effect depending on [anti-thrombin] No anti-thrombin, no action One of the advantages is that it is safe to be used during pregnancy because it cannot pass through placenta. There is also an antidote available. Protamine is a positively charged protein. It strongly binds to the negatively charged heparin and inactivates it. The downside of heparin would be the possible adverse effects. It could cause bleeding and platelet deficiency. Therefore, patients must be closely monitored after administration of heparin. Heparin has another problem too. It must work with anti-thrombin III to inhibit coagulation. If the patient has low anti-thrombin III level, its effect would be limited.
23
Heparin: LMWH Low molecular weight heparin (LMWH)
Fragmented heparin Weaker anticoagulation CANNOT inactivate thrombin CAN inactivate Factor X c Heparin c Anti-Thrombin Heparin can also come in a fragmented form. It is called low molecular weight heparin (LMWH). The reason to cut it down is to get weaker anticoagulation. Silverthorn, Fig. 16.4
24
Heparin: LMWH only Low molecular weight heparin (LMWH)
Fragmented heparin Weaker anticoagulation CANNOT inactivate thrombin CAN inactivate Factor X c only Heparin Anti-Thrombin The shorter version cannot stabilize the anti-thrombin - thrombin complex and therefore cannot inactivate thrombin. However, it can still inactivate factor ten. Silverthorn, Fig. 16.4
25
Heparin: LMWH vs. unfractionated
More predictable pharmacokinetic No monitoring is required Different onset and half-life: not interchangeable Unfractionated Low molecular weight Onset Immediate after IV; 20-60 after deep subcutaneous injection 3-6 hours after subcutaneous injection Half-life About 90 mins Varies between agents: Eg. Dalteparin 3-4 hours This selective inactivation allows its pharmacokinetics to be more predictable. Therefore, no monitoring is required, although bleeding is still a possible adverse effect. We also need to keep in mind that the onset and half-life are both very different then the unfractionated heparin. They are not interchangeable.
26
Summary Warfarin Direct Factor Xa Inhibitor (Rivaroxaban) Heparin
In summary, anticoagulants have no direct effect on an established thrombus, nor do they reverse ischemic tissue damage. However, once a thrombus has occurred, the goal of anticoagulant treatment is to prevent further extension of the formed clot and prevent complications. Today we talked about 3 of many anticoagulants, which we have summarized in this table.
27
Summary Drug Class Examples Use Mechanism of Action Pros Cons
Vitamin K Antagonist Warfarin Prevention and treatment of venous thromboembolic events (deep vein thrombosis, pulmonary embolism) Vitamin K antagonist Decreases the synthesis of clotting factor II (prothrombin) VII, IX, X Cheap Antidote available (vitamin K, fresh-frozen plasma) Narrow therapeutic index → requires frequent monitoring Can cross placenta → cause birth malformations and fatal hemorrhage in fetus Require dietary restrictions and drug interaction monitoring Direct Factor Xa Inhibitor Rivaroxaban Apixaban Edoxaban Prevention of stroke Prevention of heart attacks People with atrial fibrillation People with mechanical heart valves (warfarin) Competitive, direct, antithrombin-independent inhibition of Factor Xa Inhibits cleavage of prothrombin to thrombin to terminate thrombin burst and therefore decrease coagulation Wide therapeutic window, predictable pharmacokinetics No monitoring required Limited drug interactions and dietary restrictions and Antidote available (Andexanet, FXa decoy) No standardized test should monitoring be needed Expensive Increased risk of bleeding and secondary complications Not for use in children, pregnant women, those with renal or hepatic impairment Heparin Unfractionated heparin, LMWH In cardiac surgery (heparin) Enhanced Anti-thrombin inhibition of thrombin and factor X Safe for pregnancy Antidote available (Protamine, cationic peptide) Monitoring required Increased risk of bleeding and platelet deficiency Antithrombin-dependent
28
References CPS [Internet]. Ottawa (ON): Canadian Pharmacists Association; c2016 [updated 2018 SEP 04; cited 2019 SEP 16]. Coumadin [product monograph]. Available from: or CPS [internet]. Ottawa (ON): Canadian Pharmacists Association; c2016 [updated 2016 DEC; cited 2019 SEP 16]. Heparin: unfractionated. Available from: or CPS [internet]. Ottawa (ON): Canadian Pharmacists Association; c2016 [updated 2015 JUN; cited 2019 SEP 16]. Heparin: low molecular weight. Available from: or CPS [Internet]. Ottawa (ON): Canadian Pharmacists Association; c2016 [updated 2018 SEP 18; cited 2019 SEP 16]. Xarelto [product monograph]. Available from: or Katzung, B.G. (2018). Basic and Clinical Pharmacology, 14th Edition. McGraw Hill Education. Perzborn Elisabeth, Roehrig Susanne, Straub Alexander, Kubitza Dagmar, Mueck Wolfgang, and Laux Volker. “Rivaroxaban: A New Oral Factor Xa Inhibitor” Arteriosclerosis, Thrombosis, and Vascular Biology (2010): 376–381. Web. 16 Sep Silverthorn, D.U. (2012). Human Physiology: An Integrated Approach, 8th Edition. Pearson Education.
Similar presentations
© 2025 SlidePlayer.com. Inc.
All rights reserved.