Sign Up
You are not currently logged in. Please log in to CEUfast to enable the course progress and auto resume features.

Course Library

Anticoagulant and Fibrinolytic Therapy

2.5 Contact Hours including 2.5 Advanced Pharmacology Hours
Listen to Audio
CEUfast OwlGet one year unlimited nursing CEUs $39Sign up now
This peer reviewed course is applicable for the following professions:
Advanced Practice Registered Nurse (APRN), Certified Nurse Midwife, Certified Nurse Practitioner, Certified Registered Nurse Practitioner, Clinical Nurse Specialist (CNS), Licensed Practical Nurse (LPN), Licensed Vocational Nurses (LVN), Midwife (MW), Nursing Student, Registered Nurse (RN), Registered Nurse Practitioner
This course will be updated or discontinued on or before Sunday, July 26, 2026

Nationally Accredited

CEUFast, Inc. is accredited as a provider of nursing continuing professional development by the American Nurses Credentialing Center's Commission on Accreditation. ANCC Provider number #P0274.


Outcomes

≥ 92% of participants will know the appropriate use and limitations of various anticoagulant, antiplatelet, and fibrinolytic medications based on mechanisms of action and clinical scenarios.

Objectives

After completing this continuing education course, the participant will be able to:

  1. Analyze the risk factors for bleeding complications in patients receiving anticoagulant or fibrinolytic therapy.
  2. Evaluate the potential benefits and risks of anticoagulant and fibrinolytic therapy for patients with specific conditions.
  3. Develop a comprehensive patient education plan on using anticoagulant or fibrinolytic therapy medications safely and effectively.
  4. Identify four different classes of anticoagulant medications and their mechanisms of action.
  5. Differentiate between the indications and contraindications for various anticoagulant and fibrinolytic therapies.
  6. Implement strategies to minimize bleeding risks in patients receiving anticoagulant therapy.
  7. Compare and contrast the mechanisms of action of aspirin, heparin, direct oral anticoagulants (DOACs), and warfarin.
  8. Interpret laboratory results of common coagulation tests to monitor the effectiveness and safety of anticoagulants and fibrinolytic therapy.
CEUFast Inc. and the course planners for this educational activity do not have any relevant financial relationship(s) to disclose with ineligible companies whose primary business is producing, marketing, selling, re-selling, or distributing healthcare products used by or on patients.

Last Updated:
  • $39 Unlimited Access for 1 Year
    (Includes all state required Nursing CEs)
  • No Tests Required
    (Accepted by most states & professions)
  • Instant Reporting to CE Broker
  • Instant Access to certificates of completion
Logo Audio
Now includes
Audio Courses!
Learn More
Restart
Restart
  • 0% complete
Hide Outline
Playback Speed

Narrator Preference

(Automatically scroll to related sections.)
Done
Anticoagulant and Fibrinolytic Therapy
0:00
0:15
 
To earn a certificate of completion you have one of two options:
  1. Take test and pass with a score of at least 80%
  2. Attest that you have read and learned all the course materials.
    (NOTE: Some approval agencies and organizations require you to take a test and "No Test" is NOT an option.)
Author:    Breann Kakacek (BSN, RN)

Introduction

Nurses provide care to patients who receive anticoagulants and fibrinolytic therapy. Thrombosis is a leading cause of death in the United States, responsible for over 100,000 deaths annually (Centers for Disease Control and Prevention [CDC], 2024). Treating patients with anticoagulant and fibrinolytic drugs can be complicated. Multiple medications are available; each drug affects a different part of the clotting process, and serious side effects are possible. This course will simplify administering anticoagulant and fibrinolytic therapies by discussing the mechanism of action, the onset of effects, duration of effects, uses, dosing, adverse effects, and special consideration for each anticoagulant/fibrinolytic. Vital nursing care considerations, including patient education strategies, will be addressed to ensure optimal patient outcomes.

Anatomy and Physiology of Clotting Processes

The clotting process, coagulation or hemostasis, is a cascade of events that prevents blood loss after injury. It involves vascular, cellular, and enzymatic components contributing to clot formation (LaPelusa & Dave, 2023).

  1. Vascular Spasm
    • The initial response to vessel injury is a vascular spasm, a reflex contraction of the smooth muscle in the blood vessel wall. This constriction reduces blood flow to the area, limiting blood loss for several minutes to hours, allowing time for the subsequent phases of hemostasis. At the injury site, the extracellular matrix (ECM)/collagen from the endothelial lining becomes exposed to the different components in the blood.
  2. Platelet Plug Formation (Primary Hemostasis)
    • Upon encountering a breach in the vessel wall, platelets adhere to exposed subendothelial collagen and other ECM proteins through surface receptors, such as glycoprotein (GP) Ia/IIa and GPVI. The ECM then releases inflammatory markers and cytokines that start platelet aggregation. This adhesion is stabilized by von Willebrand factor (vWF), a multimeric protein that bridges platelets to collagen.
    • Following adhesion, platelets become activated, changing shape from smooth disks to forms with extended pseudopods, which allow them to interact more effectively with other platelets. Activation also triggers the release of granules containing adenosine diphosphate (ADP), serotonin, and thromboxane A2 (TXA2). These substances amplify the platelet response by inducing further platelet activation and recruitment to the injury site, forming a soft platelet plug.
  3. Coagulation (Secondary Hemostasis)
    • The coagulation cascade is a series of enzymatic reactions that culminate in the conversion of fibrinogen, a soluble plasma protein, into fibrin, an insoluble protein that cross-links to form a stable meshwork, anchoring the platelet plug and providing structural integrity to the clot.

This cascade is traditionally divided into three pathways:

Intrinsic Pathway(Barmore et al., 2023): This is initiated by trauma inside the vascular system and is activated by platelets. It involves factors XII, XI, IX, and VIII. Factor XI is activated by thrombin. Factor XIa and factor VIIa activate IXa. IXa combines with factor VIII and activates itself, and factor X. Factor Xa binds to factor Va and then converts prothrombin to thrombin (LaPelusa & Dave, 2023).

Extrinsic Pathway(Barmore et al., 2023): This pathway is triggered by external trauma that causes blood to escape from the vascular system. It involves the tissue factor (TF or factor III) and factor VII. Factor VII undergoes activation events that activate factor X and factor IX. Factor IXa binds to VIII. Factor Xa binds to factor V and calcium, then cleaves prothrombin into thrombin (LaPelusa & Dave, 2023).

Common Pathway(Barmore et al., 2023): Where the intrinsic and extrinsic pathways converge, involving factors X, V, II (prothrombin), and I (fibrinogen), leading to the formation of a thrombin-activated fibrin meshwork.

The coagulation cascade is activated by converting a series of proenzymes (zymogens) to active enzymes, ending in the activation of factor X to form the prothrombinase complex. This complex converts prothrombin (factor II) to thrombin (factor IIa), cleaving fibrinogen into fibrin monomers. Fibrin monomers spontaneously polymerize and are stabilized by factor XIIIa, forming cross-linked fibrin, the stable structure of the clot (Barmore et al., 2023).

Regulation of Coagulation

The clotting process is tightly regulated to prevent excessive clot formation and localize the clot to the injury site. Anticoagulant mechanisms include the action of antithrombin III, which inactivates several clotting factors, including thrombin; protein C and protein S, which inactivate factors Va and VIIIa; and the tissue factor pathway inhibitor (TFPI), which inhibits the extrinsic pathway.

Furthermore, the fibrinolytic system eventually dissolves the clot through the action of plasmin, an enzyme that degrades fibrin, ensuring that vessels return to a patent state after the repair of the injured tissue.

Coagulation Tests

Coagulation tests are diagnostic tools for assessing blood hemostatic function. They can detect abnormalities in the clotting process, guide anticoagulation therapy, and diagnose bleeding disorders. Here is a list of some of the commonly used coagulation tests.

Table 1: Commonly Used Coagulation Tests
Activated clotting time (ACT) (Kostousov, 2019)ACT measures the time it takes for blood to clot in the presence of an activator. It's primarily used to monitor high-dose heparin therapy during procedures that require rapid anticoagulation, such as cardiac bypass surgery or percutaneous coronary interventions. ACT is useful because it provides an immediate, point-of-care assessment of anticoagulation levels, allowing for real-time adjustments in heparin dosing.
Normal Levels: 70-120 seconds, therapeutic anticoagulation 150-600 seconds
Activated partial thromboplastin time (aPTT) (MedlinePlus, 2022b)aPTT assesses the intrinsic and common pathways of the coagulation cascade. It monitors patients on unfractionated heparin therapy and investigates suspected bleeding disorders like hemophilia. The test measures the time it takes for a clot to form after partial thromboplastin and an activator are added to the patient's plasma. Prolonged aPTT can indicate deficiencies in clotting factors, inhibitors like lupus anticoagulant, or the effect of heparin therapy.
Normal Levels: 25-35 seconds
Anti-factor Xa activity  (Szigeti, 2022a)This assay measures the inhibition of factor Xa, an essential enzyme in the coagulation cascade, by anticoagulants such as low molecular weight heparin (LMWH), unfractionated heparin (UH), and direct oral anticoagulants that target factor Xa (e.g., rivaroxaban, apixaban). The test monitors the efficacy and safety of these anticoagulants, ensuring that patients receive a therapeutic dose that minimizes the risk of bleeding or thrombotic complications.
Therapeutic ranges:
  • LMWH: 0.5-1.2 IU/mL
  • UH: 0.3-0.7 IU/mL
Prophylactic ranges:
  • LMWH: 0.2-0.5 IU/mL
  • UH: 0.1-0.4 IU/mL
International normalized ratio (INR) (MedlinePlus, 2022a)INR is a standardized way of reporting the prothrombin time (PT). It allows for comparing PT results across different laboratories by normalizing for variations in test performance. INR primarily monitors patients taking vitamin K antagonists (e.g., warfarin) for anticoagulation therapy.
Normal Levels: The target INR range depends on the indication for anticoagulation, with most conditions requiring a target INR between 2.0 and 3.0.
Prothrombin time (PT) (Devkota, 2020)PT measures the time it takes for blood to clot by assessing the extrinsic and common pathways of the coagulation cascade. It's susceptible to clotting factors VII, X, V, II (prothrombin), and fibrinogen deficiencies. PT monitors oral anticoagulant therapy (warfarin), assesses liver function (since clotting factors are produced in the liver), and evaluates the risk of bleeding in patients.
Normal Levels: 11 to 13.5 seconds
Thrombin time (TT) (Teruya, 2022)TT measures the time it takes for fibrinogen in the blood to convert to fibrin, forming a clot in the presence of thrombin. It's primarily used to assess fibrinogen levels and function and detect the presence of inhibitors that interfere with thrombin (e.g., unfractionated heparin or direct thrombin inhibitors like dabigatran). TT is sensitive to abnormalities in fibrinogen (dysfibrinogenemia or hypofibrinogenemia) and can be prolonged in cases of heparin contamination or the presence of fibrin degradation products (FDPs).
Normal Levels: Less than 20 seconds
D-dimer
(Szigeti, 2022b)
Detects the presence of fibrin degradation products in the blood, which are produced when a clot is broken down. Elevated D-dimer levels can indicate active thrombosis and are used to rule out conditions such as deep vein thrombosis (DVT) and pulmonary embolism (PE), especially in low-risk patients.
Normal Levels: < 250 ng/mL
Fibrinogen antigen test
(Cleveland Clinic, 2022)
Measures the fibrinogen concentration in the blood, which is critical for clot formation. It's used to diagnose suspected bleeding disorders or thrombotic conditions.
Platelet Function
(MedlinePlus, 2021)
Assess how well platelets are functioning in the clotting process. These tests, including platelet aggregation tests, diagnose platelet disorders or monitor antiplatelet therapy (e.g., aspirin, clopidogrel).

Considerations for Use in Patients

Anticoagulants

Anticoagulants and fibrinolytics have different considerations for use in patients.

Anticoagulants prevent blood clots from forming and are prescribed to patients with a condition where it is likely to form a clot. These conditions include atrial fibrillation, acute coronary syndrome, heart valve disease, and ischemic strokes (National Health Services, 2021c).

Indications(National Health Services, 2021b):

  • Venous thromboembolism prophylaxis and treatment
  • Atrial fibrillation
  • Acute coronary syndrome
  • Valvular heart disease

Contraindications and risks(Umerah & Momodu, 2023):

  • Active bleeding: Anticoagulants are contraindicated in patients with active bleeding or a high risk of bleeding, such as those with recent major surgery, peptic ulcers, or severe uncontrolled hypertension.
  • Thrombocytopenia: A low platelet count increases the risk of anticoagulant bleeding complications.
  • Liver disease: Severe liver disease can impair the metabolism and clearance of certain anticoagulants, necessitating dose adjustments or avoiding them altogether. Liver disease can also impair platelet production, leading to a lower platelet count.
  • Pregnancy: Most anticoagulants are contraindicated during pregnancy due to the risk of fetal bleeding complications.

Types of Anticoagulants(Umerah & Momodu, 2023)

  • Vitamin K antagonists: Warfarin is the most common one, requiring regular monitoring of INR for dose adjustments.
  • (DOACs): Newer alternatives like rivaroxaban, apixaban, and dabigatran offer several advantages over warfarin, including predictable dosing and minimal monitoring requirements.
  • LMWHs: Enoxaparin and dalteparin are commonly used for venous thromboembolism (VTE) prophylaxis and treatment, requiring subcutaneous administration.
  • Unfractionated Heparin (UH): This is primarily used in hospitalized patients for rapid anticoagulation, requiring continuous intravenous infusion and frequent monitoring.

Fibrinolytic Therapy

Fibrinolytics, also known as thrombolytics, can dissolve blood clots. While potentially lifesaving in specific situations, their use requires careful evaluation due to the inherent risk of bleeding complications (Baig & Bodle, 2023).

Indications(Baig & Bodle, 2023):

Acute ischemic stroke: Fibrinolytics are considered within a narrow therapeutic window (typically 3-4.5 hours) to dissolve the clot and restore blood flow to the brain, potentially minimizing neurological damage and improving patient outcomes.

Acute myocardial infarction: In specific scenarios with contraindications to other interventions, fibrinolytics may reopen occluded coronary arteries to limit the infarct size and improve the prognosis.

PE: In high-risk PE with hemodynamic instability, fibrinolytics can dissolve the clot and improve oxygenation. However, their use is not routine in other PE presentations due to the increased risk of bleeding compared to the potential benefit.

DVT:  Fibrinolytics are sometimes used in DVT to restore blood flow through blocked veins and improve circulation. This can potentially reduce the risk of long-term complications like post-thrombotic syndrome (PTS), which can cause pain, swelling, and skin changes in the leg. However, fibrinolytics are not for everyone with DVT, and there is a risk of bleeding.

Occlusion of indwelling catheters: Fibrinolytics can be used to try to clear blood clots that block catheters, such as those used for dialysis or delivering medications. This can help avoid the need to replace the catheter, which can be uncomfortable for the patient and carries some risks of infection.

Acute peripheral arterial occlusion (PAO): In acute PAO, a blood clot suddenly blocks an artery, reducing blood flow to the leg or arm. Fibrinolytics may be used to dissolve the clot and restore blood flow rapidly. This can help prevent tissue damage and potentially improve limb salvage and long-term outcomes. However, fibrinolytics are not always appropriate for everyone with PAO, and there is a risk of bleeding.

Contraindications and risks(National Health Services, 2021a):

Increased bleeding risk: The primary concern with fibrinolytics is their propensity to cause bleeding due to their systemic effect on dissolving clots. This risk is further heightened by:

  • Recent major surgery or trauma
  • Active internal bleeding
  • History of intracranial hemorrhage
  • Severe uncontrolled hypertension
  • Pregnancy
  • Known bleeding disorders

Ineligibility for timely treatment: Fibrinolytics are only effective from the onset of symptoms within a specific time window. Delays significantly decrease their efficacy and increase bleeding risks. For use in acute ischemic stroke, a tissue plasminogen activator (tPA) should be given within 3-4.5 hours of symptoms starting before the risk of intracranial bleeding increases.

Increased risk of intracranial hemorrhage: This is a severe and potentially fatal complication associated with fibrinolytic use, especially in stroke patients.

Meticulous evaluation is crucial to identifying patients who meet the strict criteria for fibrinolytic use and minimizing the risk of complications. Adherence to the narrow therapeutic window is critical for maximizing benefits and minimizing risks. Different fibrinolytic agents have specific administration protocols that require strict adherence to ensure safety and effectiveness.

Clotting Complications in Patients

Depending on the situation and risk-benefit analysis, several clotting complications may need treatment with either anticoagulants or fibrinolytics. Here's a breakdown of some common conditions:

Venous Thromboembolism

DVT and PE are types of VTE. Anticoagulants are used to treat VTE. Due to their rapid onset of action, UH and LMWHs are commonly used initially (National Heart Lung and Blood Institute [NHLBI], 2022b).

DOACs may be used as a transition from initial heparin therapy, offering long-term management with convenient oral administration.

Anticoagulants prevent further clot formation and growth, minimizing the risk of PE and preventing DVT extension. This aids natural clot resolution and reduces the risk of long-term complications like PTS (NHLBI, 2022b).

Acute Ischemic Stroke

Ischemic strokes are another clotting complication in patients and lead to severe complications without treatment. Treatment involves fibrinolytics administered within a narrow therapeutic window (3-4.5 hours) to dissolve the clot and restore blood flow, potentially limiting brain damage and improving patient outcomes. The specific agent used depends on hospital protocols and patient characteristics (NHLBI, 2023).

Fibrinolytics break down the clot, resuming blood flow and potentially preventing further neurological damage. However, the risk of bleeding complications, specifically intracranial hemorrhage, needs to be carefully considered (NHLBI, 2023).

Acute Myocardial Infarction

Acute myocardial infarctions have a window of time to be treated to prevent cell death within the heart. Fibrinolytics and anticoagulants can be used to treat myocardial infarctions (Sweis & Jivan, 2024).

Fibrinolytics may dissolve the clot and restore blood flow to the heart muscle in specific scenarios when timely access to percutaneous coronary intervention (PCI) is unavailable.

Anticoagulants such as heparin may also be used with other therapies to prevent further clot formation and improve outcomes.

Like stroke, fibrinolytics aim to dissolve the clot and salvage heart tissue by restoring blood flow. However, the risks of bleeding complications need careful consideration (Sweis & Jivan, 2024).

Disseminated Intravascular Coagulation

Disseminated intravascular coagulation (DIC) is an uncontrolled activation of the clotting cascade, leading to the widespread formation of small clots throughout the bloodstream. Paradoxically, it also causes bleeding due to the depletion of clotting factors and platelets (NHLBI, 2022a).

Treatment includes anticoagulants in specific situations where the benefits outweigh the risks; low-dose heparin may be used to prevent further clot formation and organ damage. However, this is a complex decision requiring careful evaluation by a specialist.

In some cases, controlled inhibition of further clot formation with low-dose heparin may be beneficial, alongside addressing the underlying cause of DIC. However, the risk of excessive bleeding requires extreme caution (NHLBI, 2022a).

Other Situations

Arterial thrombosis involves peripheral arterial occlusions (e.g., stroke due to carotid artery thrombosis), and anticoagulants like heparin may be used cautiously to prevent further clot formation (Cleveland Clinic, 2023).

Central venous catheters may clot, leading to a catheter-related thrombosis, and prophylactic anticoagulation with LMWHs may be used to prevent clot formation on the catheter surface.

Anticoagulants vs. Fibrinolytic Therapy

While anticoagulants and fibrinolytics are crucial in managing blood clots, they differ significantly in their cellular mechanisms, processing, mode of action, and associated risks. Understanding these differences is vital for their clinical application in managing thrombotic disorders.

Mechanism of Action

Fibrinolytic Therapy

Fibrinolytics act directly on fibrin clots to promote their dissolution. They convert plasminogen to plasmin, which enzymatically breaks down fibrin, the structural framework of blood clots (Baig & Bodle, 2023).

Target the final step of the clotting cascade, effectively dissolving clots after forming. Specifically, it targets and breaks down fibrin within clots, acting on the established thrombi.

Fibrinolytics can be given either through an intravenous (IV) pathway, such as a peripheral IV, or through a local release through a catheter by interventional radiology or cardiology (Baig & Bodle, 2023).

  1. Plasminogen Activation (Risman et al., 2023)
    • Fibrinolysis begins with activating plasminogen, a proenzyme, into its active form, plasmin. This activation can occur through various pathways:
      • tPA: Released from endothelial cells lining blood vessels, tPA is the primary physiological activator of plasminogen.
      • Urokinase-type plasminogen activator (uPA): Expressed by various cell types, uPA plays a role in physiological fibrinolysis and wound healing.
      • Other activators: Several factors, including factor XIIa and kallikrein, can activate plasminogen under specific conditions.
  2. Plasmin-Mediated Fibrin Degradation
    • Once activated, plasmin, a serine protease, is the major enzyme responsible for fibrinolysis. It cleaves fibrin, the main protein component of blood clots, at specific sites, leading to fragmentation and dissolution of the clot structure.
  3. Regulation of Fibrinolysis
    • To prevent excessive and uncontrolled breakdown of healthy blood clots, a sophisticated regulatory system exists:
      • Plasminogen activator inhibitors (PAIs): These proteins, primarily PAI-1, inactivate tPA and uPA, limiting plasminogen activation.
      • Antiplasmins: These proteins, including alpha-2-antiplasmin, directly inhibit plasmin, preventing its detrimental effects on healthy tissues.
  4. Fibrinolytic Drugs and Their Modes of Action
    • Clinically used fibrinolytic therapies act by different mechanisms to enhance fibrinolysis:
      • Recombinant tPA (rtPA): This drug mimics the action of endogenous tPA, directly activating plasminogen.
      • Streptokinase: This bacterial protein activates plasminogen by forming a complex, which leads to a conformational change that promotes its activity.
      • Other fibrinolytic therapies: Newer agents like alteplase and tenecteplase are modified forms of rtPA with improved properties.

Anticoagulants

Anticoagulants are sometimes called “blood thinners,” but this term is inaccurate as they disrupt the clotting process rather than thinning the blood. They are also different from antiplatelets such as clopidogrel and low-dose aspirin (Heestermans et al., 2022).

Anticoagulants do not directly dissolve clots but prevent clot formation or extension by inhibiting various factors within the coagulation cascade.

Mechanisms vary by class: Vitamin K antagonists (e.g., warfarin) inhibit the synthesis of vitamin K-dependent clotting factors, whereas DOACs inhibit specific factors such as factor Xa (e.g., rivaroxaban, apixaban) or thrombin (e.g., dabigatran).

These drugs prevent the formation or growth of thrombi by inhibiting various coagulation cascade components without directly dissolving existing clots (Heestermans et al., 2022).

Pharmacokinetics

Fibrinolytic Therapy

Fibrinolytics are administered via IV to ensure the rapid onset of action, which is crucial for their effectiveness in acute settings.

Typically, they have a short half-life, necessitating their administration soon after thrombotic events like stroke or myocardial infarction.

Anticoagulants

Anticoagulants can be administered orally (e.g., warfarin, DOACs) or parenterally (e.g., Heparin, LMWH) (Heestermans et al., 2022).

The pharmacokinetics, including absorption, distribution, metabolism, and excretion, vary significantly across different agents, influencing factors such as dosing schedules and monitoring (Heestermans et al., 2022).

Specific Complications, Side Effects, and Risks

Fibrinolytic Therapy

The significant risk of bleeding, including intracranial hemorrhage, is due to the systemic activation of plasminogen and subsequent degradation of fibrin not only in the thrombus but also in hemostatic plugs throughout the body (Baig & Bodle, 2023).

Anticoagulants have a significantly higher risk due to their direct action on lysing existing clots. Other side effects may include allergic reactions, hypotension, reperfusion arrhythmia, angioedema, anaphylactic shock, and fever (Baig & Bodle, 2023).

Anticoagulants

Bleeding is the primary risk, though generally less severe than with fibrinolytics, except in cases of overdose or when used concomitantly with other medications that increase bleeding risk (Umerah & Momodu, 2023).

Specific complications can vary with the agent used. For example, warfarin requires monitoring of INR to avoid over-anticoagulation, while DOACs have a lower risk of intracranial hemorrhage but require careful dose adjustment in renal impairment. However, in general, with anticoagulants (Umerah & Momodu, 2023):

  • Increased bleeding risk: The main concern is that they inhibit clot formation throughout the body. This risk is further heightened by:
    • Recent surgery or trauma
    • Active bleeding
    • Uncontrolled hypertension
    • Concurrent use of other medications that affect bleeding

Heparin-Induced Thrombocytopenia

Heparin-induced thrombocytopenia (HIT) is a significant risk with heparin and LMWH, leading to a paradoxical increase in thrombotic events. It can occur in patients receiving heparin for various indications, including VTE prophylaxis and treatment, cardiac procedures, and dialysis (Patriarcheas et al., 2020).

Onset typically occurs 5-14 days after initial heparin exposure, although it can occur earlier with subsequent exposures.

Symptoms include (Patriarcheas et al., 2020):

  • Thrombocytopenia: A decrease in platelet count, the blood cells responsible for clot formation. This can be detected through routine blood tests.
  • New or worsening thrombosis: HIT can paradoxically lead to new blood clots despite being an anticoagulant. This can manifest as:
    • DVT symptoms like leg pain, swelling, and redness.
    • PE symptoms like shortness of breath, chest pain, and coughing up blood.
    • Other potential manifestations depending on the location of the new clot.

A rapid drop in platelet count is a critical sign and requires immediate action. New symptoms of additional clot formations may suggest the development of HIT.

Causes and Risks(Patriarcheas et al., 2020):

  • Heparin exposure: All forms of heparin, including UH and LMWHs, can trigger HIT.
  • Immune response: Heparin binds to platelets, causing the immune system to mistakenly attack these platelets, leading to their destruction and subsequent thrombocytopenia.
  • Previous heparin exposure.
  • Surgery or major trauma.
  • Sepsis.
  • Cancer.
  • Advanced age.

Complications(Patriarcheas et al., 2020):

  • Thrombosis: Paradoxically, HIT can lead to new or worsening blood clots due to the combined effects of platelet activation and inhibition of normal clotting mechanisms.
  • Necrosis: In severe cases, HIT can cause skin and tissue death (necrosis) at the injection site.
  • PE: A potentially life-threatening complication of HIT due to new clot formation in the lungs.

Prevention(Patriarcheas et al., 2020):

  • Risk stratification: Identifying patients at high risk for HIT can help guide the choice of alternative anticoagulants.
  • Platelet count monitoring: Regular platelet count monitoring during heparin therapy is crucial for early detection of HIT.
  • Alternative anticoagulants: In high-risk patients, alternative anticoagulants not associated with HIT may be preferred.

Types of Anticoagulants and Antiplatelets

Both anticoagulants and antiplatelet agents prevent blood clots, but they target different stages of the clotting cascade and have distinct mechanisms of action.

Anticoagulants work by inhibiting various enzymes or factors involved in the later stages of the blood clotting cascade. They prevent the formation of fibrin, a mesh-like protein that forms the core of a blood clot.

Antiplatelets primarily target platelets, cell fragments in the blood that initiate clot formation by clumping together at the injury site. They aim to prevent platelet activation and aggregation, thereby hindering the initial stages of clot development.

Dabigatran (Pradaxa®, APO-Dabigatran®)

Dabigatran (Pradaxa®, APO-Dabigatran®) is classified as a direct thrombin inhibitor. It acts later in the clotting cascade process, binding to thrombin and preventing it from converting fibrinogen into fibrin (Medscape, n.d. -b).

Dabigatran can be used to prevent and reduce the risk of stroke in atrial fibrillation, prevent blood clots after hip or knee replacement surgeries, and treat existing blood clots such as DVTs and PEs. 

The most common side effect of dabigatran is bleeding because it directly inhibits clot formation. Other potential side effects include indigestion, nausea, vomiting, and back pain.

Dabigatran has no readily available antidote for excessive bleeding events. Careful monitoring is crucial.

Kidney function significantly impacts how the body eliminates dabigatran. Dosage adjustments may be needed for patients with impaired kidney function (Medscape, n.d. -b).

Edoxaban (Savaysa®)

Edoxaban (Savaysa®) is classified as a factor Xa inhibitor. This means it targets factor Xa in the blood clotting cascade, making it an anticoagulant. It selectively inhibits Factor Xa, which converts prothrombin into thrombin (Medscape, n.d. -d).

The most common side effect of edoxaban is bleeding. As it directly hinders clot formation, an increased risk of bleeding events (both major and minor) is a significant consideration.

Other potential side effects include indigestion, nausea, vomiting, dizziness, back pain, and low blood pressure.

There is no specific antidote for edoxaban-related bleeding. Management strategies rely on supportive care and the potential use of blood products in severe cases.

Edoxaban elimination is significantly impacted by kidney function. Dosage adjustments or alternative medications may be necessary for patients with impaired kidney function.

Edoxaban can interact with other medications that affect clotting, potentially increasing bleeding risk. Careful medication review and potential dose adjustments are essential (Medscape, n.d. -d).

Rivaroxaban (Xarelto®)

Rivaroxaban (Xarelto®) is a factor Xa inhibitor, making it an anticoagulant. Rivaroxaban selectively inhibits factor Xa, preventing prothrombin from converting to thrombin (MedlinePlus, 2023).

The most common side effect of Rivaroxaban (Xarelto®) is bleeding. As it directly hinders clot formation, an increased risk of bleeding events is a significant consideration.

Other potential side effects include indigestion, nausea, vomiting, dizziness, back pain, and low blood pressure.

There is no specific antidote for rivaroxaban-related bleeding. Management strategies rely on supportive care and the potential use of blood products in severe cases (MedlinePlus, 2023).

Enoxaparin (Lovenox®)

Enoxaparin (Lovenox®) falls under the LMWH category, making it an anticoagulant (Medscape, n.d. -e).

Unlike rivaroxaban and other factor Xa inhibitors, enoxaparin works through an indirect mechanism. It primarily enhances the activity of antithrombin III, a natural anticoagulant protein in the blood. This complex formation (enoxaparin-antithrombin III) then inactivates factor Xa and thrombin, hindering clot formation at multiple stages.

The most common side effect of enoxaparin is bleeding. As it indirectly inhibits clotting factors, there's an increased risk of bleeding events (both major and minor). Other potential side effects include injection site pain or redness, bruising, and thrombocytopenia in rare cases.

Enoxaparin can prevent blood clots such as DVTs or PEs from forming in various situations, such as after major orthopedic surgery, during medical illness causing prolonged immobilization, and in patients with unstable angina or NSTEMI. It can also be used to treat existing blood clots.

Enoxaparin carries a bleeding risk, though potentially lower than some other anticoagulants. Careful patient selection and monitoring are crucial, especially for those with a history of bleeding or high bleeding risk.

There is a potential for the development of HIT with the administration of enoxaparin. Monitoring for signs and symptoms of HIT is essential.

Enoxaparin is administered subcutaneously. Protamine sulfate can partially reverse the anticoagulant effect of enoxaparin in cases of serious bleeding (Medscape, n.d. -e).

Heparin

Heparin, often referred to as UH, is a naturally occurring glycosaminoglycan and the original injectable anticoagulant medication (Medscape, n.d. -g).

Heparin works through an indirect mechanism that primarily enhances the activity of antithrombin III, a natural anticoagulant protein in the blood. The complex formed (heparin-antithrombin III) then inactivates various clotting factors, including factor Xa and thrombin, hindering clot formation at multiple stages.

The most common side effect of heparin is bleeding. As it indirectly inhibits clotting factors, there's an increased risk of bleeding events (both major and minor).

Other potential side effects include injection site pain or redness, bruising, thrombocytopenia, HIT, which is a concern, and hair loss (less common).

Heparin can be used to prevent DVT and PE in various scenarios, including after major surgery, during medical illness causing prolonged immobilization, in patients with unstable angina or non-ST-elevation myocardial infarction. Heparin can be used to treat existing blood clots. Heparin solutions are used at low concentrations to prevent clotting within IV catheters.

Heparin is often monitored with a coagulation test called an aPTT. Heparin carries the risk of bleeding and development of HIT.

Heparin can be administered IV or subcutaneously (under the skin), depending on the situation. Protamine sulfate can partially reverse the anticoagulant effect of heparin in case of serious bleeding (Medscape, n.d. -g).

Warfarin (Coumadin®)

Warfarin (Coumadin®) is a medication classified as a vitamin K antagonist. It is an anticoagulant that directly targets clotting factors or enzymes, and warfarin works indirectly by inhibiting the production of Vitamin K-dependent clotting factors in the liver. Vitamin K is essential for the proper function of several clotting factors, including factor II (prothrombin) and factor X. By reducing Vitamin K availability, warfarin effectively diminishes the activity of these clotting factors, leading to a decreased ability to form clots (Patel et al., 2023).

The most significant side effect of warfarin is bleeding. Due to its effect on multiple clotting factors, the risk of bleeding can be substantial and requires careful monitoring. Other potential side effects include skin rash, hair loss (less common), and purple toes syndrome (uncommon).

Warfarin has traditionally been used for several purposes, including preventing blood clots. It can help prevent DVT and PE in people who have undergone certain surgeries or have risk factors for clot formation, such as atrial fibrillation and a high risk of stroke. Warfarin can also treat existing blood clots, such as DVTs and PEs.

The use of omeprazole with warfarin has an increased risk of anticoagulation side effects, including increased INR and bleeding risk.

Warfarin is highly sensitive to various foods (vitamin K-rich vegetables) and medications, significantly impacting its effectiveness. Careful dietary counseling and medication review are essential.

Warfarin takes several days to reach its full effect, and elimination can also be slow. This necessitates frequent monitoring and dose adjustments during initiation and discontinuation. Dosing is based on the condition the medication is treating.

It is monitored through a PT/INR blood test. Therapeutic INR levels for patients on Coumadin typically are between 2.0 and 3.0.

Unlike newer anticoagulants, there is no specific antidote to reverse warfarin's effects in case of serious bleeding. Vitamin K administration can help, but it takes time to counteract warfarin's action (Patel et al., 2023).

Aspirin

Aspirin (acetylsalicylic acid) is a widely used medication with various effects, including pain relief, fever reduction, and, at low doses, blood clot prevention. Aspirin works through a different mechanism for blood clot prevention. It irreversibly inhibits platelet COX-1, preventing the formation of TXA2, a potent activator of platelets that contributes to clot formation. However, this effect is relatively weak compared to dedicated antiplatelet medications (Arif & Aggarwal, 2023).

The most common side effects of aspirin include stomach upset, heartburn or indigestion, nausea, vomiting, and gastritis in some cases. At higher doses, aspirin can also cause tinnitus, dizziness, and increased bleeding risk.

Low-dose aspirin (typically 81 mg daily) can be used to help prevent blood clots in certain high-risk individuals, such as those with a history of heart attack or stroke.

Aspirin can irritate the stomach lining and increase the risk of bleeding, especially at higher doses or in combination with other medications that affect clotting.

Aspirin should not be given to children or teenagers with viral illnesses due to the risk of Reye's syndrome.

While aspirin can help prevent blood clots, it is less effective than dedicated antiplatelet medications like clopidogrel or ticagrelor (Arif & Aggarwal, 2023).

Argatroban

Argatroban is classified as a direct thrombin inhibitor. It reversibly binds to the active site of thrombin, preventing it from converting fibrinogen into fibrin (Mahat et al., 2023).

The most common side effect of argatroban is bleeding. This is because it directly inhibits clot formation. Other potential side effects include headache, nausea, vomiting, fever, and hypotension.

Argatroban is primarily used for managing HIT and performing PCI in HIT patients.

Due to its potent action, argatroban carries a significant risk of bleeding. Careful patient selection and close monitoring are crucial, especially for those with a history of bleeding or high bleeding risk. No readily available standardized test can directly monitor the argatroban effect. However, coagulation tests like aPTT can be used as a guide.

There is no specific antidote to reverse the anticoagulant effect of argatroban in case of serious bleeding. Supportive care and potential blood product administration become essential.

The liver primarily eliminates argatroban. Dosage adjustments may be necessary for patients with impaired liver function (Mahat et al., 2023).

Dipyridamole (Persantine®)

Dipyridamole is classified as an antiplatelet aggregation inhibitor (Kerndt & Nagalli, 2023).

Dipyridamole's exact mechanism is not fully understood, but it's believed to work through multiple pathways: Inhibition of phosphodiesterase enzymes, leading to increased levels of cyclic adenosine monophosphate (cAMP) within platelets; this can impair platelet activation and aggregation. Disruption of adenosine uptake by red blood cells allows more adenosine (an inhibitor of platelet aggregation) to be available and exert its antiplatelet effect. There are direct effects on the shape and function of platelets.

The most common side effects of dipyridamole include headache (especially during initial use), chest pain, dizziness, nausea, vomiting, diarrhea (less common), and flushing.

Dipyridamole, typically used in combination with other medications like aspirin (in Aggrenox®), has been used for secondary prevention of stroke and prevention of peripheral arterial disease (Kerndt & Nagalli, 2023).

Dalteparin (Fragmin®)

Dalteparin is classified as an LMWH. Dalteparin works indirectly by potentiating antithrombin III, causing a conformational change that enhances its ability to deactivate factor Xa and thrombin (Medscape, n.d. -c).

The most common side effect of dalteparin is bleeding. Other potential side effects include injection site pain or redness, bruising, and low blood cell count (thrombocytopenia) in rare cases. The Development of HIT is a risk with this medication.

Dalteparin is administered subcutaneously. Its uses include preventing blood clots, including DVTs and PEs, and treating existing blood clots. Protamine sulfate can partially reverse the anticoagulant effect of dalteparin in case of serious bleeding (Medscape, n.d. -c).

Clopidogrel (Plavix®)

Clopidogrel (Plavix®) is an antiplatelet drug. Clopidogrel works by irreversibly inhibiting the P2Y12 receptor on platelets. By blocking the P2Y12 receptor, clopidogrel prevents platelets from responding to certain chemical signals that would normally trigger them to aggregate (Beavers & Naqvi, 2023).

Clopidogrel is used for the prevention of heart attack and stroke and the placement of a stent.

Clopidogrel takes several days to take full effect and is considered to have a slow onset. There is no reversal or antidote for clopidogrel (Beavers & Naqvi, 2023).

Apixaban (Eliquis®)

Apixaban is a factor Xa inhibitor. Apixaban acts as a selective inhibitor, specifically targeting factor Xa. By inhibiting factor Xa, apixaban disrupts the clotting cascade. Apixaban works on free-floating factor Xa and factor Xa, which are already bound to a clot. This ensures broader inhibition of clot formation (Medscape, n.d. -a).

The most common side effects of apixaban are related to bleeding, such as bruising and heavy bleeding. It is typically used for atrial fibrillation, DVTs, and PEs. The antidote is called andexanet alfa (brand name Andexxa). It's the first and only Food and Drug Administration (FDA)-approved specific antidote for reversing the anticoagulant effects of apixaban. Andexanet is typically reserved for emergencies where someone is taking apixaban and experiences life-threatening or uncontrolled bleeding.

Andexanet binds to apixaban in the bloodstream, effectively neutralizing its anticoagulant effect. This can happen within minutes of administration. It's important to note that andexanet does not eliminate apixaban from the body. The effects of apixaban will gradually return as the body eliminates andexanet (Medscape, n.d. -a).

Prasugrel (Effient®)

Prasugrel is an antiplatelet medication that targets the P2Y12 receptor on platelets. It is a prodrug, meaning it needs to be metabolized to become active. Once activated, it binds irreversibly to the P2Y12 receptor. This prevents ADP from binding and activating the platelet, inhibiting platelet aggregation and reducing clot formation (Medscape, n.d. -h).

Unlike other P2Y12 receptor antagonists like clopidogrel, which have a reversible binding, prasugrel's irreversible binding provides a more consistent and potent inhibition of platelet aggregation throughout a platelet's lifespan.

The most common side effects are related to bleeding, like easy bruising, nosebleeds, or bleeding gums. More serious side effects include severe or uncontrolled bleeding in the stomach, intestines, or brain. Rarely, it can cause a blood disorder called thrombotic thrombocytopenic purpura. Symptoms include fever, confusion, and yellowing of the skin or eyes.

Prasugrel can be given to patients with acute coronary syndrome or during PCI to prevent blood clots from forming and reduce the risk of heart attack and stroke (Medscape, n.d. -h).

Ticagrelor (Brilinta®)

Ticagrelor (Brilinta) is an antiplatelet medication that targets the P2Y12 receptor on platelets. It acts as a reversible antagonist to the P2Y12 receptor. Ticagrelor binds to the P2Y12 receptor, preventing ADP from binding and activating the platelet. This binding is reversible, so ADP can displace ticagrelor and activate the platelet over time (Medscape, n.d. -i).

However, ticagrelor has a high affinity for the P2Y12 receptor, meaning it binds strongly and for a relatively long duration compared to its dissociation rate. This ensures consistent inhibition of platelet activation throughout most of a platelet's lifespan.

By reversibly blocking the P2Y12 receptor, ticagrelor reduces platelets' ability to aggregate and form clots. This decreases the risk of thrombotic events in arteries, which is beneficial for patients with acute coronary syndrome or those who have undergone procedures like PCI, where blood clot formation can be dangerous.

The most common side effect is an increased bleeding risk. This can manifest as easy bruising, nosebleeds, or bleeding gums. More serious bleeding in the stomach, intestines, or brain can also occur. Some people experience mild to moderate shortness of breath, which usually improves as the body adjusts to the medication. Other side effects may include headache, diarrhea, nausea, and dizziness.

There's currently no specific antidote to reverse the effects of ticagrelor in case of bleeding. This is because of its reversible binding to the P2Y12 receptor. While the drug eventually dissociates from the receptor, there's no way to accelerate this process rapidly (Medscape, n.d. -i).

Vorapaxar (Zontivity®)

Vorapaxar (Zontivity®) works differently from other antiplatelet medications (like prasugrel and ticagrelor) (Medscape, n.d. -j).

Vorapaxar targets a specific receptor on platelets and some vascular cells called protease-activated receptor-1 (PAR-1). Vorapaxar acts as a selective antagonist to PAR-1. By blocking PAR-1, Vorapaxar disrupts the signaling pathway that leads to platelet activation and aggregation, ultimately reducing blood clot formation.

The most concerning side effect is an increased risk of bleeding, including easy bruising, nosebleeds, or bleeding in the stomach, intestines, or brain. Vorapaxar is not suitable for everyone due to the bleeding risk. People with a history of stroke, transient ischemic attack, or active bleeding are not recommended to take it.

Vorapaxar is typically used in specific patient populations with a high risk of cardiovascular events, such as those with a history of heart attack or peripheral arterial disease when combined with aspirin and/or clopidogrel (antiplatelet medications).

There's currently no specific antidote to reverse the effects of vorapaxar in case of bleeding (Medscape, n.d. -j).

Eptifibatide (Integrilin®)

Eptifibatide (Integrilin®) falls under glycoprotein IIb/IIIa inhibitors, a cardiovascular and antiplatelet drug (Bansal et al., 2023).

Eptifibatide targets a specific protein complex on the surface of platelets called GP IIb/IIIa. It acts as a reversible antagonist to GP IIb/IIIa receptors. It binds to these receptors, preventing fibrinogen from binding and causing platelet aggregation. By blocking GPIIb/IIIa receptors, eptifibatide disrupts the final step of clot formation, thereby reducing the overall risk of blood clots.

Eptifibatide is primarily used in two main settings to prevent blood clots and reduce the risk of cardiovascular events in acute coronary syndrome, NSTEMI, and PCI.

The most common side effect of eptifibatide is bleeding. This can manifest as easy bruising, nosebleeds, or bleeding gums. More serious bleeding in the stomach, intestines, or brain can also occur. Other potential side effects include low blood pressure, fever, headache, nausea, and vomiting.

Eptifibatide is administered through IV measures in a hospital setting due to the potential for serious bleeding. Since there is no specific reversal agent for eptifibatide, careful monitoring is essential during treatment (Bansal et al., 2023).

Tirofiban (Aggrastat®)

Tirofiban (Aggrastat®) is a GP IIb/IIIa inhibitor. Tirofiban acts as a reversible antagonist to GP IIb/IIIa receptors. It binds to these receptors, preventing fibrinogen from binding and causing platelet aggregation. By blocking GP IIb/IIIa receptors, tirofiban disrupts the final step of clot formation, thereby reducing the overall risk of blood clots (Aggrastat, n.d.).

Tirofiban is primarily used in two main settings to prevent blood clots and reduce the risk of cardiovascular events such as acute coronary syndrome, NSTEMI, and PCI.

The most common side effect of tirofiban is bleeding. This can manifest as easy bruising, nosebleeds, or bleeding gums. More serious bleeding in the stomach, intestines, or brain can also occur. Other potential side effects include low blood pressure, fever, headache, nausea, and vomiting.

Tirofiban is administered via IV in a hospital setting due to the potential for serious bleeding. There's no specific reversal agent for tirofiban, so careful monitoring is essential during treatment (Aggrastat, n.d.).

Cangrelor (Kengreal®)

Cangrelor (Kengreal®) is an antiplatelet that belongs to the category of P2Y12 receptor inhibitors. It has a very rapid onset of action, working within minutes of administration to inhibit platelet aggregation. Cangrelor reversibly binds to the P2Y12 receptor. However, its dissociation rate (unbinding) is much faster than others. This rapid reversibility allows the effects of cangrelor to wear off quickly after the infusion is stopped (Awosika & Patel, 2023).

Its use is primarily limited to specific situations during PCI due to its short duration of action and potential for increased bleeding risk.

The most common side effect of cangrelor is bleeding. This can manifest as easy bruising, nosebleeds, or bleeding gums. More serious bleeding in the stomach, intestines, or brain can also occur (Awosika & Patel, 2023).

Betrixaban (Bevyxxa®)

Betrixaban (Bevyxxa®) belongs to the category of factor Xa inhibitors, a class of anticoagulant medications. Betrixaban acts as a selective inhibitor of factor Xa. Betrixaban binds directly to factor Xa, preventing it from converting prothrombin to thrombin.

Betrixaban is approved specifically for VTE prophylaxis. Its most common side effect is an increased bleeding risk. Due to limitations in monitoring and potential bleeding risks, betrixaban is typically not a first-line choice for long-term blood clot prevention. There's currently no specific antidote to reverse the effects of betrixaban in case of bleeding.

Types of Fibrinolytic Therapy

Several types of fibrinolytic therapy exist, and they can sometimes be referred to as plasminogen activators. All currently available drugs used to dissolve blood clots (thrombolytic agents) belong to a category of enzymes called serine proteases. These enzymes break down plasminogen, an inactive protein in the blood, into its active form, plasmin. Plasmin, in turn, plays a crucial role in dissolving fibrin, the main protein component of blood clots (Baig & Bodle, 2023).

Alteplase (Activase®)

Alteplase, an rtPA, is a near-identical copy of the body's natural tPA. This close resemblance translates to high fibrin specificity, primarily targeting fibrin within clots. Unlike some fibrinolytics, alteplase boasts a rapid breakdown in the bloodstream with a short plasma half-life of 4-6 minutes (Reed et al., 2023).

Important Cautions:

Despite fibrin specificity, alteplase can cause systemic fibrinolysis, leading to elevated levels of circulating fibrin degradation products and a moderate increase in bleeding risk. Alteplase has specific indications and contraindications. Time is critical for effectiveness. Alteplase has specific time windows for administration in different conditions to maximize benefits and minimize risks.

To treat ischemic strokes, tPA is most effective when given within 3-4.5 hours of the onset of symptoms (NHLBI, 2023). Patients experiencing an ischemic stroke may still benefit from tPA treatment after the 4.5-hour window. However, more research needs to be done.

Contraindications(Genentech, n.d.)

  • Active intracranial hemorrhage
  • Subarachnoid hemorrhage
  • Active internal bleeding
  • Recent (within three months) intracranial or intraspinal surgery
  • Recent serious head trauma
  • Intracranial conditions that may increase the risk of bleeding 
  • Bleeding diathesis
  • Current severe uncontrolled hypertension

Adverse Side Effects:

While less common than other fibrinolytics, alteplase can cause bleeding, especially in areas with recent injury or surgery. Other potential side effects include low blood pressure, nausea, vomiting, and allergic reactions.

Reteplase (Retavase®)

Reteplase, a second-generation rtPA, offers potential advantages over alteplase (first-generation). Reteplase exhibits weaker binding to fibrin compared to native tPA. This allows for greater diffusion through the clot, potentially leading to faster clot breakdown than other agents (UpToDate, n.d.).

Unlike alteplase, reteplase does not competitively inhibit plasminogen activation. This allows for more efficient conversion of plasminogen into clot-dissolving plasmin, contributing to faster clot resolution.

Clinical Use:

The FDA has approved reteplase for managing acute myocardial infarction in a specific regimen. It is administered in two separate 10-unit boluses, injected intravenously over two minutes each, with a 30-minute interval between doses.

Additional Considerations:

While potentially offering a faster effect, reteplase still carries a risk of bleeding, similar to alteplase. Careful patient evaluation and monitoring are essential. Like alteplase, reteplase can be re-administered, if necessary, under specific circumstances. Like alteplase, reteplase lacks antigenicity, reducing the risk of allergic reactions (UpToDate, n.d.).

Tenecteplase (TNKase®)

Tenecteplase is a modified rtPA designed for improved clot targeting. It works by binding to fibrin within clots (high fibrin specificity) and activating plasminogen, an enzyme that breaks down fibrin (Bach & Lui, 2023).

Pharmacokinetics:

Tenecteplase's longer plasma half-life (compared to alteplase) allows for a single-bolus administration, improving convenience for healthcare providers and potentially reducing medication errors. It is primarily cleared through the liver, potentially reducing concerns about kidney function compared to other fibrinolytics. Tenecteplase lacks antigenicity, meaning it's less likely to trigger allergic reactions than other fibrinolytics.

Important Cautions:

Tenecteplase is not approved for ischemic stroke in many regions, including the USA. Although potentially lower than alteplase, tenecteplase still carries a risk of bleeding, especially in areas with recent injury or surgery. Careful patient evaluation and monitoring are crucial (Bach & Lui, 2023).

Fondaparinux (Arixtra®)

Fondaparinux functions differently from the fibrinolytic medications discussed previously. It belongs to a class of drugs called factor Xa inhibitors (Medscape, n.d. -f).

Fondaparinux explicitly inhibits factor Xa, a critical enzyme in the blood clotting cascade. Blocking factor Xa prevents thrombin formation, another essential enzyme for clot formation. This targeted approach helps to prevent new blood clots from developing.

Indications:

Fondaparinux is primarily used for two purposes: to prevent the formation of DVT and PE in high-risk patients, particularly those undergoing surgery.

Also, to treat existing DVT or PE when other medications may not be suitable.

Advantages:

Unlike some anticoagulants that work broadly across the clotting cascade, fondaparinux focuses on factor Xa, potentially reducing the risk of excessive bleeding.

Fondaparinux is administered as a subcutaneous injection, offering a convenient alternative to IV medications.

Unlike medications like warfarin, fondaparinux typically does not require frequent blood tests to monitor its effects.

Important Considerations:

Not a clot buster! Fondaparinux does not dissolve existing blood clots. It works to prevent new clot formation.

While offering a more targeted approach, fondaparinux still carries a risk of bleeding. Careful patient evaluation and monitoring are essential.

Fondaparinux should not be used in patients with active bleeding or a history of HIT (Medscape, n.d. -f).

Streptokinase

Streptokinase remains a popular fibrinolytic agent globally due to its relative affordability and established efficacy and safety profile. However, compared to alteplase, it is less effective in dissolving clots while carrying a lower risk of intracranial bleeding (Edwards & Nagalli, 2023).

Important Caution:

Streptokinase's high antigenicity makes re-administration within six months unsafe due to the potential for severe allergic reactions linked to the development of anti-streptococcal antibodies.

Mechanism of Action:

Unlike other listed drugs, streptokinase is not a direct plasminogen activator. Instead, it binds to circulating plasminogen, forming a complex that converts additional plasminogen to active plasmin.

Adverse Effects:

Due to its bacterial origin (Streptococcus), streptokinase frequently causes febrile reactions and other allergic responses. Additionally, dose-dependent hypotension is another potential side effect requiring careful monitoring (Edwards & Nagalli, 2023).

Urokinase

Urokinase primarily dissolves blood clots in specific locations, such as catheters and vascular thrombi. It can effectively clear blocked catheters used for medical procedures and treat blood clots in peripheral arteries or veins outside the major vessels supplying the brain or heart (Multum, 2024).

Urokinase is a naturally occurring enzyme produced by the kidneys and can be purified from human urine for therapeutic use. A commercially available, recombinant version of urokinase is also available, eliminating the need for human urine as a source.

Mechanism of Action:

Urokinase cleaves plasminogen into plasmin, the enzyme responsible for dissolving fibrin clots.

Because urokinase has a lower affinity for fibrin than other fibrinolytics, it may offer a more targeted effect on the clot itself, potentially reducing the risk of systemic breakdown of healthy fibrin throughout the body.

Repeatability:

Due to its low antigenicity, urokinase can be re-administered without concerns about antigenic problems that might occur with some other agents (Multum, 2024).

Prourokinase

Prourokinase shares some characteristics with urokinase. However, there are key considerations for its use (Baig & Bodle, 2023).

Like urokinase, prourokinase has been explored for its potential to dissolve blood clots in specific locations, such as occluded catheters and peripheral vascular thrombi.

Mechanism of Action:

Prourokinase is a single-chain plasminogen activator. However, unlike other plasminogen activators, it requires conversion to its active form (two-chain form) on the fibrin clot surface for optimal activity.

Prourokinase's activation on the clot surface may lead to a more targeted effect on the clot itself, potentially reducing the risk of systemic breakdown of healthy fibrin throughout the body.

Limited Use and Ongoing Research:

Despite this potential benefit, prourokinase has limited clinical use due to several factors.

Research and development of prourokinase are ongoing, with efforts to improve its stability and effectiveness.

Important Considerations:

Prourokinase is not a widely used medication for clot removal due to the abovementioned factors.

Urokinase and other established medications may be preferred for targeted clot removal in catheters and peripheral vessels (Baig & Bodle, 2023).

Anistreplase

Anistreplase, a thrombolytic medication that dissolves blood clots, is an anisoylated plasminogen streptokinase activator complex. However, its use is less common than that of other options.

Anistreplase cleaves plasminogen, a blood protein, into its active form, plasmin. Plasmin then breaks down fibrin, the main component of blood clots (Baig & Bodle, 2023).

Key Considerations:

Anistreplase has limited clinical use due to several factors. Compared to other established medications like alteplase or reteplase, less robust clinical data supports its widespread use for clot removal (Hazare et al., 2024).

Like other fibrinolytics, anistreplase carries a risk of bleeding, and its potential benefits may not outweigh the risks in all situations.

Other well-established medications may be preferred for dissolving clots due to their more extensive data and potentially lower bleeding risks.

Important Notes:

Due to the limitations mentioned above, anistreplase is not typically considered a first-line option for treating blood clots. There is limited ongoing research on anistreplase compared to other fibrinolytics (Hazare et al., 2024).

Reversal Agents

While anticoagulants and fibrinolytics play crucial roles in treating various blood clot-related conditions, situations may arise where their effects need to be reversed quickly due to (Ochsner Health System, 2019):

  • Uncontrolled bleeding: This could occur following surgery, trauma, or spontaneous bleeding events.
  • Iatrogenic complications: Accidental administration of an incorrect dose or overdosage can necessitate reversal.
  • Need for urgent procedures: In some situations, reversal may be necessary before performing procedures with high bleeding risk.

When a situation arises requiring reversal of anticoagulation or fibrinolysis, swift and coordinated action is essential to minimize bleeding complications. Here's a breakdown of critical steps and considerations for nurses (Aldhaeefi et al., 2023):

  1. Stop the Offending Medication:
    • The first and most crucial step is discontinuing the anticoagulant or fibrinolytic medication immediately. This helps limit further anticoagulation or clot breakdown.
  2. Assess the Bleeding Risk:
    • Evaluate the severity and source of bleeding. Minor bleeding may not necessitate reversal, while life-threatening hemorrhage demands immediate intervention.
  3. Consider Reversal Agents:
    • Here's an overview of commonly used reversal agents for different anticoagulants and fibrinolytics:

Reversing Warfarin (Vitamin K Antagonist)

Agent: Vitamin K1 (phytonadione)

Mechanism: Replenishes vitamin K stores, which are essential for the activation of vitamin K-dependent clotting factors (II, VII, IX, X).

Administration: IV or oral, depending on the urgency of reversal.

Risks and side effects: Allergic reactions, anaphylaxis (rare), and potential drug interactions.

Reversing Heparin (UF and LMWHs)

Agent: Protamine sulfate

Mechanism: Binds to heparin molecules, neutralizing their anticoagulant effect.

Administration: IV injection, usually in a controlled setting.

Risks and side effects: Hypotension, bradycardia, allergic reactions (rare), and potential for heparin rebound (recurrence of anticoagulation effect).

Reversing Dabigatran (Direct Thrombin Inhibitor)

Agent: Idarucizumab (Praxbind)

Mechanism: A specific antibody that binds to and neutralizes dabigatran, preventing its anticoagulant activity.

Administration: IV injection.

Risks and Side Effects: Hypersensitivity reactions (rare) and limited experience with long-term safety.

Reversing Rivaroxaban and Apixaban (Direct Factor Xa Inhibitors)

Agent: Andexanet alfa (Ondexia)

Mechanism: A specific antidote that binds to and inhibits rivaroxaban and apixaban, preventing their anticoagulant effects.

Administration: Intravenous injection.

Risks and Side Effects: Hypersensitivity reactions (rare) and limited experience with long-term safety.

Reversing Alteplase and Tenecteplase (Fibrinolytics)

Agent: No specific reversal agent exists.

Management: Supportive care focused on controlling bleeding, which may include:

Fresh frozen plasma (FFP): Replaces depleted clotting factors.

Cryoprecipitate: Provides a concentrated source of fibrinogen, essential for clot formation.

Aminocaproic acid and tranexamic acid: Anti-fibrinolytic agents that inhibit further clot breakdown.

Alternative Approaches if Reversal Agents Unavailable

If specific reversal agents are unavailable, then FFP can replace depleted clotting factors, offering a broader approach but lacking specificity.

Cryoprecipitate is given to Provide a concentrated source of fibrinogen, a crucial component for clot formation, in specific scenarios.

Aminocaproic acid and tranexamic acid are anti-fibrinolytic agents that inhibit further clot breakdown (not reversing existing clots, but potentially preventing further bleeding with fibrinolytic).

Nursing Considerations

Understanding reversal agent indications and limitations: Nurses should be familiar with the specific reversal agents used for different anticoagulants and fibrinolytics, as well as their indications and limitations.

Strict adherence to prescribed protocols for each reversal agent ensures safe and effective administration.

Nurses who care for patients on these medications are at the forefront of ensuring optimal outcomes. Specific care considerations should be made when caring for patients on anticoagulants or fibrinolytics (National Health Services, 2021c).

Some of these considerations include the following (Vera, 2024):

Regular Monitoring

  • Vitamin K Agonists (Warfarin): Monitor INR to ensure proper anticoagulation and prevent bleeding complications.
  • DOACs: Specific tests may be required depending on the medication.
  • Heparins: Monitor coagulation tests like aPTT.
  • Bleeding risk assessment: Identify factors that increase bleeding risk (recent surgery, trauma, uncontrolled hypertension) and collaborate with physicians to determine if anticoagulant therapy is appropriate.

Patient Education

  • Medication education: Educate patients on their specific medication, side effects, bleeding risk signs and symptoms, and the importance of adherence to the prescribed regimen.
  • Adherence support: Address patient concerns and barriers to adherence, collaborate with healthcare professionals to ensure timely medication refills, and monitor for medication interactions.

Communication and Collaboration

  • Interdisciplinary communication: Maintain clear communication with physicians and pharmacists to ensure optimal care coordination and medication management.
  • Advocacy: Stay updated on current guidelines and advocate for the safest and most effective use of these medications to improve patient outcomes.

Specific Considerations(Gutierrez & Rocuts, 2023)

Fibrinolytic Therapy

  • Close monitoring: Following fibrinolytic administration, vital signs, neurologic function (stroke patients), and bleeding sites require continuous monitoring to detect any potential complications promptly.
  • Antithrombotic precautions: After fibrinolytic use, aspirin, other antiplatelet medications, and other anticoagulants are generally avoided for a specific period to minimize bleeding risk.

Surgery(Wagner et al., 2019)

Anticoagulants, antiplatelets, and fibrinolytics must be stopped quickly to prevent bleeding complications when a patient prepares for surgery. This includes major surgeries, endoscopies, and dental procedures.

Any DOACs should be paused for 24-48 hours prior. If there is a high bleeding risk, they should be paused for up to 96 hours, depending on the patient’s renal function and the medication's half-life.

Vitamin K agonists, such as warfarin, should be paused for 7-10 days before surgery to prevent bleeding complications.

Pregnancy

Some anticoagulants such as warfarin, rivaroxaban (Xarelto®), dabigatran (Pradaxa®), edoxaban (Lixiana®), and apixaban (Eliquis®) are not recommended in pregnancy. They are known to cause congenital disabilities and may cause excessive bleeding in the placenta or the fetus.

Breastfeeding

Warfarin and heparin are considered safe to take while breastfeeding. However, rivaroxaban (Xarelto®), dabigatran (Pradaxa®), edoxaban (Lixiana®), and apixaban (Eliquis®) are not recommended as there is not enough evidence that they are safe for the baby.

Medication Reactions to Other Medicines and Remedies

Certain medications may affect anticoagulants. These include antibiotics, steroids, non-steroidal anti-inflammatory drugs (NSAIDs), antidepressants, and anticonvulsants.

Medication Reactions to Diet

Warfarin's effectiveness relies on blocking vitamin K, which is abundant in leafy greens, chickpeas, and the liver. Consistent intake is essential, while these foods can still be included in moderation. Fluctuations in vitamin K intake can complicate accurate dosage adjustments, potentially compromising the medication's effectiveness.

Additionally, certain juices like cranberry, grapefruit, and pomegranate can interfere with warfarin's metabolism, increasing the risk of bleeding. Patients should avoid these juices altogether. Vitamin K supplements can also directly counteract warfarin's mechanism, rendering it ineffective.

Excessive alcohol consumption can also interfere with warfarin's function. Patients should limit their intake to no more than one or two drinks per day and strictly avoid binge drinking.

Patient Education(Vera, 2024)

Nurses are responsible for providing safe and effective care to patients receiving anticoagulants and fibrinolytics. This includes educating patients about the treatment. Education should include medication details, bleeding risk management, adherence strategies, and other specific considerations.

Medication Knowledge

Medication name and purpose: Explain the medication's name, what it does, and why the patient takes it.

Dosage and administration: Instruct the correct dosage, frequency, and medication administration techniques.

Side effects: Discuss potential side effects like bleeding, bruising, and nausea. Explain what to do if these occur.

Bleeding Risk Management

Recognize bleeding signs: Educate patients on bleeding signs and symptoms to watch for, including unusual bruising, red or dark urine/stool, nosebleeds, and bleeding gums. Monitor for signs of blood in urine and stool, including black tarry stool and frank red blood. Severe bruising may occur if bumped or fallen. If hit in the head, it is important to seek help in the case of a brain bleed. Women may also have heavier periods on blood thinners.

Minimize bleeding risk: Avoid activities and practices that increase bleeding risk (contact sports, using sharp objects). Use a soft bristle toothbrush to minimize gum bleeding. Use an electric razor to shave rather than a blade to reduce the risks of cutting and bleeding.

Report bleeding: Instruct patients to immediately report any bleeding, especially heavy or prolonged bleeding, to the healthcare provider.

Medication Adherence(National Health Services, 2021c)

Importance of adherence: Emphasize the importance of taking the medication exactly as prescribed, even if they feel well. This ensures consistent anticoagulation levels. Medication should be taken at the same time each day.

Refills and missed doses: Explain procedures for obtaining medication refills and what to do if a dose is missed. If a dose is missed, take it as soon as possible, but do not double the dose if it is close to the next dosing.

If taking apixaban or dabigatran twice daily and missing a dose:

More than 6 hours until your next dose: Take the missed dose as soon as you remember.

Less than 6 hours until your next dose: Skip the missed dose and take the next scheduled dose as usual.

If taking rivaroxaban once daily and missing a dose:

More than 12 hours until your next dose: Take the missed dose as soon as you remember.

Less than 12 hours until your next dose: Skip the missed dose and take the next scheduled dose as usual.

For edoxaban (once daily):

Take the missed dose as soon as you remember.

If you only remember the following day, skip the missed dose and resume your regular dosing schedule.

Never take a double dose of medication to compensate for a missed dose.

Specific Considerations

Fibrinolytic therapy: For patients receiving fibrinolytics, explain the importance of avoiding strenuous activity and specific situations that could increase bleeding risk.

Dietary restrictions: Discuss potential dietary restrictions that may affect the effectiveness of some medications, especially warfarin.

If DOACs cause an upset stomach, take the medication with food to minimize stomach upset.

Case Study

Joseph Wrenn, a 67-year-old male with a history of hypertension and hyperlipidemia, presented to the emergency department experiencing sudden sharp pain and swelling in his left calf. He reported the pain started two days prior and had progressively worsened, making it difficult to walk. Joseph denied any recent trauma, infection, or prolonged immobilization. He takes lisinopril for hypertension and simvastatin for hyperlipidemia. He is allergic to penicillin and has no prior history of blood clots or bleeding disorders.

Upon examination, Joseph's left calf was tender, swollen, and warm to the touch compared to the right. Homan's sign (increased pain with dorsiflexion of the foot) was positive. A Doppler ultrasound confirmed DVT in the left popliteal vein.

Joseph's initial laboratory tests showed:

  • Platelet count: 250,000 uL (within normal range)
  • PTT: 28 seconds (within normal range)

Based on the clinical presentation and imaging confirmation, Joseph was diagnosed with DVT and started on a continuous IV heparin drip (UH) to prevent further clot formation and promote clot resolution.

History of Present Illness

Five days after starting heparin, Joseph reported experiencing new-onset chills, low-grade fever, and worsening pain in his calf. A repeat physical examination revealed the presence of scattered petechiae on his legs and abdomen, which were not present previously. A blood test showed a significant drop in his platelet count. Platelet count: 80,000 uL (thrombocytopenia).

Assessment/Screening

During routine assessments, nurses should be vigilant for signs and symptoms suggestive of HIT, particularly in patients receiving heparin therapy. Here are key elements to consider:

Onset: New-onset or worsening signs and symptoms of thrombosis 4-14 days after starting heparin.

Bleeding: Skin bleeding, such as petechiae, purpura, or oozing from wounds or mucous membranes.

Fever: Low-grade fever may occur.

Thrombocytopenia: A significant drop in platelet count (at least 50%) compared to baseline.

Precautions for HIT

History review: Nurses should inquire about any allergies to heparin or other medications and document any previous episodes of HIT.

Baseline platelet count: Measure the platelet count before starting heparin and monitor it regularly during therapy.

Heparin administration technique: Use an aseptic technique and avoid using heparin flushes in arterial lines.

Screening for HIT

4T Score: A scoring system based on clinical features (thrombocytopenia, timing of onset, type of reaction, and other factors) can help assess the pre-test probability of HIT.

HIT antibody test: A specific blood test can confirm the presence of antibodies against heparin-platelet complexes, supporting the diagnosis of HIT.

Plan of Care

Suspecting HIT, the heparin drip was immediately discontinued. A HIT antibody test was ordered, and alternative anticoagulation therapy was considered. Based on the positive HIT test result, the plan of care included:

  • Discontinuation of heparin: All forms of heparin were stopped immediately to prevent further platelet destruction.
  • Alternative anticoagulation: Fondaparinux, a medication not affected by HIT, was initiated to prevent new clot formation.
  • Platelet count monitoring: Joseph's platelet count was monitored closely to track its recovery.
  • Supportive care: Medications were administered to manage Joseph's fever and chills.

Follow Up Evaluation

John's platelet count increased within 24 hours of stopping heparin and improved gradually over the next few days. The HIT antibody test confirmed the presence of HIT antibodies, supporting the diagnosis. Fondaparinux was continued for seven days, followed by a switch to warfarin, a vitamin K antagonist, for long-term anticoagulation therapy. John was educated about HIT and the importance of avoiding heparin-based medications in the future. He was discharged home after showing clinical improvement and tolerating the new medication regimen.

Conclusion

This course has provided a comprehensive understanding of anticoagulant and fibrinolytic therapy. We have explored the physiological underpinnings of hemostasis and thrombosis, allowing you to contextualize the use of these medications. Through a detailed examination of various coagulation tests, you are now equipped to assess a patient's clotting status and inform treatment decisions. We have differentiated between anticoagulants and fibrinolytics, analyzing their mechanisms of action and clinical applications. You have gained knowledge of reversal agents and essential nursing considerations, including patient education strategies, to optimize patient outcomes. Finally, applying these concepts through interactive case studies has solidified your understanding of anticoagulant and fibrinolytic therapy in real-world scenarios.

Select one of the following methods to complete this course.

Take TestPass an exam testing your knowledge of the course material.
OR
No TestAttest that you have read and learned all the course materials.

Implicit Bias Statement

CEUFast, Inc. is committed to furthering diversity, equity, and inclusion (DEI). While reflecting on this course content, CEUFast, Inc. would like you to consider your individual perspective and question your own biases. Remember, implicit bias is a form of bias that impacts our practice as healthcare professionals. Implicit bias occurs when we have automatic prejudices, judgments, and/or a general attitude towards a person or a group of people based on associated stereotypes we have formed over time. These automatic thoughts occur without our conscious knowledge and without our intentional desire to discriminate. The concern with implicit bias is that this can impact our actions and decisions with our workplace leadership, colleagues, and even our patients. While it is our universal goal to treat everyone equally, our implicit biases can influence our interactions, assessments, communication, prioritization, and decision-making concerning patients, which can ultimately adversely impact health outcomes. It is important to keep this in mind in order to intentionally work to self-identify our own risk areas where our implicit biases might influence our behaviors. Together, we can cease perpetuating stereotypes and remind each other to remain mindful to help avoid reacting according to biases that are contrary to our conscious beliefs and values.

References

  • Aggrastat. (n.d.) Mechanism of action. Medicure. Visit Source.
  • Aldhaeefi, M., Badreldin, H. A., Alsuwayyid, F., Alqahtani, T., Alshaya, O., Al Yami, M. S., Bin Saleh, K., Al Harbi, S. A., & Alshaya, A. I. (2023). Practical guide for anticoagulant and antiplatelet reversal in clinical practice. Pharmacy, 11(1), 34. Visit Source.
  • Arif, H. & Aggarwal, S. (2023). Salicylic acid (Aspirin). In StatPearls. StatPearls Publishing. Visit Source.
  • Awosika, A. O., & Patel, P. (2023). Cangrelor. In StatPearls. StatPearls Publishing. Visit Source.
  • Bach, K. T., & Lui, F. (2023). Tenecteplase. In StatPearls. StatPearls Publishing. Visit Source.
  • Baig, M. U. & Bodle, J. (2023). Thrombolytic Therapy. In StatPearls. StatPearls Publishing. Visit Source.
  • Bansal, A. B., Sattar, Y., Patel, P., & Jamil, R. T. (2023). Eptifibatide. In StatPearls. StatPearls Publishing. Visit Source.
  • Barmore, W., Bajwa, T., & Burns, B. (2023). Biochemistry, Clotting Factors. In StatPearls. StatPearls Publishing. Visit Source.
  • Beavers, C. J. & Naqvi, I. A. (2023). Clopidogrel. In StatPearls. StatPearls Publishing. Visit Source.
  • Centers for Disease Control and Prevention (CDC). (2024). Impact of blood clots on the United States Infographic. Centers for Disease Control and Prevention. Visit Source.
  • Cleveland Clinic. (2022). Fibrinogen test. Cleveland Clinic. Visit Source.
  • Cleveland Clinic. (2023). Thrombosis. Cleveland Clinic. Visit Source.
  • Devkota, B. P. (2020). Factor II, Prothrombin Assay. Medscape. Visit Source.
  • Edwards, Z. & Nagalli, S. (2023). Streptokinase. In StatPearls. StatPearls Publishing. Visit Source.
  • Genentech. (n.d.) Assessments to consider when diagnosing acute ischemic stroke. Genentech. Visit Source.
  • Gutierrez, J. J. & Rocuts, K. R. (2023). Perioperative Anticoagulation Management. In StatPearls. StatPearls Publishing. Visit Source.
  • Hazare, C., Bhagwat, P., Singh, S. & Pillai, S. (2024). Diverse origins of fibrinolytic enzymes: A comprehensive review. Heliyon, 10(5), e26668. Visit Source.
  • Heestermans, M., Poenou, G., Hamzeh-Cognasse, H., Cognasse, F. & Bertoletti, L. (2022). Anticoagulants: A short history, their mechanism of action, pharmacology, and indications. Cells, 11(20), 3214. Visit Source.
  • Kerndt, C. C. & Nagalli, S. (2023). Dipyridamole. In StatPearls. StatPearls Publishing. Visit Source.
  • Kostousov, V. (2019). Activated clotting time. Medscape. Visit Source.
  • LaPelusa, A. & Dave, H. D. (2023). Physiology, hemostasis. In StatPearls. StatPearls Publishing. Visit Source.
  • Mahat, K. C., Sedhai, Y. R. & Krishnan, P. (2023). Argatroban. In StatPearls. StatPearls Publishing. Visit Source.
  • MedlinePlus. (2021). Platelet tests. National Library of Medicine. Visit Source.
  • MedlinePlus. (2022a). Partial thromboplastin time (PTT) test. National Library of Medicine. Visit Source.
  • MedlinePlus. (2022b). Prothrombin Time Test and INR (PT/INR). National Library of Medicine. Visit Source.
  • MedlinePlus. (2023). Rivaroxaban. National Library of Medicine. Visit Source.
  • Medscape. (n.d. -a). apixaban (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -b). dabigatran (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -c). dalteparin (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -d). edoxaban (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -e). enoxaparin (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -f). fondaparinux (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -g). Heparin (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -h). prasugrel (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -i). ticagrelor (Rx). the heart.org. Visit Source.
  • Medscape. (n.d. -j). vorapaxar (Rx). the heart.org. Visit Source.
  • Multum. C. (2024). Urokinase. Drugs.com. Visit Source.
  • National Health Services (NHS). (2021a). Considerations: Anticoagulant medicines. National Health Services. Visit Source.
  • National Health Services (NHS). (2021b). Dosage: Anticoagulant medicines. National Health Services. Visit Source.
  • National Health Services (NHS). (2021c). Uses: Anticoagulant medicines. National Health Services. Visit Source.
  • National Heart, Lung, and Blood Institute (NHLBI). (2022a). Blood clotting disorders treatment and management. National Heart, Lung, and Blood Institute. Visit Source.
  • National Heart, Lung, and Blood Institute (NHLBI). (2022b). Venous Thromboembolism Treatment. National Heart, Lung, and Blood Institute. Visit Source.
  • Ochsner Health System. (2019). Anticoagulation reversal guideline for adults. Ochsner Health System. Visit Source.
  • Patel, S., Singh, R., Preuss, C. V., & Patel, N. (2023). Warfarin. In StatPearls. StatPearls Publishing. Visit Source.
  • Patriarcheas, V., Pikoulas, A., Kostis, M., Charpidou, A., & Dimakakos, E. (2020). Heparin-induced thrombocytopenia: Pathophysiology, diagnosis and management. Cureus, 12(3), e7385. Visit Source.
  • Reed, M., Kerndt, C. C., & Nicolas D. (2023). Alteplase. In StatPearls. StatPearls Publishing. Visit Source.
  • Risman, R. A., Kirby, N. C., Bannish, B. E., Hudson, N. E., & Tutwiler, V. (2023). Fibrinolysis: An illustrated review. Research and practice in thrombosis and haemostasis, 7(2), 100081. Visit Source.
  • Sweis, R. N. & Jivan, A. (2024). Medications for acute coronary syndromes. Merck Manual Professional Edition. Visit Source.
  • Szigeti, R. G. (2022a). Anti-Xa assay (heparin assay). Medscape. Visit Source.
  • Szigeti, R. G. (2022b). D-dimer. Medscape. Visit Source.
  • Teruya, J. (2022). Thrombin time. Medscape. Visit Source.
  • UpToDate. (n.d.) Reteplase: Drug information. UpToDate. Visit Source.
  • Umerah, C. O., & Momodu, I. I. (2023). Anticoagulation. In StatPearls. StatPearls Publishing. Visit Source.
  • Vera, M. (2024). Drugs affecting coagulation (Anticoagulants, antiplatelets, thrombolytics). Nurseslabs. Visit Source.
  • Wagner, J., Lock, J. F., Kastner, C., Klein, I., Krajinovic, K., Löb, S., Germer, C. T., & Wiegering, A. (2019). Perioperative management of anticoagulant therapy. Innovative Surgical Sciences, 4(4), 144–151. Visit Source.