Your activity: 2 p.v.

Heparin (unfractionated): Pediatric drug information

Heparin (unfractionated): Pediatric drug information
(For additional information see "Heparin (unfractionated): Drug information" and see "Heparin (unfractionated): Patient drug information")

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
Brand Names: Canada
  • Heparin Leo
Therapeutic Category
  • Anticoagulant
Dosing: Neonatal

Note: Many concentrations of heparin are available and range from 1 to 20,000 units/mL. Carefully examine each prefilled syringe, bag, or vial prior to use to ensure that the correct concentration is chosen. Heparin lock flush solution is intended only to maintain patency of IV devices and is not to be used for anticoagulant therapy.

Prophylaxis

Prophylaxis:

Central line flush; patency (intermittent doses): Limited data available: Various recommendations exist for intermittent flushes of heparin used to maintain patency of single and double lumen central catheters; dose of heparin flush used should not approach therapeutic unit per kg dose; refer to institution specific protocols. In neonates, the 10 units/mL concentration is used with frequency and volume of flushing determined by the type of catheter; capped polyvinyl chloride catheters and peripheral heparin locks require flushing more frequently (eg, every 6 to 8 hours). Volume of heparin flush is usually similar to volume of catheter (or slightly greater) or may be standardized according to specific NICU policy (eg, 0.5 to 1 mL/flush). Additional flushes should be given when stagnant blood is observed in catheter, after catheter is used for drug or blood administration, and after blood withdrawal from catheter (Ref).

Central venous access device (CVAD), patency: Continuous IV infusion: 0.5 unit/kg/hour (Ref).

Extracorporeal membrane oxygenation (ECMO) (venoarterial [VA]/cardiac), anticoagulation: Note: While used to prevent thrombosis, full anticoagulation dosing is necessary; IV: 100 units/kg prior to ECMO cannulation followed by continuous heparin infusion to maintain the activated clotting time (ACT) between 180 and 220 seconds; ACT should be checked hourly while patient is on ECMO; additional monitoring targets for heparin therapy are prolongation of the PTT to 1.5 to 2.5 times the control value or an anti-Xa level of 0.3 to 0.7 units/mL (Ref).

Parenteral nutrition (PN) additive, venous access patency: 0.5 to 1 unit/mL (final heparin concentration in the PN solution), both central and peripheral PN solutions. The final concentration of heparin used for PN solutions may need to be decreased in small neonates receiving larger PN volumes in order to avoid approaching therapeutic amounts (Ref).

Peripheral arterial catheters in situ: Intra-arterial (via arterial catheter): Continuous infusion of heparin at a final concentration of 5 units/mL at 1 mL/hour (Ref).

Umbilical artery catheter (UAC): Intra-arterial (via UAC): Continuous infusion of heparin at a final concentration of 0.25 to 1 unit/mL; total heparin dose of 25 to 200 units/kg/day to maintain patency (Ref).

Thromboprophylaxis in congenital heart defect (CHD) patients with systemic to pulmonary artery shunts (eg, Sano shunt, Blalock-Taussig shunt, central shunt) or central venous lines in certain CHD patients (eg, previous thrombosis or hypercoagulable state): Low dose: Continuous IV infusion: 10 to 15 units/kg/hour (Ref).

Thrombosis; treatment

Thrombosis; treatment:

Systemic heparinization: IV: Initial loading dose: 75 units/kg over 10 minutes; then initial continuous maintenance infusion at 28 units/kg/hour; adjust dose to maintain an anti-Xa activity of 0.35 to 0.7 units/mL or an aPTT range that correlates to this anti-Xa range or a protamine titration range of 0.2 to 0.4 units/mL (Ref).

Note: Because of variation among hospitals with reagents (lot numbers) and corresponding control of aPTT values, individual institutions should establish unique, institution-specific nomograms based on current reagent. Due to extensive variability within reagents and anti-Xa levels with corresponding aPTTs, a specific nomogram has not been provided; refer to guidelines for a specific nomogram (Ref).

Systemic to pulmonary artery shunt thrombosis (eg, Sano shunt, Blalock-Taussig shunt, central shunt); treatment in CHD patients: Bolus: 50 to 100 units/kg, ongoing continuous infusion should be considered (Ref).

Dosing: Pediatric

Note: Many concentrations of heparin are available and range from 1 to 20,000 units/mL. Carefully examine each prefilled syringe, bag, or vial prior to use to ensure that the correct concentration is chosen. Heparin lock flush solution is intended only to maintain patency of IV devices and is not to be used for anticoagulant therapy.

Prophylaxis

Prophylaxis:

Central line flush; patency (intermittent doses): Limited data available (Ref): Infants, Children, and Adolescents: When using intermittent flushes of heparin to maintain patency of single and double lumen central catheters, various recommendations exist; refer to institution specific protocols. Capped polyvinyl chloride catheters and peripheral heparin locks require flushing more frequently (eg, every 6 to 8 hours). Volume of heparin flush is usually similar to volume of catheter (or slightly greater). Dose of heparin flush used should not approach therapeutic unit per kg dose. Additional flushes should be given when stagnant blood is observed in catheter, after catheter is used for drug or blood administration, and after blood withdrawal from catheter.

Extracorporeal membrane oxygenation (ECMO) (venoarterial [VA]/cardiac), anticoagulation: Note: While used to prevent thrombosis, full anticoagulation dosing is necessary; Infants, Children, and Adolescents: IV: 100 units/kg prior to ECMO cannulation followed by continuous IV heparin infusion to maintain the activated clotting time (ACT) between 180 and 220 seconds; ACT should be checked hourly while patient is on ECMO; additional monitoring targets for heparin therapy are prolongation of the PTT to 1.5 to 2.5 times the control value or an anti-Xa level of 0.3 to 0.7 units/mL (Ref).

Parenteral nutrition (PN) additive, venous access patency: Infants, Children, and Adolescents: 1 unit/mL (final heparin concentration in PN), both central and peripheral. The final concentration of heparin used for PN solutions may need to be decreased to 0.5 units/mL in small infants receiving larger PN volumes in order to avoid approaching therapeutic amounts (Ref).

Peripheral arterial catheters in situ: Infants, Children, and Adolescents: Intra-arterial (via arterial catheter): Continuous infusion of heparin at a final concentration of 5 units/mL at 1 mL/hour (Ref).

Thromboprophylaxis in congenital heart defect (CHD) patients with systemic to pulmonary artery shunts (eg, Sano shunt, Blalock-Taussig shunt, central shunt) or central venous lines in certain CHD patients (eg, previous thrombosis or hypercoagulable states): Infants, Children, and Adolescents: Low Dose: Continuous IV infusion: 10 to 15 units/kg/hour (Ref).

Thrombosis, treatment

Thrombosis, treatment:

Systemic heparinization:

Infants: IV: Initial loading dose: 75 units/kg over 10 minutes; then initial continuous maintenance infusion at: 28 units/kg/hour; adjust dose to maintain an anti-Xa activity of 0.35 to 0.7 units/mL or an aPTT range that correlates to this anti-Xa range or a protamine titration range of 0.2 to 0.4 units/mL (Ref).

Children and Adolescents: IV: Initial loading dose: 75 units/kg over 10 minutes, then initial continuous maintenance infusion at: 20 units/kg/hour; adjust dose to maintain an anti-Xa activity of 0.35 to 0.7 units/mL or an aPTT range that correlates to this anti-Xa range or a protamine titration range of 0.2 to 0.4 units/mL (Ref). Note: A lower initial continuous maintenance infusion dose of 18 units/kg/hour in older patients has also been suggested (Ref).

Note: Because of variation among hospitals with reagents (lot numbers) and corresponding control of aPTT values, individual institutions should establish unique, institution-specific nomograms based on current reagent. Due to extensive variability within reagents and anti-Xa levels with corresponding aPTTs, a specific nomogram has not been provided; refer to guidelines for a specific nomogram (Ref).

Systemic to pulmonary artery shunt thrombosis (eg, Sano shunt, Blalock-Taussig shunt, central shunt); treatment in CHD patients: Infants, Children, and Adolescents: IV: Bolus: 50 to 100 units/kg, ongoing continuous infusion should be considered (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Pediatric

All patients: No dosage adjustment required; adjust therapeutic heparin according to aPTT or anti-Xa activity

Dosing: Hepatic Impairment: Pediatric

All patients: No dosage adjustment required; adjust therapeutic heparin according to aPTT or anti-Xa activity

Dosing: Adult

(For additional information see "Heparin (unfractionated): Drug information")

Note: Heparin may be given by continuous IV infusion or SubQ depending on the indication. For weight-based IV heparin, an institution-specific dosing nomogram may help to achieve therapeutic anticoagulation more rapidly (see example based on aPTT under "Venous thromboembolism treatment"). If unusually large doses of heparin are required to achieve therapeutic targets, consider possible heparin resistance (Ref). Safety: Many concentrations of heparin are available ranging from 1 to 20,000 units/mL. Carefully examine each prefilled syringe or vial to ensure the correct concentration is selected.

Antibiotic lock technique, adjunctive therapy

Antibiotic lock technique, adjunctive therapy (catheter-salvage strategy) (off-label use): Note: Antibiotic lock therapy is used in addition to systemic antibiotics for certain catheter-related infections when the catheter cannot be removed. Heparin is incompatible with ethanol and should not be used with ethanol lock therapy (Ref). Heparin is also incompatible with certain antibiotics; confirm compatibility prior to use (Ref).

Intracatheter: 100 to 5,000 units/mL in combination with an appropriate antibiotic. Heparin concentration depends on compatibility with the selected antibiotic, antibiotic concentration, and catheter type, which may vary by institution (Ref). For patients with end-stage renal disease requiring hemodialysis, maximum final heparin concentration should not exceed 1,000 units/mL due to increased risk of bleeding (Ref). Instill into each lumen of the catheter access port using a sufficient volume to fill the catheter (eg, 2 to 5 mL) with a maximum dwell time of ≤72 hours, depending on frequency of catheter use. Withdraw lock solution prior to catheter use; replace with fresh lock solution after catheter use (Ref).

Atrial fibrillation

Atrial fibrillation (to prevent stroke and systemic embolism): Note: When admitted for short-term hospitalization (eg, admission for a procedure or surgery), ambulatory patients taking an oral anticoagulant and not at high risk of immediate thromboembolism typically do not require bridging anticoagulation. Patients at high risk of thromboembolism (eg, recent cardioversion, high CHA2DS2-VASc score, prior cardioembolic stroke, current intracardiac thrombus) may be considered for bridging with a parenteral anticoagulant (see Transitioning between anticoagulants below) (Ref).

IV: Initial bolus of 60 to 80 units/kg (maximum: 5,000 units), followed by a continuous infusion of 12 to 18 units/kg/hour (maximum: 1,000 units/hour). Institutional dosing protocols may vary; adjust infusion rate to maintain anticoagulation target (Ref).

Frostbite

Frostbite (adjunctive agent) (off-label use): Note: Patients should be transferred to a facility that is familiar with managing patients with frostbite. Anticoagulation may be used in conjunction with thrombolytic therapy, either concurrently or following administration of the thrombolytic, but not as monotherapy (Ref). Optimal regimens, administration routes, and doses, including the use of therapeutic or low-dose anticoagulation after thrombolytic therapy, have not been identified; refer to institutional protocols.

Example regimens include:

When thrombolytic is administered IV:

IV: 500 to 1,000 units/hour as a fixed dose or targeted to an aPTT 2× control for up to 6 hours (Ref); some centers extend the duration to up to 5 days (Ref) or transition to therapeutic low-molecular-weight heparin (LMWH) (eg, enoxaparin) (Ref).

When thrombolytic is administered intra-arterial:

Intra-arterial: 500 units/hour administered through the intra-arterial catheter; targeted to a goal aPTT 50 to 70 seconds; after discontinuation of thrombolytic therapy, continue anticoagulation for 72 to 96 hours or transition to therapeutic LMWH (eg, enoxaparin) (Ref). Instead of intra-arterial heparin, some suggest the use of IV heparin after intra-arterial thrombolytic (see IV dosing above) (Ref).

Hemodialysis, anticoagulation of circuit

Hemodialysis, anticoagulation of circuit:

Note: Standard dosing has not been established for intermittent hemodialysis or CRRT. Recommendations provided below are example regimens. Refer to institutional protocols. May need to individualize dose based on patient-specific needs.

Intermittent hemodialysis:

Standard risk for bleeding:

IV: Initial: Bolus 1,000 units or 2,000 units at the beginning of hemodialysis, followed by a continuous infusion of 500 units/hour; stop the infusion 60 minutes before the end of hemodialysis (Ref).

If clotting occurs during first half of hemodialysis session: Increase IV bolus by 500 units before each subsequent session (eg, administer 1,500 units or 2,500 units, depending on the previously attempted dose, at the beginning of hemodialysis) until clotting no longer occurs or up to a maximum of 4,000 units; administer continuous infusion as described above (Ref).

If clotting occurs during second half of hemodialysis session: Administer the same IV bolus dose, but increase continuous infusion by 100 units/hour for each subsequent session (eg, administer 600 units/hour) until clotting no longer occurs or up to a maximum of 1,000 units/hour; stop the infusion 30 minutes before the end of hemodialysis (Ref).

High risk for bleeding:

Note: There is no standard practice to prevent clotting of the hemodialysis circuit among patients who are at high risk for bleeding. Dialysis without heparin is preferred, but may consider heparin anticoagulation in patients who continue to have recurrent filter thrombosis (Ref).

IV: Initial: Bolus 1,000 units at the beginning of hemodialysis; do not administer a continuous infusion during the procedure (Ref).

If clotting occurs during hemodialysis: IV: Bolus 1,000 units at the beginning of the subsequent hemodialysis session, followed by a continuous infusion of 500 units/hour; stop the infusion 60 minutes before the end of hemodialysis; if clotting continues, increase continuous infusion by 100 units/hour at each subsequent session (eg, administer 600 units/hour) until clotting no longer occurs or up to a maximum of 1,000 units/hour (Ref).

CRRT:

Note: For patients who require anticoagulation due to repeated clotting of hemofilter and when regional citrate anticoagulation is not feasible or unavailable (Ref).

IV: Initial: Bolus 500 to 1,000 units, followed by a continuous infusion of 500 units/hour; titrate to maintain an aPTT ~1.5 times the control or ~45 seconds (Ref).

Ischemic heart disease

Ischemic heart disease:

Acute coronary syndromes:

ST-elevation myocardial infarction (off-label use):

Adjunct to percutaneous coronary intervention: see Percutaneous coronary intervention for dosing guidance.

Adjunct to fibrinolysis: IV: Bolus 60 units/kg (maximum: 4,000 units), followed by 12 units/kg/hour (maximum: 1,000 units/hour); adjust infusion rate to maintain anticoagulation target based on institutional protocol; continue for ≥48 hours or until revascularization (if performed) (Ref).

No planned reperfusion: IV: Bolus 50 to 70 units/kg (maximum: 5,000 units), followed by 12 units/kg/hour; adjust infusion rate to maintain anticoagulation target based on institutional protocol; continue for ≥48 hours (Ref).

Non-ST-elevation acute coronary syndromes (off-label use):

Ischemia-guided (conservative) approach (alternative agent): IV: Bolus 60 units/kg (maximum: 5,000 units), followed by 12 units/kg/hour (maximum: 1,000 units/hour); adjust infusion rate to maintain anticoagulation target based on institutional protocol; continue for ≥48 hours or until management changes to an invasive strategy (eg, percutaneous coronary intervention [PCI]) (Ref); if PCI is performed, see Percutaneous coronary intervention for dosing guidance.

Invasive approach (adjunct to percutaneous coronary intervention): see Percutaneous coronary intervention for dosing guidance.

Percutaneous coronary intervention (off-label use):

No prior anticoagulant therapy:

No planned glycoprotein IIb/IIIa inhibitor use: IV: Initial bolus of 70 to 100 units/kg (maximum: 10,000 units) to achieve activated clotting time (ACT) of 250 to 300 seconds (goal ACT may vary depending on point-of-care device); repeat bolus as needed to maintain goal ACT throughout procedure (Ref).

Planned glycoprotein IIb/IIIa inhibitor use: IV: Initial bolus of 50 to 70 units/kg (maximum: 7,000 units) to achieve ACT of 200 to 250 seconds (regardless of point-of-care device); repeat bolus as needed to maintain goal ACT throughout procedure (Ref).

Prior anticoagulant therapy:

Prior anticoagulation with heparin:

No planned glycoprotein IIb/IIIa inhibitor use: IV: Check ACT prior to PCI and administer heparin bolus as needed (eg, 2,000 to 5,000 units) to achieve ACT of 250 to 300 seconds (goal ACT may vary depending on point-of-care device); repeat bolus (maximum: 10,000 units) as needed to maintain goal ACT throughout procedure (Ref).

Planned glycoprotein IIb/IIIa inhibitor use: IV: Check ACT prior to PCI and administer heparin bolus as needed (eg, 2,000 to 5,000 units) to achieve ACT of 200 to 250 seconds (regardless of point-of-care device); repeat bolus (maximum 7,000 units) as needed to maintain goal ACT throughout procedure (Ref).

Prior anticoagulation with enoxaparin:

If percutaneous coronary intervention occurs ≤12 hours after the last SubQ dose of enoxaparin: Transition to unfractionated heparin is not recommended. Refer to enoxaparin monograph for dosing recommendations.

If percutaneous coronary intervention occurs >12 hours after the last SubQ dose of enoxaparin: May use unfractionated heparin; refer to recommendations above for PCI with no prior anticoagulant therapy (Ref).

Prior anticoagulation with fondaparinux:

No planned glycoprotein IIb/IIIa inhibitor use: IV: Initial bolus of 85 units/kg (maximum: 10,000 units) to achieve ACT of 250 to 300 seconds (goal ACT may vary depending on point-of-care device); repeat bolus as needed to maintain goal ACT throughout procedure (Ref).

Planned glycoprotein IIb/IIIa inhibitor use: IV: Initial bolus of 60 units/kg (maximum: 7,000 units) to achieve ACT of 200 to 250 seconds (regardless of point-of-care device); repeat bolus as needed to maintain goal ACT throughout procedure (Ref).

Mechanical heart valve, bridging anticoagulation

Mechanical heart valve, bridging anticoagulation (for interruptions in warfarin therapy) (off-label use): Note: Bridging during intervals of subtherapeutic anticoagulation should be considered for patients with mechanical mitral or tricuspid valve replacement; however, for patients with mechanical aortic valve replacement, bridging is not required unless an additional thromboembolic risk factor is present or patient has an older-generation mechanical aortic valve (Ref).

IV: Limited data available: Initial: 12 to 18 units/kg/hour (no bolus) starting when INR falls below the therapeutic range; adjust infusion rate to maintain anticoagulation target based on institutional protocol. If patient is to undergo an invasive procedure, discontinue heparin 4 to 6 hours prior to procedure; reinitiate heparin infusion as soon as possible after the procedure when bleeding risk is acceptable. Continue heparin until warfarin has been reinitiated and INR is within therapeutic range for 2 consecutive days (Ref).

Mechanical heart valve, postsurgical management

Mechanical heart valve, postsurgical management (to transition to warfarin) (off-label use): Note: Initiate postoperatively when risk of bleeding is acceptable (Ref).

IV: Limited data available: Initial: 12 to 18 units/kg/hour (no bolus); adjust infusion rate to maintain anticoagulation target based on institutional protocol. Overlap with warfarin until INR is stable and within therapeutic range for ≥2 consecutive days (Ref).

Peripheral arterial occlusion, acute

Peripheral arterial occlusion, acute (off-label use): Note: Specific dosing information is limited, but anticoagulation is commonly used at the time of diagnosis to limit thrombus propagation while the patient is evaluated for other possible interventions (Ref).

IV: Initial bolus of 60 to 80 units/kg, followed by an initial continuous infusion of 12 to 18 units/kg/hour; adjust infusion rate to maintain anticoagulation target based on institutional protocol (Ref).

Venous thromboembolism prophylaxis

Venous thromboembolism prophylaxis (alternative agent): Note: Low-weight patients (eg, <50 kg) may be more sensitive to routine prophylactic doses, increasing the potential for higher than intended levels of anticoagulation; consider adhering to every-12-hour dosing interval (Ref).

Medical patients with acute illness at moderate to high risk for venous thromboembolism: SubQ: 5,000 units every 8 to 12 hours; continue for length of hospitalization or until fully ambulatory (Ref); extended prophylaxis beyond acute hospital stay is not routinely recommended (Ref).

Nonorthopedic surgery:

Patients with active cancer:

SubQ: 5,000 units 2 to 4 hours prior to surgery, then 5,000 units every 8 hours thereafter (Ref) or 5,000 units every 8 to 12 hours started ~6 to 24 hours after surgery (Ref). Note: The optimal duration of prophylaxis has not been established, but it is usually given for a minimum of 7 to 10 days; extending for up to 4 weeks may be reasonable in those undergoing major abdominal or pelvic surgery (Ref).

Patients without cancer: Note: For patients with moderate or high risk of venous thromboembolism (VTE) and low risk of bleeding.

SubQ: 5,000 units every 8 to 12 hours, with initial dose given ≥2 hours prior to surgery. Alternatively, may postpone pharmacologic prophylaxis until after surgery (eg, high bleeding risk) when it is safe to initiate. Continue until fully ambulatory and risk of VTE has diminished (typically up to 10 days) (Ref).

Orthopedic surgery (eg, hip fracture surgery, total hip arthroplasty, total knee arthroplasty): SubQ: 5,000 units every 8 to 12 hours, with initial dose administered ≥12 hours preoperatively or ≥12 hours postoperatively once hemostasis is achieved; optimal duration of prophylaxis is unknown, but it is usually given for a minimum of 10 to 14 days and can be extended for up to 35 days; some experts suggest a duration in the lower end of the range (10 to 14 days) for total knee arthroplasty or higher end of the range (~30 days) for total hip arthroplasty (Ref). For extended duration of prophylaxis, may transition to an oral anticoagulant or alternative SubQ anticoagulant with less frequent dosing (Ref).

Pregnancy: Note: For patients at moderate or high VTE risk during antepartum and postpartum periods. Dose intensity is individualized based on risk of thrombosis and bleeding complications (Ref).

Prophylactic dose (also referred to as intermediate dose to account for weight gain during pregnancy):

First trimester: SubQ: 5,000 to 7,500 units every 12 hours (Ref).

Second trimester: SubQ: 7,500 to 10,000 units every 12 hours (Ref).

Third trimester: SubQ: 10,000 units every 12 hours (reduce dose if the aPTT becomes elevated) (Ref).

Adjusted dose (therapeutic): Note: Reserved for patients at the highest VTE risk (eg, history of recurrent thrombosis, severe thrombophilia).

SubQ: 10,000 units every 12 hours; adjust dose to target an aPTT of 1.5 to 2.5 times control, measured 6 hours after injection; monitor aPTT once daily until stable and within therapeutic range, then monitor every 1 to 2 weeks (Ref).

Note: Discontinue heparin at the onset of spontaneous labor. Prior to planned induction of labor or cesarean delivery, discontinue heparin ≥12 hours before in patients receiving 7,500 to 10,000 units every 12 hours or ≥24 hours before in patients receiving >10,000 units/dose or >20,000 units/day. Consider checking coagulation parameters before delivery. Appropriate discontinuation is particularly important if neuraxial anesthesia is planned. May restart ≥4 to 6 hours after vaginal delivery or ≥6 to 12 hours after cesarean delivery, unless significant bleeding occurred or traumatic neuraxial catheter placement. Anticoagulation should continue for up to 6 weeks postpartum, but potentially longer (Ref).

Venous thromboembolism treatment, deep vein thrombosis and/or pulmonary embolism

Venous thromboembolism treatment, deep vein thrombosis and/or pulmonary embolism: Note: IV heparin may be preferred for initial therapy in patients who are hemodynamically unstable, may need invasive procedures or thrombolysis due to extensive clot burden, are obese, have renal failure, or when rapid reversal of anticoagulation may be needed (Ref). If thrombolytics are used, it is recommended to discontinue heparin during administration then resume upon completion of the thrombolytic infusion (Ref).

Inpatient treatment: IV: Initial: 80 units/kg bolus followed by a continuous infusion of 18 units/kg/hour or 5,000 unit bolus followed by 1,333 units/hour; adjust infusion rate to maintain target laboratory values based on institutional protocol (Ref). Note: Weight-based dosing is more effective than fixed dosing at reaching therapeutic anticoagulation (Ref).

Example Weight-Based IV Heparin Nomogram for Treatment of Venous Thromboembolisma,b

aHull 2022a

bThis is one example of a weight-based heparin dosing nomogram. Each institution should establish their own heparin dosing nomogram. Other heparin nomograms based on aPTT or anti-Factor Xa monitoring may be employed. Therapeutic range for aPTT must be established at each individual laboratory (Dager 2018).

cUse actual body weight for calculations.

Initial dose and monitoring

80 units/kg bolus (maximum dose: 10,000 units)c, then 18 units/kg/hour (maximum initial infusion: 2,000 units/hour)c

Obtain aPTT 6 hours after initial heparin bolus

Dosing adjustments and monitoring

If using anti-Factor Xa activity (units/mL)

Response

If using aPTT (seconds)

0 to 0.09

• Bolus 25 units/kg

• Increase infusion by 3 units/kg/hour

• Repeat assay in 6 hours

<40

0.1 to 0.19

• Increase infusion by 2 units/kg/hour

• Repeat assay in 6 hours

40 to 49

0.2 to 0.29

• Increase infusion by 1 unit/kg/hour

• Repeat assay in 6 hours

50 to 69

0.3 to 0.7

• No change (within therapeutic range)

• Repeat assay in 6 hours

• Once therapeutic for 2 consecutive assays, may change to once-daily assays

70 to 110

0.71 to 0.79

• Decrease infusion by 1 unit/kg/hour

• Repeat assay in 6 hours

111 to 120

0.8 to 0.89

• Stop infusion for 1 hour, then decrease by 2 units/kg/hour

• Repeat assay 6 hours after restarting the infusion

121 to 130

0.9 to 0.99

• Stop infusion for 1 hour, then decrease by 3 units/kg/hour

• Repeat assay 6 hours after restarting the infusion

131 to 140

1 to 1.09

• Stop infusion for 2 hours, then decrease by 4 units/kg/hour

• Repeat assay 6 hours after restarting the infusion

141 to 150

≥1.1

• Stop infusion for 2 hours, then decrease by 5 units/kg/hour and notify clinician

• Repeat assay 6 hours after restarting the infusion

>150

Outpatient treatment: Note: Alternative for patients who have a contraindication to other anticoagulants.

SubQ: Initial: 333 units/kg, followed by 250 units/kg every 12 hours (Ref).

Transitioning between anticoagulants: Note: This provides general guidance on transitioning between anticoagulants; also refer to local protocol for additional detail.

Transitioning from another anticoagulant to IV heparin:

Transitioning from a therapeutic dose of SubQ low-molecular-weight heparin or SubQ fondaparinux to a therapeutic dose of IV heparin: Start IV heparin without a bolus dose (infusion rate depends on the indication) 1 to 2 hours before the next dose of low-molecular-weight heparin (LMWH) or fondaparinux would have been due (Ref).

Transitioning from warfarin to a therapeutic dose of IV heparin: Stop warfarin and, when INR is as close as possible to the lower end of the targeted INR range, start IV heparin without a bolus dose (infusion rate depends on the indication) (Ref).

Transitioning from a direct-acting oral anticoagulant to a therapeutic dose of IV heparin: Stop direct-acting oral anticoagulant (DOAC) and, when the next DOAC dose would have been due, start IV heparin without a bolus dose (infusion rate depends on the indication) (Ref).

Transitioning from IV heparin to another anticoagulant:

Transitioning from a therapeutic dose of IV heparin to therapeutic SubQ low-molecular-weight heparin or SubQ fondaparinux: Stop IV heparin and within 1 hour start SubQ LMWH or SubQ fondaparinux. Note: If aPTT is not within therapeutic range at the time heparin is stopped, consult local protocol (Ref).

Transitioning from a therapeutic dose of IV heparin to warfarin: Start warfarin and continue IV heparin until INR is within therapeutic range (Dager 2018; Hull 2022a). Note: Overlap IV heparin with warfarin until INR is ≥2 for at least 2 measurements taken ~24 hours apart (duration of overlap is ~5 days) (Ref).

Transitioning from a therapeutic dose of IV heparin to a direct-acting oral anticoagulant: Start DOAC when the heparin infusion is stopped (consult local protocol if the aPTT is above the target range) (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Adult

The renal dosing recommendations are based upon the best available evidence and clinical expertise. Senior Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.

Altered kidney function: IV, SubQ: Mild to severe impairment: No initial dosage adjustment necessary; adjust to maintain anticoagulation target based on institutional protocol (Ref).

Renal replacement therapies: Poorly dialyzed (Ref): IV, SubQ: No supplemental dose or initial dosage adjustment necessary in patients receiving renal replacement therapies (eg, hemodialysis, peritoneal dialysis, CRRT, PIRRT); adjust to maintain anticoagulation target based on institutional protocol (Ref).

Dosing: Hepatic Impairment: Adult

No dosage adjustment required; adjust therapeutic heparin according to aPTT or anti-Factor Xa activity.

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling. [DSC] = Discontinued product

Solution, Intravenous:

Generic: 2000 units (1000 mL); 25,000 units (500 mL); 2000 units in 0.9% NaCl per liter (1000 mL); 25,000 units/500 mL (500 mL); 25,000 units/500 mL in NaCl 0.45% (500 mL)

Solution, Injection, as sodium:

Generic: 12,500 units (250 mL); 25,000 units (250 mL); 1000 units/mL (1 mL, 10 mL, 30 mL); 5000 units/mL (1 mL, 10 mL); 10,000 units/mL (1 mL, 4 mL, 5 mL); 20,000 units/mL (1 mL)

Solution, Injection, as sodium [preservative free]:

Generic: 1000 units/mL (2 mL); 5000 units/mL (1 mL); 5000 units/0.5 mL (0.5 mL)

Solution, Intravenous, as sodium:

Generic: 1000 units (500 mL); 10,000 units (250 mL); 20,000 units (500 mL); 25,000 units (250 mL); 1 units/mL (1 mL, 2 mL, 2.5 mL, 3 mL, 5 mL, 10 mL); 10 units/mL (1 mL, 2 mL, 2.5 mL, 3 mL, 5 mL, 10 mL); 100 units/mL (1 mL, 2 mL, 2.5 mL, 3 mL, 5 mL, 10 mL); 1000 units/500 mL in NaCl 0.9% (500 mL); 25,000 units/250 mL in Dextrose 5% (250 mL); 25,000 units/250 mL in NaCl 0.45% (250 mL)

Solution, Intravenous, as sodium [preservative free]:

Generic: 10 units/mL (1 mL [DSC], 3 mL [DSC], 5 mL [DSC]); 100 units/mL (1 mL [DSC], 3 mL [DSC], 5 mL [DSC]); 25,000 units/250 mL in Dextrose 5% (250 mL)

Solution Prefilled Syringe, Injection, as sodium [preservative free]:

Generic: 5000 units/0.5 mL (0.5 mL)

Generic Equivalent Available: US

Yes

Dosage Forms: Canada

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Solution, Intravenous:

Generic: 25,000 units/500 mL (500 mL)

Solution, Injection, as sodium:

Generic: 1000 units/mL (1 mL, 2 mL, 10 mL, 30 mL); 10,000 units/mL (0.5 mL, 1 mL, 5 mL); 1000 units (500 mL, 1000 mL)

Solution, Intravenous, as sodium:

Generic: 20,000 units/500 mL in Dextrose 5% (500 mL); 100 units/mL (2 mL, 10 mL); 25,000 units/250 mL in Dextrose 5% (250 mL)

Solution Prefilled Syringe, Subcutaneous:

Generic: 5000 units/0.5 mL (0.5 mL)

Administration: Pediatric

Note: Many concentrations of heparin are available and range from 1 to 20,000 units/mL. Carefully examine each prefilled syringe, bag, or vial prior to use to ensure that the correct concentration is chosen.

Parenteral: Do not administer IM due to pain, irritation, and hematoma formation.

IV:

IV bolus: Administered over 10 minutes (Ref).

Continuous IV infusion: Infuse via infusion pump.

Heparin lock: Inject via injection cap using positive pressure flushing technique. Heparin lock flush solution is intended only to maintain patency of IV devices and is not to be used for anticoagulant therapy.

Central venous catheters: Must be flushed with heparin solution when newly inserted, daily (at the time of tubing change), after blood withdrawal or transfusion, and after an intermittent infusion through an injectable cap.

SubQ: Not all preparation intended for SubQ administration, verify product selection. Inject in subcutaneous tissue only (not muscle tissue). Injection sites should be rotated (usually left and right portions of the abdomen, above iliac crest).

Administration: Adult

SubQ: Inject in subcutaneous tissue only (not muscle tissue). Injection sites should be rotated (usually left and right portions of the abdomen, above iliac crest).

IM: Do not administer IM due to pain, irritation, and hematoma formation.

Continuous IV infusion: Infuse via infusion pump. If preparing solution, mix thoroughly prior to administration.

Heparin lock: Inject via injection cap using positive pressure flushing technique. Heparin lock flush solution is intended only to maintain patency of IV devices and is not to be used for anticoagulant therapy.

Central venous catheters: Must be flushed with heparin solution when newly inserted, daily (at the time of tubing change), after blood withdrawal or transfusion, and after an intermittent infusion through an injectable cap. A volume of at least 10 mL of blood should be removed and discarded from a heparinized line before blood samples are sent for coagulation testing.

Intravesical (off-label use): Various dosage regimens of heparin (20,000 to 50,000 units) alone or with alkalinized lidocaine (1% to 4%) have been instilled into the bladder.

Intra-arterial (off-label use): Infuse via infusion pump. If preparing solution, mix thoroughly prior to administration (Ref).

Usual Infusion Concentrations: Neonatal

IV infusion: Maintenance of line patency: 0.5 unit/mL

Usual Infusion Concentrations: Pediatric

Note: Premixed solutions available

IV infusion: 100 units/mL

Storage/Stability

Heparin solutions are colorless to slightly yellow. Minor color variations do not affect therapeutic efficacy. Heparin should be stored at room temperature. Protect from freezing and temperatures >40°C.

Stability at room temperature and refrigeration:

Prepared bag: Variable (specific to solution, concentration, and/or study conditions); also refer to manufacturer's labeling.

Premixed bag: After seal is broken, 4 days.

Out of overwrap stability: 30 days.

Use

Systemic: Injection: Prophylaxis and treatment of thromboembolic disorders and anticoagulant for blood transfusions, extracorporeal circulation, and dialysis procedures (FDA approved in pediatric patients [age not specified] and adults).

Local: Heparin lock flush solution: Maintain patency of IV devices (FDA approved in adults).

Medication Safety Issues
Sound-alike/look-alike issues:

Heparin may be confused with Hespan

High alert medication:

The Institute for Safe Medication Practices (ISMP) includes this medication among its list of drugs which have a heightened risk of causing significant patient harm when used in error.

National Patient Safety Goals:

The Joint Commission (TJC) requires healthcare organizations that provide anticoagulant therapy to have approved protocols and evidence-based practice guidelines in place to reduce the risk of anticoagulant-associated patient harm. Patients receiving anticoagulants should receive individualized care through a defined process that includes medication selection, dosing (including adjustments for age, renal function, or liver function), drug-drug interactions, drug-food interactions, other applicable risk factors, monitoring, patient and family education, proper administration, reversal of anticoagulation, management of bleeding events, and perioperative management. This does not apply to routine short-term use of anticoagulants for prevention of venous thromboembolism during procedures or hospitalizations (NPSG.03.05.01).

Administration issues:

The 100 unit/mL concentration should not be used to flush heparin locks, IV lines, or intra-arterial lines in neonates or infants <10 kg (systemic anticoagulation may occur). The 10 unit/mL flush concentration may inadvertently cause systemic anticoagulation in infants <1 kg who receive frequent flushes.

Other safety concerns:

Heparin sodium injection 10,000 units/mL and Hep-Lock U/P 10 units/mL have been confused with each other. Fatal medication errors have occurred between the two whose labels are both blue. Never rely on color as a sole indicator to differentiate product identity.

Labeling changes: Effective May 1, 2013, heparin labeling is required to include the total amount of heparin per vial (rather than only including the amount of heparin per mL). During the transition, hospitals should consider only stocking the newly labeled heparin to avoid potential errors and confusion with the older labeling.

Heparin lock flush solution is intended only to maintain patency of IV devices and is not to be used for anticoagulant therapy.

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified.

Postmarketing:

Cardiovascular: Vasospasm (including cyanotic extremities, limb ischemia, and limb pain)

Dermatologic: Skin ulceration at injections site (SUBQ), transient alopecia

Endocrine & metabolic: Acute adrenocortical insufficiency, adrenal hemorrhage, hyperkalemia, ovarian hemorrhage, suppression of aldosterone synthesis

Genitourinary: Priapism

Hematologic & oncologic: Bruise, hemorrhage, heparin-induced thrombocytopenia retroperitoneal hemorrhage, thrombocytopenia (often of no clinical significance with continued therapy), thrombosis in heparin-induced thrombocytopenia (including acute myocardial infarction, cerebrovascular accident, deep vein thrombosis, gangrene of the extremities, pulmonary embolism, skin necrosis)

Hepatic: Increased serum alanine aminotransferase, increased serum aspartate aminotransferase

Hypersensitivity: Anaphylactic shock, hypersensitivity reaction, nonimmune anaphylaxis, severe infusion related reaction (skin necrosis)

Local: Erythema at injection site (SUBQ), hematoma at injection site (SUBQ), irritation at injection site (SUBQ), pain at injection site (SUBQ), tissue necrosis at injection site (SUBQ)

Neuromuscular & skeletal: Osteoporosis (with long-term use)

Contraindications

Hypersensitivity to heparin or any component of the formulation (unless a life-threatening situation necessitates use and use of an alternative anticoagulant is not possible); severe thrombocytopenia; history of heparin-induced thrombocytopenia; history of heparin-induced thrombocytopenia with thrombosis; uncontrolled active bleeding; not for use when appropriate blood coagulation tests cannot be obtained at appropriate intervals (applies to full-dose heparin only).

Note: Some products contain benzyl alcohol as a preservative; their use in neonates, infants, or pregnant or breastfeeding women is contraindicated by some manufacturers.

Warnings/Precautions

Concerns related to adverse effects:

• Bleeding: May occur, including fatal events. Use with caution in patients with an increased risk of bleeding, including subacute bacterial endocarditis; congenital or acquired bleeding disorders; active ulcerative or angiodysplastic GI diseases; continuous GI tube drainage; severe uncontrolled hypertension; history of hemorrhagic stroke; use shortly after brain, spinal, or ophthalmologic surgery or other invasive procedures including spinal tap or spinal anesthesia; concomitant treatment with platelet inhibitors; recent GI bleeding; impaired hemostasis; thrombocytopenia or platelet defects; patients with hereditary antithrombin deficiency receiving concurrent antithrombin replacement therapy; severe liver disease; hypertensive or diabetic retinopathy; renal failure; or in patients (especially women) >60 years of age. Discontinue if bleeding occurs; severe hemorrhage or overdosage may require protamine (consult Protamine monograph for dosing recommendations).

• Heparin resistance: Dose requirements >35,000 units/24 hours to maintain a therapeutic aPTT may occur in patients with antithrombin deficiency, increased heparin clearance, elevations in heparin-binding proteins, and elevations in factor VIII and/or fibrinogen; frequently encountered in patients with fever, thrombosis, thrombophlebitis, infections with thrombosing tendencies, myocardial infarction, cancer, and in postsurgical patients; measurement of anticoagulant effects using anti-Factor Xa levels may be of benefit.

• Hepatic effects: Elevations in serum aminotransferases have been observed during therapy. These elevations should be evaluated with caution as they may occur and resolve in the setting of the underlying condition for which heparin is being used.

• Hyperkalemia: Hyperkalemia may occur, especially in patients with diabetes, renal impairment, history of metabolic acidosis, history of hyperkalemia, or taking concomitant potassium-sparing medication; may suppress aldosterone production.

• Hypersensitivity reactions: Hypersensitivity reactions, including fever, chills, urticaria, asthma, rhinitis, lacrimation, and anaphylaxis, have been reported. In patients with a documented hypersensitivity reaction, heparin should only be considered in life-threatening situations when use of an alternative anticoagulant is not possible. Some products are derived from animal tissue and may be contraindicated in patients with animal allergies (ie, pork); consult individual prescribing information.

• Osteoporosis: May occur with prolonged use (>6 months) due to a reduction in bone mineral density.

• Thrombocytopenia: Mild thrombocytopenia (platelet count >100,000/m3) may occur during therapy. Heparin-induced thrombocytopenia (HIT), a serious antibody-mediated reaction resulting from irreversible aggregation of platelets, may occur. Patients who develop HIT may be at risk of developing a new thrombus (heparin-induced thrombocytopenia with thrombosis [HITT]). Discontinue therapy and consider alternatives if platelet count falls below 100,000/mm3, there is a >50% reduction in platelet count from baseline, and/or thrombosis develops while on heparin therapy. Onset of HIT or HITT is usually delayed (5 to 10 days after exposure in heparin-naive individuals) and can occur up to several weeks after discontinuation of heparin. “Rapid onset” HIT can occur within 24 hours of heparin initiation, especially in patients with recent heparin exposure within the previous 100 days. Use with extreme caution (for a limited duration) or avoid use in patients with history of HIT, especially if administered within 100 days of a HIT episode (Dager 2007; Warkentin 2001).

Special populations:

• Older adult: Use with caution in patients >60 years of age, particularly women; older adults can be more sensitive to heparin and a higher incidence of bleeding has been reported in these patients. May require lower doses.

Dosage form specific issues:

• Benzyl alcohol and derivatives: Some dosage forms may contain benzyl alcohol as a preservative. In neonates, large amounts of benzyl alcohol (≥99 mg/kg/day) have been associated with a potentially fatal toxicity ("gasping syndrome"); the "gasping syndrome" consists of metabolic acidosis, respiratory distress, gasping respirations, CNS dysfunction (including convulsions, intracranial hemorrhage), hypotension, and cardiovascular collapse (AAP ["Inactive" 1997]; CDC 1982); some data suggests that benzoate displaces bilirubin from protein binding sites (Ahlfors 2001); avoid or use dosage forms containing benzyl alcohol with caution in neonates. See manufacturer's labeling. Use in neonates, infants, or pregnant or nursing mothers is contraindicated by some manufacturers; the use of preservative-free heparin is, therefore, recommended in these populations.

• Sulfites: Some preparations contain sulfite which may cause allergic reactions.

Other warnings/precautions:

• Fatal medications errors: Many concentrations of heparin are available ranging from 1 unit/mL to 20,000 units/mL. Clinicians must carefully examine each prefilled syringe or vial prior to use ensuring that the correct concentration is chosen; fatal hemorrhages have occurred related to heparin overdose especially in pediatric patients.

Warnings: Additional Pediatric Considerations

Confirm the concentration of all heparin injection vials prior to administration; do not use heparin injection as a "catheter lock flush" as the injection is supplied in various concentrations including highly concentrated strengths. Fatal hemorrhages have occurred in pediatric patients when higher concentrations of heparin injection were confused with lower concentrations of heparin lock flush.

Heparin-induced thrombocytopenia (HIT) has been reported in pediatric patients; incidence and risk factors are not well-defined due to variability related to patient inclusion and laboratory techniques; incidence rates up to 2.3% in PICU patients have been reported, and cases observed at both low and high levels of heparin exposure (eg, venous access device line flushes and supratherapeutic doses associated with hemodialysis and during cardiopulmonary bypass). Monitor platelet count closely; if HIT develops, consider alternate anticoagulation therapy (eg, danaparoid, argatroban) (ACCP [Monagle 2012]).

Heparin resistance should be suspected in pediatric patients if unable to achieve an activated clotting time (ACT) >300 seconds after administration of >600 units/kg (ACCP [Giglia 2013]).

Metabolism/Transport Effects

None known.

Drug Interactions

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program by clicking on the “Launch drug interactions program” link above.

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program

Acalabrutinib: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Agents with Antiplatelet Properties (e.g., P2Y12 inhibitors, NSAIDs, SSRIs, etc.): May enhance the anticoagulant effect of Heparin. Management: Decrease the dose of heparin or agents with antiplatelet properties if coadministration is required. Risk D: Consider therapy modification

Alemtuzumab: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Aliskiren: Heparin may enhance the hyperkalemic effect of Aliskiren. Risk C: Monitor therapy

Andexanet Alfa (Coagulation Factor Xa [Recombinant], Inactivated): May diminish the therapeutic effect of Heparin. Risk X: Avoid combination

Angiotensin II Receptor Blockers: Heparin may enhance the hyperkalemic effect of Angiotensin II Receptor Blockers. Risk C: Monitor therapy

Angiotensin-Converting Enzyme Inhibitors: Heparin may enhance the hyperkalemic effect of Angiotensin-Converting Enzyme Inhibitors. Risk C: Monitor therapy

Antithrombin: May enhance the anticoagulant effect of Heparin. Risk C: Monitor therapy

Apixaban: May enhance the anticoagulant effect of Anticoagulants. Refer to separate drug interaction content and to full drug monograph content regarding use of apixaban with vitamin K antagonists (eg, warfarin, acenocoumarol) during anticoagulant transition and bridging periods. Risk X: Avoid combination

Bromperidol: May enhance the adverse/toxic effect of Anticoagulants. Risk C: Monitor therapy

Caplacizumab: May enhance the anticoagulant effect of Anticoagulants. Management: Avoid coadministration of caplacizumab with antiplatelets if possible. If coadministration is required, monitor closely for signs and symptoms of bleeding. Interrupt use of caplacizumab if clinically significant bleeding occurs. Risk D: Consider therapy modification

Collagenase (Systemic): Anticoagulants may enhance the adverse/toxic effect of Collagenase (Systemic). Specifically, the risk of injection site bruising and/or bleeding may be increased. Risk C: Monitor therapy

Corticorelin: Heparin may enhance the adverse/toxic effect of Corticorelin. Significant hypotension and bradycardia have been previously attributed to this combination. Risk X: Avoid combination

Dabigatran Etexilate: May enhance the anticoagulant effect of Anticoagulants. Refer to separate drug interaction content and to full drug monograph content regarding use of dabigatran etexilate with vitamin K antagonists (eg, warfarin, acenocoumarol) during anticoagulant transition and bridging periods. Risk X: Avoid combination

Dasatinib: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Deferasirox: Anticoagulants may enhance the adverse/toxic effect of Deferasirox. Specifically, the risk for GI ulceration/irritation or GI bleeding may be increased. Risk C: Monitor therapy

Deoxycholic Acid: Anticoagulants may enhance the adverse/toxic effect of Deoxycholic Acid. Specifically, the risk for bleeding or bruising in the treatment area may be increased. Risk C: Monitor therapy

Desirudin: Anticoagulants may enhance the anticoagulant effect of Desirudin. Management: Discontinue treatment with other anticoagulants prior to desirudin initiation. If concomitant use cannot be avoided, monitor patients receiving these combinations closely for clinical and laboratory evidence of excessive anticoagulation. Risk D: Consider therapy modification

Drospirenone-Containing Products: May enhance the hyperkalemic effect of Heparin. Risk C: Monitor therapy

Edoxaban: May enhance the anticoagulant effect of Anticoagulants. Refer to separate drug interaction content and to full drug monograph content regarding use of edoxaban with vitamin K antagonists (eg, warfarin, acenocoumarol) during anticoagulant transition and bridging periods. Management: Some limited combined use may be indicated during periods of transition from one anticoagulant to another. See the full edoxaban drug monograph for specific recommendations on switching anticoagulant treatment. Risk X: Avoid combination

Eplerenone: Heparin may enhance the hyperkalemic effect of Eplerenone. Risk C: Monitor therapy

Factor X (Human): Anticoagulants (Inhibitors of Factor Xa) may diminish the therapeutic effect of Factor X (Human). Risk C: Monitor therapy

Hemin: May enhance the anticoagulant effect of Anticoagulants. Risk X: Avoid combination

Herbal Products with Anticoagulant/Antiplatelet Effects (eg, Alfalfa, Anise, Bilberry): May enhance the adverse/toxic effect of Anticoagulants. Bleeding may occur. Risk C: Monitor therapy

Ibritumomab Tiuxetan: Anticoagulants may enhance the adverse/toxic effect of Ibritumomab Tiuxetan. Both agents may contribute to an increased risk of bleeding. Risk C: Monitor therapy

Ibrutinib: May enhance the adverse/toxic effect of Anticoagulants. Risk C: Monitor therapy

Icosapent Ethyl: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Inotersen: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Kanamycin: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Limaprost: May enhance the adverse/toxic effect of Anticoagulants. The risk for bleeding may be increased. Risk C: Monitor therapy

Lipid Emulsion (Fish Oil Based): May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Mesoglycan: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

MiFEPRIStone: May enhance the adverse/toxic effect of Anticoagulants. Specifically, the risk of bleeding may be increased. Risk X: Avoid combination

Nintedanib: Anticoagulants may enhance the adverse/toxic effect of Nintedanib. Specifically, the risk for bleeding may be increased. Risk C: Monitor therapy

Nitroglycerin: May diminish the anticoagulant effect of Heparin. Nitroglycerin may decrease the serum concentration of Heparin. Risk C: Monitor therapy

Nonsteroidal Anti-Inflammatory Agents: May enhance the anticoagulant effect of Heparin. Management: Decrease the dose of heparin or nonsteroidal anti-inflammatory agents (NSAIDs) if coadministration is required. Risk D: Consider therapy modification

Nonsteroidal Anti-Inflammatory Agents (Topical): May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Obinutuzumab: Anticoagulants may enhance the adverse/toxic effect of Obinutuzumab. Specifically, the risk of serious bleeding-related events may be increased. Risk C: Monitor therapy

Omacetaxine: Anticoagulants may enhance the adverse/toxic effect of Omacetaxine. Specifically, the risk for bleeding-related events may be increased. Management: Avoid concurrent use of anticoagulants with omacetaxine in patients with a platelet count of less than 50,000/uL. Risk X: Avoid combination

Omega-3 Fatty Acids: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Oritavancin: May diminish the therapeutic effect of Heparin. Specifically, oritavancin may artificially increase the results of laboratory tests commonly used to monitor IV heparin effectiveness, which could lead to incorrect decisions to decrease heparin doses. Risk X: Avoid combination

Palifermin: Heparin may increase the serum concentration of Palifermin. Management: If heparin is used to maintain an intravenous line, rinse the line with saline prior to and after palifermin administration. Risk C: Monitor therapy

Pentosan Polysulfate Sodium: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Pentoxifylline: May enhance the anticoagulant effect of Heparin. Risk C: Monitor therapy

Potassium Salts: Heparin may enhance the hyperkalemic effect of Potassium Salts. Risk C: Monitor therapy

Potassium-Sparing Diuretics: Heparin may enhance the hyperkalemic effect of Potassium-Sparing Diuretics. Management: Monitor serum potassium concentrations closely. The spironolactone Canadian product monograph lists its combination with heparin or low molecular weight heparins as contraindicated. Risk C: Monitor therapy

Prostacyclin Analogues: May enhance the adverse/toxic effect of Anticoagulants. Specifically, the antiplatelet effects of these agents may lead to an increased risk of bleeding with the combination. Risk C: Monitor therapy

Rivaroxaban: Anticoagulants may enhance the anticoagulant effect of Rivaroxaban. Refer to separate drug interaction content and to full drug monograph content regarding use of rivaroxaban with vitamin K antagonists (eg, warfarin, acenocoumarol) during anticoagulant transition and bridging periods. Risk X: Avoid combination

Salicylates: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Streptokinase: May enhance the anticoagulant effect of Heparin. Risk X: Avoid combination

Sugammadex: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Sulodexide: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Telavancin: May diminish the therapeutic effect of Heparin. Specifically, telavancin may artificially increase the results of laboratory tests commonly used to monitor IV heparin effectiveness, which could lead to incorrect decisions to decrease heparin doses. Risk X: Avoid combination

Thrombolytic Agents: May enhance the anticoagulant effect of Anticoagulants. Management: Monitor for signs and symptoms of bleeding if these agents are combined. For the treatment of acute ischemic stroke, avoidance with anticoagulants is often recommended, see full Lexicomp or drug interaction monograph for details. Risk C: Monitor therapy

Tibolone: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Tipranavir: May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Tobacco (Smoked): May decrease the serum concentration of Heparin. Risk C: Monitor therapy

Urokinase: May enhance the anticoagulant effect of Anticoagulants. Risk X: Avoid combination

Vitamin E (Systemic): May enhance the anticoagulant effect of Anticoagulants. Risk C: Monitor therapy

Vitamin K Antagonists (eg, warfarin): Anticoagulants may enhance the anticoagulant effect of Vitamin K Antagonists. Risk C: Monitor therapy

Vorapaxar: May enhance the adverse/toxic effect of Anticoagulants. More specifically, this combination is expected to increase the risk of bleeding. Risk X: Avoid combination

Zanubrutinib: May enhance the adverse/toxic effect of Anticoagulants. Risk C: Monitor therapy

Pregnancy Considerations

Heparin does not cross the placenta (ESC [Regitz-Zagrosek 2018]).

Due to pregnancy-induced physiologic changes, the risk of thromboembolism is increased during pregnancy and the immediate postpartum period. Heparin may be used for anticoagulation in pregnancy (ACOG 196 2018). Due to a better safety profile and ease of administration, the use of low molecular weight heparin (LMWH) is generally preferred over heparin (unfractionated heparin [UFH]) in pregnancy (ACOG 196 2018; Bates 2018; ESC [Regitz-Zagrosek 2018]). Anticoagulant therapy for the prevention and treatment of thromboembolism in pregnant patients can be discontinued prior to induction of labor or a planned cesarean delivery (Bates 2018) or LMWH can be converted to UFH in higher risk patients (ESC [Regitz-Zagrosek 2018]). Consult current recommendations for appropriate use in pregnancy.

Patients with mechanical heart valves have an increased risk of adverse maternal and fetal outcomes and these risks are greater without appropriate anticoagulation. UFH or LMWH may be used in pregnant patients with mechanical heart valves. Increased monitoring is required to maintain adequate therapeutic concentrations during pregnancy (consult current recommendations for details) (ESC [Regitz-Zagrosek 2018]; ACC/AHA [Otto 2021]).

Some products contain benzyl alcohol as a preservative; their use in pregnant patients is contraindicated by some manufacturers; use of a preservative-free formulation is recommended.

Monitoring Parameters

Hemoglobin, hematocrit, signs of bleeding; fecal occult blood test; aPTT (or antifactor Xa activity levels) or activated clotting time (ACT) depending upon indication. Thromboelastogram (TEG) may be useful in monitoring overall hemostasis and fibrinolysis in patients receiving unfractionated heparin for therapeutic anticoagulation (AHA [Giglia 2013]).

Platelet counts should be routinely monitored (eg, every 2 to 3 days on days 4 to 14 of heparin therapy) when the risk of heparin‐induced thrombocytopenia (HIT) is >1% (eg, receiving therapeutic dose heparin, postoperative antithrombotic prophylaxis), if the patient has received heparin or low molecular weight heparin (eg, enoxaparin) within the past 100 days, if preexposure history is uncertain, or if anaphylactoid reaction to heparin occurs. When the risk of HIT is <1% (eg, medical/obstetrical patients receiving heparin flushes), routine platelet count monitoring is not recommended (ACCP [Guyatt 2012]).

For patients on extracorporeal membrane oxygenation (ECMO), ACT should be checked hourly while patient is on ECMO (AHA [Giglia 2013]).

Reference Range

Venous thromboembolism: Heparin: pediatric patients: 0.35 to 0.7 unit/mL; adults: 0.3 to 0.7 unit/mL anti-Xa activity (by chromogenic assay) or 0.2 to 0.4 unit/mL (by protamine titration); aPTT: 1.5 to 2.5 times control (usually reflects an aPTT of 60 to 85 seconds) (Garcia 2012; ACCP [Monagle 2012])

ECMO anticoagulation: Maintain the ACT between 180 and 220 seconds; additional monitoring targets for heparin therapy are prolongation of the PTT to 1.5 to 2.5 times the control value and an anti-Xa level of 0.3 to 0.7 units/mL (AHA [Giglia 2013])

Mechanism of Action

Potentiates the action of antithrombin III and thereby inactivates thrombin (as well as other coagulation factors IXa, Xa, XIa, XIIa, and plasmin) and prevents the conversion of fibrinogen to fibrin; heparin also stimulates release of lipoprotein lipase (lipoprotein lipase hydrolyzes triglycerides to glycerol and free fatty acids)

Pharmacokinetics (Adult data unless noted)

Note: Increased interpatient variability of pharmacokinetic parameters in pediatric patients compared to adults; however, age-related decreases in volume of distribution and clearance with increasing pediatric patient age have been reported (ACCP [Monagle 2012]; McDonald 1981).

Onset of action: Anticoagulation: IV: Immediate; SubQ: ~20 to 30 minutes.

Absorption: Oral, rectal: Erratic at best from these routes of administration; SubQ absorption is also erratic, but considered acceptable for prophylactic use.

Distribution:

Premature neonates (data based on single dose of 100 units/kg within 4 hours of birth) (McDonald 1981): Inversely proportional to gestational age (GA).

GA 25 to 28 weeks: 81 ± 41 mL/kg.

GA 29 to 32 weeks: 73.3 ± 24.8 mL/kg.

GA 33 to 36 weeks: 57.8 ± 32.2 mL/kg.

Adults: Following a single 75 unit/kg dose: 36.6 ± 7.4 mL/kg (McDonald 1981).

Metabolism: Complex; thought to occur by depolymerization and desulphation via the reticuloendothelial system primarily in the liver and spleen (ACCP [Garcia 2012]; Dawes 1979; Estes 1980; Kandrotas 1992).

Half-life elimination:

Age-related: Shorter half-life reported in premature neonates compared to adult patients.

Premature neonates GA 25 to 36 weeks (data based on single dose of 100 units/kg within 4 hours of birth): Mean range: 35.5 to 41.6 minutes (McDonald 1981).

Dose-dependent: IV bolus: 25 units/kg: 30 minutes (Bjornsson 1982); 100 units/kg: 60 minutes (de Swart 1982); 400 units/kg: 150 minutes (Olsson 1963).

Mean: 1.5 hours; Range: 1 to 2 hours; affected by obesity, renal function, malignancy, presence of pulmonary embolism, and infections.

Note: At therapeutic doses, elimination occurs rapidly via nonrenal mechanisms. With very high doses, renal elimination may play more of a role; however, dosage adjustment remains unnecessary for patients with renal impairment (Kandrotas 1992).

Excretion: Urine (small amounts as unchanged drug); Note: At therapeutic doses, elimination occurs rapidly via nonrenal mechanisms. With very high doses, renal elimination may play more of a role; however, dosage adjustment remains unnecessary for patients with renal impairment (Kandrotas 1992).

Clearance: Age-related changes; within neonatal population, slower clearance with lower GA; however, when compared to adults, the overall clearance in neonatal and pediatric patients is faster than adults (ACCP [Monagle 2012 ]; McDonald 1981).

Pharmacokinetics: Additional Considerations

Altered kidney function: The half-life may be increased.

Hepatic function impairment: The half-life may be increased or decreased.

Older adult: Plasma levels may be higher.

Additional Information

Updates to the United States Pharmacopeia (USP) heparin monograph were made in response to over 200 deaths linked to contaminated heparin products in 2007-2008. Serious adverse effects (including hypersensitivity reactions) were associated with a heparin-like contaminant (oversulfated chondroitin sulfate). At the time, the available quality assurance tests did not test for oversulfated chondroitin sulfate. Effective October 1, 2009, a new reference standard for heparin and a new test to determine potency (the chromogenic antifactor IIa test) were established by USP. The updated USP heparin monograph also harmonized the USP unit with the WHO international standard (IS) unit (ie, international unit). The new standard may result in a 10% reduction in potency for heparin marketed in the United States. The FDA has requested that all manufacturers differentiate (from "old" heparin products) heparin products manufactured by the new standards. The labels of products manufactured according to the new standard will have an "N" in the lot number or following the expiration date. Additionally, products manufactured by Hospira may be identified by the number "82" or higher (eg, 83, 84) at the beginning of their lot numbers. For therapeutic use, practitioners may or may not notice that larger doses of heparin are required to achieve "therapeutic" activity of anticoagulation. The impact of this change in potency should be less significant when heparin is administered by subcutaneous injection due to low and variable bioavailability. Heparin dosing should always be individualized according to the patient-specific clinical situation. Appropriate clinical judgment is essential in determining heparin dosage (Smythe 2010).

Pricing: US

Solution (Heparin (Porcine) in NaCl Intravenous)

1000UT/500ML 0.9% (per mL): $0.01

2000UNIT/L 0.9% (per mL): $0.01

12500UT/250ML 0.45% (per mL): $0.04

25000UT/250ML 0.45% (per mL): $0.04 - $0.05

25000UT/500ML 0.45% (per mL): $0.02

Solution (Heparin Sod (Porcine) in D5W Intravenous)

40UNIT/ML 5% (per mL): $0.03

100 units/mL (per mL): $0.04 - $0.07

25000UT/500ML 5% (per mL): $0.02 - $0.04

Solution (Heparin Sod (Pork) Lock Flush Intravenous)

1 units/mL (per mL): $1.16 - $1.25

10 units/mL (per mL): $0.13 - $0.72

100 units/mL (per mL): $0.19 - $0.74

Solution (Heparin Sodium (Porcine) Injection)

1000 units/mL (per mL): $0.27 - $0.91

5000 units/mL (per mL): $1.45 - $5.20

10000 units/mL (per mL): $2.52 - $9.16

20000 units/mL (per mL): $12.00 - $20.95

Solution (Heparin Sodium (Porcine) PF Injection)

5000 units/0.5 mL (per 0.5 mL): $2.58 - $7.20

5000 units/mL (per mL): $7.20

Solution Prefilled Syringe (Heparin Sodium (Porcine) Injection)

5000 units/0.5 mL (per 0.5 mL): $6.84 - $6.84

Disclaimer: A representative AWP (Average Wholesale Price) price or price range is provided as reference price only. A range is provided when more than one manufacturer's AWP price is available and uses the low and high price reported by the manufacturers to determine the range. The pricing data should be used for benchmarking purposes only, and as such should not be used alone to set or adjudicate any prices for reimbursement or purchasing functions or considered to be an exact price for a single product and/or manufacturer. Medi-Span expressly disclaims all warranties of any kind or nature, whether express or implied, and assumes no liability with respect to accuracy of price or price range data published in its solutions. In no event shall Medi-Span be liable for special, indirect, incidental, or consequential damages arising from use of price or price range data. Pricing data is updated monthly.

Brand Names: International
  • Agglutek (TW);
  • Anticlot (VN);
  • Caprin (IN);
  • Coaparin (PL);
  • Haidyparin (EG);
  • Hemastat (PH);
  • Hemonor (PE);
  • Hepaflex (FI, NO);
  • Hepagusan (ID);
  • Heparen (LK);
  • Heparil (UA);
  • Heparin (AT, BF, BG, BJ, CH, CI, CZ, DE, ET, FI, GB, GH, GM, GN, GR, IL, KE, LR, MA, ML, MR, MU, MW, NE, NG, NO, SC, SD, SE, SL, SN, TN, TZ, UG, ZA, ZM, ZW);
  • Heparin Injection B.P. (AU);
  • Heparin Leo (DK, HK, ID, MY, PH, TW);
  • Heparin Novo (TW);
  • Heparin Sodium B Braun (ID, MY);
  • Heparina (ES);
  • Heparine (BE, NL);
  • Heparine Choay (FR);
  • Heparine Novo (BE, NL);
  • Heprin (PH);
  • Hepsal (MT, SA, SG);
  • Heptin (PH);
  • Inhepar (CR, DO, GT, HN, NI, PA, SV);
  • Inviclot (ID);
  • Kabihep (PH);
  • Lioton (HR, RU, UA);
  • Lioton Gel (RO);
  • Liquemin (DE, IT);
  • Liquemine (BE, TR);
  • Lyogel (UA);
  • Minihep (AE, QA);
  • Monoparin (QA, SA);
  • Multiparin (NZ, PK);
  • Nuparin (PH);
  • Pharepa (LB);
  • Proparin (CR, DO, GT, HN, MX, NI, PA, SV);
  • Rhoneparina (PY);
  • Sakarin (PH);
  • Thrombophob (DE);
  • Thrombophob-S (EG);
  • Thromboreduct (DE);
  • Unihep (AE, QA);
  • Wellparin (VN)


For country code abbreviations (show table)
  1. Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Oral anticoagulant therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e44S-e88S. doi: 10.1378/chest.11-2292. [PubMed 22315269]
  2. Ahlfors CE. Benzyl alcohol, kernicterus, and unbound bilirubin. J Pediatr. 2001;139(2):317-319. [PubMed 11487763]
  3. American College of Obstetricians and Gynecologists (ACOG) Committee on Practice Bulletins-Obstetrics. ACOG Practice Bulletin No. 196: Thromboembolism in pregnancy. Obstet Gynecol. 2018;132(1):e1-e17. doi: 10.1097/AOG.0000000000002706. [PubMed 29939938]
  4. Amsterdam EA, Wenger NK, Brindis RG, et al; ACC/AHA Task Force Members. 2014 AHA/ACC guideline for the management of patients with non-ST-elevation acute coronary syndromes: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published correction appears in Circulation. 2014;130(25):e433-e444]. Circulation. 2014;130(25):e344-e426. doi: 10.1161/CIR.0000000000000134. [PubMed 25249585]
  5. Andrew M, Marzinotto V, Massicotte P, et al, “Heparin Therapy in Pediatric Patients: A Prospective Cohort Study,” Pediatr Research, 1994, 35(1):78-83. [PubMed 8134203]
  6. Antman EM, Morrow DA, McCabe CH, et al; ExTRACT-TIMI 25 Investigators. Enoxaparin versus unfractionated heparin with fibrinolysis for ST-elevation myocardial infarction. N Engl J Med. 2006;354(14):1477-1488. doi: 10.1056/NEJMoa060898. [PubMed 16537665]
  7. Balestrino D, Quintana M, Charbonnel N, Forestier C, Lartigue C, Souweine B. Compatibility of injectable anticoagulant agents in ethanol; in vitro antibiofilm activity and impact on polyurethane catheters of enoxaparin 400 U/mL in 40% v/v ethanol. PLoS One. 2016;11(7):e0159475. doi: 10.1371/journal.pone.0159475. [PubMed 27441664]
  8. Barletta JF, DeYoung JL, McAllen K, Baker R, Pendleton K. Limitations of a standardized weight-based nomogram for heparin dosing in patients with morbid obesity. Surg Obes Relat Dis. 2008;4(6):748-753. doi: 10.1016/j.soard.2008.03.005 [PubMed 18586569]
  9. Based on expert opinion.
  10. Bates SM, Greer IA, Middeldorp S, Veenstra DL, Prabulos AM, Vandvik PO. VTE, thrombophilia, antithrombotic therapy, and pregnancy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e691S-e736S. doi: 10.1378/chest.11-2300. [PubMed 22315276]
  11. Bates SM, Rajasekhar A, Middeldorp S, et al. American Society of Hematology 2018 guidelines for management of venous thromboembolism: venous thromboembolism in the context of pregnancy. Blood Adv. 2018;2(22):3317-3359. doi: 10.1182/bloodadvances.2018024802. [PubMed 30482767]
  12. Bates SM, Weitz JI, Johnston M, Hirsh J, Ginsberg JS. Use of a fixed activated partial thromboplastin time ratio to establish a therapeutic range for unfractionated heparin. Arch Intern Med. 2001;161(3):385-391. [PubMed 11176764]
  13. Bauer KA. Risk and prevention of venous thromboembolism in adults with cancer. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed October 4, 2021.
  14. Bauer KA. Use of anticoagulants during pregnancy and postpartum. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 20, 2022.
  15. Bauer SR, Ou NN, Dreesman BJ, et al. Effect of body mass index on bleeding frequency and activated partial thromboplastin time in weight-based dosing of unfractionated heparin: a retrospective cohort study. Mayo Clin Proc. 2009;84(12):1073-1078. doi: 10.4065/mcp.2009.0220 [PubMed 19955244]
  16. Bjornsson T, Wolfram B, Kitchell B. Heparin kinetics determined by three assay methods. Clin Pharmacol Ther. 1982;31(1):104–113. [PubMed 7053298]
  17. Bookstaver PB, Rokas KE, Norris LB, Edwards JM, Sherertz RJ. Stability and compatibility of antimicrobial lock solutions. Am J Health Syst Pharm. 2013;70(24):2185-2198. doi: 10.2146/ajhp120119. [PubMed 24296841]
  18. Bookstaver PB, Williamson JC, Tucker BK, Raad II, Sherertz RJ. Activity of novel antibiotic lock solutions in a model against isolates of catheter-related bloodstream infections. Ann Pharmacother. 2009;43(2):210-219. doi: 10.1345/aph.1L145. [PubMed 19193593]
  19. Bradford NK, Edwards RM, Chan RJ. Heparin versus 0.9% sodium chloride intermittent flushing for the prevention of occlusion in long term central venous catheters in infants and children: A systematic review. Int J Nurs Stud. 2016;59:51-59. [PubMed 27222450]
  20. Braun JD. Embolism to the lower extremities. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 5, 2022.
  21. Bruen KJ, Ballard JR, Morris SE, Cochran A, Edelman LS, Saffle JR. Reduction of the incidence of amputation in frostbite injury with thrombolytic therapy. Arch Surg. 2007;142(6):546-551. doi:10.1001/archsurg.142.6.546 [PubMed 17576891]
  22. Burch HB. Drug effects on the thyroid. N Engl J Med. 2019;381(8):749-761. doi: 10.1056/NEJMra1901214. [PubMed 31433922]
  23. Butt W, Shann F, McDonnell G, Hudson I. Effect of heparin concentration and infusion rate on the patency of arterial catheters. Crit Care Med. 1987;15(3):230-232. [PubMed 3545674]
  24. Centers for Disease Control (CDC). Neonatal deaths associated with use of benzyl alcohol—United States. MMWR Morb Mortal Wkly Rep. 1982;31(22):290-291. http://www.cdc.gov/mmwr/preview/mmwrhtml/00001109.htm [PubMed 6810084]
  25. Cloherty JP, Eichenwald EC, Martin CR, eds. Manual of Neonatal Care. 8th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2017.
  26. Cohen M, Demers C, Gurfinkel EP, et al. A comparison of low-molecular-weight heparin with unfractionated heparin for unstable coronary artery disease. Efficacy and Safety of Subcutaneous Enoxaparin in Non-Q-Wave Coronary Events Study Group. N Engl J Med. 1997;337(7):447-452. doi: 10.1056/NEJM199708143370702. [PubMed 9250846]
  27. Conway MA, McCollom C, Bannon C. Central venous catheter flushing recommendations: a systematic evidence-based practice review. J Pediatr Oncol Nurs. 2014;31(4):185-190. [PubMed 24794887]
  28. Conway SE, Hwang AY, Ponte CD, Gums JG. Laboratory and clinical monitoring of direct acting oral anticoagulants: what clinicians need to know. Pharmacotherapy. 2017;37(2):236-248. doi:10.1002/phar.1884 [PubMed 27983747]
  29. Corkin MR, ed. Pediatric Nutrition Support Core Curriculum. 2nd edition. Silver Spring, MD: American Society for Parenteral and Enteral Nutrition; 2015.
  30. Cruickshank MK, Levine MN, Hirsh J, et al, “A Standard Heparin Nomogram for the Management of Heparin Therapy,” Arch Intern Med, 1991, 151(2):333-7. [PubMed 1789820]
  31. Cutlip D, Levin T. Antithrombotic therapy for elective percutaneous coronary intervention: general use. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed February 6, 2020b.
  32. Cutlip D, Lincoff AM. Anticoagulant therapy in non-ST elevation acute coronary syndromes. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 7, 2020a.
  33. Dager WE, Dougherty JA, Nguyen PH, et al. Heparin-Induced Thrombocytopenia: Treatment Options and Special Considerations. Pharmacotherapy. 2007;27(4):564-587. [PubMed 17381384]
  34. Dager WE, Gulseth MP, Nutescu EA, eds. Anticoagulation Therapy: A Clinical Practice Guide. 2nd ed. American Society of Health-System Pharmacists; 2018.
  35. Dager WE, Kiser TH. Systemic anticoagulation considerations in chronic kidney disease. Adv Chronic Kidney Dis. 2010;17(5):420-427. doi:10.1053/j.ackd.2010.06.002 [PubMed 20727512]
  36. Dager WE, Tsu LV, Pon TK. Considerations for Systemic Anticoagulation in ESRD. Semin Dial. 2015;28(4):354-362. doi:10.1111/sdi.12376 [PubMed 25951901]
  37. Davenport A. Anticoagulation for continuous renal replacement therapy. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. August 7, 2020.
  38. Davies MJ, D'Alessio DA, Fradkin J, et al. Management of hyperglycemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2018;41(12):2669-2701. doi:10.2337/dci18-0033 [PubMed 30291106]
  39. Dawes J, Papper DS. Catabolism of low-dose heparin in man. Thromb Res. 1979;14(6):845-860. [PubMed 473122]
  40. Decousus HA, Croze M, Levi FA, et al, “Circadian Changes in Anticoagulant Effect of Heparin Infused at a Constant Rate,” Br Med J (Clin Res Ed), 1985, 290(6465):341-4. [PubMed 3917812]
  41. de Swart C, Nijmeyer B, Roelofs J, Sixma JJ. Kinetics of intravenously administered heparin in normal humans. Blood. 1982;60(6):1251–1258. [PubMed 7139119]
  42. Douketis JD, Mithoowani S. Prevention of venous thromboembolic disease in adult nonorthopedic surgical patients. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 27, 2019.
  43. Douketis JD, Spyropoulos AC, Spencer FA, et al. Perioperative management of antithrombotic therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines [published correction appears in Chest. 2012;141(4):1129]. Chest. 2012;141(2 suppl):e326S-e350S. doi: 10.1378/chest.11-2298. [PubMed 22315266]
  44. Estes JW. Clinical pharmacokinetics of heparin. Clin Pharmacokinet. 1980;5(3):204-220. doi:10.2165/00003088-198005030-00002 [PubMed 6993082]
  45. Falck-Ytter Y, Francis CW, Johanson NA, et al. Prevention of VTE in orthopedic surgery patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e278S-e325S. doi: 10.1378/chest.11-2404. [PubMed 22315265]
  46. Faust AC, Kanyer D, Wittkowsky AK. Managing transitions from oral factor Xa inhibitors to unfractionated heparin infusions. Am J Health Syst Pharm. 2016;73(24):2037-2041. doi:10.2146/ajhp150596 [PubMed 27919873]
  47. Ferguson JJ, Califf RM, Antman EM, et al; SYNERGY Trial Investigators. Enoxaparin vs unfractionated heparin in high-risk patients with non-ST-segment elevation acute coronary syndromes managed with an intended early invasive strategy: primary results of the SYNERGY randomized trial. JAMA. 2004;292(1):45-54. doi: 10.1001/jama.292.1.45. [PubMed 15238590]
  48. Flaker GC, Bartolozzi J, Davis V, et al, “Use of a Standardized Heparin Nomogram to Achieve Therapeutic Anticoagulation after Thrombolytic Therapy in Myocardial Infarction. TIMI 4 investigators. Thrombolysis in Myocardial Infarction,” Arch Intern Med, 1994, 154(13):1492-6. [PubMed 8018004]
  49. Freed MD, Keane JF, and Rosenthal A, “The Use of Heparinization to Prevent Arterial Thrombosis After Percutaneous Cardiac Catheterization in Children,” Circulation, 1974, 50(3):565-9. [PubMed 4607271]
  50. Frontera JA, Lewin JJ 3rd, Rabinstein AA, et al; Guideline for reversal of antithrombotics in intracranial hemorrhage: a statement for healthcare professionals from the Neurocritical Care Society and Society of Critical Care Medicine. Neurocrit Care. 2016;24(1):6-46. [PubMed 26714677]
  51. Garcia DA, Baglin TP, Weitz JI, Samama MM. Parenteral anticoagulants: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines [published corrections appear in Chest. 2012;141(5):1369; Chest. 2013;144(2):721]. Chest. 2012;141(2 suppl):e24S-e43S. doi: 10.1378/chest.11-2291. [PubMed 22315264]
  52. Gerhard-Herman MD, Gornik HL, Barrett C, et al. 2016 AHA/ACC guideline on the management of patients with lower extremity peripheral artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines [published correction appears in Circulation. 2017;135(12):e791-e792]. Circulation. 2017;135(12):e726-e779. doi: 10.1161/CIR.0000000000000471. [PubMed 27840333]
  53. Gerlach AT, Folino J, Morris BN, Murphy CV, Stawicki SP, Cook CH. Comparison of heparin dosing based on actual body weight in non-obese, obese and morbidly obese critically ill patients. Int J Crit Illn Inj Sci. 2013;3(3):195-199. doi:10.4103/2229-5151.119200 [PubMed 24404457]
  54. Giglia TM, Massicotte MP, Tweddell JS, et al. Prevention and treatment of thrombosis in pediatric and congenital heart disease: a scientific statement from the American Heart Association. Circulation. 2013;128(24):2622-2703. [PubMed 24226806]
  55. Gonzaga T, Jenabzadeh K, Anderson CP, Mohr WJ, Endorf FW, Ahrenholz DH. Use of intra-arterial thrombolytic therapy for acute treatment of frostbite in 62 patients with review of thrombolytic therapy in frostbite. J Burn Care Res. 2016;37(4):e323-e334. doi:10.1097/BCR.0000000000000245 [PubMed 25950290]
  56. Gould MK, Garcia DA, Wren SM, et al. Prevention of VTE in nonorthopedic surgical patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines [published correction appears in Chest. 2012;141(5):1369]. Chest. 2012;141(2 suppl):e227S-e277S. doi: 10.1378/chest.11-2297. [PubMed 22315263]
  57. Guyatt GH, Akl EA, Crowther M, Gutterman DD, Schuünemann HJ; American College of Chest Physicians Antithrombotic Therapy and Prevention of Thrombosis Panel. Executive summary: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines [published corrections appear in Chest. 2012;141(4):1129; Chest. 2012;142(6):1698]. Chest. 2012;141(2 suppl):7S-47S. doi: 10.1378/chest.1412S3 [PubMed 22315257]
  58. Heparin sodium in 0.45% sodium chloride [prescribing information]. Lake Forest, IL: Hospira; July 2020.
  59. Heparin sodium in 0.9% sodium chloride [prescribing information]. New York, NY: Pfizer Labs; September 2019.
  60. Heparin sodium in dextrose injection [prescribing information]. Lake Forest, IL: Hospira, Inc; August 2019.
  61. Heparin sodium in dextrose injection [prescribing information]. Lake Forest, IL: Hospira, Inc; September 2019.
  62. Heparin sodium in sodium chloride 2 units/mL [prescribing information]. Deerfield, IL: Baxter Healthcare Corporation; January 2022.
  63. Heparin sodium injection (single- and multi-dose vials) [prescribing information]. Lake Zurich, IL: Fesenius Kabi; June 2021.
  64. Heparin sodium injection (single- and multi-dose vials) [prescribing information]. Lake Zurich, IL: Fesenius Kabi; July 2021.
  65. Heparin sodium injection [prescribing information]. Rockford, IL: Mylan; September 2019.
  66. Heparin Sodium (Porcine) Lock Flush [prescribing information]. Glendale Heights, IL: Medefil Inc; December 2016.
  67. Hepponstall M, Chan A, Monagle P. Anticoagulation therapy in neonates, children and adolescents. Blood Cells Mol Dis. 2017;67:41-47. [PubMed 28552474]
  68. Hickey S, Whitson A, Jones L, et al. Guidelines for thrombolytic therapy for frostbite. J Burn Care Res. 2020;41(1):176-183. doi:10.1093/jbcr/irz148 [PubMed 31899512]
  69. Hirsh J, Fuster V. Guide to anticoagulant therapy. Part 1: heparin. American Heart Association. Circulation. 1994;89(3):1449-1468. [PubMed 8124829]
  70. Hirsh J, Raschke R, Warkentin TE, Dalen JE, Deykin D, Poller L. Heparin: mechanism of action, pharmacokinetics, dosing considerations, monitoring, efficacy, and safety. Chest. 1995;108(4)(suppl):258S-275S. doi:10.1378/chest.108.4_supplement.258s [PubMed 7555181]
  71. Holbrook A, Schulman S, Witt, DM, et al. Evidence-based management of anticoagulant therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e152S-e184S. doi: 10.1378/chest.11-2295. [PubMed 22315259]
  72. Hosch LM, Breedlove EY, Scono LE, Knoderer CA. Evaluation of an unfractionated heparin pharmacy dosing protocol for the treatment of venous thromboembolism in nonobese, obese, and severely obese patients. Ann Pharmacother. 2017;51(9):768-773. doi:10.1177/1060028017709819 [PubMed 28511582]
  73. Hull RD, Garcia DA. Heparin and LMW heparin: dosing and adverse effects. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 21, 2022a.
  74. Hull RD, Lip GYH. Venous thromboembolism: Anticoagulation after initial management. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 12, 2022b.
  75. Hull RD, Raskob GE, Rosenbloom D, et al, “Optimal Therapeutic Level of Heparin Therapy in Patients with Venous Thromboembolism,” Arch Intern Med, 1992, 152(8):1589-95. [PubMed 1497392]
  76. Ibrahim AE, Goverman J, Sarhane KA, Donofrio J, Walker TG, Fagan SP. The emerging role of tissue plasminogen activator in the management of severe frostbite. J Burn Care Res. 2015;36(2):e62-e66. doi:10.1097/BCR.0000000000000135 [PubMed 25687362]
  77. "Inactive" ingredients in pharmaceutical products: update (subject review). American Academy of Pediatrics (AAP) Committee on Drugs. Pediatrics. 1997;99(2):268-278. [PubMed 9024461]
  78. Institute for Safe Medication Practice (ISMP) and Vermont Oxford Network, “Standard Concentrations of Neonatal Drug Infusions,” 2011. Available at https://www.ismp.org/Tools/PediatricConcentrations.pdf
  79. Jaff MR, McMurtry MS, Archer SL, et al, “Management of Massive and Submassive Pulmonary Embolism, Iliofemoral Deep Vein Thrombosis, and Chronic Thromboembolic Pulmonary Hypertension: A Scientific Statement from the American Heart Association,” Circulation, 2011, 123(16):1788-830. [PubMed 21422387]
  80. January CT, Wann LS, Alpert JS, et al; ACC/AHA Task Force Members. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the Heart Rhythm Society. Circulation. 2014;130(23):e199-e267. doi: 10.1161/CIR.0000000000000041. [PubMed 24682347]
  81. January CT, Wann LS, Calkins H, et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. Heart Rhythm. 2019. pii: S1547-5271(19)30037-2. doi: 10.1016/j.hrthm.2019.01.024 [PubMed 30703530]
  82. Johnson AR, Jensen HL, Peltier G, DelaCruz E. Efficacy of intravenous tissue plasminogen activator in frostbite patients and presentation of a treatment protocol for frostbite patients. Foot Ankle Spec. 2011;4(6):344-348. doi:10.1177/1938640011422596 [PubMed 21965579]
  83. Kahn SR, Lim W, Dunn AS, et al. Prevention of VTE in nonsurgical patients: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e195S-e226S. doi: 10.1378/chest.11-2296. [PubMed 22315261]
  84. Kandrotas RJ. Heparin pharmacokinetics and pharmacodynamics. Clin Pharmacokinet. 1992;22(5):359-374. [PubMed 1505142]
  85. Kearon C, Akl EA, Comerota AJ, et al. Antithrombotic therapy for VTE disease: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines [published correction appears in Chest. 2012;142(6):1698-1704]. Chest. 2012;141(2)(suppl):e419S-e496S. doi: 10.1378/chest.11-2301. [PubMed 22315268]
  86. Kearon C, Akl EA, Ornelas J, et al. Antithrombotic Therapy for VTE disease: CHEST guideline and expert panel report. Chest. 2016;149(2):315-352. doi: 10.1016/j.chest.2015.11.026. [PubMed 26867832]
  87. Key NS, Khorana AA, Kuderer NM, et al. Venous thromboembolism prophylaxis and treatment in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol. 2020;38(5):496-520. doi:10.1200/JCO.19.01461 [PubMed 31381464]
  88. Kincaid EH, Hammon JW. Cardiopulmonary Bypass. In: Yuh DD, Vricella LA, Yang SC, Doty JR. eds. Johns Hopkins Textbook of Cardiothoracic Surgery, Second Edition. New York, NY: McGraw-Hill; 2014.
  89. Konkle BA, Nkomo VT. Antithrombotic therapy for mechanical heart valves. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 24, 2022.
  90. Konkle BA. Antithrombotic therapy for prosthetic heart valves: management of bleeding and invasive procedures. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 27, 2019b.
  91. Kovalik EC, Davenport A. Anticoagulation for the hemodialysis procedure. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 26, 2020.
  92. Kroon C, de Boer A, Kroon JM, Schoemaker HC, van den Meer FJ, Cohen AF. Influence of skinfold thickness on heparin absorption. Lancet. 1991;337(8747):945-946. doi:10.1016/0140-6736(91)91573-d [PubMed 1678033]
  93. Kulik A, Rubens FD, Wells PS, et al. Early postoperative anticoagulation after mechanical valve replacement: a systematic review. Ann Thorac Surg. 2006;81(2):770-781. doi: 10.1016/j.athoracsur.2005.07.023. [PubMed 16427905]
  94. Lawton JS, Tamis-Holland JE, Bangalore S, et al. 2021 ACC/AHA/SCAI guideline for coronary artery revascularization: a report of the American College of Cardiology/American Heart Association joint committee on clinical practice guidelines. J Am Coll Cardiol. 2022;79(2):e21-e129. doi:10.1016/j.jacc.2021.09.006 [PubMed 34895950]
  95. Lee OK, Johnston L. A systematic review for effective management of central venous catheters and catheter sites in acute care paediatric patients. Worldviews Evid Based Nurs. 2005;2(1):4-13. [PubMed 17040550]
  96. Levito MN, Coons JC, Verrico MM, et al. A systemwide approach for navigating the dilemma of oral factor Xa inhibitor interference with unfractionated heparin anti-factor Xa concentrations. Ann Pharmacother. Published online September 4, 2020. doi:10.1177/1060028020956271 [PubMed 32885997]
  97. Lincoff AM, Cutlip D. Acute ST-elevation myocardial infarction: Management of anticoagulation. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 26, 2020.
  98. Lip GYH, Hull RD. Venous thromboembolism: Initiation of anticoagulation. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 24, 2022.
  99. McDonald MM, Jacobson LJ, Hay WW Jr, Hathaway WE. Heparin clearance in the newborn. Pediatr Res. 1981;15(7):1015-1018. [PubMed 7254945]
  100. McIntosh SE, Freer L, Grissom CK, et al. Wilderness Medical Society clinical practice guidelines for the prevention and treatment of frostbite: 2019 update. Wilderness Environ Med. 2019;30(4S):S19-S32. doi:10.1016/j.wem.2019.05.002 [PubMed 31326282]
  101. Mermel LA, Allon M, Bouza E, et al. Clinical practice guidelines for the diagnosis and management of intravascular catheter-related infection: 2009 update by the Infectious Diseases Society of America [published corrections appear in Clin Infect Dis. 2010;50(7):1079; Clin Infect Dis. 2010;50(3):457]. Clin Infect Dis. 2009;49(1):1-45. doi: 10.1086/599376. [PubMed 19489710]
  102. Monagle P, Chan A, Goldenberg NA, et al, "Antithrombotic Therapy in Neonates and Children: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines (9th Edition)," Chest, 2012, 141(2 Suppl):e737-801. [PubMed 22315277]
  103. Morimoto Y, Niwa H, Minematsu K. Risk factors affecting hemorrhage after tooth extraction in patients undergoing continuous infusion with unfractionated heparin. J Oral Maxillofac Surg. 2012;70(3):521-526. [PubMed 22177816]
  104. Nemecek BD, Hammond DA, eds. Demystifying Drug Dosing in Renal Dysfunction. American Society of Health-System Pharmacists; 2019.
  105. Newall F, Johnston L, Ignjatovic V, et al, "Unfractionated Heparin Therapy in Infants and Children," Pediatrics, 2009, 123(3):e510-8. [PubMed 19221154]
  106. Nygaard RM, Lacey AM, Lemere A, et al. Time matters in severe frostbite: assessment of limb/digit salvage on the individual patient level. J Burn Care Res. 2017;38(1):53-59. doi:10.1097/BCR.0000000000000426 [PubMed 27606554]
  107. O'Gara PT, Kushner FG, Ascheim DD, et al. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;61(4):e78-e140. doi: 10.1016/j.jacc.2012.11.019. [PubMed 23256914]
  108. Olsson P, Lagergren H, Ek S. The elimination from plasma of intravenous heparin: An experimental study on dogs and humans. Acta Med Scand. 1963;173:619–630. [PubMed 13940090]
  109. Ortel TL, Neumann I, Ageno W, et al. American Society of Hematology 2020 guidelines for management of venous thromboembolism: treatment of deep vein thrombosis and pulmonary embolism. Blood Adv. 2020;4(19):4693-4738. doi:10.1182/bloodadvances.2020001830 [PubMed 33007077]
  110. Osataphan S, Patell R, Chiasakul T, Khorana AA, Zwicker JI. Extended thromboprophylaxis for medically ill patients with cancer: a systemic review and meta-analysis. Blood Adv. 2021;5(8):2055-2062. doi:10.1182/bloodadvances.2020004118 [PubMed 33861298]
  111. Otto CM, Nishimura RA, Bonow RO, et al. 2020 ACC/AHA guideline for the management of patients with valvular heart disease: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. 2021;143(5):e72-e227. doi:10.1161/CIR.0000000000000923 [PubMed 33332150]
  112. Pai M, Douketis JD. Prevention of venous thromboembolic disease in acutely ill hospitalized medical adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 27, 2019a.
  113. Pai M, Douketis JD. Prevention of venous thromboembolism in adult orthopedic surgical patients. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 7, 2020.
  114. Paine RE, Turner EN, Kloda D, Falank C, Chung B, Carter DW. Protocoled thrombolytic therapy for frostbite improves phalangeal salvage rates. Burns Trauma. 2020;8:tkaa008. doi:10.1093/burnst/tkaa008 [PubMed 32341921]
  115. Pasquale LD, Ferneini EM. Heparin and Lovenox: What the Oral and Maxillofacial Surgeon Needs to Know. Oral Maxillofac Surg Clin North Am. 2016;28(4):507-513. [PubMed 27624773]
  116. Patanwala AE, Seaman SM, Kopp BJ, Erstad BL. Heparin dosing for venous thromboembolism prophylaxis in obese hospitalized patients: an observational study. Thromb Res. 2018;169:152-156. doi:10.1016/j.thromres.2018.07.027 [PubMed 30071480]
  117. Pearl G, Gilani R. Acute mesenteric arterial occlusion. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed August 5, 2020.
  118. Raschke RA, Reilly BM, Guidry JR, et al, “The Weight-Based Heparin Dosing Nomogram Compared With a “Standard Care” Nomogram: A Randomized Controlled Trial,” Ann Intern Med, 1993, 119(9):874-81. [PubMed 8214998]
  119. Regitz-Zagrosek V, Roos-Hesselink JW, Bauersachs J, et al; ESC Scientific Document Group. 2018 ESC guidelines for the management of cardiovascular diseases during pregnancy. Eur Heart J. 2018;39(34):3165-3241. doi: 10.1093/eurheartj/ehy340. [PubMed 30165544]
  120. Riney JN, Hollands JM, Smith JR, Deal EN. Identifying optimal initial infusion rates for unfractionated heparin in morbidly obese patients. Ann Pharmacother. 2010;44(7-8):1141-1151. doi:10.1345/aph.1P088 [PubMed 20587743]
  121. Samuel S, Iluonakhamhe EK, Adair E, et al. High dose subcutaneous unfractionated heparin for prevention of venous thromboembolism in overweight neurocritical care patients. J Thromb Thrombolysis. 2015;40(3):302-307. doi:10.1007/s11239-015-1202-x [PubMed 25736986]
  122. Sharma A, Chatterjee S, Lichstein E, Mukherjee D. Extended thromboprophylaxis for medically ill patients with decreased mobility: does it improve outcomes? J Thromb Haemost. 2012;10(10):2053-2060. doi: 10.1111/j.1538-7836.2012.04874.x. [PubMed 22863355]
  123. Shen JI, Winkelmayer WC. Use and safety of unfractionated heparin for anticoagulation during maintenance hemodialysis. Am J Kidney Dis. 2012;60(3):473-486. doi:10.1053/j.ajkd.2012.03.017 [PubMed 22560830]
  124. Shin S, Harthan EF. Safety and efficacy of the use of institutional unfractionated heparin protocols for therapeutic anticoagulation in obese patients: a retrospective chart review. Blood Coagul Fibrinolysis. 2015;26(6):655-660. doi:10.1097/MBC.0000000000000336 [PubMed 26236941]
  125. Shlensky JA, Thurber KM, O'Meara JG, et al. Unfractionated heparin infusion for treatment of venous thromboembolism based on actual body weight without dose capping. Vasc Med. 2020;25(1):47-54. doi:10.1177/1358863X19875813 [PubMed 31623539]
  126. Smythe MA, Nutescu EA, and Wittkowsky AK, "Changes in the USP Heparin Monograph and Implications for Clinicians," Pharmacotherapy, 2010, 30(5):428-31. [PubMed 20411994]
  127. Steg PG, Jolly SS, Mehta SR, et al; FUTURA/OASIS-8 Trial Group. Low-dose vs standard-dose unfractionated heparin for percutaneous coronary intervention in acute coronary syndromes treated with fondaparinux: the FUTURA/OASIS-8 randomized trial. JAMA. 2010;304(12):1339-1349. doi: 10.1001/jama.2010.1320. [PubMed 20805623]
  128. Steg PG, van 't Hof A, Hamm CW, et al; EUROMAX Investigators. Bivalirudin started during emergency transport for primary PCI. N Engl J Med. 2013;369(23):2207-2217. doi: 10.1056/NEJMoa1311096. [PubMed 24171490]
  129. Stone GW, McLaurin BT, Cox DA, et al; ACUITY Investigators. Bivalirudin for patients with acute coronary syndromes. N Engl J Med. 2006;355(21):2203-2216. doi: 10.1056/NEJMoa062437. [PubMed 17124018]
  130. Streiff MB, Holmstrom B, Ashrani A, et al. Cancer-associated venous thromboembolic disease, version 1.2015. J Natl Compr Canc Netw. 2015;13(9):1079-1095. doi:10.6004/jnccn.2015.0133 [PubMed 26358792]
  131. Tapson VF, Weinberg AS. Thrombolytic (fibrinolytic) therapy in acute pulmonary embolism and lower extremity deep vein thrombosis. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 27, 2019.
  132. Topjian AA, Raymond TT, Atkins D, et al. Part 4: Pediatric basic and advanced life support: 2020 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Circulation. 2020;142(16 Suppl 2):S469-S523. doi:10.1161/CIR.0000000000000901 [PubMed 33081526]
  133. Tsujimoto H, Tsujimoto Y, Nakata Y, et al. Pharmacological interventions for preventing clotting of extracorporeal circuits during continuous renal replacement therapy. Cochrane Database Syst Rev. Published online March 13, 2020. doi:10.1002/14651858.CD012467.pub2 [PubMed 32164041]
  134. Vandiver JW, Vondracek TG. Antifactor Xa levels versus activated partial thromboplastin time for monitoring unfractionated heparin. Pharmacotherapy. 2012;32(6):546-558. doi: 10.1002/j.1875-9114.2011.01049.x. [PubMed 22531940]
  135. Wada H, Thachil J, Di Nisio M, Mathew P, et al; The Scientific Standardization Committee on DIC of the International Society on Thrombosis Haemostasis. Guidance for diagnosis and treatment of DIC from harmonization of the recommendations from three guidelines [published online February 4, 2013]. J Thromb Haemost. doi: 10.1111/jth.12155. [PubMed 23379279]
  136. Wallentin L, Goldstein P, Armstrong PW, et al. Efficacy and safety of tenecteplase in combination with the low-molecular-weight heparin enoxaparin or unfractionated heparin in the prehospital setting: the Assessment of the Safety and Efficacy of a New Thrombolytic Regimen (ASSENT)-3 PLUS randomized trial in acute myocardial infarction. Circulation. 2003;108(2):135-142. doi: 10.1161/01.CIR.0000081659.72985.A8. [PubMed 12847070]
  137. Warkentin TE and Kelton JG, “Temporal Aspects of Heparin-Induced Thrombocytopenia,” N Engl J Med, 2001, 344(17):1286-92. [PubMed 11320387]
  138. Whitlock RP, Sun JC, Fremes SE, Rubens FD, Teoh KH. Antithrombotic and thrombolytic therapy for valvular disease: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e576S-e600S. doi: 10.1378/chest.11-2305. [PubMed 22315272]
  139. Yamaji H, Murakami T, Hina K, et al. Differences in activated clotting time and initial heparin dosage during atrial fibrillation ablation for patients with edoxaban compared with warfarin. J Cardiovasc Electrophysiol. 2018;29(6):835-843. doi: 10.1111/jce.13483. [PubMed 29533476]
  140. Yevzlin AS, Sanchez RJ, Hiatt JG, et al. Concentrated heparin lock is associated with major bleeding complications after tunneled hemodialysis catheter placement. Semin Dial. 2007;20(4):351-354. doi: 10.1111/j.1525-139X.2007.00294.x. [PubMed 17635828]
  141. You JJ, Singer DE, Howard PA, et al; American College of Chest Physicians. Antithrombotic therapy for atrial fibrillation: Antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 suppl):e531S-e575S. doi: 10.1378/chest.11-2304. [PubMed 22315271]
  142. Zafren K, Mechem CC. Frostbite: Emergency care and prevention. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed July 1, 2022.
Topic 13349 Version 525.0