Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 3

Glycoprotein IIb/IIIa Inhibitors.

Consequently, platelet transfusion would not be able to absorb excess drug and would not be
GPIs inhibit GP IIb/IIIa receptors on platelets, blocking the binding of fibrinogen to activated helpful in the management of bleeding with eptifibatide or tirofiban.
GP IIb/IIIa receptors, which is the final step in platelet aggregation.
- Platelet transfusion is a lifesaving procedure that is carried out to prevent bleeding
These agents must be given with unfractionated heparin (UFH) or a low-molecular-weight or stop ongoing bleeding in patients with low platelet count or functional platelet
heparin (LMWH) because coadministration of heparin provides improved protection against disorders
persistent platelet activation, thereby improving outcome after percutaneous coronary
intervention however it should be discontinued immediately after PCI to reduce the risk of In general:
major bleeding. • GPIs are not beneficial and should not be used in patients with NSTE-ACS undergoing an
ischemia-driven approach. GPIs should also be avoided in patients with STEMI receiving
Note that these agents are only available as an IV infusion. reperfusion with fibrinolytics because of significant increases in major bleeding and ICH.
• Besides bleeding, GPIs cause significant thrombocytopenia in about 1% of patients
• Use of GPIs has been declining in recent years; patients likely to benefit most are those receiving abciximab and 0.5% with eptifibatide and tirofiban.
receiving PCI for NSTE-ACS (Non-ST elevation-acute coronary syndrome) with elevated
troponin levels and patients with STEMI who have not been preloaded with a P2Y12 inhibitor GPIs are given with heparin because coadministration of heparin provides improved
and are not being treated with bivalirudin. protection against persistent platelet activation, thereby improving outcome after
percutaneous coronary intervention, however it is important to differentiate GPI-induced
Recommended doses are as follows: thrombocytopenia from heparin-induced thrombocytopenia (HIT). Thrombocytopenia from a
GP IIb/IIIa inhibitor occurs more rapidly (within hours) and the platelet count nadir is typically
✓ Abciximab: 0.25 mg/kg IV bolus given 10–60 minutes before the start of PCI, followed by lower (about 20,000) compared to HIT.
0.125 mcg/kg/min (maximum 10 mcg/min) for 12 hours; alternatively, 0.25 mg/kg
intracoronary bolus only. Anticoagulants
• Although patients with ACS are typically on at least two antiplatelet agents for a year or
Abciximab is a chimeric human-murine monoclonal antibody Fab fragment that blocks the GP more, a single anticoagulant is usually given for a short time (the initial few days of
IIb/IIIa receptor–fibrinogen binding. It irreversibly binds to the GP IIb/IIIa receptor, with hospitalization). UFH can be used across the spectrum of ACS and regardless of the
platelet function recovery occurring over approximately 24 hours after discontinuation of the management strategy.
infusion.
Unfractionated heparin provides its anticoagulant activity by binding to the endogenous
Due to the strong binding of abciximab to the GP IIb/IIIa receptor, the drug to receptor ration anticoagulant antithrombin (AT) via a unique pentasaccharide sequence, increasing its
is approximately 2:1. Therefore, platelet transfusions can absorb the excess abciximab, and affinity for clotting factor inhibition by 1000-fold.
would be effective in management of a major bleeding episode.
These UFH–AT complexes can then inhibit clotting factors IXa, Xa, XIa, XIIa, and thrombin,
✓ Eptifibatide: 180 mcg/kg IV bolus, repeated in 10 minutes, followed by IV infu- sion of 2 with most of the impact provided through inhibition of factor Xa and thrombin. Inhibition of
mcg/kg/min for 18–24 hours after PCI; reduce infusion dose by 50% if creatinine clearance thrombin requires a tertiary binding between the UFH–AT and thrombin molecules. This
(CrCl) is <50 mL/min (0.83 mL/sec). requires that the chain length of the inhibitory molecule be at least 18 saccharides long. Since
✓ Tirofiban: 25 mcg/kg IV bolus, then 0.15 mcg/kg/min for up to 18–24 hours after PCI; most UFH molecules are approximately 45-50 saccharides long, UFHs can inhibit factor Xa
reduce infusion dose by 50% if CrCl is ≤60 mL/min (1.0 mL/sec). and thrombin in an equal 1:1 ratio.

Eptifibatide and tirofiban are commonly referred to as “small molecule” as they are peptide Currently, the recommended dose of UFH is an IV bolus of 60 units/kg (initial maximum total
(Eptifibatide) and nonpeptide (tirofiban) inhibitors of the GP IIb/IIIa receptor. dose of 4000 units) and an initial infusion rate of 12 units/kg/hr (initial maximum 1000
units/hr). This is the recommended dose regardless of the ACS diagnosis or management
And in comparison to abciximab, they have a much smaller molecular weight. In addition, strategy.
these agents have reversibly binds of the GP IIb/IIIa receptor. Therefore, platelet function
recovery occurs in 2 to 4 hours after discontinuation of the infusion. The reversible binding of ***The anticoagulant effect of UFH has significant interpatient variability. This is due to
the small molecule agents requires that they overwhelm the ability of fibrinogen to bind to additional binding of UFH with endothelial cells, plasma protein, and ingestion by
the GP IIb/IIIa receptor with high concentrations and a drug to receptor ratio that is macrophages.
approximately 500 to 1000:1.
Because of significant interpatient variability in anticoagulant response, therapy must be
monitored with the activated partial thromboplastin time (aPTT) every 6 hours until two
consecutive readings are within the institution’s therapeutic range (1.5–2 times the control
value), then every 24 hours for the duration of UFH therapy. The activated clotting time (ACT) CONTRAINDICATION: Active bleeding, history of heparin-induced thrombocytopenia, severe
is monitored during PCI because it can be measured at the bedside with rapid results. Platelet bleeding risk, recent stroke
counts should also be monitored daily or every other day to detect HIT. If HIT is suspected,
discontinue UFH and provide anticoagulation with an IV direct thrombin inhibitor. ADE: Bleeding, heparin-induced thrombocytopenia

CONTRAINDICATION: Active bleeding, history of heparin-induced thrombocytopenia, severe Fondaparinux the only available pentasaccharide worldwide and is a synthetic molecule
bleeding risk, recent stroke existing of only the five saccharides. Again, it needs to bind to and potentiate the activity of
AT. And, due to the small size of the molecule, once it binds to AT it can only inhibit factor Xa
ADE: Bleeding, heparin-induced thrombocytopenia and has no activity against thrombin.

Low-Molecular-Weight Heparins Provides a predicable anticoagulant dose response with no need for therapeutic monitoring
Similar to UFHs, LMWHs must first bind to AT to provide their anticoagulant activity. LMWHs (similar to LMWH).
are created through chemical or enzymatic depolymerization of UFH molecules and it results
to a mixture of lower molecular weight fragments compared to the larger intact UFH Based on the lack of antibody cross-reactivity, it is reasonable to consider use of
molecule. LMWHs are heterogeneous compounds about one-third the size of unfractionated fondaparinux in patients with a history of HIT. Based on the long half- life of fondaparinux,
heparin as they are only less than 18 saccharides long. the SC dose (2.5 mg) is only given once daily. Fondaparinux is contraindicated in patients with
a CrCl of less than 30 mL/min (0.5 mL/s) due to the significant degree of renal elimination
LMWH’s anticoagulant activity results mostly from inhibition of factor Xa with only the few
larger fragments able to inhibit thrombin. The ratio of factor Xa to thrombin inhibition with a A clinical trial in NSTE-ACS patients receiving either an ischemia-driven or invasive
LMWH is typically 3:1 or 4:1, depending on the process of depolymerization. management strategy demonstrated similar efficacy between fondaparinux and enoxaparin,
with significantly less major bleeding in patients receiving fondaparinux. Fondaparinux can be
Compared to UFHs, LMWHs provide a predicable anticoagulant dose response with no need considered in patients undergoing an ischemia-driven approach who are at high risk of
for routine therapeutic monitoring. bleeding.

Anti-Xa monitoring may be helpful would be pediatrics, pregnancy, obesity (greater than 190 ***Fondaparinux is not recommended in patients receiving primary PCI for STEMI because of
kg), and patients with severe renal insufficiency (eg, creatinine clearance [CrCl] less than 30 clinical trial results demonstrating higher rates of catheter-related thrombosis compared to
mL/min [0.5 mL/s]). While pediatric and pregnant patients rarely have ACS, obesity and UFH. In patients with STEMI receiving fibrinolytics, fondaparinux had efficacy and safety
severe renal insufficiency are more common in patients with ACS. Since patients with ACS similar to UFH. However, fondaparinux is rarely used in patients with STEMI based on the lack
typically receive anticoagulant therapy for only a few days, the utility of anti-Xa monitoring in of superiority to UFH and the benefit shown with enoxaparin over UFH in this population.
these patients is limited.
CONTRAINDICATION: Active bleeding, history of heparin-induced thrombocytopenia, severe
The incidence of HIT is lower with LMWHs (<2%) than with UFH (2%–5%), monitoring of bleeding risk, recent stroke
platelet counts is still warranted. Due to 90% cross reactivity between HIT antibodies from
LMWH and UFH, LMWH is not a safe alternative in patients who develop HIT from UFH and ADE: Bleeding
vice versa.
Fibrin-bound thrombin is still enzymatically active, but the large AT–anticoagulant complexes
Enoxaparin is the most widely studied agent in ACS and is the only LMWH recommended in are unable to gain access and, therefore, exert anticoagulant activity.
current guidelines. Data support its efficacy in patients with STEMI and NSTE-ACS, regardless
of the perfusion or management strategy used. However, enoxaparin dosing varies across Bivalirudin is a direct thrombin inhibitor means bivalirudin does not have to first bind to AT to
these different situations. In patients with STEMI receiving reperfusion with fibrinolytics, provide its anticoagulant effect and because of the lack of AT binding, bivalirudin can inhibit
enoxaparin demonstrated a significant 17% reduction in death and MI compared to UFH. not only free or soluble thrombin, similar to UFH and LMWH, but also fibrin-bound thrombin.
Based on clinical trial data, either UFH or enoxaparin is recommended in patients with NSTE-
ACS. It is used only in patients with ACS who receive PCI and can be monitored with the ACT in the
catheterization laboratory. Although current guidelines recommend bivalirudin use, UFH is
In patients with NSTE-ACS undergoing an ischemia-driven approach, use often used instead because recent trials suggested that bivalirudin does not offer efficacy or
of subcutaneous (SC) enoxaparin at 1 mg/kg every 12 hours for up to 3 days is supported by safety benefits over UFH alone in the setting of primary PCI for STEMI.
two clinical trials that demonstrated a significant reduction in MACE compared to IV UFH
without increasing major bleeding.
Bivalirudin has not been evaluated in patients with STEMI receiving reperfusion with
fibrinolytics or in patients with NSTE-ACS undergoing an ischemia-driven approach.

CONTRAINDICATION: Active bleeding, history of heparin-induced thrombocytopenia, severe


bleeding risk, recent stroke

ADE: Bleeding

SECONDARY PREVENTION OF ISCHEMIC EVENTS


For most patients, the initial 24 hours of ACS care are focused on reperfusion (if appropriate),
antithrombotic therapy, and acute supportive measures. After a diagnosis of ACS, patients
are considered to have atherosclerotic cardiovascular disease (ASCVD) and should be treated
aggressively because they are at the highest risk of recurrent MACE. Again remember that
Major adverse cardiac events (MACE) of acute coronary syndrome (ACS) often occur
suddenly resulting in high mortality and morbidity.

Secondary prevention strategies proved to accomplish these goals typically include anti-
ischemic, antiplatelet, lipid- lowering, and antihypertensive therapies (Table 33-8). Specific
pharmacotherapy proved to decrease mortality, HF, reinfarction or stroke, and stent
thrombosis should be initiated prior to hospital discharge in all patients without
contraindications. Medication reconciliation at discharge should include assessment for
DAPT, β-blocker, angiotensin-converting enzyme (ACE) inhibitor or angiotensin receptor
blocker (ARB), and statin therapy unless a contraindication exists (Fig. 33-2). In addition,
short-acting NTG should be prescribed as needed for any subsequent episode of acute angina
for patients not taking phosphodiesterase-5 inhibitors. Select patients may also meet criteria
for benefit from an aldosterone antagonist.

In addition to evaluating patients for the use of medications proved to reduce the risk of
MACE or recurrent symptoms of angina, additional interventions may be appropriate to
optimize outcomes and improve safety. Aggressive risk factor modification strategies such as
increased physical activity, dietary modification, weight loss, blood pressure modification,
and smoking cessation should be communicated to all patients, initiated, and continued
indefinitely.

Proton pump inhibitors provide a protective benefit in patients at highest risk for GI bleeding
from DAPT and may be considered for select patients (eg, history of GI bleeding, triple
therapy with DAPT, and an oral anticoagulant with or without a history of GI bleeding).

All patients should refrain from chronic use of nonsteroidal anti-inflammatory drugs with
high degree of cyclo-oxygenase-2 selectivity such as

as they are associated with increased cerebrovascular and cardiovascular events.

You might also like