Open access peer-reviewed chapter

Non-ST Elevation Myocardial Infarction: Diagnosis and Management

Written By

Yaser Al Ahmad and Mohammed T. Ali

Submitted: 25 July 2017 Reviewed: 06 March 2018 Published: 03 January 2019

DOI: 10.5772/intechopen.76241

From the Edited Volume

Myocardial Infarction

Edited by Burak Pamukçu

Chapter metrics overview

3,143 Chapter Downloads

View Full Metrics

Abstract

Cardiovascular disease is expected to be the main cause of death globally due to the rapidly increasing prevalence of obesity, hypertension and diabetes mellitus. Atherosclerotic lesions and plaque rupture are the most common cause of myocardial infarction. Resting 12-lead ECG is the first diagnostic test for patients with chest pain and should be performed and interpreted within the first 10 min of the patient’s admission to the emergency department. Cardiac biomarkers preferably, high-sensitivity cardiac troponin, is mandatory in all patients with suspected NSTEMI for the diagnosis, risk stratification and treatment. Rapid, efficient diagnosis and risk stratification of patients with chest pain will help to administer the appropriate medication and plan for the timing of invasive strategy and the choice of revascularization. This chapter helps to simply but elaborately discuss the diagnosis, risk stratification and the management of patients with non-ST elevation of myocardial infarction.

Keywords

  • myocardial infarction
  • percutaneous intervention
  • antiplatelet

1. Introduction

1.1. Definition of acute coronary syndrome

Acute coronary syndrome (ACS) is a term that describes an acute ischemic insult to the myocardium resulting from sudden reduction in coronary blood flow. The findings on the ECG will help to categorize patients into two major subdivision of major diagnostic and therapeutic consequences [1]:

  1. Patients with acute chest pain and persistent >1 mm ST-segment elevation in ≥2 anatomically contiguous leads. This condition is termed ST-elevation ACS and generally reflects an acute total coronary occlusion. The mainstay of treatment in these patients is immediate reperfusion with primary angioplasty or fibrinolytic therapy [2]. While biomarkers are useful for confirmatory and prognostic purposes, they are not required for the diagnosis of STEMI and should not delay treatment.

  2. Patients with acute chest pain but no persistent ST-segment elevation. This condition is termed non-ST elevation ACS (NSTE-ACS). The ECG may be normal or there may be transient ST-segment elevation, persistent or transient ST-segment depression, T-wave inversion, flat T waves or pseudo-normalization of T waves. The NSTE-ACS can be sub-classified to:

    1. Non-ST elevation myocardial infarction (NSTEMI) which denote cardiomyocyte necrosis and death by a rise in serum troponin levels.

    2. Unstable angina is defined as myocardial ischemia at rest or minimal exertion in the absence of cardiomyocyte necrosis (cardiac biomarkers are not increased) [3, 4].

The application of high-sensitivity cardiac troponin measurements in daily clinical practice instead standard troponin assays showed increased detection of MI (4% absolute and 20% relative increase) and decreased diagnosis of unstable angina [5, 6, 7]. In comparison with NSTEMI patients, unstable angina patients do not have necrosis in their myocardial tissue and have a substantially lower risk of death. Unstable angina patients benefit less from intensified antiplatelet therapy and early invasive strategy [5, 6, 7, 8, 9, 10, 11, 12]. NSTEMI encompasses a broad spectrum of ischemic injury to the myocardium, which is detected by elevation of serum cardiac biomarkers. It can be distinguished from unstable angina pectoris by normal serial cardiac biomarkers [1].

1.2. Non-ST elevation myocardial infarction

NSTEMI is an acute ischemic event causing cardiomyocyte death by necrosis in a clinical setting consistent with acute myocardial ischemia [8]. The leading symptom that initiates the diagnostic and therapeutic cascade in patients with suspected ACS is chest pain but to make a diagnosis of NTEMI, one major criteria is typical rise and gradual fall in cardiac biomarkers (troponin or CKMB) in addition to one or more of the following:

  1. Symptoms of ischemia.

  2. ECG changes.

  3. Imaging evidence of new or presumed new loss of viable myocardium or regional wall motion abnormality.

  4. Intracoronary thrombus detected on angiography or autopsy [8].

1.3. Classification of myocardial infarction

The development of myocardial tissue-specific biomarkers and sensitive cardiac imaging techniques allows for early detection of very small amounts of myocardial injury or necrosis. Consequently MI has been redefined to encompass any necrosis in the setting of myocardial ischemia by any of the following possible etiologies [3, 8, 13]:

  1. Type 1 MI: spontaneous MI caused by atherosclerotic plaque rupture, ulceration, fissure, erosion or dissection with resulting intraluminal thrombus in one or more coronary arteries leading to decreased myocardial blood flow and/or distal embolization and subsequent myocardial necrosis. The patient may have underlying severe CAD but in 5–20% of cases there may be non-obstructive coronary atherosclerosis or no angiographic evidence of CAD, particularly in women [8, 10, 11, 14].

  2. Type 2 MI: MI secondary to an increase in oxygen demand or decrease in supply. The myocardial necrosis results from causes other than coronary plaque instability [8]. Mechanisms include coronary artery spasm, coronary endothelial dysfunction, tachyarrhythmias, bradyarrhythmias, anemia, respiratory failure, hypotension and severe hypertension. In addition, in critically ill patients and in patients undergoing major non-cardiac surgery, myocardial necrosis may be related to injurious effects of pharmacological agents and toxins [9].

  3. Type 3 MI: Sudden unexpected cardiac death before cardiac biomarkers obtained.

  4. Type 4a MI: MI associated with percutaneous coronary intervention (PCI) where there is a greater than 5-fold rise in troponin during the first 48 h following the intervention [8].

  5. Type 4b MI: MI associated with stent thrombosis.

  6. Type 5 MI: MI associated with coronary bypass graft surgery (CABG), a greater than 10-fold rise from normal baseline levels in troponin during the first 48 h following the intervention [8].

Advertisement

2. Epidemiology and pathogenesis

2.1. Epidemiology

Cardiovascular disease (CVD) is the number one cause of death worldwide, accounting for 17.5 million deaths per year. Coronary heart disease mortality is decreasing in many developed countries, but it is increasing in developing and transitional countries, partly as a result of increasing longevity, urbanization, and lifestyle changes. Epidemiological data have shown that acute coronary syndrome cases with STEMI appear to be declining and that NSTEMI occurs more frequently than STEMI [15, 16]. In the United States, it is estimated that >780,000 people will experience an ACS each year, and approximately 70% of these will have NSTEMI [17]. Trends from the world’s largest database of patients with ACS show that the percentage of patients with a diagnosis of NSTEMI is rising dramatically [18]. This is likely to be due to the advent of more sensitive assays for myocardial injury, earlier pharmacotherapy, and reperfusion (and prevention) of STEMI [13, 18].

2.2. Pathophysiology

NSTEMI is a result of an acute imbalance between myocardial oxygen demand and supply, most commonly due to a reduction in myocardial perfusion. Type 1 MI is most commonly caused by a non-occlusive thrombus that develops in a disrupted atherosclerotic plaque, and leads to non-occlusive or near-complete thrombosis of a vessel supplying the myocardium.

Plaque rupture usually occurs at the weakest and thinnest part of the atherosclerotic cap (often at the shoulder region). Ruptured plaques contain large numbers of inflammatory cells including monocytes, macrophages, and T lymphocytes [19, 20]. Although one third of occlusions occur at a site with the greatest stenosis, most (66–78%) arise from lesions with <50% stenosis, and <5% arise from lesions exhibiting >70% stenosis [19]. It is thought that the lack of ST elevation is because the infarct does not involve the full thickness of the myocardium (not a transmural infarction). The severity of myocardial damage in NSTEMI depends on:

  • Duration of ischemia and time to reperfusion

  • Extent of underlying atherosclerosis

  • Presence of collateral blood flow to the affected region

  • Diameter of affected coronary vessel

  • Degree of occlusion

  • Presence of other comorbidities (i.e., diabetes, renal failure, or HTN).

Classically it is thought that NSTEMI patients ultimately have a diagnosis of a non-Q-wave MI; however, 25% of patients with NSTEMI and elevated biomarkers go on to develop Q-wave MI in the weeks to follow [21]. In addition, approximately 25% of patients with a diagnosis of NSTEMI have a 100% occlusion of the affected artery on coronary angiography [22].

NSTEMI may also be caused by other mechanisms, such as dynamic obstruction (i.e., focal coronary artery spasm or Prinzmetal angina), severe progressive atherosclerosis, restenosis following percutaneous coronary intervention, recreational drug use (e.g., cocaine or other stimulants), arterial inflammation (i.e., vasculitis), or extrinsic causes leading to myocardial supply–demand mismatch (such as hypotension, hypovolemia, or hypoxia) [1].

Advertisement

3. Diagnostic approach

3.1. Clinical presentation

Patients presenting with chest pain or discomfort with suspected ACS require urgent evaluation. The clinical spectrum of NSTEMI may range from patients free of symptoms at presentation to individuals with ongoing ischemia, electrical or hemodynamic instability due to large myocardium in jeopardy or cardiac arrest secondary to malignant ventricular ischemia. Therefore, it is essential to establish if the patient has ACS and if so, what is the likelihood the patient will have adverse clinical event [1]. Physicians will need to stratify the patients according to their risk status and according to the initial risk assessment to choose an appropriate management strategy. The initial risk assessment includes the history, examination, ECG, and cardiac biomarkers [1, 23].

3.2. History and examination

Angina pectoris is a kind of pain described as a sensation of tightness, heaviness, aching, burning, pressure, or squeezing typically localized at the retrosternal region. The pain can often radiate to the left arm but may also radiate to the lower jaw, neck, both arms, back, and epigastrium. It is associated with exertion or emotional stress and relieved by rest or administration of sublingual nitroglycerin [1].

In ACS patients other symptoms including sweating, nausea, abdominal pain, dyspnea and syncope may be present. Atypical presentations are also possible and characterized by epigastric pain, indigestion-like symptoms and isolated dyspnea. Atypical complaints are more often observed in the elderly, in women and in patients with diabetes mellitus, chronic renal disease or dementia [24, 25]. The relief of pain at rest increase the probability of myocardial ischemia while the relief of symptoms after nitrates administration is not specific for angina pectoris [25]. In patients presenting with suspected MI to the emergency department, overall, the diagnostic performance of chest pain characteristics for MI is limited [25].

Risk factors increase the likelihood of NSTEMI include: Older age, male gender, family history of CAD, diabetes, hyperlipidemia, hypertension, renal insufficiency, previous manifestation of CAD as well as peripheral or carotid artery disease.

Physical examination is frequently unremarkable in patients with suspected NSTEMI but may reveal HTN or hypotension, the presence of third and fourth heart sounds, and paradoxical splitting of the second heart sound. Cardiac auscultation may reveal a systolic murmur due to ischemic mitral regurgitation, which is associated with poor prognosis [26] or a mechanical complication (i.e. papillary muscle rupture or ventricular septal defect) of a subacute and possibly undetected MI. Signs of heart failure (raised jugular venous pressure, bilateral crepitation on auscultation of the lungs) or cardiogenic shock may also be present, and these signify a worse prognosis.

3.3. Initial tests

3.3.1. Electrocardiogram

Resting 12-lead ECG is the first diagnostic test for patients with chest pain and should be performed and interpreted within the first 10 min of the initial admission to the hospital [27]. ECG is critical for the diagnosis of STEMI as the cause for the chest pain, this has a tremendous therapeutic implication for the patient.

While the ECG in the setting of NSTEMI may be normal in more than one-third of patients, a serial ECG at 15- to 30-min intervals should be performed to detect the developing abnormalities. Classic ECG findings of ischemia in NSTEMI include horizontal or down sloping ST depression >0.5 mm and/or symmetrically inverted T waves >2.0 mm (Figure 1) [2, 28]. Standard leads may be inconclusive in some patients and additional leads may be necessary (e.g. in case of left circumflex artery occlusion or right ventricular MI may be detected only in V7–V9 and V3R and V4R, respectively) [8]. If it is possible a comparison with previous ECG’s may be valuable. Diffuse precordial ST depression more pronounced in leads V4–V6 may indicate a culprit lesion located in the mid left anterior descending coronary artery, while changes more evident in leads V2–V3 may be more suggestive of a culprit lesion located in the left circumflex artery [29]. Diffuse ST depression including both precordial and extremity leads associated with ST-elevation ≥1 mm in lead aVR may indicate either left main coronary artery as the culprit lesion or proximal occlusion of the left anterior descending coronary artery in the presence of severe three-vessel CAD [30, 31].

Figure 1.

ECG showing ST depression in the inferolateral leads suggestive of inferior-lateral ischemia.

3.3.2. Blood tests

  • CBC: hemoglobin and hematocrit measurements may help to evaluate a secondary cause of NSTEMI (e.g., acute blood loss, anemia) and to evaluate thrombocytopenia to estimate risk of bleeding.

  • BUN and serum creatinine: creatinine clearance should be estimated in NSTEMI patients and the doses of renally cleared drugs should be adjusted appropriately. In chronic kidney disease patients undergoing angiography, iso-osmolar contrast agents may be preferred [1, 15].

  • Serum electrolytes: electrolyte derangements may predispose to cardiac arrhythmias.

  • Liver function tests: useful if treatment with drugs that undergo hepatic metabolism is considered.

  • Brain natriuretic peptide (BNP) and N-terminal pro-BNP (NT-pro-BNP): measurement of BNP or NT-pro-BNP may be considered to supplement assessment of global risk in patients with suspected ACS, particularly cardiogenic shock associated with MI type 1 [1].

  • Lipid profile: this test is indicated in the first 24 h of admission to the hospital to assess for lipid abnormalities and therefore the need for any lipid-lowering therapy.

3.3.3. Biomarkers

Clinical assessment,12-lead ECG and biomarkers are crucial for the diagnosis, risk stratification and treatment of patients with suspected NSTEMI. Measurement, preferably, high-sensitivity cardiac troponin, is mandatory in all patients with suspected NSTEMI [7, 8, 9]. Cardiac troponins are more sensitive and specific markers of cardiomyocyte injury than creatine kinase (CK), its MB isoenzyme (CK-MB) and myoglobin. In patients with suspected myocardial ischemia, a dynamic elevation of cardiac troponin above the 99th percentile of healthy individuals indicates MI. Cardiac troponin levels rise rapidly (i.e. usually within 1 h if using high-sensitivity assays) after symptom onset and remain elevated for several days [8, 9].

The use of high-sensitivity assays, has shortened the time interval to the second cardiac troponin, reduced substantially the delay to diagnosis, translating into shorter stays in the emergency department and lower costs [6, 7, 8, 9, 32, 33, 34, 35]. In patients presenting very early, the second cardiac troponin level should be obtained at 3 h, due to the time dependency of troponin release; serial cardiac troponin testing should be pursued if the clinical suspicion remains high or whenever the patient develops recurrent chest pain [36, 37]. The negative predictive value for MI in patients assigned ‘rule-out ‘exceeded 98% [35, 36, 37, 38, 39, 40, 41] used in conjunction with clinical and ECG findings. The positive predictive value for MI in those patients meeting the ‘rule-in’ criteria was 75–80%.

3.3.4. Noninvasive imaging

Transthoracic echocardiography is useful to identify abnormalities suggestive of myocardial ischemia or necrosis (i.e. segmental hypokinesia or akinesia). Strain and strain rate imaging can detect subtle reduced regional function in the absence of overt wall motion abnormalities, which improve the diagnostic and prognostic value of conventional echocardiography [42, 43]. Evaluation of left ventricular systolic function by echocardiography, at the indexed hospital admission, is important to estimate prognosis. Echocardiography can help in discrimination of other pathologies including acute aortic dissection, pericardial effusion, aortic valve stenosis, hypertrophic cardiomyopathy or right ventricular dilatation associated with acute pulmonary embolism. Echocardiography is the diagnostic tool of choice for patients with hemodynamic instability of suspected cardiac origin [44].

3.3.5. Functional stress testing

In patients without ischemic changes on 12-lead ECGs and negative cardiac troponins (preferably high-sensitivity) who are free of chest pain for several hours, stress imaging can be performed during admission or shortly after discharge [1, 45, 46]. The sensitivity and specificity of these tests increase when combined with either nuclear imaging to look for myocardial perfusion defects or echocardiography to assess wall motion abnormalities. Stress imaging is preferred over exercise ECG due to its greater diagnostic accuracy and superior prognostic value [47]. While studies have shown that normal exercise or pharmacological stress echocardiograms have high negative predictive value for ischemia and are associated with excellent patient outcomes ECG [48, 49, 50]. The addition of contrast to improve endocardial border detection and facilitate detection of ischemia [51].

To evaluate the extent of the CAD, a functional assessment using a submaximal exercise testing can be performed at 4 to 7 days after myocardial infarction, while symptom limited testing can be performed at 14 to 21 days post-myocardial infarction, when the patient has been free of active ischemic or heart failure symptoms [52].

3.3.6. Cardiac magnetic resonance

Cardiac magnetic resonance (CMR) can be used in the assessment of myocardial perfusion and wall motion abnormalities. Patients presenting with acute chest pain with a normal stress CMR have an excellent short and midterm prognosis [53]. CMR also permits detection of scar tissue (using late gadolinium enhancement) and can differentiate this from recent infarction (using T2-weighted imaging to delineate myocardial edema) [54, 55]. Moreover, CMR can facilitate the differential diagnosis between infarction and myocarditis or Takotsubo cardiomyopathy [56].

3.3.7. Nuclear myocardial perfusion imaging

Nuclear myocardial perfusion imaging has been shown to be useful for risk stratification in patients with acute chest pain suggestive for ACS. The presence of an area of myocardium that becomes deprived of perfusion during increased myocardial demand and reperfuses on stopping the activity on nuclear imaging stress tests is a reversible defect (Figure 2).

Figure 2.

Myocardial nuclear perfusion scan showing anterior, lateral and inferior reversible scan. Coronary angiogram confirmed three vessel disease.

Resting myocardial scintigraphy, can be helpful for the diagnosis of patients presenting with chest pain without ECG changes or elevated cardiac troponins [57]. Combined stress–rest imaging may further enhance assessment of ischemia, while a normal study is associated with excellent outcome [58, 59].

Advertisement

4. Anatomical evaluation

Multidetector computed tomography (MDCT) provide noninvasive evaluation of coronary anatomy and atherosclerosis. Due to the high negative predictive value of coronary computed tomography angiography (CCTA), evidence suggests that CCTA is useful in patients with low to moderate risk of NSTEMI where a normal scan excludes CAD. When compared with the standard care (observation, serial enzymes followed by stress testing) for low-risk patients, CCTA reduced time to diagnosis, reduced length of emergency department stay, and had similar safety [60]. CCTA had high negative predictive values to exclude ACS and excellent outcome in patients presenting to the emergency department with low to intermediate pre-test probability for ACS and a normal coronary CT angiogram [61]. CCTA was proven beneficial in the triage of low- to intermediate-risk patients presenting with acute chest pain to emergency departments without signs of ischemia on ECG and/or inconclusive cardiac troponins. At 6 months follow-up, there were no difference in the incidence of MI, post discharge emergency department visits or rehospitalizations, and no deaths in comparison to traditional management. Also, there were reduction in the cost and length of stay associated with MDCT [60, 62, 63, 64, 65]. But there was an increase in the use of invasive angiography [65]. CCTA is not indicated for patients with high-risk features and it is not useful in patients with known CAD [66]. Other factors limiting CCTA include severe calcifications and tachycardia. CT imaging can effectively exclude other causes of acute chest pain that, if untreated, are associated with high mortality, namely pulmonary embolism, aortic dissection and tension pneumothorax [67].

Advertisement

5. Risk assessment and outcomes

ACS management requires continuous risk stratification for death or recurrent MI. Quantitative assessment of ischemic risk by means of scores is superior to the clinical assessment alone to further triage and assist in the selection of treatment options [1]. A number of risk scores exist which incorporate a number of variables, the GRACE risk score and the TIMI risk score are examples.

The GRACE risk score provides the most accurate stratification of risk both on admission and at discharge [68, 69]. The GRACE 2.0 risk calculator provides a direct estimation, of mortality while in hospital, at 6 months, at 1 year and at 3 years. The combined risk of death or MI at 1 year is also provided [70]. Variables used in the GRACE 2.0 risk calculation include age, systolic blood pressure, pulse rate, serum creatinine, Killip class at presentation, cardiac arrest at admission, elevated cardiac biomarkers and ST deviation. The TIMI risk score uses seven variables in an additive scoring system: age ≥ 65 years, three or more CAD risk factors, known CAD, aspirin use in the past 7 days, severe angina (two or more episodes within 24 h), ST change ≥0.5 mm and positive cardiac marker [71]. Patients with a TIMI score of 0–2 are low risk, 3–4 are intermediate risk, and 5–7 are high risk. All-cause mortality, rate of MI, and rate of urgent revascularization at 14 days increase in proportion to the number of risk factors present on the TIMI score. It is simple to use, but its discriminative accuracy is inferior to that of the GRACE risk score [1, 71].

Advertisement

6. Hospital care and standard medical therapies

The aim of initial evaluation is to relieve pain and ischemia, prevent further thrombosis or embolism, and correct hemodynamic abnormalities and treat life-threatening complication.

All patients should undergo early risk estimation based on the medical history, physical exam, ECG findings, and cardiac markers.

Advertisement

7. Initial management

Initial medical therapy is indicated in all patients, with variation in some choices of agent according to risk stratification.

7.1. Cardiac rhythm monitoring

Early revascularization, effective antithrombotic therapy and administration of beta-blockers have reduced the incidence of life threatening arrhythmias in the acute phase of MI to <3%, with most of the arrhythmic events occurring within 12 h of symptom onset [72, 73]. Patients with life-threatening arrhythmias frequently had prior heart failure, low LV ejection fraction (EF < 30%) and triple vessel CAD.

NSTEMI patients at low risk for cardiac arrhythmias require rhythm monitoring for ≤24 h or until coronary revascularization (whichever comes first) in an intermediate or coronary care unit, while individuals at intermediate to high risk for cardiac arrhythmia may require rhythm monitoring for >24 h in an intensive or coronary care unit or in an intermediate care unit, depending on the clinical presentation, degree of revascularization and early post-revascularization course.

All patients require oxygen saturation measurement using pulse oximetry [1]. Although in the past oxygen was routinely given to all patients, there is no evidence to support this practice [74]. Moreover, results of the Air Versus Oxygen in ST-elevation MyocarDial Infarction (AVOID) trial have shown that routine supplemental oxygen may increase myocardial infarct size, and raise rates of recurrent MI and cardiac arrhythmia in patients with ST-elevation MI but without hypoxia. Guidelines now recommend supplemental oxygen therapy only in patients who are hypoxemic (arterial oxygen saturation < 90%), or in those who have respiratory distress or other high-risk features for hypoxemia [1, 15, 75].

7.2. Pharmacological treatment of ischemia

The goal of pharmacological anti-ischemic therapy is to decrease myocardial oxygen demand (secondary to a decrease in heart rate, blood pressure, preload or myocardial contractility) or to increase myocardial oxygen supply (by administration of oxygen or through coronary vasodilation).

Pain relief is indicated in the initial management of all patients. Those with ongoing ischemic discomfort should receive a trial of sublingual nitroglycerin (0. 4 mg) every 5 min for a total of three doses. Sublingual nitroglycerin reduces myocardial oxygen demand and enhances myocardial oxygen delivery. Intravenous nitroglycerin is recommended in patients with no symptom relief after sublingual nitroglycerin. Under careful blood pressure monitoring, the dose should be titrated upwards until symptoms are relieved, and in hypertensive patients until blood pressure is normalized, unless side effects (notably headache or hypotension) occur. Beyond symptom control, there is no indication for nitrate treatment [76]. In patients with recent intake of a phosphodiesterase type 5 inhibitor (i.e. within 24 h for sildenafil or vardenafil and 48 h for tadalafil), nitrates should not be administered due to the risk of severe hypotension. Nitroglycerin should not be given if systolic BP is <90 mmHg or there is a concern about right ventricular infarction [77]. If the patient does not respond to nitroglycerin, intravenous morphine can be administered in the absence of any contraindications [1]. Morphine causes vasodilation and may produce reductions in heart rate (through increased vagal tone) and systolic BP to further reduce myocardial oxygen demand. It should be given instead of nitroglycerin when nitroglycerin is contraindicated. Morphine should be used with caution, one randomized, double-blind trial found that morphine delays and attenuates ticagrelor exposure and action in patients with myocardial infarction [78, 79].

7.2.1. Beta-blockers

Oral beta-blockers are recommended for routine use in all patients unless contraindicated. Beta-blockers competitively inhibit the myocardial effects of circulating catecholamines and reduce myocardial oxygen consumption by lowering heart rate, blood pressure and myocardial contractility. Randomized trials with threatened or evolving MI have shown lower rates of progression to MI with beta-blocker treatment [80].

The beneficial effects of beta-blockers derived from several meta-analyses were a significant 8 and 13% relative risk reduction for in-hospital and first week mortality following MI respectively with no increase in cardiogenic shock [81, 82].

A registry study of NSTEMI patients found that the use of B-Blocker blockers within 24 h of hospital admission in patients at risk of developing cardiogenic shock (i.e. age > 70 years, heart rate > 110 beats/min, systolic blood pressure < 120 mmHg), the observed shock or death rate was significantly increased [83]. Therefore, early administration of beta-blockers should be avoided in these patients if the ventricular function is unknown.

Contraindications include heart rate <60 bpm, systolic BP <100 mmHg, moderate or severe associated left ventricular failure, PR interval on the ECG >0.24 s, second- or third-degree heart block, active asthma/reactive airways disease, severe COPD, hypotension, right ventricular infarction, and cardiogenic shock. Beta-blockers should not be administered in patients with symptoms possibly related to coronary vasospasm or cocaine use, as they might favor spasm by leaving alpha-mediated vasoconstriction unopposed by beta-mediated vasodilation.

7.3. Initial antiplatelet/anticoagulant

7.3.1. Bleeding risk assessment

The CRUSADE bleeding risk score considered baseline patient characteristics (i.e. female gender, history of diabetes, history of peripheral vascular disease or stroke), admission clinical variables (i.e. heart rate, systolic blood pressure, signs of heart failure) and admission laboratory values (i.e. hematocrit, calculated creatinine clearance) to estimate the patient’s likelihood of an in-hospital major bleeding event [84].

The Acute Catheterization and Urgent Intervention Triage strategy (ACUITY) bleeding risk score was derived from a pooled cohort recruited in the ACUITY and HORIZONS-AMI trials [85]. Six independent baseline predictors were identified including: female gender, advanced age, elevated serum creatinine, white blood cell count, anemia and presentation as NSTEMI or STEMI and one treatment-related variable [use of unfractionated heparin and a glycoprotein IIb/IIIa (GPIIb/IIIa) inhibitor rather than bivalirudin alone]. This risk score identified patients at increased risk for non-CABG-related major bleeds at 30 days and subsequent 1-year mortality. However, it has not been validated in an independent cohort.

Changes in interventional practice, such as increasing use of radial access, reduction in the dose of UFH, use of bivalirudin, diminished use of GPIIb/IIIa inhibitors and administration of more effective inhibitors of the platelet adenosine diphosphate (ADP) receptor P2Y12 (P2Y12 inhibitors), may all modify the predictive value of risk scores. Ischemic and bleeding risks need to be weighed in the individual patient, although many of the predictors of ischemic events are also associated with bleeding complications [84, 85]. Overall, CRUSADE and ACUITY scores have reasonable predictive value for major bleeding in ACS patients undergoing coronary angiography, with CRUSADE found to be the most discriminatory [86].

7.4. Platelet inhibition

7.4.1. Aspirin

Aspirin (chewed) is indicated immediately for all patients suspected of having an acute coronary syndrome unless contraindicated or already taken [1]. Aspirin should be continued at a daily maintenance dose thereafter [1]. Aspirin, an irreversible COX-1 inhibitor, suppresses thromboxane A2 production preventing platelet aggregation, and reduces the incidence of death and nonfatal MI in patients with unstable angina or acute MI [87, 88]. Aspirin has been shown to achieve a 30–51% reduction in future coronary events [89]. A meta-analysis suggests that aspirin administration (up to 2 years) is associated with a highly significant 46% odds reduction in major vascular events [90]. There was no difference between higher-dose (300–325 mg/day) and lower dose (75–100 mg/day) aspirin [91].

7.4.2. P2Y12 inhibitors

7.4.2.1. Clopidogrel

Clopidogrel (300–600 mg loading and 75 mg/day maintenance dose) is an inactive prodrug that requires oxidation by the hepatic cytochrome P450 (CYP) system to generate an active metabolite. Clopidogrel is a selective and irreversible inhibitor of platelet P2Y12 receptors and thus inhibits ADP-induced platelet aggregation [92, 93]. Dual antiplatelet therapy (DAPT) comprising aspirin and clopidogrel has been shown to reduce recurrent ischemic events in the NSTE-ACS setting compared with aspirin alone [94, 95]. However, up to 10% of patients treated with the combination of aspirin and clopidogrel will have a recurrent ischemic event in the first year after an ACS, with a rate of stent thrombosis of up to 2% [96]. There is substantial inter individual variability in the antiplatelet response to this drug and an increased risk of ischemic and bleeding events in Clopidogrel hypo- and hyper-responders, respectively [97, 98, 99, 100]. There is evidence that key gene polymorphisms are involved in both the variability of active metabolite generation and clinical efficacy of Clopidogrel [101, 102, 103, 104].

7.4.2.2. Prasugrel

Prasugrel (60 mg loading and 10 mg/day maintenance dose) is a prodrug that irreversibly blocks platelet P2Y12 receptors with a faster onset and a more profound inhibitory effect than clopidogrel. In the TRITON-TIMI 38, Prasugrel reduced recurrent CV event in ACS patients scheduled for PCI in comparison to clopidogrel, significantly driven by reduction in MI [105]. There were more severe bleeding complications with prasugrel, due to an increase in spontaneous and fatal bleeds [106]. Based on the marked reduction in definite or probable stent thrombosis observed in the TRITON-TIMI 38 prasugrel should be considered in patients with stent thrombosis despite compliance with clopidogrel therapy [100, 107]. Prasugrel is contraindicated in patients with prior stroke/transient ischemic attack due to evidence of net harm in this group in TRITON-TIMI 38. In addition, the study showed no apparent benefit in patients >75 years of age or with low bodyweight (<60 kg) [105].

7.4.2.3. Ticagrelor

Ticagrelor is an oral, reversibly binding P2Y12 inhibitor with a plasma half-life of 6–12 h.

Like prasugrel, ticagrelor has a more rapid and consistent onset of action compared with Clopidogrel, as well as a faster offset of action with more rapid recovery of platelet function [108].

In the PLATO trial, the primary composite efficacy endpoint (death from CV causes, MI or stroke) was significantly reduced with ticagrelor compared with similar reductions for CV and all-cause mortality [109, 110]. There was increased risk of non-CABG-related major bleeds with ticagrelor compared with Clopidogrel but no difference in life-threatening or fatal bleeds [110]. There was a reduction in definite stent thrombosis with ticagrelor in the NSTE-ACS subgroup. In addition to increased rates of minor or non-CABG-related major bleeding events with ticagrelor, adverse effects included dyspnea (without bronchospasm), increased frequency of asymptomatic ventricular pauses and increases in uric acid [109, 111, 112].

All patients should be given dual antiplatelet therapy with a P2Y12 receptor inhibitor in addition to aspirin. If the patient is intolerant of aspirin or it is otherwise contraindicated, a P2Y12 receptor inhibitor can be given instead of aspirin, but two different P2Y12 receptor inhibitors should not be given together. P2Y12 receptor inhibitors can reduce mortality and morbidity, but they are associated with an increased risk of bleeding [113, 114]. Ticagrelor and prasugrel are newer P2Y12 agents, which trials have shown to have a faster onset of action and greater efficacy compared with Clopidogrel [1, 115]. However, the risk of bleeding is also greater with these two P2Y12 agents compared with Clopidogrel [116, 117].

Clinicians need to tailor therapy to strike a balance between a newer agent that may have a faster onset of action and greater antiplatelet effect, but could potentiate bleeding (especially in those with prior TIA or stroke). Regardless of which P2Y12 receptor inhibitor is chosen, a loading dose should be given as soon as possible in most patients and then a maintenance dose continued for a minimum of 12 months [118].

7.5. Anticoagulation

Anticoagulation therapy (subcutaneous low molecular weight heparin, intravenous unfractionated heparin, or the alternative agents fondaparinux or bivalirudin) should be started on earliest recognition of NSTEMI. The anticoagulant is used in conjunction with antiplatelet therapy already started (i.e., aspirin and a P2Y12 receptor inhibitor). If fondaparinux is used during angiography/PCI, guidelines recommend that UFH be used in addition [1].

Anticoagulation should not be given if there are contraindications like major bleeding, history of adverse drug reaction or heparin-induced thrombocytopenia.

The antiplatelet and anticoagulation regimens should be started before the diagnostic angiogram. Triple antiplatelet therapy, in which an intravenous GP IIb/IIIa inhibitor is added to a P2Y12 receptor inhibitor, aspirin, and anticoagulation, can be considered for high-risk patients; however, it should be avoided in patients at high risk of bleeding [1]. Although guidelines recommend the use of GP IIb/IIIa inhibitors in NSTEMI, the level of evidence for their routine use is weak at best, particularly as results from randomized trials are conflicting [119, 120].

Advertisement

8. Conservative approach

Anticoagulation treatment should be added to aspirin and a P2Y12 receptor inhibitor at the earliest recognition of NSTEMI and continued for at least 48 h to hospital discharge and/or until symptoms abide and objective markers demonstrate a trend toward normal [121]. Agents include subcutaneous LMWH, intravenous UFH, or fondaparinux, according to clinician choice.

Advertisement

9. Ischemia-guided strategy versus early invasive strategies

9.1. Rationale and timing for early invasive strategy

Once initial management is instigated, the decision should be made as to whether the patient requires treatment using an invasive or noninvasive approach. The decision to pursue an invasive approach or medical management is made on an individual basis [122]. Invasive strategy carries risks but the benefit includes diagnostic accuracy, risk stratification and revascularization. The timing for coronary angiography and the selection of the revascularization modality depend on numerous factors, including clinical presentation, comorbidities, risk stratification, presence of high-risk features specific for a revascularization modality, frailty, cognitive status, estimated life expectancy and functional and anatomic severity as well as pattern of CAD. Guidelines recommend that high-risk patients routinely undergo early (12–24 h) coronary angiography and angiographically directed revascularization if possible unless patients have serious comorbidities, including cancer or end-stage liver disease, or clinically obvious contraindications, including acute or chronic (CKD 4 or higher) renal failure or multi-organ failure [1, 123, 124].

9.2. Routine invasive coronary angiography

Invasive coronary angiography allows to confirm the diagnosis of ACS related to obstructive epicardial CAD, to guide antithrombotic treatment, identify the culprit lesions and assess the suitability of coronary anatomy for PCI or CABG. Routine invasive strategy in NSTEMI has been shown to improve clinical outcomes and lower risk of death, reduce recurrent ischemic episodes, subsequent rehospitalization and revascularization [125, 126, 127].

Urgent and immediate angiography is indicated if patients do not stabilize with intensive medical treatment [1]. Guidelines recommend that an invasive approach is appropriate if any of the following high-risk features are present [1, 15]:

  • Recurrent angina or ischemia at rest or with low-level activities despite intensive medical therapy

  • Rise and fall in cardiac biomarkers (troponin T or I) consistent with MI

  • New or dynamic ST-T wave changes

  • Signs or symptoms of heart failure, or new or worsening mitral regurgitation

  • Hemodynamic instability

  • Life-threatening arrhythmia

  • PCI within 6 months

  • Prior CABG

  • High-risk score (i.e., TIMI, GRACE)

  • Mild to moderate renal dysfunction

  • Diabetes mellitus

  • Reduced left ventricular function (ejection fraction <40%).

9.3. Pattern of coronary artery disease

Angiographic patterns of CAD in NSTEMI patients are diverse, ranging from normal epicardial coronary arteries to a severely and diffusely diseased coronary arteries. Up to 20% of patients with NSTE-ACS have no lesions or non-obstructive lesions of epicardial coronary arteries, while among patients with obstructive CAD, 40–80% have multivessel disease [128, 129, 130].

Culprit lesions in the infarct-related artery are more often located within the proximal and mid segments, the left anterior descending coronary artery is the most frequent culprit vessel in both STEMI and NSTEMI-ACS (in up to 40% of patients). Left main coronary artery disease may be the underlying condition in 10% and a failure of bypass graft in 5% [128, 129, 130, 131, 132].

9.4. Identification of the culprit lesion

Culprit lesion on coronary angiography usually have features suggestive of acute plaque rupture. Vulnerable plaques are usually consisted from thin-cap fibroatheroma, and when rupture of the plaque happens they are characterized morphologically by the presence of at least two of the following features: intraluminal filling defects consistent with thrombus, plaque ulceration (i.e. presence of contrast and hazy contour beyond the vessel lumen), plaque irregularity (i.e. irregular margins or overhanging edges), dissection or impaired flow [132, 133, 134]. Multiple complex plaques observed in up to 40% of NSTEMI patients with obstructive CAD [132, 134, 135, 136, 137, 138]. One-quarter of NSTEMI patients present with an acute occluded coronary artery and two-thirds of the occlusions are already collateralized at the time of angiographic examination [138, 139].

Identification of the culprit lesion or the differentiation between an acute/subacute and chronic occlusion may sometimes be challenging based solely on angiography data. The additive value of the information from the ECG using lead localization and the regional wall motion abnormalities by Echocardiography can help identify the culprit lesion. Intracoronary imaging like optical coherence tomography can help to identify non-obstructive thin-cap fibroatheroma while vasospasm can be provoked by test such as acetylcholine [140, 141, 142]. The value of Fractional flow reserve (FFR) guided PCI in NSTEMI patient has not been properly addressed. The achievement of maximal hyperemia may be unpredictable in NSTEMI because of the dynamic nature of coronary lesions and the associated acute microvascular dysfunction. As a result, FFR may be overestimated and the hemodynamic relevance of a coronary stenosis underestimated [142].

9.5. Timing of invasive strategy

Routine intervention has been associated with an improved outcome [143, 144, 145, 146] however, the optimal timing of the intervention has not been well established. Early intervention might prevent ischemic events that could occur while the patient is awaiting a delayed procedure [147]. Alternatively, by treating a patient with intensive antithrombotic therapy and delaying intervention for several days, procedure-related complications might be avoided with intervention on a more stable plaque [148]. Thus, the question of when to intervene in patients with acute coronary syndromes without ST-segment elevation has not been definitively answered.

Immediate invasive strategy (<2 h from hospital admission) is recommended in very-high-risk NSTE-ACS patients with intent to perform vascularization because of the poor short- and long-term prognosis if left untreated.

Early invasive strategy (<24 h): Early invasive strategy is defined as coronary angiography performed within 24 h of hospital admission. Multiple studies showed no significant difference between early or delayed intervention groups in the rate of death, MI, stroke or major bleeds [130, 149, 150, 151].

In the early versus delayed invasive intervention in acute coronary syndromes clinical trial, prespecified analyses showed that early intervention improved the primary outcome in the third of patients who were at highest risk (GRACE risk score > 140) but not in the two thirds at low-to-intermediate risk (GRACE risk score ≤ 140) [129]. Early invasive strategy is recommended in patients with at least one high-risk criterion.

Delayed invasive strategy (<72 h): This is the recommended maximal delay for angiography in patients with low to intermediate risk [127, 149].

9.6. Selective invasive strategy

Patients with no recurrence of symptoms and none of the risk criteria (low risk patient), a non-invasive stress test preferably with imaging for inducible ischemia is recommended before deciding on an invasive strategy [152].

9.7. Conservative treatment

A conservative, early medical management strategy may be appropriate in patients with a low risk score, such subpopulations may not benefit from early invasive management especially low-risk women with NSTEMI [123, 124, 126]. Older patients may be considered at high risk for invasive approach regarding complications, but the benefit may be satisfactory from such approach in this subgroup [153, 154, 155]. Patients in whom an invasive strategy may be withheld by the treating physicians may include very elderly or frail patients, patients with comorbidities such as dementia, severe chronic renal insufficiency, or cancer and patients at high risk of bleeding complication. Ultimately patients care should be individualized and left at the discretion of the treating physician.

In the medically managed NSTE-ACS patients, the CURE study demonstrated that treatment with clopidogrel in addition to aspirin for 3–12 months, significantly lower the primary outcome (a composite of death from CV causes, non-fatal MI or stroke at 1 year) but there were significantly more major bleeds [94].

The association between clopidogrel use and the composite of death or MI was significant among patients presenting with NSTEMI compared with those presenting with unstable angina [156].

In the TRILOGY ACS trial, prasugrel was not associated with a statistically significant reduction in the primary endpoint (death, MI or stroke) but there were more frequent TIMI major and minor bleeding [157]. In the PLATO study, the incidence of the primary endpoint was lower with ticagrelor than with clopidogrel, but at the expense of higher incidence of TIMI major bleeds in the ticagrelor-treated patients [158].

Advertisement

10. Percutaneous coronary intervention: technical aspects and challenges

Stent implantation in the setting of NSTE-ACS helps to reduce abrupt vessel closure and restenosis associated with balloon angioplasty and it should be considered the standard treatment strategy (Figure 3 and movies online). New-generation drug eluting stents are recommended over bare metal stents in NSTE-ACS [159, 160, 161]. Dual antiplatelet therapy (DAPT) is recommended for 12 months irrespective of stent type, but DAPT may be extended depending on the number of stents and the total stents’ length used, patients with high risk of ischemic events recurrence and if patient’s bleeding risk is low. The benefit of thrombectomy has not been assessed prospectively in NSTE-ACS but cannot be recommended, considering the lack of benefit observed in STEMI [162].

Figure 3.

Angiogram of 54 years old gentleman presented with NSTEMI, ECG showed ST depression in the anterior leads. The angiogram confirmed a severe stenotic lesion in the proximal LAD (A) which stented successfully (B). Video clips of the angiogram available online.

Complications of PCI include PCI-induced MI; coronary perforation, dissection, or rupture; cardiac tamponade; malignant arrhythmias; cholesterol emboli; and bleeding from the access site. Contrast-induced nephropathy is a common and potentially serious complication, especially in patients with baseline impaired renal function [163]. Early and late stent thromboses are catastrophic complications. Radial access, performed by experienced operators, is associated with lower bleeding risk and recommended over the transfemoral access in ACS [164, 165].

11. Revascularization strategies and outcomes

In patients with complex, multivessel disease presenting with NSTEMI, the decision whether to do complete vs. incomplete revascularization and weather to do the complete revascularization at the index admission or to stage it is challenging and need to be tailored to age, general patient condition and comorbidities. A complete revascularization strategy of significant lesions should be pursued in multivessel disease patients with NSTE-ACS based on several studies showing the benefit of early intervention when compared with the conservative approach [143, 166, 167]. Also, recent trials have shown a detrimental prognostic effect of incomplete revascularization [168, 169].

Pursuing completeness of revascularization for some patients with complex coronary anatomy may mean increasing the risk of PCI especially in the presence of complex chronic total occlusions or referring to CABG.

The decision to treat all the significant lesions in the same setting or to stage the procedures should be based on clinical presentation, comorbidities, complexity of coronary anatomy, ventricular function, revascularization modality and patient preference.

With respect to outcomes, periprocedural complications of PCI as well as the long-term ischemic risk remain higher in NSTE-ACS than in stable patients, despite contemporary management. Accordingly, the risk of CV death, MI or stroke in NSTE-ACS patients in recent trials was approximately 10 and 15% at 1 and 2 years follow-up, respectively [110, 170]. For ACS patients who underwent PCI, revascularization procedures represent the most frequent, most costly and earliest cause for rehospitalization [171, 172].

12. Coronary artery bypass surgery

Approximately 10% of NSTEMI patients may require CABG during their index hospitalization [173]. The proportion of patient with NSTEMI undergoing CABG for NSTEMI decreased from 2001 to 2009, while the proportion of patients undergoing coronary angiography and PCI markedly increased [174]. CABG in the setting of NSTEMI is challenging mainly because of the difficulties in balancing ischemic and bleeding risks in relation to the timing of surgery and perioperative antithrombotic therapy. In addition, NSTEMI patients present with a higher proportion of surgical high-risk characteristics, including older age, female gender, left main coronary disease and LV dysfunction compared with patients undergoing elective CABG [175].

13. Percutaneous coronary intervention vs. coronary artery bypass surgery

The main advantages of PCI in the setting of NSTEMI are faster revascularization of the culprit lesion, a lower risk of stroke and the absence of deleterious effects of cardiopulmonary bypass on the ischemic myocardium, on the other hand, CABG may more frequently offer complete revascularization in advanced multivessel CAD. The decision to perform PCI or CABG was left to the discretion of the investigator. A post hoc analysis of NSTE-ACS patients with multivessel CAD included in the ACUITY trial showed that 78% underwent PCI while the remaining patients were treated surgically [176]. There were no differences in mortality at 1 month and 1 year between the two modalities. PCI treated patients experienced lower rates of stroke, MI, major bleeds and renal injury, but had significantly higher rates of unplanned revascularization than CABG during the periprocedural period and at 1 year [177, 178, 179].

While the majority of patients with single-vessel CAD should undergo ad hoc PCI of the culprit lesion, the revascularization strategy in an individual NSTE-ACS patient with multivessel CAD should be discussed in the context of a Heart Team and be based on the clinical status as well as the severity and distribution of the CAD and the lesion characteristics. The SYNTAX score was found to be useful in the prediction of death, MI and revascularization among NSTE-ACS patients undergoing PCI and may help guide the choice between revascularization strategies [180].

14. Long-term management post-stabilization

Cardiac rehabilitation is a structured program that provides heart attack survivors with the tools, motivation, and support needed to change behavior and increase chance of survival. Typically, cardiac rehabilitation programs use group therapy to supervise and promote beneficial exercise, as well as to provide emotional support. The aims of cardiac rehabilitation are to:

  • Increase functional capacity

  • Stop cigarette smoking

  • Modify lipids and lipoproteins

  • Decrease body weight and fat stores

  • Reduce BP

  • Improve psychosocial well-being

  • Prevent progression and promote plaque stability

  • Restore and maintain optimal physical, psychological, emotional, social, and vocational functioning.

Cardiac rehabilitation should be started on discharge and after clearance by an outpatient physician. The basic prescription should include aerobic and weight-bearing exercise 4–5 times per week for >30 min.

15. Pharmacologic strategies include the following

  • Aspirin should be continued indefinitely at a low dose if the patient is tolerant and not contraindicated.

  • A P2Y12 receptor inhibitor should be continued for up to 12 months. For patients with aspirin allergy, long-term P2Y12 receptor inhibitor use is suggested [1, 181].

  • Oral beta-blockers should be continued indefinitely, especially in patients with reduced left ventricular function.

  • All patients with NSTEMI should start high-intensity statin therapy (moderate-intensity if not a candidate for high-intensity statin) in hospital regardless of cholesterol levels, and if there are no contraindications [182]. Two trials demonstrated superior outcomes in patients treated with atorvastatin within 12 h of receiving PCI, and it may provide benefit when given early in NSTEMI [183, 184]. A high-intensity statin is defined as a daily dose that lowers LDL-C by approximately >50%, while a moderate-intensity statin daily dose lowers LDL-C by approximately 30–50%. Statin therapy is particularly important in patients who have hyperlipidemia, diabetes, prior MI, or CAD. Statins inhibit the rate-limiting step in cholesterol synthesis. They may also reduce vascular inflammation, improve endothelial function, and decrease thrombus formation in addition to lowering LDL [185]. The addition of ezetimibe to the statin regimen may also be considered to achieve lower LDL targets [186].

  • ACE inhibitors should be started in all patients with left ventricular systolic dysfunction (ejection fraction <40%), heart failure, HTN, diabetes, stable chronic kidney disease [1, 15]. They are started after 24 h. The goal BP is at least <140/90 mmHg (including patients with CKD or diabetes) [187].

  • Aldosterone antagonists should be used in all patients with left ventricular dysfunction (ejection fraction ≤40%), a history of diabetes mellitus, or evidence of congestive heart failure. Aldosterone blockade should not be used in patients with serum creatinine >2.5 mg/dL in men or > 2.0 mg/dL in women, as well as in patients with hyperkalemia (potassium >5.0 mEq/L) [188].

16. Prognosis

Patients who have experienced NSTEMI have a high risk of morbidity and death from a future event. The rate of sudden death in patients who have had an MI is 4–6 times the rate in the general population [189]. Life-threatening ventricular arrhythmias (sustained VT or VF) occurring after 48 h from the index acute coronary syndrome portend a poor prognosis, and are most frequently associated with left ventricular dysfunction. The benefit of implantable cardioverter-defibrillators, for both primary and secondary prevention, in patients with significant left ventricular dysfunction has been well demonstrated [190, 191]. Implantation for primary prevention should be considered at a minimum of 40 days following hospital discharge based on current recommendations [192].

Data from the era prior to medical therapy and revascularization suggest that the risk of cardiovascular death following an MI in the absence of treatment is approximately 5% per year, with a death rate after hospital discharge in the first year of about 10%. Pharmacotherapy, lifestyle changes, and cardiac rehabilitation are well demonstrated to be beneficial and together are additive in reducing mortality [193].

17. Monitoring

Patient monitoring after discharge is essential part of patient care. A follow-up should be arranged within the first 1 to 2 weeks of discharge and monthly visits should be scheduled thereafter. Lipids should be monitored at least every 6 months until a target LDL <70 mg/dL is reached in patients who have had an MI or have CAD. The need for follow-up cardiac ultrasounds is at the discretion of the physician. However, cardiac ultrasounds are necessary to evaluate and monitor ventricular function [1].

Smoking cessation, promotion of physical activity and joining the cardiac rehabilitation is extremely helpful. Psychosocial risk factors such as anxiety and depression should be addressed. Depression in particular has been associated with a poor prognosis [194]. All medications should be reviewed at every follow-up visit to encourage patient compliance and optimal dosing [1].

In patients who have undergone direct reperfusion, further noninvasive stress testing or further imaging is indicated only if stenosis of intermediate severity (luminal narrowing of 50–70%) is present in a non-culprit artery. Patients with recurrent ischemic-type pain after reperfusion may need angiography after medical therapy to evaluate for further stenosis or occlusion [195].

All patients, regardless of whether a stent was placed, should be treated with a P2Y12 receptor inhibitor for up to 12 months and low-dose aspirin daily as long as tolerated. This should be given for 1 month after bare-metal stent implantation, 3 months after sirolimus drug-eluting stent implantation, 6 months after paclitaxel drug-eluting stent implantation, and ideally up to 12 months if they are not at high risk for bleeding [195]. A scientific advisory from several major health organizations describes the risks of premature discontinuation of dual antiplatelet therapy in patients with coronary artery stents [196].

18. Management of patients with cardiogenic shock

Cardiogenic shock may develop in up to 3% of NSTE-ACS patients during hospitalization and has become the most frequent cause of in-hospital mortality in this setting [197, 198, 199]. One or more partial or complete vessel occlusions may result in severe heart failure, especially in cases of pre-existing LV dysfunction, reduced cardiac output and ineffective peripheral organ perfusion. More than two-thirds of patients have three-vessel CAD. Cardiogenic shock may also be related to mechanical complications of NSTEMI, including mitral regurgitation related to papillary muscle dysfunction or rupture and ventricular septal or free wall rupture. In patients with cardiogenic shock, immediate coronary angiography is indicated and PCI is the most frequently used revascularization modality. If the coronary anatomy is not suitable for PCI, patients should undergo emergent CABG. The value of intra-aortic balloon counter pulsation in MI complicated by cardiogenic shock has been challenged [200]. Extracorporeal membrane oxygenation and/or implantable LV assist devices may be considered in selected patients.

References

  1. 1. Amsterdam EA et al. 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. Journal of the American College of Cardiology. 2014;64(24):e139-e228
  2. 2. Steg G et al. ESC guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation. Revista Española de Cardiología (English Edition). 2013;66(1):53
  3. 3. Roffi M et al. 2015 ESC guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. European Heart Journal. 2015;37(3):267-315
  4. 4. Grech ED. Acute coronary syndrome: ST segment elevation myocardial infarction. British Medical Journal. 2003;326(7403):1379-1381
  5. 5. Reichlin T et al. Risk stratification in patients with unstable angina using absolute serial changes of 3 high-sensitive troponin assays. American Heart Journal. 2013;165(3):371-378 e3
  6. 6. Reichlin T et al. Introduction of high-sensitivity troponin assays: Impact on myocardial infarction incidence and prognosis. The American Journal of Medicine. 2012;125(12):1205-1213 (e1)
  7. 7. Mueller C. Biomarkers and acute coronary syndromes: An update. European Heart Journal. 2014;35(9):552-556
  8. 8. Thygesen K et al. Third universal definition of myocardial infarction. European Heart Journal. 2012;33(20):2551-2567
  9. 9. Thygesen K et al. How to use high-sensitivity cardiac troponins in acute cardiac care. European Heart Journal. 2012;33(18):2252-2257
  10. 10. Roe MT et al. Clinical and therapeutic profile of patients presenting with acute coronary syndromes who do not have significant coronary artery disease. Circulation. 2000;102(10):1101-1106
  11. 11. Reynolds HR et al. Mechanisms of myocardial infarction in women without angiographically obstructive coronary artery disease. Circulation. 2011;124(13):1414-1425
  12. 12. Braunwald E, Morrow DA. Unstable angina: Is it time for a requiem? Circulation. 2013;127(24):2452-2457
  13. 13. Alpert JS et al. Myocardial infarction redefined – A consensus document of The Joint European Society of Cardiology/American College of Cardiology Committee for the redefinition of myocardial infarction. Journal of the American College of Cardiology. 2000;36(3):959-969
  14. 14. Larsen AI et al. Characteristics and outcomes of patients with acute myocardial infarction and angiographically normal coronary arteries. The American Journal of Cardiology. 2005;95(2):261-263
  15. 15. Roffi M et al. 2015 ESC guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation: Task force for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation of the European Society of Cardiology (ESC). European Heart Journal. 2016;37(3):267-315
  16. 16. Mozaffarian D et al. Heart disease and stroke statistics – 2015 update: A report from the American Heart Association. Circulation. 2015;131(4):e29-e322
  17. 17. Amsterdam EA, Wenger NK. The 2014 American College of Cardiology (ACC)/American Heart Association guideline for the management of patients with non-ST-elevation acute coronary syndromes. Clinical Cardiology. 2015;38(2):121-123
  18. 18. Rogers WJ et al. Temporal trends in the treatment of over 1.5 million patients with myocardial infarction in the U.S. from 1990 through 1999. Journal of the American College of Cardiology. 2000;36(7):2056-2063
  19. 19. Sheridan PJ. Critical review of unstable angina and non-ST elevation myocardial infarction. Postgraduate Medical Journal. 2002;78(926):717-726
  20. 20. Jonasson L et al. Regional accumulations of T cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis. Thrombosis, and Vascular Biology. 1986;6(2):131-138
  21. 21. Hoffmeister HM et al. Alterations of coagulation and fibrinolytic and kallikrein-kinin systems in the acute and postacute phases in patients with unstable angina pectoris. Circulation. 1995;91(10):2520-2527
  22. 22. Wang TY et al. Incidence, distribution, and prognostic impact of occluded culprit arteries among patients with non-ST-elevation acute coronary syndromes undergoing diagnostic angiography. American Heart Journal. 2009;157(4):716-723
  23. 23. Padley SPG, Roditi G, Nicol ED. Chest pain of recent onset: Assessment and diagnosis (CG95). A step change in the requirement for cardiovascular CT. Clinical Radiology. 2017;72(9):751-753
  24. 24. Canto JG et al. Atypical presentations among medicare beneficiaries with unstable angina pectoris. The American Journal of Cardiology. 2002;90(3):248-253
  25. 25. Rubini Gimenez M et al. Sex-specific chest pain characteristics in the early diagnosis of acute myocardial infarction. JAMA Internal Medicine. 2014;174(2):241-249
  26. 26. Persson A et al. Long-term prognostic value of mitral regurgitation in acute coronary syndromes. Heart. 2010;96(22):1803-1808
  27. 27. Diercks DB et al. Frequency and consequences of recording an electrocardiogram >10 minutes after arrival in an emergency room in non-ST-segment elevation acute coronary syndromes (from the CRUSADE initiative). The American Journal of Cardiology. 2006;97(4):437-442
  28. 28. Savonitto S et al. Prognostic value of the admission electrocardiogram in acute coronary syndromes. Journal of the American Medical Association. 1999;281(8):707-713
  29. 29. Sgarbossa EB, Birnbaum Y, Parrillo JE. Electrocardiographic diagnosis of acute myocardial infarction: Current concepts for the clinician. American Heart Journal. 2001;141(4):507-517
  30. 30. Kurisu S et al. Electrocardiographic features in patients with acute myocardial infarction associated with left main coronary artery occlusion. Heart. 2004;90(9):1059-1060
  31. 31. de Winter RJ et al. A new ECG sign of proximal LAD occlusion. The New England Journal of Medicine. 2008;359(19):2071-2073
  32. 32. Reichlin T et al. Early diagnosis of myocardial infarction with sensitive cardiac troponin assays. The New England Journal of Medicine. 2009;361(9):858-867
  33. 33. Keller T et al. Sensitive troponin I assay in early diagnosis of acute myocardial infarction. The New England Journal of Medicine. 2009;361(9):868-877
  34. 34. Haaf P et al. High-sensitivity cardiac troponin in the distinction of acute myocardial infarction from acute cardiac noncoronary artery disease. Circulation. 2012;126(1):31-40
  35. 35. Rubini Gimenez M et al. Rapid rule out of acute myocardial infarction using undetectable levels of high-sensitivity cardiac troponin. International Journal of Cardiology. 2013;168(4):3896-3901
  36. 36. Bandstein N et al. Undetectable high-sensitivity cardiac troponin T level in the emergency department and risk of myocardial infarction. Journal of the American College of Cardiology. 2014;63(23):2569-2578
  37. 37. Reichlin T et al. Prospective validation of a 1-hour algorithm to rule-out and rule-in acute myocardial infarction using a high-sensitivity cardiac troponin T assay. Canadian Medical Association Journal. 2015;187(8):E243-E252
  38. 38. Reichlin T et al. One-hour rule-out and rule-in of acute myocardial infarction using high-sensitivity cardiac troponin T. Archives of Internal Medicine. 2012;172(16):1211-1218
  39. 39. Body R et al. Rapid exclusion of acute myocardial infarction in patients with undetectable troponin using a high-sensitivity assay. Journal of the American College of Cardiology. 2011;58(13):1332-1339
  40. 40. Zhelev Z et al. Diagnostic accuracy of single baseline measurement of Elecsys Troponin T high-sensitive assay for diagnosis of acute myocardial infarction in emergency department: Systematic review and meta-analysis. British Medical Journal. 2015;350(jan12 11):h15
  41. 41. Rubini Gimenez M et al. One-hour rule-in and rule-out of acute myocardial infarction using high-sensitivity cardiac troponin I. The American Journal of Medicine. 2015;128(8):861-870 e4
  42. 42. Tong KL et al. Myocardial contrast echocardiography versus Thrombolysis in myocardial infarction score in patients presenting to the emergency department with chest pain and a nondiagnostic electrocardiogram. Journal of the American College of Cardiology. 2005;46(5):920-927
  43. 43. Grenne B et al. Acute coronary occlusion in non-ST-elevation acute coronary syndrome: Outcome and early identification by strain echocardiography. Heart. 2010;96(19):1550-1556
  44. 44. Lancellotti P, Price S, Edvardsen T, Cosyns B, Neskovic AN, Dulgheru R, Flachskampf FA, Hassager C, Pasquet A, Gargani L, Galderisi M, Cardim N, Haugaa KH, Ancion A, Zamorano JL, Donal E, Bueno H, Habib G. The use of echocardiography in acute cardiovascular care: Recommendations of the European Association of Cardiovascular Imaging and the Acute Cardiovascular Care Association. Eur Heart J Acute Cardiovasc Care. 2015 Feb;4(1):3-5. DOI: 10.1177/2048872614568073. [Epub 2015 Jan 29]
  45. 45. Lindahl B et al. Risk stratification in unstable coronary artery disease: Additive value of troponin T determinations and pre-discharge exercise tests. European Heart Journal. 1997;18(5):762-770
  46. 46. Nyman I et al. The predictive value of silent ischemia at an exercise test before discharge after an episode of unstable coronary artery disease. American Heart Journal. 1992;123(2):324-331
  47. 47. Task Force M et al. 2013 ESC guidelines on the management of stable coronary artery disease: The task force on the management of stable coronary artery disease of the European Society of Cardiology. European Heart Journal. 2013;34(38):2949-3003
  48. 48. Shah BN et al. Incremental diagnostic and prognostic value of contemporary stress echocardiography in a chest pain unit: Mortality and morbidity outcomes from a real-world setting. Circulation: Cardiovascular Imaging. 2013;6(2):202-209
  49. 49. Sicari R et al. Stress echocardiography expert consensus statement – executive summary: European Association of Echocardiography (EAE) (a registered branch of the ESC). European Heart Journal. 2009;30(3):278-289
  50. 50. Gaibazzi N, Reverberi C, Badano L. Usefulness of contrast stress-echocardiography or exercise-electrocardiography to predict long-term acute coronary syndromes in patients presenting with chest pain without electrocardiographic abnormalities or 12-hour troponin elevation. The American Journal of Cardiology. 2011;107(2):161-167
  51. 51. Gaibazzi N et al. Contrast stress-echocardiography predicts cardiac events in patients with suspected acute coronary syndrome but nondiagnostic electrocardiogram and normal 12-hour troponin. Journal of the American Society of Echocardiography. 2011;24(12):1333-1341
  52. 52. Fletcher GF et al. Exercise standards for testing and training: A scientific statement from the American Heart Association. Circulation. 2013;128(8):873-934
  53. 53. Ingkanisorn WP et al. Prognosis of negative adenosine stress magnetic resonance in patients presenting to an emergency department with chest pain. Journal of the American College of Cardiology. 2006;47(7):1427-1432
  54. 54. Kwong RY et al. Detecting acute coronary syndrome in the emergency department with cardiac magnetic resonance imaging. Circulation. 2003;107(4):531-537
  55. 55. Cury RC et al. Cardiac magnetic resonance with T2-weighted imaging improves detection of patients with acute coronary syndrome in the emergency department. Circulation. 2008;118(8):837-844
  56. 56. Lockie T et al. Use of cardiovascular magnetic resonance imaging in acute coronary syndromes. Circulation. 2009;119(12):1671-1681
  57. 57. Udelson JE et al. Myocardial perfusion imaging for evaluation and triage of patients with suspected acute cardiac ischemia: A randomized controlled trial. Journal of the American Medical Association. 2002;288(21):2693-2700
  58. 58. Lim SH et al. Stress myocardial perfusion imaging for the evaluation and triage of chest pain in the emergency department: A randomized controlled trial. Journal of Nuclear Cardiology. 2013;20(6):1002-1012
  59. 59. Nabi F et al. Assessing risk in acute chest pain: The value of stress myocardial perfusion imaging in patients admitted through the emergency department. Journal of Nuclear Cardiology. 2012;19(2):233-243
  60. 60. Hoffmann U et al. Coronary CT angiography versus standard evaluation in acute chest pain. The New England Journal of Medicine. 2012;367(4):299-308
  61. 61. Samad Z et al. A meta-analysis and systematic review of computed tomography angiography as a diagnostic triage tool for patients with chest pain presenting to the emergency department. Journal of Nuclear Cardiology. 2012;19(2):364-376
  62. 62. Goldstein JA et al. The CT-STAT (coronary computed tomographic angiography for systematic triage of acute chest pain patients to treatment) trial. Journal of the American College of Cardiology. 2011;58(14):1414-1422
  63. 63. Goldstein JA et al. A randomized controlled trial of multi-slice coronary computed tomography for evaluation of acute chest pain. Journal of the American College of Cardiology. 2007;49(8):863-871
  64. 64. Litt HI et al. CT angiography for safe discharge of patients with possible acute coronary syndromes. The New England Journal of Medicine. 2012;366(15):1393-1403
  65. 65. Hulten E et al. Outcomes after coronary computed tomography angiography in the emergency department: A systematic review and meta-analysis of randomized, controlled trials. Journal of the American College of Cardiology. 2013;61(8):880-892
  66. 66. Hendel RC et al. ACCF/ACR/SCCT/SCMR/ASNC/NASCI/SCAI/SIR 2006 appropriateness criteria for cardiac computed tomography and cardiac magnetic resonance imaging: A report of the American College of Cardiology Foundation Quality Strategic Directions Committee Appropriateness Criteria Working Group, American College of Radiology, Society of Cardiovascular Computed Tomography, Society for Cardiovascular Magnetic Resonance, American Society of Nuclear Cardiology, North American Society for Cardiac Imaging, Society for Cardiovascular Angiography and Interventions, and Society of Interventional Radiology. Journal of the American College of Cardiology. 2006;48(7):1475-1497
  67. 67. Ayaram D et al. Triple rule-out computed tomographic angiography for chest pain: A diagnostic systematic review and meta-analysis. Academic Emergency Medicine. 2013;20(9):861-871
  68. 68. Aragam KG et al. Does simplicity compromise accuracy in ACS risk prediction? A retrospective analysis of the TIMI and GRACE risk scores. PLoS One. 2009;4(11):e7947
  69. 69. de Araujo Goncalves P et al. TIMI, PURSUIT, and GRACE risk scores: Sustained prognostic value and interaction with revascularization in NSTE-ACS. European Heart Journal. 2005;26(9):865-872
  70. 70. Fox KA et al. Should patients with acute coronary disease be stratified for management according to their risk? Derivation, external validation and outcomes using the updated GRACE risk score. BMJ Open Journal. 2014;4(2):e004425
  71. 71. Antman EM et al. The TIMI risk score for unstable angina/non-ST elevation MI: A method for prognostication and therapeutic decision making. Journal of the American Medical Association. 2000;284(7):835-842
  72. 72. Piccini JP et al. Sustained ventricular tachycardia and ventricular fibrillation complicating non-ST-segment-elevation acute coronary syndromes. Circulation. 2012;126(1):41-49
  73. 73. Rahimi K et al. Incidence, time course, and predictors of early malignant ventricular arrhythmias after non-ST-segment elevation myocardial infarction in patients with early invasive treatment. European Heart Journal. 2006;27(14):1706-1711
  74. 74. Cabello JB, Burls A, Emparanza JI, Bayliss S, Quinn T. Oxygen therapy for acute myocardial infarction. Cochrane Database Syst Rev. 2016 Dec 19;12:CD007160. DOI: 10.1002/14651858.CD007160.pub4
  75. 75. Stub D et al. Air versus oxygen in ST-segment-elevation myocardial infarction. Circulation. 2015;131(24):2143-2150
  76. 76. Borzak S et al. Effects of prior aspirin and anti-ischemic therapy on outcome of patients with unstable angina. The American Journal of Cardiology. 1998;81(6):678-681
  77. 77. Schwartz BG, Kloner RA. Drug interactions with phosphodiesterase-5 inhibitors used for the treatment of erectile dysfunction or pulmonary hypertension. Circulation. 2010;122(1):88-95
  78. 78. Meine TJ et al. Association of intravenous morphine use and outcomes in acute coronary syndromes: Results from the CRUSADE quality improvement initiative. American Heart Journal. 2005;149(6):1043-1049
  79. 79. Kubica J et al. Morphine delays and attenuates ticagrelor exposure and action in patients with myocardial infarction: The randomized, double-blind, placebo-controlled IMPRESSION trial. European Heart Journal. 2015;37(3):245-252
  80. 80. Yusuf S, Wittes J, Friedman L. Overview of results of randomized clinical trials in heart disease. II. Unstable angina, heart failure, primary prevention with aspirin, and risk factor modification. Journal of the American Medical Association. 1988;260(15):2259-2263
  81. 81. Yusuf S. Overview of results of randomized clinical trials in heart disease. Journal of the American Medical Association. 1988;260(14):2088
  82. 82. Chatterjee S et al. Early intravenous beta-blockers in patients with acute coronary syndrome – A meta-analysis of randomized trials. International Journal of Cardiology. 2013;168(2):915-921
  83. 83. Kontos MC et al. Treatment and outcomes in patients with myocardial infarction treated with acute beta-blocker therapy: Results from the American College of Cardiology's NCDR(R). American Heart Journal. 2011;161(5):864-870
  84. 84. Subherwal S et al. Baseline risk of major bleeding in non-ST-segment-elevation myocardial infarction: The CRUSADE (can rapid risk stratification of unstable angina patients suppress adverse outcomes with early implementation of the ACC/AHA guidelines) bleeding score. Circulation. 2009;119(14):1873-1882
  85. 85. Mehran R et al. A risk score to predict bleeding in patients with acute coronary syndromes. Journal of the American College of Cardiology. 2010;55(23):2556-2566
  86. 86. Abu-Assi E et al. Comparing the predictive validity of three contemporary bleeding risk scores in acute coronary syndrome. European Heart Journal. Acute Cardiovascular Care. 2012;1(3):222-231
  87. 87. Lewis Jr HD et al. Protective effects of aspirin against acute myocardial infarction and death in men with unstable angina. Results of a veterans administration cooperative study. The New England Journal of Medicine. 1983;309(7):396-403
  88. 88. Cairns JA et al. Aspirin, sulfinpyrazone, or both in unstable angina. Results of a Canadian multicenter trial. The New England Journal of Medicine. 1985;313(22):1369-1375
  89. 89. Antithrombotic Trialists C. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. British Medical Journal. 2002;324(7329):71-86
  90. 90. Collaboration AT. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients. British Medical Journal. 2002;324(7329):71-86
  91. 91. Investigators C-O et al. Dose comparisons of clopidogrel and aspirin in acute coronary syndromes. The New England Journal of Medicine. 2010;363(10):930-942
  92. 92. Savi P et al. P2y(12), a new platelet ADP receptor, target of clopidogrel. Biochemical and Biophysical Research Communications. 2001;283(2):379-383
  93. 93. Savi P, Herbert JM. Clopidogrel and ticlopidine: P2Y12 adenosine diphosphate-receptor antagonists for the prevention of atherothrombosis. Seminars in Thrombosis and Hemostasis. 2005;31(2):174-183
  94. 94. Yusuf S et al. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. The New England Journal of Medicine. 2001;345(7):494-502
  95. 95. Mehta SR et al. Effects of pretreatment with clopidogrel and aspirin followed by long-term therapy in patients undergoing percutaneous coronary intervention: The PCI-CURE study. Lancet. 2001;358(9281):527-533
  96. 96. Parodi G et al. High residual platelet reactivity after clopidogrel loading and long-term cardiovascular events among patients with acute coronary syndromes undergoing PCI. Journal of the American Medical Association. 2011;306(11):1215-1223
  97. 97. Hochholzer W et al. Impact of the degree of peri-interventional platelet inhibition after loading with clopidogrel on early clinical outcome of elective coronary stent placement. Journal of the American College of Cardiology. 2006;48(9):1742-1750
  98. 98. Sibbing D et al. Platelet reactivity after clopidogrel treatment assessed with point-of-care analysis and early drug-eluting stent thrombosis. Journal of the American College of Cardiology. 2009;53(10):849-856
  99. 99. Aradi D et al. Expert position paper on the role of platelet function testing in patients undergoing percutaneous coronary intervention. European Heart Journal. 2014;35(4):209-215
  100. 100. Pena A et al. Can we override clopidogrel resistance? Circulation. 2009;119(21):2854-2857
  101. 101. Simon T et al. Genetic determinants of response to clopidogrel and cardiovascular events. The New England Journal of Medicine. 2009;360(4):363-375
  102. 102. Collet JP et al. Cytochrome P450 2C19 polymorphism in young patients treated with clopidogrel after myocardial infarction: A cohort study. Lancet. 2009;373(9660):309-317
  103. 103. Gurbel PA et al. Genotyping: One piece of the puzzle to personalize antiplatelet therapy. Journal of the American College of Cardiology. 2010;56(2):112-116
  104. 104. Cayla G et al. Clinical, angiographic, and genetic factors associated with early coronary stent thrombosis. Journal of the American Medical Association. 2011;306(16):1765-1774
  105. 105. Wiviott SD et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. The New England Journal of Medicine. 2007;357(20):2001-2015
  106. 106. De Servi S et al. Clinical outcomes for prasugrel versus clopidogrel in patients with unstable angina or non-ST-elevation myocardial infarction: An analysis from the TRITON-TIMI 38 trial. European Heart Journal. Acute Cardiovascular Care. 2014;3(4):363-372
  107. 107. Wiviott SD et al. Intensive oral antiplatelet therapy for reduction of ischaemic events including stent thrombosis in patients with acute coronary syndromes treated with percutaneous coronary intervention and stenting in the TRITON-TIMI 38 trial: A subanalysis of a randomised trial. Lancet. 2008;371(9621):1353-1363
  108. 108. Gurbel PA et al. Randomized double-blind assessment of the ONSET and OFFSET of the antiplatelet effects of ticagrelor versus clopidogrel in patients with stable coronary artery disease: The ONSET/OFFSET study. Circulation. 2009;120(25):2577-2585
  109. 109. Wallentin L et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. The New England Journal of Medicine. 2009;361(11):1045-1057
  110. 110. Lindholm D et al. Ticagrelor vs. clopidogrel in patients with non-ST-elevation acute coronary syndrome with or without revascularization: Results from the PLATO trial. European Heart Journal. 2014;35(31):2083-2093
  111. 111. Steg PG et al. Stent thrombosis with ticagrelor versus clopidogrel in patients with acute coronary syndromes: An analysis from the prospective, randomized PLATO trial. Circulation. 2013;128(10):1055-1065
  112. 112. Storey RF et al. Characterization of dyspnoea in PLATO study patients treated with ticagrelor or clopidogrel and its association with clinical outcomes. European Heart Journal. 2011;32(23):2945-2953
  113. 113. Motovska Z, Kala P. Benefits and risks of clopidogrel use in patients with coronary artery disease: Evidence from randomized studies and registries. Clinical Therapeutics. 2008;30:2191-2202
  114. 114. Squizzato A1, Bellesini M, Takeda A, Middeldorp S, Donadini MP. Clopidogrel plus aspirin versus aspirin alone for preventing cardiovascular events. Cochrane Database Syst Rev. 2017 Dec 14;12:CD005158. DOI: 10.1002/14651858.CD005158.pub4
  115. 115. Mahaffey KW et al. Ticagrelor effects on myocardial infarction and the impact of event adjudication in the PLATO (platelet inhibition and patient outcomes) trial. Journal of the American College of Cardiology. 2014;63(15):1493-1499
  116. 116. Varenhorst C et al. Causes of mortality with ticagrelor compared with clopidogrel in acute coronary syndromes. Heart. 2014;100(22):1762-1769
  117. 117. Bavishi C et al. Meta-analysis of comparison of the newer oral P2Y12 inhibitors (prasugrel or ticagrelor) to clopidogrel in patients with non-ST-elevation acute coronary syndrome. The American Journal of Cardiology. 2015;116(5):809-817
  118. 118. Bonaca MP et al. Long-term use of ticagrelor in patients with prior myocardial infarction. The New England Journal of Medicine. 2015;372(19):1791-1800
  119. 119. de-Castellanos CA1, Colunga-Lozano LE, Delgado-Figueroa N, Magee K.Heparin versus placebo for non-ST elevation acute coronary syndromes.Cochrane Database Syst Rev. 2014 Jun 27;(6):CD003462. doi: 10.1002/14651858.CD003462.pub3
  120. 120. Hernandez AV et al. Effects of platelet glycoprotein IIb/IIIa receptor blockers in non-ST segment elevation acute coronary syndromes: Benefit and harm in different age subgroups. Heart. 2007;93(4):450-455
  121. 121. de-Castellanos CA1, Colunga-Lozano LE, Delgado-Figueroa N, Magee K.Heparin versus placebo for non-ST elevation acute coronary syndromes.Cochrane Database Syst Rev. 2014 Jun 27;(6):CD003462. DOI: 10.1002/14651858.CD003462.pub3
  122. 122. Topol EJ. A guide to therapeutic decision-making in patients with non-ST-segment elevation acute coronary syndromes. Journal of the American College of Cardiology. 2003;41(4):S123-S129
  123. 123. Kolh P, Windecker S. ESC/EACTS myocardial revascularization guidelines 2014. European Heart Journal. 2014;35(46):3235-3236
  124. 124. Unstable Angina and NSTEMI: The Early Management of Unstable Angina and Non-ST-Segment-Elevation Myocardial Infarction. National Institute for Health and Clinical Excellence: Guidance. London, UK: Royal College of Physicians, National Clinical Guideline Centre; 2010
  125. 125. Bavry AA et al. Benefit of early invasive therapy in acute coronary syndromes: A meta-analysis of contemporary randomized clinical trials. Journal of the American College of Cardiology. 2006;48(7):1319-1325
  126. 126. O'Donoghue M et al. Early invasive vs conservative treatment strategies in women and men with unstable angina and non-ST-segment elevation myocardial infarction: A meta-analysis. Journal of the American Medical Association. 2008;300(1):71-80
  127. 127. Fox KA et al. Long-term outcome of a routine versus selective invasive strategy in patients with non-ST-segment elevation acute coronary syndrome a meta-analysis of individual patient data. Journal of the American College of Cardiology. 2010;55(22):2435-2445
  128. 128. Kastrati A et al. Bivalirudin versus unfractionated heparin during percutaneous coronary intervention. The New England Journal of Medicine. 2008;359(7):688-696
  129. 129. Mehta SR et al. Early versus delayed invasive intervention in acute coronary syndromes. The New England Journal of Medicine. 2009;360(21):2165-2175
  130. 130. Thiele H et al. Optimal timing of invasive angiography in stable non-ST-elevation myocardial infarction: The leipzig immediate versus early and late percutaneous coronary intervention trial in NSTEMI (LIPSIA-NSTEMI trial). European Heart Journal. 2012;33(16):2035-2043
  131. 131. Ndrepepa G et al. Patterns of presentation and outcomes of patients with acute coronary syndromes. Cardiology. 2009;113(3):198-206
  132. 132. Kerensky RA et al. Revisiting the culprit lesion in non-Q-wave myocardial infarction. Results from the VANQWISH trial angiographic core laboratory. Journal of the American College of Cardiology. 2002;39(9):1456-1463
  133. 133. Ambrose JA et al. Angiographie morphology and the pathogenesis of unstable angina pectoris. Journal of the American College of Cardiology. 1985;5(3):609-616
  134. 134. Goldstein JA et al. Multiple complex coronary plaques in patients with acute myocardial infarction. The New England Journal of Medicine. 2000;343(13):915-922
  135. 135. Cheruvu PK et al. Frequency and distribution of thin-cap fibroatheroma and ruptured plaques in human coronary arteries: A pathologic study. Journal of the American College of Cardiology. 2007;50(10):940-949
  136. 136. Shishehbor MH et al. In unstable angina or non-ST-segment acute coronary syndrome, should patients with multivessel coronary artery disease undergo multivessel or culprit-only stenting? Journal of the American College of Cardiology. 2007;49(8):849-854
  137. 137. Libby P. Inflammation in atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2012;32(9):2045-2051
  138. 138. Virmani R et al. Pathology of the vulnerable plaque. Journal of the American College of Cardiology. 2006;47(8 Suppl):C13-C18
  139. 139. Kastrati A et al. Abciximab and heparin versus bivalirudin for non-ST-elevation myocardial infarction. The New England Journal of Medicine. 2011;365(21):1980-1989
  140. 140. Tanaka A et al. Conformational change in coronary artery structure assessed by optical coherence tomography in patients with vasospastic angina. Journal of the American College of Cardiology. 2011;58(15):1608-1613
  141. 141. Kato M et al. Presentations of acute coronary syndrome related to coronary lesion morphologies as assessed by intravascular ultrasound and optical coherence tomography. International Journal of Cardiology. 2013;165(3):506-511
  142. 142. Pijls NH, Tanaka N, Fearon WF. Functional assessment of coronary stenoses: Can we live without it? European Heart Journal. 2013;34(18):1335-1344
  143. 143. Cannon CP et al. Comparison of early invasive and conservative strategies in patients with unstable coronary syndromes treated with the glycoprotein IIb/IIIa inhibitor tirofiban. The New England Journal of Medicine. 2001;344(25):1879-1887
  144. 144. Invasive compared with non-invasive treatment in unstable coronary-artery disease: FRISC II prospective randomised multicentre study. Fragmin and fast revascularisation during instability in coronary artery disease investigators. Lancet. 1999;354(9180):708-715
  145. 145. Fox KA et al. 5-year outcome of an interventional strategy in non-ST-elevation acute coronary syndrome: The British Heart Foundation RITA 3 randomised trial. Lancet. 2005;366(9489):914-920
  146. 146. Spacek R et al. Value of first day angiography/angioplasty in evolving non-ST segment elevation myocardial infarction: An open multicenter randomized trial. The VINO study. European Heart Journal. 2002;23(3):230-238
  147. 147. Neumann FJ et al. Evaluation of prolonged antithrombotic pretreatment (“cooling-off” strategy) before intervention in patients with unstable coronary syndromes: A randomized controlled trial. Journal of the American Medical Association. 2003;290(12):1593-1599
  148. 148. de Winter RJ et al. Early invasive versus selectively invasive management for acute coronary syndromes. The New England Journal of Medicine. 2005;353(11):1095-1104
  149. 149. Navarese EP et al. Optimal timing of coronary invasive strategy in non-ST-segment elevation acute coronary syndromes: A systematic review and meta-analysis. Annals of Internal Medicine. 2013;158(4):261-270
  150. 150. Riezebos RK et al. Immediate versus deferred coronary angioplasty in non-ST-segment elevation acute coronary syndromes. Heart. 2009;95(10):807-812
  151. 151. Montalescot G et al. Immediate vs delayed intervention for acute coronary syndromes: A randomized clinical trial. Journal of the American Medical Association. 2009;302(9):947-954
  152. 152. Amsterdam EA et al. Testing of low-risk patients presenting to the emergency department with chest pain: A scientific statement from the American Heart Association. Circulation. 2010;122(17):1756-1776
  153. 153. Tegn N et al. Invasive versus conservative strategy in patients aged 80 years or older with non-ST-elevation myocardial infarction or unstable angina pectoris (after eighty study): An open-label randomised controlled trial. The Lancet. 2016;387(10023):1057-1065
  154. 154. Bach RG et al. The effect of routine, early invasive management on outcome for elderly patients with non-ST-segment elevation acute coronary syndromes. Annals of Internal Medicine. 2004;141(3):186-195
  155. 155. Devlin G et al. Management and 6-month outcomes in elderly and very elderly patients with high-risk non-ST-elevation acute coronary syndromes: The global registry of acute coronary events. European Heart Journal. 2008;29(10):1275-1282
  156. 156. Solomon MD et al. Comparative effectiveness of clopidogrel in medically managed patients with unstable angina and non-ST-segment elevation myocardial infarction. Journal of the American College of Cardiology. 2014;63(21):2249-2257
  157. 157. Roe MT et al. Prasugrel versus clopidogrel for acute coronary syndromes without revascularization. The New England Journal of Medicine. 2012;367(14):1297-1309
  158. 158. James SK et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes intended for non-invasive management: Substudy from prospective randomised platelet inhibition and patient outcomes (PLATO) trial. British Medical Journal. 2011;342(Jun 17):d3527
  159. 159. Raber L et al. Effect of biolimus-eluting stents with biodegradable polymer vs bare-metal stents on cardiovascular events among patients with acute myocardial infarction: The comfortable AMI randomized trial. Journal of the American Medical Association. 2012;308(8):777-787
  160. 160. Sabate M et al. Everolimus-eluting stent versus bare-metal stent in ST-segment elevation myocardial infarction (Examination): 1 year results of a randomised controlled trial. Lancet. 2012;380(9852):1482-1490
  161. 161. Valgimigli M et al. Two-year outcomes after first- or second-generation drug-eluting or bare-metal stent implantation in all-comer patients undergoing percutaneous coronary intervention: A pre-specified analysis from the PRODIGY study (prolonging dual antiplatelet treatment after grading stent-induced intimal hyperplasia study). JACC: Cardiovascular Interventions. 2014;7(1):20-28
  162. 162. Lagerqvist B et al. Outcomes 1 year after thrombus aspiration for myocardial infarction. The New England Journal of Medicine. 2014;371(12):1111-1120
  163. 163. McCullough PA et al. Epidemiology and prognostic implications of contrast-induced nephropathy. The American Journal of Cardiology. 2006;98(6):5-13
  164. 164. Jolly SS et al. Radial versus femoral access for coronary angiography and intervention in patients with acute coronary syndromes (RIVAL): A randomised, parallel group, multicentre trial. Lancet. 2011;377(9775):1409-1420
  165. 165. Rao SV et al. A registry-based randomized trial comparing radial and femoral approaches in women undergoing percutaneous coronary intervention: The SAFE-PCI for women (study of access site for enhancement of PCI for women) trial. JACC. Cardiovascular Interventions. 2014;7(8):857-867
  166. 166. Fox KA et al. Interventional versus conservative treatment for patients with unstable angina or non-ST-elevation myocardial infarction: The British Heart Foundation RITA 3 randomised trial. Randomized intervention trial of unstable angina. Lancet. 2002;360(9335):743-751
  167. 167. Wallentin L et al. Outcome at 1 year after an invasive compared with a non-invasive strategy in unstable coronary-artery disease: The FRISC II invasive randomised trial. The Lancet. 2000;356(9223):9-16
  168. 168. Farooq V et al. Quantification of incomplete revascularization and its association with five-year mortality in the synergy between percutaneous coronary intervention with taxus and cardiac surgery (SYNTAX) trial validation of the residual SYNTAX score. Circulation. 2013;128(2):141-151
  169. 169. Genereux P et al. Quantification and impact of untreated coronary artery disease after percutaneous coronary intervention: The residual SYNTAX (synergy between PCI with taxus and cardiac surgery) score. Journal of the American College of Cardiology. 2012;59(24):2165-2174
  170. 170. Tricoci P et al. Thrombin-receptor antagonist vorapaxar in acute coronary syndromes. The New England Journal of Medicine. 2012;366(1):20-33
  171. 171. Curtis JP et al. All-cause readmission and repeat revascularization after percutaneous coronary intervention in a cohort of medicare patients. Journal of the American College of Cardiology. 2009;54(10):903-907
  172. 172. Meadows ES et al. Rehospitalization following percutaneous coronary intervention for commercially insured patients with acute coronary syndrome: A retrospective analysis. BMC Research Notes. 2012;5(1):342
  173. 173. Ranasinghe I et al. Risk stratification in the setting of non-ST elevation acute coronary syndromes 1999-2007. The American Journal of Cardiology. 2011;108(5):617-624
  174. 174. Martensson S et al. Trends in time to invasive examination and treatment from 2001 to 2009 in patients admitted first time with non-ST elevation myocardial infarction or unstable angina in Denmark. BMJ Open Journal. 2014;4(1):e004052
  175. 175. Fukui T et al. Early and long-term outcomes of coronary artery bypass grafting in patients with acute coronary syndrome versus stable angina pectoris. The Journal of Thoracic and Cardiovascular Surgery. 2013;145(6):1577-1583 (1583 e1)
  176. 176. Ben-Gal Y et al. Surgical versus percutaneous revascularization for multivessel disease in patients with acute coronary syndromes: Analysis from the ACUITY (acute catheterization and urgent intervention triage strategy) trial. JACC: Cardiovascular Interventions. 2010;3(10):1059-1067
  177. 177. Mohr FW et al. Coronary artery bypass graft surgery versus percutaneous coronary intervention in patients with three-vessel disease and left main coronary disease: 5-year follow-up of the randomised, clinical SYNTAX trial. Lancet. 2013;381(9867):629-638
  178. 178. Farkouh ME et al. Strategies for multivessel revascularization in patients with diabetes. The New England Journal of Medicine. 2012;367(25):2375-2384
  179. 179. Windecker S et al. 2014 ESC/EACTS guidelines on myocardial revascularization. EuroIntervention. 2015;10(9):1024-1094
  180. 180. Palmerini T et al. Prognostic value of the SYNTAX score in patients with acute coronary syndromes undergoing percutaneous coronary intervention: Analysis from the ACUITY (acute catheterization and urgent intervention triage strategy) trial. Journal of the American College of Cardiology. 2011;57(24):2389-2397
  181. 181. Harrington RA et al. Antithrombotic therapy for non-ST-segment elevation acute coronary syndromes. Chest. 2008;133(6):670S-707S
  182. 182. Stone NJ et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: A report of the American College of Cardiology/American Heart Association task force on practice guidelines. Journal of the American College of Cardiology. 2014;63(25 Pt B):2889-2934
  183. 183. Briguori C et al. Novel approaches for preventing or limiting events (Naples) II trial: Impact of a single high loading dose of atorvastatin on periprocedural myocardial infarction. Journal of the American College of Cardiology. 2009;54(23):2157-2163
  184. 184. Di Sciascio G et al. Efficacy of atorvastatin reload in patients on chronic statin therapy undergoing percutaneous coronary intervention: Results of the ARMYDA-RECAPTURE (atorvastatin for reduction of myocardial damage during angioplasty) randomized trial. Journal of the American College of Cardiology. 2009;54(6):558-565
  185. 185. Sposito AC. Statin therapy in acute coronary syndromes: Mechanistic insight into clinical benefit. Arteriosclerosis, Thrombosis, and Vascular Biology. 2002;22(10):1524-1534
  186. 186. Cannon CP et al. Ezetimibe added to statin therapy after acute coronary syndromes. The New England Journal of Medicine. 2015;372(25):2387-2397
  187. 187. James PA et al. 2014 Evidence-based guideline for the management of high blood pressure in adults. Journal of the American Medical Association. 2014;311(5):507
  188. 188. Pitt B et al. Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. The New England Journal of Medicine. 2003;348(14):1309-1321
  189. 189. Zaman S, Kovoor P. Sudden cardiac death early after myocardial infarction: Pathogenesis, risk stratification, and primary prevention. Circulation. 2014;129(23):2426-2435
  190. 190. Moss AJ et al. Improved survival with an implanted defibrillator in patients with coronary disease at high risk for ventricular arrhythmia. New England Journal of Medicine. 1996;335(26):1933-1940
  191. 191. Moss AJ et al. Prophylactic implantation of a defibrillator in patients with myocardial infarction and reduced ejection fraction. The New England Journal of Medicine. 2002;346(12):877-883
  192. 192. Hohnloser SH et al. Prophylactic use of an implantable cardioverter-defibrillator after acute myocardial infarction. New England Journal of Medicine. 2004;351(24):2481-2488
  193. 193. Law MR, Watt HC, Wald NJ. The underlying risk of death after myocardial infarction in the absence of treatment. Archives of Internal Medicine. 2002;162(21):2405
  194. 194. Lichtman JH et al. Depression as a risk factor for poor prognosis among patients with acute coronary syndrome: Systematic review and recommendations: A scientific statement from the American Heart Association. Circulation. 2014;129(12):1350-1369
  195. 195. Smith SC. ACC/AHA/SCAI 2005 guideline update for percutaneous coronary intervention – summary article: A report of the American College of Cardiology/American Heart Association task force on practice guidelines (ACC/AHA/SCAI writing committee to update the 2001 guidelines for percutaneous coronary intervention). Circulation. 2005;113(1):156-175
  196. 196. Grines CL et al. Prevention of premature discontinuation of dual Antiplatelet therapy in patients with coronary artery stents: A science advisory from the American Heart Association, American College of Cardiology, Society for Cardiovascular Angiography and Interventions, American College of Surgeons, and American Dental Association, with representation from the American College of Physicians. Circulation. 2007;115(6):813-818
  197. 197. Hasdai D et al. Platelet glycoprotein IIb/IIIa blockade and outcome of cardiogenic shock complicating acute coronary syndromes without persistent ST-segment elevation. Journal of the American College of Cardiology. 2000;36(3):685-692
  198. 198. Holmes DR et al. Cardiogenic shock in patients with acute ischemic syndromes with and without ST-segment elevation. Circulation. 1999;100(20):2067-2073
  199. 199. Hollenberg SM, Kavinsky CJ, Parrillo JE. Cardiogenic shock. Annals of Internal Medicine. 1999;131(1):47-59
  200. 200. Thiele H et al. Intra-aortic balloon counterpulsation in acute myocardial infarction complicated by cardiogenic shock (IABP-SHOCK II): Final 12 month results of a randomised, open-label trial. Lancet. 2013;382(9905):1638-1645

Written By

Yaser Al Ahmad and Mohammed T. Ali

Submitted: 25 July 2017 Reviewed: 06 March 2018 Published: 03 January 2019