The Protocol Book Soumitra Kumar
INDEX
A
Abciximab 37, 38
Aborted MI 12
ACE-inhibitors (ACEIs) 91
Acute cryptogenic organizing pneumonia 259
Acute eosinophilic pneumonia 259
Acute hypersensitivity pneumonitis 259
Acute interstitial pneumonia 258
Acute limb ischaemia 186
Acute tubular necrosis 289
Adenosine 131, 156, 356
Adenosine A1 receptor blocker 81
Adenosine receptor blockers 81
Adenosine triphosphate 81, 109, 152, 153
Adrenal crisis 300
Adrenaline 363
Aldosterone receptor antagonists 94
Aliskiren 105
Alprostadil 192, 357
Aminoglycoside 244, 248
Aminophylline 237
Amiodarone 96, 155, 356
Amphotericin B 352
Aneurysmectomy 105
Angiodysplasia 268
Angiogenesis 99
Angiotensin-receptor blockers 92
Anion gap 213
Anticonvulsants 276
Antimicrobial drugs 376
Antiphospholipid antibody syndrome 321
Aortic dissection 183
Apache II 342, 380, 381
Apache III 380, 381
Apixban 121
Arrhythmogenic right ventricular cardiomyopathy 142
Arrhythmogenic RV dysplasia 110
Aspirin 34, 119, 160, 275
Atenolol 359
Atropine 358
Azimilide 125
Aztreonam 340
B
B-type natriuretic peptide 68, 87
Balloon atrial septostomy 192
Balloon dilatation catheters 163
Barotrauma 251
Base excess 213
Basic life support 151
Beta-blockers 34, 93, 132, 160
Beta-blockers, acute 359
Bivalirudin 45
BNP (B-type natriuretic peptide) 31
Bosentan 97
Brugada syndrome 110, 143
Bundle-branch reentrant tachycardia 142
C
C-reactive protein 31
Calponin 185
Captopril 200
Cardiac index 54, 63, 66, 75
Cardiac rehabilitation 171
Cardiac resynchronization therapy 99
Cardiac transplantation 104
Cardioembolic stroke 270
Cardiogenic shock 54
Cardiomyoplasty 104
Cariporide 125
Carotid sinus hypersensitivity 111
Carotid sinus massage 109
Carvedilol 200
Catecholaminergic polymorphic ventricular tachycardia 145
Catheter ablation 123
Central venous pressure 73
Cephalosporin 340, 341, 350, 351
Cerebellar haemorrhage 280
Choline 31
Ciprofloxacin 247, 248, 249, 340
Clopidogrel 35, 160, 275
Coagulase negative staphylococci 345
Colloid 248, 335, 354
Congenital long Q-T syndrome 147
Congrelor 36
Conivaptan 97
Coronary artery bypass grafting 55
Coronary stenting 164
Corticosteroids 278
Creatine kinase-MB 30
Creatinine clearance 380
Crystalloid 335
Cyclophosphamide 325
Cyclosporine 326
D
D-dimer assay 177
Dabigatran 121
Darbopoetin 106
Dabigatran etexilate 121
Deep venous thrombosis 172
Dengue 353, 354
Diabetic ketoacidosis 309
Diazoxide 210
Dieulafoy's lesion 267, 268
Diffuse alveolar haemorrhage 259
Digitalis 94, 132
Digoxin 200, 360
Dilated cardiomyopathy 141
Diltiazem 132
Diltiazem 156
Diltiazem (IV) 361
Dipyridamole 275
Direct thrombin inhibitors 20
Disopyramide 122
Diuretics 78, 90
Dobutamine 79, 200, 361, 372
Dofetilide 118, 128
Dopamine 79, 200, 362, 372
Dressler's syndrome 27
Dronedarone 125
Drotrecogin 348, 349
Drotrecogin alfa 342, 343
E
Echocardiography portable 68
Eclampsia 206
Electrical cardioversion 117
Electrophysiology studies 109
Enalapril 200
Enalaprilat 210, 274
Endoaneurysmorrhaphy 105
Endoscopic therapy 266
Enoximone, vesanarinone 95
Enterococcus 350
Epinephrine 79, 363
Eplerenone 95
Eptifibatide 37, 38, 375
Esmolol 359
Esmolol hydrochloride 210
Etanercept 97
Euro heart failure survey II 85
Extended spectrum beta-lactamases 350
F
Facilitated PCI 14
Fascicular tachycardia 145
Fatty acid binding protein 31
Fenoldopam mesylate 209
Fibrinolytic agents 363
Flecainide 118, 122
Fluconazole 331, 352
Flucytosine 352
Fondaparinux 21, 40, 44, 183
Free wall rupture 26, 56, 81
Fungal infections 351
Furosemide 200
G
Glycerol 276
Glyceryl trinitrate 78
Glycoprotein IIb/IIIa inhibitors 21, 36, 160, 363
Grace risk score 32
Guiding catheter 161
H
Haemobilia 268
Haemodialysis 298
Haemofiltration 298
Hanta virus infection 353
Hemicraniectomy 276
Heparin 160, 363
Heparin-induced thrombocytopenia (HIT) 176
Hirudin 45
Hirulog 45
Hydralazine 95, 205
Hypercalcaemia 318
Hyperglycaemic hyperosmolar state 313
Hyperglycemia 316, 344
Hyperkalaemia 196, 224
Hypermagnesemia 227
Hypernatremia 222
Hypertensive crisis 202, 304
Hypertensive emergencies 202
Hypertensive urgencies 203
Hypertrophic cardiomyopathy 143
Hypocalcaemia 319
Hypokalemia 225
Hyponatremia 221
Hypotension 251
I
IABP 60
Ibutilide 122
Idraparinux 183
Imipenem 248, 249, 340, 378
Implantable cardioverter defibrillator 103
Implantable loop recorder 109
Infliximab 97, 327
Inotropic agents 79, 80
Intra-aortic balloon pump 169
Intracerebral haemorrhage 277
Ipratropium bromide 231
Isoprenaline 364
Isoproterenol 364
Istaroxime 81
Itraconazole 352
J
Japanese B encephalitis 354
L
Labetolol 274
Labetolol hydrochloride 210
Lacunar stroke 270
Left ventricular assist devices 169
Leptospirosis 353
Levosalbutamol 237
Levosimendan 79
Lignocaine 156, 364
Linezolid 340, 350, 351
Low molecular weight heparin 19, 42, 174, 183
M
Magnesium sulpate 231, 365
Malaria 353
Mallory-Weiss syndrome 265
Mannitol 276, 278
Mechanical ventilation 241
Meropenem 340
Metabolic acidoses 215
Metabolic alkalosis 215
Methicillin-resistant Staphylococcus aureus 350
Methylprednisolone 325
Metolazone 78, 200
Metoprolol 193, 359
Midodrine 112
Milrinone 79, 200, 365
Monobactam 340
Morphine sulfate 193, 366
MPM II 380, 381
Multivessel disease 51
Myeloperoxidase 31
Myocardial infarction 1
Myoglobin 31
Myxoedema coma 308
N
Neonatal lupus 326
Nesiritide 79
Neurally mediated reflex 107
Neurally mediated syncope 111
Nicardipine 274
Nicardipine hydrochloride 209
Nifedipine 274
NIH stroke scale 272
Nitroglycerine, acute 366
Nitroprusside 78, 200, 367, 373
Noninvasive ventilation 238
No-reflow 166
Non-sustained ventricular tachycardia 140, 146
Noninvasive ventilation 233
Norepinephrine 79, 368, 373
O
Obscure 283, 337
Omapatrilat 97
Open artery hypothesis 16
Orthostatic syncope 112
P
Pentasaccharides 183
Percutaneous coronary intervention 58
Percutaneous mechanical thrombectomy 189
Perhexiline 81
Pericarditis 26
Peritoneal dialysis 298
Phaeochromocytoma 304
Phenoxybenzamine 369
Phentolamine 210
Phenylephrine 193, 369
Piboserod 125
Piperacillin-tazobactam 340
Pituitary apoplexy 302
Plasmapheresis 326
Polymorphic VT 147
Potassuim sparing diuretics 91
Prasugrel 36
Prednisolone 231, 325
Pregnancy-associated plasma protein A 31
Pre-hospital thrombolysis 13
Primary PCI 7
Procainamide 157
Procalcitonin 339
Propranolol 193
Prostaglandin 192
Prostaglandin E1 357
Proton pump inhibitor 266
Pseudomonas aeruginosa 351
Pulmonary artery catheter 73
Pulmonary artery systolic pressure 72
Pulmonary embolism 177
Pulmonary thromboendarterectomy 175
Pulmonary vascular resistance 72, 74
Q
Quinidine 122
R
Recombinant factor VIIa 279
Reperfusion injury 168
Rescue PCI 12
Respiratory acidosis 215
Respiratory alkalosis 215
RF ablation 132
Right ventricular infarction 61
Rivaroxaban 121
Rotigaptide 125
rtPA 188
RV function assessment 72
S
Salbutamol 231, 237
SAPS 380, 381
SCD40 ligand 31
Scrub typhus 353
Septic shock 336
Short QT syndrome 150
Sodium nitroprusside 209
Somatostatin 266
Sotalol 118, 131
Spironolactone 95
Statins 97, 161
Steptokinase 188
Supraventricular tachycardia 132
Surgical ablation 124
Syncope
cardiac 108
neurally mediated reflex 108
orthostatic 108
Systemic inflammatory response syndrome 332
Systemic vascualr resistance 63, 74
T
Tedisamil 125
Teicoplanin 340
Tezosentan 97
Thrombin-receptor antagonists 36
Thrombolysis 181, 272
Thrombolytic therapy 4
Thrombophilias 172
Thrombus 166
Thyroid storm 305
Thyrotoxic crisis 305
Ticarcillin-clavulanate 340
Tilt-table testing 109
TIMI risk score 32
Tirofiban 37, 38, 374
Tissue Doppler echocardiography 100
Tolvaptan 97
Torsade de pointes 136
Transmyocardial laser revascularization 99
Troponins 30
U
Unfractionated heparin 18, 41, 174, 182
Urodilatin 81
Urokinase 188
V
Vancomycin 340
Variceal bleeding 266
Vasodilators 78, 80
Vasopressin 370
Vasopressin antagonists 81
Vasopressors 348
Vasovagal syncope 111
Ventricular associated pneumonia 246
Ventricular fibrillation 136
Ventricular flutter 136
Ventricular tachyarrhythmias 135
Verapamil 132, 156
Verapamil (intravenous) 370
Voriconazole 352
W
Warfarin 96, 119
Weaning 261
Wide complex tachycardia 133
X
Ximelagatran 121, 175
Z
Zoniporide 125
×
Chapter Notes

Save Clear


Acute ST-Elevation Myocardial InfarctionChapter 1

Vidya Verma,
Soumitra Kumar
 
UNIVERSAL DEFINITION OF MYOCARDIAL INFARCTION
(Joint ESC/ACCF/AHA/WHF Task Force for the Redefinition of Myocardial Infarction 2007)
 
Clinical Classification of Different Types of Myocardial Infarction
 
Type 1
Spontaneous myocardial infarction related to ischaemia due to a primary coronary event such as plaque erosion and/or rupture, fissuring or dissection.
 
Type 2
Myocardial infarction secondary to ischaemia due to either increased oxygen demand or decreased supply, e.g. coronary artery spasm, coronary embolism, anaemia, arrhythmias, hypertension, or hypotension.
 
Type 3
Sudden unexpected cardiac death, including cardiac arrest, often with symptoms suggestive of myocardial ischaemia, accompanied by presumably new ST elevation, or new LBBB or evidence of fresh thrombus in a coronary artery by angiography and/or at autopsy, but death occurring before blood samples could be obtained or at a time before the appearance of cardiac biomarkers in the blood.
 
Type 4A
Myocardial infarction associated with PCI.
 
Type 4B
Myocardial infarction associated with stent thrombosis as documented by angiography or at autopsy.
 
Type 5
Myocardial infarction associated with CABG.2
 
Criteria for Acute Myocardial Infarction
The term myocardial infarction should be used when there is evidence of myocardial necrosis in a clinical setting consistent with myocardial ischaemia. Under these conditions, any one of the following criteria meets the diagnosis for myocardial infarction:
  • Detection of rise and/or fall of cardiac biomarkers (preferably troponin) with at least one value above the 99th percentile of the upper reference limit (URL) together with evidence of myocardial ischaemia with at least one of the following:
    • Symptoms of ischaemia
    • ECG changes indicative of new ischaemia (new ST-T changes or new left bundle branch block [LBBB])
    • Development of pathological Q waves in the ECG
    • Imaging evidence of new loss of viable myocardium or new regional wall motion abnormality.
  • Sudden, unexpected cardiac death, involving cardiac arrest, often with symptoms suggestive of myocardial ischaemia, and accompanied by presumably new ST elevation, or new LBBB, and/or evidence of fresh thrombus by coronary angiography and/or at autopsy, but death occurring before blood samples could be obtained, or at a time before the appearance of cardiac biomarkers in the blood.
  • For percutaneous coronary interventions (PCI) in patients with normal baseline troponin values, elevations of cardiac biomarkers above the 99th percentile URL are indicative of peri-procedural myocardial necrosis. By convention, increases of biomarkers greater than 3 × 99th percentile, URL have been designated as defining PCI-related myocardial infarction. A subtype related to a documented stent thrombosis is recognized.
  • For coronary artery bypass grafting (CABG) in patients with normal baseline troponin values, elevations of cardiac biomarkers above the 99th percentile URL are indicative of peri-procedural myocardial necrosis. By convention, increases of biomarkers greater than 5 × 99th percentile URL plus either new pathological Q waves or new LBBB, or angiographically documented new graft or native coronary artery occlusion, or imaging evidence of new loss of viable myocardium have been designated as defining CABG-related myocardial infarction.
  • Pathological finding of an acute myocardial infarction.
 
CRITERIA FOR PRIOR MYOCARDIAL INFARCTION
Any one of the following criteria meets the diagnosis for prior myocardial infarction:
  • Development of new pathological Q waves with or without symptoms3
    zoom view
    4
  • Imaging evidence of a region or loss of viable myocardium that is thinned and fails to contract, in the absence of a non-ischaemic cause
  • Pathological findings of a healed or healing myocardial infarction.
 
ECG MANIFESTATIONS OF ACUTE MYOCARDIAL ISCHAEMIA(IN ABSENCE OF LVH AND LBBB)
 
ST Elevation
New ST elevation at the J-point in two contiguous leads with the cut-off points: ≥ 0.2 mV in men or ≥ 0.15 mV in women in leads V2-V3 and/or ≥ 0.1 mV in other leads.
 
ST Depression and T-wave Changes
New horizontal or down-sloping ST-depression ≥ 0.05 mV in two contiguous leads; and/or T inversion ≥ 0.1 mV in two contiguous leads with prominent R-wave or R/S ratio > 1.
 
THROMBOLYTIC (FIBRINOLYTIC) THERAPY
Fibrinolytic therapy revolutionized the management of STEMI in 1980s. The lifesaving capability of early fibrinolytic therapy has been well established beginning with GISSI I trial in 1986. Pooled data shows a relative reduction in mortality of 18% and an absolute reduction of nearly 2%. Even more dramatic Longterm mortality benefit may be the result of preservation of normal LV function. There are three generations of fibrinolytic agents:
a. First generation fibrinolytics:
Streptokinase, urokinase, APSAC (Antistreplase)
b. Second generation fibrinolytics: (Fibrin-specific)
Alteplase (rt-PA or recombinant tissue plasminogen activator), duteplase, saruplase
c. Third generation fibrinolytics: (Fibrin-specific)
Reteplase (r-PA), tenecteplase, (TNK-tPA), recombinant staphylokinase
 
INDICATION OF THROMBOLYSIS
 
Class I (Level of Evidence A) (Table 1.1)
• Chest pain consistent with angina within prior 12 hours
• ECG changes:
ST≥1 mm, ≥ 2 contiguous limb leads
ST≥2 mm,≥2 contiguous precordial leads
New left bundle branch block
• Absence of contraindications (vide algorithm).
5
Table 1.1   Applying classification of recommendations and level of evidence “Size of Treatment Effect”
Class I
Class IIa
Class IIb
Class III
“Estimate of Certainty (Precision) of Treatment Effect”
Benefit >>> Risk
Benefit >> Risk Additional studies with focused objectives needed
Benefit ≥ Risk Additional studies with broad objectives needed; additional registry data would be helpful
Risk ≥Benefit No additional studies needed
Procedure/treatment should be performed/administered
It is reasonable to perform procedure/ administer treatment
Procedure/treatment may be considered
Procedure/treatment should not be performed/administered since it is not helpful and may be harmful
Lavel A
Multiple (3–5) population risk strata evaluated* General consistency of direction and magnitude of effect
Recommendation that procedure or treatment is useful/effective Sufficient evidence from multiple randomized trials or meta-analyses
Recommendation in favour of treatment of procedure being useful/effective Some conflicting evidence from multiple randomized trails or metaanalyses
Recommendation's usefulness/efficacy less well established Greater conflicting evidence from multiple randomized trials or metaanalyses
Recommendation that procedure or treatment no useful/effective and may be harmful Sufficient evi-dence from multiple randomized trials or meta-analyses
Level B
Limited (2–3) population risk strata evaluated*
Recommendation that procedure or treatment is useful/effective Limited evidence from single randomized trial or non-randomized studies
Recommendation in favour of treatment or procedure being useful/effective Some conflicting evidence from single randomized trial or non-randomized studies
Recommendation's usefulness/efficacy less well established Greater conflicting evidence from single randomized trial or non-randomized studies
Recommendation that procedure or treatment not useful/effective and may be harmful Limited evidence from single randomized trial or non-randomized studies
Level C
Very limited (1–2) population risk strata evaluated*
Recommendation that procedure or treatment is useful effective
Recommendation in favour of treatment or procedure being useful/effective
Recommendation's usefulness/efficacy less well established
Recommendation that procedure or treatment no useful/effective and may be harmful
Only expert opinion, case studies, or standard-of-care should is
Only diverging expert opinion, case studies, or standard-of-care
Only diverging expert opinion, case studies, or standard-of-care
Only expert opinion, case studies, or standard-of care
Suggested phrases for writing recom-mendations
Recommended is indicated Is useful/effective/ beneficial
Is reasonable can be useful/effective/ beneficial Is probably recommended or indicated
May/might be considered may/might be Reasonable usefulness/effectiveness is unknown/unclear/uncertain or not well established
Is not recommended is not indicated should not Is not useful/effective/beneficial may be harmful
*Data available from clinical trials or registries about the usefulness/efficacy in different sub-populations, such as gender, age, history of diabetes, history of prior MI, history of heart failure, and prior aspirin use. A recommendation will Level of Evidence B or C does not imply that the recommendation is weak. Many important clinical questions addressed in the guidelines do not lend themselves to clinical trails even though randomized trials are not available, there may be a very clear clinical consensus that a particular test or therapy is useful or effective.
ACC/AHA Task Force on Practice Guidelines developed a list of suggested phrases to use when writing recommendations. All recommendations in this guideline have been written in full sentences that express a complete thought, such that a recommendation, even if separated and presented apart from the rest of the document (including headings above sets of recommendations), would still convey the full intent of the recommendation. It is hoped that this will increase readers' comprehension of the guidelines and will allow queries at the individual recommendation level.
6
 
Class IIa (Level of Evidence C)
In presence of chest pain (within prior 12 hours), ECG findings consistent with true posterior MI.
 
STATUS OF DIFFERENT THROMBOLYTIC AGENTS (TABLE 1.2)
Streptokinase:
Approved for general use.
Alteplase:
Established standard
Reteplase:
Approved for general use.
TNK-tPA:
Approved for general use; like to replace Alteplase because:
  1. Bolus injection simplifies administration even in pre-hospital setting and reduces potential for medication errors
  2. Increased fibrin specificity provided by TNK-tPA does confer a significant decrease in major systemic bleeding.
 
TIMING OF FIBRINOLYTIC THERAPY
Time from onset of symptoms to fibrinolytic therapy is an important predictor of MI size and patient outcome and efficacy diminishes with passage of time. Fibrinolytic therapy administrated within the first two hours (especially the first hour) can occasionally abort MI and dramatically reduce mortality.
Table 1.2   Comparison of thrombolytic agents
Property
SK
tPA
r-PA
TNK-tPA
SAK
Fibrin specificity
+ +
+
+ + +
+ + + +
Dose (most frequently used/tested)
1.5Mu/ 60min
100mg/ 90min
2×10 Mu bolus 30 min apart
0.5mg/ kg bolus
20-30mg/ 30min
Antigenic
+
+
Hypotension
+
Patency at 90 min
+
+ + +
+ + + +
+ + +
+ + + (+?)
Haemorrhagic stroke
+
+ +
+ +
+ +
?
Mortality reduction
+
+ + +
+ + +
+ + + (?)
+ + (?)
Cost
+
+ + +
+ + +
+ + + (?)
+ + (?)
Concomitant heparin
?
+
+
+
+
Bleeding (non-cerebral)
+ + +
++
+ +
+
?
SK = Streptokinase t-PA = Recombinant tissue − type plasminogen activator (alteplase), SAK = Recombinant staphylokinase TNK − tPA = Tenecteplase rPA = Reteplase.
7
zoom view
Management of patients with haemorrhagic complications following fibrinolytic therapy
The Task Force on management of AMI of ESC and ACC/AHA, both recommended a target of door (or medical contact) to needle time of 30 minutes.
 
ASSESSMENT OF REPERFUSION (NON-INVASIVE)
  • Relief of symptoms
  • Maintenance or restoration of haemodynamic and/or electrical stability
  • Reduction of atleast 50% of initial ST segment elevation injury pattern on a follow-up of ECG 60–90 minutes after initiation of therapy.
 
PRIMARY PCI
Basic Requisites:
  • Door-balloon time < 90 minutes, individual doing > 75 PCI/year, 11 Primary PCI for STEMI.
  • Lab performing > 200 PCI/yr, 36 primary PCI for STEMI.
 
Indications
  1. Symptom duration < 3 hours and door to balloon - door to needle is < 1 hr, primary PCI is preferable, otherwise thrombolysis with a fibrin specific agent is preferred.
    8
  2. Symptom duration > 3 hrs - primary PCI is generally preferred.
  3. Patients less than 75 years old, developing shock within 36 hours of MI and are suitable for revascularization that can be performed within 18 hours of shock.
  4. Severe CHF and/or pulmonary oedema (Killip class 3) and onset of symptoms within 12 hours.
 
Primary PCI is Reasonable in Following
  1. Patients of 75 years or older, develop shock within 36 hours of MI and are suitable for revascularization that can be performed within 18 hours of shock.
  2. 12–24 hours of symptom onset and one or more of the following:
    1. Severe CHF
    2. Haemodynamic or electrical instability
    3. Persistent ischaemic symptoms.
PCI-treated patients experience lower shortterm mortality rates, less non-fatal reinfarction and less haemorrhagic stroke than those treated by fibrinolysis but with an increased risk for major bleeding. PCI appears to have its greatest mortality benefit in high-risk patients. In patients with cardiogenic shock, an absolute 9% reduction in 30-days' mortality with coronary revascularization instead of immediate medical stabilization was reported in the SHOCK trial. In NRMI-II, patients with CHF and a 33% relative risk reduction with primary PCI compared with a 9% relative risk reduction with fibrinolytic therapy. Primary PCI in patients with anterior STEMI reduces mortality compared with fibrinolytic therapy, but there is no difference in patients with non-anterior STEMI. Findings from a recent meta-analysis suggest that for STEMI populations with a mortality risk < 4.5%, primary PCI is unlikely to improve 30 days mortality, relative to fibrinolytic therapy. Time from symptom onset to reperfusion is an important predictor of patient outcome. The mortality benefit with PCI exist when treatment is delayed by no more than 60 minutes. If the expected door-to-balloon time exceeds the expected door-to-needle time by more than 60 minutes, fibrinolytic treatment with a fibrin-specific agent should be considered unless it is contraindicated. This is particularly important when symptom duration is less than 3 hours but is less important with longer symptom duration, when less ischaemic myocardium can be salvaged. In both, the CAPTIM trial, which showed lower mortality with prehospital fibrinolysis than with primary PCI, and the PRAGUE-2 trial, which showed lower mortality with primary PCI after interhospital transfer than with on-site fibrinolysis. PCI was superior to fibrinolysis when symptom duration was greater than 2 to 3 hours but not when symptom duration was shorter. In the early hours of STEMI, prompt fibrinolytic therapy can decrease infarct size and the risk of developing cardiogenic shock.9
Potential complications of an invasive strategy for treating STEMI include problems with the arterial access site; adverse reactions to volume loading, contrast medium, and antithrombotic medications, technical complications, and reperfusion events. Reocclusion occurs in 10 to 15% of patients after PTCA but in fewer than 5% after stent implantation. Likewise, angiographic restenosis occurs in 30 to 40% of patients after PTCA but in 15 to 20% after stent implantation.
Primary PCI might be considered in hospitals without on-site cardiac surgery, provided that a proven plan for rapid transport to a cardiac surgery operating room exists in a nearby hospital with appropriate haemodynamic support capability for transfer.
 
INTERHOSPITAL TRANSFER FOR PRIMARY PCI
To achieve optimal results, time from the first hospital door to the balloon inflation in the second hospital should be as short as possible, with a goal of less than 90 minutes. In case of delay in door-to-balloon time versus door-to-needle time of more than 60 minutes, immediate intravenous fibrinolysis with a fibrin-specific agent is preferred over interhospital transfer for primary PCI especially with patients presenting early (< 3 hours).
 
TIME TO PCI DOES MATTER
  • Door-to-balloon (DBT): “SHORTER IS BETTER”
  • Optimal quality target DBT < 90 minutes
  • STEMI Guidelines of ESC presented at ESC Congress in Munich 2008 has extended the first medical contact (FMC) to balloon time to < 2 hours in all cases except in those presenting < 2 hours after pain onset where FMC to balloon time remains 90 minutes.
  • Time has major impact in
    • High risk patients
    • Patients presenting < 3 hours
  • Time is a lesser factor for presentations > 3–4 hours and in such situations, PCI remains the preferrable treatment even if anticipated DBT > 90 minutes.
 
RECOMMENDATIONS TO IMPROVE TRANSFER DELAYS AND REPERFUSION TIME
 
Pre-Hospital ECG
  1. Direct transfer to PCI centre (bypassing closest hospital).
  2. EMS (Emergency Medical Service) authority to mobilize catheteri-zation laboratory.
10
 
Hospital without Percutaneous Coronary Intervention Capacity
  1. Ambulance remains at hospital, patient not taken off stretcher.
  2. Avoid IV infusion pumps (delays in switching equipment).
  3. Authority to transfer patient to tertiary care centre without delay of speaking to a cardiologist.
 
Tertiary Care Centre
  1. Direct transfer to catheterization laboratory (bypassing emergency, residents, etc.)
  2. Phone call to mobilize team, have paient pre-registered, prepare catheterization laboratory supplies.
  3. Expedited patient preparation.
  4. Engage suspected infarct artery with guiding catheter and prompt reperfusion.
 
Real-world Challenges in Implementing Primary Pci Programmes
  • Availability of skilled PCI labs: what constitutes safe minimum volume for a PCI-capable centre?
  • Transportation delays: National Registry of Myocardial Infarction (NRMI) data from USA indicate that only 4.2% of patients transferred for PCI meet recommended ≤ 90 min DBT time
  • Impact of time of day and day of the week on timely initiation of therapy
  • Availability of hospital beds in populated countries like India
  • Lack of an organized plan for interfacility transfer in developing countries like India
  • Ready availability of fund and problem of reimbursement strategies (insurance, etc.) at moments when every minute counts
  • Issues related to staff availability for 24/7 PCI coverage.
 
TIME TO REPERFUSION: TIME IS MUSCLE
Extent of Myocardial salvage and mortality reduction show best results when reperfusion is achieved within first 3 hours. Although opening up the infarct related artery (IRA) beyond first few hours may not be very productive in terms of myocardial salvage, yet mortality rates after AMI appear to be inversely related to ability to achieve TIMI-3 flow. The greatest benefit of primary PCI may be its ability to achieve TIMI-3 flow in more than 90% of patients, even when the patients are treated in late stages of infarction. In contrast, thrombolytic therapy has a marked decrease in thrombolytic efficacy in patients treated more than a few hours after symptom onset.11
Improvement in ejection fraction (EF) from baseline to 6 months was substantial when reperfusion was achieved < 2 hours from symptom onset, but was modest and relatively independent of time to reperfusion after 2 hours. There were no differences in 1 or 6 months mortality by time to reperfusion by PCI. There were also no differences in other clinical outcomes by time to reperfusion by PCI, except that re-infarction and infarct artery reocclusion at 6 months were more frequent with later reperfusion. This is different from lytic therapy, where time to reperfusion is inversely related to outcome.
Justifying the aphorism “TIME IS MUSCLE”, a multivariate analysis by O'Neill (2005) demonstrated that prolonged door-to-balloon (P < 0.0001) and pain onset to-door time (P = 0.025) remained independent predictors of final infarct size. However, adding to the controversy, in patients with late presentation, an RCT study (Schomig et al 2005) demonstrated that in patients with AMI without persisting symptoms presenting 12–48 hours after symptom onset, primary PCI reduces infarct size compared with usual care. This suggests that although reperfusion time does matter for primary PCI, the window may be extended beyond 12 hours.
 
PRIMARY PCI VS FIBRINOLYTIC THERAPY
Primary PCI
Thrombolysis
Defined as intervention in the culprit vessel within 12h after the onset of chest pain or other symptoms, with- out prior (full or concomitant) thrombolytic or other clot- dissolving therapy.
Thrombolytic treatment is a valid option if primary PCI cannot be delivered in a timely manner. If the patient presents within first 3 hours, thrombolytic therapy can provide results comparable to primary PCI.
Advantages:
Advantages:
• High rate of reperfusion success and improved clinical outcomes.
• Limited contraindications
• Early risk stratification made possi-ble by angiography
• Widespread availability
• Ease of use
• Rapid administration
• Operator independent
Disadvantages:
Disadvantages:
• Lack of ready availability and resources (Even in US, < 25% have facilities for primary PCI and even fewer have 24 hours availability; however, greater percentage are located within a 90 minute transfer time
• Operator dependent
• Failure to open IRA in 20%
• Failure to provide complete reperfusion in additional 30%
• Reocclusion in 20% when not followed by PCI
• Risk of ICH is 1–2% (substantially higher in elderly)
12
Rescue (also known as salvage) PCI is defined as PCI within 12 hours after failed fibrinolysis for patients with continuing or recurrent myocardial ischaemia. Rescue PCI has resulted in higher rates of early infarct artery patency, improved regional infarct-zone wall motion, and greater freedom from adverse in-hospital events than with a deferred PCI strategy or medical therapy. A major problem in adopting a strategy of rescue PCI lies in the limitation of accurate identification of patients for whom fibrinolytic therapy has not restored antegrade coronary flow. Unless unsuccessful fibrinolysis is recognized and corrected quickly (within 3 to 6 hours of onset of symptoms) salvage of ischaemic myocardium is unlikely. Unfortunately, clinical markers of reperfusion, such as relief of ischaemic type chest discomfort, partial resolution of ST-segment elevation, and reperfusion arrhythmias, have limited predictive value in identifying failure of fibrinolysis. In a prior era, in which the practice of PCI was less mature, immediate catheterization of all patients after fibrinolytic therapy to identify those with an occluded infarct artery was found to be impractical, costly, often associated with bleeding complications. This strategy is being re-evaluated in clinical trials testing facilitated PCI in the contemporary PCI setting.
Among patients who fail fibrinolytic therapy, rescue PCI was associated with consistent improvements in clinical outcomes despite time delays from symptom onset to treatment that ranged from 4.3 to 6.9 hours as was shown by two recent major trials of rescue PCI (MERLIN and REACT). Primary composite end point in these trials included death, reinfarction, stroke or severe heart failure at any time within six months. Benefits of rescue PCI were obtained in MERLIN and REACT despite inclusion of patients in these trials that required transfer to institutions with interventional capacities. Medium transfer time was only 85 min in REACT. This supports the fact that improved systems are urgently needed if full benefits of rescue PCI are to be realized in transferred patients. Most appropriate adjunctive pharmacotherapy in patients undergoing rescue PCI also needs to be outlined for improving outcomes. Unsuccessful rescue PCI is associated with a high mortality rate.
In contrast, repeat fibrinolytic therapy was not associated with significant improvements in all-cause mortality or reinfarction but was associated with an increased risk for minor bleeding.
A 3% increase in absolute risk of stroke was observed with rescue PCI and contrary to expectations, majority of strokes in MERLIN trial were thromboembolic rather than haemorrhagic. There was a 13% increase in risk of minor bleeding with rescue PCI but there was no excess of major bleeding.
ABORTED MI: A new target for reperfusion therapy: Treatment within an hour after symptom onset may result in 25% of aborted infarction and is in combination with complete (70%) ST-segment resolution (without 13enzyme rise). Aborted MI is associated with better survival and may become a new end-point in clinical trials of STEMI.
 
EVOLVING STRATEGIES TO MEET REAL-WORLD CHALLENGES
  1. Pre-Hospital Thrombolysis: Prehospital fibrinolysis is reasonable in those setting in which physicians or fully trained paramedics are present in the ambulance or prehospital transport times are more than 60 minutes.
    This has been successfully practised in European countries. The concept of prehospital thrombolysis appears particularly relevant to the developing countries with few tertiary care centres, predominantly rural population and patients travelling long distances to avail of medical facility. These circumstances warrant an urgent need for prehospital thrombolysis, however, this needs tremendous infrastructure and a coordinated programme by government or private sector. If the transport to a medical facility is likely to be time consuming, a competent physician after explaining the benefits and life saving nature can administer thrombolytic therapy in pre-hospital phase. The CAPTIM (comparison of angioplasty and pre-hospital thrombolysis in acute myocardial infarction) trial randomized early (< 2 hours), there was a strong trend for lower mortality with pre-hospital fibrinolytic therapy (2.2% vs 5.7%, p = 0.053), while in patients randomized later (> 2 hours) there was no benefit to early fibrinolytic therapy, suggesting that time delays are very important early after the onset of symptoms but are less important later.
    Using data from a national registry, investigators in France reported 1-year mortality from STEMI of 6% in patients receiving prehospital fibrinolysis compared with 11% in patients receiving inhospital fibrinolysis or primary PCI.
    Bjorklund et al reported similar data from Swedish Register of cardiac intensive care on patients admitted to 75 Swedish hospitals in 2001–04. When compared with regular in-hospital thrombolysis, pre-hospital diagnosis and thrombolysis with trained paramedics in the ambulances are associated with reduced time to thrombolysis by almost 1 hour and reduced adjusted 1 year mortality by 30% in real-life STEMI patients.
    Requisites for Pre-hospital Thrombolysis (PHT)
    • Critical care ambulances staffed with physicians (ideally or paramedics trained to send pre-hospital ECG to corresponding hospital's CCUs using telemedicines (appropriate ECG machines are connected to Laptop computer with internet connection and the paramedic discusses inclusion and exclusion criteria over phone with physician-on-call).
      14
    • Improving public awareness about value of time to treatment after onset of chest pain (“TIME IS MUSCLE”)
    • Emergency dial numbers for hospitals pertaining to a locality.
  2. Facilitated PCI: Facilitated PCI refers to a strategy of planned immediate PCI after an initial pharmacological regimen such as fulldose fibrinolysis or half-dose fibrinolysis or a GP IIb/IIIa inhibitor, or a combination or reduced-dose fibrinolytic therapy and a platelet GP IIb/IIIa inhibitor.
    The benefit of a facilitated PCI approach depends in part on the impact that treatment delays with primary PCI have on clinical outcomes. This has been a controversial subject, but observational data suggest that incremental treatment delays in performing primary PCI for STEMI which occur in the first 2–3 hours after the onset of symptoms are critical and can greatly compromise outcomes. After 2–3 hours, incremental treatment delays with primary PCI appear to have less effect on outcomes. This is probably related to the fact that there is a window of opportunity for myocardial salvage within the first two hours after the onset of symptoms, after which the opportunity for salvage is very modest. The benefits of very early reperfusion are related to myocardial salvage, and this is a very time dependent process. The benefits of later reperfusion are more related to the effects of an open artery in preventing remodelling and promoting electrical stability, and these effects are less time dependent.
    Potential advantages of facilitated PCI include earlier time to reperfusion, improved patient stability, greater procedural success rates, higher TIMI flow rates, and improved survival rates. However, preliminary studies (PACT, BRAVE, ON-TIME, GRACIA-2) have not demonstrated any benefit in reducing infarct size or improving outcomes.
The ASSENT-4 PCI trial randomized 1667 patients to PCI with or without full-dose TNK with a primary endpoint of 90 days death, cardiogenic shock or congestive heart failure. The TNK plus PCI patient had significantly higher rates of repeated myocardial infarction (p = 0.0279), repeat target vessel revascularization (p = 0.0041), stroke (p< 0.0001) and, in the primary trial, end point of death, congestive heart failure or shock (p = 0.0045). There are several hypotheses to explain this failure such as:
  1. Given the concern of major bleeding, activation of platelets by thrombolysis was not covered by intravenous GPIIb/IIIa antagonists.
  2. The fact that only a minority of patients were enrolled in the pre-hospital or ambulance setting or in the first 2 hours, i.e. when and where facilitated PCI would be expected to yield its greatest benefits.
A recent meta-analysis of RCTs by Keeley and Grines to compare facilitated and primary PCI also showed higher event rates (death, non-fatal MI, TVR rates) in facilitated PCI arm.15
Following this, a silver lining for facilitated PCI has been the results of Canadian WEST trial (2006). This was a pilot randomized trial of approximately 300 patients with STEMI, comparing three strategies: pre-hospital lysis and usual care, prehospital lysis and provisional rescue PCI in case of failure to achieve ST segment resolution and mandatory invasive management within 24 hours in other cases and primary PCI. There are two interesting features to this trial, which makes it complementary to ASSENT-4 PCI: the average time from symptom to randomization was short (approximately within 2 hours) and patients received state of the art antithrombotic therapy (with patients undergoing PCI after lytics receiving clopidogrel and in many instances GPIIb/IIIa blockers). Finally, PCI was performed early but not immediately after lysis, which makes it logistically convenient and gives ample time for potent antithrombotic therapy. Interestingly, mortality was lowest and very similar in the two groups which underwent intervention: lytic facilitated PCI and primary PCI (1%). Thus, WEST trial suggests that there may be room for facilitated PCI, provided lysis can be implemented very early and PCI can be preceded by state-of-the-art anti-thrombotic and antiplatelet therapy. Further information about future of facilitated PCI have become available from results of FINESSE and CARESS IN AMI trials which were recently presented at the ESC Congress, held in Vienna in September 2007. FINESSE had enrolled 2452 patients in various centres in Europe and USA. Mean age was 60 years and 65% of patients had high risk MI and 46% had anterior wall MI. Symptom duration was 20 minutes to 6 hours. Exclusion criteria included situations when angiography was expected to be done before 60 minutes or after 4 hours of presentation. Patients were randomized to three arms (i) Primary PCI (with in-lab abciximab) (ii) Facilitated PCI following abciximab started upfront (iii) Facilitated PCI following half dose reteplase and abciximab. No significant improvement of primary end points were noted in any group (death, cardiogenic shock, readmission for heart failure). Pre-PCI TIMI3 flow was best in the reteplase group but post-PCI TIMI3 flow was same in all the groups. Major bleeding and intra-cerebral bleeding was higher in the reteplase group but did not reach statistical significance. The study investigators concluded that primary PCI (with in-lab abciximab) within 4 hours of medical contact is more beneficial than facilitated abciximab or facilitated reteplase (half dose + abciximab groups). Reasons cited for failure of facilitated arms were that probably wrong study population (up to 6 hours of symptom onset) was chosen, no upfront clopidogrel was used routinely (FINESSE was a pre-CLARITY trial) and enoxaparin was used only in a subset of patients. The investigators concluded that the case for “facilitated PCI” (or preferably it should be called “Pharmaco-invasive PCI”) is still probably valid if (i) presentation is within 2–3 hours of symptom onset (ii) anticipated long delay to primary PCI is present (iii) large viable 16myocardium is present (iv) pre-PCI TIMI3 flow is already present. Such speculations have been kept alive by reports of Thiele et al (2006) and PATCAR Pilot trial (2007). In the latter trial, the group which received pre-hospital reteplase followed by urgent PCI had initial IRA TIMI flow grades of 2 to 3 in 82% and full reperfusion (TIMI angiographic perfusion score ≥ 20) in 67% compared to 37 and 22% respectively in the group which received primary PCI. Further evaluation of the strategy of pre-hospital fibrinolytic acceleration of STEMI treatment coupled with urgent PCI (FAST-PCI) in a large multicentre randomized trial multicentre randomized trial compared with primary PCI seems warranted.
CARESS IN AMI enrolled 1650 patients from seven countries in Europe between 2005–07. STEMI patients presenting before 12 hours from symptom onset and less than 75 years old with ST-elevation/new onset LBBB along with chest pain were admitted to centres without PCI facilities. Patients were in “high-risk” category (Killip class > 2). Mean age was 60 years. All patients were given aspirin, reteplase, unfractionated heparin and were randomized to either facilitated PCI or medical therapy (±Rescue PCI if needed). Abciximab and Clopidogrel were given in-lab. Median time from pain onset to reteplase therapy was 170 minutes and from reteplase to coronary angiography was 136 minutes in the facilitated group and 212 minutes in the rescue group. Incidence of death, re-infarction and refractory ischaemia was 11.1% in medical rescue PCI group and 4% in facilitated PCI group. Bleeding however, was higher in the facilitated group. Thus, this trial is the fourth one (after SIAM III, GRACIA-1 and CAPITAL-AMI) which confirms ESG guidelines for early angioplasty after lysis, i.e. “high-risk” STEMI patients should be immediately transferred for PCI after lytic therapy. TRANSFER- AMI trial presented at ACC'08 confirmed the superiority of routine PCI within 6 hours following conventional TNK-tPA and upfront clopidogrel compared to standard lytic therapy. 12-month follow-up of CARESS-AMI presented at ESC'08 showed persistent significant benefit in the PCI arm for refractory ischaemia and recurrent Ml but no difference in terms of death and admission for heart failure. More support in this regard has come from very recent GRACIA-2 and FAST-MI trials. STEMI guidelines presented at the same conference has accorded Class IIa status to PCI after successful lysis within 3–24 hours of lytic therapy initiation. There is no similar recommendation from ACC/AHA.
 
OPEN ARTERY HYPOTHESIS
Survivors of STEMI with a persistently occluded artery after fibrinolysis, PCI or no reperfusion therapy have:
  1. Increased LV dilatation and poorer LV function.
    17
  2. Greater incidence of spontaneous and inducible ventricular arrhythmias.
  3. Poorer prognosis.
It has been hypothesized that infarct related artery (IRA) patency may favourably influence LV remodelling and electrical stability, even if accomplished at a time when salvage of ischaemic myocardium (i.e. more than 12 hours to days after coronary artery occlusion). Most studies, however, showed no significant benefit.
The Occluded Artery Trial (OAT) was undertaken randomizing patients to test whether routine PCI days to weeks after MI improves Longterm clinical outcomes in asymptomatic high-risk patients with an occluded IRA. The OAT investigators (Hochman et al 2006) published their report in over 2000 high risk stable patients with total infarct related coronary occlusion 3–28 days post MI (Median 8 days). In this open label randomized trial of PCI, usually employing bare metal stenting, a high rate of initial procedural success with good 1 year patency (among a subset) was achieved. Despite successful mechanical intervention, the expected decline in death, MI and heart failure did not occur. In fact, the reverse tended to be true. Hence, there was a statistically greater incidence of fatal and nonfatal MI in the intervention vs medical group as ascertained by investigators.
Explanations forwarded to explain the negative impact of PCI in occluded IRAs were:
  1. Periprocedural MI associated with branch occlusion and distal embolization.
  2. Since much of the healing and remodelling after MI occurs within the first week, a quicker intervention might have yielded a different result.
  3. Improved outcomes with modern day medical therapy (aspirin, betablockers, ACEIs, statins).
 
“OPEN ARTERY HYPOTHESIS”: WHERE DO WE STAND?
Some issues pertaining to OAT deserve further exploration and these are:
  1. It is impossible to know from current study whether persisting occlusion vs reperfusion followed by reocclusion may have occurred.
  2. The question of whether persisting myocardial viability would signal the potential for benefit vs harm with late mechanical intervention and the need for viability studies before PCI (e.g. stress thallium, stress echo, etc.) should be addressed.
A recently published (2008) meta-analysis of randomized controlled trials of 3,560 patients undergoing late PCI (> 12 hours from AMI onset) [Abbate et al.] showed that PCI of infarct related artery performed late (12 hours to 60 days) after AMI is associated with significant improvement in cardiac function and survival making the “Open Artery Hypothesis” a still valid one within 3 months from AMI.18
Table 1.3   ACC/AHA/SCAI and ESC guidelines for patients after successful fibrinolysis and for patients not undergoing primary reperfusion (Latecomers)
Clinical condition
ACC/AHA/SCAI
ESC
After successful fibrinolysis (upto 24 hours) independent of angina and/or ischaemia
No recommendation
Routine angiography and PCI if applicable (Class I, Level of Evidence: A)
Patients not undergoing reperfusion (latecomers) or after fibrinolysis with residual moderate to severe ischaemia
PCI if applicable (Class I, Level of Evidence: B)
PCI if applicable; (Class I, Level of Evidence: B)
Latecomers (12 to 24 hours) asymptomatic and stable
No recommendation
Class IIb
Latecomers (> 24 hours) with haemodynamically significant stenosis in a patient IRA
PCI may be considered part of an invasive strategy (Class IIb, Level of Evidence: B)
No recommendation
Latecomers (> 24 hours) asymptomatic and stable with totally occluded IRA, without evidence of ischaemia
PCI not recommended (Class III, Level of Evidence: B)
No recommendation
Patients symptomatic for angina or heart failure and those with residual ischaemia or documented viability are more likely to benefit from late PCI at a Longterm follow up. Patients with uncomplicated AMI, especially with reduced life-expectancy, however may not benefit from routine PCI. Current recommendations from ACC/AHA/SCAI and ESC for late PCI after AMI are shown in Table 1.3.
 
ADJUNCTIVE ANTITHROMBOTIC THERAPY
  1. Unfractionated Heparin (UFH)
    • Catalyzes the action of anti-thrombin, which inactivates factor IIa (thrombin), factor IXa and factor Xa
    • Dose varies according to different reperfusion strategies and adjuvant therapies
    • For both tPA and TNK, UFH (intravenous) has class I recommendation although evidence is not strong.
      Dose: IV bolus of 60 u/Kg (maximum 4000u) followed by infusion of 12 u/Kg/hour (maximum 1000u/hr) Target APTT: 50–70 seconds.19
      Duration of treatment: 48 hours followed by gradual cessation of therapy; continuation beyond 48 hours should be individualized based on risk of pulmonary and systemic embolization and other factors.
    • For streptokinase, support for use of IV UFH is less certain and there appears to be no obvious advantage over that provided by subcutaneous heparin
      STEMI guidelines, 2008 presented at ESC Congress, Munich has accorded Class Ila recommendation to heparin for use following streptokinase.
    • Platelet counts should be monitored daily
    • Initial anticoagulation with UFH should be followed by warfarin in patients at high risk for systemic embolism (large or anterior MI, atrial fibrillation, previous embolus or known LV thrombus).
  2. Low Molecular Weight Heparin (LMWH)
    [Enoxaparin and Dalteparin]
    • Binds preferentially to factor Xa rather than to thrombin
    • ASSENT-3 trial, in which patients received tenecteplase and either UFH (bolus 60 u/Kg followed by IV infusion of 12u/kg/hour for 48 hours) or enoxaparin (bolus 30 mg IV, subcutaneous injection 1.0 mg/kg every 12 hours; duration of treatment equals duration of hospital stay). Each of the elements of composite end point of 30 days mortality, inhospital reinfarction or inhospital recurrent ischaemia were reduced with enoxaparin treatment. However, there were increases in total stroke and intracranial haemorrhage seen in patients > 75 years of age.
    • In EXTRACT-TIMI 25 trial, over 20,000 patients with STEMI who were scheduled to undergo fibrinolysis with a variety of agents (tPA and analogues, streptokinase) received either enoxaparin throughout index hospitalization or UFH for atleast 48 hours. Patients with creatinine clearance less than 30 ml/minute had one dose of enoxaparin every 24 hours (versus a 12 hours regimen) and patients over 75 years did not receive 30 mg IV bolus and their subcutaneous dose was reduced to 0.75 mg/kg every 12 hours. The composite of death, nonfatal reinfarction or urgent revascularization occurred in 14.5% of patients given UFH and 11.7% of those given enoxaparin (P < 0.001); major bleeding occurred in 1.4 and 2.1% respectively (P < 0.001). The composite of death, non-fatal reinfarction or non-fatal intracranial haemorrhage (a measure of net clinical benefit) occurred in 12.2% of patients given UFH and 10.1% of those given enoxaparin.
    • Thus, LMWH may be considered an acceptable alternative to UFH for patients aged less than 75 years who are receiving fibrinolytic 20therapy, provided that significant renal dysfunction (serum creatinine greater than 2.5 mg/dL in men or 2.0 mg/dL in women) is not present.
      Dose: Enoxaparin 30 mg IV bolus followed by 1.0 mg/kg SC every 12 hours until hospital discharge.
    • STEMI Guidelines, 2008 presented at ESC Congress, Munich has given Class I recommendation to Enoxaparin for use with tpA and analogues and Class Ila recommendation for use with streptokinase.
    • It is reasonable that STEMI patients not undergoing reperfusion therapy, who do not have a contraindication to anticoagulation be treated with intravenous or subcutaneous LMWH for at least 48 hours.
    • TETAMI trial investigated the efficacy and safety of treatment with enoxaparin or tirofiban alone or in combination for 2 to 8 days in patients with AMI who were not eligible for early reperfusion therapy. The trial failed to demonstrate any benefit with either agent alone or in combination compared to placebo.
 
DIRECT THROMBIN INHIBITORS (DTIs)
  • Bind directly to thrombin, both circulating and bound forms.
  • Promise of DTIs emerged from phase II trials, e.g. HERO suggesting that the outcome of STEMI patients treated with steptokinase and bivalirudin as compared with UFH was associated with enhanced coronary reperfusion and no excess in systemic bleeding.
  • HERO II study compared bivalirudin and unfractionated heparin (UFH) in patients receiving streptokinase for STEMI within 6 hours of symptom onset was undertaken. Although a clear reduction in the rate of reinfarction was evident in patients treated with bivalirudin, there was a tendency toward excess systemic and intracranial bleeding and no reduction in mortality.
  • Hence, bivalirudin's current role in STEMI appears to be as an alternative when UFH is contraindicated as in case of Heparin-induced thrombocytopenia. However, HORIZONS-AMI trial presented at TCT'07 demonstrated the superior efficacy and safety of Bivalirudin mono-therapy in STEMI patients undergoing primary PCI (vide Chapter 10).
However in view of nearly four times higher incidence of acute stent thrombosis in bivalirudin arm (provisional GPIIb/IIIa inhibitor use) in contrast to UFH + GPIIb/IIIa inhibitor arm, STEMI Guidelines (ESC 2008) has granted only Class Ila recommendation for use of bivalirudin as an adjunct to primary PCI.21
 
FACTOR Xa INHIBITOR (SYNTHETIC PENTASACCHARIDE)
  • Binding site of heparin to antithrombin consists of five sugar molecules which have become the basis for the creation of the synthetic pentasaccharides, of which fondaparinux is the first one of the extensively studied.
  • OASIS-6 trial evaluated the impact of fondaparinux in 12,092 patients with STEMI in preventing the primary and composite outcome of death or reinfarction at 30 days. Patients were randomized to a double blind comparison of fondaparinux 2.5 mg once daily or control. The results showed that rates of death or reinfarction at 30 days was significantly reduced from 11.2% in the control group to 9.7% in the fondaparinux group (p = 0.008); significant benefits were observed in those receiving thrombolytic therapy and those not receiving any reperfusion therapy. However, there was no benefit in those undergoing PCI (7 vs 6% mortality in both groups). STEMI Guidelines (ESC 2008) has accorded Class Ila recommendation to Fondaparinux for use with lytics but has given Class III recommendation for use in primary PCI.
 
GLYCOPROTEIN IIb/IIIa INHIBITORS
Fuelled by the success of GPIIb/IIIa inhibitors in NSTEMI, efficacy of abciximab in patients with STEMI was tested in ADMIRAL and CADILLAC trials. In ADMIRAL trial, 300 patients were randomized to have abciximab or placebo in the ambulance, in the emergency room or cardiac catheterization laboratory. The composite end-point for death, re-infarction and urgent target vessel revascularization (TVR) was 7.7% in the stent plus abciximab group and 14.6% in the stent plus placebo group (P=0.004). This benefit was maintained at 6 months (8.0 vs 15.9%) respectively, P=0.02). The better outcomes were related to improvements of TIMI-3 flow before the procedure and a better LV ejection at 24 hours.
These results were not reproduced by the larger CADILLAC trial. In this trial, composite of death, reinfarction, disabling stroke and ischaemia-driven TVR was similar in patients treated with stents with or without abciximab (9.5 vs 10.4%, P-NS). Some observers speculate that the strategy of GPI randomization after angiography, the late administration of abciximab in the CADILLAC trial may have blunted the effect of GPI on stented patients.
A recent meta-analysis of the studies involving GPIs in patients with STEMI (11 trials, involving 27,115 patients) demonstrated that when compared with the control group, abciximab was associated with a significant reduction in short-term (30 days) mortality (2.4 vs 3.4%, P = 0.047) and Longterm (6–12 months) mortality (4.4 vs 6.2%, P = 0.01). This was observed in patients undergoing primary PCI but not in those treated with fibrinolysis or in all trials combined.22
zoom view
STEMI Reperfusion Guidelines
  • (*) Prolonged transport is anticipated
  • (**) PCI is reliably available within 60 minutes of time to fibrinolysis
  • (***) As evidenced by SIAMIII, GRACIA-1, CAPITAL-AMI and CARESS-INAMI in high risk subset of AMI patients.
Abciximab was associated with a significant reduction in 30 day re-infarction, both in all trials combined (2.1 vs 3.3%, P<0.001), in primary angioplasty (1 vs 1.9%, P=0.03) and in fibrinolysis trials (2.3 vs 3.6% P<0.001). Abiciximab did not result in an increased risk of intracranial bleeding (0.61 vs 0.62%, P = 0.62) overall, but was associated with an increased risk of major bleeding complications when combined with fibrinolysis (5.2 vs 3.1%, P<0.001).23
zoom view
Evidence-based approach to need for Coronary Angiography (CAG) and revascularization
* 5–7 days post-MI
** 14–21 days post-MI
Inadequate data exist regarding use of small molecular weight GPIIb/IIIa inhibitor agents in AMI, hence use of epitfibatide or tirofiban for STEMI is not advocated at present.24
zoom view
ONTIME-2 trial presented at ESC Congress 2008 (Munich) where higher dose of Tirofiban was used in the ambulance prior to primary PCI, demonstrated higher TIMI 3 flow and ST-segment resolution with decreased MACE and mortality. Earlier administration (< 75 minutes) produced better results. STEMI Guidelines (ESC 2008) has given Class IIa recommendation to Abciximab and Class IIb to Eptifibatide and Tirofiban for use in primary PCI. However, the guidelines did not have ONTIME-2 results at the time they were drafted.25
zoom view
26
 
OTHER COMPLICATIONS OF ACUTE MYOCARDIAL INFARCTION
  1. Ventricular Septal Rupture (VSR):
    Incidence: 1–2% in prethrombolytic era, now dramatically reduced.
    Time of occurrence: Usually 2–5 days after MI
    Therapy: Surgery, Percutaneous Closure
  2. Mitral regurgitation (MR):
    Incidence: Mild to moderate MR, 13–45%.
    Severe MR leading to Cardiogenic shock: 1%
    Time of occurrence: 2–7 days after MI.
    Therapy: Surgical repair or less commonly replacement of mitral valve. PCI has no role.
  3. Cardiac rupture:
    Incidence: 3% of post-MI patients.
    Time of occurrence: 50% occur in first 5 days; 90% occur within 2 weeks.
    Therapy: Immediate pericardiocentesis in case of subacute rupture; Acute rupture is often immediately fatal.
  4. Pseudoaneurysm (Contained rupture):
    Communicate with body of left ventricle through a narrow neck. May be clinically silent; diagnosed by Echo/CT/MRI.
    Therapy: Surgical resection because spontaneous rupture may occur
  5. Ventricular aneurysm:
    1. Acute aneurysm: Occur with transmural anteroapical infarcts and expand during systole. May result in severe heart failure or even cardiogenic shock.
    2. Chronic aneurysm: Are those which persist more than 6 weeks. They are less compliant and rarely expand during systole.
      Therapy: Anticoagulation if a mural thrombus is demonstrated surgical therapy (Aneurysmorrhaphy) in patients with refractory heart failure and ventricular arrhythmias.
      PCI after 12 hours of MI but before 24 hours of MI in patients not reperfused earlier may be beneficial in those with acute aneurysm.
  6. Dynamic LVOT obstruction:
    Uncommon complication after acute anterior MI resulting from hyperkinesis of basal and mid segments of LV. Resultant venturi effect cause LVOT obstruction and MR. Free wall rupture can occur.
    Therapy: Slow addition of beta-blockers, small boluses of I.V. normal saline may reduce LVOT gradient.
  7. Pericarditis:
    1. Early pericarditis:
      Incidence: 10% (approx)
      Time of occurrence: 24–96 hours after MI.
      Therapy: Aspirin 650 mg every 4–6 hours. Avoid NSAIDs and corticosteroids Colchicine for recurrent pericarditis.
      27
    2. Late pericarditis or Dressler's syndrome:
      Incidence: 1–3%
      Time of occurrence: 1–8 weeks.
      Therapy: Aspirin If > 4 weeks have elapsed after MI, NSAIDs and even steroids may be started for severe symptoms.
zoom view
Management of patients with postinfarction mechanical complications
 
SECONDARY PREVENTION AFTER STEMI
  • Smoking: Complete cessation and also avoid second hand smoke
  • Physical activity: Minimum goal: 30 minutes 3–4 days per week; optimal: daily
  • Weight management: Goal: BMI 18.5–24.9 kg/m2
  • Blood pressure control: Goal-less than 140/90 mmHg or less than 130/80 mmHg if chronic kidney disease or diabetes
  • Diabetes management: Goal: HbA1C < 7%
  • Lipid management: Primary goal: LDL-C < 100 mg/dl Non-HDL-C < 130 mg/dl
    28
  • Antiplatelet agents or anticoagulants: Start and continue indefinitely aspirin 75 to 162 mg/day if not contraindicated. Consider clopidogrel 75 mg/day or warfarin if aspirin is contraindicated. Manage warfarin to INR of 2.5 to 3.5 in post-STEMI patients when clinically indicated or for those not able to take aspirin or clopidogrel.
  • Renin-angiotensin-aldosterone system:
ACE-inhibitors: An ACE-inhibitor should be prescribed at discharge for all patients without contraindications after STEMI; start early in stable high risk patients (anterior MI, prevents MI, Killip Class > II (S3 gallop, rales, radiographic CHF, LVEF < 0.40).
Angiotensin receptor blocker indicated in patients who are intolerant of ACE inhibitors and with either clinical or radiologic signs of heart failure or LVEF < 0.40 (Candesartan and Valsartan have established efficacy)
Aldosterone blockade: in post-STEMI patients with serum cretinine 2.5 mg/dl in men or < 2.0 mg/dl in women or potassium < 5.0 mEq/l, already on ACEIs, LVEF < 0.40 and have either symptomatic heart failure or diabetes.
Beta-blocker: Start in all patients, continue indefinitely. Observe usual contraindications.
SUGGESTED READING
  1. Abbate A, Giuseppe GL, Biondi-Zoccai GL et al. Survival and Cardiac Remodelling benefits in patients undergoing late percutaneous coronary intervention of the infarct-related artery. J. Am Coll Cardiol 2008;51:956–64.
  1. ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction-executive summary: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 2004;110:588–636.
  1. Becker RC, Burns M, Gore JM, for the NRMI-2 Investigators. Early assessment and in-hospital management of patients with acute myocardial infarction at increased risk for adverse infarction: A nationwide perspective of current clinical practice. Am Heart J 1998;135:786–96.
  1. Gershlick AH, Stephens-Llyod A, Hughes S, et al. Rescue angioplasty after failed thrombolytic therapy for acute myocardial infarction N Engl J Med 2005;353(26):2758–68.
  1. Huber K, De Caterina R, Kristensen SD, et al. Prehospital reperfusion therapy: A strategy to improve therapeutic outcome in patients with ST-elevation myocardial infarction. Eur. Heart J 2005;26: 2063–74.
  1. Keeley EC, Boura JA, Grines CL. Comparison of primary and facilitated percutaneous coronary interventions for ST-elevation myocardial infarction: quantitative review of randomized trials. Lancet 2006;367(9510):579–88.
  1. Keeley EC, Boura JA, Gunies CL. Primary angioplasty versus intravenous thrombolytic therapy for acute myocardial infarction: A quantitative review of 23 randomized trials. Lancet 2003;36:13–20.
  1. Universal Definition of Myocardial Infarction. Thygesen K, Alpert JS, White HD, Joint ESC/ACCF/AHA/WHF Task Force for the Redefinition of Myocardial Infarction. J Am Coll Cardiol published online Oct 19, 2007; doi: 10.1016/j.jacc.2007.09.11.