Cardiac Drugs Kanu Chatterjee, Eric J Topol
INDEX
Page numbers followed by f refer to figure and t refer to table.
A
α-adrenergic blocking agent 421
α-adrenergic receptor antagonists 153
Abciximab 208, 277, 306, 307, 309
doses 306
ACE inhibitors 16
Acebutolol 113, 125, 151, 280
Acetazolamide 168
Acetylsalicylic acid 292
Adenosine 381, 382, 512
diphosphate 276
pathway 449
receptor antagonists 295
monophosphate 42
triphosphate 42, 198, 462
binding cassette A1 194
Adipose triglyceride lipase 198
Adrenergic agents 45, 60
Adrenergic receptor antagonists, dosing for 127t
Adrenergic receptor blockers
cautions 151t
drugs, dosing for 124t
side effects 151t
Adult treatment panel 259
Alacepril 129
Alanine transaminase 195
Albumin 115
Albuminuria 115t
Aldosterone 18, 88, 103, 120
antagonist 325, 416
spironolactone 417f
blockade 434
evaluation study, randomized 19
receptor antagonists 16
synthase inhibitors 434
Aliskiren 130, 143, 145, 156
observation of heart failure treatment trial 433
Aliskiren/amlodipine 143
Aliskiren/valsartan 143
Allopurinol 467
Alpha-adrenergic receptor antagonists 127
Alprenolol 124
Alteplase 313
doses 313
Ambrisentan 481, 488
use of 488
American Diabetes Association 92, 259
American Heart Association 93
Amiloride 120, 184, 185, 188
Amiodarone 205, 346, 347, 352, 369, 371, 374, 391, 393, 512, 513
class III 350
infusion, intravenous 373
Amlodipine 7, 16, 132, 145, 157, 159, 206, 460
and valsartan 145
Amlodipine/benazepril 143
Amlodipine/olmesartan 144
Amlodipine/telmisartan 144
Amlodipine/valsartan 144
Ammi visnaga plant 370
Amosulalol 126
Angina and myocardial ischemia, relief of 458
Angina pectoris 442
Angina
chronic stable 442, 444t
classic 446
drugs for stable 442
drugs to 454t
relieve 454
effort 446
linked 468
management of stable 447
mixed 468
pathophysiology of 462f
revascularization therapy in chronic stable 447t
subsets of stable 445, 446t
treatment of stable 460t
Angiogenesis 468
Angiotensin-converting enzyme 12t, 16, 101, 103
inhibitors 3, 9, 12t, 41, 88, 91, 92, 94, 100, 117, 146, 148, 156, 321, 322, 408, 412, 413, 413t
side effects of 413f
with diuretics, combinations of 141t
Angiotensin I 12t
Angiotensin II 12t, 14
receptor blockers 16, 88, 94, 117, 148, 156, 408, 411, 413
subserves 11
Angiotensin inhibitors 321, 410
Angiotensin receptor 12t
blocker 3, 15, 91, 92, 101, 146, 255
blocking agent 321
valsartan in postmyocardial infarction, effects of 321f
Angiotensin-inhibiting drugs 452
Antianginal drugs 508
Antianginal strategies, current non-pharmacologic 464t
Antiarrhythmic agents, newer 383
Antiarrhythmic drug 342, 343
class I 344
class IA 353
class IB 357
class IC 360
class II 363
class III 364
class IV 381
device interactions 392
in lactation 391
in pregnancy 391
major drug interactions of 352t
on defibrillation and pacing thresholds 393t
pharmacokinetic properties of common 347t
properties of class I 354t
selection in atrial fibrillation 390
therapy 343, 391
Vaughan Williams classification of 345t
Antiarrhythmic therapy 388
future of 383
Antiarrhythmics 71
Anticoagulant therapies 284
Anticoagulation drugs in pregnancy and lactation 516t
Antihypertensive and lipid-lowering treatment to prevent heart attack trial 103
Antihypertensive drugs 81, 99, 103, 117, 146t
and dosing 116
and effects on serum lipids 117t
by class 96f, 100t
by compelling contraindications 103t
by compelling indications 103t
combinations of two 145t
dosing for 146t
in morbid conditions 104t
Antihypertensive medication 85
Antihypertensive therapy 255
Anti-ischemic therapies 277
Antiobesity drugs 254
Antiplatelet agents 507, 508
Antiplatelet drugs 317, 448
aspirin 317
aspirin dosage 317
Antiplatelet therapies 256, 265, 292
Antithrombotic therapies
primary prevention 265
secondary prevention 266
Antithrombotic trialists’ collaboration 266
Antithrombotics 507
Aortic impedance, components of 2t
Aortic stenosis, severe 26
Apolipoprotein 224
Arachidonic acid 476
Aranidipine 132
Arginine vasopressin 11, 13
antagonists 428
use of 429
Arotinolo 126
Arrhythmia 113
in pregnancy and lactation 511
mechanisms 343
Arrhythmogenic right ventricular dysplasia 351
Arterial disease, prevention of 265
Arterial revascularization therapies study-I 267
Arterial system, dilatation of 457
Arteriolar vasodilators 4
Artery disease in pregnancy, stable coronary 506
Aspirin 292, 449
complications 294
doses 293
higher dose 294
low-dose 293
pharmacology 292
resistance 294
Asthma 104
Asymptomatic organ damage 101
Atenolol 113, 125, 140, 151, 280
Atherosclerotic cardiovascular disease 193
Atorvastatin 201, 210, 221, 229, 327
amlodipine, double-blind 451
prevention
primary 212
secondary 213
Atrial fibrillation 351
Atrial natriuretic factor 13
Atrial septostomy 494
Autoregulatory reserve 444
Azelnidipine 132
Azilsartan 130
Azilsartan/chlorthalidine 142
Azimilide 346, 348, 380
dihydrochloride 380
prolongs 380
Azosemide 119
B
Balloon atrial septostomy 497
Barnidipine 132
BB and diuretic combination 137
Bedtime and evening versus morning dosing 97
Bemetizide/triamterene 139
Benazepril 16, 129, 141, 155
Bendroflumethiazide 118, 179
Benzothiazepines 156
block l-type channels 131
Benzthiazide 118
Beta 1-adrenergic receptor blockers 117
Beta adrenergic receptor 42
actions of 123t
Beta adrenoceptor blockade 345
Beta blockers 100, 280, 393
class II 345
class II 350
in contemporary 180t
pharmacology 280
with diuretics, dosing for combinations of 140t
with vasodilating properties 117, 126, 152
Beta-adrenergic antagonists 324
Beta-adrenergic blocking agents in stable angina 456t
Beta-adrenergic receptor antagonists 16, 453
Beta-adrenoreceptor blockers, uses of 113t
Betaxolol 113, 125, 280
Bethanidine 122, 150
Bevantolol 126
Bezafibrate 218, 222
Bile acid sequestrants 223, 224, 224f, 226, 227, 228t
Bioavail 119
Bisoprolol 16, 113, 125, 140, 280, 324, 510
Bivalirudin 290
doses 290
Blood pressure 81, 101, 259, 413, 443
classification 83t, 84t
comparison between JNC 6 and 7 83t
initial drug therapy for adults with hypertension, comparisons of goal 91t, 92t
regulation
pathophysiology of 81
physiology of 81
systemic 60
systolic 84, 93
thresholds 84t
with proteinuria 259
Blood urea nitrogen 156, 158
Blood vessels 123
Boceprevir 205
Body mass index 259
Bopindolol (prodrug for pindolol) 124
Bosentan 481, 487
Bretylium 343
Brisk walking 254
Bronchospasm 363
Bucindolol 126
Bumetanide 119, 171
Bunazosin 127
Bupranolol (similar to propranolol) 124
Buthiazide 118
Butizide 118
Butizide/spironolactone 139
C
Cadralazine 128
Calcium 42, 81, 346
antagonist 100
combination 137
channel blockers 71, 88, 91, 92, 94, 103, 117, 147, 158, 283, 352, 422, 459, 475, 479, 497
agents 460t
class IV 346, 351
on cardiac function 159t
on vascular functions 159t
side effects 156t
release channel, activation of 62
sensitizers 65
upregulation 432
Canadian hypertension education program 92
Candesartan 16, 130, 142
Cangrelor 304
Cangrelor pharmacokinetics 304
Captopril 16, 141, 155
sulfhydryl-containing 129
Captopril, converted to 129
Carbonic anhydrase inhibitors 168
Cardiac adrenergic activity with systolic heart failure 414f
Cardiac and arrhythmic death 372
Cardiac arrhythmia suppression trial I 361
Cardiac drugs in pregnancy and lactation 502
Cardiac dysrhythmias 42
Cardiac myocyte 11
Cardiac output 81
Cardiodysmetabolic syndrome 250, 250t, 251, 252, 268
Cardiometabolic syndrome 250t
Cardiomyocyte sarcolemma 67
Cardiomyopathies 509
Cardioselective 152
Cardiovascular control with established type 2 diabetes 258
Cardiovascular diseases 92, 259, 502
Cardiovascular drugs with teratogenic effects 503t
Cardioverter defibrillator, implantable 342, 351
Carteolol 113, 124
Carvedilol 16, 55, 66, 113, 126, 151, 280
Catechol-phenylethylamine molecule 45
Catheter-based coronary intervention 52
Celiprolol 125
Cell adhesion molecule, vascular 1 274
Cellular calcium handling 388
Central imidazoline receptor agonists
cautions 149t
side effects 149t
Central nervous system 150, 153
Central sympatholytic action 149
Chemical structure of LCZ696 23f
Chlorothiazide 118, 179
Chlorothiazide/reserpine 144
Chlorthalidone 119, 148, 169, 179
Chlorthalidone/reserpine 144
Cholesterol 197, 241
absorption inhibitor 241
Cholesteryl ester transfer protein 237, 253
Cholestyramine 226, 227
dose 225
Chronic kidney disease 88, 91, 92, 120, 148, 150, 154, 156, 158
Cigarette smoking 259
Cilazapril 129
Cilnidipine 132
Cilostazol 450
Cinchonism 356
Clarithromycin 205
Clevidipine 133
Clonidine 121, 149
Clonidine/chlorthalidone 145
Clopamide 118
Clopidogrel 297300, 319, 449
doses 297
indications 319
resistance 449
therapy, duration of 299
Clotting cascade
extrinsic 276f
intrinsic 276f
Coarctation of aorta 82
Colchicine 206
Colesevelam 229
dose 225
Colestipol 227
dose 225
Congestive heart failure 113, 154, 158, 177, 412, 428, 462
Constipation 461
Coronary artery bypass surgery 52
Coronary artery disease 43, 87, 150, 154, 259, 404
stable 508t
Coronary drug project 233
Coronary revascularization 53
Coronary syndrome, acute 308, 317, 506, 507t
drugs for 274
management of 280t
pathogenesis of 275f
statin doses of statins in 326
Corticotropin-releasing hormone 87
Cough, intractable 323
Creatinine kinase 195, 452
Cromakalim 134
Cushing's syndrome 82
Cyclic adenosine monophosphate 8, 42, 198, 490
Cyclopenthiazide 118
Cyclopenthiazide/amiloride 139
Cyclosporine 205
Cyclothiazide 118
D
Dalteparin 287
doses 287
Danazol 205
Debrisoquine 122, 150
Deep vein thrombosis 515
Delapril 129
Diabetes 261
and cardiodysmetabolic syndrome, drugs for 249
management in 259t
mellitus 88, 106, 257
management of coronary artery disease in 266
prevented, type 2 254
prevention of 265
Diabetic cardiomyopathy 268
Diacylglycerol acyltransferase 237
Diarylaminopropylamine ester 133, 158
Diastolic blood pressure 84, 93
Diazoxide 119
Diet intervention 253
Digitalis-digoxin 67
Digoxin 16, 206, 393, 424, 479, 510, 512
chronic 70
induced or -toxic dysrhythmias 72
use of intravenous 68
Dihydralazine 128
Dihydropyridine 100, 157, 159
calcium channel blockers 8, 460
second generation 131
Diltiazem 100, 131, 156, 159, 206, 348, 460, 512
injectable 131
slow release 460
sustained-release 131
Diltiazem/enalapril 143
Dipyridamole 449
Disease, vascular 261
Disopyramide 343, 347, 357, 389, 391
class IA 349, 354
use of 357
Diuresis dosing 173
Diuretic adverse effects 187
Diuretic classification of 168
Diuretic compounds 168
Diuretic sites of action in nephron 169f
Diuretic therapy 425
aggressive 423
Diuretic use in renal disease 176t
Diuretic, electrolyte effect of 187t
Diuretic, metabolic effect of 187t
Diuretic-based therapy 181
Diuretics 168, 423, 479
antial-dosterone 100
complications 423
dosing for 118t, 423
dosing for loop 172
high-ceiling or loop 171t
in hospitalized patients 60
intravenous 175t
loop 100, 170
oral dosing of loop 175t
DNA-drug interactions, basis of 520
Dobutamine 43, 45, 510
chronotropic properties of 52
Dofetilide 346, 347, 352, 367, 391, 393
class III 351
dose of 368t
Dopamine 58, 425, 510
agonists 162t
pharmacologic effects 58
Doxazosin 127, 153
Dronedarone 206, 346, 348, 352, 375, 391
class III 351
Drug class 100
Drug combinations of antihypertensive treatment 136t
Drug interactions 189, 420
Drug therapy in pregnancy 507t, 508t
Drug-related, drug-induced or other causes 114
Drugs as “first-line” agents 94t
Drugs new again, old 389
Drugs with novel antiarrhythmic mechanisms 390
Dysfunction, left ventricle systolic 515
Dysgeusia 15
Dyslipidemia, combined 239
Dyslipidemias, drugs for 193
Dysrhythmias 68, 114, 249
development of 59
drugs for 342
E
Efonidipine 132
Electrolytes 188
Enalapril 16, 129, 141, 155
Enalaprilat 129
Endopeptidase inhibitors, neutral 433
Endothelial dysfunction 249
Endothelial function 252
Endothelial lipase 194
Endothelial nitric oxide synthase 490
Endothelial no synthase 476
Endothelin 476
converting enzyme 476
receptor antagonists 481, 486, 487f, 497
side effects of 489
Endralazine 128
End-stage renal disease 101
Enoxaparin 288
doses 288
Epinephrine 61
Epitizide/triamterene 139
Eplerenone 16, 185
post-acute myocardial infarction heart failure efficacy 325
Epoprostenol 8, 481, 484
Eprosartan 16, 130, 142
Erectile dysfunction 107
Erythromycin 205
Esatenolol 125
Esmolol 113, 151, 280
Ethacrynic acid 120, 171
Ethylglycinexylidide 358
Etozolin 120
European Society of Cardiology 91
European Society of Hypertension 91
Extracellular matrix, degradation of 274
Ezetimibe 241
dose of 242
F
Farnesoid X receptor 224
Fat cells 123
Fatty acid oxidation 462
Fatty acid, free 198
Felodipine 157, 159, 460
Felodipine extended-release/enalapril 143
Fenofibrate 217, 221, 222
Fenofibric acid 221, 221t
Fenoldopam mesylate 162
Fibrate clinical trials 222t
Fibrates 214
Fibrillation, ventricular 351
Fibrinolytic agents 312
Flecainide 347, 352, 360, 361, 391, 393, 512, 513
class IC 350, 354
Fluconazole 205
Fluvastatin 202, 209
secondary prevention 212
Fondaparinux 289
doses 289
Fosinopril 16, 141, 155
phosphonate-containing 129
Furosemide 120, 169, 171, 172, 510
G
G protein-coupled receptor 198, 230
Gallopamil 133
Ganglionic and postganglionic blockers 150t
cautions 150t
dosing for 122t
side effects 150t
Ganglionic blockers 122, 150
Gemfibrozil 205, 218, 222
Gene therapy in cardiac disease, percutaneous of 432
Genetics in drug therapy, role of 520
Genomic guidance 522
Glaucoma 107
Glomerular filtration rate 120, 148, 153, 176, 201, 413
Glucose 259
elevated 253
fasting 239
infusion in acute myocardial infarction 267
mediated osmotic polyuria 170
oxidation 462
tolerance 257
Glycemic control 263
Glycinexylidide 358
Glycoprotein 276
IIB/IIIA pharmacology 305
Glycosylated haemoglobin 259
Gout 107
Guanabenz 121
guanfacine 149
Guanadrel 122, 150
Guanethidine 122, 145, 150
Guanfacine 121
Guanosine monophosphate 490
Guanosine triphosphate 490
Guanylate cyclase stimulator, soluble 490, 491
Gynecomastia
cause of 72
incidence of 419
H
Hemorrhage, intracranial 285
HDL cholesterol, low 253
Heart 123
Heart block (second and third degrees) 107
Heart catheterization, right 473
Heart failure 11, 13f, 70, 88, 107, 403, 410
angiotensin II receptor blockers in 413t
chronic 47
systolic 324
correction of hyponatremia in 427
diagnosis of 404
drugs for 403
drugs in managing chronic 16t
effects of drug classes in chronic 408t
eplerenone in chronic 21
in adults, management of 53, 59, 64
in African-American heart failure trial 7f
in diabetes 268
in pregnancy and lactation 509
in pregnancy, drugs in management of t 510
management of 403, 416, 425, 429
mineralocorticoid in 19
mortality curves with 6f
moxonidine in congestive 415
neurohormonal activation with systolic 411f
newer pharmacologic agents for treatment of 430
of intravenous inotropic support, acute 66
prognosis of 4
severe 26
syndromes, drug treatment for acute 425
systolic 18, 88, 103, 158, 403, 461
trial, vasodilator 6f
trial, veteran administration 421
with ejection fraction
management of
preserved 429
reduced 407
preserved functional changes in 406t
preserved morphologic in 406t
reduced functional changes in 406t
reduced morphologic in 406t
with preserved ejection fraction 436
with reduced ejection fraction 408t, 431
Heart outcomes prevention evaluation study 103
Heart rate 42
Heart, lung, and blood institute, national 93
Heberden's angina 446
Helicobacter pylori infection 301
Hemodynamic classification of pH 475
Hemodynamic effect
of milrinone, comparison of 63f
on coronary 457
on systemic 457
Hemodynamic profile 421
Hemodynamic responses to thiazides 181f
Heparin doses, unfractionated 284
Heparin pharmacology, unfractionated 284
Heparin, low-molecular weight 287, 515
Heparin, unfractionated 284, 285
Heparin-induced thrombocytopenia 286
Hepatic lipase 194
Hepatic transaminase levels 242
Hepatic-splanchnic flow 49
Hepatotoxicity 374
High blood pressure 253
High-density lipoproteins 93, 117, 148, 153, 154, 156, 158, 194, 224, 234, 237, 253, 259
High-risk subjects, high- and very 93t
HIV protease inhibitors 205
Hormone sensitive lipase 198, 237
Human immunodeficiency virus 202
Hydralazine 4, 5, 128, 145, 505, 510
and nitrates 420
combination of 422
or amlodipine 4
Hydralazine-isosorbide dinitrate 16
Hydrochlorothiazide 118, 120, 169, 179, 505, 510
Hydrochlorothiazide/reserpine 144
Hydroflumethiazide 118
Hypercalcemia 82, 249, 263
prevention of 257
Hyperinsulinemia 249
Hyperkalemia 326
Hyperlipidemia 258
Hyperpolarization-activated cyclic nucleotide-gated 389
Hypertension 11, 113, 182, 186, 260
benefits of treating 84t
causes of resistant 114t
classifications of 82
elderly and isolated systolic 106
in adolescents 114
in children 114
in pregnancy, drugs for treatment 505t
in special groups 99
management of 93, 260
medication for essential 92
resistant 99
severe 135
systolic 96
therapy in essential 93
treatment of 82, 84, 98
principles for 85t
with measurements 84t
Hypertensive disease in
lactation 504
pregnancy 504
Hypertensive emergencies 101
intravenous for 128t
Hyperthyroidism 108
Hypertriglyceridemia, severe 240
Hyperuricemia 189
Hypokalemia 87
Hyponatremia 427
drug treatment of 427
I
Ibutilide 346, 347, 368, 393
use of 369
Idiopathic pulmonary arterial hypertension 497
Iloprost 485
Imidapril 129
Imidazoline receptor antagonist 121t
Immediate release niacin 231
Indapamide 119, 179
Indenolol 124
Indoline derivatives 119
Indoramin 127
Inodilator 65
Inotropes, positive 60
Inotropic agents, positive 423
Inotropic drugs 431
positive 41
Inotropic interventions, intravenously positive 65
Inotropic therapy 425
intravenous short-term positive 43
Insulin sensitivity 252
Intestines 123
Intra-aortic balloon counterpulsation 52
Intravenously administered positive inotropes 66
Intrinsic sympathomimetic activity 113, 117
Irbesartan 16, 130, 142
Ischemic syndromes 289
Ishchemia threshold in stable angina 445f
Isolated systolic hypertension 101
Isoproterenol 60
Isosorbide dinitrate 458
Isosorbide dinitrate/hydralazine 6f
Isosorbide mononitrate 458
Isradipine 159, 460
Itraconazole 205
Ivabradine 385, 386, 434, 466
dose 467
side effects 467
J
JNC 8 with JNC 7 guidelines, comparison of current recommendations from 89t
K
Ketanserin 162
Ketoconazole 205
Kidney 123
disease, chronic 82, 88, 183, 220
hydrophilic and removed by 152
L
Labetalol 113, 126, 280, 505
Lacidipine 132
LCZ696 23
Lecithin cholesterol acyltransferase 194
Left ventricular
dysfunction and afterload stress, relationship between various degrees of 1f
ejection fraction 403
end-diastolic pressure 475
hypertrophy 18, 101
hypotrophy 93
stroke work index 57
Lercanidipine 132
Levosimendan 65
Liddle's syndrome 187
Lidocaine 347, 353, 358, 359, 393
class IB 354
Lifestyle modification 87t, 252
Lipid disorders, treatment of 193
Lipid management 255
Lipid solubility of β-adrenergic receptors 151t
Lipid-lowering agents 450
Lipid-lowering drugs 508
Lipophilic 151
Lipoprotein levels with
bile acid sequestrant therapy, changes in 229t
fenofibrate, changes in 221
niacin therapy, changes in 234t
statin therapy, changes in 209t
Lipoprotein lipase 237
Lipoprotein, high-density 193, 250
Lipoprotein, intermediate density 193
Lipoprotein, low-density 250
Lisinopril 16, 129, 141, 155
Liver 123
disease, active 232
disease, chronic 105
enzymes, elevated 47
function test 482
X receptor 224
Losartan 16, 130, 142
Lovastatin 200, 209
dose of 452
primary prevention 212
Low lipid solubility/hydrophilic 151
Low-density lipoprotein 93, 117, 148, 153, 154, 156, 193, 194, 224, 234, 237, 253, 255, 521
cholesterol 259
receptors 237, 452
Ltizide/spironolactone 139
Lung 123
and combined heart 493
injury ventilator evaluation, acute 383
transplantation 493
M
Macitentan 481, 488
Magnesium 81
Manidipine 132
Mannitol 170
Mecamylamine 122
Mecamylamine (oral tablet) 150
Mefruside 118
Membrane stabilizing activity 113
Menopausal symptoms 108
Mepindolol 124, 140
Metabolic abnormalities 252
Metabolic dysfunction, decreased 463
Metabolic efficiency with ranolazine 387
Metabolic modulation 464
Metabolic syndrome 108, 251
management of 253t
to cardiovascular disease 256
to diabetes 256
Metabolism 26
Methyclothiazide 118
Methyclothiazide/deserpidine 144
Methyldopa 121, 149, 505
Methylxanthines 383
Meticrane 118
Metipamide 119
Metolazone 119, 179
Metoprolol 55, 113, 125, 140, 280, 347, 505, 510, 512, 513
extended release 125
succinate 16
Metyrosine 134, 160, 347, 352, 359, 391
Mexiletine 343, 353
class IB 349, 354
Microvascular disease 264
Migraine headaches (prevention) 108
Milrinone 43, 49, 55, 61, 62, 510
effect of 65
molecular structure of 63f
Mineralocorticoid (aldosterone) receptor blockers 18
Minoxidil 128, 134, 161
Mitogen-activated protein 256
Moexipril 16, 129, 141, 155
Monoamine oxidase inhibitors 150
Monotherapy 93, 229
Monotherapy and combination therapy 93
Monotherapy of ranolazine in stable angina trial 463
Monotherapy versus combination therapy strategy 95f
Moxonidine 121
rilmenidine 149
Muscle creatine kinase 327
Myocardial disease 403
Myocardial infarction 84, 88, 113, 154, 412
acute 315
triage and intervention 314
Myocardial ischemia 454t, 464t
manifestation of 443
reduction with aggressive cholesterol lowering trial 326
with stable angina 454
Myocardial metabolic dysfunction 462
Myocardial oxygen demand 443t
Myocyte apoptosis 405
Myocyte hypertrophy 405
Myocyte necrosis 405
N
N-acetyl procainamide 356
Nadolol 113, 124, 280
Nadolol/bendroflumethazide 140
Narcotics 279
doses 279
pharmacology 279
National Health and Nutrition Examination Survey 249
Nebivolol 16, 113, 126, 153
Nefazodone 205
Nephrotic syndrome 169, 174, 175
Neprilysin inhibitor 434
Nesiritide 27, 49
Neuregulins 433
Neurohormonal activation, effects of 409t
Neurohormonal modulation 409
Neurohormonal systems 14
activation of 14
Neurohormone modulator 1
Neurohormones, activation of 13
Neurohumoral hypothesis 14
Neurohumoral modulating drugs 33
Niacin 229, 231
extended release 231, 234
preparations 232
Niacin/lovastatin, extended release 234
Nicardipine 159, 460
immediate-release 132
slow-release 132
Nicorandil 161, 465
dose 466
side effects 466
Nicotinic acid 233
Nifedipine 159, 460, 505
Nimodipine 132, 157, 159, 460
Nisoldipine 132, 159, 460
coatcore tablet 132
hydrogel tablet 132
Nitrates 8, 61, 421, 456458, 510
dosing for 133t
Nitrendipine 132, 460
Nitric oxide 12t, 24, 113, 476, 490
pathway 489
Nitroglycerin 49, 277, 278, 456, 458, 505
and nitrates
action of 458t
dose of 458t
duration of 458t
preparations 458
causes, intravenous 27
degenerates in vivo 277
doses 278
in treatment of patients with heart failure, intravenous 27
intravenous 5f, 27, 133, 160
patch 458
pharmacokinetics 278
pharmacology 277
sublingual 447
syncope 457
with early tolerance 5f
Nitroprusside 25, 26, 49, 510
Nondihydropyridine calcium channel blockers 461
Nondiuretic/nonsaluretic thiazide 119
Nonsteroidal anti-inflammatory drugs 148, 517
Norepinephrine 49, 61, 426
Nutrition, adverse 259
O
Obesity, central 250
Olmesartan 16, 130, 142, 145
Omega-3 fatty acids 236, 237f
Optic neuropathy, anterior ischemic 97, 482
Oral D-sotalol, survival with 366
Oral flecainide 362
Oral nitrates 8
Oral procainamide 342
Oral sotalol 515
Oral β-adrenergic blocking drugs 30
Orally administered positive inotropic agents 72
Orally antiarrhythmic agents
side effects of 349t
uses of 349t
Osmotic agents 170
Ototoxicity 190
Oxprenolol 124
Oxygen 478
P
P2Y12 antagonists 296t
Pancreas 123
Paradoxical response of nitroglycerin 457
Parathyroid gland 123
Pargyline 122
Penbutolol 113, 124
Peptic ulcer
disease 108
exacerbation of 233
Perindopril 16, 129, 155
Perindopril/amlodipine 143
Peripheral arteriolar vasoconstriction 2
Peripheral resistance, total 81
Peripheral vascular disease 108
Peroxisome proliferator receptor-α 224
P-glycoprotein 420
Pharmacologic effects 47
Phenoxybenzamine 127
Phentolamine 127
Phenylalkylamines 158
Phenylalkylamines (block l-type channels) 133
Phenylephrine 61
Phenytoin 206, 343
Phosphodiesterase 42
5 inhibitors 482, 490
side effects of 491
inhibitors 55, 62
isoenzymes 23
type 5 inhibitors 23
Phthalimidine derivatives 119
Pindolol 113, 124, 280
Pindolol/clopamide 140
Piretanide 120
Pitavastatin 203, 210
Pituitary, posterior 123
Plasma glucose levels 188
Plasma renin activity 13
Plasminogen activator inhibitor 12t, 18
Platelet plug, primary 275
Platelet receptors
identified therapeutic targets 277t
inhibition in ischemic syndrome management 309
use of antiplatelet agents on 276f
Polythiazide 118, 144
Posaconazole 205
Postganglionic blockers 122, 150
Potassium 42, 81, 413
channel blockers, class III 346
channel openers or activators, dosing for 134t
rectifier current
rapid delayed 346
slow delayed 346
sparing diuretics 71, 120, 170, 183, 185t
Potential duration, action of 346
Pranidipine 133
Prasugrel 299, 300, 302, 319
doses 302
Pravastatin 200, 210
primary prevention 212
secondary prevention 213
Prazosin 6f, 127, 153
Pregnancy 109, 478
Premature ventricular contraction 51
Prerenal azotemia 47
Prinzmetal angina 446
Proarrhythmia 344
Procainamide 347, 356, 391, 393, 513
class IA 349, 354
intravenous 356
Proinflammatory state 253
Proliferators-activated receptor gamma 255
Propafenone 347, 352, 362, 391, 512, 513
class IC 350, 354
Propranolol 113, 140, 151, 152, 280, 505
timolol 152
Prostacyclin 483f
analogues 480, 481
side effects 486
synthase 476
Prostaglandins 12t
Protease-activated receptor-1 276, 310
Protein 115
K 42
kinase A 198
Proteinuria 115t
Prothrombin time 233
Prothrombotic state 253, 259
Psychiatric comorbidity 110
Pulmonary arterial hypertension 473475, 487f, 497
action in 483f
approved drugs 480
disease-specific therapies for 481t, 492
prognosis in 484t
specific therapies 477f
treatment for 477, 497f
Pulmonary arterial pressure, mean 473
Pulmonary artery wedge pressure 475
Pulmonary capillary wedge pressure 57, 410
Pulmonary disease, chronic obstructive 153
Pulmonary hypertension 473, 474
drugs for 473
WHO classification of 474t
with unclear multifactorial mechanism 474
Pulmonary thromboendarterectomy 494
Pulmonary vasculature 473
Pyrophosphate 197
Q
Quinapril 16, 129, 141, 155
Quinazoline derivatives 119
Quinethazone 119
Quinidine 343, 347, 352, 353, 356, 389, 391, 393
class IA 349, 354
R
Ramipril 16, 129, 155
Ranolazine 206, 348, 386, 461, 463
side effects of 463
Rauwolfia alkaloids and antihypertensive agents, combinations of 144t
Raynaud's phenomenon 110
Rehabilitation and exercise training 477
Remedial cardiac surgery 47
Renal artery stenosis 110
Renal disease 176
Renal dysfunction 65
Renal failure 176
Renin inhibitors 433
with antihypertensives, dosing for combinations of 143t
Renin-angiotensin system 23
dosing for inhibitors of 129t
inhibitors of 155t
cautions 155t
side effects 155t
Renin-angiotensin-aldosterone
blocking agents 433
system 2, 82, 262
blockers 9
Renovascular disease 110
Reserpine 144
Reteplase 313
doses 313
Rho-kinase inhibition 464, 467
Rifampicin 206
Rilmenidine 121
Riociguat 482, 491
Rosuvastatin 202, 209, 327
Rosuvastatin primary prevention 212
Ryanodine receptor stabilizers 432
S
Sarcoendoplasmic reticulum atpase 388
Sarcolemmal sodium/potassium, inhibitor of 67
Sarcoplasmic reticulum 62
calcium adenosine triphosphatase activators 432
Scavenger receptor B1 194
Selexipag 486
Serotonin receptor agents 162t
Serum digoxin 70
concentrations, alter 71t
Sibutramine cardiovascular 255
Sildenafil 24, 482, 490
and tadalafil 23, 489
Simvastatin 201, 211, 221, 229
secondary prevention 213
Sinoatrial 158
Sinus node inhibitors, selective 434
Skeletal muscle 123
aches and pains 327
Sodium 42, 81, 346
channel blockers, class I 345
current 383
guanylate cyclase stimulators 476, 482, 497
inward current 462
nitroprusside 128
Sotalol 113, 346, 347, 352, 365, 391, 393, 512, 513
class III 351
with creatinine clearance, dose of 365t
Spirapril 129
Spironolactone 16, 21, 170, 185, 188
action of 21
Statin 193, 200, 326
cautions 205t
doses 205t
high-, moderate- and low-intensity 195t
interactions 205t
pharmacology of 199t
prevention trials 212t
Sterol heterodimer partner 224
Sterol regulatory element binding protein 2 224
Stomach 123
Streptokinase 313
Streptokinase doses 313
Stroke 111
work index 410
Subendothelial tissue factor 274
Subintimal layer of foam cells 274
Supraventricular tachyarrhythmias in pregnancy, drugs for treatment of 512t, 513t
Sympathetic overactivity 82
Sympatholytic agents, dosing for central 121t
Sympathomimetic activity, intrinsic 153
Syndrome “X” 469
Systemic lupus erythematosus 154, 156, 158
T
Tachyarrhythmias, rapid atrial 364
Tachychardia, ventricular 351
Tadalafil 24, 482, 491
Takotsubo cardiomyopathy 58
Talinolol 125
Tartrate 125
Tedisamil 385
Telaprevir 205
Telithromycin 205
Telmisartan 16, 130, 142
Temocapril 129
Tenecteplase 312, 314
doses 314
Teratogenic effects 502
Terazosin 127, 153
Tertatolol 125
Therapeutic considerations, general 182
Thiazide diuretics 100, 117, 177
and thiazide-like 169, 179t
Thiazides 118, 183
Thiazide-type diuretics 147, 186
Thiocyanate ion 26
Thiocyanate toxicity 26
Third generation dihydropyridines 132
Thrombin inhibitors 289
pharmacology 289
Thrombin receptor antagonists 310
Thromboembolic pulmonary hypertension, chronic 474
Thrombolysis, contraindications to 315t
Thrombolytic agents 312
Thrombolytics 507
Thromboplastin time, activated partial 284
Thromboprophylaxis in
lactation 515
pregnancy 515
Thromboxane 276
Thyroid 123
Ticagrelor 299, 303, 320
doses 303
Ticlopidine 295
complications 297
doses 295
Tilisolol 126
Timolol 113, 125, 280
Tirofiban 308
doses 308
Tissue plasminogen activator 312
recombinant 312
Tocainide 342
Tolazoline 127
Torsemide 120, 169, 171
Toxicity of nitroprusside 26
Trandolapril 16, 130, 155
Tremor, essential 107
Treprostinil 481, 485
Triamterene 120, 170, 185, 188
Trichlormethiazide 118
Triglycerides 198, 259
Trimetazidine 435, 464
side effects 465
Trimethaphan 122
intravenous 150
Tripamide 119
Tyrosine hydroxylase blocker 134t
U
Urapidil 162
Urinary bladder 123
USFDA pregnancy risk category for medications 503t
Uterus 123
V
Valsartan 16, 130, 142
Valvular heart disease 4
Vascular resistance, systemic 2
Vasodilator drugs 3
with systolic heart failure, arterial versus venous effects of 4
Vasodilator therapy 426
Vasodilators modulator 1
Vasodilators, dosing for direct 128t
oral acting 128t
Vasodilators—relaxin 431
Vasopeptidase inhibitors 467
Vasopressor infusion 49
Vasopressor therapy 425
Vasospastic angina 446, 468
Venous thromboembolism 515
Ventilation-perfusion mismatch 26
Ventricular tachycardia 343
Verapamil 100, 158, 159, 206, 348, 352, 393, 460, 512, 513
extended-release at bedtime 133
immediate-release 133
intravenous 133
slow release 460
sustained-release 133
Vernakalant 348, 383
Very low-density lip 198
Vitamin D supplementation 436
Vorapaxar 310
doses 310
pharmacology 310
W
Waist circumference 250
Walk-through-angina 446, 468
Warfarin 206, 450
Wilcoxon test 6f
Wolff-Parkinson-White syndrome 351
X
Xipamide 118
Z
Zofenopril 16, 130
×
Chapter Notes

Save Clear


Vasodilators and Neurohormone ModulatorsCHAPTER 1

Gary S Francis,
Jason A Bartos,
Thenappan Thenappan,
Suma H Konety
 
INTRODUCTION OF THE AFTERLOAD REDUCTION CONCEPT
It has long been recognized that impedance to left ventricular (LV) outflow (afterload or increased wall stress during systole) is a critical determinant of cardiac performance.14 This is especially true of patients with impaired LV systolic performance, such as that occurs in systolic heart failure (Fig. 1).5 Ultimately, the failing heart's ability to respond to increased impedance is diminished. We now know that specific drugs lower aortic impedance, thus, restoring myocardial systolic function to some extent.
zoom view
Fig. 1: The relationship between various degrees of left ventricular dysfunction and afterload stress.
2
The factors that makeup impedance and impair LV ejection of blood in patients with heart failure are multiple, complex, and highly interactive. They include:
  • Peripheral arteriolar vasoconstriction: This is due to heightened neurohormonal activity including augmentation of the sympathetic nervous system, and intense activation of the renin-angiotensin-aldosterone system (RAAS). There is also release of vasopressin and endothelin, potent peripheral vasoconstrictors.
  • Increased LV wall stress during cardiac myocyte shortening—a concept known as “afterload”. Afterload is a term that emerged years ago from the study of isolated muscles. It is a laboratory but not a bedside measurement.
  • Diminished distensibility of the large vessels such as the aorta and its major branches.
  • Reduction in small vessel caliber and compliance.
  • Increased blood viscosity and inertia.
Each of these forces can act collectively to impair LV outflow tract flow or cardiac output during contraction and may improve to some extent with vasodilator therapy.
Heightened resistance or impedance to LV ejection is often referred to as “afterload”, but the term afterload originates from isolated muscle studies done in the mid-1970s and is not, strictly speaking, appropriately applied to the clinical setting. Afterload is defined as ventricular wall stress during myocyte shortening, and cannot be easily measured in the intact circulation. It is a product of LV cavity size (LaPlace relationship) and systolic arterial pressure and is inversely related to wall thickness or hypertrophy. In clinical practice, systemic vascular resistance (SVR) is a surrogate for afterload that is frequently calculated from right heart catheterization data [SVR = (mean arterial pressure – CVP) × 80/cardiac output], but this calculation is largely an estimate of small peripheral vessel resistance. SVR is, therefore, only a part of the total impedance that affects LV ejection (Table 1). The failing ventricles (both left and right) are exquisitely sensitive to afterload conditions, and it is a logical extension of this concept that drugs that reduce aortic impedance will improve cardiac systolic performance, independent of any effect on myocardial contractility.
Table 1   Components of aortic impedance
  • Large vessel distensibility
  • Small vessel caliber (systemic vascular resistance)
  • Small vessel compliance
  • Blood viscosity
  • Inertia
3
 
Vasodilator Drugs and Low Blood Pressure
Patients with moderate-to-severe heart failure often have low blood pressure that is asymptomatic. Low brachial systolic pressure is sometimes perceived by physicians as a contraindication to the use of arteriolar dilator drugs such as nitrates, angiotensin converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), or carvedilol. However, vasodilator drugs can maintain systolic blood pressure by increasing stroke volume in patients with impaired systolic function. Observations from large clinical trials have challenged the belief that vasodilators are deleterious in patients with low systolic blood pressure.68 Generally speaking, vasodilator drugs should be continued in patients with systolic heart failure and asymptomatic low systolic blood pressure in the range of 80–110 mmHg is not necessarily a contraindication to vasodilator therapy. Severe, symptomatic hypotension can sometimes occur in a volume-depleted patient in response to the first dose of an ACE inhibitor. For example, this might occur following a robust diuresis. Such brisk falls in blood pressure can be treated by leg elevation. Clinicians recognize that symptomatic hypotension is a well described adverse event that can occur when ACE inhibitors or ARBs are used in the context of hypovolemia and an activated RAAS. Symptomatic reduction in systolic blood pressure in response to vasodilators in a euvolemic or volume overloaded patient with severe LV dysfunction is another matter. Such patients are said to be truly intolerant of vasodilators, and symptomatic hypotension in response to drug therapy is a very powerful sign of poor prognosis. Low blood pressure without symptoms is far more common and is usually tolerated by patients using vasodilator drugs.
To avoid symptomatic hypotension when using vasodilator drugs to treat heart failure, it is best to begin with the lowest tolerable dose, and then gradually titrate the drugs over several weeks. This requires great patience and frequent contact with the patient. However, the goal is to prevent adverse effects, such as dizziness, light-headedness, syncope, extreme fatigue, and re-admission to the hospital for generalized malaise. It is now a common practice to have highly trained nurses with expertise in heart failure manage such patients soon after hospital discharge (1 week or sooner at our medical center) to carefully proceed with medication titration in a highly monitored setting. In addition to monitoring patient response, serum electrolytes and renal function are frequently assessed. Frequent follow-up soon after hospital discharge with measurement of electrolytes and careful physical examination seems to be an important element in reducing re-hospitalization.4
 
Arterial versus Venous Effects of Vasodilator Drugs in Patients with Systolic Heart Failure
The hemodynamic effects of vasodilator drugs are dependent on the relative effects of the drug on resistance and capacitance vessels. Arterial vasodilators such as hydralazine or amlodipine reduce aortic impedance and thereby increase the velocity of shortening during LV ejection. LV end-systolic volume is thus reduced, and LV ejection fraction increases. With hydralazine, LV end-diastolic volume (i.e., preload) is not acutely altered, so the stroke volume response can be markedly increased.9 When venodilator drugs such as nitrates are employed in patients with systolic heart failure, blood volume may acutely redistribute into the large capacitance veins, and LV end-diastolic volume or preload is reduced. The reduced LV end-diastolic volume can limit the increase in stroke volume to some extent.10 With balanced arteriolar-venous vasodilator drugs, such as sodium nitroprusside, a combination of decreased venous pressure (decongestion) and decreased aortic impedance is achieved, which results in improved stroke volume. In patients with severe regurgitant lesions such as mitral or aortic regurgitation, vasodilator drugs also reduce the regurgitant fraction and increase forward cardiac output, thus adding to their beneficial effects. It should be so noted that there are no adequately powered randomized controlled trials with the use of vasodilators in valvular heart disease that support their use to improve long-term outcomes.
The reflex tachycardia observed in normal subjects in response to arterial vasodilators is not seen in patients with advanced systolic heart failure.11 This is likely due to a reduction in cardiac norepinephrine spillover rate in the setting of heart failure with unloading of the baroreceptors and low pressure mechanoreceptors in response to systemic vasodilation.12 In fact, the magnitude of the blunted neurohumoral response to nitroprusside infusion in patients with systolic heart failure (i.e., lack of reflex tachycardia) may be a marker of the severity and prognosis of heart failure.11
In general, the beneficial response to vasodilator drug therapy is most pronounced in patients with systolic heart failure and a dilated LV. Patients with normal LV cavity size may be more sensitive to changes in preload reduction, and hypotension can occur in response to reduced afterload if the heart is small or there is a relatively reduced preload.
 
ARTERIOLAR VASODILATORS
 
Hydralazine
Hydralazine was one of the first drugs used to treat hypertension in the 1950s.5
zoom view
Fig. 2: Nitroglycerin (NTG) alone is associated with the development of early tolerance, whereas the combination of NTG and hydralazine (HYD) 75 mg four times per day is associated with less NTG tolerance. (*: Statistically significant changes).
Its mechanism of action is still not completely understood, but it appears to be a direct and potent arteriolar dilator that relaxes the smooth muscles of small resistance vessels. It has essentially no venodilating effects. Hydralazine primarily dilates the renal and peripheral resistant arterioles, and has little effect on coronary or liver blood flow. It may also have antioxidant effects, and can prevent tolerance to nitrates (Fig. 2).
Hydralazine is well-known to cause reflex tachycardia when used in patients without heart failure. For example, in patients with systemic hypertension large doses can produce a reflex tachycardia, edema and may rarely worsen angina. Reflex tachycardia and excess salt and water retention in response to hydralazine is not typically observed in patients with more advanced systolic heart failure because of a blunted baroreceptor response.
Hydralazine can be given orally, where it is rapidly absorbed from the gastrointestinal tract. However, the actual bioavailability is highly variable, and depends on the rate of acetylation by the liver, a genetically determined trait. In the United States, about half of people are fast acetylators and half are slow acetylators. Acetylation activity is not routinely measured in patients. A lupus-like syndrome from hydralazine is more likely to occur in slow acetylators, and this typically wanes when hydralazine is stopped. Fast acetylators may require higher doses of hydralazine. Chronic hydralazine use can cause vitamin B6 deficiency.
The hemodynamic response to chronic oral hydralazine therapy in patients with systolic heart failure is usually characterized by no change in heart rate, a fall in SVR, and about 6a 50% increase in cardiac output.13 Most commonly, blood pressure does not change much with hydralazine. Patients with chronic mitral or aortic regurgitation demonstrate a reduction in the regurgitant jet by echo and auscultation, and forward stroke volume is markedly increased. There is no long-term improvement in exercise capacity despite a modest, persistent improvement in ejection fraction. The combination of hydralazine and isosorbide dinitrate ushered in the vasodilator era for the treatment of heart failure in the 1980s (Fig. 3).
Even today we do not know entirely how to properly dose hydralazine for individual patients with advanced heart failure. Because of the high success rate of other vasodilator drugs, such as ACE inhibitors and ARBs, hydralazine has been relegated to second-tier therapy. The one important exception is the safety and efficacy of hydralazine and isosorbide dinitrate in the African-American Heart Failure Trial (A-HeFT) (Fig. 4).14 In this trial, the fixed combination of hydralazine and isosorbide dinitrate (BiDil®) added to standard therapy markedly improved survival and other outcomes among self-identified black patients with systolic heart failure. One rationale for the trial was that isosorbide dinitrate might augment nitric oxide production, and therefore improve endothelial function. Hydralazine may also work as an antioxidant and can reduce nitrate tolerance.15 The results of the A-HeFT trial have not been robustly translated into clinical practice for a number of reasons.
zoom view
Fig. 3: Mortality curves of patients with heart failure randomized to placebo, prazosin or isosorbide dinitrate/hydralazine in the first Vasodilator Heart Failure Trial (V-HeFT 1); p= 0.046 on the generalized Wilcoxon test, which gives more weight to the treatment differences in the early part of the mortality curves.
7
zoom view
Fig. 4: Mortality curves of African-American patients randomized to placebo or isosorbide dinitrate/hydralazine in addition to standard therapy for heart failure in the African-American Heart Failure Trial (A-HeFT).14
The combination of hydralazine and isosorbide dinitrate today is generally considered as an add-on therapy, superimposed on more conventional therapy, when patients are demonstrating signs and symptoms of worsening heart failure.
Typically, hydralazine is prescribed along with isosorbide dinitrate to improve cardiac output and reduce pulmonary capillary wedge pressure. The initial hydralazine dose used in A-HeFT was 37.5 mg three times per day and gradually increased to 75 mg three times per day. Isosorbide dinitrate was slowly titrated to a dose of 80 mg three times per day. Doses of hydralazine as high as 1,200 mg/day have been used to treat systolic heart failure, but onset of the lupus syndrome is seen in 15–20% of patients receiving more than 400 mg/day. Fluid retention is also more common when higher doses of hydralazine are used. There is likely a survival advantage associated with long-term hydralazine therapy when taken with isosorbide dinitrate to treat systolic heart failure.
 
Amlodipine
Amlodipine is a dihydropyridine L-type calcium channel blocking agent that is widely used to treat hypertension and angina. It is a long-acting, potent, arteriolar dilating drug that is well-tolerated. The typical starting dose is 2.5 or 5 mg/day and the target dose for many patients with hypertension is 10 mg/day. Calcium channel drugs are vasodilators and have anti-ischemic effects, so it is logical that they would be investigated in patients with systolic heart failure. The most promising calcium channel blocker to emerge from these studies as potential heart failure therapy was amlodipine. A minor drawback to amlodipine is the frequent development 8of pedal edema with the higher dose, but this is assumed to be due to benign vasodilation in the small arterioles and venules in the ankles, and not due to heart failure per se. Other non-dihydropyridine calcium channel blockers such as verapamil and diltiazem have negative inotropic properties, may cause cardiac electrical conduction problems, and are not very powerful vasodilators. They have never played a primary role in the treatment of heart failure.
The effect of amlodipine on outcomes in patients with chronic systolic heart failure was evaluated in two PRAISE (Prospective Randomized Amlodipine Survival Evaluation) studies.16,17 The earlier of the two studies demonstrated that all-cause mortality might be lower in a subset of patients with nonischemic dilated cardiomyopathy treated with amlodipine, though overall, the trial was neutral. The PRAISE II trial was then focused solely in patients with nonischemic dilated cardiomyopathy. In PRAISE II, an overall neutral effect of amlodipine was once again observed. It seems clear that amlodipine is safe to use in patients with systolic heart failure when needed to control hypertension or angina. However, amlodipine is not effective as a life-saving therapy for the treatment of systolic heart failure, despite its powerful vasodilating properties. The observations suggest that vasodilation alone is not enough to provide a mortality benefit. Several other potent vasodilators have failed to improve mortality in patients with heart failure, including prazosin, flosequinan, nesiritide, and synthetic prostacyclin (epoprostenol) or Flolan. Presumably, it is not simple “vasodilation” that provides for the survival benefit, but there should be some neurohumoral modulation property or some other mechanisms beyond simple reduction in afterload.
 
Oral Nitrates
Nitrates have been widely used to treat angina by physicians for well over 100 years. It is only in the past 25 years that they have been used to treat systolic heart failure. Their favorable effects on angina, systolic heart failure, mitral regurgitation and coronary spasm are now well known. The mechanism of action of nitrates is complex, but these molecules appear to undergo a metabolic biotransformation in vascular smooth muscle, which leads to the formation of nitric oxide or a related S-nitrosothiol. These breakdown products of nitrates stimulate the enzyme guanylate cyclase, leading to the formulation of cyclic guanosine monophosphate (cGMP). cGMP in turn reduces calcium influx, which leads to venous and arterial vasodilation.18 It is also likely that the vascular endothelium responds to nitrates with the synthesis and release of prostacyclin,19 thus improving endothelial function. Nitrates primarily cause venodilation, which typically increases venous capacitance and reduces 9preload, thus lowering end-diastolic volume, reducing cardiac wall tension and diminishing pulmonary capillary wedge pressure.
zoom view
Fig. 5: The data indicate that tolerance can develop to intravenous nitroglycerin (NTG) over 24 hours. There is a brisk initial response to IV NTG manifested by a fall in pulmonary capillary wedge pressure (PCWP) during titration; but during 24 hours of infusion, PCWP increases back toward control in both the NTG and the placebo arms of the study.
Dyspnea is relieved. Larger doses lead to arteriolar dilation, further reducing afterload and improving forward flow. LV cavity size diminishes, reducing mitral regurgitation.20 It is not surprising that oral nitrate therapy has emerged as an important treatment for systolic heart failure. Nitrates are among the few vasodilators that are able to increase exercise tolerance in patients with systolic heart failure.21,22 However, nitrate tolerance occurs in many patients (Fig. 5), thus casting suspicion on long-term efficiency. This can be offset to some extent by concomitant use of hydralazine.15
 
RENIN-ANGIOTENSIN-ALDOSTERONE SYSTEM BLOCKERS
 
Angiotensin Converting Enzyme Inhibitors
Angiotensin converting enzyme (ACE) inhibitors were introduced into clinical practice in the 1980s for the treatment of hypertension and heart failure.10
zoom view
Figs 6A and B: In the CONSENSUS Trial, the difference between treatments is even more striking, as the patients likely had more advanced heart failure. Kaplan-Meier survival curves (A) from The CONSENSUS Trial Study Group. Effects of enalapril on mortality in severe congestive heart failure. Results of the Cooperative North Scandinavian Enalapril Survival Study (CONSENSUS).28
11
This class of drug therapy has revolutionized the treatment for these two conditions, and has been demonstrated to improve survival in patients with systolic heart failure (Figs 6A and B). The success of ACE inhibitors for the treatment of heart failure was predicated on the observation that the RAAS is activated in chronic systolic heart failure,23 and this activation contributes importantly to heightened afterload and to the LV remodeling process.
Angiotensinogen is produced in the liver and is converted in the blood by renin to form a small peptide, angiotensin I (Fig. 7). Angiotensin I is then further cleaved to form angiotensin II, a very small peptide, but potent arteriole constrictor. Angiotensin II subserves a host of other biological activities primarily through the angiotensin II receptor, including promotion of volume retention, activation of and sensitization to the sympathetic nervous system, thirst, regulation of salt and water balance, modulation of potassium balance, cardiac myocyte and vascular smooth muscle growth, to name but a few. Its actions are central to the development of acute and chronic systolic heart failure.
Early, overly simplistic thinking was that systolic heart failure was essentially a vasoconstricted state caused by excessive sympathetic nervous system activity and heightened levels of other vasoconstrictor neurohormones, including angiotensin II, arginine vasopressin, (AVP) and endothelin. When it became apparent that ACE inhibitors could block the production of angiotensin II, ACE inhibitors became an attractive candidate for the treatment of patients with hypertension and systolic heart failure. ACE inhibitors would be expected to reduce afterload, and in turn would increase cardiac output and forward flow. Although the initial clinical studies indeed supported this hypothesis,24 it soon became clear that ACE inhibitors were doing much more than reducing afterload. Long-term clinical improvement was accompanied by reduced LV remodeling and improved patient survival when applied to postmyocardial infarction patients,25 very similar to the seminal animal work of Pfeffer and colleagues.26 ACE inhibitors were no longer thought of as simple arteriolar dilators, but were neurohormone modulators that could very favorably alter the natural history of systolic heart failure and improve survival by inhibiting the progression of LV remodeling (Fig. 8).
We now recognize that neurohormonal activation plays a key role in the initiation and progression of heart failure. The RAAS is central to this neurohormonal cascade, as patients with systolic heart failure and high renin levels seemingly derive the most acute benefit from blocking the RAAS.27 It is now well established that ACE inhibitors slow the progression of heart failure and improve survival in patients with a reduced ejection fraction and congestive heart failure.28
12
zoom view
Fig. 7: The renin-angiotensin-aldosterone system.
13
zoom view
Fig. 8: Heart failure is a complex clinical syndrome characterized by extensive neuroendocrine activation. The release of neurohormones appears to be in response to reduced cardiac function and a perceived reduction in effective circulatory volume. It is as if neuroendocrine activity is attempting to protect the blood pressure and maintain circulatory homeostasis. Although this may be adaptive early on, chronic neuroendocrine activation leads to peripheral vasoconstriction, left ventricular remodeling and worsening left ventricular performance, and thus becomes an attractive therapeutic target. Drugs designed to block the exuberant neuroendocrine response, such as ACE inhibitors, have now become the cornerstone of treatment for heart failure.
Much of this improvement is believed to be due to “reverse remodeling”. Even patients with a reduced ejection fraction, but few or no heart failure symptoms, derive clinical benefit from ACE inhibitor therapy.29 The development of symptomatic heart failure is delayed in these patients. The activation of neurohormones (renin, norepinephrine and AVP) appears to occur early in the natural history of the syndrome, before symptoms occur.30 This observation suggested that early introduction of neurohormonal blocking drugs before symptoms ensue may slow the progression of systolic heart failure or even delay its onset of signs and symptoms.29 Indeed, today neurohumoral modulating drugs are recommended for patients who demonstrate impaired LV systolic function even in the absence of symptoms. Many investigators observed that the RAAS was markedly activated during decompensated heart failure, but returns to normal once the patient clinically stabilizes, even though severe LV dysfunction may persist.31 The concept that blocking the RAAS improves patients with systolic heart failure became widely recognized in the 1990s.14
In the 1980s, a number of hypotheses and concepts emerged that challenged the long-standing notion that systolic heart failure was fundamentally a mechanical problem. Katz introduced the idea that heart failure may be a disorder of abnormal gene expressional growth response to injury,32 and many others believed that the myocardial remodeling was at least in part due to activation of neurohormonal systems,33 which were well known to also be cardiac growth factors when studied in vitro.34 Alteration of loading conditions due to increased LV chamber size and increased wall stress also undoubtedly led to progressive LV remodeling.34 Both mechanical and neurohormonal signals regulated the remodeling process, as did altered gene expression. It became clear that the all-important LV remodeling process was largely structural and not functional.35 Additional data emerged indicating that excessive angiotensin II caused cardiac myocyte necrosis under experimental conditions.36 Eventually a coherent story emerged suggesting that systolic heart failure was at least in part driven by excessive neurohormonal activation,37,38 setting up a vicious cycle of worsening heart failure and death (Fig. 8). These neurohormonal systems are likely adaptive in an evolutionary sense,39 and are not simple biomarkers or epiphenomena. They are known to directly contribute to LV remodeling4042 and subsequent patient mortality.43 The strong notion emerged that pharmacological inhibition of the RAAS (and the sympathetic nervous system) might reduce the progression of LV remodeling,4447 and, therefore, such drugs should improve patient survival.28
The ACE inhibitors were the first class of drugs to really test the neurohumoral hypothesis (Figs 6A and B). Needless to say, they have now become a standard of care for patients with hypertension, systolic heart failure, acute myocardial infarction and advanced cardiovascular disease. The ACE inhibitor class of drugs reduces afterload, presumably by inhibiting angiotensin II arteriolar constriction reducing sympathetic tone. There is also venodilation with a fall in pulmonary capillary wedge pressure, presumably due to reduction in sympathetic activity to veins and desensitization of venous capacitance vessels to norepinephrine. Angiotensin II does not directly dilate veins, so there is no direct effect of ACE inhibitors on venous capacitance vessels. There is modest improvement in cardiac index with ACE inhibitors, and heart rate may slightly slow. As previously mentioned, if the patient is acutely hyper-reninemic as a consequence of vigorous diuresis, there can be a substantial and prolonged fall in blood pressure with even small doses of ACE inhibition. This is why many physicians prefer to use short-acting ACE inhibitors such as captopril in hospitalized patients with acute systolic heart failure, as patients are less likely to develop prolonged symptomatic hypotension. If symptomatic hypotension ensues, the patient 15should lie down and the feet should be elevated until these symptoms resolve and the blood pressure improves. Usually a sense of improved well-being is established with the use of ACE inhibitors despite chronically low arterial pressures. Rarely, dysgeusia or loss of taste occurs, sometimes requiring withdrawal of the drug. Rash is uncommon with the smaller doses of ACE inhibitors used today. A dry, non-productive cough occurs in some patients receiving ACE inhibitors, and the drug is discontinued in 5–10% of patients for this reason. The mechanism of the cough is not entirely clear, but is believed to be due to the effects of bradykinin on sensory neurons in the proximal airways.
There is now a long list of ACE inhibitors to choose from (Table 2). They have somewhat dissimilar pharmacodynamics, pharmacokinetics, and rates of elimination. In general, it is best to start with small doses of an ACE inhibitor that has been tested in a large clinical trial and slowly titrates up over days to weeks to a target dose established as safe and effective by use in large clinical trials. It is expected that many patients with advanced systolic heart failure will have about a 20% increase in serum creatinine with ACE inhibitor use. This is usually not reason to discontinue or lower the dose of the ACE inhibitor. However, this class of drug is contraindicated in patients with cardiogenic shock or acute renal failure, and can cause renal insufficiency when used in patients with renal artery stenosis. Occasionally, hyperkalemia can occur requiring alteration of dose or temporary/permanent discontinuation of the ACE inhibitor. Careful, regular follow-up with monitoring of electrolytes, BUN (blood urea nitrogen) and serum creatinine is important in the care of these patients when making decisions about altering the dose of ACE inhibitors. Renal function and serum electrolytes should be checked at about one week following initiation of ACE inhibitor therapy.
 
Angiotensin Receptor Blockers
Angiotensin receptors of the AT1 subtype bind angiotensin II with a high structural specificity but limited binding capacity.48 The remarkable success of ACE inhibitors in the treatment of hypertension, arterial disease, myocardial hypertrophy, heart failure and diabetic renal disease encouraged the exploration of alternative drugs to block the RAAS. It was eventually recognized that ACE inhibitors blocked only one of several pathways that normally increase angiotensin II activity, and that angiotensin II could “escape” from chronic ACE inhibition. ARBs do not demonstrate this “escape” phenomenon. ARBs do not cause cough. They can be used safely in patients who develop angioedema during treatment with an ACE inhibitor. Increased levels of angiotensin II peptides seen with the use of ARBs do not appear to have unexpected off-target effects despite activating AT2 receptors.16
Table 2   Common drugs used in managing chronic heart failure in the United States
Drug
Trade name
Heart failure indication
Post-myocardial infarction indication
Dosing
Angiotensin-converting enzyme (ACE inhibitors)
Benazepril
Lotensin
No
No
5–40 mg QD
Captopril
Capoten
Yes
No
6.25–150 mg TID
Enalapril
Vasotec
Yes
No
2.5–20 mg BID
Fosinopril
Monopril
Yes
No
10–80 mg QD
Lisinopril
Prinivil, Zestril
Yes
No
5–20 mg QD
Moexipril
Univasc
No
No
7.5–60 mg QD
Perindopril
Aceon
No
No
2–16 mg QD
Quinapril
Accupril
Yes
No
5–20 mg BID
Ramipril
Altace
Yes
Yes
2.5–20 mg QD
Trandolapril
Mavik
No
Yes
1–4 mg QD
Zofenopril
Bifril
NA
NA
7.5–60 mg QD
Angiotensin II receptor blockers (ARBs)
Candesartan
Atacand
Yes
No
8–32 mg QD/BID
Eprosartan
Teveten
No
No
400–800 mg QD
Irbesartan
Avapro
No
No
150–300 QD
Losartan
Cozaar
No
No
50–100 mg QD/BID
Telmisartan
Telma
No
No
40–80 QD
Olmesartan
Benicar
No
No
20–40 mg QD
Valsartan
Diovan
Yes
No
80–320 mg QD
β-adrenergic receptor antagonists
Carvedilol
Coreg
Yes
Yes
3.125–25 mg BID
Metoprolol succinate
Toprol XL
Yes
No
25–200 mg QD
Bisoprolol
Zebeta
No
No
1.25–10 mg QD
Nebivolol
Nabilet
No
No
1.25–10 mg QD
Aldosterone receptor antagonists
Spironolactone
Aldactone
Yes
No
25–50 mg QD
Eplerenone
Inspra
No
Yes
25–50 mg QD
Others
Amlodipine
Norvasc
No
No
2.5–10 mg QD
Hydralazine-isosorbide dinitrate
BiDil (37.5/20)
Yes
No
1–2 tablets TID
Digoxin
Digitek
Yes
No
0.125–0.25 mg QD
BID, twice daily; QD, once daily; TID, three times daily. Italics indicate drugs that are currently not indicated by the US Food and Drug Administration for treating patients with heart failure).
17
First-dose hypotension is not typically seen when ARBs are given to diuretic-treated patients, as often occurs with ACE inhibitors. This is probably because ARBs have a much slower onset of action. Orthostatic hypotension is rare. Rebound hypertension upon withdrawal of ARBs does not appear to be a problem. As with ACE inhibitors, acute renal failure may occur with ARBs if administered to patients with renal artery stenosis or cardiogenic shock. The incidence of renal dysfunction and hyperkalemia is comparable with ARBs and ACE inhibitors.49 It is now reasonably clear that ACE inhibitors and ARBs should not be used together, as the likelihood of hyperkalemia, hypotension and worsening renal function is greater.50
Many randomized controlled trials of ARBs have been performed in patients with chronic systolic heart failure,51,52 in patients with acute myocardial infarction complicated by heart failure or LV dysfunction,53 and in patients at high-risk for vascular events.54 Several important points have emerged from these large trials:
  • ARBs and ACE inhibitors appear to have very similar efficacy in these patient groups
  • If the patient does not tolerate an ACE inhibitor, an ARB is a suitable substitution
  • Although generally more expensive, ARBs are better tolerated than ACE inhibitors
  • The combination of an ACE inhibitor and an ARB (dual RAAS blocking effect) is not more effective and is associated with more hypotension, worsening renal function, and hyperkalemia55
  • Despite earlier favorable reports, ARBs do not appear to prevent recurrent atrial fibrillation.56
The dose of ARBs has generally been determined by pharmaceutical generated data and subsequent verification of these doses in large clinical trials (Table 2). Extensive experience with RAAS blockers over the years has led to changes in dose recommendations. For example, the Heart failure Endpoint evaluation of Angiotensin II Antagonist Losartan (HEAAL) trial demonstrated that losartan 150 mg daily reduced the rate of death or admission for heart failure to a greater extent than a dose of 50 mg/day.57
Similar to ACE inhibitors, we now have data to suggest that inhibition of the RAAS with ARBs also results in favorable structural and functional changes. Treatment with the ACE inhibitor captopril, the ARB valsartan, or the combination of captopril plus valsartan resulted in similar changes in cardiac volume, ejection fraction and infarct segment length in patients 20 months following acute myocardial infarction.58 These observations suggest that ARBs are similar to ACE 18inhibitors with regard to their anti-remodeling properties. Neither ACE inhibitors nor ARBs improve outcomes in patients with heart failure with preserved ejection fraction.59
 
MINERALOCORTICOID (ALDOSTERONE) RECEPTOR BLOCKERS
 
Aldosterone and Systolic Heart Failure
Aldosterone was structurally identified more than 50 years ago, and was soon after designated a mineralocorticoid due to its salt retaining properties. It also promotes loss of potassium from the kidney, gastrointestinal tract, sweat and salivary glands. It has long been known to play a pathophysiologic role in cardiovascular disease, including congestive heart failure (Fig. 9).60,61 In addition to its mineralocorticoid properties, which can cause hypokalemia and hypomagnesemia, aldosterone contributes in many ways to the development of heart failure. It likely causes vascular and cardiac remodeling, endothelial dysfunction, inhibits norepinephrine reuptake, and causes baroreceptor dysfunction (Fig. 9). It expands intravascular and extravascular volume. Inhibition of aldosterone is believed to be favorable due to:
  • Reduced collagen deposition and possibly anti-remodeling effects
  • Blood pressure reduction
  • Prevention of hypokalemia and associated arrhythmias
  • Modulation of nitric oxide synthesis.
zoom view
Fig. 9: Aldosterone is a mineralocorticoid that has a central role in a host of biological activities. Many of these activities can be excessive due to dysregulation of aldosterone activity, thus contributing to cardiovascular disease.
19
The major mineralocorticoid in heart failure is cortisol and not aldosterone. Serum aldosterone levels are not consistently elevated in patients with heart failure in the absence of diuretics. Accordingly, it is not aldosterone blockade per se, but mineralocorticoid receptor blockade that is important. Spironolactone and eplerenone are thus mineralocorticoid receptor blockers more than simply aldosterone receptor blockers.
ACE inhibitors were originally believed to chronically suppress angiotensin II in patients with heart failure, a major determinant of aldosterone production by the adrenal glands. This notion probably led to some initial loss of interest in aldosterone receptor inhibitors for the treatment of systolic heart failure. We now know that ACE inhibitors do not suppress angiotensin II long-term, and that there is an aldosterone escape phenomenon. Three landmark studies, the Randomized Aldosterone Evaluation Study (RALES) (Fig. 10),62 the Eplerenone Post-acute Myocardial Infarction Heart Failure Efficacy and Survival Study (EPHESUS) (Fig. 11)63 and the Eplerenone in Mild Patients Hospitalization and Survival Study in Heart Failure (EMPHASIS-HF)64 have remarkably increased the role of aldosterone mineralocorticoid antagonists for the everyday treatment of systolic heart failure. Eplerenone, as compared with placebo, reduced both the risk of death and the risk of hospitalization among patients with systolic heart failure and mild symptoms in the EMPHASIS-HF trial.
zoom view
Fig. 10: Survival curves of patients with advanced heart failure randomly allocated to spironolactone or placebo. Most patients were not receiving β-adrenergic blockers. There was a 30% reduction in mortality in patients randomized to spironolactone compared to patients in the placebo group. From the Randomized Aldactone Evaluation Study (RALES).62
20
zoom view
Fig. 11: Kaplan-Meier estimates of the rate of death from any cause in the EPHESUS trial.63
This finding is particularly important as it expands the use of mineralocorticoid antagonists for the New York Heart Association (NYHA) functional class II patients. Spironolactone62 and eplerenone63 are now widely used to treat chronic systolic heart failure and postmyocardial infarction heart failure. Despite their greater use today, in the USA it was estimated that less than one-third of eligible patients hospitalized for heart failure received appropriate, guideline-recommended aldosterone antagonist therapy.65 Some of the reluctance to use aldosterone blockers in patients with systolic heart failure may be justified because of the advanced age of patients, the frequency of chronic renal insufficiency, other common comorbidities such as diabetes mellitus, and the serious threat of hyperkalemia.66 However, when used according to protocol, hyperkalemia is seemingly not such a major problem. Careful follow-up of patients and frequent measurement of renal function and serum potassium are necessary to ensure safety when using aldosterone receptor blocking drugs.
The RAAS is likely an ancient (~400–600 million years) system that evolved in such a way as to allow them to adapt to salt and volume depletion, as might have occurred during transition from the sea to land eons ago. The notion is that regulation of salt and water retention is adaptive, perhaps by protecting intravascular volume, blood pressure and perfusion to vital organs. We now know that chronic stimulation of the RAAS in patients with heart failure can be maladaptive, and that pharmacologically blocking the RAAS can improve patient survival. Blockade of aldosterone membrane receptors is a widely accepted form of therapy for systolic heart failure. The RALES and EPHESUS studies provide strong evidence that aldosterone mineralocorticoid receptor blockade is effective 21therapy for patients with heart failure across all degrees of severity. Postmyocardial infarction heart failure is also improved by mineralocorticoid receptor blockers.63 The role of nuclear aldosterone receptors is less clear, but given the complex array of regulatory properties that angiotensin II and aldosterone demonstrate, including inflammation, collagen synthesis, cytokine production, regulation of nitric oxide and cell adhesion molecules, one has to suspect that the activation of nuclear aldosterone receptors with resultant regulation of selective gene expression is also responsible for many of the biological activities of aldosterone, some of which are seen in systolic heart failure.
 
Spironolactone and Eplerenone in Chronic Heart Failure
The mechanism of action of spironolactone is complex, as aldosterone mineralocorticoid modulates many features of the heart failure syndrome. Although spironolactone is still used as an antihypertensive agent, it is not considered to be a “vasodilator” in the usual sense. Patients taking spironolactone need to be frequently and carefully monitored (patients in RALES were seen monthly for the first 12 weeks), as hyperkalemia and azotemia can occur with spironolactone,65 particularly if non-steroidal anti-inflammatory drugs are used concomitantly. Diabetes mellitus, chronic kidney disease, volume depletion, advanced age and use of other potassium sparing agents and non-steroidal anti-inflammatory drugs are all risk factors for the development of hyperkalemia when using RAAS blocking drugs.61 With careful monitoring, however, serious hyperkalemia is uncommon.62
Because of the central importance of aldosterone in the pathophysiology of heart failure, it is not surprising that the aldosterone receptor blocker spironolactone has emerged as an important therapy. Spironolactone is an old drug that was primarily used in large doses to treat ascites, edema and refractory hypertension. Excessive mineralocorticoid, common in patients with heart failure, promotes sodium retention, loss of magnesium and potassium, sympathetic nervous system activation, parasympathetic nervous system inhibition, myocardial and vascular fibrosis, baroreceptor dysfunction, and impaired arterial compliance.67
The definitive RALES was published in 199962 and clearly demonstrated that spironolactone (25–50 mg/day) added to standard therapy (β-blockers were not yet in widespread use) was safe and reduced mortality by 30% (Fig. 7). Death from progressive heart failure and sudden death were both reduced by spironolactone. The patients who participated in RALES were primarily NYHA class III (70%) and IV (30%).22
Eplerenone, a newer, more selective aldosterone mineralocorticoid receptor blocker, causes less gynecomastia and breast tenderness than spironolactone. It is more mineralocorticoid specific than spironolactone. EPHESUS63 was conducted in patients who experienced a recent acute myocardial infarction with an ejection fraction of 40% or less who had heart failure, or had a history of diabetes mellitus. The patients in EPHESUS were randomly allocated to eplerenone or placebo in addition to standard therapy for acute myocardial infarction. In EPHESUS, eplerenone (average dose 42.6 mg/day) reduced all-cause mortality by 15%, cardiovascular mortality by 17%, and significantly lowered the need for subsequent hospitalization (Fig. 11). Sudden cardiac death was also reduced. As with RALES, serious hyperkalemia was unusual.
The EMPHASIS-HF trial suggests that eplerenone is effective in patients with systolic heart failure and NYHA functional class II symptoms. In EMPHASIS-HF hospitalizations for heart failure and for any cause were also reduced with eplerenone. A serum potassium level exceeding 5.5 mmol/L occurred in 11.8% of patients in the eplerenone group and 7.2% of those in the placebo group (P <0.001). Today, aldosterone mineralocorticoid antagonists are widely used to treat advanced heart failure and selected patients with acute myocardial infarction. However, less than one-third of eligible patients hospitalized for heart failure are receiving guideline-recommended aldosterone receptor blocking drugs.64 This is perhaps due in part to the need for more frequent and careful follow-up, and the fear of hyperkalemia. There is a perception by some physicians that this class of drugs poses more risk than other RAAS blockers. Nevertheless, aldosterone receptor blockers are effective and safe when properly prescribed and monitored, and their indications are seemingly expanding. There appears to be considerably less reverse remodeling in patients with mild-to-moderate heart failure and LV systolic dysfunction randomly assigned to eplerenone, even though there is a reduction in collagen turnover and a reduction in brain natriuretic peptide (BNP) factor.68 Despite these surprising neutral effects on reverse remodeling, the results of EMPHASIS-HF trial suggest that patients with mild-to-moderate systolic heart failure still derive a favorable effect on morbidity and mortality from eplerenone.
Recently, the Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist (TOPCAT) trial has been completed and published.69 This was a study aimed at patients with heart failure with a preserved ejection fraction rather than heart failure with a reduced ejection fraction. Overall, there was no improvement in cardiovascular mortality. However, spironolactone was associated with a significant (P = 0.04) reduction in hospitalization (17%). It is unclear how physicians will adopt the results of TOPCAT.23
zoom view
Fig. 12: Chemical structure of LCZ696.
 
LCZ696
A new therapy for hypertension and heart failure has been developed by Novartis. LCZ696 is a novel molecule that includes both valsartan and a neprilysin inhibitor. The valsartan moiety of the molecule suppresses the renin-angiotensin system (RAS) while the neprilysin inhibitor reduces the degradation of brain natriuretic peptide (BNP), thereby increasing circulating BNP in plasma (Fig. 12). LCZ696 is taken twice per day. In a recent large clinical trial, 8436 patients with a reduced ejection fraction and NYHA class II to IV symptoms were randomly allocated to LCZ696 at 200 mg twice daily or enalapril at 10 mg twice daily. A favorable effect on survival was observed with LCZ696 relative to enalapril.70 It is possible that this new form of therapy may emerge as a prominent form of treatment for patients with heart failure and reduced ejection fraction.
 
PHOSPHODIESTERASE TYPE 5 INHIBITORS
 
Sildenafil and Tadalafil
Phosphodiesterases are enzymes that hydrolyze the cyclic nucleotides—cGMP and cyclic adenosine monophosphate. At least eleven families of phosphodiesterase isoenzymes have been identified. Phosphodiesterase 5 (PDE 5) degrades cGMP 24via hydrolysis, thus influencing cGMP's ability to modulate smooth muscle tone,71 particularly in the venous system of the penile corpus cavernosum and in the pulmonary vasculature. The discovery of sildenafil, a highly selective inhibitor of PDE 5, was initially aimed to be a novel treatment for coronary artery disease. The initial clinical studies in the early 1990s were not promising for this target, but the off-target effect of enhancement of penile erections did not escape the notice of investigators. The use of PDE 5 inhibitors was then redirected toward erectile dysfunction and more recently pulmonary hypertension.
Nitric oxide activates soluble guanylate cyclase, stimulating the production of cGMP. PDE 5 normally hydrolyzes cGMP. Sildenafil inhibits PDE 5, leading to increased cGMP and vasodilation in response to nitric oxide. For years it was known that PDE 5 was not present in normal cardiac myocytes, and the heart itself was not considered an appropriate target. This was challenged by Kass and colleagues72 who demonstrated that inhibiting PDE 5 in hypertrophied RVs induces a positive inotropic response.73 In fact, PDE 5 is markedly upregulated in hypertrophied ventricles, and PDE 5 inhibition may lead to regression of RV hypertrophy.73 PDE 5 has long been known to be highly expressed in lung vasculature, and so it is not surprising that sildenafil is beneficial for the treatment of patients with pulmonary hypertension. As of this writing, it is still not clear if normal cardiac myocytes express PDE 5, but hypertrophied and/or failing myocytes do express it, and PDE 5 inhibition can be clinically helpful in patients with pulmonary hypertension and some element of right ventricular hypertrophy or failing right ventricle.
Sildenafil and tadalafil are both PDE 5 inhibitors that are indicated for use in patients with pulmonary arterial hypertension who have mild to moderately severe symptoms.74 Preliminary data on sildenafil suggest that its use may also be safe and beneficial in patients with disproportionate pulmonary hypertension and LV dysfunction.75,76 Sildenafil citrate is prescribed in doses of 20 mg TID and tadalafil is much longer acting and is prescribed in doses of 40 mg/day to control pulmonary hypertension. Hypotension can occur with PDE 5 inhibitors, especially when they are used with nitrates. Visual disturbances and priapism have also been observed with this class of drugs. There is no specific antidote for PDE 5 induced hypotension. Sildenafil and tadalafil are not approved for use in patients with heart failure, but they are being investigated. A small case series (3 patients) has recently implied that a combination of sildenafil and nitrates can be used in patients with heart failure and pulmonary hypertension,77 though clearly more robust clinical trials are needed. Experimental data indicate that PDE 5 levels are increased in severely failing 25hearts78 and that sildenafil reduces myocardial remodeling.79 Recent data also suggest that PDE 5 is regulated in the LV by oxidative stress.80 Clearly this story is still unfolding and we have much to learn. Nevertheless, drugs such as sildenafil and tadalafil that selectively restore right ventricular contractility, limit right ventricular hypertrophy and reduce pulmonary artery remodeling are intriguing as potential therapy for right heart failure due to disproportionately increased pulmonary artery pressure. Perhaps PDE 5 inhibitors will also favorably affect left-sided systolic heart failure, particularly if there is associated pulmonary hypertension. More studies are needed, and use of these drugs for the treatment of heart failure remains investigational for now.
Accordingly, the Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in Heart Failure with Preserved Ejection Fraction (RELAX) trial was designed to test the hypothesis that, compared with placebo, therapy with the PDE-5 inhibitor sildenafil would improve exercise capacity in heart failure with preserved ejection fraction (HFpEF) after 24 weeks of therapy, assessed by the change in peak oxygen consumption.81 Among these patients with HFpEF, sildenafil did not significantly improve exercise capacity or clinical status relative to placebo. Nevertheless, many experts believe that there still may be a role for phosphodiesterase-5 inhibitors in selected patients with HFpEF and disproportionate pulmonary hypertension.
 
INTRAVENOUS VASODILATORS
 
Nitroprusside
Sodium nitroprusside can be dramatic in reversing the deleterious hemodynamics of acute systolic heart failure. Those who have had experienced using the drug in this setting are often astonished how quickly the drug lowers pulmonary capillary wedge pressure (PCWP) and improves cardiac output, leading to prompt and often striking clinic improvement. The drug is usually started as doses of 10 μg/min, and gradually titrated up to no more than 400 μg/min, as needed to control hemodynamic abnormalities and symptoms. Some clinicians give nitroprusside according to body weight, with the typical dose starting at 10–20 μg/kg/min. Our extensive experience with nitroprusside suggests that with low dose infusion rates (<3 μg/kg/min) used for less than 72 hours, toxicity is almost never observed.82 The systolic blood pressure should not be allowed to be less than 90 mmHg or to a level that includes hypotensive symptoms. Invasive monitoring with a pulmonary artery catheter and an arterial catheter can be useful if the patient has marginal blood pressure. Persistent 26or severe hypotension will nearly always dissipate as soon as nitroprusside is stopped.
 
Metabolism and Toxicity of Nitroprusside
Nitroprusside has been used to treat severe heart failure for many years,83 though the Food and Drug Administration (FDA) has approved it only for severe hypertension and for certain neurosurgical procedures. It must be used carefully by experienced nurses and clinicians. Thiocyanate toxicity can rarely occur, and thiocyanate levels should be monitored, particularly if the patient has received a high dose for a prolonged period of time. Measurement of thiocyanate is a simple, inexpensive colorimetric test, normal levels being less than 10 mg/mL. Metabolic acidosis, anuria, and a prolonged high dose of nitroprusside (>400 μg/min) can predispose to thiocyanate toxicity, prompting measurement of thiocyanate levels. The thiocyanate ion is also readily removed by hemodialysis. When thiocyanate toxicity does occur, the patient may present with confusion, hyperreflexia and convulsions. Occasionally, mild hypoxemia occurs from nitroprusside due to ventilation-perfusion mismatch, but it is usually of little clinical consequence, as cardiac output rises and the delivery of oxygen to tissues increases. Coronary “steal” can occur when nitroprusside is used in the setting of acute myocardial infarction, and it should not be used routinely in this setting.84,85 If intravenous vasodilator therapy is used for patients with acute myocardial infarction and severe heart failure, intravenous nitroglycerin may be preferred. Nevertheless, nitroprusside has been used successfully in this setting when given in the subacute phase.84 If nitroprusside is used to treat severe heart failure related to acute myocardial infarction, it should be given later, perhaps 12 hours after admission to the hospital.85
 
Nitroprusside and Severe Heart Failure
Nitroprusside quickly improves hemodynamics and symptoms in patients with severe heart failure.86 Even patients with hypotension and shock may improve with nitroprusside,87 as blood pressure may stabilize or even improve with large increases in cardiac output. Patients with severe mitral regurgitations or aortic regurgitation may also demonstrate dramatic clinical improvement with nitroprusside. Patients with severe aortic stenosis and worsening heart failure can be improved with nitroprusside used prior to aortic value replacement,88 provided they are not hypotensive. It can also be used to stabilize acute heart failure in patients who demonstrate a ruptured interventricular septum following acute myocardial infarction. Recent observational data indicate that in patients hospitalized with advanced, low-output heart failure, those 27stabilized in the hospital with nitroprusside may have a more favorable long-term clinical outcome.89
 
Intravenous Nitroglycerin
Similar to nitroprusside, intravenous nitroglycerin has an immediate onset and offset of action. The infusion rate is usually initiated at 10–20 μg/min with gradual titration to 200–400 μg/min as needed to control symptoms and improve hemodynamic parameters. It is not approved by the FDA for the treatment of heart failure, but has been widely used for this indication over the past 20 years. Intravenous nitroglycerin is endothelium dependent, and unlike nitroprusside, it has more effect on the venous circulation than on the arterial circulation. However, higher doses of intravenous nitroglycerin have arteriolar dilating properties and may decrease afterload. Therefore, cardiac output may increase and blood pressure can be maintained. PCWP is reduced. Mitral regurgitation improves. There are few data available on the effects of intravenous nitroglycerin on coronary circulation in patients with heart failure. Coronary blood flow appears to improve. This suggests that both the epicardial conductance vessels and the coronary arteriolar resistance vessels are favorably influenced by intravenous nitroglycerin.
 
Limitations of Intravenous Nitroglycerin in the Treatment of Patients with Heart Failure
Intravenous nitroglycerin causes headaches in about 20% of patients, and when severe, may require cessation of the infusion. Hypotension (10%), nausea and bradycardia occasionally occur. Some patients are relatively resistant to intravenous nitroglycerin and seemingly require very large doses to afford a hemodynamic effect. The reason for this is not particularly clear, but very large doses in excess of 500 μg/min are best avoided. Nitrate tolerance is said to occur when there is a robust initial hemodynamic response, but by 1–2 hours the dose of intravenous nitroglycerin must be increased to establish a continued hemodynamic response. About one-half of patients develop nitrate tolerance, and it cannot be predicted by baseline hemodynamic values (Fig. 5). The mechanism of resistance to intravenous nitroglycerin is not clear, but it is possibly prevented by concomitant use of oral hydralazine (Fig. 2).
 
Nesiritide
Nesiritide is pure, human BNP synthesized using recombinant DNA techniques. It has the same 32-amino acid sequence as endogenous BNP released from the heart where it is synthesized and stored. When infused intravenously into the 28circulation of patients with heart failure, the mean terminal elimination half-life of nesiritide is about 18 minutes. Plasma BNP levels increase about three to six-fold with a nesiritide infusion. Human BNP is eliminated from the circulation through complex, multiple mechanisms. Most of the BNP is cleared by c-receptors on cell surfaces, but some is cleared by neutral endopeptidases in renal tubular and vascular cells, and a smaller amount is cleared by renal filtration that is proportional to body weight.
The earliest clinical trial of nesiritide, Vasodilation in the Management of Acute CHF (VMAC), was a comparison study with intravenous nitroglycerin.90 It demonstrated that nesiritide improved hemodynamic function and self-reported symptoms more effectively than intravenous nitroglycerin or placebo (Figs 13A and B). On this basis, nesiritide was approved by the FDA for heart failure and became widely used. Nesiritide has venous, arterial and coronary vasodilator properties. Cardiac output improves and PCWP is reduced. Hypotension occurs in about 4% of patients, and unlike intravenous nitroglycerin, it can be prolonged (~20 minutes) because of nesiritide's relatively longer half-life. The effects of nesiritide on renal function are variable, but generally only a modest or neutral renal effect is observed, though worsening renal function has been reported.91,92
In 2005 Sackner-Bernstein and colleagues reported that nesiritide may be associated with an increased risk of death after treatment for acute decompensated heart failure.93 At about this time, infusions of nesiritide were also being widely performed in outpatient clinics, and the drug came under severe criticism.94,95 Ultimately, an outpatient randomized controlled trial of nesiritide vs. placebo was performed which demonstrated that serial outpatient nesiritide infusions did not provide a demonstrable clinical benefit over standard therapy.96 The drug rapidly fell out of favor.97 Ultimately, the Acute Study of Clinical Effectiveness of Nesiritide in Decompensated Heart Failure (ASCEND-HF) trial was designed to evaluate the effect of nesiritide, in addition to standard care, on rates of self-reported dyspnea at 6 and 24 hours, re-hospitalization for heart failure or death from any cause at 30 days, and renal dysfunction. The study included more than 7,000 patients and concluded that the IV vasodilator nesiritide did not improve survival or re-hospitalization relative to placebo, but had a small, non-significant effect on dyspnea when used in combination with other therapies. It also did not compromise renal function within a month of its use in acute decompensated heart failure. Therefore, the use of nesiritide for patients with acute decompensated heart failure has further waned over the years, while less expensive intravenous vasodilators continue to be employed.29
zoom view
Figs 13a and b: Changes in pulmonary capillary wedge pressure from baseline in response to intravenous nitroglycerin, nesiritide and placebo in patients with heart failure.90
30
 
ORAL β-ADRENERGIC BLOCKING DRUGS
There is a fundamental belief that the biologically powerful adrenergic nervous system compensates for the failing heart by increasing myocyte size (hypertrophy), heart rate and force of contraction (inotropy). The sympathetic nervous system also activates the RAAS, thus conserving intravascular volume and redirecting blood flow to vital organs. However, an overly active sympathetic nervous system has repeatedly been shown to be essentially toxic to myocardial cells in both animals and humans.98 There have been numerous large randomized trials supporting the concept that blocking the sympathetic nervous system with β-adrenergic blocking drugs in patients with systolic heart failure slows the progression of systolic heart failure and improves patient survival (Fig. 14).
The importance of dysfunctional adrenergic activation in heart failure was first elucidated by work of Braunwald and colleagues at the National Institutes of Health in the 1960s.99 Since then, there has been an enormous basic and clinical research effort testing the rather counterintuitive concept that blocking the β1 and β2-adrenergic receptors will benefit patients with systolic heart failure.100 It is well known that β-adrenergic receptors downregulate in response to excessive sympathetic drive,101 presumably in an attempt to protect the cardiac myocyte from overstimulation. Such biological behavior suggests that blocking the receptors pharmacologically may also protect the heart.102 Moreover, pheochromocytoma (a classic example of long-term hyperadrenergic activity) is well known to express itself as dilated or hypertrophic cardiomyopathy.103 This provides additional proof of concept that the overly active sympathetic nervous system (SNS) and its dysfunctional status is central to the pathophysiology of heart failure,104107 similar to the overly active RAAS.
The first use of β-adrenergic blockers to treat patients with heart failure was the product of a series of carefully written case reports from Göteborg, Sweden.108110 This experience was a source of both great excitement and profound skepticism. Eventually, a small clinical trial (Metoprolol in Dilated Cardiomyopathy [MDC]) was launched, but showed only marginal benefit of metoprolol in patients with heart failure.111 Other clinical trials were performed using bisoprolol (The Cardiac Insufficiency Bisoprolol Study [CIBIS] and CIBIS II)112,113 and metoprolol succinate (the Metoprolol CR/XL Randomized Intervention Trial in Congestive Heart Failure [MERIT-HF]).114 Carvedilol, an α1 and non-selective β-adrenergic blocker, was also demonstrated to improve survival in patients with moderate and even very severe heart failure (The Carvedilol Prospective Randomized Cumulative Survival [COPERNICUS] Trial).115 Some would argue that the α1-adrenergic receptor blockade induced by carvedilol provides an 31additional advantage to standard β-adrenergic blockade,115117 but this has remained controversial.
zoom view
Fig. 14: Placebo-controlled studies of β-blocker therapy.
32
Today β-adrenergic blockers are widely used throughout the world to treat patients with systolic heart failure.118 They are considered “evidence-based” therapy. The suggested initial dose and evidence-based maximal dose are shown in Table 2.
Although it is unusual to see patients with heart failure who are naive to either RAAS inhibitors or β-blockers, occasionally the issue of which class of drug to initiate first arises. Experience indicates that either RAAS blockers (i.e., ACE inhibitor or ARB) or a β-blocker may be initiated first,119 but that eventually full-doses of both classes of drugs should be attempted. The titration schedule of β-adrenergic drugs should be slow, that is over several weeks. The magnitude of heart rate reduction is significantly associated with the survival benefit of β-blockers in patients with systolic heart failure, whereas the dose of β-blocker is not.120 There is also a strong correlation between change in heart rate and improvement in LV ejection fraction.121 It appears as though decreased heart rate, improved LV chamber performance and afterload reduction each contribute to enhanced LV ejection fraction with use of carvedilol.122
β-adrenergic blocking drugs are now widely used to treat all stages of heart failure. Some patients admitted to hospital with NYHA class III or IV systolic heart failure may not tolerate β-blockers because of symptomatic hypotension or low cardiac output, but most hospitalized patients with acute heart failure do tolerate these drugs. The continuation of β-blocker therapy in patients hospitalized with acute decompensated systolic heart failure is associated with lower post-discharge mortality risk and improved treatment rates.123 Withdrawal of β-blocker therapy in the hospital is associated with a higher risk. β-blocker therapy before and during hospitalization for acute systolic heart failure is associated with improved outcomes.124 In our experience, the most common documented cause of discontinuance of β-blockers in patients with heart failure is failure to restart β-blockers after they have been stopped during hospitalization.125
Not all patients with systolic heart failure improve with β-blocking therapy. One possibility is that functional improvement from β-blockers may be related to changes in myocardial contractile protein gene expression,126 which could vary from patient to patient. Another possibility is that β-blocking drugs are quite different from each other. Metoprolol and bisoprolol are both β-receptor subtype selective (i.e., β1). Bucindolol, labetalol and carvedilol are each non-selective, and labetalol and carvedilol have α1 blocking properties that produce ancillary vasodilation. Bucindolol, though not generally available, has been intensely studied and has mild vasodilator properties, probably mediated by cGMP. 33Additionally, bucindolol has meager “inverse agonism”, so there is less negative chronotropism and inotropic effects. Bucindolol can also lower systemic norepinephrine levels substantially in some patients, and therefore has the potential to be a powerful sympatholytic agent. The norepinephrine lowering effects of bucindolol, as well as the clinical response to the drug, are strongly influenced by the pre-synaptic α2c-adrenergic receptors, which modulate exocytosis and exhibit substantial genetic variation in humans. It is believed that a α2c-adrenergic receptor polymorphism affects the sympatholytic effects of bucindolol in patients with systolic heart failure.127 Patients with the α2c-Del 322-325 polymorphism appear to have a marked increase in the sympatholytic response to bucindolol, and these carriers exhibit no evidence of clinical efficacy when treated with bucindolol. This concept is consistent with observations from other studies that indicate a marked decrease in plasma norepinephrine levels as a consequence of certain drug therapy, such as moxonidine, is associated with increased mortality and more heart failure hospitalizations. This seems also true with regard to the response to bucindolol where carriers of the α2c-Del 322-325 variant exhibit very low plasma norepinephrine levels during bucindolol use, and a poor response to treatment. The frequency of this genetic variant is ~0.04 in whites and ~0.40 in blacks.
In addition to their favorable effects on LV performance and patient survival, β-adrenergic blockers, like RAAS blockers, slow the progression of LV remodeling. This occurs in patients with heart failure secondary to an acute myocardial infarction and in patients with chronic heart failure from dilated cardiomyopathy. LV end-diastolic volume tends to improve, the LV becomes less spherical and assumes a more natural ellipsoid shape. Mitral regurgitation is ameliorated or improved, and on average the LV ejection fraction goes up by about 5–7 ejection fraction units. In some cases, there is spectacular reverse remodeling, and in other cases this is less apparent or may not be seen at all. Reverse remodeling of the LV is associated with improved survival. We now have three major heart failure therapeutic strategies aimed at producing reverse remodeling: RAAS blocking drugs, cardiac resynchronization therapy (CRT), and β-adrenergic blocking drugs. Of course, coronary revascularization can also improve LV size and performance in selected patients. These therapies have proven to be powerful drivers of improved patient survival.
 
CONCLUSION
Neurohumoral modulating drugs now have a central role in the treatment of patients with systolic heart failure. This was not the case 35 years ago when only digitalis and diuretics were used. 34Annualized mortality has fallen from approximately 20% to less than 10% per year commensurate with the use of RAAS and sympathetic nervous system blocking drugs. Of course, ICDs and CRT have also importantly contributed to this mortality reduction. The total cardiovascular death rate burden has fallen substantially in accordance with the widespread use of these therapies. Although, the incidence of STEMI (ST segment elevation myocardial infarction) has also fallen dramatically, incident systolic heart failure continues to be a major cause of hospitalization. There is now much better treatment for hypertension and hyperlipidemia. Paradoxically, as people live longer, we are now seeing a wave of heart failure in the elderly, the fastest growing segment of our population. The scourge of heart failure has not gone away, but has rather been shifted to people in their 70s, 80s, and 90s. In the end, prevention of heart failure by life-long control of known risk factors and mechanistic enlightenment though additional genomic studies may reduce the burden of heart failures even more, as systolic heart failure is likely a largely preventable disorder.
 
ACKNOWLEDGMENT
We acknowledge the outstanding help of Marisa Tirimacco in the preparation of this manuscript.
REFERENCES
  1. Imperial ES, Levy MN, Zieske H Jr. Outflow resistance as an independent determinant of cardiac performance. Circ Res. 1961;9:1148–55.
  1. Sonnenblick EH, Downing SE. Afterload as a primary determinant of ventricular performance. Am J Physiol. 1963;204:604–10.
  1. Wilcken DE, Charlier AA, Hoffman JI, et al. Effects of alterations in aortic impedance on the performance of the ventricles. Circ Res. 1964;14:283–93.
  1. Ross J Jr, Braunwald E. The Study of left ventricular function in man by increasing resistance to ventricular ejection with angiotensin. Circulation. 1964;29:739–49.
  1. Cohn JN. Blood pressure and cardiac performance. Am J Med. 1973;55:351–61.
  1. Meredith PA, Ostergren J, Anand I, et al. Clinical outcomes according to baseline blood pressure in patients with a low ejection fraction in the CHARM (Candesartan in Heart Failure: Assessment of Reduction in Mortality and Morbidity) Program. J Am Coll Cardiol. 2008;52:2000–7.
  1. Anand IS, Tam SW, Rector TS, et al. Influence of blood pressure on the effectiveness of a fixed-dose combination of isosorbide dinitrate and hydralazine in the African-American Heart Failure Trial. J Am Coll Cardiol. 2007;49:32–9.
  1. Rouleau JL, Roecker EB, Tendera M, et al. Influence of pretreatment systolic blood pressure on the effect of carvedilol in patients with severe chronic heart failure: the Carvedilol Prospective Randomized Cumulative Survival (COPERNICUS) study. J Am Coll Cardiol. 2004;43:1423–9.
  1. Franciosa JA, Pierpont G, Cohn JN. Hemodynamic improvement after oral hydralazine in left ventricular failure: a comparison with nitroprusside infusion in 16 patients. Ann Intern Med. 1977;86:388–93.
  1. Franciosa JA, Blank RC, Cohn JN. Nitrate effects on cardiac output and left ventricular outflow resistance in chronic congestive heart failure. Am J Med. 1978;64:207–13.35
  1. Olivari MT, Levine TB, Cohn JN. Abnormal neurohumoral response to nitroprusside infusion in congestive heart failure. J Am Coll Cardiol. 1983;2:411–7.
  1. Kaye DM, Jennings GL, Dart AM, et al. Differential effect of acute baroreceptor unloading on cardiac and systemic sympathetic tone in congestive heart failure. J Am Coll Cardiol. 1998;31:583–7.
  1. Chatterjee K, Parmley WW, Massie B, et al. Oral hydralazine therapy for chronic refractory heart failure. Circulation. 1976;54:879–83.
  1. Taylor AL, Ziesche S, Yancy C, et al. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med. 2004;351:2049–57.
  1. Elkayam U. Nitrates in the treatment of congestive heart failure. Am J Cardiol. 1996;77:41C–51C.
  1. Packer M, Carson P, Elkayam U, et al. Effect of amlodipine on the survival of patients with severe chronic heart failure due to a nonischemic cardiomyopathy: Results of the PRAISE-2 study (prospective randomized amlodipine survival evaluation 2). JACC Heart failure. 2013;1:308–14.
  1. Packer M, O'Connor CM, Ghali JK, et al. Effect of amlodipine on morbidity and mortality in severe chronic heart failure. Prospective Randomized Amlodipine Survival Evaluation Study Group. N Engl J Med. 1996;335:1107–14.
  1. Ignarro LJ, Lippton H, Edwards JC, et al. Mechanism of vascular smooth muscle relaxation by organic nitrates, nitrites, nitroprusside and nitric oxide: evidence for the involvement of S-nitrosothiols as active intermediates. J Pharmacol Exp Ther. 1981;218:739–49.
  1. De Caterina R, Dorso CR, Tack-Goldman K, et al. Nitrates and endothelial prostacyclin production: studies in vitro. Circulation. 1985;71:176–82.
  1. Franciosa JA, Nordstrom LA, Cohn JN. Nitrate therapy for congestive heart failure. JAMA. 1978;240:443–6.
  1. Leier CV, Huss P, Magorien RD, et al. Improved exercise capacity and differing arterial and venous tolerance during chronic isosorbide dinitrate therapy for congestive heart failure. Circulation. 1983;67:817–22.
  1. Franciosa JA, Goldsmith SR, Cohn JN. Contrasting immediate and long-term effects of isosorbide dinitrate on exercise capacity in congestive heart failure. Am J Med. 1980;69:559–66.
  1. Chatterjee K, Parmley WW. Vasodilator therapy for acute myocardial infarction and chronic congestive heart failure. J Am Coll Cardiol. 1983;1:133–53.
  1. A placebo-controlled trial of captopril in refractory chronic congestive heart failure. Captopril Multicenter Research Group. J Am Coll Cardiol. 1983;2:755–63.
  1. Pfeffer MA, Braunwald E, Moye LA, et al. Effect of captopril on mortality and morbidity in patients with left ventricular dysfunction after myocardial infarction. Results of the survival and ventricular enlargement trial. The SAVE Investigators. N Engl J Med. 1992;327:669–77.
  1. Pfeffer JM, Pfeffer MA, Braunwald E. Influence of chronic captopril therapy on the infarcted left ventricle of the rat. Circ Res. 1985;57:84–95.
  1. Curtiss C, Cohn JN, Vrobel T, et al. Role of the renin-angiotensin system in the systemic vasoconstriction of chronic congestive heart failure. Circulation. 1978;58:763–70.
  1. Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. The SOLVD Investigators. N Engl J Med. 1991;325:293–302.
  1. Effect of enalapril on mortality and the development of heart failure in asymptomatic patients with reduced left ventricular ejection fractions. The SOLVD Investigators. N Engl J Med. 1992;327:685–91.
  1. Francis GS, Benedict C, Johnstone DE, et al. Comparison of neuroendocrine activation in patients with left ventricular dysfunction with and without congestive heart failure. A substudy of the Studies of Left Ventricular Dysfunction (SOLVD). Circulation. 1990;82:1724–9.
  1. Dzau VJ, Colucci WS, Hollenberg NK, et al. Relation of the renin-angiotensin-aldosterone system to clinical state in congestive heart failure. Circulation. 1981;63:645–51.36
  1. Katz A. Molecular biology in cardiology, a paradigmatic shift. J Mol Cell Cardiol. 1988;20:12.
  1. Packer M. The neurohormonal hypothesis: a theory to explain the mechanism of disease progression in heart failure. J Am Coll Cardiol. 1992;20:248–54.
  1. Hill JA, Olson EN. Cardiac plasticity. N Engl J Med. 2008;358:1370–80.
  1. McDonald KM, Garr M, Carlyle PF, et al. Relative effects of alpha 1-adrenoceptor blockade, converting enzyme inhibitor therapy, and angiotensin II subtype 1 receptor blockade on ventricular remodeling in the dog. Circulation. 1994;90:3034–46.
  1. Tan LB, Jalil JE, Pick R, et al. Cardiac myocyte necrosis induced by angiotensin II. Circ Res. 1991;69:1185–95.
  1. Levine TB, Francis GS, Goldsmith SR, et al. Activity of the sympathetic nervous system and renin-angiotensin system assessed by plasma hormone levels and their relation to hemodynamic abnormalities in congestive heart failure. Am J Cardiol. 1982;49:1659–66.
  1. Packer M. Neurohormonal interactions and adaptations in congestive heart failure. Circulation. 1988;77:721–30.
  1. Harris P. Evolution and the cardiac patient. Cardiovasc Res. 1983;17:313–445.
  1. Pfeffer MA, Braunwald E. Ventricular remodeling after myocardial infarction. Experimental observations and clinical implications. Circulation. 1990;81:1161–72.
  1. Cohn J. Structural basis for heart failure. Ventricular remodeling and its pharmacological inhibition. Circulation. 1995;91:2504–7.
  1. Cohn JN, Ferrari R, Sharpe N. Cardiac remodeling—concepts and clinical implications: a consensus paper from an international forum on cardiac remodeling. Behalf of an International Forum on Cardiac Remodeling. J Am Coll Cardiol. 2000;35:569–82.
  1. Packer M, Lee WH, Kessler PD, et al. Role of neurohormonal mechanisms in determining survival in patients with severe chronic heart failure. Circulation. 1987;75:IV80–92.
  1. Pfeffer MA, Lamas GA, Vaughan DE, et al. Effect of captopril on progressive ventricular dilatation after anterior myocardial infarction. N Engl J Med. 1988;319: 80–6.
  1. Konstam MA, Kronenberg MW, Rousseau MF, et al. Effects of the angiotensin converting enzyme inhibitor enalapril on the long-term progression of left ventricular dilatation in patients with asymptomatic systolic dysfunction. SOLVD (Studies of Left Ventricular Dysfunction) Investigators. Circulation. 1993;88:2277–83.
  1. Greenberg B, Quinones MA, Koilpillai C, et al. Effects of long-term enalapril therapy on cardiac structure and function in patients with left ventricular dysfunction. Results of the SOLVD echocardiography substudy. Circulation. 1995;91:2573–81.
  1. St John Sutton M, Pfeffer MA, Plappert T, et al. Quantitative two-dimensional echocardiographic measurements are major predictors of adverse cardiovascular events after acute myocardial infarction. The protective effects of captopril. Circulation. 1994;89:68–75.
  1. Goodfriend TL, Elliott ME, Catt KJ. Angiotensin receptors and their antagonists. The New England journal of medicine. 1996;334:1649–54.
  1. Burnier M, Brunner HR. Angiotensin II receptor antagonists. Lancet. 2000;355: 637–45.
  1. Phillips CO, Kashani A, Ko DK, et al. Adverse effects of combination angiotensin II receptor blockers plus angiotensin-converting enzyme inhibitors for left ventricular dysfunction: a quantitative review of data from randomized clinical trials. Arch Intern Med. 2007;167:1930–6.
  1. Cohn JN, Tognoni G, Valsartan Heart Failure Trial I. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure. N Engl J Med. 2001;345:1667–75.
  1. Young JB, Dunlap ME, Pfeffer MA, et al. Mortality and morbidity reduction with Candesartan in patients with chronic heart failure and left ventricular systolic dysfunction: results of the CHARM low-left ventricular ejection fraction trials. Circulation. 2004;110:2618–26.37
  1. Pfeffer MA, McMurray JJ, Velazquez EJ, et al. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med. 2003;349:1893–906.
  1. Investigators O. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358:1547–59.
  1. Messerli FH. The sudden demise of dual renin-angiotensin system blockade or the soft science of the surrogate end point. J Am Coll Cardiol. 2009;53:468–70.
  1. GISSI-AF Investigators, Disertori M, Latini R, et al. Valsartan for prevention of recurrent atrial fibrillation. N Engl J Med. 2009;360:1606–17.
  1. Konstam MA, Neaton JD, Dickstein K, et al. Effects of high-dose versus low-dose losartan on clinical outcomes in patients with heart failure (HEAAL study): a randomised, double-blind trial. Lancet. 2009;374:1840–8.
  1. Solomon SD, Skali H, Anavekar NS, et al. Changes in ventricular size and function in patients treated with valsartan, captopril, or both after myocardial infarction. Circulation. 2005;111:3411–9.
  1. Massie BM, Carson PE, McMurray JJ, et al. Irbesartan in patients with heart failure and preserved ejection fraction. N Engl J Med. 2008;359:2456–67.
  1. Weber KT. Aldosterone in congestive heart failure. N Engl J Med. 2001;345:1689–97.
  1. Tang WH, Parameswaran AC, Maroo AP, et al. Aldosterone receptor antagonists in the medical management of chronic heart failure. Mayo Clinic proceedings. 2005;80:1623–30.
  1. Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med. 1999;341:709–17.
  1. Pitt B, Remme W, Zannad F, et al. Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. N Engl J Med. 2003;348:1309–21.
  1. Zannad F, McMurray JJ, Krum H, et al. Eplerenone in patients with systolic heart failure and mild symptoms. N Engl J Med. 2011;364:11–21.
  1. Albert NM, Yancy CW, Liang L, et al. Use of aldosterone antagonists in heart failure. JAMA. 2009;302:1658–65.
  1. Juurlink DN, Mamdani MM, Lee DS, et al. Rates of hyperkalemia after publication of the Randomized Aldactone Evaluation Study. N Engl J Med. 2004;351:543–51.
  1. Weber KT, Villarreal D. Aldosterone and antialdosterone therapy in congestive heart failure. Am J Cardiol. 1993;71:3A–11A.
  1. Udelson JE, Feldman AM, Greenberg B, et al. Randomized, double-blind, multicenter, placebo-controlled study evaluating the effect of aldosterone antagonism with eplerenone on ventricular remodeling in patients with mild-to-moderate heart failure and left ventricular systolic dysfunction. Circ Heart fail. 2010;3:347–53.
  1. Pitt B, Pfeffer MA, Assmann SF, et al. Spironolactone for heart failure with preserved ejection fraction. N Engl J Med. 2014;370:1383–92.
  1. McMurray JJ, Packer M, Desai AS, et al. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med. 2014;371:993–1004.
  1. Kumar P, Francis GS, Tang WH. Phosphodiesterase 5 inhibition in heart failure: mechanisms and clinical implications. Nat Rev Cardiol. 2009;6:349–55.
  1. Kass DA. Hypertrophied right hearts get two for the price of one: can inhibiting phosphodiesterase type 5 also inhibit phosphodiesterase type 3? Circulation. 2007;116:233–5.
  1. Takimoto E, Champion HC, Li M, et al. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med. 2005;11:214–22.
  1. Archer SL, Michelakis ED. Phosphodiesterase type 5 inhibitors for pulmonary arterial hypertension. N Engl J Med. 2009;361:1864–71.
  1. Semigran MJ. Type 5 phosphodiesterase inhibition: the focus shifts to the heart. Circulation. 2005;112:2589–91.38
  1. Guazzi M, Samaja M, Arena R, et al. Long-term use of sildenafil in the therapeutic management of heart failure. J Am Coll Cardiol. 2007;50:2136–44.
  1. Stehlik J, Movsesian MA. Combined use of PDE5 inhibitors and nitrates in the treatment of pulmonary arterial hypertension in patients with heart failure. J Card Fail. 2009;15:31–4.
  1. Pokreisz P, Vandenwijngaert S, Bito V, et al. Ventricular phosphodiesterase-5 expression is increased in patients with advanced heart failure and contributes to adverse ventricular remodeling after myocardial infarction in mice. Circulation. 2009;119:408–16.
  1. Nagayama T, Hsu S, Zhang M, et al. Sildenafil stops progressive chamber, cellular, and molecular remodeling and improves calcium handling and function in hearts with pre-existing advanced hypertrophy caused by pressure overload. J Am Coll Cardiol. 2009;53:207–15.
  1. Lu Z, Xu X, Hu X, et al. Oxidative stress regulates left ventricular PDE5 expression in the failing heart. Circulation. 2010;121:1474–83.
  1. Redfield MM, Chen HH, Borlaug BA, et al. Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. JAMA. 2013;309:1268–77.
  1. Cohn JN, Burke LP. Nitroprusside. Ann Intern Med. 1979;91:752–7.
  1. Mikulic E, Cohn JN, Franciosa JA. Comparative hemodynamic effects of inotropic and vasodilator drugs in severe heart failure. Circulation. 1977;56:528–33.
  1. Franciosa JA, Limas CJ, Guiha NH, et al. Improved left ventricular function during nitroprusside infusion in acute myocardial infarction. Lancet. 1972;1:650–4.
  1. Cohn JN, Franciosa JA, Francis GS, et al. Effect of short-term infusion of sodium nitroprusside on mortality rate in acute myocardial infarction complicated by left ventricular failure: results of a Veterans Administration cooperative study. N Engl J Med. 1982;306:1129–35.
  1. Guiha NH, Cohn JN, Mikulic E, et al. Treatment of refractory heart failure with infusion of nitroprusside. N Engl J Med. 1974;291:587–92.
  1. Cohn JN, Mathew KJ, Franciosa JA, et al. Chronic vasodilator therapy in the management of cardiogenic shock and intractable left ventricular failure. Ann Intern Med. 1974;81:777–80.
  1. Khot UN, Novaro GM, Popovic ZB, et al. Nitroprusside in critically ill patients with left ventricular dysfunction and aortic stenosis. N Engl J Med. 2003;348:1756–63.
  1. Mullens W, Abrahams Z, Francis GS, et al. Sodium nitroprusside for advanced low-output heart failure. J Am Coll Cardiol. 2008;52:200–7.
  1. VMAC. Intravenous nesiritide vs nitroglycerin for treatment of decompensated congestive heart failure: a randomized controlled trial. JAMA. 2002;287:1531–40.
  1. Sackner-Bernstein JD, Skopicki HA, Aaronson KD. Risk of worsening renal function with nesiritide in patients with acutely decompensated heart failure. Circulation. 2005;111:1487–91.
  1. Teerlink JR, Massie BM. Nesiritide and worsening of renal function: the emperor's new clothes? Circulation. 2005;111:1459–61.
  1. Sackner-Bernstein JD, Kowalski M, Fox M, et al. Short-term risk of death after treatment with nesiritide for decompensated heart failure: a pooled analysis of randomized controlled trials. JAMA. 2005;293:1900–5.
  1. Topol EJ. Nesiritide - not verified. N Engl J Med. 2005;353:113–6.
  1. O'Connor CM, Starling RC, Hernandez AF, et al. Effect of nesiritide in patients with acute decompensated heart failure. N Engl J Med. 2011;365:32–43.
  1. Yancy CW, Krum H, Massie BM, et al. Safety and efficacy of outpatient nesiritide in patients with advanced heart failure: results of the Second Follow-Up Serial Infusions of Nesiritide (FUSION II) trial. Circ Heart Fail. 2008;1:9–16.
  1. Hauptman PJ, Schnitzler MA, Swindle J, et al. Use of nesiritide before and after publications suggesting drug-related risks in patients with acute decompensated heart failure. JAMA. 2006;296:1877–84.
  1. Mann DL, Kent RL, Parsons B, et al. Adrenergic effects on the biology of the adult mammalian cardiocyte. Circulation. 1992;85:790–804.39
  1. Braunwald E, Chidsey CA, Pool PE, et al. Congestive heart failure. Biochemical and physiological considerations. Combined clinical staff conference at the National Institutes of Health. Ann Intern Med. 1966;64:904–41.
  1. Braunwald E, Bristow MR. Congestive heart failure: fifty years of progress. Circulation. 2000;102:IV14–23.
  1. Bristow MR, Ginsburg R, Umans V, et al. Beta 1- and beta 2-adrenergic-receptor subpopulations in nonfailing and failing human ventricular myocardium: coupling of both receptor subtypes to muscle contraction and selective beta 1-receptor down-regulation in heart failure. Circ Res. 1986;59:297–309.
  1. Eichhorn EJ, Bristow MR. Medical therapy can improve the biological properties of the chronically failing heart. A new era in the treatment of heart failure. Circulation. 1996;94:2285–96.
  1. Dalby MC, Burke M, Radley-Smith R, et al. Pheochromocytoma presenting after cardiac transplantation for dilated cardiomyopathy. J Heart Lung Transplant. 2001; 20:773–5.
  1. Cohn J. Sympathetic nervous system in heart failure. Circulation. 2002;106:2417–8.
  1. Bristow M. Antiadrenergic therapy of chronic heart failure: surprises and new opportunities. Circulation. 2003;107:1100–2.
  1. Triposkiadis F, Karayannis G, Giamouzis G, et al. The sympathetic nervous system in heart failure physiology, pathophysiology, and clinical implications. J Am Coll Cardiol. 2009;54:1747–62.
  1. Floras JS. Sympathetic nervous system activation in human heart failure: clinical implications of an updated model. J Am Coll Cardiol. 2009;54:375–85.
  1. Waagstein F, Hjalmarson A, Varnauskas E, et al. Effect of chronic beta-adrenergic receptor blockade in congestive cardiomyopathy. Br Heart J. 1975;37:1022–36.
  1. Swedberg K, Hjalmarson A, Waagstein F, et al. Beneficial effects of long-term beta-blockade in congestive cardiomyopathy. Br Heart J. 1980;44:117–33.
  1. Swedberg K, Hjalmarson A, Waagstein F, et al. Adverse effects of beta-blockade withdrawal in patients with congestive cardiomyopathy. Br Heart J. 1980;44:134–42.
  1. Waagstein F, Bristow MR, Swedberg K, et al. Beneficial effects of metoprolol in idiopathic dilated cardiomyopathy. Metoprolol in Dilated Cardiomyopathy (MDC) Trial Study Group. Lancet. 1993;342:1441–6.
  1. A randomized trial of beta-blockade in heart failure. The Cardiac Insufficiency Bisoprolol Study (CIBIS). CIBIS Investigators and Committees. Circulation. 1994;90: 1765–73.
  1. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial. Lancet. 1999;353:9–13.
  1. Hjalmarson A, Goldstein S, Fagerberg B, et al. Effects of controlled-release metoprolol on total mortality, hospitalizations, and well-being in patients with heart failure: the Metoprolol CR/XL Randomized Intervention Trial in congestive heart failure (MERIT-HF). MERIT-HF Study Group. JAMA. 2000;283:1295–302.
  1. Packer M, Fowler MB, Roecker EB, et al. Effect of carvedilol on the morbidity of patients with severe chronic heart failure: results of the carvedilol prospective randomized cumulative survival (COPERNICUS) study. Circulation. 2002;106:2194–9.
  1. Poole-Wilson PA, Swedberg K, Cleland JG, et al. Comparison of carvedilol and metoprolol on clinical outcomes in patients with chronic heart failure in the Carvedilol Or Metoprolol European Trial (COMET): randomised controlled trial. Lancet. 2003;362:7–13.
  1. Packer M. Do beta-blockers prolong survival in heart failure only by inhibiting the beta1-receptor? A perspective on the results of the COMET trial. J Card Fail. 2003;9:429–43.
  1. Klapholz M. Beta-blocker use for the stages of heart failure. Mayo Clinic proceedings. 2009;84:718–29.
  1. Willenheimer R, van Veldhuisen DJ, Silke B, et al. Effect on survival and hospitalization of initiating treatment for chronic heart failure with bisoprolol followed by enalapril, as compared with the opposite sequence: results of the randomized Cardiac Insufficiency Bisoprolol Study (CIBIS) III. Circulation. 2005;112:2426–35.40
  1. McAlister FA, Wiebe N, Ezekowitz JA, et al. Meta-analysis: beta-blocker dose, heart rate reduction, and death in patients with heart failure. Ann Intern Med. 2009;150:784–94.
  1. Flannery G, Gehrig-Mills R, Billah B, et al. Analysis of randomized controlled trials on the effect of magnitude of heart rate reduction on clinical outcomes in patients with systolic chronic heart failure receiving beta-blockers. Am J Cardiol. 2008;101:865–9.
  1. Maurer MS, Sackner-Bernstein JD, El-Khoury Rumbarger L, et al. Mechanisms underlying improvements in ejection fraction with carvedilol in heart failure. Circ Heart Fail. 2009;2:189–96.
  1. Fonarow GC, Abraham WT, Albert NM, et al. Influence of beta-blocker continuation or withdrawal on outcomes in patients hospitalized with heart failure: findings from the OPTIMIZE-HF program. J Am Coll Cardiol. 2008;52:190–9.
  1. Butler J, Young JB, Abraham WT, et al. Beta-blocker use and outcomes among hospitalized heart failure patients. Journal of the American College of Cardiology. 2006;47:2462–9.
  1. Parameswaran AC, Tang WH, Francis GS, et al. Why do patients fail to receive beta-blockers for chronic heart failure over time? A “real-world” single-center, 2-year follow-up experience of beta-blocker therapy in patients with chronic heart failure. Am Heart J. 2005;149:921–6.
  1. Lowes BD, Gilbert EM, Abraham WT, et al. Myocardial gene expression in dilated cardiomyopathy treated with beta-blocking agents. N Engl J Med. 2002;346: 1357–65.
  1. Bristow MR, Murphy GA, Krause-Steinrauf H, et al. An alpha2C-adrenergic receptor polymorphism alters the norepinephrine-lowering effects and therapeutic response of the beta-blocker bucindolol in chronic heart failure. Circ Heart Fail. 2010;3:21–8.