* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download Clinical and research issues regarding chronic advanced coronary
Cardiac contractility modulation wikipedia , lookup
Cardiac surgery wikipedia , lookup
Antihypertensive drug wikipedia , lookup
History of invasive and interventional cardiology wikipedia , lookup
Jatene procedure wikipedia , lookup
Remote ischemic conditioning wikipedia , lookup
Quantium Medical Cardiac Output wikipedia , lookup
Results of Expert Meetings Clinical and research issues regarding chronic advanced coronary artery disease: Part I: Contemporary and emerging therapies E. Marc Jolicoeur, MD, MSc, a Christopher B. Granger, MD, a Timothy D. Henry, MD, b David J. Holmes, MD, c Carl J. Pepine, MD, d Daniel Mark, MD, a Bernard R. Chaitman, MD, e Bernard J. Gersh, MB, ChB, DPhil, f and E. Magnus Ohman, MD a on behalf of the Working Group Members g Durham, NC; Rochester, MN; Gainesville, FL; and St. Louis, MO The following report is based on a working group meeting about advanced coronary artery disease for patients with refractory ischemia who cannot receive revascularization. The aims were to review currently available treatment strategies, define unmet clinical needs, explore clinical trial design issues, and identify promising novel therapeutic targets and approaches for patients with chronic ischemia. The Working Group brought together medical experts in the management of refractory angina with representatives from regulatory agencies, Centers for Medicare and Medicaid Services, and industry. The meeting began with presentations reviewing the limitations of the current medical therapies and revascularization strategies and focused on lessons learned from past therapeutic attempts to optimize outcomes and on what are considered to be the most promising new approaches. Perspectives from clinical experts and from regulatory agencies were juxtaposed against needs and concerns of industry regarding development of new therapeutic strategies. This report presents the considerations and conclusions of the meeting on December 4-5, 2006. This document has been developed as a 2-part article, with contemporary and emerging therapies for advanced coronary artery disease reviewed first. Trial design, end points, and regulatory issues will be discussed in the second part of the article. (Am Heart J 2008;155:418-34.) Part I. Contemporary and emerging therapies The population of patients with advanced coronary artery disease (CAD) is growing as a result of the aging of the general population, more extensive use of revascularization, and more effective therapies that have prolonged the survival of patients with severe atherosclerosis. Patients with refractory angina who have exhausted most therapeutic options comprise a growing clinical entity and a therapeutic dilemma. Nonetheless, advanced CAD receives relatively little attention from the medical and research communities. As a result, the scope of the disease is not well defined, From the aDuke University, Durham, NC, bMinneapolis Heart Institute Foundation, MN, c Mayo Clinic, Rochester, MN, dUniversity of Florida, Gainesville, FL, eSaint Louis University School of Medicine, St. Louis, MO, and fMayo Clinic and Foundation, Rochester, MN. g See Appendix A for complete group membership. Dr. E. Marc Jolicoeur is supported by the Montreal Heart Institute Foundation and by the Quebec Cardiologists Association. Submitted November 30, 2007; accepted December 6, 2007. Reprint requests: E. Marc Jolicoeur, Box 3850, 2400 Pratt Street Room 0311, Terrace Level, Durham, NC 27705. E-mail: [email protected] 0002-8703/$ - see front matter © 2008, Mosby, Inc. All rights reserved. doi:10.1016/j.ahj.2007.12.004 its coverage in guidelines from professional associations is scant,1,2 and few new medical options are available.3 This report reviews the current status of advanced CAD, the limits of the contemporary available therapies, and the difficulties encountered in the development of new approaches. Definition of advanced CAD Advanced CAD has been recognized under several terms and classifications: intractable angina pectoris, nooption CAD, nonrevascularizable CAD, and end-stage CAD. Truly, refractory angina embodies several chronic coronary artery syndromes that vary in their manifestation and pathophysiology. The syndromes of chronic CAD, their definitions, and suggested pathophysiology are summarized in Table I. Recently, a European Society of Cardiology Joint Study Group on the Treatment of Refractory Angina proposed the following definition: “A chronic condition (N3 months) characterized by the presence of angina caused by coronary insufficiency in the presence of CAD, which cannot be controlled by a combination of medical therapy, angioplasty, and coronary bypass surgery. The presence of reversible myocardial ischemia should be clinically established to be the cause of the symptoms.” 4 Advanced CAD could American Heart Journal Volume 155, Number 3 Jolicoeur et al 419 Table I. Clinical syndromes of chronic CAD Syndromes Clinical definition Suggested pathophysiology CSA Deep chest/arm discomfort, ↑ with exercise/psychological stress, ↓ by rest/nitroglycerin Increase in myocardial oxygen demand with epicardial flow limitation Isolated angina Symptoms similar to CSA with no prior MI or revascularization Same as CSA Revascularization-resistant angina Symptoms similar to CSA but successful prior revascularization Combination of epicardial endothelial dysfunction, microvascular angina, and sensitive heart syndrome Refractory angina Symptoms similar to CSA but unsuccessful prior revascularization Increase in myocardial oxygen demand with marked epicardial flow limitation Persistent angina Symptoms similar to CSA with recurrence on medical therapy Same as CSA with pharmacologic resistance and microvascular angina Microvascular angina (syndrome X) Symptoms may or may not be similar to CSA with evidence for myocardial; ischemia and “normal” coronary angiogram (no flow-limiting stenosis) Altered cardiac nociception, endothelial dysfunction, impaired microvascular dilation ± vascular smooth muscle dysfunction Variant angina (Prinzmetal) Symptoms similar to CSA, usually at rest and early am hours, episodic coronary spasm with ST-segment elevation VSMC hypersensitivity to vasoconstrictor stimuli + ↓ endothelium-derived relaxing factors. May involve microvascular spasm Silent ischemia Asymptomatic episodes in patients with obstructive CAD with or without CSA Defective nociception through narcotics, neuropathy or advanced age Sensitive heart syndrome Lack of evidence of ischemia with cardiac related pain Decreased threshold for nociception ± sympathovagal imbalance CSA, Chronic stable angina; VSMC, vascular smooth muscle cells. be more practically defined as symptomatic multivessel CAD in patients who are not candidates for revascularization with either percutaneous coronary intervention (PCI) or coronary artery bypass grafting (CABG) surgery. The reasons why a patient may not be revascularizable include unsuitable coronary anatomy, surgery-related issues (previous CABG, lack of surgical conduits, multiple thoracotomies, extensive aortic calcification), extracardiac comorbid illnesses (renal failure, chronic obstructive pulmonary disease), and advanced age (which often combines the above-mentioned features). However, the “nonrevascularization” status also will be influenced by local expertise, operator tolerance to risk, and patient agreeing to the procedure. Likewise, the threshold for revascularization may be modified depending on the clinical context. Prevalence of advanced CAD Precise estimates of the prevalence of advanced CAD in a global population are not available. However, rough estimates have been obtained from population surveys and catheterization laboratory registries. In the late 1990s, Mukherjee et al5 reported that 12% of the patients referred for symptomatic CAD were not amenable to PCI or CABG. In absolute numbers, this would represent more than 100 000 patients per year in the United States. This proportion is somewhat similar to Swedish surveys in which 10% of the patients did not undergo revascularization despite severe angina symptoms.6,7 Inferences from catheterization laboratory registries should be interpreted with caution, however, because referrals for angiography are generally biased by patients initially deemed by their physicians to be candidates for revascularization. This referral bias likely leads to an underestimate of the true prevalence of chronic refractory angina because the most morbid patients are probably not included in the denominator. Using the same logic, this referral bias may lead to an overrepresentation of unsuitable anatomy (or lack of graft material) as a reason for revascularization ineligibility. Mortality associated with advanced CAD In patients with chronic stable angina, the annual mortality rate is approximately 1.5%.8 For advanced CAD, few mortality and morbidity data have been American Heart Journal March 2008 420 Jolicoeur et al Table II. Mortality and adverse cardiac events associated with optimal medical therapy in refractory angina trials Study therapy, year Design (n) Relevant inclusion/ exclusion criteria Mortality and morbidity in control group ⁎ Comments Schofield et al15 TMR, 1999 RCT (188) Control: medical E: LVEF N30%, unable to do treadmill, life expectancy b12 mo Death: 4% at 12 mo ACS: 63% at 12 mo Medical therapy not detailed ACS expressed as 0.8 per patient-year (n = 83) ATLANTIC11 TMR, 1999 RCT (182) Death: 10% at 12 mo Control: medical I: At least one protected vascular territory E: LVEF b30%, unable to do treadmill Protected region defined as unobstructed blood flow (lesion b50%) or patent CABG: 65% had a protected LAD Frazier et al12 TMR, 1999 RCT (192) Control: medical E: LVEF b20%, concurrent major illness Death: 21% ACS: 69% at 12 mo 18 patients underwent revascularization procedure despite being nonrevascularizable initially. At 5 y; the number goes up to 35 Allen et al10 TMR, 1999 RCT (275) Control: medical I: Class IV angina only E: LVEF b25%, severe COPD Death: 11% at 12 mo ACS: 69% at 12 mo The incapacity to undergo a stress test was not an inclusion criterion. PACIFIC14 TMR, 2000 RCT (221) Death: 3% at 12 mo ACS: 53% at 12 mo CHF: 10% at 12 mo Control: medical E: LVEF b30%, unable to do treadmill, symptomatic heart failure, renal insufficiency, moderate AS, severe PAD 24 patients had PTCA/CABG or TMR during follow-up. Nonrevascularization assessment poorly defined Aaberge et al9 TMR, 2000 RCT (100) Control: medical E: age N75 y, LVEF b30%, overt heart failure Death: 8% at 12 mo ACS: 40% at 12 mo Medical therapy is optimal with rate of administration higher than 80% Leon at al 13 TMR, 2005 RCT (298) E: unable to do treadmill, chronic AF Death: 5% at 12 mo ACS: 3% at 12 mo MACE: 11% at 12 mo Medical therapy group underwent sham invasive procedure (may affect survival) E: LVEF b30%, unstable cardiac condition or revascularization within the past 3 mo, renal dysfunction Death: 1% at 6 mo ACS: 15% at 6 mo MACE: at 6 mo Placebo patients underwent intracoronary placebo injection with no complication. Hypotension associated with dosing was reported Control: placebo FIRST19 rFGF-2, 2002 RCT (337) Control: placebo ACS: 77% at 12 mo CHF: 10% at 12 mo VIVA16 rhVEGF, 2003 RCT (178) Control: placebo I: age 40-75 y E: LVEF b25%, previous TMR, unable to do treadmill, history cancer/proliferative diabetic retinopathy, renal dysfunction Death: 3% at 4 mo ACS: 11% at 4 mo Placebo patients underwent intracoronary placebo injection with no complication Euroinject One17 phVEGF-A165, 2005 RCT (80) Control: placebo E: LVEF b45%, history of cancer, proliferative diabetic retinopathy, premenopausal women Death: 3% at 6 mo ACS: 15% at 6 mo Placebo patients underwent sham injections (2 procedural complications reported: STEMI and complete AV block) REVASC18 Ad-VEGF121, 2006 RCT (67) Control: medical Death: 3% at 6 mo MACE: 26% at 6 mo Several exclusion criteria, including past history of cancer AGENT III and IV21 RCT (532) I: Angina CCS II-IV E: age N80 y, LVEF b25%, unprotected proximal LAD stenosis/left main equivalent, ICD, severe CHF, anemia unable to do treadmill, renal dysfunction I: age 30-75 y, angina CCS II-IV MACE: 22% at 12 mo Mortality rates not provided American Heart Journal Volume 155, Number 3 Jolicoeur et al 421 Table Table II (continued) II (continued ) Study therapy, year Design (n) Relevant inclusion/ exclusion criteria Mortality and morbidity in control group ⁎ Comments E: LVEF b30%, previous TMR, unable to do treadmill, history cancer/proliferative diabetic retinopathy, renal dysfunction, unprotected left main stenosis Ad5FGF-4, 2007 Control: placebo AF, Atrial fibrillation; AS, aortic stenosis; AV, auriculoventricular; COPD, chronic obstructive pulmonary disease; E, exclusion; I, inclusion; ICD, implantable cardioverter defibrillator; LAD, left anterior descending artery; MACE, major adverse cardiac events; PAD, peripheral arterial disease; PTCA, percutaneous coronary transluminal angioplasty; RCT, randomized-controlled trial; STEMI; ST-elevation myocardial infarction; TMR, transmyocardial laser revascularization. ⁎ACS represents all hospitalizations for unstable angina, non-STEMI, and STEMI. reported. In recent years, several controlled trials have explored the efficacy of alternative therapies including laser transmyocardial revascularization9-15 and therapeutic angiogenesis.16-20 In general, these trials enrolled patients with refractory angina despite optimal medical therapy, so that the outcomes experienced by the patients randomized to the control arm of these trials may give a fair representation of the outcomes experienced by patients with advanced CAD ( Table II). In the control arm of these alternative therapy trials, annual mortality rates varied from 3% to 21%, whereas coronary events rates (including hospitalization for decompensated angina and acute coronary syndromes [ACSs]) varied from 11% to 69%. Of note, most of these trials excluded patients unable to perform an exercise treadmill test (ETT) or patients with left ventricular ejection fraction (LVEF) b30% (detailed in Table II). Thus, these exclusion criteria are biased toward a less severe picture than what may actually be the case in an unselected population of advanced CAD because inability to perform an exercise test is associated with increased mortality. Registries provide additional but variable information. The Mediators of Social Support Study reported mortality and myocardial rates of 38% and 10%, respectively, after a median of 2.2 years of follow-up for patients with medically treated advanced CAD (defined as significant 3-vessel or left main disease and either LVEF b50% or severe angina).22 Hospitalization occurred in 89% of patients. In the OPTions In Myocardial Ischemic Syndrome Therapy Program (Mineapolis Heart Intitute, Minneapolis, MN), the largest reported prospective series of patients with refractory angina reported, total mortality was 11.7% after an average follow-up of 5.4 years.23 Despite the presence of extensive CAD, cardiovascular mortality was low (6.2%)—an annualized death rate of only 1%. Of note, 8.3% of all the patients experienced myocardial infarction (MI). Patients who died were more likely to have diabetes, peripheral arterial disease, chronic renal disease, valvular disease, history of congestive heart failure (CHF), lower ejection fraction, and more angina at baseline. In aggregate, these data suggest that mortality associated with advanced CAD is variable and depends on multiple factors including associated comorbidities. In addition to improving mortality and cardiovascular events, newer therapies directed at advanced CAD should also improve functional status and health-related quality of life. Contemporary treatments of advanced CAD Current pharmacological and nonpharmacological therapies are outlined in Tables III and IV. Most antianginal medications currently in use decrease myocardial oxygen demand either by limiting heart rate, lowering systolic pressure, or by decreasing contractility. Additional lusitropic and vasodilatory properties also contribute significantly. Combination of antianginal medication is often needed to relieve refractory angina. Simple principles may be used to enhance the efficacy of these possible combinations. For instance, the circadian variation of ischemic episodes shows 2 peaks (one in the early morning and one in the late evening).25 An appropriate chronopharmacological strategy using different preparations of extended-release nitrates or long-acting calcium antagonists may help achieve a significantly better therapeutic response in these critical periods of the day.26 Orthostatic hypotension frequently limits the total dose of antianginal medication that can be administered to a patient. Likewise, orthostatic hypotension by itself may beget angina due to a decrease of the coronary perfusion pressure. Simplification of the drug regimen should be attempted whenever possible to enhance medical adherence. Secondary causes of angina, like anemia, poorly controlled hypertension, thyroid dysfunction, or atrial fibrillation with rapid ventricular rate response, should be identified and treated accordingly. American Heart Journal March 2008 422 Jolicoeur et al Table III. Contemporary treatments for chronic refractory angina: pharmacological therapies Pharmacological therapy Proposed mechanisms of action Comments Aspirin and clopidogrel No known antianginal effect: prevention of thrombotic events Warfarin may be substituted in case of AF or decreased LVEF β-Blockers 1. 2. 3. 4. Selectivity to β-receptor affects antianginal properties. Only atenolol, metoprolol, nadolol, propanolol, and bisoprolol approved for treatment of angina Nitrates 1. Coronary vasodilatation 2. Ventricular wall stress reduction (↓ both pre- and postcharge) 3. Antithrombotic effect? Tachyphylaxis development: nitrate-free periods required Nondihydropiridine calcium antagonists 1. 2. 3. 4. Especially useful when prominent vasospastic component present Dihydropiridine calcium antagonists 1. Coronary vasodilatation 2. Afterload reduction (anti-HTN) May cause reflex tachycardia: should be used in combination to a β-blockers. Aggravation of angina possible if severe fixed coronary obstruction Ranolazine 1. Partial inhibition of late Na+ current (lNa) 2. Partial inhibition of fatty acid metabolism Doubt have been emitted about action of ranolazine on fatty acid oxidation Statins 1. Unknown: improved endothelial function of epicardial conductance vessels 2. ↓ coronary events 3. Plaque burden regression Prolonged use required before therapeutic effect Opioids 4,24 1. Direct analgesia 2. Possible decrease in adrenergic tone with epidural analgesia Reserved for selected patients, often as a palliative solution. Frequent adverse events. Epidural analgesia hampered by several side effects Chronotrope-negative Lusitrope-positive Inotrope-negative Afterload reduction (anti-HTN) Chronotrope-negative Lusitrope-positive Inotrope-negative Coronary vasodilatation AF, Atrial fibrillation; HTN, hypertension. Pharmacological treatments Rationale for the administration of β-blockers, calcium antagonists, and nitrates in refractory angina relates to the evidence for their use in the wider population of patients with chronic stable angina ( Table III).1,2 In addition to this traditional pharmacopeia, newer antianginal drugs like ranolazine are now available. Antiplatelet agents and intensive dyslipidemia management should be part of a comprehensive approach to risk factor modification. Antiplatelet agents combination: aspirin and clopidogrel. Neither aspirin nor clopidogrel has intrinsic antianginal properties. Aspirin is a mandatory component of the treatment of CAD because of its ability to prevent atherothrombotic events.2 Several trials have assessed whether the combination of aspirin with clopidogrel would result in a further reduction of coronary events. The Clopidogrel versus Aspirin in Patients at Risk of Ischemic Events trial showed that clopidogrel was slightly better than aspirin in preventing vascular complications in a population of patients with stable vascular disease.27 The Clopidogrel in Unstable angina to prevent Recurrent Events trial demonstrated a 20% relative reduction of cardiovascular events when clopidogrel was added to aspirin in patients with ACS.28 The Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management, and Avoidance trial assessed whether the addition of clopidogrel (75 mg, daily) to aspirin (75-162 mg, daily) was superior to aspirin alone in reducing adverse cardiovascular events in high-risk stable patients for over 4 years.29 The combination led to a nonsignificant reduction of the composite end point (cardiovascular death/MI/stroke) from 7.3% in the placebo plus aspirin arm to 6.8% in the clopidogrel plus aspirin arm (P = .22). However, aspirin plus clopidogrel significantly increased moderate bleeding (2.1% vs 1.3%, P b .001). In a prespecified subgroup analysis, patients with documented atherothrombotic disease at baseline did derive benefit from clopidogrel (6.9% vs 7.9% in the placebo group, P = .046). However, there is currently no strong evidence for routinely combining antiplatelet therapy American Heart Journal Volume 155, Number 3 Jolicoeur et al 423 Table IV. Contemporary treatments for chronic refractory angina: mechanical treatments and advances Nonpharmacological therapy Proposed mechanisms of action Comments CTO recanalization 1. Restoration of epicardial blood flow Success depends on technical skills and technology used EECP 1. Diastolic augmentation + enhanced coronary perfusion pressure 2. ↓ LV afterload 3. Release of growth factors, mobilization of progenitor cells, ↑ NO and ↓ endothelin Contraindications: impossible ECG gating, decompensated heart failure, severe AI, uncontrolled systemic hypertension, severe pulmonary hypertension, severe lower extremity PAD, aortic aneurysm, recent DVT Spinal cord stimulation 1. Pain inhibition: gate control theory 2. Anti-ischemia: a. ↓ myocardial oxygen consumption b. Redistribution of coronary flow c. ↓ sympathetic tone Class A therapeutic option for refractory angina according to ESC Joint Study Group Heart transplantation Organ replacement Described indication: few cases reported AI, Aortic insufficiency; DVT, deep vein thrombosis; ECG, echocardiogram; ESC, European Society of Cardiology; LV, left ventricle; NO, nitric oxide; PAD, peripheral arterial disease. in patients who remain symptomatic despite optimal medical therapy. Statin and dyslipidemia management. Intensive statin therapy reduces death and vascular events in patients with established CAD.30 Whether statins have a true anti-ischemic effect on top of usual therapy is unknown.31 Statin-related improvement in endothelial function provides a rationale for how angina could be improved.32 The regression of atherosclerotic plaque burden observed with intensive statin therapy or with reconstituted high-density lipoprotein (HDL) ApoA-1 Milano or with recombinant HDL has generated hope for a reversal of symptoms in patients with advanced CAD.33-37 Fueled by the observational studies suggesting protection by elevated HDL, modulation of HDL intermediate metabolism has been considered a promising target for halting progression of atherosclerosis. In the Investigation of Lipid Level Management to Understand Its Impact in Atherosclerotic Events trial, however, the experimental HDL-raising drug torcetrapid (a cholesteryl-ester-transfer-protein [CETP] inhibitor) in combination with atorvastatin was associated with an increased risk of death. Aside from the significant increase in the blood pressure associated with the drug, it has been speculated that the type of HDL produced by CETP inhibition may be proatherogenic rather than antiatherosclerotic.38 Ranolazine. Ranolazine (Ranexa) is the first drug in more than 20 years to be approved by United States Food and Drug Administration (FDA) for the treatment of chronic angina. The antianginal mechanisms of action of ranolazine are not entirely known. It had been postulated that ranolazine acted as a partial inhibitor of fatty acid oxidation, which would facilitate energy production in hypoxic cardiomyocytes by partially shifting cardiac lipid oxidation to glucose oxidation.39-41 More recent studies in human cardiac myocytes indicate this is an unlikely explanation for the antianginal/antiischemic properties because the pFOX inhibitory effect is small at the concentrations of ranolazine used to treat chronic angina.42 More recently, ranolazine has been found to have electrophysiological properties similar to amiodarone, which could explain its anti-ischemic and antianginal properties.43 At the cardiomyocytes level, ranolazine inhibits the late Na+ current,44 which is found to be abnormally increased during hypoxia or in the presence of reactive oxygen species and ischemic metabolites.45 In experimental hypoxic conditions, this inhibition of the Na+ current leads to an intracellular Na+ overload,46 which activates several exchangers to maintain cellular Na+ homeostasis. One of the most important exchangers is the Na+/Ca++ exchanger that transports excess Na+ out of the cell in exchange for Ca++, resulting in abnormal intramyocyte Ca++ concentration. This abnormal Ca++ overload impairs both the contraction-relaxation coupling and as a consequence increases diastolic tension, thereby reducing coronary perfusion. Ranolazine, through a partial inhibition of the late Na+ current, could limit this pathological process and attenuate the abnormal increase in intracellular Ca++.44 The Monotherapy Assessment of Ranolazine in Stable Angina (MARISA) study was the first placebo-controlled crossover trial with the sustained release preparation to demonstrate the anti-ischemic effect of ranolazine as monotherapy among patients with chronic severe angina. In MARISA, a 1-week treatment of ranolazine resulted in a dose-related increase in the exercise duration.47 In the Combination Assessment of Ranolazine In Stable Angina (CARISA) trial, the addition of ranolazine to standard dose of atenolol, diltiazem, or amlodipine significantly (but modestly) increased exercise capacity and time to 424 Jolicoeur et al electrographic ischemia during stress testing.41 Of note, ranolazine provided additional anti-ischemic benefits in a setting where some of the traditional anti-ischemic agents have failed to do so.48-50 The findings in CARISA were confirmed by the Efficacy of Ranolazine in Chronic Angina trial, which assessed ranolazine in patients with persistent angina despite maximum recommended dosage of amlodipine.51 Long-term safety information is now available through the Ranolazine Open Label Experience registry that followed on the MARISA and CARISA trials.52 After 2 years, less than 10% of the patients discontinued ranolazine because of unacceptable adverse events: annualized death rate was of 3%. The recently completed MERLIN trial also established the 1-year safety record of ranolazine in a large placebo-controlled trial of 6560 patients with ACS.53 Because ranolazine exerts its antianginal effect differently than the modes of action of nitrates, β-blockers, or calcium antagonists and has minimal effects on heart rate or blood pressure, it is well-suited for add-on therapy in clinically eligible refractory angina patients.41,47 This includes patients who do not tolerate additional chronotropic- or inotropic-negative therapy.54 Opioids. Opioids are generally reserved for selected cases of refractory angina when other therapeutic options have failed. Under close surveillance, opioids may be administered with reasonable efficacy and with limited side effects.24,55 For several years, opioids have been used with success in Denmark for the treatment of chronic refractory angina.55 Importantly, the use of opioids is limited by frequent side effects such as constipation, nausea, and disorientation.56 Mechanical treatments and advances Although highly effective for the treatment of symptomatic angina, PCI may be challenging with complex coronary anatomy including diffuse disease and chronic total occlusions (CTOs). Significant nonpharmacologic advances are now being proposed to patients with refractory angina ( Table IV). Chronic total occlusion recanalization. By definition, a CTO of a coronary artery is present for more than 3 months.57 Chronic total occlusions have been identified in up to 30% of diagnostic catheterizations.58 Advances in coronary interventional techniques have enabled recanalization of CTOs with good procedural success rates in selected cases. Although routine opening of occluded arteries in the days after acute MI does not reduce the occurrence of death, reinfarction, or heart failure,59 recanalization of chronic occlusions may improve symptoms and possibly even the prognosis of patients with advanced CAD.60,61 Even with the sophisticated interventional tools currently available, CTO recanalization is attempted less frequently than it was 10 years ago. In the 7-year period American Heart Journal March 2008 covered by the National Heart, Lung, and Blood Institute's (NHLBI) Dynamic Registry, the percentage of attempted CTOs recanalization dropped from 9.6% of all PCI in 1997 to 5.7% in 2004 (P b .0001).62 The success rate of the procedure remained stable at 71% in spite of a significant increase in the length of lesions attempted (from 17.0 to 22.4 mm, P = .006). In the largest reported observational study of patients undergoing PCI for a CTO, a successful recanalization was associated with a significant 10year survival advantage when compared with a failed revascularization.60 In the stent era, the rate of target vessel revascularization at 1 year after CTO recanalization has declined by approximately 50%.61 In return, CTO attempts are associated with periprocedural complication rates exceeding 5%,60,63,64 including MI, emergency cardiac surgery, and death. Progress in CTO recanalization is anticipated with emerging technologies, including new tapered hydrophilic guidewires,65 laser excimer,66 and plaque softening formulation.67,68 Enhanced external counterpulsation. By generating precisely timed diastolic pressure augmentation, enhanced external counterpulsation (EECP) produces externally what aortic counterpulsation does internally. Diastolic augmentation is accomplished with inflatable cuffs that sequentially compress the lower limbs during diastole and rapidly deflate before systole. This systemic diastolic pressure augmentation increases coronary perfusion pressure, decreases ventricular afterload, and increases venous return.69 The efficacy of EECP in patients with angina was assessed by a single randomized sham placebo–controlled Multicenter Study of EECP (MUST-EECP).70 The trial assessed whether a treatment regimen of 1 hour of active counterpulsation daily for 35 days would improve subjective angina and objective ischemia parameters when compared with sham lower limb compression. When compared with baseline, EECP did not increase ETT duration but did significantly improve the time to ≥1mm ST-segment depression. Likewise, patients randomized to EECP reported an improvement of their angina symptoms when compared with control patients. Of note, 55% of the patients in active treatment arm reported adverse events (such as lower limb bruise, edema, and leg pain), but only 10% were severe enough to cause withdrawal. A number of hypotheses have been proposed for the sustained antianginal effect of EECP, including a pressure-driven recruitment of myocardial collaterals,71 hemodynamic changes, the release of proangiogenic cytokines, and a peripheral training effect.72 The peripheral training effect seems mediated by a decrease in peripheral vascular resistance and a more appropriate heart rate response to exercise.73 The vascular shear stress provided by EECP is thought to American Heart Journal Volume 155, Number 3 Jolicoeur et al 425 Table V. Emerging therapies for advanced CAD Therapy Proposed mechanisms of action Comments Ivabradine 87 Sinus node slowing: inhibits pacemaker current (If) Approved in Europe for patients with chronic stable angina with sinus rhythm and contraindication to beta-blockers. Limited use for patient with refractory angina Nicorandil 1. Opening of ATP-sensitive potassium channels (KATP): dilatation of coronary resistance arterioles 2. Nitrate-like effect 3. Mimetic of ischemic preconditioning Approved in Europe. May be use to bridge the nitrate-free periods required to avoid tachyphylaxis Trimetazidine 1. Mitochondrial 3-KetoAcyl CoA thiolase inhibitor 2. Shift ischemic myocardium from fatty acid to carbohydrate use Approved in Europe for patients with chronic stable angina Perhexilene 1. Mitochondrial carnitine-palmitoyl-transferase inhibitor 2. Shift ischemic myocardium from fatty acid to carbohydrate utilization Serum level monitoring required due to risk of hepatic toxicity Therapeutic angiogenesis (protein and gene therapy) 1. Sustained tissular expression of proangiogenic factors 2. Neovascularization See Table VI for review of randomized control trials of gene therapy Cellular therapy 1. Paracrine secretion of proangiogenic factors 2. Neovascularization 3. Myogenesis (?) Various type of cell available with additional possibility to combine stem cell therapy and gene therapy Thrombolytic agent 1. Depletion in fibrinogen 2. Improved blood rheology and microcirculatory flow Urokinase administrated through intermittent dose. Treatment never compared to placebo in a trial Chelation therapy 1. Reduced LDL oxidation 2. PTH-related vasodilatory effect 3. Enhanced t-PA production No direct evidence of clinical efficacy. Transmyocardial laser revascularization Direct myocardial revascularization through microchannels formation No direct evidence of clinical efficacy Extracorporeal cardiac shock wave therapy 1. Cavitation effect Efficacy and safety of SW have been reported 2. Upregulation of VEGF expression by the myocardium 88 in a phase 1 trial including 9 patients 3. Enhanced coronary angiogenesis Neovasc reducer coronary sinus stent Controlled and permanent narrowing of coronary sinus. Pressure-driven redistribution of antegrade blood flow toward region of lesser resistance First-in-man reported in 10 patients in 2006 PICVA Evolving technology. Associated with several procedural flaws and with high complications rates Shunting of arterial blood into the coronary veins with subsequent retroperfusion of capillaries LDL, Low-density lipoprotein; SW, shock wave. result in a systemic release of growth factors and proangiogenic cytokines.74-76 The use of EECP is limited by the need to commit to 35 days of in-laboratory treatment. Enhanced external counterpulsation is contraindicated in several situations such as decompensated heart failure, severe peripheral arterial disease, and severe aortic insufficiency. These comorbid conditions are frequently encountered in patients with advanced CAD. When applied to the appropriate patient, EECP is well tolerated and associated with low risk of adverse events.77-80 Neuromodulation. Neuromodulation includes transcutaneous electric nerve stimulation and spinal cord stimulation. Long-term transcutaneous electric nerve stimulation therapy is limited by electrode-related skin irritation associated with prolonged use. Spinal cord stimulation has been used in the past 30 years to relieve various pain syndromes. It is now recognized as a valid therapeutic option in Europe (class IA) for the treatment of refractory angina.4 Modern spinal cord stimulators are entirely subcutaneously implantable and can be installed under local anesthesia using a minimally invasive American Heart Journal March 2008 426 Jolicoeur et al Table VI. Randomized placebo-controlled trials of therapeutic angiogenesis in patients with CAD Study Therapy/Delivery method Outcomes Safety Comments Laitinen et al,93 2000 (15 patients) 1000 μg plasmid-liposome + mouse VEGF/IC infusion Trial intended to assess the effect of local VEGF therapy on post-PCI restenosis: no effect. Ischemia not assessed No major serious adverse events reported at 6 mo Murine gene used. Strong suspicion of inefficient gene transfer. Patients with advanced CAD excluded FIRST,19 2002 (337 patients) Ascending doses of rFGF2/IC No effect on ETT total time at 90 and 180 d 5 deaths (1%) reported in the rFGF-2 treated patients; similar to placebo (1%) Angina frequency ↓ by rFGF-2 at 90 d: difference no longer present at 180 d Losordo et al,20 2002 (19 patients) Ascending doses of plasmid hDNA VEGF-2/Transendo Reduction in CCS angina class significantly better in VEGF-2 patients at 12 wk No major serious adverse events reported Patients with class III or IV angina. Trial interrupted by FDA due to a death in an unrelated adenoviral gene therapy trial AGENT,94 2002 (79 patients) Ascending doses of Ad5-FGF4/IC infusion Nonsignificant trend toward improvement of ETT total time and time to angina No major serious adverse event (mean f/u, 311 d). One case of brain tumor Patients with class II or III angina. No significant dose-response effect AGENT II,95 2003 (52 patients) 1010 pfu Ad5-FGF4/IC infusion Nonsignificant change in SPECT reversible perfusion defect size No major serious adverse event reported Total perfusion defect size (necrosis + ischemia) significantly improved with Ad5-FGF4 (4.2% vs 0%, P = .001). Clinically significant reduction of angina in the active group VIVA,16 2003 (178 patients) rhVEGF low and high doses/IC infusion day 0 then IV infusion days 3, 6, and 9 No effect on ETT total time, myocardial perfusion (SPECT) or on angina at day 60 Significant flushing. One case of severe hypotension per-infusion in high-dose group Significant improvement of in angina class by day 120 in the high-dose group KAT,96 2003 (103 patients) 2 × 1010 pfu AdVEGF-165 vs 2 mg plasmid-liposome VEGF/IC infusion Improvement in SPECT reversible perfusion defect size in AdVEGF group. No effect on ETT parameter Two new cancers diagnosed in the plasmid arm Moderate angina patients (CCS 2-3). Study designed to assess effect of VEGF on restenosis post-PCI Euroinject 1,17,97 2005 (80 patients) 0.5 mg of plasmid VEGF-A165/ Transendo No effect on SPECT reversible perfusion defect size. Improved regional wall motion in the active group NOGA procedure related events in 5 patients No-option patients with severe angina. First trial to use a gene vector (blind plasmid) as a placebo control AGENT III and IV,21,98 2007 (pooled analysis 532 patients) 1010 pfu vs 1011 pfu Ad5-FGF4 vs placebo/IC infusion No effect on total ETT time. No difference in the rate of coronary events/death No major serious adverse event reported Significant improvement in total ETT time and reduction of angina in women treated with either 1010 or 1011 pfu Ad5-FGF4 vs placebo Ad, Adenovirus; f/u, follow-up; IC, intracoronary; IV, intravenous; pfu, plaque forming units; Transendo, transendocardial injection. procedure. The therapy is self-administrated by the patient and requires stimulation during 1 hour, 3 times a day, and whenever angina is felt. The analgesic effect of neuromodulation is not fully understood but likely results from several simultaneous mechanisms. In addition to the analgesic modulation of A-beta neurons abnormal activity and to the restoration of normal gamma (γ)-aminobutyric acid levels in the dorsal horn, relief of angina may also be the consequence of adenosine-mediated coronary vasodilatation and of reduced sympathetic afference.81 An anti-ischemic effect has been demonstrated both with exercise stress testing and ambulatory electrocardiogram monitoring.82 Spinal cord stimulation has been compared with transmyocardial laser revascularization in patients with refractory angina 83; no difference in the mean total exercise time, angina-free exercise capacity, and Canadian Cardiovascular Society (CCS) class could be identified between the 2 groups. Overall, spinal cord stimulation appears safe. The initial concerns regarding a possible deprivation of a American Heart Journal Volume 155, Number 3 pain warning signal, as would be the case during a threatened MI, do not appear to be warranted.84 Animal studies even suggest that thoracic spinal cord stimulation reduces the risk of ischemic ventricular arrhythmias in postinfarction heart failure.85 The only randomized placebo-controlled trial, the Stimulation Therapy for Angina Refractory to Standard Treatments, Interventions, and Medications trial, was prematurely suspended in 2006. Results from this study have not been published.86 Other therapeutic options such as thoracic epidural anesthesia, thoracic sympathectomy, and left stellate ganglion blockade have been associated with anecdotal success but are rarely used because of their frequent side effects and invasive nature.4 Emerging therapies for chronic refractory angina Although some of the emerging therapies presented in the Table V are experimental, others are currently accepted for clinical use and are gaining wider acceptance. Nicorandil Nicorandil has been assessed among patients with established CAD and recently diagnosed as having angina during the IONA trial.89 In addition to a nitratelike effect, nicorandil opens the mitochondrial adenosine triphosphate (ATP)-sensitive potassium channels (KATP), a property presumed to mimic the ischemic preconditioning phenomenon. Compared with placebo, nicorandil reduced the combined end point of cardiovascular death, non-fatal MI, and hospital admission for cardiac chest pain (13.1% vs 15.5%, P = .014). Although nicorandil favorably influenced death and MI in a combined end point, neither component was improved when taken separately. Despite its apparent clinical benefit, nicorandil is currently not approved for use in North America. Trial data on the efficacy of nicorandil as an antianginal drug are inconsistent. The IONA trial highlights issues around the FDA requirements for approval of new antiangina medication (please see Part II: Trial Design, End points, and Regulatory Issues). Like many other therapies discussed in this section, nicorandil has not been specifically investigated in patients with refractory angina; little information is available on the safety of nicorandil in combination with traditional antiangina therapy. Metabolic modulation of ischemic myocardium In contrast to the traditional antianginal agents that operate through negative-chronotropic or negativeinotropic means, newer agents that favorably modulate energy metabolism of the myocardium are being developed. In normal conditions, free fatty acids are Jolicoeur et al 427 the main source of energy for cardiomyocytes. Agents like trimetazidine 90 and perhexiline inhibit fatty acids β-oxidation pathways at the mitochondrial level. As a result, in cardiomyocytes glucose metabolism is favored over the fatty acids metabolism.91 When compared with free fatty acids, glucose metabolism results in a higher ATP production for the same level of oxygen requirements; accordingly the imbalance between oxygen level and supply takes place at higher work loads. Metabolic modulators minimally affect blood pressure and pulse rate so that they may be useful in hypotensive and bradycardic patients already on maximal traditional therapy. Therapeutic angiogenesis Therapeutic angiogenesis could be defined as the use of angiogenic growth factors, genes that encode for growth factors, or cell-based therapies to enhance the natural process of collateral vessel development in ischemic tissue.92 Enthusiasm for gene therapy expanded in response to the lack of definitive efficacy of short-lived recombinant protein therapy 16,19 ( Table VI). Unlike protein therapy, gene therapy has the potential to induce a sustained tissue-based expression of proangiogenic factors. Several phase I trials established the safety of gene therapy for patients with CAD.99 Although phase II trials have generally been disappointing given their negative primary end point (total exercise time or reduction of ischemia using single-photon emission computed tomography [SPECT] imaging), some have however demonstrated a reduction in angina and an improvement in quality of life17,20,21,96 ( Table VI). Losordo et al20 have shown a positive effect of vascular endothelial growth factor-2 (VEGF-2) gene therapy in a randomized placebo-controlled trial. Using plasmid DNA delivered by a NOGA transendocardial delivery system (Biosense Webster, Diamond Bar, CA), not only could they objectify a reduction of the selfreported angina in the active treatment group, but they also were able to document a clinically encouraging trend in exercise stress test time improvement (1.5 minutes prolongation on a Bruce protocol in the active treatment group; P = .26). The trial was interrupted prematurely at FDA request after a patient died from gene therapy in an unrelated adenoviral trial. In 2003, the Kuopio Angiogenesis Trial suggested that intracoronary adenoviral VEGF infusion was superior to plasmid VEGF or placebo at reducing the size of SPECT reversible perfusion defect. However, exercise stress test parameters did not improve.96 Although encouraging, the Kuopio trial needs to be interpreted with caution because it was primarily intended to assess the effect of VEGF gene therapy on post-PCI restenosis. Likewise, the variability brought by the concomitant PCI complicates the interpretation of the anti-ischemic effect observed in the trial. 428 Jolicoeur et al More recently, Henry et al21 reported the analysis of the pooled results of the Angiogenic GENe Therapy (AGENT) III and IV trials that assessed Ad5FGF-4 in patients with advance CAD. In the overall study population, total exercise time was not significantly modified by the different doses of Ad5FGF-4 when compared with placebo. However, a prespecified analysis suggested that women significantly benefited from the gene therapy. The beneficial effect was consistently observed in several angina or ischemia measures: total ETT time, time to 1 mm ST-segment depression, and CCS angina class. As reported by the investigators, a negligible placebo in women and a large placebo effect in men might explain the difference observed in sexes.21 Interpretation of gene therapy should take into account the gene, its carrying vector, and the delivery method used.100 To date, most clinical data have been obtained with genes encoding for different isoforms of VEGF and fibroblast growth factor (FGF). These genes have been selected for their potent proangiogenic capacity.101 The choice of vectors depends on the goal of the therapy and the admissible side effects. DNA plasmids are regarded as safe, low-immunogenic agents; however, they have poor transduction efficiency and result in a transitory gene expression. Adenovirus results in more efficient transduction rates. However, adenoviruses show transient cellular expression and provoke immunogenic reaction.102 Alternative vectors such as adeno-associated virus and retrovirus, although endowed with attracting properties, have not yet been studied in the cardiovascular clinical arena.100 The delivery methods currently used in clinic are direct intramyocardial injection (either epicardial or endocardial) and catheter-based intracoronary perfusion. Direct injection assures an efficient albeit focal myocardial gene delivery but is invasive and may result in major complications.17 Innovative delivery methods such as ultrasound-targeted microbubble destruction and coronary sinus retroinfusion may well be used soon in clinical trials.100 Cellular therapy Aside from their theoretical regenerative capacity, certain cells, including endothelial progenitor cells, have the potential to promote angiogenesis in patients with advanced CAD. Stem cells of various types are capable of secreting cardioprotective and proangiogenic factors to support the development of new blood vessels when transplanted in severely ischemic myocardium.103,104 It is hoped that the neovascularization resulting from stem cell transplantation will reduce the ischemic burden and relieve symptoms like angina or heart failure.105,106 Although the clinical experience with cellular therapy in acute MI and CHF is growing, the number of trials directed at patients with advanced CAD remains limited.107 The analysis of clinical trials using stem cell American Heart Journal March 2008 therapy in patients with refractory angina should take into account the type of cell and the mode of delivery. In patients with refractory angina, 3 distinct types of cell preparations have been studied so far: skeletal myoblasts, bone marrow mononuclear cells (BMMCs), and circulating CD34+ progenitor cells. With the exception of the POZNAN trial,108 all of the clinical experiments using skeletal myoblasts were performed concomitantly with CABG surgery. Although early phase 1 trials consistently demonstrated the feasibility and safety of myoblast transplantation for ischemic heart failure,109,110 proof of efficacy is still awaited. In the first randomized placebo control Myoblast Autologous Grafting in Ischemic Cardiomyopathy trial, patients undergoing CABG surgery were concomitantly transplanted with either autologous skeletal myoblasts or placebo within their nonrevascularizable myocardial segments. The trial was terminated prematurely after the independent data monitoring board advised that a therapeutic effect of skeletal myoblasts was unlikely to be shown.111 It is generally felt that the variability brought by a concomitant CABG surgery significantly hampered the detection of any potential beneficial effect of skeletal myoblasts. Skeletal muscle cells have not been assessed properly in patients with refractory ischemia. Bone marrow mononuclear cells are obtained from the centrifugation of a filtered bone marrow harvest. The BMMCs contain several subpopulations of cells, among which hematopoietic and mesenchymal stem cells have been shown to possess stem cell properties: They are estimated to represent 2% to 4% of BMMCs' cellular composition.112 In an open label trial, Perin et al113 reported a significant reduction of the reversible ischemic defect (assessed by SPECT) among patients with nonrevascularizable ischemia who where treated with BMMCs using a NOGA Myostar catheter (Biosense Webster). Four months after transplantation, BMMCs patients have shown a significant improvement in LVEF (from 20% to 29%, P = .003). At least one other phase I to II trial reported similar conclusions.114 Circulating progenitor cells represent a heterogeneous group of cells expressing the hematopoietic marker proteins CD133, CD34, and the endothelial marker VEGFR2. Despite the debate around the definition of circulating progenitors cells, the clinical experience with circulating CD34+ cells in chronic or refractory angina is probably the most advanced. CD34+ circulating progenitor cells are usually collected from the blood after being mobilized from the bone marrow with granulocyte colony-stimulating factor for several days. One potential problem with granulocyte colony-stimulating factor in patients with advanced CAD is that it may transiently increase angina as a result of increases in blood viscosity, metabolic demand, and thrombocytosis.115,116 Erbs et al117 compared the efficacy of an intracoronary infusion of CD34+ circulating cells to a sham placebo in American Heart Journal Volume 155, Number 3 26 patients with CTO successfully recanalized. When compared with placebo, patients treated with CD34+ circulating cells experienced a significant decline in the number of hibernating segments in the target region. Hence, a concordant improvement of the regional wall motion (assessed by magnetic resonance imaging) was observed.117 More recently, Losordo et al115 demonstrated the safety and the feasibility of injecting autologous CD 34+ cells into the myocardium using electromechanical mapping to guide the injections toward potentially viable regions of the left ventricle (NOGA Myostar catheter, Biosense Webster). These results served as the basis for a larger phase IIb trial, which is currently under way. Several challenges still await cellular therapy before its clinical use is accepted. Unlike MI, advanced CAD has not been assessed properly with randomized control stem cell therapy trials. In the near future, genetic manipulation of stem cells either to prolong their survival 118 or to enhance their cytokine secretion profile 119 should bring additional hope to this already promising field. Antioxidant and chelation therapy In 1993, Grier and Meyers120 entitled their review article on 37 years of ethylenediaminetetraacetic acid (EDTA) chelation therapy, “So much writing, so little science.” The authors came to the conclusion that, collectively, the reported literature did not provide reliable evidence to support a benefit of chelation therapy in atherosclerosis. At that time, the medical literature primarily consisted of testimonials, case reports, and case series. Since then, no less than 5 randomized controlled trials have been designed to test the efficacy of chelation therapy in patients with advanced atherosclerosis. EDTA is an amino acid complex with high affinity for cations such as lead, magnesium, zinc, iron, and calcium. Once in contact with EDTA, cations are chelated into an inactive state and subsequently excreted by the kidneys. Over the years, several putative mechanisms have been proposed to explain how EDTA may reduce atherosclerosis progression. Current hypotheses state that EDTA reduces low-density lipoprotein oxidation by binding copper and iron.121 The resultant antioxidant effect is presumed to decrease the risk of developing atherosclerosis.122,123 Indirect mechanisms involving parathyroid hormone (PTH) may also be involved. By inducing hypocalcemia, EDTA stimulates the secretion of PTH, which is known to relax smooth muscle cells (inhibition of L calcium channel), induce vasodilatation, and decrease systemic blood pressure.124,125 Other mechanisms of action, linking PTH and t-PA production, have also been proposed.126,127 The Program to Assess Alternative Treatment Strategies to Achieve Cardiac Health trial was a double-blind, randomized controlled trial comparing EDTA with Jolicoeur et al 429 placebo in 81 patients with stable angina. Seven months of EDTA administration resulted in a trivial 9-second improvement in the exercise treadmill ischemic time (as assessed by the time to 1 mm ST-segment deviation) when compared with placebo. Likewise, total exercise capacity and quality of life scores improved by similar degrees in both groups, again suggesting an important placebo effect.128 The Trial to Assess Chelation Therapy (TACT), sponsored by the NHLBI and the National Center for Complementary and Alternative Medicine, is a phase III trial currently recruiting patients in the United States. TACT aims to randomly assign 1950 patients with recent MI to receive 40 infusions of either the standard chelation solution or placebo. The primary end point will be a composite of all-cause mortality, MI, stroke, and hospitalization for angina or for CHF over a mean follow-up of 2.5 years. TACT is expected to bring a significant positive result or an informative null result upon which rational clinical decision making and health policy can be based. Results are expected to be available by 2010. Although not directly related to angina and refractory ischemia, the TACT trial should provide important safety information for an agent frequently used to treat refractory angina. The risks associated with chelation therapy have been reported to be substantial (renal failure, arrhythmia, bone marrow depression, and even death), but the clinical proofs of efficacy are still awaited.129 More recently, the Aggressive Reduction of Inflammation Stops Events (ARISE) study assessed the antioxidant succinobucol (AGI-1067) in the prevention of recurrent ischemic events on patients with recent ACS. Before ARISE, atherosclerosis regression was demonstrated by intravascular ultrasound using this agent.130 In ARISE, however, succinobucol did not significantly influence the combined end point of death, resuscitated cardiac arrest, MI, stroke, coronary revascularization, and hospitalization for unstable angina.131 Percutaneous bypasses and retroperfusion technologies Retroperfusion technologies exploit the coronary sinus to redistribute either flow or pressure from the venules to the capillaries. To date, both flow-driven retroperfusion and pressure-driven retroperfusion have been explored in clinical studies. Both of these techniques are the earliest stage of development but represent important new possibilities in the treatment of advanced CAD. With flow-driven retroperfusion, blood from an artery is shunted to the coronary vein that drains the ischemic myocardial region. Using this principle, oxygenated blood shunts through the venules and subsequently reaches the capillaries, assuring a backward irrigation of the ischemic myocardium. Recently, percutaneous in situ coronary venous arterialization (PICVA) has been attempted in patients with severe angina and no revascularization options.132,133 This technology links American Heart Journal March 2008 430 Jolicoeur et al the coronary artery proximal to its site of occlusion to its correspondent coronary vein. The coronary sinus is selectively occluded to permit an appropriate retrograde redistribution of the oxygenated blood to the venules. The PICVA requires either intravascular ultrasound positioning or advanced NOGA navigation system to be performed. Pressure-driven retroperfusion exploits the principle that coronary flow distribution varies according to coronary perfusion pressure. The anti-anginal effect of a partial occlusion of the coronary sinus has been known for several decades.134 By selectively occluding venous drainage of a given region of the heart, the resulting imbalance in regional capillary pressure will lead to a redistribution of antegrade blood toward other regions of lesser resistance. As such, it is possible to favor blood flow in diseased coronary segments by selectively occluding venous drainage of other flow-competing, nondiseased coronary segments. The Reducer (Neovasc Medical, Israel) is a stainless steel, balloon-expandable, hourglassshaped stent that creates a selective, permanent, and controlled narrowing of the coronary sinus. First-in-man series reported in 2007 has shown the safety and the feasibility of this technique.135 Conclusions Aside for the traditional nitrates, calcium blockers, and β-blockers, few therapies have been deemed effective to control refractory angina. Ranolazine is the first medical therapy approved in more than a decade for chronic angina and should be considered in the treatment of patients with advanced CAD. Among mechanical therapies, EECP and transmyocardial laser revascularization are approved for the treatment of refractory angina. Finally, CTO recanalization and metabolic modulators have been shown to consistently relieve angina on top of standard treatment but formal evidence of efficacy are still lacking. Despite these advances, many patients remain symptomatic. It is time to evaluate whether clinical programs dedicated to these no-option patients may provide improved care. Those clinical programs, as was the case for heart failure clinics in the 1990s, would permit a better characterization of the disease and improve accessibility to sophisticated treatments. More importantly, development of these clinical programs would accelerate our ability to provide new and innovative ways to treat CAD in the future. Developing new therapies for patients with advanced coronary disease is difficult for several reasons. The prevalence of advanced CAD is poorly defined, and the incidence of mortality and coronary events is not known. Because of that, selecting the appropriate trial design, sample size, and outcomes are difficult. Additional challenges stem from the substantial placebo effect encountered with a subjective outcome like angina. This has been illustrated by several trials presented in this review, especially when therapies involving invasive procedures were being investigated. Indeed, such a placebo effect calls for properly blinded, sham controlled trials. In the second part of this article, we will discuss the difficulties encountered by investigators when designing trials for refractory angina in terms of trial design, end point selection, and compliance with regulatory agencies. The authors thank Jonathan McCall for his excellent editorial assistance. References 1. Fox K, Garcia MA, Ardissino D, et al. Guidelines on the management of stable angina pectoris: executive summary: the Task Force on the Management of Stable Angina Pectoris of the European Society of Cardiology. Eur Heart J 2006;27:1341-81. 2. Gibbons RJ, Abrams J, Chatterjee K, et al. ACC/AHA 2002 guideline update for the management of patients with chronic stable angina—summary article: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (Committee on the Management of Patients With Chronic Stable Angina). Circulation 2003;107:149-58. 3. Opie LH, Commerford PJ, Gersh BJ. Controversies in stable coronary artery disease. Lancet 2006;367:69-78. 4. Mannheimer C, Camici P, Chester MR, et al. The problem of chronic refractory angina; report from the ESC Joint Study Group on the Treatment of Refractory Angina. Eur Heart J 2002;23:355-70. 5. Mukherjee D, Bhatt DL, Roe MT, et al. Direct myocardial revascularization and angiogenesis-how many patients might be eligible? Am J Cardiol 1999;84:598-600, A8. 6. Bernstein SJ, Brorsson B, Aberg T, et al. Appropriateness of referral of coronary angiography patients in Sweden. SECOR/SBU Project Group. Heart 1999;81:470-7. 7. Brorsson B, Bernstein SJ, Brook RH, et al. Quality of life of patients with chronic stable angina before and four years after coronary revascularisation compared with a normal population. Heart 2002; 87:140-5. 8. Thom T, Haase N, Rosamond W, et al. Heart disease and stroke statistics—2006 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2006;113:e85-e151. 9. Aaberge L, Nordstrand K, Dragsund M, et al. Transmyocardial revascularization with CO2 laser in patients with refractory angina pectoris: clinical results from The Norwegian Randomized Trial. J Am Coll Cardiol 2000;35:1170-7. 10. Allen KB, Dowling RD, Fudge TL, et al. Comparison of transmyocardial revascularization with medical therapy in patients with refractory angina. N Engl J Med 1999;341:1029-36. 11. Burkhoff D, Schmidt S, Schulman SP, et al. Transmyocardial laser revascularisation compared with continued medical therapy for treatment of refractory angina pectoris: a prospective randomised trial. Lancet 1999;354:885-90. 12. Frazier OH, March RJ, Horvath KA, et al. Transmyocardial revascularization with a carbon dioxide laser in patients with end-stage coronary artery disease. N Engl J Med 1999;341: 1021-8. American Heart Journal Volume 155, Number 3 13. Leon MB, Kornowski R, Downey WE, et al. A blinded, randomized, placebo-controlled trial of percutaneous laser myocardial revascularization to improve angina symptoms in patients with severe coronary disease. J Am Coll Cardiol 2005; 46:1812-9. 14. Oesterle SN, Sanborn TA, Ali N, et al. Percutaneous transmyocardial laser revascularisation for severe angina: the PACIFIC randomised trial. Lancet 2000;356:1705-10. 15. Schofield PM, Sharples LD, Caine N, et al. Transmyocardial laser revascularisation in patients with refractory angina: a randomised controlled trial. Lancet 1999;353:519-24. 16. Henry TD, Annex BH, McKendall GR, et al. The VIVA trial: vascular endothelial growth factor in ischemia for vascular angiogenesis. Circulation 2003;107:1359-65. 17. Kastrup J, Jorgensen E, Ruck A, et al. Direct intramyocardial plasmid vascular endothelial growth factor-A165 gene therapy in patients with stable severe angina pectoris. A randomized double-blind placebo-controlled study: the Euroinject One trial. J Am Coll Cardiol 2005;45:982-8. 18. Stewart DJ, Hilton JD, Arnold JM, et al. Angiogenic gene therapy in patients with nonrevascularizable ischemic heart disease: a phase 2 randomized, controlled trial of AdVEGF(121) (AdVEGF121) versus maximum medical treatment. Gene Ther 2006;13:1503-11. 19. Simons M, Annex BH, Laham RJ, et al. Pharmacological treatment of coronary artery disease with recombinant fibroblast growth factor2: double-blind, randomized, controlled clinical trial. Circulation 2002;105:788-93. 20. Losordo DW, Vale PR, Hendel RC, et al. Phase 1/2 placebocontrolled, double-blind, dose-escalating trial of myocardial vascular endothelial growth factor 2 gene transfer by catheter delivery in patients with chronic myocardial ischemia. Circulation 2002;105: 2012-8. 21. Henry TD, Grines CL, Watkins MW, et al. Effects of Ad5FGF-4 in patients with angina: an analysis of pooled data from the AGENT-3 and AGENT-4 trials. J Am Coll Cardiol 2007;50:1038-46. 22. Kandzari DE, Lam LC, Eisenstein EL, et al. Advanced coronary artery disease: appropriate end points for trials of novel therapies. Am Heart J 2001;142:843-51. 23. Henry TD, Satran D, Johnson RJ, et al. Natural history of patients with refractory angina. J Am Coll Cardiol 2006;47:231. 24. Mouallem M, Schwartz E, Farfel Z. Prolonged oral morphine therapy for severe angina pectoris. J Pain Symptom Manage 2000;19: 393-7. 25. Uen S, Un I, Fimmers R, et al. Myocardial ischemia during everyday life in patients with arterial hypertension: prevalence, risk factors, triggering mechanism and circadian variability. Blood Press Monit 2006;11:173-82. 26. Smolensky MH, Portaluppi F. Chronopharmacology and chronotherapy of cardiovascular medications: relevance to prevention and treatment of coronary heart disease. Am Heart J 1999;137(4 Pt 2): S14-S24. 27. CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet 1996;348:1329-39. 28. The Clopidogrel in Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med 2001;345:494-502. 29. Bhatt DL, Fox KAA, Hacke W, et al. Clopidogrel and aspirin versus aspirin alone for the prevention of atherothrombotic events. N Engl J Med 2006;354:1706-17. Jolicoeur et al 431 30. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 2005;366:1267-78. 31. Claeys MJ, Cosyns B, Hoffer E, et al. Short-term effect of atorvastatin on ischaemic threshold in hypercholesterolaemic patients with stable ischaemic heart disease. Acta Cardiol 2004;59:269-74. 32. Calabro P, Yeh ET. The pleiotropic effects of statins. Curr Opin Cardiol 2005;20:541-6. 33. Nissen SE, Tuzcu EM, Schoenhagen P, et al. Statin therapy, LDL cholesterol, C-reactive protein, and coronary artery disease. N Engl J Med 2005;352:29-38. 34. Nissen SE, Nicholls SJ, Sipahi I, et al. Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial. JAMA 2006;295:1556-65. 35. Nissen SE, Tsunoda T, Tuzcu EM, et al. Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. JAMA 2003;290: 2292-300. 36. Tardif JC, Gregoire J, L'Allier PL, et al. Effects of reconstituted highdensity lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial. JAMA 2007;297:1675-82. 37. Deedwania P, Stone PH, Bairey CN, et al. Effects of intensive versus moderate lipid-lowering therapy on myocardial ischemia in older patients with coronary heart disease: results of the Study Assessing Goals in the Elderly (SAGE). Circulation 2007;115: 700-7. 38. Nissen SE, Tardif JC, Nicholls SJ, et al. Effect of torcetrapib on the progression of coronary atherosclerosis. N Engl J Med 2007;356: 1304-16. 39. McCormack JG, Barr RL, Wolff AA, et al. Ranolazine stimulates glucose oxidation in normoxic, ischemic, and reperfused ischemic rat hearts. Circulation 1996;93:135-42. 40. Wolff AA, Rotmensch HH, Stanley WC, et al. Metabolic approaches to the treatment of ischemic heart disease: the clinicians' perspective. Heart Fail Rev 2002;7:187-203. 41. Chaitman BR, Pepine CJ, Parker JO, et al. Effects of ranolazine with atenolol, amlodipine, or diltiazem on exercise tolerance and angina frequency in patients with severe chronic angina: a randomized controlled trial. JAMA 2004;291:309-16. 42. Zerumsky K, McBride BF. Ranolazine in the management of chronic stable angina. Am J Health Syst Pharm 2006;63:2331-8. 43. Antzelevitch C, Belardinelli L, Zygmunt AC, et al. Electrophysiological effects of ranolazine, a novel antianginal agent with antiarrhythmic properties. Circulation 2004;110:904-10. 44. Belardinelli L, Shryock JC, Fraser H. Inhibition of the late sodium current as a potential cardioprotective principle: effects of the late sodium current inhibitor ranolazine. Heart 2006;92(Suppl 4): iv6-iv14. 45. Eng S, Maddaford TG, Kardami E, et al. Protection against myocardial ischemic/reperfusion injury by inhibitors of two separate pathways of Na+ entry. J Mol Cell Cardiol 1998;30: 829-35. 46. Eigel BN, Hadley RW. Contribution of the Na(+) channel and Na (+)/H(+) exchanger to the anoxic rise of [Na(+)] in ventricular myocytes. Am J Physiol 1999;277(5 Pt 2):H1817-22. 47. Chaitman BR, Skettino SL, Parker JO, et al. Anti-ischemic effects and long-term survival during ranolazine monotherapy in patients with chronic severe angina. J Am Coll Cardiol 2004;43:1375-82. 48. Hung J, Lamb IH, Connolly SJ, et al. The effect of diltiazem and propranolol, alone and in combination, on exercise performance and left ventricular function in patients with stable effort angina: a 432 Jolicoeur et al 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. double-blind, randomized, and placebo-controlled study. Circulation 1983;68:560-7. Pehrsson SK, Ringqvist I, Ekdahl S, et al. Monotherapy with amlodipine or atenolol versus their combination in stable angina pectoris. Clin Cardiol 2000;23:763-70. Fox KM, Mulcahy D, Findlay I, et al. The Total Ischaemic Burden European Trial (TIBET). Effects of atenolol, nifedipine SR and their combination on the exercise test and the total ischaemic burden in 608 patients with stable angina. The TIBET Study Group. Eur Heart J 1996;17:96-103. Stone PH, Gratsiansky NA, Blokhin A, et al. Antianginal efficacy of ranolazine when added to treatment with amlodipine: the ERICA (Efficacy of Ranolazine in Chronic Angina) trial. J Am Coll Cardiol 2006;48:566-75. Koren MJ, Crager MR, Sweeney M. Long-term safety of a novel antianginal agent in patients with severe chronic stable angina: the Ranolazine Open Label Experience (ROLE). J Am Coll Cardiol 2007;49:1027-34. Morrow DA, Scirica BM, Karwatowska-Prokopczuk E, et al. Effects of ranolazine on recurrent cardiovascular events in patients with non–ST-elevation acute coronary syndromes: the MERLIN-TIMI 36 randomized trial. JAMA 2007;297:1775-83. O'Meara E, McMurray JJV. Myocardial metabolic manipulation: a new therapeutic approach in heart failure? Heart 2005;91:131-2. Sorensen HT, Rasmussen HH, Moller-Petersen JF, et al. Epidemiology of pain requiring strong analgesics outside hospital in a geographically defined population in Denmark. Dan Med Bull 1992;39:464-7. Ballantyne JC, Mao J. Opioid therapy for chronic pain. N Engl J Med 2003;349:1943-53. Stone GW, Kandzari DE, Mehran R, et al. Percutaneous recanalization of chronically occluded coronary arteries: a consensus document: part I. Circulation 2005;112:2364-72. Srinivas VS, Brooks MM, Detre KM, et al. Contemporary percutaneous coronary intervention versus balloon angioplasty for multivessel coronary artery disease: a comparison of the National Heart, Lung and Blood Institute Dynamic Registry and the Bypass Angioplasty Revascularization Investigation (BARI) study. Circulation 2002;106:1627-33. Hochman JS, Lamas GA, Buller CE, et al. Coronary intervention for persistent occlusion after myocardial infarction. N Engl J Med 2006; 355:2395-407. Suero JA, Marso SP, Jones PG, et al. Procedural outcomes and longterm survival among patients undergoing percutaneous coronary intervention of a chronic total occlusion in native coronary arteries: a 20-year experience. J Am Coll Cardiol 2001;38:409-14. Prasad A, Rihal CS, Lennon RJ, et al. Trends in outcomes after percutaneous coronary intervention for chronic total occlusions: a 25-year experience from the Mayo Clinic. J Am Coll Cardiol 2007; 49:1611-8. Abbott JD, Kip KE, Vlachos HA, et al. Recent trends in the percutaneous treatment of chronic total coronary occlusions. Am J Cardiol 2006;97:1691-6. Olivari Z, Rubartelli P, Piscione F, et al. Immediate results and oneyear clinical outcome after percutaneous coronary interventions in chronic total occlusions: data from a multicenter, prospective, observational study (TOAST-GISE). J Am Coll Cardiol 2003;41: 1672-8. Ruocco Jr NA, Ring ME, Holubkov R, et al. Results of coronary angioplasty of chronic total occlusions (the National Heart, Lung, and Blood Institute 1985-1986 Percutaneous Transluminal Angioplasty Registry). Am J Cardiol 1992;69:69-76. American Heart Journal March 2008 65. Stone GW, Reifart NJ, Moussa I, et al. Percutaneous recanalization of chronically occluded coronary arteries: a consensus document: part II. Circulation 2005;112:2530-7. 66. Contreras G, Cieza T, Hardy N, et al. Uncrossable coronary obstruction treated by the new laser guidewire. J Invasive Cardiol 2005;17:560-2. 67. Abbas AE, Brewington SD, Dixon SR, et al. Intracoronary fibrinspecific thrombolytic infusion facilitates percutaneous recanalization of chronic total occlusion. J Am Coll Cardiol 2005;46:793-8. 68. Strauss BH, Goldman L, Qiang B, et al. Collagenase plaque digestion for facilitating guide wire crossing in chronic total occlusions. Circulation 2003;108:1259-62. 69. Sinvhal RM, Gowda RM, Khan IA. Enhanced external counterpulsation for refractory angina pectoris. Heart 2003;89: 830-3. 70. Arora RR, Chou TM, Jain D, et al. The Multicenter Study of Enhanced External Counterpulsation (MUST-EECP): effect of EECP on exerciseinduced myocardial ischemia and anginal episodes. J Am Coll Cardiol 1999;33:1833-40. 71. Niebauer J, Cooke JP. Cardiovascular effects of exercise: role of endothelial shear stress. J Am Coll Cardiol 1996;28:1652-60. 72. Michaels AD, Raisinghani A, Soran O, et al. The effects of enhanced external counterpulsation on myocardial perfusion in patients with stable angina: a multicenter radionuclide study. Am Heart J 2005; 150:1066-73. 73. Lawson WE, Hui JC, Zheng ZS, et al. Improved exercise tolerance following enhanced external counterpulsation: cardiac or peripheral effect? Cardiology 1996;87:271-5. 74. Urano H, Ikeda H, Ueno T, et al. Enhanced external counterpulsation improves exercise tolerance, reduces exercise-induced myocardial ischemia and improves left ventricular diastolic filling in patients with coronary artery disease. J Am Coll Cardiol 2001;37:93-9. 75. Masuda D, Nohara R, Hirai T, et al. Enhanced external counterpulsation improved myocardial perfusion and coronary flow reserve in patients with chronic stable angina; evaluation by(13)Nammonia positron emission tomography. Eur Heart J 2001;22: 1451-8. 76. Wu G, Du Z, Hu C, et al. Angiogenic effects of long-term enhanced external counterpulsation in a dog model of myocardial infarction. Am J Physiol Heart Circ Physiol 2006;290:H248-54. 77. Lawson WE, Hui JC, Lang G. Treatment benefit in the enhanced external counterpulsation consortium. Cardiology 2000;94:31-5. 78. Peterson PN, Spertus JA, Magid DJ, et al. The impact of diabetes on one-year health status outcomes following acute coronary syndromes. BMC Cardiovasc Disord 2006;6:41. 79. Soran O, Kennard ED, Kfoury AG, et al. Two-year clinical outcomes after enhanced external counterpulsation (EECP) therapy in patients with refractory angina pectoris and left ventricular dysfunction (report from The International EECP Patient Registry). Am J Cardiol 2006;97:17-20. 80. Michaels AD, McCullough PA, Soran OZ, et al. Primer: practical approach to the selection of patients for and application of EECP. Nat Clin Pract Cardiovasc Med 2006;3:623-32. 81. Oakley JC, Prager JP. Spinal cord stimulation: mechanisms of action. Spine 2002;27:2574-83. 82. Hautvast RW, De Jongste MJ, Staal MJ, et al. Spinal cord stimulation in chronic intractable angina pectoris: a randomized, controlled efficacy study. Am Heart J 1998;136:1114-20. 83. McNab D, Khan SN, Sharples LD, et al. An open label, singlecentre, randomized trial of spinal cord stimulation vs. percutaneous myocardial laser revascularization in patients with refractory angina pectoris: the SPiRiT trial. Eur Heart J 2006;27:1048-53. American Heart Journal Volume 155, Number 3 84. Buchser E, Durrer A, Albrecht E. Spinal cord stimulation for the management of refractory angina pectoris. J Pain Symptom Manage 2006;31(4 Suppl):S36-S42. 85. Issa ZF, Zhou X, Ujhelyi MR, et al. Thoracic spinal cord stimulation reduces the risk of ischemic ventricular arrhythmias in a postinfarction heart failure canine model. Circulation 2005;111:3217-20. 86. STARTSTIM (Stimulation Therapy for Angina Refractory To Standard Treatments, Interventions and Medications) www.clinicaltrial.gov (id: NCT00200070). Last access on 10/10/2007. 87. Tardif JC, Ford I, Tendera M, et al. Efficacy of ivabradine, a new selective I(f) inhibitor, compared with atenolol in patients with chronic stable angina. Eur Heart J 2005;26:2529-36. 88. Nishida T, Shimokawa H, Oi K, et al. Extracorporeal cardiac shock wave therapy markedly ameliorates ischemia-induced myocardial dysfunction in pigs in vivo. Circulation 2004;110:3055-61. 89. The IONA Investogators. Effect of nicorandil on coronary events in patients with stable angina: the Impact Of Nicorandil in Angina (IONA) randomised trial. Lancet 2002;359:1269-75. 90. Ciapponi A, Pizarro R, Harrison J. Trimetazidine for stable angina. Cochrane Database Syst Rev 2005:CD003614. 91. Lee L, Horowitz J, Frenneaux M. Metabolic manipulation in ischaemic heart disease, a novel approach to treatment. Eur Heart J 2004;25:634-41. 92. Henry TD. Therapeutic angiogenesis. BMJ 1999;318:1536-9. 93. Laitinen M, Hartikainen J, Hiltunen MO, et al. Catheter-mediated vascular endothelial growth factor gene transfer to human coronary arteries after angioplasty. Hum Gene Ther 2000;11:263-70. 94. Grines CL, Watkins MW, Helmer G, et al. Angiogenic Gene Therapy (AGENT) trial in patients with stable angina pectoris. Circulation 2002;105:1291-7. 95. Grines CL, Watkins MW, Mahmarian JJ, et al. A randomized, double-blind, placebo-controlled trial of Ad5FGF-4 gene therapy and its effect on myocardial perfusion in patients with stable angina. J Am Coll Cardiol 2003;42:1339-47. 96. Hedman M, Hartikainen J, Syvanne M, et al. Safety and feasibility of catheter-based local intracoronary vascular endothelial growth factor gene transfer in the prevention of postangioplasty and in-stent restenosis and in the treatment of chronic myocardial ischemia: phase II results of the Kuopio Angiogenesis Trial (KAT). Circulation 2003;107:2677-83. 97. Gyongyosi M, Khorsand A, Zamini S, et al. NOGA-guided analysis of regional myocardial perfusion abnormalities treated with intramyocardial injections of plasmid encoding vascular endothelial growth factor A-165 in patients with chronic myocardial ischemia: subanalysis of the EUROINJECT-ONE multicenter double-blind randomized study. Circulation 2005;112 (9 Suppl):I157-65. 98. Penny WF, Hammond HK. Clinical use of intracoronary gene transfer of fibroblast growth factor for coronary artery disease. Curr Gene Ther 2004;4:225-30. 99. Staudacher DL, Preis M, Lewis BS, et al. Cellular and molecular therapeutic modalities for arterial obstructive syndromes. Pharmacol Ther 2006;109:263-73. 100. Muller OJ, Katus HA, Bekeredjian R. Targeting the heart with gene therapy–optimized gene delivery methods. Cardiovasc Res 2007; 73:453-62. 101. Tomanek RJ, Sandra A, Zheng W, et al. Vascular endothelial growth factor and basic fibroblast growth factor differentially modulate early postnatal coronary angiogenesis. Circ Res 2001;88:1135-41. 102. Assessment of adenoviral vector safety and toxicity: report of the National Institutes of Health Recombinant DNA Advisory Committee. Hum Gene Ther 2002;13:3-13. Jolicoeur et al 433 103. Kinnaird T, Stabile E, Burnett MS, et al. Bone-marrow–derived cells for enhancing collateral development: mechanisms, animal data, and initial clinical experiences. Circ Res 2004;95:354-63. 104. Kocher AA, Schuster MD, Szabolcs MJ, et al. Neovascularization of ischemic myocardium by human bone-marrow–derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med 2001;7:430-6. 105. Kinnaird T, Stabile E, Burnett MS, et al. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res 2004;94:678-85. 106. Kinnaird T, Stabile E, Burnett MS, et al. Local delivery of marrowderived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation 2004;109:1543-9. 107. Jolicoeur EM, Granger CB, Fakunding JL, et al. Bringing cardiovascular cell-based therapy to clinical application: perspectives based on a National Heart, Lung, and Blood Institute Cell Therapy Working Group meeting. Am Heart J 2007;153:732-42. 108. Siminiak T, Fiszer D, Jerzykowska O, et al. Percutaneous transcoronary-venous transplantation of autologous skeletal myoblasts in the treatment of post-infarction myocardial contractility impairment: the POZNAN trial. Eur Heart J 2005;26:1188-95. 109. Hagege AA, Marolleau JP, Vilquin JT, et al. Skeletal myoblast transplantation in ischemic heart failure: long-term follow-up of the first phase I cohort of patients. Circulation 2006;114(1 Suppl): I108-13. 110. Menasche P, Hagege AA, Scorsin M, et al. Myoblast transplantation for heart failure. Lancet 2001;357:279-80. 111. Mills JM, Adam GL, Tricoci P, et al. Highlights from the 20th Scientific Sessions of the American Heart Association: November 12 to 15, 2006, Chicago, Illinois. Am Heart J 2007;153:213-23. 112. Fuchs S, Satler LF, Kornowski R, et al. Catheter-based autologous bone marrow myocardial injection in no-option patients with advanced coronary artery disease: a feasibility study. J Am Coll Cardiol 2003;41:1721-4. 113. Perin EC, Dohmann HF, Borojevic R, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003;107:2294-302. 114. Tse HF, Kwong YL, Chan JK, et al. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003;361:47-9. 115. Losordo DW, Schatz RA, White CJ, et al. Intramyocardial transplantation of autologous CD34+ stem cells for intractable angina: a phase I/IIa double-blind, randomized controlled trial. Circulation 2007;115:3165-72. 116. Wilson RF, Henry TD. Granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor: double-edged swords. J Am Coll Cardiol 2005;46:1649-50. 117. Erbs S, Linke A, Adams V, et al. Transplantation of blood-derived progenitor cells after recanalization of chronic coronary artery occlusion: first randomized and placebo-controlled study. Circ Res 2005;97:756-62. 118. Mangi AA, Noiseux N, Kong D, et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med 2003;9:1195-201. 119. Eliopoulos N, Al-Khaldi A, Crosato M, et al. A neovascularized organoid derived from retrovirally engineered bone marrow stroma leads to prolonged in vivo systemic delivery of erythropoietin in nonmyeloablated, immunocompetent mice. Gene Ther 2003;10:478-89. 120. Grier MT, Meyers DG. So much writing, so little science: a review of 37 years of literature on edetate sodium chelation therapy. Ann Pharmacother 1993;27:1504-9. American Heart Journal March 2008 434 Jolicoeur et al 121. Lamb DJ, Leake DS. The effect of EDTA on the oxidation of low density lipoprotein. Atherosclerosis 1992;94:35-42. 122. Klatt P, Esterbauer H. Oxidative hypothesis of atherogenesis. J Cardiovasc Risk 1996;3:346-51. 123. Steinberg D, Parthasarathy S, Carew TE, et al. Beyond cholesterol. Modifications of low-density lipoprotein that increase its atherogenicity. N Engl J Med 1989;320:915-24. 124. Bukoski RD, Ishibashi K, Bian K. Vascular actions of the calciumregulating hormones. Semin Nephrol 1995;15:536-49. 125. Meltzer LE, Kitchell JR, Palmon Jr F. The long term use, side effects, and toxicity of disodium ethylenediamine tetraacetic acid (EDTA). Am J Med Sci 1961;242:11-7. 126. Catherwood BD, Titus L, Evans CO, et al. Increased expression of tissue plasminogen activator messenger ribonucleic acid is an immediate response to parathyroid hormone in neonatal rat osteoblasts. Endocrinology 1994;134:1429-36. 127. Leloup G, Delaisse JM, Vaes G. Relationship of the plasminogen activator/plasmin cascade to osteoclast invasion and mineral resorption in explanted fetal metatarsal bones. J Bone Miner Res 1994;9:891-902. 128. Knudtson ML, Wyse DG, Galbraith PD, et al. Chelation therapy for ischemic heart disease: a randomized controlled trial. JAMA 2002; 287:481-6. 129. Ernst E. Chelation therapy for coronary heart disease: an overview of all clinical investigations. Am Heart J 2000;140:139-41. 130. Tardif JC, Gregoire J, L'Allier PL, et al. Effects of the antioxidant succinobucol (AGI-1067) on human atherosclerosis in a randomized clinical trial. Atherosclerosis 2007. In press. 131. Adams GL, Mills JS, Melloni C, et al. Highlights from the 56th Annual Scientific Sessions of the American College of Cardiology: March 25 to 27, 2007, Atlanta, Georgia. Am Heart J 2007;154: 247-59. 132. Oesterle SN, Reifart N, Hauptmann E, et al. Percutaneous in situ coronary venous arterialization: report of the first human catheterbased coronary artery bypass. Circulation 2001;103:2539-43. 133. Oesterle SN, Reifart N, Hayase M, et al. Catheter-based coronary bypass: a development update. Catheter Cardiovasc Interv 2003; 58:212-8. 134. Robertson HF. The reestablishment of cardiac circulation during progressive coronary occlusion: an experimental study on dogs. Am Heart J 1935;10:533-41. 135. Banai S, Ben MS, Parikh KH, et al. Coronary sinus reducer stent for the treatment of chronic refractory angina pectoris: a prospective, open-label, multicenter, safety feasibility first-in-man study. J Am Coll Cardiol 2007;49:1783-9. Appendix A Members of the Working Group (in addition to coauthors): Karen Alexander, MD, Duke University Medical Center, Durham, NC; Steve C. Mockrin, PhD, NHLBI, NIH, Bethesda, MD. Presenters to the Working Group: Jonathan Abrams, MD, University of New Mexico Hospital, Albuquerque, NM; Brian Annex, MD, Duke VA Medical Center, Durham, NC; Gregory Barsness, MD, Mayo Clinic Rochester, Rochester, MN; Deepak Bhatt, MD, Cleveland Clinic, Cleveland, OH; Robert Califf, MD, Duke Clinical Research Institute, Durham, NC; Bernard R. Chaitman, MD, Saint Louis University School of Medicine, St. Louis, MO; Bernard J. Gersh, MD, Mayo Clinic, Rochester, MN; David Holmes, MD, Mayo Clinic, Rochester, MN; Timothy D. Henry, MD, Minneapolis Heart Institute, Minneapolis, MN; Andrew Koren, MD, CV Therapeutics Palo Alto, CA; Daniel Mark, MD, Duke Clinical Research Institute, Durham, NC; Alice M. Mascette, MD, NIH, NHLBI, Bethesda, MD; Magnus Ohman, MD, Duke University Medical Center, Durham, NC; Carl Pepine, MD, University of Florida, Gainesville, FL; Ileana Pina, MD, Case Western Reserve University, Cleveland Height, OH; Jyme Schafer, MPH, CMS, Baltimore, MD; Sidney Smith, MD, University of North Carolina, Chapel Hill, NC; Norman Stockbridge MD, PhD, FDA, Silver Spring, MD; Anders Svensson, MD, PhD, AstraZeneca, Molndal, Sweden; Julie Swain, FDA, CDRH, Fallbrook, CA; Robert Temple, MD, FDA, Silver Spring, MD; Lars Wallentin, MD, PhD, Uppsala Clinical Center University Hospital, Uppsala, Sweden; Doug Weaver, MD, Henry Ford Health System, Detroit, MI; Cecelia Witten, MD, PhD, FDA/ CBER, Rockville, MD; Bram Zuckerman, MD, FDA, Rockville, MD.