Open access peer-reviewed chapter

Coronary Arteries Bypass Grafting as a Salvage Surgery in Ischemic Heart Failure

Written By

Samuel Jacob, Pankaj Garg, Games Gramm and Saqib Masroor

Submitted: 05 March 2022 Reviewed: 14 April 2022 Published: 21 May 2022

DOI: 10.5772/intechopen.104939

From the Edited Volume

Coronary Artery Bypass Grafting

Edited by Takashi Murashita

Chapter metrics overview

136 Chapter Downloads

View Full Metrics

Abstract

Ischemic cardiomyopathy accounts for approximately two-thirds of all Heart Failure (HF) cases. Recent studies indicates that revascularization provides superior outcomes compared with optimal medical therapy (OMT) alone. Current European and American guidelines recommend an invasive approach in patients with reduced left ventricular ejection fraction (LVEF) less than 35% and with multivessel disease (MVD). Randomized controlled trials in these patients have proven that long-term survival is greater following coronary artery bypass grafting (CABG) than with OMT alone. Patients with ischemic cardiomyopathy and coronary artery disease that is amenable to surgical revascularization should undergo combination of surgical revascularization and medical therapy rather than medical therapy alone. In some cases, combined CABG with other surgeries are vital salvage procedures, such as atrial fibrillation, mitral valve, tricuspid valve, and LV remodeling. Based on small but, nontrivial, early mortality risk associated with CABG surgery as well as other post-CABG morbidities, patients may also reasonably choose medical therapy as initial treatment option. Revascularization remains an important treatment option for patients with ongoing anginal symptoms despite optimal medical therapy. In this chapter, we will highlight the role of CABG in heart failure treatment and when to use it as a salvage surgery before referring the patient for heart transplantation.

Keywords

  • CABG
  • ischemic heart failure
  • cardiac surgery
  • salvage surgery
  • cardiomyopathy
  • combined CABG and MVR
  • combined CABG and TVR
  • ventricular remodeling

1. Introduction

There is no universally accepted definition of ischemic cardiomyopathy (ICM). However, the term ischemic cardiomyopathy generally refers to significantly impaired left ventricular function (left ventricular ejection fraction [LVEF] ≤35–40%) that results from coronary artery disease (CAD) [1, 2, 3]. In 2002, Felker et al. suggested that the symptomatic patients with LVEF ≤40% and presence of left main or proximal left anterior descending coronary artery stenosis ≥75% or two or more epicardial coronary artery stenosis ≥75% or a prior history of coronary artery revascularization [percutaneous coronary intervention (PCI) or coronary artery bypass grafting (CABG)] or prior history of myocardial infarction should only be classified as having ICM [3].

Ischemic heart disease is a global pandemic, and its incidence continues to increase. In an estimate, 125 million people across the globe suffer from ischemic heart disease. In the United States itself, every year 720,000 people develop their first myocardial infarction (MI) resulting in hospitalization and/or death [4, 5]. Thirty-five percent of the patients who experience coronary event in a given year die due to it; and each death is associated with an average of 16 years of lost life. Patients who survive after the myocardial infarction are at an increased risk of developing ICM and eventually heart failure (HF). Etiopathogenesis of heart failure is multifactorial; however, ischemic cardiomyopathy is the single most common cause of heart failure. More than 64.3 million people across the world and 6 million people in the United States currently experience HF [3, 6]. In addition to increase in human toll, the estimated cost of HF exceeds $60 billion each year [7, 8].

Advertisement

2. Pathophysiology of ischemic cardiomyopathy

In patients with coronary artery disease, rupture of atherosclerotic plaque followed by in situ thrombus formation leads to sudden cessation of coronary blood flow. If the coronary blood flow is not established early enough either by spontaneous, pharmacological, or interventional recanalization, the death of ischemic myocytes ensues. With time, dead myocytes are replaced with fibrous tissue. Once the amount of scarred myocardium is significant enough after single or multiple episodes of MI, the left ventricle remodels with dilatation, regional deformation, and decrease in overall contractility. Remodeling and alteration of LV geometry especially the inferior wall may also lead to papillary muscle malalignment and mitral regurgitation (MR). Left ventricle volume overloading due to chronic MR in association with poor left ventricular contractility sets up a vicious cycle of worsening LV remodeling and MR [9].

The replacement of the dead myocardium with fibrous tissue is the most important mechanism in the development of ICM. Other pathophysiological processes such as myocardial stunning and hibernation that render the viable myocardial cells unable to perform their mechanical work and also contribute to the development of ICM. Both myocardial stunning and hibernation are reversible forms of myocardial contractile dysfunction that have the potential of mechanical work restoration if the blood flow supply can be improved [10]. In any given heart with ICM, all three stages of myocardium, i.e., normal, viable but hypocontractile and scarred myocardium often coexist within a single cross section of LV. Thus, ischemic cardiomyopathy is extremely heterogeneous and particularly challenging for accurate viability assessment with imaging studies [11].

The concept of hibernating myocardium is interesting as well as mysterious. Our present understanding about the hibernating myocardium is limited [12, 13, 14, 15, 16]. Rahimtoola [17] described the hibernating myocardium as “resting left ventricular dysfunction due to reduced coronary blood flow that can be partially or completely reversed by myocardial revascularization and/or by reducing myocardial oxygen demand.” Hibernating myocardium is usually limited to subendocardial tissues. Histologically, in hibernating myocardium, there is loss of contractile proteins and sarcoplasmic reticulum without the change in the cell volume. Presumably, hibernation is a protective dedifferentiation of myocardial cells or switch to a quiescent state of decreased mechanical work in times of chronically decreased oxygen supply [13]. This adaptive mechanism probably allows the myocytes to avoid the ischemic imbalance and remain alive in the milieu of decreased coronary blood flow that would otherwise lead to cell death. Alternative mechanism for ventricular dysfunction in ICM may be myocardial stunning. Myocardial stunning apparently occurs due to repeated episodes of ischemic insult that result in viable but chronically hypocontractile myocardium (i.e., repetitive stunning). Due to extremely low ischemic threshold of the myocytes, any decrease in coronary blood flow during stress leads to ischemia and ischemia–reperfusion changes in the myocytes despite normal or insignificantly decreased resting coronary perfusion [13, 18]. This repetitive stunning of the myocytes results in chronic LV dysfunction. Thus, in patients with ICM, territories with high numbers of cardiomyocytes with excess glycogen reserve and less fibrosis in all probabilities are reversible after revascularization. These myocytes also demonstrate higher blood flow and glucose uptake on positron emission tomography (PET) scan [19].

Advertisement

3. Preoperative considerations

Patients with ICM present with myriad of signs and symptoms depending upon the severity of heart failure and degree of physiological compensation. Some patients may be asymptomatic or minimally symptomatic with mild anginal chest pain and dyspnea on exertion while other patients may present with overt heart failure symptoms, e.g., dyspnea, orthopnea, poor exercise tolerance, and increased fatigability. Patients usually have a longstanding history of coronary artery disease and a prior history of myocardial infarctions. Physical examination can reveal bibasilar crackles, S3 gallop, displaced apical impulse, carotid bruits, jugular venous distension, positive hepato-jugular reflex, and bilateral lower extremity edema.

3.1 Diagnostic testing

In patients with ICM, multivessel disease, low LVEF, and increased LV end-systolic volumes are important prognostic factors. Therefore, all these factors must be taken into consideration when making the difficult decision regarding revascularization. Suitability of the patient for CABG depends upon: A) suitability of the diseased coronary arteries for bypass grafting; B) the amount of viable myocardium present and whether the viable myocardium is present in the territory of CAD; C) severity of right and left heart failure; and D) associated cardiac lesions. All the diagnostic investigations should be directed toward determining whether the patient is a suitable candidate for CABG or not.

Transthoracic echocardiography (TTE): Transthoracic echocardiography is an essential investigation in assessing myocardial viability in a patient with ICM. Echocardiography is useful in evaluating cardiac anatomy, valvular function, ventricular systolic/diastolic function, cardiac wall motion, and pericardial pathology. All this information is useful in diagnosing ischemic cardiomyopathy, especially in patients with HF and other high-risk features.

Coronary angiography: Coronary angiography allows direct visualization of the coronary arteries for assessment of severity of obstruction, collateralization, and the blood flow to the myocardium. Coronary angiography is most important in defining the extent and severity of coronary artery disease and whether the coronaries arteries are suitable for grafting. Computed tomography coronary angiography can also be performed in place of conventional coronary angiography to assess coronary arteries in patients with low to intermediate risk of CAD [20].

Cardiac stress test: There are different stress tests available depending on the patient’s health, functional status, baseline heart rhythm, and exercise tolerance. The goal of these stress tests is to assess for cardiac ischemia and myocardial viability. Late gadolinium enhancement cardiac magnetic resonance (LGE-CMR), dobutamine stress echocardiography, single-photon emission computed tomography (SPECT), and F-18- fluorodeoxyglucose positron emission tomography (FDG-PET) imaging can be used to assess myocardial viability [21]. Dobutamine stress echocardiography is widely used to assess myocardial contractility reserve and viability. With continuous dobutamine infusion, initially myocardial perfusion increases along with increased contractility. However, as the dobutamine dose increases, blood flow cannot be escalated further leading to reduced myocardial contractility. This phenomenon known as biphasic reaction can predict the recovery of the myocardial function after revascularization.

Late gadolinium enhancement cardiovascular magnetic resonance (LGE-CMR) can detect increase in extracellular space due to myocardial apoptosis and necrosis and can predict the reversibility of the myocardial contractility after successful revascularization while dobutamine stress CMR can detect the ischemic myocardium. In patients with ICM with transmural infarct, minimal LGE (<25%) in dysfunctional myocardial segment indicates a high likelihood of recovery while the chance of recovery is minimal in segments with >50% LGE.13 In segments with 25–50% LGE involvement, the recovery prediction is not consistent [22].

Single-photon emission computed tomography (SPECT) and positron emission tomography (PET) had been widely utilized in the past to assess myocardial viability. Thallium-based SPECT scan demonstrate delayed distribution but has increased risk of ionizing radiations while technetium-based SPECT has less risk of radiation, but it cannot demonstrate a delayed distribution. Another nuclear imaging modality to assess myocardial viability is cardiac PET. PET imaging is based on the principle that in an ICM, ischemic myocardium switches to glucose-based metabolism instead of fatty acids. 18F-fluorodeoxyglucose (18F-FDG) can detect this shift in viable but ischemic myocardium. PET has higher spatial resolution, lower risk of radiation, and better attenuation correction compared with SPECT. PET, however, cannot distinguish between normal and ischemic or hibernating myocardium in patients with insulin resistance, and results may be inaccurate in patients with variable uptake of FDG due to heart failure [23].

Brain natriuretic peptide (BNP) test: BNP is synthesized in the ventricles, and it is secreted when the myocardial muscle has a high wall tension. BNP is an important biomarker for heart failure patients. Increasing trend in BNP suggests worsening of heart failure; however, it cannot detect myocardial ischemia.

Advertisement

4. Clinical studies and randomized trials in patients with ischemic cardiomyopathy

Coronary artery bypass grafting for CAD started in the mid-1960s. Since then, numerous clinical trials and studies have tried to address different questions related to the management of CAD. All these trials and studies have established an undisputed role of surgical revascularization in patients with CAD in terms of improved survival, risk of reintervention, and quality of life [24, 25, 26, 27]. Nevertheless, prior to Surgical Treatment for Ischemic Heart Failure (STICH) trial [28], none of the studies specifically addressed the management of patients with ICM. The coronary artery surgery study (CASS) trial registry that followed the patients who were excluded from the main study reported that patients with LVEF <35% had better survival with CABG than with medical therapy, if they had associated three-vessel disease and if the presenting symptom was angina [29]. Similarly, a 25-year observational study involving 1391 patients (medical therapy (n = 1052) or CABG (n = 339)) from Duke Cardiovascular Disease Databank also reported an improved survival with CABG over medical therapy alone after 30 days to more than 10 years in patients with NYHA class ≥II, CAD with at least one vessel stenosis ≥75%, and LVEF <40%. The benefit with CABG was observed irrespective of the extent of coronary artery involvement (P < 0.001) [30].

These observational studies pointed toward the role of CABG in patients with ischemic cardiomyopathy; however, lack of randomized clinical studies in patients with ICM led to different therapeutic approaches driven by the physician bias regarding the potential benefit of myocardial revascularization [9]. The resulting equipoise formed the basis for the multiinstitutional STICH randomized controlled clinical trial [28]. STICH trial was the first and only large-scale randomized clinical trial to compare surgical revascularization with medical therapy in patients with LVEF ≤35% and CAD amenable to CABG. The STICH trial randomly assigned 1212 patients to three groups (medical therapy alone, medical therapy with CABG, and medical therapy with CABG and SVR). To evaluate the superiority of either procedure, two hypotheses were developed. In Hypotheses 1, the investigators evaluated medical therapy against medical therapy with CABG. All patients underwent coronary angiography to define the extent of CAD; patients with critical left main disease or unstable coronary syndromes were excluded from the trial. The primary outcome of the study was all-cause mortality, and secondary outcomes were cardiovascular mortality, combination of all-cause mortality and hospitalization for cardiac causes. At a median follow-up of 56 months, medical therapy plus CABG surgery resulted in a nonsignificant trend toward improvement in the primary outcome (36% vs. 41% with medical therapy alone) as well as significantly lower cardiovascular mortality and improved quality of life (at 4, 12, 24, and 36 months as assessed by the Kansas City Cardiomyopathy Questionnaire) [31]. However, this trial was fraught with certain limitations. First, during the study period, 9% patients in medical therapy plus CABG group crossed over to medical therapy group only while 17% patients in medical therapy alone group crossed over to medical therapy and CABG group. This crossover may have led to a diminished treatment benefit, thereby preventing the primary outcome from reaching statistical significance. Second, the STICH trial was designed to maximize both medical and surgical outcomes using strict criteria for surgical expertise (e.g., documented surgical expertise by volume and outcome criteria) and regular review of both surgical center conduct and intensity of medical therapy. Clinical equipoise had to be present, and both the surgeon and cardiologist had to believe revascularization was technically feasible. Both these issues may limit the generalizability of the trial to routine clinical practice.

In 2016, results of extended follow-up of STICH trial patients, i.e., the STICH Extension Study (STICHES), were published extending the median follow-up to 9.8 years [32]. After 9.8 years, the primary outcome (all-cause mortality) was significantly lower in the medical therapy and CABG group compared with medical therapy alone group (59% vs. 66%; hazard ratio [HR] 0.84; 95% CI, 0.73–0.97). Medical therapy and CABG group also experienced significant reductions in cardiovascular mortality (40.5% vs. 49.3%; HR 0.79; 95% CI, 0.66–0.93) and the combination of all-cause mortality and cardiovascular hospitalization (76.6% vs. 87%; HR 0.72; 95% CI, 0.64–0.82). Another large population based observational study related to CAD with LV systolic dysfunction was reported [33], it is recommended to do CABG and medical therapy for patients with ICM who have coronaries amenable to surgical revascularization.

Advertisement

5. Myocardial viability and treatment decisions

Observational studies done in early 2000s focused on the potential benefit of viable myocardium on the patient survival and LV function after the revascularization. Initial potential survival benefit from revascularization in patients with ICM and viable myocardium was reported in a meta-analysis published in 2002. This meta-analysis included 24 nonrandomized viability studies involving 3088 patients with CAD and LV dysfunction who had a mean LVEF of 32% [34]. Patients with myocardial viability had 80% reduction in annual mortality with revascularization (3.2% vs. 16% with medical therapy alone), while there was no significant change in annual mortality with revascularization in patients without myocardial viability (7.7% vs. 6.2% with medical therapy alone). Potential effect of viable myocardium on LVEF was also illustrated in a review published in 2004 that involved 29 observational studies including 758 patients [35]. In this review, LVEF increased after revascularization when myocardial viability was present (37–45%) but did not change significantly in the absence of viability. Further, studies have also demonstrated that 25–30% of the dysfunctional myocardium needs to be viable to result in improvement of LVEF. On the contrary, in a substudy of the STICH trial, 601 of the 1212 patients were evaluated for myocardial viability, and outcomes were analyzed according to those assigned to receive medical therapy plus CABG or medical therapy alone. Study showed minimal improvement in LVEF with revascularization (from 28% pre-CABG to 30% post-CABG). Following adjustment for differences in baseline variables and with follow-up extending beyond 10 years, there was no significant improvement in mortality with medical therapy plus CABG compared with medical therapy alone. Myocardial viability was associated with reduced mortality but did not predict a benefit from revascularization. This raises the question of whether viability assessment is needed prior to surgical revascularization. However, myocardial viability in STICH trial was assessed using stress echocardiography and SPECT radionuclide myocardial perfusion imaging; more contemporary techniques such as CMR and positron emission tomography (PET) were not studied and are an important limitation of the STICH findings [36]. Presence of myocardial viability does lead to improvement in contractility and myocardial thickness following revascularization subject to the presence of at least 25–30% of viable myocardium and scar burden <25% (as detected by LGE-CMR) [37]. However, inconsistencies in the criteria and the methods used to diagnose myocardial viability between various studies have led to blurring of the evidence of benefit of revascularization.

In the absence of firm evidence, routine viability assessment prior to consideration for CABG in patients with ICM is not recommended. However, situations that require greater precision in defining large infarcts either due to associated excessive surgical morbidity (e.g., renal failure) or risk of suboptimal outcome (e.g., evidence of LV remodeling, inability to achieve complete revascularization); viability assessment with more contemporary techniques such as LGE-CMR or FDG-PET may help further refine the potential risks and benefits.

Advertisement

6. Impact of left ventricular size and remodeling

Left ventricular size is an important determinant of outcome after surgical revascularization in patients with ICM. However, our present understanding of impact of preoperative LV size on postoperative LV function and survival is still limited. The impact of left ventricular enlargement on the improvement in LV function after revascularization was illustrated in a review of 61 patients with ischemic heart disease and a mean LVEF of 28%, all of whom had an evidence of substantial myocardial viability [38]. One-third of the patients had no significant improvement in the LVEF (≥5%). The study showed that the patients with a significant improvement in LVEF after CABG had a significantly smaller left ventricular end-systolic volume (LVESV) on preoperative echocardiography than those without improvement (121 mL vs. 153 mL). The observational data are in contrast with the findings from the STICH trial, which found greater benefit with respect to mortality in patients with greater baseline remodeling (e.g., larger left ventricle end-systolic volume index [LVESVI]) [28].

Advertisement

7. Percutaneous coronary intervention versus surgical revascularization

Percutaneous coronary intervention (PCI) is an established treatment for revascularization in acute myocardial infarction. Role of PCI in management of ICM is still unclear due to the lack of well-designed randomized studies. In the lack of randomized controlled study, best available data come from the observational study comparing PCI with CABG in 4616 patients with LVEF ≤35% who were enrolled in New York State registries (1351 underwent PCI with drug eluting stents and 3265 underwent CABG), from which 2126 patients were chosen for evaluation based on propensity score matching [39]. At a median follow-up of 2.9 years, there was no significant difference in mortality between contemporary PCI and CABG (HR 1.01; 95% CI 0.81–1.28). PCI was associated with a greater risk of myocardial infarction (HR 2.16; 95% CI 1.42–3.28) and need for repeat revascularization (HR 2.54; 95% CI 1.88–3.44), but a significantly lower risk of stroke compared with CABG (HR 0.57; 95% CI 0.33–0.97).

In a separate post hoc analysis of AWESOME trial, in which 454 patients who had medically refractory unstable or provocable ischemia were randomized to PCI or CABG. Ninety-four patients had LVEF <35% (mean 25%) [40]. Among patients with LVEF <35%, there was no difference in mortality between CABG and PCI. However, limitation of this trial was that all patients included in the study had angina and acute coronary syndromes and not heart failure.

Advertisement

8. Role of CABG in patients with ischemic cardiomyopathy

The mechanism of survival advantage conferred by CABG in patients with heart failure irrespective of myocardial viability still remains speculative, although, post hoc analysis of STICH trial has been able to shed some interesting insight on this topic. In STICH trial, a subanalysis evaluating cause-specific cardiac mortality in patients with ICM demonstrated that sudden cardiac death (SCD) was the most frequent mode of death and outnumbered pump failure deaths by approximately twofold [41]. Further, both SCD and death from HF were significantly reduced after the CABG (as was death from myocardial infarction). Predictors of increased risk of SCD in this analysis were increased LVESVI and elevated BNP level. Interestingly, same variables along with regional myocardial sympathetic denervation were found to be significant risk factors for SCD in patients with ICM in the Prediction of Arrhythmic Events with Positron Emission Tomography (PAREPET) Study [42, 43]. Thus, the survival benefit of CABG in patients with ICM is largely due to the significant effect of revascularization on reducing the death due to arrhythmia with a smaller contribution from reducing the deaths from pump failure and fatal MI.

Advertisement

9. Our approach to patients with ischemic cardiomyopathy

We suggest the combined CABG and medical therapy instead of medical therapy alone for patients with ICM and CAD that is amenable to surgical revascularization. This suggestion is based primarily on a 7% absolute reduction in overall mortality over 10 years (STICH trial) and superior relief of anginal symptoms following CABG. However, as significant morbidity and early mortality (compared with medical management alone) are associated with CABG in patients with ICM, patients may also reasonably choose medical therapy alone as the initial treatment option. Following initiation of medical therapy, patients should be reevaluated on an ongoing basis for any changes in clinical status or symptoms and consideration for surgical revascularization should be discussed with the patient.

Other clinical features that should be considered while tailoring the decision for any given patient are greater functional capacity (6-minute walk >300 m), greater burden of CAD (e.g., three-vessel disease), coexistent moderate to severe mitral regurgitation (MR), lower ejection fraction (e.g., LVEF <35%), and greater remodeling (e.g., LVESVI >79 mL/m2) (associated with improved outcomes in STICH trial).

Additionally, we do not recommend routine viability assessment prior to consideration for surgical revascularization and consideration should be case-to-case basis especially in patients in whom the risk-to-benefit profile is not as clear (e.g., patients with significantly elevated surgical risk). We believe that viability study may not aid in decision-making; however, the presence of significant viability and < 25–30% scar on LGE-CMR gives reassurance to the surgeon for improved surgical outcome.

Considering the advantage with CABG from the STICHES trial, it seems that patients with suitable targets for revascularization in the setting of an EF < 35% with two or three vessel CAD should be considered for CABG irrespective of the results of viability testing. However, competing risk factors such as severity of heart failure, age of the patient, and risks for noncardiac mortality need to be carefully weighed in considering the recommendation for revascularization and decision should be made on individual basis.

Advertisement

10. Preoperative optimization and perioperative temporary mechanical support

Factors that have been consistently associated with adverse outcomes after CABG for patients with ICM include preoperative renal dysfunction, advanced HF, recent myocardial infarction, and hemodynamic instability. Perioperative shock in this patient population more than doubles the rate of perioperative mortality [44, 45, 46]. Therefore, preoperative optimization of the patient status can improve the patient outcome after the surgery. The specific mode of optimization should be individualized to patients’ needs and driven by their response to initial therapy. If medical therapy alone is ineffective, more invasive measures should be considered. In the preoperative setting, prophylactic intra-aortic balloon pump (IABP) decreases afterload, increases coronary artery perfusion, provides a modest increase in cardiac output [47, 48]. In a variety of analyses, IABP therapy before the operation has been noted to result not only in improved patient condition before CABG, but also in reduced perioperative morbidity and mortality. Two meta-analyses of randomized clinical trials examining the utility of preoperative IABP therapy in patients with ICM demonstrated a strong association between preoperative use of IABP and reduced hospital mortality, lower incidence of low cardiac output syndrome, and shorter duration of ICU stay. Patients with high-risk profile including low LVEF, left main disease >70%, prior heart surgery, poor coronary artery targets, and unstable angina typically benefit from preoperative IABP [47, 48, 49, 50].

In patients who present with cardiogenic shock resulting from acute myocardial infarction or decompensated HF with end-organ dysfunction, IABP may be inadequate for stabilization or preoperative optimization. In these patients, transvalvular devices such as microaxial surgical heart pump can be used. These devices reduce left ventricular end-diastolic pressure (LVEDP) and volume workload and provide the circulatory support necessary to allow native heart recovery. In a recent analysis, the use of these micro-axial pump was associated with reduced mortality, without significant increase in device-related stroke, hemolysis, or limb ischemia [51, 52]. Finally, in patients with cardiogenic shock that is refractory to inotropic support, IABP, and/or microaxial pumps, ventricular assist device (VAD) implantation should be considered [47, 53, 54].

Patients with ICM with cardiogenic shock, who have organ dysfunction at the time of presentation, temporary VAD can be used as bridge to decision. Patients who reverse their organ dysfunction and acidosis after the insertion of temporary MCS and demonstrate an adequate contractile reserve and response to inotropic stimulation can successfully bridge to CABG. This is contingent to good coronary targets and absence of unfavorable anatomic and physiologic profiles [27]. Otherwise, they should be evaluated for heart transplant and should be considered for more durable VAD option as bridge to transplant.

11. Coronary artery bypass graft surgery strategy

11.1 On-pump arrested-heart CABG

The goal of CABG in patients with ICM is to achieve expeditious and complete revascularization. On-pump arrested-heart CABG is the most commonly used strategy that allows a bloodless and still field that facilitates complete revascularization [55]. Excellent myocardial protection especially right ventricle is paramount in the setting of ischemic cardiomyopathy as myocardial ischemia and injury are poorly tolerated when myocardial reserve is limited [56].

In patients undergoing on-pump CABG, controversy still remains about type of cardioplegic solution, temperature, and route of administration that provides the optimal myocardial protection. This becomes critical in patients with ICM as any amount of further myocardial damage may be deleterious. In a meta-analysis of 12 studies including 2866 patients, lower prevalence of perioperative myocardial infarction was found in patients who received blood cardioplegia [57]. Another meta-analysis of 41 randomized clinical trials (RCT) found that warm cardioplegia did not improve clinical outcomes but was associated with a mild reduction of cardiac enzyme release [58]. Single-dose cardioplegia benefit is limited to a reduction in ischemia and bypass time and does not translate into a major morbidity or mortality advantage [59]. There is no systematic comparison of different routes of cardioplegia administration (i.e., antegrade vs. retrograde vs. combined); however, isolated retrograde cardioplegia should be avoided due to its heterogeneous perfusion and unpredictable right ventricle myocardial protection [60]. On the other hand, retrograde cardioplegia may be useful in adjunct to antegrade cardioplegia in patients with severe CAD and in redo CABG to reach territories not otherwise reachable by antegrade delivery and to flush potential embolic debris from inadvertently manipulated diseased vein grafts [61, 62]. Although data are scarce, it has been reported that antegrade cardioplegia supplemented with venous graft perfusion can significantly improve myocardial protection. The most suitable myocardial protection strategy may be a combination of antegrade, retrograde, and delivery down the vein grafts.

11.2 Off-pump CABG

Utilization of off-pump CABG (OPCABG) is limited to few centers and selected patients in the developed countries. There have been no large RCTs comparing on-pump CABG versus OPCABG and small RCTs that did compare these two modalities have reported inferior or non-superior long-term outcome with OPCABG. Most of these studies are limited by smaller sample size, short duration of follow-up, and limited experience of the operator. This is of particular relevance given that OPCABG may lead to inferior long-term outcomes if performed by inexperienced operators and/or accompanied by incomplete revascularization [63]. In a meta-analysis of 23 individual nonrandomized studies published in 2011 that involved 7759 CABG patients with LVEF <40%, 2822 patients underwent OPCABG. Overall early mortality was significantly reduced (odds ratio [OR], 0.64; 95% CI, 0.51–0.81) in OPCABG group. Similar results were observed on subgroup analysis of 1915 patients with LVEF <30% (OR 0.61; 95% CI 0.47–0.80) [64]. A recent meta-analysis published in 2020 comprising 16 studies with 32,354 patients with LV dysfunction (defined as LVEF <40%) also reported a significant reduction in 30-day mortality (OR 0.84; 95% CI 0.73–0.97), perioperative complications, and transfusion requirements with OPCABG [65]. In a report published in 2016 from the Japan Adult Cardiovascular Surgery Database including 918 pairs of propensity-matched CABG patients with LVEF <30%, there was reduced perioperative and 30-day mortality with OPCABG (1.7% vs. 3.7%; P < 0.01) and reduced incidence of mediastinitis, reoperation for bleeding, and need for prolonged ventilation, but there was no difference in incidence of stroke or renal failure compared to on-pump CABG [66].

11.3 On-pump beating-heart CABG

On-pump beating-heart CABG has been proposed as an alternative strategy to on-pump cardioplegic arrest CABG, particularly in higher-risk patients including patients with impaired LV function [67]. This technique is more of historical significance as it is rarely used nowadays. In a review of 11 studies, comprising two RCTs and nine observational studies comparing on-pump beating-heart CABG and on-pump arrested heart CABG, lower mortality was reported with on-pump beating-heart CABG in five of the nine observational studies while mortality was similar with both techniques in two RCTs. However, due to the lack of randomization and the absence of propensity matching, the possibility of selection bias accounting for the difference in mortality cannot be discounted. Intraoperative myocardial injury with on-pump beating heart may increase due to inadequate coronary perfusion distal to areas of stenosis [68].

In the absence of more definitive evidence about the superiority of one technique of CABG over the other, the operative strategy should be tailored based on patient factors such as extent of CAD and associated comorbidities, surgeon’s expertise and comfort level of the cardiac anesthetist, and center experience. When off-pump technique is used, maintenance of appropriate perfusion pressure and when on-pump CABG is utilized, appropriate myocardial protection is imperative to minimize further myocardial injury.

12. Bypass conduits

Presently, use of left internal mammary artery (LIMA) for bypassing left anterior descending coronary artery and reverse saphenous vein grafts for bypassing rest of the coronary arteries is the standard of care across the globe. Evidence from the recent studies has shown the superiority of multi-arterial grafting in improving long-term patient survival after CABG. The impact on survival becomes even more significant with increasing duration of follow-up [69, 70, 71]. The evidence of beneficial effects of multi-arterial grafting in patients with ICM, however, is limited to few studies and a small number of patients [72, 73, 74]. Further, multi-arterial grafting in patients with ICM still remains controversial as the overriding priority in these patients is to mitigate the upfront risk of surgery and avoidance of perioperative myocardial ischemia. In a risk predictive model based on STS database review of patients operated for CABG, the HR for perioperative mortality after isolated CABG was 1.19 (95% CI, 1.17–1.22) for every 10% reduction in LVEF [75], and operative risk was further compounded with the addition of noncardiac organ dysfunction and other comorbidities.

There are four reasons why caution should be used when contemplating multi-arterial grafting in patients with ICM [56]. First, perioperative administration of high doses of vasopressors may be necessary in these patients, and this is an important predisposing factor for the development of spasm in the arterial grafts [76]. Radial and gastroepiploic arteries are particularly vulnerable to spasm compared with IMAs. Second, adequacy of blood flow in a fresh arterial graft may not be as robust as in a vein graft, with the potential for clinically significant perioperative coronary artery hypoperfusion [77, 78, 79]. Third, multi-arterial grafting usually adds to the complexity and length of the operation and prolongs myocardial ischemic time. This may not be well tolerated by the patients with ICM. Fourth, arterial grafts may not be of adequate length in massively dilated hearts, especially if sequential anastomoses are contemplated. A patient-level combined analysis of six RCTs associated radial artery grafts in addition to LIMA with improved clinical outcomes compared with venous grafts [80]. The benefit of radial artery grafting was persistent even on subgroup analysis of patients with severe LV dysfunction (LVEF <35%). However, the number of patients in subgroup were limited (25 (4.7%) and 32 (6.4%) in the radial artery and saphenous vein groups, respectively). The results of other observational studies have yielded mixed results with the use of multi-arterial grafting in patients with ICM [73, 81, 82, 83, 84]. The probable reason is variable cutoff for LVEF with different studies (lowest limit <30%), which adds to uncertainty regarding multi-arterial grafting benefits [85]. Observational evidence also suggests that the benefit of multi-arterial grafting is lost in patients with ICM with limited life expectancy or severe associated comorbidities [83, 86, 87, 88].

We believe that multi-arterial grafting should not be routinely recommended for patients with ICM. Patient selection for multi-arterial grafting should be based on patient factors and surgeon’s experience and comfort. Young patients with compensated HF having good target for bypass may be considered for multi-arterial grafting if the risk–benefit ratio is favorable and prolonged survival is anticipated after revascularization.

13. CABG combined with other procedures

13.1 Atrial fibrillation

Atrial fibrillation (AFib) is present in 5–10% of patients undergoing CABG. It is associated with increased risk of complications including stroke and renal failure, prolonged hospital stay as well as increased mortality despite adjustment for potential confounders [89]. Therefore, current North American and European guidelines for CABG recommend concomitant AFib ablation procedure in symptomatic patients or asymptomatic patients having low operative risk [90, 91]. The evidence supporting the surgical ablation of AFib in patients with ICM undergoing CABG is minimal and limited by selection bias [92]. Theoretically, patients with a reduced ejection fraction would benefit from the restoration of sinus rhythm and atrial contraction [93]. However, concomitant AFib ablation procedure adds to the technical complexity of the surgery and prolongs the duration of aortic cross clamp and cardiopulmonary bypass. Despite this, some studies reported that surgical AFib ablation is safe and effective in patients with heart failure [94, 95].

13.2 Mitral valve surgery

Up to 10% patients develop chronic moderate or severe MR following acute myocardial infarction. Chronic ischemic mitral regurgitation (CIMR) is associated with an increased incidence of heart failure and increased risk of mortality in patients with LV dysfunction [96]. Furthermore, LV dysfunction can lead to gradual dilatation and geometric change in the left ventricle that results in distortion of the mitral valve and worsening of MR. Although, there is a general consensus to repair or replace the mitral valve in patients with severe CIMR undergoing CABG, the management of moderate (Grade II) mitral regurgitation still remains controversial.

In the Cardiothoracic Surgical Trials Network study, adding surgical mitral valve repair to CABG in patients with moderate CIMR had no significant effect on survival or LV reverse remodeling at 2 years follow-up but was associated with increased duration of hospital stay and morbidity including neurological events and atrial arrhythmias [97]. Smaller RCTs have shown benefit in surrogate outcomes for CABG and mitral valve repair versus CABG alone in patients with moderate CIMR [98, 99]. However, none of the trials has specifically focused on patients with ICM. In patients with severe CIMR, mitral valve replacement has been shown to provide more reliable and durable relief of MR than repair, but without survival benefit [100]. Mitral valve replacement rather than repair is also favored in patients with LV basal aneurysm/dyskinesis or other potential risk factors for recurrent MR after repair, e.g., significant leaflet tethering and/or severe left ventricular dilatation (LV end-diastolic dimension >6.5 cm). Preserving the subvalvular apparatus is also strongly recommended when replacing mitral valve in these patients. Concerns about persistent tethering of the posterior leaflet and recurrent MR after CABG in patients with prior inferior wall MI have prompted some to combine mitral anuloplasty with a subvalvular procedure such as papillary muscle approximation and papillary muscle relocation. All these procedures result in improved echocardiographic and cardiovascular outcomes but fail to influence all-cause mortality or quality of life [101, 102, 103]. Therefore, this remains an area for further study and evaluation.

13.3 Tricuspid valve surgery

Tricuspid regurgitation (TR) is an established risk factor in patients undergoing CABG [104]. In patients with CIMR, although progression of unrepaired mild to moderate TR after revascularization is uncommon, presence and progression of moderate or greater TR are associated with increased incidence of clinical events [105]. The underlying etiology of TR in ICM includes tricuspid annular dilatation and leaflet tethering in the setting of RV remodeling due to right ventricle infarction with or without pulmonary hypertension, tricuspid annular dilatation associated with AFib, and iatrogenic or lead related injury to tricuspid leaflets. Current AHA/ACC guidelines assign class I recommendation for tricuspid valve repair at the time of left sided valve surgery for severe TR and class IIa for less than severe TR in the presence of annular dilatation (>4.0 cm) or right-sided HF [106].

Concomitant mitral valve repair can be considered in patients with ICM undergoing CABG in the presence of atrial arrhythmias, left atrial dilation, or in the setting of severe LV dilation. Replacement, rather than repair, should be considered in patients with limited viability in the posterolateral wall of the LV [97]. Tricuspid valve repair should be considered at the time of left sided valve surgery for severe TR and less than severe TR in the presence of annular dilatation (>4.0 cm), right-sided HF or iatrogenic, or lead-related injury to tricuspid leaflets. Severe TR in the presence of significant RV dysfunction is a marker of poor outcome after coronary revascularization and warrants evaluation and consideration for advanced HF therapies.

13.4 Surgical ventricular restoration

In patients with ICM, gradual dilatation of LV results in transition from elliptical to a more spherical geometry. This impairs the structure–function relationship of the left ventricle [107]. The concept of surgical ventricular restoration (SVR) procedure for the patients with ICM is more than four decades old; however, the procedure is yet to gain acceptance as not only the procedure is technically challenging but also, no study so far has been able to show consistent benefit with concomitant SVR. Doctrine of SVR operation assumes that resection of scarred myocardium, reducing the ventricular size, and restoring an anatomically elliptical shape can improve the left ventricular function [108]. However, studies so far have not been able to prove this assumption. A randomized study including 137 patients with LVEF <50% and LV end systolic volume index (LVESVI) >80 ml/m2 showed that CABG alone was inferior to CABG with SVR in terms of improvement in LVEF, MR, and NYHA class. However, study was limited to only 2 years of follow-up [109]. Similarly, Prucz et al. reported this result [110]. Both these studies were limited by short duration of follow-up and failed to show any benefit of SVR procedure on survival. Consequently, the STICH trial was conducted to evaluate the long-term outcome of concomitant SVR procedure in patients with LV dysfunction, LV akinesis/dyskinesis, presence of scar, and LV dilatation [111]. To evaluate the benefit of SVR, patients enrolled in STICH trial in CABG arm were divided into two groups (medical therapy with CABG versus medical therapy with CABG and SVR). The study found no difference in mortality between the groups at median follow-up of 48 months (hazard ratio 1.00, 95% CI 0.79–1.26, P = 0.98) [111]. Results of these studies led to abandonment of the SVR procedure by majority surgeons [112].

It still remains uncertain which patients should receive SVR as part of CABG operation and what is its impact on long-term survival and functional outcome [112, 113, 114]. Therefore, consideration for SVR should still be given to patients with true large ventricular aneurysms who present with medically refractory heart failure or ventricular arrhythmias.

14. Postcardiotomy shock and temporary MCS

Patients with ICM undergoing CABG are at increased risk of postcardiotomy shock and the risk increases further in patients with ischemic MR and/or right ventricular infarct. Patients with postcardiotomy shock who are unable to separate from cardiopulmonary bypass or require high-dose inotropic therapy, MCS should be considered [115].

14.1 Intra-aortic balloon pump (IABP)

Intra-aortic balloon pump has been considered as first line therapy for PCS as it is safe, widely available, and easy to place. Intra-aortic balloon pump improves the coronary perfusion, decreases the left ventricular afterload, and improves the cardiac output by 0.5–1 L/min. However, the hemodynamic support provided by an IABP is usually insufficient in reversing cardiogenic shock [116, 117]. In a recent analysis of 4550 patients operated for CABG between 2004 and 2008, 5% patients required an intraoperative or postoperative IABP, with overall mortality of 37%. IABP was equally effective in patients with predominantly right-sided failure with 50% increase in cardiac index and associated mortality of 31%. This study specifically addressed the issue of IABP effectiveness in both right- and left-sided failure [118].

14.2 Impella

Impella is a percutaneous or surgically implanted axial-flow device that is used for all types of cardiogenic shock. Impella devices significantly reduce LV end-diastolic pressure and volume, reduce myocardial oxygen demand, and support the systemic perfusion while allowing the heart to recover. Engstrom and colleagues [119] reported their experience with Impella 5.0 for treating 46 postcardiotomy shock patients mostly after CABG at three European centers. Half of the patients received an IABP before the Impella placement. Overall survival was 40% at 30 days. More recently, David and colleagues [120] reported on use of the Impella 5.0/Impella LD in 29 patients (40% with isolated CABG) treated for PCS between 2010 and 2015. Mortality was nearly 40%, similar to the aforementioned study. The best results for PCS treatment were reported by Griffith and colleagues [121] in the RECOVER I study, wherein an Impella 5.0 was placed in 16 patients having difficulty weaning from cardiopulmonary bypass. Fifteen patients were successfully supported, with 30-day survival of 94%. Results of this study should however be interpreted carefully as all the patients in the study were on low level of inotropic support before the Impella placement as opposed to the study protocol requirement of high inotropic support prior to Impella placement.

14.3 Extracorporeal membrane oxygenation

Veno-arterial extracorporeal membrane oxygenation (VA-ECMO) is second most commonly used device after IABP for postcardiotomy shock. Veno-arterial ECMO significantly unloads the right ventricle, improves the coronary perfusion, and supports the systemic perfusion while allowing the right heart to recover. However, VA-ECMO significantly increases the left ventricular afterload. Therefore, in patients supported with VA-ECMO, it is imperative to maintain left ventricular ejection either spontaneous or with inotropes. Otherwise, left side of the heart should be vented either by atrial septostomy, left atrial/left ventricular vent, or Impella [122]. There are no RCTs regarding the effectiveness of VA-ECMO in PCS, but several retrospective studies have shown 60–70% mortality in patients with PCS despite use of VA-ECMO [122, 123, 124, 125]. In a recent report of the European registry of 781 patients receiving VA-ECMO for PCS, institution of VA-ECMO was associated with increased mortality (odds ratio 1.54; 95% CI, 1.09–2.18), reoperation for bleeding/tamponade (odds ratio, 1.96; 95% CI, 1.37–2.81), and blood transfusion of >9 units (odds ratio, 2.42; 95% CI, 1.59–3.67). The authors also did a systematic review of 2491 patients with PCS who received VA-ECMO and reported 66.6% pooled prevalence of in-hospital/30-day mortality (95% CI, 64.7–68.4%), and lower in-hospital/30-day mortality in patients with peripheral ECMO (risk ratio, 0.92; 95% CI, 0.87–0.98). Switching the patients from central to peripheral cannulation appeared to provide close to a 10% mortality benefit [126]. Finally, studies evaluating the role of LV unloading during VA-ECMO for cardiogenic shock have reported 10–20% mortality benefit with LV unloading with either Impella or IABP [127, 128].

15. Post discharge management

In patients with ICM, the importance of adhering to guideline-directed medical therapy (GDMT), secondary prevention, and cardiac rehabilitation after revascularization cannot be overemphasized [129, 130]. Close follow-up of these patients is recommended for the titration of heart failure medications and continued assessment for needed additional interventions, including device implantation (e.g., automated implantable cardioverter-defibrillator (AICD)/Cardiac resynchronization therapy device (CRT) or advanced surgical therapies for persistent HF. In patients with ICM, initial 90 days after CABG are most vulnerable and associated with several-fold increase in HF-associated rehospitalization and mortality. Thus, these patients should undergo a close clinical monitoring after discharge. Initial post-discharge follow-up should be done at 7–14 days to review the volume status of the patient and titrate guideline-directed medications [131]. Although studies directly evaluating and comparing the impact of GDMT on ICM patients who have or have not undergone CABG are limited, conventional medical opinion supports that GDMT goals for post-CABG patients should not differ from those without CABG. Post hoc analysis has revealed that in patients with ICM, maintenance of optimal medical therapy after discharge is associated with best short-term and long-term outcomes [132].

16. Summary

Patients with ischemic cardiomyopathy and coronary artery disease that is amenable to surgical revascularization should undergo combination of surgical revascularization and medical therapy rather than medical therapy alone. This suggestion is based primarily on the long-term absolute reduction in mortality over the 10 years following CABG balanced against the early mortality risk of CABG. Routine assessment of viability to evaluate advisability of multivessel coronary revascularization to improve total mortality is not recommended. Based on the small but nontrivial early mortality risk associated with CABG surgery as well as other post-CABG morbidities, patients may also reasonably choose medical therapy as the initial treatment option. Revascularization remains an important treatment option for patients with ongoing anginal symptoms despite optimal medical therapy. For such patients, the relative efficacy of percutaneous coronary intervention (PCI) compared with CABG for revascularization is unknown. Nonrandomized registry suggests that there was no difference in mortality between CABG and PCI.

References

  1. 1. Maron BJ, Towbin JA, Thiene G, et al. Contemporary definitions and classification of the cardiomyopathies: An American Heart Association scientific statement from the council on clinical cardiology, heart failure and transplantation committee; quality of care and outcomes research and functional genomics and translational biology interdisciplinary working groups; and council on epidemiology and prevention. Circulation. 2006;113:1807
  2. 2. Elliott P, Andersson B, Arbustini E, et al. Classification of the cardiomyopathies: A position statement from the European Society of Cardiology working group on myocardial and pericardial diseases. European Heart Journal. 2008;29:270
  3. 3. Felker GM, Shaw LK, O’Connor CM. A standardized definition of ischemic cardiomyopathy for use in clinical research. Journal of the American College of Cardiology. 2002;39:210-218
  4. 4. Khan MA, Hashim MJ, Mustafa H, Baniyas MY, Al Suwaidi S, AlKatheeri R, et al. Global epidemiology of ischemic heart disease: Results from the global burden of disease study. Cureus. 2020;12:e9349
  5. 5. Virani SS, Alonso A, Aparicio HJ, Benjamin EJ, Bittencourt MS, Callaway CW, et al. Heart disease and stroke statistics–2021 update: A report from the American Heart Association. Circulation. 2021;143:e254-e743
  6. 6. Elgendy IY, Mahtta D, Pepine CJ. Medical therapy for heart failure caused by ischemic heart disease. Circulation Research. 2019;124:1520-1535
  7. 7. Heidenreich PA, Albert NM, Allen LA, Bluemke DA, Butler J, Fonarow GC, et al. Forecasting the impact of heart failure in the United States: A policy statement from the American Heart Association. Circulation. Heart Failure. 2013;6:606-619
  8. 8. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr, Drazner MH, et al. 2013 ACCF/AHA guideline for the management of heart failure: A report of the American College of Cardiology Foundation/American Heart Association task force on practice guidelines. Journal of the American College of Cardiology. 2013;62:e147-e239
  9. 9. Khera S, Panza JA. Surgical revascularization for ischemic cardiomyopathy in the post-STICH era. Cardiology in Review. 2015;23(4):153-160. DOI: 10.1097/CRD.0000000000000061 PMID: 25715329
  10. 10. Heusch G, Schulz R. Characterization of hibernating and stunned myocardium. European Heart Journal. 1997;18(suppl D):D102-D110
  11. 11. Armstrong WF. “Hibernating” myocardium: Asleep or part dead? Journal of the American College of Cardiology. 1996;28:530-535
  12. 12. Wijns W, Vatner SF, Camici PG. Hibernating myocardium. The New England Journal of Medicine. 1998;339:173-181
  13. 13. Schuster A, Morton G, Chiribiri A, et al. Imaging in the management of ischemic cardiomyopathy: Special focus on magnetic resonance. Journal of the American College of Cardiology. 2012;59:359-370
  14. 14. Rahimtoola SH. A perspective on the three large multicenter randomized clinical trials of coronary bypass surgery for chronic stable angina. Circulation. 1985;72(6 Pt 2):V123-V135
  15. 15. Braunwald E, Rutherford JD. Reversible ischemic left ventricular dysfunction: Evidence for the “hibernating myocardium”. Journal of the American College of Cardiology. 1986;8:1467-1470
  16. 16. Heusch G, Schulz R, Rahimtoola SH. Myocardial hibernation: A delicate balance. American Journal of Physiology. Heart and Circulatory Physiology. 2005;288:H984-H999
  17. 17. Rahimtoola SH. The hibernating myocardium. American Heart Journal. 1989;117:211-221
  18. 18. Vanoverschelde JL, Wijns W, Borgers M, et al. Chronic myocardial hibernation in humans. From bedside to bench. Circulation. 1997;95:1961-1971
  19. 19. Gerber BL, Vanoverschelde JL, Bol A, et al. Myocardial blood flow, glucose uptake, and recruitment of inotropic reserve in chronic left ventricular ischemic dysfunction. Implications for the pathophysiology of chronic myocardial hibernation. Circulation. 1996;94:651-659
  20. 20. Depré C, Vanoverschelde JL, Melin JA, et al. Structural and metabolic correlates of the reversibility of chronic left ventricular ischemic dysfunction in humans. The American Journal of Physiology. 1995;268(3 Pt 2):H1265-H1275
  21. 21. Im DJ, Youn JC, Lee HJ, Nam K, Suh YJ, Hong YJ, et al. Role of cardiac computed tomography for Etiology evaluation of newly diagnosed heart failure with reduced ejection fraction. Journal of Clinical Medicine. 2020;9(7):2270. doi: 10.3390/jcm9072270
  22. 22. Gatti M, Carisio A, D'Angelo T, Darvizeh F, Dell'Aversana S, Tore D, et al. Cardiovascular magnetic resonance in myocardial infarction with non-obstructive coronary arteries patients: A review. World Journal of Cardiology. 2020;12(6):248-261
  23. 23. Subramanyam P, Palaniswamy SS. Does myocardial viability detection improve using a novel combined 99mTc sestamibi infusion and low dose dobutamine infusion in high risk ischemic cardiomyopathy patients? Anatolian Journal of Cardiology. 2020;24(2):83-91
  24. 24. Garrett HE, Dennis EW, DeBakey ME. Aortocoronary bypass with saphenous vein graft. Seven-year follow-up. Journal of the American Medical Association. 1973;223:792-794
  25. 25. Sabiston DC Jr, The WF, Rienhoff J, lecture. The coronary circulation. The Johns Hopkins Medical Journal. 1974;134:314-329
  26. 26. Noon GP. Evolution of surgical treatment of coronary artery occlusive disease. Journal of the American Medical Association. 2009;301:970-971
  27. 27. Bakaeen FG, Gaudino M, Whitman G, Doenst T, Ruel M, Taggart DP, et al. 2021: The American Association for Thoracic Surgery expert consensus document: Coronary artery bypass grafting in patients with ischemic cardiomyopathy and heart failure. The Journal of Thoracic and Cardiovascular Surgery. 2021;162(3):829-850.e1. DOI: 10.1016/j.jtcvs.2021.04.052
  28. 28. Velazquez EJ, Lee KL, Deja MA, et al. Coronary-artery bypass surgery in patients with left ventricular dysfunction. The New England Journal of Medicine. 2011;364(17):1607-1616
  29. 29. Alderman EL, Fisher LD, Litwin P, et al. Results of coronary artery surgery in patients with poor left ventricular function (CASS). Circulation. 1983;68:785-795
  30. 30. O’Connor CM, Velazquez EJ, Gardner LH, et al. Comparison of coronary artery bypass grafting versus medical therapy on long-term outcome in patients with ischemic cardiomyopathy (a 25-year experience from the Duke cardiovascular disease databank). The American Journal of Cardiology. 2002;90:101-107
  31. 31. Mark DB, Knight JD, Velazquez EJ, Wasilewski J, Howlett JG, Smith PK, et al. Quality-of-life outcomes with coronary artery bypass graft surgery in ischemic left ventricular dysfunction: A randomized trial. Annals of Internal Medicine. 2014;161(6):392-399. DOI: 10.7326/M13-1380
  32. 32. Velazquez EJ, Lee KL, Jones RH, Al-Khalidi HR, Hill JA, Panza JA, et al. Coronary-artery bypass surgery in patients with ischemic cardiomyopathy. The New England Journal of Medicine. 2016;21(374):1511-1520
  33. 33. Sun LY, Gaudino M, Chen RJ, Bader Eddeen A, Ruel M. Long-term outcomes in patients with severely reduced left ventricular ejection fraction undergoing percutaneous coronary intervention vs coronary artery bypass grafting. JAMA Cardiology. 2020;5:631-641
  34. 34. Allman KC, Shaw LJ, Hachamovitch R, Udelson JE. Myocardial viability testing and impact of revascularization on prognosis in patients with coronary artery disease and left ventricular dysfunction: A meta-analysis. Journal of the American College of Cardiology. 2002;39(7):1151-1158. DOI: 10.1016/s0735-1097(02)01726-6 PMID: 11923039
  35. 35. Bax JJ, van der Wall EE, Harbinson M. Radionuclide techniques for the assessment of myocardial viability and hibernation. Heart. 2004;90(Suppl. 5):v26-v33
  36. 36. Panza JA, Ellis AM, Al-Khalidi HR, Holly TA, Berman DS, Oh JK, et al. Myocardial viability and long-term outcomes in ischemic cardiomyopathy. The New England Journal of Medicine. 2019;381(8):739-748
  37. 37. Shah DJ, Kim HW, James O, Parker M, Wu E, Bonow RO, et al. Prevalence of regional myocardial thinning and relationship with myocardial scarring in patients with coronary artery disease. Journal of the American Medical Association. 2013;309(9):909-918
  38. 38. Schinkel AF, Poldermans D, Rizzello V, Vanoverschelde JL, Elhendy A, Boersma E, et al. Why do patients with ischemic cardiomyopathy and a substantial amount of viable myocardium not always recover in function after revascularization? The Journal of Thoracic and Cardiovascular Surgery. 2004;127(2):385-390
  39. 39. Bangalore S, Guo Y, Samadashvili Z, Blecker S, Hannan EL. Revascularization in patients with multivessel coronary artery disease and severe left ventricular systolic dysfunction: Everolimus-eluting stents versus coronary artery bypass graft surgery. Circulation. 2016;133(22):2132-2140
  40. 40. Sedlis SP, Ramanathan KB, Morrison DA, Sethi G, Sacks J, Henderson W, et al. Outcome of percutaneous coronary intervention versus coronary bypass grafting for patients with low left ventricular ejection fractions, unstable angina pectoris, and risk factors for adverse outcomes with bypass (the AWESOME randomized trial and registry). The American Journal of Cardiology. 2004;94(1):118-120
  41. 41. Carson P, Wertheimer J, Miller A, O’Connor CM, Pina IL, Selzman C, et al. The STICH trial (surgical treatment for ischemic heart failure): Mode-of death results. JACC: Heart Failure. 2013;1:308-314
  42. 42. Fallavollita JA, Dare JD, Carter RL, Baldwa S, Canty JM Jr. Denervated myocardium is preferentially associated with sudden cardiac arrest in ischemic cardiomyopathy: A pilot competing risks analysis of cause-specific mortality. Circulation. Cardiovascular Imaging. 2017;10(8):e006446
  43. 43. Fallavollita JA, Heavey BM, Luisi AJ Jr, Michalek SM, Baldwa S, Mashtare TL Jr, et al. Regional myocardial sympathetic denervation predicts the risk of sudden cardiac arrest in ischemic cardiomyopathy. Journal of the American College of Cardiology. 2014;63:141-149
  44. 44. Vickneson K, Chan SP, Li Y, Bin Abdul Aziz MN, Luo HD, Kang GS, et al. Coronary artery bypass grafting in patients with low ejection fraction: What are the risk factors? The Journal of Cardiovascular Surgery. 2019;60:396-405
  45. 45. Kusu-Orkar TE, Kermali M, Oguamanam N, Bithas C, Harky A. Coronary artery bypass grafting: Factors affecting outcomes. Journal of Cardiac Surgery. 2020;35:3503-3511
  46. 46. O’Brien SM, Feng L, He X, Xian Y, Jacobs JP, Badhwar V, et al. The Society of Thoracic Surgeons 2018 adult cardiac surgery risk models: Part 2—Statistical methods and results. The Annals of Thoracic Surgery. 2018;105:1419-1428
  47. 47. Pichette M, Liszkowski M, Ducharme A. Preoperative optimization of the heart failure patient undergoing cardiac surgery. The Canadian Journal of Cardiology. 2017;33:72-79
  48. 48. Christenson JT, Schmuziger M, Simonet F. Effective surgical management of high-risk coronary patients using preoperative intra-aortic balloon counterpulsation therapy. Cardiovascular Surgery. 2001;9:383-390
  49. 49. Sa MP, Ferraz PE, Escobar RR, Martins WN, Nunes EO, Vasconcelos FP, et al. Prophylactic intra-aortic balloon pump in high-risk patients undergoing coronary artery bypass surgery: A meta-analysis of randomized controlled trials. Coronary Artery Disease. 2012;23:480-486
  50. 50. Pilarczyk K, Boening A, Jakob H, Langebartels G, Markewitz A, Haake N, et al. Preoperative intra-aortic counterpulsation in high-risk patients undergoing cardiac surgery: A meta-analysis of randomized controlled trialsdagger. European Journal of Cardio-Thoracic Surgery. 2016;49:5-17
  51. 51. Ramzy D, Soltesz E, Anderson M. New surgical circulatory support system outcomes. ASAIO Journal. 2020;66:746-752
  52. 52. Silvestri ERG, Pino JE, Donath E, et al. Impella to unload the left ventricle in patients undergoing venoarterial extracorporeal membrane oxygenation for cardiogenic shock: A systematic review and meta-analysis. Journal of Cardiac Surgery. 2020;35:1237-1242
  53. 53. Rihal CS, Naidu SS, Givertz MM, et al. 2015 SCAI/ACC/HFSA/STS clinical expert consensus statement on the use of percutaneous mechanical circulatory support devices in cardiovascular care: Endorsed by the American Heart Association, the Cardiological Society of India, and Sociedad Latino Americana de Cardiologia Intervencion; affirmation of value by the Canadian Association of Interventional Cardiology-Association Canadienne de Cardiologie d’intervention. Journal of the American College of Cardiology. 2015;65:e7-e26
  54. 54. Robinson NB, Audisio K, Bakaeen FG, Gaudino M. Coronary artery bypass grafting in low ejection fraction: State of the art. Current Opinion in Cardiology. 2021;36(6):740-747
  55. 55. Bakaeen FG, Shroyer AL, Gammie JS, Sabik JF, Cornwell LD, Coselli JS, et al. Trends in use of off-pump coronary artery bypass grafting: Results from the Society of Thoracic Surgeons adult cardiac surgery database. The Journal of Thoracic and Cardiovascular Surgery. 2014;148(856-63):864.e1 discussion 863-4
  56. 56. Bakaeen FG, Gaudino M, Whitman G, Doenst T, Ruel M, Taggart DP, et al. The American Association for Thoracic Surgery expert consensus document: Coronary artery bypass grafting in patients with ischemic cardiomyopathy and heart failure. The Journal of Thoracic and Cardiovascular Surgery. 2021, 2021;162(3):829-850.e1
  57. 57. Zeng J, He W, Qu Z, Tang Y, Zhou Q , Zhang B. Cold blood versus crystalloid cardioplegia for myocardial protection in adult cardiac surgery: A meta-analysis of randomized controlled studies. Journal of Cardiothoracic and Vascular Anesthesia. 2014;28:674-681
  58. 58. Fan Y, Zhang AM, Xiao YB, Weng YG, Hetzer R. Warm versus cold cardioplegia for heart surgery: A meta-analysis. European Journal of Cardio-Thoracic Surgery. 2010;37:912-919
  59. 59. Gambardella I, Gaudino MFL, Antoniou GA, Rahouma M, Worku B, Tranbaugh RF, et al. Single- versus multidose cardioplegia in adult cardiac surgery patients: A meta-analysis. The Journal of Thoracic and Cardiovascular Surgery. 2020;160:1195-202.e12
  60. 60. Oriaku G, Xiang B, Dai G, Shen J, Sun J, Lindsay WG, et al. Effects of retrograde cardioplegia on myocardial perfusion and energy metabolism in immature porcine myocardium. The Journal of Thoracic and Cardiovascular Surgery. 2000;119:1102-1109
  61. 61. Siddiqi S, Blackstone EH, Bakaeen FG. Bretschneider and del Nido solutions: Are they safe for coronary artery bypass grafting? If so, how should we use them? Journal of Cardiac Surgery. 2018;33:229-234
  62. 62. Borger MA, Rao V, Weisel RD, Floh AA, Cohen G, Feindel CM, et al. Reoperative coronary bypass surgery: Effect of patent grafts and retrograde cardioplegia. The Journal of Thoracic and Cardiovascular Surgery. 2001;121:83-90
  63. 63. Puskas JD, Gaudino M, Taggart DP. Experience is crucial in off-pump coronary artery bypass grafting. Circulation. 2019;139:1872-1875
  64. 64. Jarral OA, Saso S, Athanasiou T. Off-pump coronary artery bypass in patients with left ventricular dysfunction: A meta-analysis. The Annals of Thoracic Surgery. 2011;92:1686-1694
  65. 65. Guan Z, Guan X, Gu K, Lin X, Lin J, Zhou W, et al. Short-term outcomes of on vs off-pump coronary artery bypass grafting in patients with left ventricular dysfunction: A systematic review and meta-analysis. Journal of Cardiothoracic Surgery. 2020;15:84
  66. 66. Ueki C, Miyata H, Motomura N, Sakaguchi G, Akimoto T, Takamoto S. Off pump versus on-pump coronary artery bypass grafting in patients with left ventricular dysfunction. The Journal of Thoracic and Cardiovascular Surgery. 2016;151:1092-1098
  67. 67. Al Jaaly E, Chaudhry UA, Harling L, Athanasiou T. Should we consider beating-heart on-pump coronary artery bypass grafting over conventional cardioplegic arrest to improve postoperative outcomes in selected patients? Interactive Cardiovascular and Thoracic Surgery. 2015;20:538-545
  68. 68. Pegg TJ, Selvanayagam JB, Francis JM, Karamitsos TD, Maunsell Z, Yu LM, et al. A randomized trial of on-pump beating heart and conventional cardioplegic arrest in coronary artery bypass surgery patients with impaired left ventricular function using cardiac magnetic resonance imaging and biochemical markers. Circulation. 2008;118:2130-2138
  69. 69. Aldea GS, Bakaeen FG, Pal J, Fremes S, Head SJ, Sabik J, et al. The Society of Thoracic Surgeons clinical practice guidelines on arterial conduits for coronary artery bypass grafting. The Annals of Thoracic Surgery. 2016;101:801-809
  70. 70. Neumann FJ, Sousa-Uva M, Ahlsson A, Alfonso F, Banning AP, Benedetto U, et al. 2018 ESC/EACTS guidelines on myocardial revascularization. European Heart Journal. 2019;40:87-165
  71. 71. Gaudino M, Benedetto U, Taggart DP. Radial-artery grafts for coronary-artery bypass surgery. The New England Journal of Medicine. 2018;379:1967-1968
  72. 72. Pu A, Ding L, Shin J, Price J, Skarsgard P, Wong DR, et al. Long-term outcomes of multiple arterial coronary artery bypass grafting: A population-based study of patients in British Columbia, Canada. JAMA Cardiology. 2017;2:1187-1196
  73. 73. Weiss AJ, Zhao S, Tian DH, Taggart DP, Yan TD. A meta-analysis comparing bilateral internal mammary artery with left internal mammary artery for coronary artery bypass grafting. Annals of Cardiothoracic Surgery. 2013;2:390-400
  74. 74. Shahian DM, O’Brien SM, Filardo G, Ferraris VA, Haan CK, Rich JB, et al. The Society of Thoracic Surgeons 2008 cardiac surgery risk models: Part 1—Coronary artery bypass grafting surgery. The Annals of Thoracic Surgery. 2009;88:S2-S22
  75. 75. He GW, Taggart DP. Spasm in arterial grafts in coronary artery bypass grafting surgery. The Annals of Thoracic Surgery. 2016;101:1222-1229
  76. 76. Silva M, Rong LQ , Naik A, Rahouma M, Hameed I, Robinson B, et al. Intraoperative graft flow profiles in coronary artery bypass surgery: A metaanalysis. Journal of Cardiac Surgery. 2020;35:279-285
  77. 77. Jones EL, Lattouf OM, Weintraub WS. Catastrophic consequences of internal mammary artery hypoperfusion. The Journal of Thoracic and Cardiovascular Surgery. 1989;98:902-907
  78. 78. Navia D, Cosgrove DM III, Lytle BW, Taylor PC, McCarthy PM, Stewart RW, et al. Is the internal thoracic artery the conduit of choice to replace a stenotic vein graft? The Annals of Thoracic Surgery. 1994;57:40-44
  79. 79. Gaudino M, Benedetto U, Fremes S, Biondi-Zoccai G, Sedrakyan A, Puskas JD, et al. Radial-artery or saphenous-vein grafts in coronary-artery bypass surgery. The New England Journal of Medicine. 2018;378:2069-2077
  80. 80. Lytle BW, Blackstone EH, Sabik JF, Houghtaling P, Loop FD, Cosgrove DM. The effect of bilateral internal thoracic artery grafting on survival during 20 postoperative years. The Annals of Thoracic Surgery. 2004;78:2005-2014
  81. 81. Schwann TA, Al-Shaar L, Tranbaugh RF, Dimitrova KR, Hoffman DM, Geller CM, et al. Multi versus single arterial coronary bypass graft surgery across the ejection fraction spectrum. The Annals of Thoracic Surgery. 2015;100:810-818
  82. 82. Samadashvili Z, Sundt TM III, Wechsler A, Chikwe J, Adams DH, Smith CR, et al. Multiple versus single arterial coronary bypass graft surgery for multivessel disease. Journal of the American College of Cardiology. 2019;74:1275-1285
  83. 83. Chikwe J, Sun E, Hannan EL, Itagaki S, Lee T, Adams DH, et al. Outcomes of second arterial conduits in patients undergoing multivessel coronary artery bypass graft surgery. Journal of the American College of Cardiology. 2019;74:2238-2248
  84. 84. Mohammadi S, Kalavrouziotis D, Cresce G, Dagenais F, Dumont E, Charbonneau E, et al. Bilateral internal thoracic artery use in patients with low ejection fraction: Is there any additional long-term benefit? European Journal of Cardio-Thoracic Surgery. 2014;46:425-431
  85. 85. Gaudino M, Bakaeen F, Benedetto U, Rahouma M, Di Franco A, Tam DY, et al. Use rate and outcome in bilateral internal thoracic artery grafting: Insights from a systematic review and meta-analysis. Journal of the American Heart Association. 2018;7:e009361
  86. 86. Benedetto U, CodispotiM. Age cutoff for the loss of survival benefit from use of radial artery in coronary artery bypass grafting. The Journal of Thoracic and Cardiovascular Surgery. 2013;146:1078-1085
  87. 87. Benedetto U, Amrani M, Raja SG. Guidance for the use of bilateral internal thoracic arteries according to survival benefit across age groups. The Journal of Thoracic and Cardiovascular Surgery. 2014;148:2706-2711
  88. 88. Gaudino M, Samadashvili Z, Hameed I, Chikwe J, Girardi LN, Hannan EL. Differences in long-term outcomes after coronary artery bypass grafting using single vs multiple arterial grafts and the association with sex. JAMA Cardiology. 2020;6:401-409
  89. 89. Ad N, Barnett SD, Haan CK, et al. Does preoperative atrial fibrillation increase the risk for mortality and morbidity after coronary artery bypass grafting? The Journal of Thoracic and Cardiovascular Surgery. 2009;137:901-906
  90. 90. Badhwar V, Rankin JS, Damiano RJ, et al. The Society of Thoracic Surgeons 2017 clinical practice guidelines for the surgical treatment of atrial fibrillation. The Annals of Thoracic Surgery. 2017;103:329-341
  91. 91. Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). European Heart Journal. 2021;42:373-498
  92. 92. Malaisrie SC, McCarthy PM, Kruse J, et al. Ablation of atrial fibrillation during coronary artery bypass grafting: Late outcomes in a Medicare population. The Journal of Thoracic and Cardiovascular Surgery. 2019;161:1251.e1-1261.e1
  93. 93. Guglin M, Chen R, Curtis AB. Sinus rhythm is associated with fewer heart failure symptoms: Insights from the AFFIRM trial. Heart Rhythm. 2010;7:596-601
  94. 94. Ad N, Henry L, Hunt S. The impact of surgical ablation in patients with low ejection fraction, heart failure, and atrial fibrillation. European Journal of Cardio-Thoracic Surgery. 2011;40:70-76
  95. 95. Pecha S, Ahmadzade T, Schafer T, et al. Safety and feasibility of concomitant surgical ablation of atrial fibrillation in patients with severely reduced left ventricular ejection fraction. European Journal of Cardio-Thoracic Surgery. 2014;46:67-71
  96. 96. Goliasch G, Bartko PE, Pavo N, et al. Refining the prognostic impact of functional mitral regurgitation in chronic heart failure. European Heart Journal. 2018;39:39-46
  97. 97. Michler RE, Smith PK, Parides MK, Ailawadi G, Thourani V, Moskowitz AJ, et al. Two-year outcomes of surgical treatment of moderate ischemic mitral regurgitation. The New England Journal of Medicine. 2016;374:1932-1941
  98. 98. Chan KM, Punjabi PP, Flather M, Wage R, Symmonds K, Roussin I, et al. Coronary artery bypass surgery with or without mitral valve annuloplasty in moderate functional ischemic mitral regurgitation: Final results of the randomized ischemic mitral evaluation (RIME) trial. Circulation. 2012;126:2502-2510
  99. 99. Fattouch K, Guccione F, Sampognaro R, Panzarella G, Corrado E, Navarra E, et al. POINT: Efficacy of adding mitral valve restrictive annuloplasty to coronary artery bypass grafting in patients with moderate ischemic mitral valve regurgitation: A randomized trial. The Journal of Thoracic and Cardiovascular Surgery. 2009;138:278-285
  100. 100. Goldstein D, Moskowitz AJ, Gelijns AC, Ailawadi G, Parides MK, Perrault LP, et al. Two-year outcomes of surgical treatment of severe ischemic mitral regurgitation. The New England Journal of Medicine. 2016;374:344-353
  101. 101. Nappi F, Lusini M, Spadaccio C, Nenna A, Covino E, Acar C, et al. Papillary muscle approximation versus restrictive annuloplasty alone for severe ischemic mitral regurgitation. Journal of the American College of Cardiology. 2016;67:2334-2346
  102. 102. Fattouch K, Lancellotti P, Castrovinci S, Murana G, Sampognaro R, Corrado E, et al. Papillary muscle relocation in conjunction with valve annuloplasty improve repair results in severe ischemic mitral regurgitation. The Journal of Thoracic and Cardiovascular Surgery. 2012;143:1352-1355
  103. 103. Malhotra A, Sharma P, Garg P, Bishnoi A, Kothari J, Pujara J. Ring annuloplasty for ischemic mitral regurgitation: A single center experience. Asian Cardiovascular & Thoracic Annals. 2014;22(7):781-786
  104. 104. Haywood N, Mehaffey JH, Chancellor WZ, Beller JP, Speir A, Quader M, et al. Burden of tricuspid regurgitation in patients undergoing coronary artery bypass grafting. The Annals of Thoracic Surgery. 2021;111:44-50
  105. 105. Bertrand PB, Overbey JR, Zeng X, Levine RA, Ailawadi G, Acker MA, et al. Progression of tricuspid regurgitation after surgery for ischemic mitral regurgitation. Journal of the American College of Cardiology. 2021;77:713-724
  106. 106. Otto CM, Nishimura RA, Bonow RO, Carabello BA, Erwin JP III, Gentile F, et al. 2020 ACC/AHA guideline for the management of patients with valvular heart disease: A report of the American College of Cardiology/American Heart Association joint committee on clinical practice guidelines. Journal of the American College of Cardiology. 2021;77:e25-e197
  107. 107. Buckberg G, Athanasuleas C, Conte J. Surgical ventricular restoration for the treatment of heart failure. Nature Reviews. Cardiology. 2012;9:703-716
  108. 108. O’Neill JO, Starling RC, McCarthy PM, Albert NM, Lytle BW, Navia J, et al. The impact of left ventricular reconstruction on survival in patients with ischemic cardiomyopathy. European Journal of Cardio-Thoracic Surgery. 2006;30:753-759
  109. 109. Ribeiro GA, da Costa CE, Lopes MM, et al. Left ventricular reconstruction benefits patients with ischemic cardiomyopathy and nonviable myocardium. European Journal of Cardio-Thoracic Surgery. 2006;29:196-201
  110. 110. Prucz RB, Weiss ES, Patel ND, et al. Coronary artery bypass grafting with or without surgical ventricular restoration: A comparison. The Annals of Thoracic Surgery. 2008;86:806-814
  111. 111. Jones RH, Velazquez EJ, Michler RE, Sopko G, Oh JK, O’Connor CM, et al. Coronary bypass surgery with or without surgical ventricular reconstruction. The New England Journal of Medicine. 2009;360:1705-1717
  112. 112. Buckberg GD, Athanasuleas CL, Wechsler AS, Beyersdorf F, Conte JV, Strobeck JE. The STICH trial unravelled. European Journal of Heart Failure. 2010;12:1024-1027
  113. 113. Isomura T, Hoshino J, Fukada Y, Kitamura A, Katahira S, Kondo T, et al. Volume reduction rate by surgical ventricular restoration determines late outcome in ischaemic cardiomyopathy. European Journal of Heart Failure. 2011;13:423-431
  114. 114. Di Donato M, Castelvecchio S, Menicanti L. End-systolic volume following surgical ventricular reconstruction impacts survival in patients with ischaemic dilated cardiomyopathy. European Journal of Heart Failure. 2010;12:375-381
  115. 115. Antman EM, Anbe DT, Armstrong PW, Bates ER, Green LA, Hand M, et al. ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction—Executive summary: A report of the American College of Cardiology/American Heart Association task force on practice guidelines (writing committee to revise the 1999 guidelines for the management of patients with acute myocardial infarction). [published correction appears in circulation. 2005;111:2013]. Circulation. 2004;110:588-636
  116. 116. Morici N, Oliva F, Ajello S, Stucchi M, Sacco A, Cipriani MG, et al. Management of cardiogenic shock in acute decompensated chronic heart failure: The ALTSHOCK phase II clinical trial. American Heart Journal. 2018;204:196-201
  117. 117. McGee MG, Zillgitt SL, Trono R, Turner SA, Davis GL, Fuqua JM, et al. Retrospective analyses of the need for mechanical circulatory support (intraaortic balloon pump/abdominal left ventricular assist device or partial artificial heart) after cardiopulmonary bypass. A 44 month study of 14,168 patients. The American Journal of Cardiology. 1980;46:135-142
  118. 118. Boeken U, Feindt P, Litmathe J, Kurt M, Gams E. Intraaortic balloon pumping in patients with right ventricular insufficiency after cardiac surgery: Parameters to predict failure of IABP support. The Thoracic and Cardiovascular Surgeon. 2009;57:324-328
  119. 119. Engstrom AE, Granfeldt H, Seybold-Epting W, Dahm M, Cocchieri R, Driessen AH, et al. Mechanical circulatory support with the Impella 5.0 device for postcardiotomy cardiogenic shock: A three-center experience. Minerva Cardioangiologica. 2013;61:539-546
  120. 120. David CH, Quessard A, Mastroianni C, Hekimian G, Amour J, Leprince P, et al. Mechanical circulatory support with the Impella 5.0 and the Impella left direct pumps for postcardiotomy cardiogenic shock at La Pitie-Salpetriere hospital. European Journal of Cardio-Thoracic Surgery. 2020;57:183-188
  121. 121. Griffith BP, Anderson MB, Samuels LE, Pae WE Jr, Naka Y, Frazier OH. The RECOVER I: A multicenter prospective study of Impella 5.0/LD for postcardiotomy circulatory support. The Journal of Thoracic and Cardiovascular Surgery. 2013;145:548-554
  122. 122. Lorusso R, Whitman G, MilojevicM RG, McMullan DM, Boeken U, et al. EACTS/ELSO/STS/AATS expert consensus on post-cardiotomy extracorporeal life support in adult patients. The Journal of Thoracic and Cardiovascular Surgery. 2020;2021(161):1287-1331
  123. 123. Rastan AJ, Dege A, Mohr M, Doll N, Falk V, Walther T, et al. Early and late outcomes of 517 consecutive adult patients treated with extracorporeal membrane oxygenation for refractory postcardiotomy cardiogenic shock. The Journal of Thoracic and Cardiovascular Surgery. 2010;139:302-311
  124. 124. Elsharkawy HA, Li L, Esa WA, Sessler DI, Bashour CA. Outcome in patients who require venoarterial extracorporeal membrane oxygenation support after cardiac surgery. Journal of Cardiothoracic and Vascular Anesthesia. 2010;24:946-951
  125. 125. Wang L, Wang H, Hou X. Clinical outcomes of adult patients who receive extracorporeal membrane oxygenation for postcardiotomy cardiogenic shock: A systematic review and meta-analysis. Journal of Cardiothoracic and Vascular Anesthesia. 2018;32:2087-2093
  126. 126. Mariscalco G, Salsano A, Fiore A, Dalen M, Ruggieri VG, Saeed D, et al. Peripheral versus central extracorporeal membrane oxygenation for postcardiotomy shock: Multicenter registry, systematic review, and meta-analysis. The Journal of Thoracic and Cardiovascular Surgery. 2020;160:1207-1216
  127. 127. Schrage B, Becher PM, Bernhardt A, Bezerra H, Blankenberg S, Brunner S, et al. Left ventricular unloading is associated with lower mortality in patients with cardiogenic shock treated with venoarterial extracorporeal membrane oxygenation: Results from an international, multicenter cohort study. Circulation. 2020;142:2095-2106
  128. 128. Russo JJ, Aleksova N, Pitcher I, Couture E, Parlow S, Faraz M, et al. Left ventricular- unloading during extracorporeal membrane oxygenation in patients with cardiogenic shock. Journal of the American College of Cardiology. 2019;73:654-662
  129. 129. Murphy SP, Ibrahim NE, Januzzi JL Jr. Heart failure with reduced ejection fraction: A review. Journal of the American Medical Association. 2020;324:488-504
  130. 130. Maddox TM, Januzzi JL Jr, Allen LA, Breathett K, Butler J, Davis LL, et al. 2021 update to the 2017 ACC expert consensus decision pathway for optimization of heart failure treatment: Answers to 10 pivotal issues about heart failure with reduced ejection fraction: A report of the American College of Cardiology solution set oversight committee. Journal of the American College of Cardiology. 2021;77:772-810
  131. 131. Hernandez AF, Greiner MA, Fonarow GC, Hammill BG, Heidenreich PA, Yancy CW, et al. Relationship between early physician follow-up and 30-day readmission among Medicare beneficiaries hospitalized for heart failure. Journal of the American Medical Association. 2010;303:1716-1722
  132. 132. Farsky PS, White J, Al-Khalidi HR, Sueta CA, Rouleau JL, Panza JA, et al. O'Connor CM; working group and surgical treatment for IsChemic heart failure trial Investigators. Optimal medical therapy with or without surgical revascularization and long-term outcomes in ischemic cardiomyopathy. The Journal of Thoracic and Cardiovascular Surgery. 2021;S0022-5223(20):33449-33448

Written By

Samuel Jacob, Pankaj Garg, Games Gramm and Saqib Masroor

Submitted: 05 March 2022 Reviewed: 14 April 2022 Published: 21 May 2022