Review Article

Cardiogenic Shock Management and Research: Past, Present, and Future Outlook

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

Although great strides have been made in the pathophysiological understanding, diagnosis and management of cardiogenic shock (CS), morbidity and mortality in patients presenting with the condition remain high. Acute MI is the commonest cause of CS; consequently, most existing literature concerns MI-associated CS. However, there are many more phenotypes of patients with acute heart failure. Medical treatment and mechanical circulatory support are well-established therapeutic options, but evidence for many current treatment regimens is limited. The issue is further complicated by the fact that implementing adequately powered, randomized controlled trials are challenging for many reasons. In this review, the authors discuss the history, landmark trials, current topics of medical therapy and mechanical circulatory support regimens, and future perspectives of CS management.

Disclosure:SO has received research and study grants from Novartis; EP is proctor and consultant for and has received institutional grants from Abbott, Medtronic, and Abiomed; BO is a consultant for Teleflex and has received research funding from the British Heart Foundation and the National Institute for Health Science Research. All other authors have no conflict of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Sascha Ott, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany. E: sott@dhzb.de

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

In 1939, Harrison introduced cardiogenic shock (CS) as a specific entity and differentiated it from other forms of shock.1 Acute MI (AMI) is the most common cause of CS and has a mortality rate of up to 50%, which has changed little in the past two decades.2

In the early years of interventional AMI therapy, intracoronary thrombolytic agents were used to dissolve thrombi.3,4 Later, in 1977, Gruentzig performed the first percutaneous coronary artery balloon angioplasty. Nearly one decade later in 1986, the first bare metal stent was implanted. Since then, coronary artery stents have undergone significant development, spawning generations of drug-eluting stents.5 In 1999, Hochman et al. published one of the first major randomized controlled trials (RCTs) in the field, the SHOCK trial, proving early revascularization in AMI-associated CS (AMICS) to be the cornerstone of successful treatment and reduction in mortality for these patients.6

Another fundamental pillar for supporting AMICS patients is to bridge hemodynamic instability with mechanical circulatory support (MCS) devices. More than 50 years after the intra-aortic balloon pump (IABP) was developed in 1968 as the first MCS technology, the arsenal has increased considerably. Currently, IABP, the Impella (a miniaturized ventricular assist device; Abiomed) and extracorporeal life support (ECLS) circuits are the most common devices for acute and short-term MCS in CS.

Technical advances in the performance and manageability of MCS devices have led to their widespread availability and more frequent use. However, despite the remarkable increase in short-term MCS use, there is still little evidence from RCTs showing any significant improvement in strong outcome parameters.

Landmark Trials and a Slow Evolution

Table 1 provides an overview of the key trials in medical and mechanical therapy of CS in the past 20 years. The initial trials of MCS were predominantly retrospective cohort analyses with all their known limitations. Since the first use of a heart-lung machine by Gibbon in 1965, meaningful scientific interrogation of MCS modalities has evolved slowly.

First, a major issue in performing and comparing trials in the field of CS and MCS related to CS is the absence of a standard definition. Table 2 summarizes different definitions of CS by the US and European societies as well as those of some landmark trials. In addition, differing primary endpoints among study groups further complicate the situation.

Key Trials in Cardiogenic Shock

Article image

Key Trials in Cardiogenic Shock

Article image

Different Definitions of Cardiogenic Shock

Article image

Second, although AMI is the most common cause of CS, there are many more phenotypes of patients with acute heart failure, which adds complexity to screening and randomizing patients for a trial in time-pressured circumstances.2 Defining a specific study population can be problematic and obtaining patients’ informed consent difficult. Furthermore, blinding is normally not possible.

Consequently, the first randomized controlled trials (RCTs) in the field of MCS were small trials enrolling fewer than 50 patients. Table 1 provides an overview of the RCTs in MCS. Most of the trials were aborted because of low recruitment rates, highlighting one of the major problems in prospective RCTs in MCS.7 Other than increasing the number of recruiting centers in multicenter trials, the hub and spoke model proposed by van Diepen et al. could improve recruitment yield.8

Furthermore, there are ethical issues around randomizing critically ill patients to be supported with MCS, which is often thought to be a last-chance treatment option, and such issues will require careful consideration when designing a trial. In this context, the study protocol of the DAWN trial, explained by Samsky et al., could be helpful.9,10

Moreover, the necessary infrastructure participating sites need to establish to successfully undertake trials in MCS is complex, costly, and can be delivered by only a limited number of select centers.11

Finally, volume/outcome relationships for MCS programs are increasingly well documented, and the need for dedicated cardiogenic shock centers has become apparent.12,13

Current Topics and Updates from Recent Studies

Definition and Diagnosis of Cardiogenic Shock

Therapeutic intervention is often time critical, not least to minimize secondary end-organ damage, so early diagnosis is key.

Recently, Chioncel et al. again highlighted the significance of early detection of tissue hypoperfusion in patients with evolving or established CS.14 Shortly afterwards, Chioncel et al. published a position statement on behalf of the European Society of Cardiology Heart Failure Association, presenting a new definition of CS that focuses on the importance of hypoperfusion and organ dysfunction.15 In this context, hypotension is no longer a required criterion, and the definition now includes normotensive CS.

Baran et al. presented the Society for Cardiovascular Angiography and Interventions (SCAI) clinical expert consensus statement on the classification of cardiogenic shock, defining a grading system of CS ranging from stage A with patients At risk, through to B and C (Beginning and Classic CS) – then to D and E (Deteriorating patients and those in Extremis).16 This classification was shown to correlate with both in-hospital mortality and mortality after hospital discharge.17–20

Ceglarek et al. published the results of a CULPRIT-SHOCK biomarker sub-study and presented a novel, fast, and objective, biomarker-based mortality risk score for patients with AMICS.21 Based on an evaluation of cystatin c, lactate, interleukin-6 and N-terminal pro-B-type natriuretic peptide, they established and validated the CLIP score as a mortality risk predictor.

Hemodynamic Monitoring

Cardiac output (CO) or cardiac index (CI) are key parameters in evaluating patients with CS. There is an ongoing debate over the role of the pulmonary catheter (PAC). Although the PAC was first introduced and studied in the mid-1970s, 50 years later there is still no clear evidence concerning its significance.22

The use of PACs has decreased over the years, probably influenced by the ESCAPE trial and other studies which showed there was no benefit from PAC monitoring.23,24 However, this study did not enroll CS patients. To date, PACs have never been studied specifically in a CS population. Existing data were obtained from different study populations and reveal contradictory results.25 In a propensity score-matched retrospective cohort study with the remarkable number of 9,431,944 patients, Hernandez et al. showed that the use of PACs in patients with heart failure was, indeed, associated with increased mortality (9.9% versus 3.3%; OR 3.96; p<0.001); however, in patients with CS, PACs were associated with lower mortality (35.1% versus 39.2%; OR 0.91; p< 0.001).26 Another recently published study and two expert consensus articles support the use of PACs in CS.27–29 Consequently, CS treatment algorithms in current studies include PAC monitoring.12,30

However, because of concerns about the safety of PACs, alternatives, such as transpulmonary thermodilution (TPTD), are being investigated. Only a few studies have addressed the accuracy and utility of TPTD methods in patients with CS. Schmid et al. compared values derived from TPTD and PAC in a population of 11 patients with CS and found they gave identical results.31 Zhang et al. prospectively randomized 60 patients with AMICS to a pulse contour (invasive) continuous cardiac output (PiCCO)-guided therapy group versus standard care and demonstrated a mortality benefit in patients treated with PiCCO, a TPTD method.32

Technical advances in the past decade have opened up new avenues of noninvasive cardiac output monitoring (NICOM). While setting out to assess these innovations, the recently published NICOM study reported disappointing results, showing one NICOM modality to be unreliable in measuring cardiac output in patients with decompensated heart failure and CS.33

Reperfusion strategies

Early revascularization has been accepted for many years as the key intervention to treat patients with AMI to prevent deterioration to CS and is firmly established in current guidelines and recommendations.6,34,35 A significant number of patients with acute MI, however, present with more than one coronary lesion. The CULPRIT-SHOCK trial prospectively showed a culprit-lesion-only percutaneous coronary intervention (PCI) strategy to be superior to an immediate multivessel PCI in patients presenting with AMICS.36

Another aspect was identified in a sub-study of the CULPRIT-SHOCK trial. Guedeney et al. were able to show that transradial artery access compared with transfemoral artery access was associated with a lower 30-day death rate (34.7% versus 49.7%; adjusted OR 0.56; 95% CI [0.33–0.96]) and a lower rate of renal replacement therapy requirement (5.9% versus 15.9%; adjusted OR 0.40; 95% CI [0.16-0.97]).37 However, this benefit could not be confirmed at the 1-year follow up. The causes for the improved short-term endpoints warrant further research.

Mechanical Circulatory Support

Intra-aortic Balloon Pump

In 2013, Thiele et al. published the IABP-SHOCK II trial, the first adequately powered prospective, randomized multicenter trial comparing IABP against controls in a CS population, which showed no improvement in 30-day, 12-month, or 6-year mortality rates.38,39 Consequently, European guidelines do not recommend routine IABP implantation in CS (class 3b).40 The last US guideline for managing AMICS from 2013 did not yet include the IABP-SHOCK II data and issued a class 2a recommendation.35 Later updates, however, downgraded the recommendation for the routine use of IABP.28, 41

In a subgroup analysis of the IABP-SHOCK II trial, Fuernau et al. investigated the impact of timing of IABP on mortality in CS and found no difference whether IABP was implanted before or after PCI.42

Impella

Although the Impella has been shown to provide superior hemodynamic support to IABP, there are conflicting results with respect to hard outcome parameters.43–45 However, these conflicting results originate mostly from studies comparing Impella to other MCS strategies.44,46-48

To date there is only one study, a retrospective, single-center cohort analysis, comparing Impella to medical treatment in patients after out-of-hospital cardiac arrest due to AMI who subsequently present with CS.49 This analysis suggests an Impella-associated survival benefit at hospital discharge and after 6 months.

Scherer et al. recently presented propensity-score matched data from a retrospective analysis comparing Impella CP (n=70) with non-MCS-treated CS patients (n=70).50 While, naturally, there were more bleeding complications in the Impella groups, mortality rates did not differ.

The ISAR-SHOCK trial compared Impella 2.5 (n=12) with IABP (n=13) in patients presenting with AMICS, and revealed a higher CI in the Impella group.44 Mortality, however, was not influenced.

Manzo-Silberman et al. retrospectively compared Impella 2.5 with IABP in 78 patients and found no difference in mortality, but a higher rate of bleeding complications in the Impella group.46 Patients in the Impella group were on higher catecholamine doses (epinephrine 2.3 mg/h versus 1.0 mg/h; p=0.04) and their left ventricular (LV) ejection fraction was lower (25% versus 35%; p=0.01), indicating the groups were not balanced with respect to illness severity.

In 2017, Ouweneel et al. presented the IMPRESS study, the first prospective multicenter trial comparing Impella CP (n=24) with IABP (n=24).47 This study, again, showed no significant difference in mortality rate (50% versus 46%; p=0.92); differences in bleeding complications also failed to reach statistical significance. Notably, all patients in the Impella group and 83% of the IABP group underwent cardiopulmonary resuscitation before device implantation.

A study by Pieri et al., though retrospective, single-centered and non-randomized, also identified aspects warranting further evaluation.48 As in the study by Manzo-Silberman et al., patients in the Impella group (2.5 and CP models) were more critically ill, as indicated by more frequent catecholamine support (93% versus 57%; p=0.002), and were on higher doses of inotropes (indicated by an inotropic score of 8 versus 5; p=0.02). Nevertheless, the 30-day mortality rate tended to be lower in the Impella group (79% versus 94%; p=0.11). In any case, mortality rates of 79% or 94% appear conspicuously high. At 6-month follow-up, LV ejection fraction and cardiac recovery rate were higher in the Impella group. A retrospective comparison of historical cohorts on Impella versus IABP support by Alushi et al. yielded similar results.51

Another remarkable study, by Schrage et al., compared Impella 2.5 and CP with a historical control group from the IABP-SHOCK II trial treated with IABP or medical treatment; it showed no survival benefit for the Impella group, but more bleeding and peripheral vascular complications.52 Adjusting the control group to IABP patients alone did not change the results; however, a comparative analysis after adjusting for medical treatment alone was not performed.

The recently published propensity-matched, registry-based, retrospective cohort study by Dhruva et al. attracted attention after reporting a higher adjusted risk of in-hospital death or major bleeding complications under Impella support compared to IABP.53 This study, however, has been heavily criticized for statistical limitations and incomplete conditions for comparison.54

In contrast, Lemor et al. retrospectively analyzed data of 6,290 patients from the US National Inpatient Sample register to compare Impella with veno-arterial extracorporeal membrane oxygenation (VA-ECMO) in patients with AMICS.55 In this propensity score-matched study, patients treated with Impella had a significantly lower in-hospital mortality rates than those receiving V-A ECMO (26.7% versus 43.3%; OR: 2.10; p=0.021). However, these data are based on the International Classification of Diseases (ICD) and CS was thus identified on the basis of the ICD code, not on hemodynamic parameters. Furthermore, there was no discrimination as to the specific devices or cannulation strategies. Despite these substantial limitations, the impressive number of patients must be acknowledged, and the results are consistent with current findings that VA-ECMO is not, at least not in isolation, the panacea for CS.

On the basis of available data, there are growing calls for limiting the unrestricted use of Impella in AMICS.56 The limitations of these data, however, bring us back to the root of the problem. While propensity-score matched analyses mitigate numerous limiting factors of retrospective studies, they are still not RCTs and significant limitations remain. Well-powered, prospective, multicenter RCTs producing high-quality data to delineate the significance of Impella are still lacking, and numerous other questions, such as those concerning timing of implantation, choice of Impella device, combination with other devices or just suitable patient selection, have not yet been sufficiently addressed.

For example, Nersesian et al. identified a lactate level >8 mmol/l or having received cardiopulmonary resuscitation before implantation as predictors for increased 30-day mortality in a mixed etiology cohort of patients with CS treated with Impella 5 or 5.5.57 Indeed, the use of Impella 5 or 5.5 in CS after cardiopulmonary resuscitation was associated with an increase in 30-day mortality (92% versus 41%, p=0.001).

Another approach, intended to reduce bleeding and other complications, is the ECMELLA 2.0 concept, where single arterial access is used for VA-ECLS and Impella.58 In this context, the results from the currently recruiting DanGer Shock trial are eagerly awaited.59 In this multicenter RCT, Udesen et al. are prospectively comparing Impella CP versus conventional therapy in patients with AMICS.

The recently published consensus statement by the European Association of Percutaneous Cardiovascular Interventions and the Association for Acute Cardiovascular Care recommends Impella CP to be considered for short-term MCS in CS stage C and D with a potentially reversible underlying cause, as a bridge to transplant, or in ventricular assist device candidates.60

Veno-arterial Extracorporeal Life Support

The Extracorporeal Life Support Organization published a position paper in 2019 addressing the nomenclature of ECLS.61 According to this, ECLS is defined as a set of therapies that focus on oxygenation, carbon dioxide removal, cardiac support, or a combination thereof. ECMO is one ECLS entity used for temporary support of patients with respiratory and/or cardiac failure. Therefore, in general, the term ECLS is used in this article; when reviewing former studies that used the term ECMO, this terminology was maintained.

 

Even though VA-ECLS is significantly older than Impella, high-quality data are even more scarce. The first RCTs that systematically addressed the role of VA-ECLS in CS have only been published in the past 2 years.

The retrospective cohort analyses by Lemor et al. discussed above, which has significant limitations, showed VA-ECLS to be inferior to Impella in treating CS.55

Pozzi et al. recently published a retrospective observational analysis from their institutional database of patients treated with VA-ECMO in AMICS.62 Between 2007 and 2017, they treated 56 patients with VA-ECMO and demonstrated a survival-to-discharge rate of 41.1% (n=23). Notably, the results of a subgroup analysis showed that patients aged ≤60 years had a better chance of survival. This matches the findings of Muller et al., who identified an age of ≥60 years as an independent risk factor for death during an ICU stay.63 However, in Pozzi et al.’s study, the survival rate with VA-ECMO did not substantially exceed common survival rates of CS treated conventionally. Relatively little is known about their local MCS protocols, and it is questionable whether a median of less than six patients per year can support a complex intervention like ECLS to become standard care.

The only published randomized trial comparing VA-ECMO treatment of CS with standard care is the ECLS-SHOCK trial.64,65 Forty-one patients with AMICS were randomized to receive VA-ECMO or not. There was no difference in the primary endpoint of LV recovery. All-cause mortality after 1 year showed no difference either but a trend of lower mortality in the VA-ECLS group was observed (19% versus 38%; p=0.31). Mortality was a secondary endpoint though, and the study was underpowered to detect a difference.

Left Ventricular Unloading and ECMELLA

Despite the ability of VA-ECLS to support cardiac and pulmonary function, there are considerable limitations and disadvantages to this approach. The increase in LV afterload and consecutive rise in LV wall stress impeding recovery has been known about for many years.66–68

The mortality-reducing effect of LV unloading regardless of the method applied (IABP, Impella, right upper pulmonary vein drainage, or transseptal left atrial cannula) has been underlined by recent meta-analyses.69-71

Ongoing Trials in Cardiogenic Shock

Article image

The latest study addressing the combination of VA-ECLS with an Impella for LV-unloading, called ECMELLA, was recently published by Schrage et al.72 This retrospective, international, multicenter, 1:1 propensity-score matched cohort analysis compared 510 patients with CS treated with VA-ECMO with or without LV unloading by Impella. LV unloading was associated with a lower 30-day mortality (HR 0.79; 95% CI [0.63–0.98]; p=0.03). This reduction in mortality was seen even though patients with LV unloading were more likely to experience complications such as severe bleeding (38.4% versus 17.9%; p<0.01), access site-related ischemia (21.6% versus 12.3%; p<0.01), abdominal compartment syndrome (9.4% versus 3.7%; p=0.02), and a requirement for renal replacement therapy (58.5% versus 39.1%; p<0.01). Even though the data give a signal for the beneficial effect of LV unloading, this concept, again, is based on retrospective analysis of observational studies and adequate RCTs are missing.

Extracorporeal Life Support in Cardiopulmonary Resuscitation

Since VA-ECLS is quite easily and rapidly implanted at the bedside, provides biventricular and pulmonary support, and is furthermore of comparatively low cost, it distinguishes itself as a firstline MCS for patients in cardiac arrest.

In 2020, Yannopoulos et al. presented the ARREST trial, comparing ECMO-facilitated resuscitation with standard advanced cardiac life support treatment in patients with out-of-hospital cardiac arrest and refractory VF.73 For just short of a year, they randomly assigned 15 patients to each group, and there was a 36 percentage point better survival rate in the ECMO-facilitated resuscitation group (43% versus 7% survival; HR 0.16; 95% CI [0.06 – 0.41]; p<0.0001). Notably, all survivors in the ECMO group had good cerebral performance scores at 6 months. Although the trial was planned to enroll 77 patients, it was discontinued after the first interim analysis because of the superiority of VA-ECMO.

Hypothermia

The discussion whether mild hypothermia in patients with AMICS but not specifically after cardiac arrest improves morbidity and mortality has been ongoing for several years.74

The SHOCK-COOL trial investigated the impact of therapeutic hypothermia (33°C) for 24 hours in AMICS patients without a history of cardiac arrest.75 The primary endpoint was the cardiac power index after 24 hours; secondary endpoints were several hemodynamic parameters and lactate levels. There was no difference in the cardiac power index or hemodynamic parameters. Lactate levels were higher in the hypothermia group; there were no significant differences in 30-day mortality (60% versus 50%; HR 1.27; 95% CI [0.55–2.94]; p=0.55).

The HYPO-ECMO trial, a prospective, multicenter RCT examining the impact of moderate hypothermia (33–34°C) during VA-ECLS in CS patients, was recently completed and is expected to be published in 2022.76

Current Clinical Trials

Table 3 and Supplementary Material Table 1 provide an overview of ongoing clinical trials in the field of CS. Besides some trials investigating medical therapies, a considerable number of MCS studies have been initiated.

Interestingly, despite the conflicting evidence regarding the significance of Impella in CS, only the DanGer trial and the UNLOAD-AMI trial (NCT04562272) are focusing on this question.59 The ECMO-CS trial (NCT02301819) and the EURO-SHOCK trial (NCT03813134) will examine the impact of early ECLS intervention in patients with CS. Eagerly anticipated are the results of the REVERSE trial (NCT03431467) by Schrage et al., who are prospectively investigating the potential superiority of the ECMELLA concept compared with VA-ECLS alone.72

Unresolved Issues in Cardiogenic Shock Care

Article image

Conclusion

CS remains a leading cause of death in patients with acute cardiac diseases. Despite a considerable number of studies in the field of CS, major areas of care are poorly understood. Table 4 provides an overview of unresolved issues in CS care.

The key question in management of CS is how to interrupt the vicious cycle of CS progression. With AMICS, revascularization is crucial; nonetheless, evolving CS has to be treated symptomatically. Inotropic and vasopressor support has shown to have limited benefit or even cause harm in CS.77,78 The effect MCS can have on stabilizing hemodynamics has led to its widespread use, but is yet to be shown to improve clinical outcomes that matter to patients and caregivers.

Although we have seen an increasing number of studies in the field of MCS in recent years, the optimal strategy remains unclear. Results to date suggest that stratification is necessary and there is no one-size-fits-all solution.

Large RCTs have to answer questions on rational selection of patients, the best modality of MCS in different clinical circumstances, the efficacy of combining different types of MCS, and the optimal timing for implementation of MCS.

Furthermore, basic science needs to help improve our understanding of the underlying mechanisms and importance of areas such as immunomodulation, endothelial function, and genetics.

Difficulties in designing and performing high-quality clinical trials in this very sick patient population complicate the evolution of systematic and consistent evidence-based CS management protocols. Despite this, well-designed trials in clearly-defined CS patient populations must now be established.79

Click here to view Supplementary Material.

References

  1. Harrison T. Failure of the Circulation. Baltimore (MD): Williams and Wilkins; 1939.
  2. Chioncel O, Mebazaa A, Harjola VP, et al. Clinical phenotypes and outcome of patients hospitalized for acute heart failure: the ESC Heart Failure Long-Term Registry. Eur J Heart Fail 2017;1:1242–54.
    Crossref | PubMed
  3. Verstraete M. Thrombolytic therapy in recent myocardial infarction. Textbook of Coronary Care. Amsterdam: Excerpta Medica 1972:643–59.
  4. Fletcher AP, Alkjaersig N, Smyrniotis FE, Sherry S. The treatment of patients suffering from early myocardial infarction with massive and prolonged streptokinase therapy. Trans Assoc Am Physicians 1958;71:287–96.
    PubMed
  5. Tomberli B, Mattesini A, Baldereschi GI, Di Mario C. A brief history of coronary artery stents. Rev Esp Cardiol (Engl Ed) 2018;71:312–9.
    Crossref | PubMed
  6. Hochman JS, Sleeper LA, Webb JG, et al. Early revascularization in acute myocardial infarction complicated by cardiogenic shock. N Engl J Med 1999;341:625–34.
    Crossref | PubMed
  7. Vallabhajosyula S, Prasad A, Sandhu GS, et al. Ten-year trends, predictors and outcomes of mechanical circulatory support in percutaneous coronary intervention for acute myocardial infarction with cardiogenic shock. EuroIntervention 2021;16:e1254–61.
    Crossref | PubMed
  8. van Diepen S, Morrow DA. Potential growth in cardiogenic shock research though an international registry collaboration: the merits and challenges of a Hub-of-Spokes model. Eur Heart J Acute Cardiovasc Care 2021;10:3–5.
    Crossref | PubMed
  9. Nogueira RG, Jadhav AP, Haussen DC, et al. Thrombectomy 6 to 24 hours after stroke with a mismatch between deficit and infarct. N Engl J Med 2018;378:11–21.
    Crossref | PubMed
  10. Samsky M, Krucoff M, Althouse AD, et al. Clinical and regulatory landscape for cardiogenic shock: a report from the Cardiac Safety Research Consortium ThinkTank on cardiogenic shock. Am Heart J 2020;219:1–8.
    Crossref | PubMed
  11. Berg DD, Barnett CF, Kenigsberg BB, et al. Clinical practice patterns in temporary mechanical circulatory support for shock in the Critical Care Cardiology Trials Network (CCCTN) registry. Circ Heart Fail 2019;12:e006635.
    Crossref | PubMed
  12. Rab T, Ratanapo S, Kern KB, et al. Cardiac shock care centers: JACC review topic of the week. J Am Coll Cardiol 2018;72:1972–80.
    Crossref | PubMed
  13. Tchantchaleishvili V, Hallinan W, Massey HT. Call for organized statewide networks for management of acute myocardial infarction-related cardiogenic shock. JAMA Surgery 2015;150:1025–6.
    Crossref | PubMed
  14. Chioncel O, Mebazaa A, Maggioni AP, et al. Acute heart failure congestion and perfusion status – impact of the clinical classification on in-hospital and long-term outcomes; insights from the ESC-EORP-HFA Heart Failure Long-Term Registry. Eur J Heart Fail 2019;21:1338–52.
    Crossref | PubMed
  15. Chioncel O, Parissis J, Mebazaa A, et al. Epidemiology, pathophysiology and contemporary management of cardiogenic shock – a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2020;22:1315–41.
    Crossref | PubMed
  16. Baran DA, Grines CL, Bailey S, et al. SCAI clinical expert consensus statement on the classification of cardiogenic shock. Catheter Cardiovasc Interv 2019;94:29–37.
    Crossref | PubMed
  17. Jentzer JC, van Diepen S, Barsness GW, et al. Cardiogenic shock classification to predict mortality in the cardiac intensive care unit. J Am Coll Cardiol 2019;74:2117–28.
    Crossref | PubMed
  18. Jentzer JC, Baran DA, van Diepen S, et al. Admission Society for Cardiovascular Angiography and Intervention shock stage stratifies post-discharge mortality risk in cardiac intensive care unit patients. Am Heart J 2020;219:37–46.
    Crossref | PubMed
  19. Schrage B, Dabboura S, Yan I, et al. Application of the SCAI classification in a cohort of patients with cardiogenic shock. Catheter Cardiovasc Interv 2020;96:E213–9.
    Crossref | PubMed
  20. Thayer KL, Zweck E, Ayouty M, et al. Invasive hemodynamic assessment and classification of in-hospital mortality risk among patients with cardiogenic shock. Circ Heart Fail 2020;13:e007099.
    Crossref | PubMed
  21. Ceglarek U, Schellong P, Rosolowski M, et al. The novel cystatin C, lactate, interleukin-6, and N-terminal pro-B-type natriuretic peptide (CLIP)-based mortality risk score in cardiogenic shock after acute myocardial infarction. Eur Heart J 2021;42:2344–52.
    Crossref | PubMed
  22. Kuhn LA. The treatment of cardiogenic shock. I. The nature of cardiogenic shock. Am Heart J 1967;74:578–81.
    Crossref | PubMed
  23. Binanay C, Califf RM, Hasselblad V, et al. Evaluation study of congestive heart failure and pulmonary artery catheterization effectiveness: the ESCAPE trial. JAMA 2005;294:1625–33.
    Crossref | PubMed
  24. Harvey S, Harrison DA, Singer M, et al. Assessment of the clinical effectiveness of pulmonary artery catheters in management of patients in intensive care (PAC–Man): a randomised controlled trial. Lancet 2005;366:472–7.
    Crossref | PubMed
  25. Hadian M, Pinsky MR. Evidence-based review of the use of the pulmonary artery catheter: impact data and complications. Crit Care 2006;10(Suppl 3):S8.
    Crossref | PubMed
  26. Hernandez GA, Lemor A, Blumer V, et al. Trends in utilization and outcomes of pulmonary artery catheterization in heart failure with and without cardiogenic shock. J Card Fail 2019;25:364–71.
    Crossref | PubMed
  27. Saxena A, Garan AR, Kapur NK, et al. Value of hemodynamic monitoring in patients with cardiogenic shock undergoing mechanical circulatory support. Circulation 2020;141:1184–97.
    Crossref | PubMed
  28. van Diepen S, Katz JN, Albert NM, et al. Contemporary management of cardiogenic shock: a scientific statement from the American Heart Association. Circulation 2017;136:e232–68.
    Crossref | PubMed
  29. Sorajja P, Borlaug BA, Dimas VV, et al. SCAI/HFSA clinical expert consensus document on the use of invasive hemodynamics for the diagnosis and management of cardiovascular disease. Catheter Cardiovasc Interv 2017;89:e233–47.
    Crossref | PubMed
  30. Basir MB, Kapur NK, Patel K, et al. Improved outcomes associated with the use of shock protocols: updates from the national cardiogenic shock initiative. Catheter Cardiovasc Interv 2019;93:1173–83.
    Crossref | PubMed
  31. Schmid B, Fink K, Olschewski M, et al. Accuracy and precision of transcardiopulmonary thermodilution in patients with cardiogenic shock. J Clin Monit Comput 2016;30:849–56.
    Crossref | PubMed
  32. Zhang YB, Zhang ZZ, Li JX, et al. Application of pulse index continuous cardiac output system in elderly patients with acute myocardial infarction complicated by cardiogenic shock: a prospective randomized study. World J Clin Cases 2019;7:1291–301.
    Crossref | PubMed
  33. Rali AS, Buechler T, Van Gotten B, et al. Non-invasive cardiac output monitoring in cardiogenic shock: the NICOM study. J Card Fail 2020;26:160–5.
    Crossref | PubMed
  34. Neumann FJ, Sousa-Uva M, Ahlsson A, et al. 2018 ESC/EACTS guidelines on myocardial revascularization. EuroIntervention 2019;14:1435–534.
    Crossref | PubMed
  35. O’Gara PT, Kushner FG, Ascheim DD, et al. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation 2013;127:e362–425.
    Crossref | PubMed
  36. Thiele H, Akin I, Sandri M, et al. PCI strategies in patients with acute myocardial infarction and cardiogenic shock. N Engl J Med 2017;377:2419–32.
    Crossref | PubMed
  37. Guedeney P, Thiele H, Kerneis M, et al. Radial versus femoral artery access for percutaneous coronary artery intervention in patients with acute myocardial infarction and multivessel disease complicated by cardiogenic shock: Subanalysis from the CULPRIT–SHOCK trial. Am Heart J 2020;225:60–8.
    Crossref | PubMed
  38. Thiele H, Zeymer U, Neumann FJ, et al. Intra-aortic balloon counterpulsation in acute myocardial infarction complicated by cardiogenic shock (IABP–SHOCK II): final 12 month results of a randomised, open-label trial. Lancet 2013;382:1638–45.
    Crossref | PubMed
  39. Thiele H, Zeymer U, Thelemann N, et al. Intraaortic balloon pump in cardiogenic shock complicating acute myocardial infarction: long-term 6-year outcome of the randomized IABP-SHOCK II trial. Circulation 2019;139:395–403.
    Crossref | PubMed
  40. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Eur J Heart Fail 2016;18:891–975.
    Crossref | PubMed
  41. Sandhu A, McCoy LA, Negi SI, et al. Use of mechanical circulatory support in patients undergoing percutaneous coronary intervention: insights from the National Cardiovascular Data Registry. Circulation 2015;132:1243–51.
    Crossref | PubMed
  42. Fuernau G, Ledwoch J, Desch S, et al. Impact of timing of intraaortic balloon counterpulsation on mortality in cardiogenic shock – a subanalysis of the IABP–SHOCK II trial. Eur Heart J Acute Cardiovasc Care 2021;10:54–61.
    Crossref | PubMed
  43. Sjauw KD, Remmelink M, Baan J Jr, et al. Left ventricular unloading in acute ST-segment elevation myocardial infarction patients is safe and feasible and provides acute and sustained left ventricular recovery. J Am Coll Cardiol 2008;51:1044–6.
    Crossref | PubMed
  44. Seyfarth M, Sibbing D, Bauer I, et al. A randomized clinical trial to evaluate the safety and efficacy of a percutaneous left ventricular assist device versus intra-aortic balloon pumping for treatment of cardiogenic shock caused by myocardial infarction. J Am Coll Cardiol 2008;52:1584–8.
    Crossref | PubMed
  45. Amin AP, Spertus JA, Curtis JP, et al. The evolving landscape of Impella use in the United States among patients undergoing percutaneous coronary intervention with mechanical circulatory support. Circulation 2020;141:273–84.
    Crossref | PubMed
  46. Manzo-Silberman S, Fichet J, Mathonnet A, et al. Percutaneous left ventricular assistance in post cardiac arrest shock: comparison of intra aortic blood pump and IMPELLA Recover LP2.5. Resuscitation 2013;84:609–15.
    Crossref | PubMed
  47. Ouweneel DM, Eriksen E, Sjauw KD, et al. Percutaneous mechanical circulatory support versus intra-aortic balloon pump in cardiogenic shock after acute myocardial infarction. J Am Coll Cardiol 2017;69:278–87.
    Crossref | PubMed
  48. Pieri M, Sorrentino T, Oppizzi M, et al. The role of different mechanical circulatory support devices and their timing of implantation on myocardial damage and mid-term recovery in acute myocardial infarction related cardiogenic shock. J Interv Cardiol 2018;31:717–24.
    Crossref | PubMed
  49. Karatolios K, Chatzis G, Markus B, et al. Impella support compared to medical treatment for post-cardiac arrest shock after out of hospital cardiac arrest. Resuscitation 2018;126:104–10.
    Crossref | PubMed
  50. Scherer C, Lüsebrink E, Kupka D, et al. Long-term clinical outcome of cardiogenic shock patients undergoing Impella CP treatment vs standard of care. J Clin Med 2020;9:3803.
    Crossref | PubMed
  51. Alushi B, Douedari A, Froehlig G, et al. Impella versus IABP in acute myocardial infarction complicated by cardiogenic shock. Open Heart 2019;6:e000987.
    Crossref | PubMed
  52. Schrage B, Ibrahim K, Loehn T, et al. Impella support for acute myocardial infarction complicated by cardiogenic shock. Circulation 2019;139:1249–58.
    Crossref | PubMed
  53. Dhruva SS, Ross JS, Mortazavi BJ, et al. Association of use of an intravascular microaxial left ventricular assist device vs intra-aortic balloon pump with in-hospital mortality and major bleeding among patients with acute myocardial infarction complicated by cardiogenic shock. JAMA 2020;323:734–45.
    Crossref | PubMed
  54. Ravn HB, Helgestad OKL, Møller JE. Intravascular microaxial left ventricular assist device vs intra-aortic balloon pump for cardiogenic shock. JAMA 2020;324:302–3.
    Crossref | PubMed
  55. Lemor A, Hosseini Dehkordi SH, Basir MB, et al. Impella versus extracorporeal membrane oxygenation for acute myocardial infarction cardiogenic shock. Cardiovasc Revasc Med 2020;21`:1465–71.
    Crossref | PubMed
  56. Wernly B, Bhatt DL, Thiele H, Jung C. Impella in cardiogenic shock: Is it time to hit the break? Shock 2021;55:693–4.
    Crossref | PubMed
  57. Nersesian G, Tschöpe C, Spillmann F, et al. Prediction of survival of patients in cardiogenic shock treated by surgically implanted Impella 5+ short-term left ventricular assist device. Interact Cardiovasc Thorac Surg 2020;31:475–82.
    Crossref | PubMed
  58. Eulert-Grehn JJ, Starck C, Kempfert J, et al. ECMELLA 2.0 – single arterial access technique for a staged approach in cardiogenic shock. Ann Thorac Surg 2020.
    Crossref | PubMed
  59. Udesen NJ, Møller JE, Lindholm MG, et al. Rationale and design of DanGer shock: Danish-German cardiogenic shock trial. Am Heart J 2019;214:60–8.
    Crossref | PubMed
  60. Chieffo A, Dudek D, Hassager C, et al. Joint EAPCI/ACVC expert consensus document on percutaneous ventricular assist devices. Eur Heart J Acute Cardiovasc Care 2021;10:570–83.
    Crossref | PubMed
  61. Broman LM, Taccone FS, Lorusso R, et al. The ELSO Maastricht Treaty for ECLS Nomenclature: abbreviations for cannulation configuration in extracorporeal life support – a position paper of the Extracorporeal Life Support Organization. Critical Care 2019;23:36.
    Crossref | PubMed
  62. Pozzi M, Flagiello M, Armoiry X, et al. Extracorporeal life support in the multidisciplinary management of cardiogenic shock complicating acute myocardial infarction. Catheter Cardiovasc Interv 2020;95:e71–7.
    Crossref | PubMed
  63. Muller G, Flecher E, Lebreton G, et al. The ENCOURAGE mortality risk score and analysis of long-term outcomes after VA-ECMO for acute myocardial infarction with cardiogenic shock. Intensive Care Med 2016;42:370–8.
    Crossref | PubMed
  64. Lackermair K, Brunner S, Orban M, et al. Outcome of patients treated with extracorporeal life support in cardiogenic shock complicating acute myocardial infarction: 1-year result from the ECLS–Shock study. Clin Res Cardiol 2021;110:1412–20
    Crossref | PubMed
  65. Brunner S, Guenther SPW, Lackermair K, et al. Extracorporeal life support in cardiogenic shock complicating acute myocardial infarction. J Am Coll Cardiol 2019;73:2355–7.
    Crossref | PubMed
  66. Bavaria JE, Ratcliffe MB, Gupta KB, et al. Changes in left ventricular systolic wall stress during biventricular circulatory assistance. Ann Thorac Surg 1988;45:526–32.
    Crossref | PubMed
  67. Strauer BE, Beer K, Heitlinger K, Höfling B. Left ventricular systolic wall stress as a primary determinant of myocardial oxygen consumption: comparative studies in patients with normal left ventricular function, with pressure and volume overload and with coronary heart disease. Basic Res Cardiol 1977;72:306–13.
    Crossref | PubMed
  68. Lucas SK, Schaff HV, Flaherty JT, et al. The harmful effects of ventricular distention during postischemic reperfusion. Ann Thorac Surg 1981;32:486–94.
    Crossref | PubMed
  69. Russo JJ, Aleksova N, Pitcher I, et al. Left ventricular unloading during extracorporeal membrane oxygenation in patients with cardiogenic shock. J Am Coll Cardiol 2019;73:654–62.
    Crossref | PubMed
  70. Al-Fares AA, Randhawa VK, Englesakis M, et al. Optimal strategy and timing of left ventricular venting during veno-arterial extracorporeal life support for adults in cardiogenic shock: a systematic review and meta-analysis. Circ Heart Fail 2019;12:e006486.
    Crossref | PubMed
  71. Li Y, Yan S, Gao S, et al. Effect of an intra-aortic balloon pump with venoarterial extracorporeal membrane oxygenation on mortality of patients with cardiogenic shock: a systematic review and meta-analysis†. Eur J Cardiothorac Surg 2019;55:395–404.
    Crossref | PubMed
  72. Schrage B, Becher PM, Bernhardt A, 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;142:2095–106.
    Crossref | PubMed
  73. Yannopoulos D, Bartos J, Raveendran G, et al. Advanced reperfusion strategies for patients with out-of-hospital cardiac arrest and refractory ventricular fibrillation (ARREST): a phase 2, single centre, open-label, randomised controlled trial. Lancet 2020;396:1807–16.
    Crossref | PubMed
  74. Stegman BM, Newby LK, Hochman JS, Ohman EM. Post-myocardial infarction cardiogenic shock is a systemic illness in need of systemic treatment: is therapeutic hypothermia one possibility? J Am Coll Cardiol 2012;59:644–7.
    Crossref | PubMed
  75. Fuernau G, Beck J, Desch S, et al. Mild hypothermia in cardiogenic shock complicating myocardial infarction. Circulation 2019;139:448–57.
    Crossref | PubMed
  76. Jacquot A, Lepage X, Merckle L, et al. Protocol for a multicentre randomised controlled trial evaluating the effects of moderate hypothermia versus normothermia on mortality in patients with refractory cardiogenic shock rescued by venoarterial extracorporeal membrane oxygenation (VA-ECMO) (HYPO-ECMO study). BMJ Open 2019;9:e031697.
    Crossref | PubMed
  77. De Backer D, Biston P, Devriendt J, et al. Comparison of dopamine and norepinephrine in the treatment of shock. N Engl J Med 2010;362:779–89.
    Crossref | PubMed
  78. Levy B, Clere-Jehl R, Legras A, et al. Epinephrine versus norepinephrine for cardiogenic shock after acute myocardial infarction. J Am Coll Cardiol 2018;72:173–82.
    Crossref | PubMed
  79. Schrage B, Beer BN, Savarese G, et al. Eligibility for mechanical circulatory support devices based on current and past randomised cardiogenic shock trials. Eur J Heart Fail 2021; 23:1942–51.
    Crossref | PubMed
  80. White HD, Assmann SF, Sanborn TA, et al. Comparison of percutaneous coronary intervention and coronary artery bypass grafting after acute myocardial infarction complicated by cardiogenic shock: results from the Should We Emergently Revascularize Occluded Coronaries for Cardiogenic Shock (SHOCK) trial. Circulation 2005;112:1992–2001.
    Crossref | PubMed
  81. Alexander JH, Reynolds HR, Stebbins AL, et al. Effect of tilarginine acetate in patients with acute myocardial infarction and cardiogenic shock: the TRIUMPH randomized controlled trial. JAMA 2007;297:1657–66.
    Crossref | PubMed
  82. Dzavík V, Cotter G, Reynolds HR, et al. Effect of nitric oxide synthase inhibition on haemodynamics and outcome of patients with persistent cardiogenic shock complicating acute myocardial infarction: a phase II dose-ranging study. Eur Heart J 2007;28:1109–16.
    Crossref | PubMed
  83. Fuhrmann JT, Schmeisser A, Schulze MR, et al. Levosimendan is superior to enoximone in refractory cardiogenic shock complicating acute myocardial infarction. Crit Care Med 2008;36:2257–66.
    Crossref | PubMed
  84. Tousek P, Rokyta R, Tesarova J, et al. Routine upfront abciximab versus standard periprocedural therapy in patients undergoing primary percutaneous coronary intervention for cardiogenic shock: the PRAGUE-7 Study. An open randomized multicentre study. Acute Card Care 2011;13:116–22.
    Crossref | PubMed
  85. Thiele H, Sick P, Boudriot E, et al. Randomized comparison of intra-aortic balloon support with a percutaneous left ventricular assist device in patients with revascularized acute myocardial infarction complicated by cardiogenic shock. Eur Heart J 2005;26:1276–83.
    Crossref | PubMed
  86. Burkhoff D, Cohen H, Brunckhorst C, O’Neill WW. A randomized multicenter clinical study to evaluate the safety and efficacy of the TandemHeart percutaneous ventricular assist device versus conventional therapy with intraaortic balloon pumping for treatment of cardiogenic shock. Am Heart J 2006;152:469.e1–8.
    Crossref | PubMed
  87. Thiele H, Zeymer U, Neumann FJ, et al. Intraaortic balloon support for myocardial infarction with cardiogenic shock. N Engl J Med 2012;367:1287–96.
    Crossref | PubMed
  88. Varshney AS, Berg DD, Katz JN, et al. Use of temporary mechanical circulatory support for management of cardiogenic shock before and after the united network for organ sharing donor heart allocation system changes. JAMA Cardiol 2020;5:703–8.
    Crossref | PubMed
  89. Thiele H, Ohman EM, Desch S, et al. Management of cardiogenic shock. Eur Heart J 2015;36:1223–30.
    Crossref | PubMed