Abstract
Despite the rapidly evolving evidence base in modern cardiology, progress in the area of cardiogenic shock remains slow, with short-term mortality still reaching 40-50%, relatively unchanged in recent years. Despite advances with an increase in the number of clinical trials taking place in this admittedly difficult-to-study area, the evidence base on which we make day-to-day decisions in clinical practice remains relatively sparse. With only definitive evidence for early revascularisation and the relative ineffectiveness of intra-aortic balloon pumping, most aspects of patient management are based on expert consensus, rather than randomised controlled trials. This updated 2020 review will outline the management of CS mainly after acute myocardial infarction with major focus on state-of-the-art treatment based on randomised clinical trials or matched comparisons if available.
Introduction
Left or right ventricular failure subsequent to acute myocardial infarction (AMI) remains the most frequent cause of cardiogenic shock (CS), accounting for approximately 80% of cases. Mechanical complications of AMI represent less frequent causes of CS (ventricular septal rupture [4%], free wall rupture [2%], and acute severe mitral regurgitation [7%]) 1. Given the relatively heterogeneous nature of the causes of non-infarct-related CS in comparison to AMI-CS, such as decompensated acute on chronic heart failure, valvular heart disease, acute myocarditis, Takotsubo syndrome, and arrhythmias 2, treatment of each potential underlying causative factor will not be addressed in detail, but overarching guiding principles will be elucidated.
The incidence of CS complicating AMI has historically been found to be of the order of 8% for STEMI, and 5% for NSTEMI 3. Although there has been some degree of disagreement in recent registries with regard to the rate of change of this figure, and whether it is in fact increasing or decreasing, overall, the most recent data do not differ markedly 4 5. Despite evidence-based therapeutic advances, foremost coronary revascularisation with subsequent survival benefit, mortality rates still remain unacceptably high at 40-50% 6 7. Indeed, with an ageing population coupled with an increase in comorbidities, there are data to suggest increasing mortality rates 8 9. These observations appear logical in the setting of an increased prevalence of diabetes mellitus and renal disease. In the setting of the recent COVID-19 pandemic, the picture is heterogeneous 10. Admissions for AMI in general were significantly reduced in Italy, along with an increase in complications including CS-AMI 11. However, in Denmark and Austria, no change in the rates and mortality for CS-AMI was noted. This may, however, reflect the severity of the COVID-19 situation in each country during the period for which data were collated 12 13.
The underlying causes, pathophysiology, and treatment of AMI-CS have been reviewed previously 2 14. This 2020 update will focus on evidence-based therapeutic management of AMI-CS with major emphasis on current guideline recommendations, revascularisation strategies, intensive care unit (ICU) treatment, adjunctive medication, and mechanical circulatory support (MCS) devices. The main focus is set on randomised controlled trials (RCT) or, where not available, on relevant matched comparisons. Furthermore, major research areas and gaps in evidence will be elucidated.
Definition of cardiogenic shock
The central pathophysiological hallmark of CS is critical end-organ hypoperfusion and hypoxia due to reduced cardiac output as a consequence of primary cardiac disorders 2. The diagnosis of CS can be made in routine practice on the basis of clinical criteria reflective of persistent hypoperfusion without adequate response to volume replacement as manifested by cold extremities, oliguria, or altered mental status in combination with biochemical manifestations of inadequate tissue perfusion such as elevated arterial lactate.
Most accepted definitions of CS include specific blood pressure parameters, generally a systolic blood pressure <90 mmHg for ≥30 minutes, or mechanical/pressor support required to maintain values above this. It should be noted, however, that in certain cases compensatory mechanisms may preserve blood pressure through vasoconstriction, while at the same time tissue perfusion and oxygenation may be significantly decreased, the so-called “normotensive CS” 14. Urine output <30 ml/hr and arterial lactate >2.0 mmol/l satisfy the criteria for clinical and biochemical manifestations of inadequate tissue perfusion.
Although not mandatory in clinical practice, objective haemodynamic parameters such as reduced cardiac index and increased pulmonary capillary wedge pressure are helpful for diagnosis confirmation, and are essential in terms of more clearly defining the status of the right heart and systemic vasculature, particularly in those patients who do not respond in the expected manner to initial therapy. The pulmonary artery pulsatility index (PAPI) may have a particular role for right ventricular function assessment here 15.
Prognosis assessment by biomarkers and scores
Understanding of the complexity of CS has evolved over the last decades and in recent years additional insights have been gained in the understanding of CS severity and phenotyping such as left, right or biventricular predominance. In general terms, there is a profound depression of myocardial contractility resulting in a potentially deleterious downward spiral of reduced cardiac index, low blood pressure, hypoperfusion and maladaptive cycles of ischaemia, inflammation, initial vasoconstriction and later vasodilation, culminating in various degrees of multiorgan failure with subsequent death if untreated.
Although not specific to CS, arterial lactate has been shown to be strongly predictive of mortality. Evidence for lactate in CS had been relatively sparse. A recent sub-analysis of the Intraaortic Balloon Pump in Cardiogenic Shock II (IABP-SHOCK II) trial and registry attempted to clarify the prognostic value for mortality of lactate clearance versus single measurements at admission and after 8 hours in AMI-CS 16. The 8-hour value was a significantly better predictive parameter than baseline or lactate clearance. Lactate at 8 hours ≥3.1 mmol/l and clearance <-3.45%/hr remained independently predictive for time to death. Another new score (CLIP score) based on biomarkers has recently been introduced. Based on the CULPRIT-SHOCK biomarker substudy, the four most relevant prognostic biomarkers are cystatin C, lactate, interleukin-6 and NT-proBNP 17. The CLIP score outperformed clinical CS scores and is objective without requirement for input of subjective parameters. Multiple other novel biomarkers - involving metabolomics, proteomics, genomics and transcriptomics - in addition to lactate and creatinine measuring the degree of inflammation, renal function, and liver involvement, respectively, have been shown to be associated with impaired prognosis (Table 1).
Table 1. Biomarkers and their relevance in cardiogenic shock 59, 93, 94.
“Omics” | Tests | Current applications | Current candidates | Current clinical value |
Metabolomics | Biochemical tests | Follow-up and risk scores | Glucose and lactate | Indicators of disease severity/prognostication |
Genomics | GWAs (nil to date) and sequencing | Genotype-based risk prediction and discovery of gene candidates | RANTES-403G/A, TNF-α308a>g, IFN-γ874t>a, MnSOD Ala16Val | Fixed from birth, risk prediction, easy to obtain, minimal measurement error |
Transcriptomics | RNAseq and microarrays | Biomarker discovery and drug safety assessment | miRNAs and LncRNAs | No predictive value (to date) |
Secretomics (extracellular vesicles) | Ultracentrifugation and size-exclusion chromatography | Biomarker discovery and pathogenesis | Leukocyte and platelet-derived EVs | Potential indicators of disease severity |
Peptides and proteins | ELISA assays | Biomarker discovery and pathogenesis | Cardiac: BNP, NT-proBNP, cTn, ST2.
Non-cardiac: IFN-γ, TNF-α, MIP-1β, Ang-1 and Ang-2, G-CSF, Th17/Treg, FGF-23, OPG, GDF-15, MCP-1β, I-FABP, L-FABP, Catalytic iron, Syndecan-1, CS4P (B2M, ALDOB, L-FABP, IC1) |
Indicators of disease severity, prognosis and clinical outcomes |
In this Table, the current landscape of biomarker utilisation in cardiogenic shock is outlined, classified by “-omics” type. ALDOB: aldolase B; Ang-1 and Ang-2: angiopoietin-1 and -2; AUC: area under the curve on receiver operating plot; B2M: beta-2-microglobulin; BNP: brain natriuretic peptide; CS4P: biomarker panel consisting of B2M, ALDOB, L-FABP and IC1; cTn: cardiac troponin; EVs: extracellular vesicles; FGF23: fibroblast growth factor 23; G-CSF: granulocyte colony-stimulating factor; GDF15: growth-differentiation factor 15; GWAs: Genome-Wide Association studies; I-FABP: intestinal fatty acid binding protein; IC1: plasma protease C1 inhibitor; IFN-γ: interferon-gamma; L-FABP: liver fatty acid binding protein; LncRNAs: long non-coding RNAs; MCP-1β: monocyte chemoattractant protein-1β; MIP-1β: macrophage inflammatory protein-1β; miRNAs: microRNAs; MnSOD: manganese superoxide dismutase; NT-proBNP: N-terminal prohormone of brain natriuretic peptide; OPG: osteoprotegerin; RANTES-403G/A: regulated on activation, normal T cell expressed and secreted polymorphism; ST2: interleukin-1 receptor-like 1; TNF-α: tumour necrosis factor alpha |
Clinical and biological factors used for prognosis assessment have been summarised in multiple scores in the I) pre-shock, II) full CS, and III) venoarterial extracorporeal membrane oxygenation (VA-ECMO) settings previously 18.
Of these, the IABP-SHOCK II score, which was developed exclusively in AMI-CS patients, incorporates the biomarkers lactate, creatinine and glucose, and has both internal and external validation 19. The SCAI CS classification with the stages A (at risk), B (beginning), C (classic), D (deteriorating), and E (extremis) was developed based on clinical considerations by expert consensus for both AMI-CS and non-AMI-CS 20. It has been validated in several retrospective cohorts and one prospective study (Table 2) 21 22 23 24 25 26 27 28. Overall, on the basis of these data, the SCAI classification appears to fulfil its goal as a tool correlating well with prognosis. The SCAI score may have its advantage as a dynamic score evaluating the course of CS over time but is currently not well suited as a numerical immediate score for decision making in the cath lab. Furthermore, the SCAI shock classification validation is not always very objective because many variables were not available for score validation, leading to subjective classification. In addition, as for biomarkers alone, none of the scores (including SCAI and IABP-SHOCK II) have been used so far to guide CS therapy.
Table 2. Characteristics of studies validating the association between SCAI shock stage and mortality. Duplicate data from the same cohort are not shown.
Study | Population | Design | Patients (n) | Primary outcome |
Jentzer, JACC 2019 26 | CICU | Retrospective single-centre | 10,004 | In-hospital mortality |
Jentzer, AHJ 2020 21 | CICU survivors | Retrospective single-centre | 9,096 | Post-discharge survival |
Schrage, CCI 2020* 22 | CS or large MI | Retrospective single-centre | 1,007 | 30-day survival |
Baran, CCI 2020 25 | CS | Prospective single-centre | 166 | 30-day survival |
Thayer, Circ HF 2020 27 | CS | Retrospective multicentre | 1,414 | In-hospital mortality |
Hanson, CCI 2020 24 | AMICS | Retrospective multicentre | 300 | Survival to discharge |
Pareek, CCI 2021 23 | OHCA | Retrospective single-centre | 393 | 30-day mortality |
Jentzer, EHJ ACC 2021* 28 | CS | Retrospective single-centre | 934 | 30-day survival |
Lawler, CCM 2021; in press | CICU or CS | Retrospective multicentre | 1,991 | In-hospital mortality |
*CS patients from Schrage et al Catheter Cardiovasc Interv 2020 were included in Jentzer et al Eur Heart J Acute Cardiovasc Care 2021, so only the non-duplicated patients are reported for Jentzer et al. |
Management and treatment
The Central illustration provides a general overview of the necessary steps in assessment and management of the AMI-CS patient. Figure 1 provides an up-to-date summary of RCT in AMI-CS and the respective mortality indicating relative risk (RR) and 95% confidence interval (CI).
SYSTEMS OF CARE
Where local protocol and logistical feasibility allow, patients with CS should be treated at specialised tertiary CS care centres, with the ability to start and escalate MCS, and dedicated cardiac ICU and cardiac surgery facilities on site 2 29. Studies in the USA have shown that mortality rates are lower in those centres with the highest quartile of mean annual CS case volume, even when controlling for early revascularisation, and that lower-volume hospitals were less likely to offer aggressive and specific treatments for CS 30. This holds true both for the case of CS-AMI and for CS due to other causes 31. Furthermore, the collaboration of an expert “Shock Team” at these centres, consisting generally of an interventional cardiologist, cardiac surgeon, advanced heart failure cardiologist and cardiac intensivist, has been shown to be an independent factor in improving outcomes 32. At a bare minimum, when transfer to a level one centre is not immediately logistically feasible, the patient should be brought to a centre with 24/7 PCI facilities. The definition of “immediate” will of course need to take local circumstances into account, but a recommended transfer time of <120 minutes has been proposed 29.
REVASCULARISATION
The key issues surrounding revascularisation for CS-AMI are summarised in this section with important interim developments addressed. Although nowadays the results of the SHOCK trial 33, which compared early revascularisation with initial medical stabilisation, may be viewed in a different light in that it failed to meet its primary 30-day endpoint of decreased mortality in the early revascularisation group compared to the medically managed group, the long-term results showing reduced mortality at 6 months, 1 year and 6 years 34, as well as the evidence borne out in subsequent registries, appear to justify the Class IB recommendation for early revascularisation 35 36 37. In summary, as in the case for AMI without CS, early revascularisation is key; multiple registries have shown a delay in revascularisation in the setting of cardiogenic shock to be associated with poorer outcomes 38 39.
The vast majority (between 70 and 80%) 18 of patients who present with AMI-CS have multivessel coronary disease. Until the results of the randomised, multicentre Culprit Lesion Only PCI versus Multivessel PCI in Cardiogenic Shock (CULPRIT-SHOCK) trial 7, there was a dearth of evidence to guide decision making. However, both the 30-day and one-year results of this trial clarify that there is significant net clinical benefit to culprit-only revascularisation, driven principally by a difference in mortality. This was consistent across all subgroups 40. Specifically, the rate of death and renal replacement therapy, as a composite endpoint, in the culprit lesion-only PCI group was 45.9%, compared to 55.4% in the multivessel PCI group (RR 0.83, 95% CI: 0.71-0.96; p=0.01) including a significant mortality reduction. Importantly, the majority of surviving patients in CULPRIT-SHOCK underwent staged protocol-recommended revascularisation during follow-up in the initial culprit lesion-only PCI group. Thus, the preferred revascularisation strategy is culprit lesion PCI with subsequent staged revascularisation after clinical stabilisation similar to the ST-elevation myocardial infarction (STEMI) setting without CS.
Current guidelines recommend early revascularisation by PCI or coronary artery bypass grafting (CABG) depending on coronary anatomy and amenability to PCI 35 36 37. Based on evidence from four observational reports, comparing PCI versus CABG, the type of revascularisation did not influence the outcome in AMI-CS, and thus there is little evidence to recommend one strategy in preference to the other 41. A trial of culprit lesion-only PCI with staged revascularisation versus immediate CABG in patients with multivessel disease and CS may clarify matters and is currently in development in the USA and in Germany.
ACCESS SITE
Current guidelines recommend radial access as default strategy in non-shock STEMI 36 or non-ST-elevation acute coronary syndromes (NSTE-ACS) 37, and also stable coronary artery disease 35, on the basis of a survival benefit and a lower risk of vascular complications. In CS, the benefit does not have the same breadth of evidence, but that which is available appears to favour radial access. Radial access was associated with a reduction in all-cause mortality in a meta-analysis of observational data analysing 8,131 patients with AMI-CS 42. Recent data from the CULPRIT-SHOCK trial confirmed the benefit of the radial approach in CS with lower mortality at 30-day follow-up (37.3% vs 53.2%, adjusted odds ratio [aOR] 0.57, 95% CI: 0.34-0.96) 43. Where the femoral approach is chosen, use of ultrasound-guided access may reduce bleeding complications 44.
PERI-INTERVENTIONAL ANTIPLATELET AND ANTITHROMBOTIC MEDICATIONS
There is a lack of RCT-derived evidence for antiplatelet and antithrombotic medication specifically in the setting of CS, and thus all recommendations are derived from more general AMI trials. Caveats to be aware of include the presence of impaired enteral resorption in CS for oral antiplatelets, often potentiated by the co-administration of opioids. The ongoing Dual Antiplatelet Therapy for Shock patients with Acute Myocardial Infarction (DAPT-SHOCK-AMI) trial (ClinicalTrials.gov: NCT03551964), assessing intravenous cangrelor versus crushed oral antiplatelets, may shed light on this matter. The benefit of routine upstream use of glycoprotein IIb/IIIa inhibitors has not been shown to be superior to standard treatment (Figure 1) 45. These considerations are discussed further in the recent ESC position paper on antithrombotic therapy in patients with ACS complicated by CS or out-of-hospital cardiac arrest (OHCA) 46.
INTENSIVE CARE UNIT TREATMENT
FLUIDS, VASOPRESSORS, INOTROPES
Due to the complexity of most CS presentations, these patients are best treated in specialised ICUs, allowing close monitoring of volume status, vasopressor and inotropic support, and the prophylaxis and treatment of multiorgan dysfunction syndrome (MODS) 2 29. Fluid administration in CS is based mainly on pathophysiological considerations and, according to current guidelines, a fluid challenge as first-line therapy should be considered unless there are signs of overt fluid overload (class 1C recommendation). Despite the frequency with which inotropes and vasopressors are administered in patients in CS (approximately 90%) 6, it should be remembered these drugs increase myocardial oxygen consumption and vasoconstriction, and may impair microcirculation and increase afterload. Thus, as a general rule they should be administered at the lowest possible dose for the shortest possible duration.
In the SOAP II trial 47, involving 1,679 shock patients, 280 of whom had CS, norepinephrine appeared favourable compared to dopamine given the propensity of dopamine to cause arrhythmia in the overall cohort. Furthermore, when only the CS subgroup was considered, this group was noted to have a lower mortality with norepinephrine. As shown in the OPTIMA-CC trial in AMI-CS, norepinephrine also appears favourable over epinephrine in terms of minimising both metabolic changes (including lactic acidosis) and heart rate, without having an appreciable difference in effect on cardiac index 48. Indeed, there was in fact more refractory CS with epinephrine than norepinephrine (37% vs 7%; p=0.008), leading to the trial being terminated early. Thus, norepinephrine appears to be the vasoconstrictor of choice in refractory CS, reflected in ESC recommendations, albeit with a class IIb B recommendation. There are no data on vasopressin in the CS setting.
The target mean arterial pressure (MAP) is not well defined in CS. Previously, in contrast to the recommendations for septic shock, consensus cautioned aiming for a MAP >65 mmHg as it had been shown to be potentially associated with more side effects 49. However, a recent combined analysis from two RCT assessing two different MAP goals in patients after OHCA showed lower indirect infarct size as measured by cardiac troponin in the subgroup of AMI-CS patients randomised to a target MAP of 85-100 mmHg 50. This is currently only hypothesis-generating and requires a large-scale RCT to prove the optimal MAP in CS.
Inotropes, e.g., dobutamine, may be given simultaneously with norepinephrine in an attempt to improve cardiac contractility (class IIb, level of evidence C) 51. Despite pathophysiological considerations potentially favouring the mechanism of action of inodilators such as levosimendan or phosphodiesterase inhibitors, their current evidence base is very limited. Proof of concept was suggested in a very small trial of 32 CS patients with lower mortality in levosimendan in comparison to enoximone (Figure 1) 52. However, to date, these findings have not been replicated in larger-scale studies 53 54 55 56.
GENERAL INTENSIVE CARE MEASURES
Although there is no specific evidence base in CS, ICU treatment should follow general best practice guidelines, with regard to lung protective ventilation (6 ml/kg predicted body weight tidal volume), thromboprophylaxis, stress-ulcer prophylaxis, glycaemic control, and nutritional supplementation.
Indications for renal replacement therapy do not differ from those in the general ICU setting (uraemia, refractory volume overload, metabolic acidosis [pH <7.2] and refractory hyperkalaemia [>6.0 mmol/l]). There is also now conclusive evidence from two large RCT showing no benefit of earlier initiation of renal replacement therapy 57 58.
Elevated liver transaminases, as a surrogate for liver hypoperfusion, have been shown to be associated with increased mortality, and so care should be taken to stabilise haemodynamics in this setting 59.
BLEEDING IN CARDIOGENIC SHOCK
Moderate/severe bleeding is common in CS, ranging from 20-90% depending on the definition used, and is also influenced by concomitant use of MCS 60. A recent pre-specified post hoc analysis of the CULPRIT-SHOCK trial revealed that a total of 21.5% of patients with AMI-CS experienced at least one bleeding event up to 30 days 61. Most frequent were Bleeding Academic Research Consortium (BARC) category 3a bleeding events (33%); 5.4% of bleeding events were fatal. Treatment with active MCS by VA-ECMO or Impella™ (Abiomed, Danvers, MA, USA) emerged as the major risk factor for bleeding, as was noted in a 2014 meta-analysis 62. Next to the requirement for significantly larger access sheaths, the risk of bleeding may be increased both by consumptive coagulopathy and by acquired platelet dysfunction in the setting of high shear stress in case of MCS use. Use of glycoprotein IIb/IIIa inhibitors was also found on multivariate testing to be a statistically significant predictor of bleeding. Bleeding was associated with a significantly higher mortality at 30 days (hazard ratio [HR] 2.11, 95% CI: 1.63-2.75; p<0.0001) and, when patients with only severe bleeding events were compared to those without, the HR was even higher (HR 2.80, 95% CI: 1.94-4.05; p<0.0001).
Trials in non-CS patients with bleeding demonstrated that a restrictive transfusion regimen can improve outcome. Generally accepted ICU strategies avoid correction of haemoglobin levels >7 g/dl (>4.3 mmol/l) unless there is a clinical bleeding problem. Transfusion of stored blood serves, on top of the pre-existing systemic inflammatory state in CS, as a source of further inflammation, with alterations in normal nitric oxide biology contributing to vasoconstriction, platelet aggregation and impaired oxygen delivery.
HYPOTHERMIA
Target temperature management is part of standard of care in OHCA and should therefore be a basic element in the treatment of CS patients suffering cardiac arrest. However, there is little clinical trial evidence in the CS case itself. Theoretical assumptions suggested a possible beneficial effect of hypothermia in non-resuscitated CS patients; this was not borne out in the SHOCK-COOL trial including 40 patients. In fact, lactate clearance was found to be impaired, suggesting even harm 63.
MECHANICAL COMPLICATIONS
A detailed description of mechanical complications is beyond the scope of this review and has been summarised previously 64. However, given the presence of moderate or greater mitral regurgitation in 5-10% of patients with CS, of note is some recent work on percutaneous mitral valve repair (PMVr) using edge-to-edge repair as salvage therapy in patients with mitral regurgitation and refractory CS. A recent multicentre pooled patient-level systematic review looked at 141 patients at 14 institutions who presented with SCAI stage B-E CS and 3+ or 4+ mitral regurgitation - not necessarily as a consequence of AMI - and were treated with PMVr 65. No periprocedural complications were reported; successful PMVr was achieved in about 89% of cases. When stratified by procedural results, successful PMVr reduced rates of in-hospital (HR 0.36, 95% CI: 0.13-0.98; p=0.04) and 90-day mortality (HR 0.36, 95% CI: 0.16-0.78; p=0.01).
MECHANICAL CIRCULATORY SUPPORT
Although the theoretical concept of MCS is appealing, in terms of reducing the dependency on inotropes/vasopressors while the heart is bridged to recovery or decision, at present there exist only limited data derived from RCT based on clinical outcomes.
INTRA-AORTIC BALLOON PUMPING
Despite decades of familiarity with the IABP, only in recent years has conclusive evidence established a lack of benefit in AMI-CS. The IABP-SHOCK II trial randomised 600 patients with AMI-CS to early revascularisation with or without IABP 6. No difference in the primary study endpoint of 30-day mortality was noted, or in terms of any of the trial’s secondary endpoints. Longer-term follow-up, up to six years, clarified that these results were sustained, and that they held for the intention-to-treat as well as for the as-treated group 66 67. Thus, nowadays IABP may only be considered for the mechanical complications group (III B recommendation without and IIb C recommendation in the setting of mechanical complication) 35 36 51.
The neutral results of IABP-SHOCK II together with the downgrading in guidelines led to a decrease in IABP use to <30% in the USA 68, <10% in Germany 69, and <2% in Denmark 70, respectively. The decline in IABP use was associated with an increase of active MCS including Impella/TandemHeart™ (LivaNova, London, United Kingdom) and VA-ECMO in CS from approximately 1% in 2006 to 8% in 2014 in the USA 68. Similarly, VA-ECMO implementation has developed towards a routine procedure with a more than eightfold increase in Germany from 2010 to 2015 71 72.
ACTIVE PERCUTANEOUS LEFT VENTRICULAR MCS
The mechanism of action of various MCS devices has been outlined previously 18 73. New developments include the Impella ECP (expandable CP) device with the ability to achieve peak flows >3.5 L/min despite only requiring access though a 9 Fr sheath. It is unsheathed in the descending aorta and expands to approximately 18 Fr. When being removed, it is first resheathed back down to 9 Fr. Figure 2 shows the currently available devices, including a brief overview of technical features and unloading properties on the left and/or right ventricle.
Randomised data on clinical outcomes with regard to the use of percutaneous MCS devices in CS are still limited (Figure 3A). A meta-analysis of active MCS devices against control showed no difference in mortality for the 148 included patients. There were improvements in arterial lactate and MAP after device insertion. On the other hand, there were no effects on other haemodynamic parameters and, more importantly, haemodynamic effects were counterbalanced by significantly more bleeding complications 60. A more recent small (n=15) trial in AMI-CS (IMPELLA-STIC) attempted to assess the potential additional benefit of Impella 5.0 LP in patients already managed with an IABP 74. There was no difference in the primary endpoint, change in cardiac power index at 12 hours. Adverse events, especially major bleeding, were common in the Impella + IABP group (0% vs 71.4%).
In the setting of a lack of sufficient data from RCT, matched comparisons provide the best second option evidence. A matched-pair mortality analysis of 237 Impella-treated versus 237 IABP-treated AMI-CS patients confirmed a lack of mortality benefit with the Impella device (30-day mortality 48.5% vs 46.4%, p=0.64) 75. Of note, severe or life-threatening bleeding (8.5% vs 3.0%, p<0.01) and peripheral vascular complications (9.8% vs 3.8%, p=0.01) were observed more frequently with the Impella device. A second propensity-matched analysis, involving 1,680 pairs of AMI-CS patients, added further weight to this observation, actually suggesting potential harm with the use of Impella rather than IABP 76. Among the pairs, there was a significantly higher risk of in-hospital death associated with use of Impella versus IABP (absolute risk difference 10.9%, 95% CI: 7.6-14.2%; p<0.001) and a higher risk of in-hospital major bleeding (absolute risk difference 15.4%, 95% CI: 12.5-18.2%; p<0.001). These associations were consistent regardless of whether patients received a device before or after initiation of PCI.
A third recent publication assessed 4,782 patients undergoing PCI treated with MCS in the USA 77. CS was present in 50%. After propensity adjustment, and accounting for clustering of patients by hospitals, Impella use compared to IABP was associated with an increased risk of death (OR 1.24, 95% CI: 1.13-1.36), bleeding (OR 1.10, 95% CI: 1.00-1.21) and stroke (OR 1.34, 95% CI: 1.18-1.53). Interestingly, patients treated by Impella in comparison to IABP were less sick. Thus, a selection bias inherent to any observational data is less likely to be the cause of higher mortality with the Impella device.
Taken together, these results suggest that very careful patient selection for Impella is warranted, particularly in terms of weighing up the haemodynamic benefits against potential device-related complications. Due to the retrospective and non-randomised nature of these studies, it is possible, even allowing for complex statistical propensity matching, that unmeasured confounding is occurring. The DanGer clinical trial (NCT01633502), currently ongoing, with the aim of recruiting 360 participants with STEMI complicated by CS and randomising them to either treatment with the Impella CP or conventional guideline-driven treatment, may shed some additional light on the role of Impella 78. The trial protocol stipulates that the device should be placed prior to PCI and has a hard clinical endpoint of all-cause mortality. Patients with OHCA who remain comatose after return of spontaneous circulation (ROSC) are excluded. Over 200 patients have been included to date. Patients recruited thus far are profoundly unwell, with 100% of patients having a lactate >2.5 mmol/L and median LVEF 20%. This trial will provide valuable information on this difficult-to-treat cohort but may limit generalisability to the entire CS spectrum because usually 50% of CS patients have undergone resuscitation.
EXTRACORPOREAL MEMBRANE OXYGENATION
In initial iterations, surgical insertion of ECMO was associated with substantial complications such as lower extremity ischaemia (16.9%), compartment syndrome (10.3%), amputation (4.7%), stroke (5.9%), major bleeding (40.8%), and significant infections (30.4%) 62. The development of miniaturised systems and percutaneous cannula insertion has led to a significant uptake of VA-ECMO by interventional cardiologists for CS treatment. It offers the advantages of low costs in comparison to other percutaneous MCS devices, high flow providing full circulatory support even in resuscitation situations, the ability to provide full oxygenation, and also combined support of the right and left ventricle.
Outcome data on VA-ECMO in CS remain relatively sparse. A significant mortality benefit with VA-ECMO use has been shown in a single meta-analysis, which relied only on four small observational studies 79. In CS without ongoing cardiopulmonary resuscitation (CPR), VA-ECMO resulted in a 33% higher 30-day survival compared to control (95% CI: 14-52%; p<0.001; number needed to treat 3) 79.
A single small RCT (n=42) has assessed the efficacy of VA-ECMO in AMI-CS in terms of left ventricular recovery at 30 days 80. Left ventricular ejection fraction at 30 days was not found to be significantly different amongst surviving patients in the VA-ECMO and control groups (p=0.86). All-cause 30-day mortality was low and not different between groups, questioning the inclusion of severe AMI-CS patients (19% VA-ECMO vs 33% control, p=0.37).
Currently, further RCT to assess VA-ECMO in the setting of AMI-CS are in the early or more advanced phase of patient recruitment (Figure 3B). These trials are adequately powered and most use 30-day mortality as the primary endpoint. Among them, the ECLS-SHOCK trial (NCT03637205) currently has the highest number of enrolled patients (>160) 81. It only includes high-risk CS patients after AMI with lactate >3 mmol/l and recommends VA-ECMO insertion before revascularisation. In addition, it has a dedicated protocol for ECMO venting and also an escalation strategy in the non-ECMO arm. In contrast, the EURO-SHOCK trial (approximately 10 AMI-CS patients so far) (NCT03813134) has no protocol-defined venting strategy and specifies the addition of ECMO post rather than pre PCI 82. The ANCHOR trial (NCT04184635) is still in the planning phase and will combine VA-ECMO with IABP in comparison to control (n=400). A major problem of this trial is the chosen primary study endpoint of death in the ECMO group and death or rescue ECMO in the control group. The allowance of crossover in the control arm will make interpretation of trial results difficult. The ECMO-CS trial (NCT02301819) is only powered to assess a composite endpoint of death, resuscitated cardiac arrest and implantation of another mechanical circulatory device, rather than mortality directly. Until more data are available, thorough consideration must be given to identifying appropriate candidates for VA-ECMO support to avoid unnecessary use, which might consume resources and expose patients to possible complications.
A common issue related to peripheral cannula insertion is an increase in afterload which may lead to inadequate left ventricular unloading. Multiple venting manoeuvres have been described to prevent volume overload such as combining VA-ECMO with IABP, Impella, atrial septostomy, or other (Figure 4).
A recently published multicentre cohort study assessed whether venting in patients with VA-ECMO was associated with lower mortality 83. Patients (n=225) with severe CS treated with VA-ECMO and Impella unloading (ECMELLA) were propensity matched with 225 patients treated with VA-ECMO without Impella. Left ventricular unloading was associated with lower 30-day mortality (HR 0.79, 95% CI: 0.63-0.98; p=0.03) without differences in various subgroups. However, complications were noted to occur more frequently in the venting cohort, specifically severe bleeding (HR 2.87, 95% CI: 1.92-4.35; p<0.01) and access site-related ischaemia (HR 1.96, 95% CI: 1.22-3.20; p<0.01).
This study is in agreement with previous meta-analyses which have also shown a mortality benefit with unloading VA-ECMO. Russo et al identified 17 observational studies which included 3,997 patients receiving a concomitant LV unloading strategy while on VA-ECMO (IABP 91.7%, percutaneous ventricular assist device 5.5%, pulmonary vein or transseptal left atrial cannulation 2.8%) 84. Mortality was 60% in the total cohort. The risk ratio (RR) for mortality was lower in those with venting than in those without (RR 0.79, 95% CI: 0.72-0.87; p<0.00001). There was no interaction between the specific unloading modalities and mortality. Kowalewski et al conducted a similar meta-analysis, including 7,581 patients from 62 observational studies 85. An unloading strategy was associated with a lower mortality risk (RR 0.88, 95% CI: 0.82-0.93; p<0.0001) and higher probability of VA-ECMO weaning (RR 1.35, 95% CI: 1.21-1.51; p<0.00001). A recent review article summarises all aspects of unloading in VA-ECMO use 86.
In the special situation of cardiac arrest with ongoing CPR, VA-ECMO use (ECMO cardiopulmonary resuscitation [eCPR]) was associated with an absolute 13% increase of 30-day survival compared to control (95% CI: 6-20%; p<0.001; number needed to treat 7.7) in the above-mentioned meta-analysis 79. At present, the Prague out-of-Hospital Cardiac Arrest trial (ClinicalTrials.gov: NCT01511666), assessing eCPR in refractory OHCA, is ongoing, and powered to assess superiority with regard to mortality (with good neurological outcomes) at six months.
GENERAL REFLECTIONS ON MECHANICAL CIRCULATORY SUPPORT
Appropriate patient selection for MCS remains a key consideration. Based on IABP-SHOCK II, approximately 50-60% of CS patients survive without any device 8. In these patients, device utilisation will have no impact on survival and, in the worst case scenario, may lead to complications associated with device insertion, up to and including death. Among the 40-50% not surviving, there may also be futile situations for patients with severe CS or those with anoxic brain injury, where even the best available device will be unable to change the ultimate clinical outcome. Thus, we conclude that it may be estimated that there is a key kernel of only approximately 25% of CS patients who are, in fact, appropriate candidates for MCS. Those factors best identifying the members of this group remain to be fully elucidated.
Appropriate patient selection is also influenced by the balance between efficacy, institutional experience, and device-related complications. Particularly in terms of the data on Impella, the key determinant of overall clinical benefit seems to be the balance of increased haemodynamic support versus the risk of bleeding complications. The availability of the Impella ECP may alter this dynamic, given the smaller sheath size needed to facilitate its entry. However, this remains to be seen and needs to be borne out in RCT.
Current guidelines recommend considering the use of percutaneous MCS in selected patients depending on age, comorbidities, and neurological function in particular, in refractory CS without any preference for device selection (IIa C recommendation) 35 36 51. It is important to note that the recommendation for the use of MCS only in the setting of “refractory shock” is based on limited available evidence where it is possible that the initiation of MCS at this point may, at least in some cases, be too late. The optimal timing of initiation of MCS remains a matter of debate, and has often been left to the discretion of the operator in randomised trials. Results from registry studies are conflicting, with some showing a benefit 87 88 89 90 91, whereas other larger analyses have even shown harm with pre-revascularisation insertion of Impella 76.
In terms of the role for complete revascularisation versus culprit-only PCI specifically in the setting of MCS-supported revascularisation, registry data suggest - in contrast to the large-scale randomised CULPRIT-SHOCK trial - no significant differences in survival and rates of acute kidney injury. However, if anything, this is only hypothesis-generating based on the non-randomised evidence 92. Overall, it appears that, as in non-CS STEMI, rapid diagnosis and initiation of treatment are critical, but formal RCT are required to clarify the findings to date, particularly for the case of Impella in light of the disparate data outlined above.
Conclusion
In general, RCT are difficult to perform in the CS setting and, to date, only a small number of trials have managed to recruit the required number of patients to assess key clinical outcomes. Furthermore, despite the proliferation of primary PCI networks and advances in antiplatelet and antithrombotic pharmacology, the 30-day mortality rate of CS has changed little. There is hope on the horizon, however, with more clinical trials than ever actively recruiting (Figure 3B), with the aim of clarifying the role of pharmacology and MCS. Especially with regard to Impella (DanGer), levosimendan (LevoHeartShock) and VA-ECMO (ECLS-SHOCK, EURO-SHOCK, ANCHOR), it is hoped that we will soon have more data to clarify the best management approach and that these data may lead to an improvement in short- and long-term outcomes.
Acknowledgments
Conflict of interest statement
The authors have no conflicts of interest to declare.
Abbreviations
- AMI
acute myocardial infarction
- BARC
Bleeding Academic Research Consortium
- CI
confidence interval
- CICU
cardiac intensive care unit
- CS
cardiogenic shock
- ECLS
extracorporeal life support
- ECMELLA
extracorporeal membrane oxygenation combined with Impella support
- eCPR
ECMO-assisted cardiopulmonary resuscitation
- HR
hazard ratio
- IABP
intra-aortic balloon pump
- ICU
intensive care unit
- LVAD
left ventricular assist device
- MAP
mean arterial pressure
- MCS
mechanical circulatory support
- MODS
multiorgan dysfunction syndrome
- OHCA
out-of-hospital cardiac arrest
- OR
odds ratio
- PAPI
pulmonary artery pulsatility index
- PMVr
percutaneous mitral valve repair
- RCT
randomised controlled trial
- RR
relative risk/risk ratio
- SCAI
Society for Cardiovascular Angiography and Interventions
- STEMI
ST-elevation myocardial infarction
- VA-ECMO
veno-arterial extracorporeal membrane oxygenation
Contributor Information
Holger Thiele, Heart Center Leipzig at University of Leipzig, Leipzig, Germany; Leipzig Heart Institute, Leipzig, Germany.
Suzanne de Waha-Thiele, University Heart Center Luebeck, Luebeck, Germany.
Anne Freund, Heart Center Leipzig at University of Leipzig, Leipzig, Germany; Leipzig Heart Institute, Leipzig, Germany.
Uwe Zeymer, Klinikum Ludwigshafen, Ludwigshafen, Germany.
Steffen Desch, Heart Center Leipzig at University of Leipzig, Leipzig, Germany; Leipzig Heart Institute, Leipzig, Germany.
Sean Fitzgerald, Heart Center Leipzig at University of Leipzig, Leipzig, Germany.
References
- Hochman JS, Buller CE, Sleeper LA, Boland J, Džavík V, Sanborn TA, Godfrey E, White HD, Lim J, LeJemtel T. Cardiogenic shock complicating acute myocardial infarction--etiologies, management and outcome: a report from the SHOCK Trial Registry. SHould we emergently revascularize Occluded Coronaries for cardiogenic shocK? J Am Coll Cardiol. 2000;36:1063–70. doi: 10.1016/S0735-1097(00)00879-2. [DOI] [PubMed] [Google Scholar]
- van Diepen S, Katz JN, Albert NM, Henry TD, Jacobs AK, Kapur NK, Kilic A, Menon V, Ohman EM, Sweitzer NK, Thiele H, Washam JB, Cohen MG American Heart Association Council on Clinical Cardiology; Council on Cardiovascular and Stroke Nursing; Council on Quality of Care and Outcomes Research; Mission: Lifeline. Contemporary management of cardiogenic shock: a Scientific Statement from the American Heart Association. Circulation. 2017;136:e232–e68. doi: 10.1161/CIR.0000000000000525. [DOI] [PubMed] [Google Scholar]
- McManus DD, Gore J, Yarzebski J, Spencer F, Lessard D, Goldberg RJ. Recent trends in the incidence, treatment, and outcomes of patients with STEMI and NSTEMI. Am J Med. 2011;124:40–7. doi: 10.1016/j.amjmed.2010.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lauridsen MD, Rørth R, Lindholm MG, Kjaergaard J, Schmidt M, Møller JE, Hassager C, Torp-Pedersen C, Gislason G, Køber L, Fosbøl EL. Trends in first-time hospitalization, management, and short-term mortality in acute myocardial infarction-related cardiogenic shock from 2005 to 2017: A nationwide cohort study. Am Heart J. 2020;229:127–37. doi: 10.1016/j.ahj.2020.08.012. [DOI] [PubMed] [Google Scholar]
- García-García C, Oliveras T, El Ouaddi N, Rueda F, Serra J, Labata C, Ferrer M, Cediel G, Montero S, Martínez MJ, Resta H, de Diego O, Vila J, Dégano IR, Elosua R, Lupón J, Bayés-Genís A. Short- and Long-Term Mortality Trends in STEMI-Cardiogenic Shock over Three Decades (1989-2018): The Ruti-STEMI-Shock Registry. J Clin Med. 2020;9:2398. doi: 10.3390/jcm9082398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thiele H, Zeymer U, Neumann FJ, Ferenc M, Olbrich HG, Hausleiter J, Richardt G, Hennersdorf M, Empen K, Fuernau G, Desch S, Eitel I, Hambrecht R, Fuhrmann J, Böhm M, Ebelt H, Schneider S, Schuler G, Werdan K IABP-SHOCK II Trial Investigators. Intraaortic balloon support for myocardial infarction with cardiogenic shock. N Engl J Med. 2012;367:1287–96. doi: 10.1056/NEJMoa1208410. [DOI] [PubMed] [Google Scholar]
- Thiele H, Akin I, Sandri M, Fuernau G, de Waha S, Meyer-Saraei R, Nordbeck P, Geisler T, Landmesser U, Skurk C, Fach A, Lapp H, Piek JJ, Noc M, Goslar T, Felix SB, Maier LS, Stepinska J, Oldroyd K, Serpytis P, Montalescot G, Barthelemy O, Huber K, Windecker S, Savonitto S, Torremante P, Vrints C, Schneider S, Desch S, Zeymer U CULPRIT-SHOCK Investigators. PCI Strategies in Patients with Acute Myocardial Infarction and Cardiogenic Shock. N Engl J Med. 2017;377:2419–32. doi: 10.1056/NEJMoa1710261. [DOI] [PubMed] [Google Scholar]
- Redfors B, Angerås O, Råmunddal T, Dworeck C, Haraldsson I, Ioanes D, Petursson P, Libungan B, Odenstedt J, Stewart J, Lodin E, Wahlin M, Albertsson P, Matejka G, Omerovic E. 17-year trends in incidence and prognosis of cardiogenic shock in patients with acute myocardial infarction in western Sweden. Int J Cardiol. 2015;185:256–62. doi: 10.1016/j.ijcard.2015.03.106. [DOI] [PubMed] [Google Scholar]
- Wayangankar SA, Bangalore S, McCoy LA, Jneid H, Latif F, Karrowni W, Charitakis K, Feldman DN, Dakik HA, Mauri L, Peterson ED, Messenger J, Roe M, Mukherjee D, Klein A. Temporal Trends and Outcomes of Patients Undergoing Percutaneous Coronary Interventions for Cardiogenic Shock in the Setting of Acute Myocardial Infarction: A Report From the CathPCI Registry. JACC Cardiovasc Interv. 2016;9:341–51. doi: 10.1016/j.jcin.2015.10.039. [DOI] [PubMed] [Google Scholar]
- Zeymer U, Gitt A, Thiele H. [COVID-19 pandemic: Effects on clinical care of cardiovascular patients in spring 2020]. [Article in German]. Herz. 2021;46:115–9. doi: 10.1007/s00059-020-05015-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Rosa S, Spaccarotella C, Basso C, Calabrò MP, Curcio A, Filardi PP, Mancone M, Mercuro G, Muscoli S, Nodari S, Pedrinelli R, Sinagra G, Indolfi C Società Italiana di Cardiologia and the CCU Academy investigators group. Reduction of hospitalizations for myocardial infarction in Italy in the COVID-19 era. Eur Heart J. 2020;41:2083–8. doi: 10.1093/eurheartj/ehaa409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lauridsen MD, Butt JH, Østergaard L, Møller JE, Hassager C, Gerds T, Kragholm K, Phelps M, Schou M, Torp-Pedersen C, Gislason G, Køber L, Fosbøl EL. Incidence of acute myocardial infarction-related cardiogenic shock during corona virus disease 19 (COVID-19) pandemic. Int J Cardiol Heart Vasc. 2020;31:100659. doi: 10.1016/j.ijcha.2020.100659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bugger H, Gollmer J, Pregartner G, Wünsch G, Berghold A, Zirlik A, von Lewinski D. Complications and mortality of cardiovascular emergency admissions during COVID-19 associated restrictive measures. PLoS One. 2020;15:e0239801. doi: 10.1371/journal.pone.0239801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chioncel O, Parissis J, Mebazaa A, Thiele H, Desch S, Bauersachs J, Harjola VP, Antohi EL, Arrigo M, Gal TB, Čelutkienė J, Collins SP, DeBacker D, Iliescu VA, Jankowska E, Jaarsma T, Keramida K, Lainscak M, Lund LH, Lyon AR, Masip J, Metra M, Miro O, Mortara A, Mueller C, Mullens W, Nikolaou M, Piepoli M, Price S, Rosano G, Vieillard-Baron A, Weinstein JM, Anker SD, Filippatos G, Ruschitzka F, Coats AJS, Seferovic P. 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. doi: 10.1002/ejhf.1922. [DOI] [PubMed] [Google Scholar]
- Kapur NK, Esposito ML, Bader Y, Morine KJ, Kiernan MS, Pham DT, Burkhoff D. Mechanical Circulatory Support Devices for Acute Right Ventricular Failure. Circulation. 2017;136:314–26. doi: 10.1161/CIRCULATIONAHA.116.025290. [DOI] [PubMed] [Google Scholar]
- Fuernau G, Desch S, de Waha-Thiele S, Eitel I, Neumann FJ, Hennersdorf M, Felix SB, Fach A, Böhm M, Pöss J, Jung C, Ouarrak T, Schneider S, Werdan K, Zeymer U, Thiele H. Arterial Lactate in Cardiogenic Shock: Prognostic Value of Clearance Versus Single Values. JACC Cardiovasc Interv. 2020;13:2208–16. doi: 10.1016/j.jcin.2020.06.037. [DOI] [PubMed] [Google Scholar]
- Ceglarek U, Schellong P, Rosolowski M, Scholz M, Willenberg A, Kratzsch J, Zeymer U, Fuernau G, de Waha-Thiele S, Büttner G, Jobs A, Freund A, Desch S, Feistritzer H, Isermann B, Thiery J, Pöss J, Thiele H. 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. doi: 10.1093/eurheartj/ehab110. [DOI] [PubMed] [Google Scholar]
- Thiele H, Ohman EM, de Waha-Thiele S, Zeymer U, Desch S. Management of cardiogenic shock complicating myocardial infarction: an update 2019. Eur Heart J. 2019;40:2671–83. doi: 10.1093/eurheartj/ehz363. [DOI] [PubMed] [Google Scholar]
- Pöss J, Köster J, Fuernau G, Eitel I, de Waha S, Ouarrak T, Lassus J, Harjola VP, Zeymer U, Thiele H, Desch S. Risk Stratification for Patients in Cardiogenic Shock After Acute Myocardial Infarction. J Am Coll Cardiol. 2017;69:1913–20. doi: 10.1016/j.jacc.2017.02.027. [DOI] [PubMed] [Google Scholar]
- Baran DA, Grines CL, Bailey S, Burkhoff D, Hall SA, Henry TD, Hollenberg SM, Kapur NK, O’Neill W, Ornato JP, Stelling K, Thiele H, van Diepen S, Naidu SS. SCAI clinical expert consensus statement on the classification of cardiogenic shock: This document was endorsed by the American College of Cardiology (ACC), the American Heart Association (AHA), the Society of Critical Care Medicine (SCCM), and the Society of Thoracic Surgeons (STS) in April 2019. Catheter Cardiovasc Interv. 2019;94:29–37. doi: 10.1002/ccd.28329. [DOI] [PubMed] [Google Scholar]
- Jentzer JC, Baran DA, van Diepen S, Barsness GW, Henry TD, Naidu SS, Bell MR, Holmes DR Jr. 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. doi: 10.1016/j.ahj.2019.10.012. [DOI] [PubMed] [Google Scholar]
- Schrage B, Dabboura S, Yan I, Hilal R, Neumann JT, Sorensen NA, Gossling A, Becher PM, Grahn H, Wagner T, Seiffert M, Kluge S, Reichenspurner H, Blankenberg S, Westermann D. Application of the SCAI classification in a cohort of patients with cardiogenic shock. Catheter Cardiovasc Interv. 2020;96:E213–9. doi: 10.1002/ccd.28707. [DOI] [PubMed] [Google Scholar]
- Pareek N, Dworakowski R, Webb I, Barash J, Emezu G, Melikian N, Hill J, Shah A, Maccarthy P, Byrne J. SCAI cardiogenic shock classification after out of hospital cardiac arrest and association with outcome. Catheter Cardiovasc Interv. 2021;97:E288–97. doi: 10.1002/ccd.28984. [DOI] [PubMed] [Google Scholar]
- Hanson ID, Tagami T, Mando R, Kara Balla A, Dixon SR, Timmis S, Almany S, Naidu SS, Baran D, Lemor A, Gorgis S, O’Neill W, Basir MB National Cardiogenic Shock Investigators. SCAI shock classification in acute myocardial infarction: Insights from the National Cardiogenic Shock Initiative. Catheter Cardiovasc Interv. 2020;96:1137–42. doi: 10.1002/ccd.29139. [DOI] [PubMed] [Google Scholar]
- Baran DA, Long A, Badiye AP, Stelling K. Prospective validation of the SCAI shock classification: Single center analysis. Catheter Cardiovasc Interv. 2020;96:1339–47. doi: 10.1002/ccd.29319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jentzer JC, van Diepen S, Barsness GW, Henry TD, Menon V, Rihal CS, Naidu SS, Baran DA. Cardiogenic Shock Classification to Predict Mortality in the Cardiac Intensive Care Unit. J Am Coll Cardiol. 2019;74:2117–28. doi: 10.1016/j.jacc.2019.07.077. [DOI] [PubMed] [Google Scholar]
- Thayer KL, Zweck E, Ayouty M, Garan AR, Hernandez-Montfort J, Mahr C, Morine KJ, Newman S, Jorde L, Haywood JL, Harwani NM, Esposito ML, Davila CD, Wencker D, Sinha SS, Vorovich E, Abraham J, O'Neill W, Udelson J, Burkhoff D, Kapur NK. Invasive Hemodynamic Assessment and Classification of In-Hospital Mortality Risk Among Patients With Cardiogenic Shock. Circ Heart Fail. 2020;13:e007099. doi: 10.1161/CIRCHEARTFAILURE.120.007099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jentzer JC, Schrage B, Holmes DR, Dabboura S, Anavekar NS, Kirchhof P, Barsness GW, Blankenberg S, Bell MR, Westermann D. Influence of age and shock severity on short-term survival in patients with cardiogenic shock. Eur Heart J Acute Cardiovasc Care. 2021 doi: 10.1093/ehjacc/zuaa035. [DOI] [PubMed] [Google Scholar]
- Rab T, Ratanapo S, Kern KB, Basir MB, McDaniel M, Meraj P, King SB 3rd, O'Neill W. Cardiac Shock Care Centers: JACC Review Topic of the Week. J Am Coll Cardiol. 2018;72:1972–80. doi: 10.1016/j.jacc.2018.07.074. [DOI] [PubMed] [Google Scholar]
- Shaefi S, O’Gara B, Kociol RD, Joynt K, Mueller A, Nizamuddin J, Mahmood E, Talmor D, Shahul S. Effect of cardiogenic shock hospital volume on mortality in patients with cardiogenic shock. J Am Heart Assoc. 2015;4:e001462. doi: 10.1161/JAHA.114.001462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang JI, Lu DY, Feldman DN, McCullough SA, Goyal P, Karas MG, Sobol I, Horn EM, Kim LK, Krishnan U. Outcomes of Hospitalizations for Cardiogenic Shock at Left Ventricular Assist Device Versus Non-Left Ventricular Assist Device Centers. J Am Heart Assoc. 2020;9:e017326. doi: 10.1161/JAHA.120.017326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tehrani BN, Truesdell AG, Sherwood MW, Desai S, Tran HA, Epps KC, Singh R, Psotka M, Shah P, Cooper LB, Rosner C, Raja A, Barnett SD, Saulino P, deFilippi CR, Gurbel PA, Murphy CE, O’Connor CM. Standardized Team-Based Care for Cardiogenic Shock. J Am Coll Cardiol. 2019;73:1659–69. doi: 10.1016/j.jacc.2018.12.084. [DOI] [PubMed] [Google Scholar]
- Hochman JS, Sleeper LA, Webb JG, Sanborn TA, White HD, Talley JD, Buller CE, Jacobs AK, Slater JN, Col J, McKinlay SM, LeJemtel TH. Early revascularization in acute myocardial infarction complicated by cardiogenic shock. SHOCK Investigators. Should We Emergently Revascularize Occluded Coronaries for Cardiogenic Shock. N Engl J Med. 1999;341:625–34. doi: 10.1056/NEJM199908263410901. [DOI] [PubMed] [Google Scholar]
- Hochman JS, Sleeper LA, Webb JG, Džavík V, Buller CE, Aylward P, Col J, White HD SHOCK Investigators. Early revascularization and long-term survival in cardiogenic shock complicating acute myocardial infarction. JAMA. 2006;295:2511–5. doi: 10.1001/jama.295.21.2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neumann FJ, Sousa-Uva M, Ahlsson A, Alfonso F, Banning AP, Benedetto U, Byrne RA, Collet JP, Falk V, Head SJ, Jüni P, Kastrati A, Koller A, Kristensen SD, Niebauer J, Richter DJ, Seferovic PM, Sibbing D, Stefanini GG, Windecker S, Yadav R, Zembala MO ESC Scientific Document Group. 2018 ESC/EACTS Guidelines on myocardial revascularization. Eur Heart J. 2018 [Google Scholar]
- Ibanez B, James S, Agewall S, Antunes MJ, Bucciarelli-Ducci C, Bueno H, Caforio ALP, Crea F, Goudevenos JA, Halvorsen S, Hindricks G, Kastrati A, Lenzen MJ, Prescott E, Roffi M, Valgimigli M, Varenhorst C, Vranckx P, Widimský P ESC Scientific Document Group. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC). Eur Heart J. 2018;39(3):119–77. doi: 10.1093/eurheartj/ehx393. [DOI] [PubMed] [Google Scholar]
- Collet JP, Thiele H, Barbato E, Barthelemy O, Bauersachs J, Bhatt DL, Dendale P, Dorobantu M, Edvardsen T, Folliguet T, Gale CP, Gilard M, Jobs A, Jüni P, Lambrinou E, Lewis BS, Mehilli J, Meliga E, Merkely B, Mueller C, Roffi M, Rutten FH, Sibbing D, Siontis GCM ESC Scientific Document Group. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Eur Heart J. 2021;42:1289–367. doi: 10.1093/eurheartj/ehaa575. [DOI] [PubMed] [Google Scholar]
- Kochar A, Al-Khalidi HR, Hansen SM, Shavadia JS, Roettig ML, Fordyce CB, Doerfler S, Gersh BJ, Henry TD, Berger PB, Jollis JG, Granger CB. Delays in Primary Percutaneous Coronary Intervention in ST-Segment Elevation Myocardial Infarction Patients Presenting With Cardiogenic Shock. JACC Cardiovasc Interv. 2018;11:1824–33. doi: 10.1016/j.jcin.2018.06.030. [DOI] [PubMed] [Google Scholar]
- Scholz KH, Maier SKG, Maier LS, Lengenfelder B, Jacobshagen C, Jung J, Fleischmann C, Werner GS, Olbrich HG, Ott R, Mudra H, Seidl K, Schulze PC, Weiss C, Haimerl J, Friede T, Meyer T. Impact of treatment delay on mortality in ST-segment elevation myocardial infarction (STEMI) patients presenting with and without haemodynamic instability: results from the German prospective, multicentre FITT-STEMI trial. Eur Heart J. 2018;39:1065–74. doi: 10.1093/eurheartj/ehy004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thiele H, Akin I, Sandri M, de Waha-Thiele S, Meyer-Saraei R, Fuernau G, Eitel I, Nordbeck P, Geisler T, Landmesser U, Skurk C, Fach A, Jobs A, Lapp H, Piek JJ, Noc M, Goslar T, Felix SB, Maier LS, Stepinska J, Oldroyd K, Serpytis P, Montalescot G, Barthelemy O, Huber K, Windecker S, Hunziker L, Savonitto S, Torremante P, Vrints C, Schneider S, Zeymer U, Desch S CULPRIT-SHOCK Investigators. One-Year Outcomes after PCI Strategies in Cardiogenic Shock. N Engl J Med. 2018;379:1699–710. doi: 10.1056/NEJMoa1808788. [DOI] [PubMed] [Google Scholar]
- Mehta RH, Lopes RD, Ballotta A, Frigiola A, Sketch MH, Bossone E, Bates ER. Percutaneous coronary intervention or coronary artery bypass surgery for cardiogenic shock and multivessel coronary artery disease? Am Heart J. 2010;159:141–7. doi: 10.1016/j.ahj.2009.10.035. [DOI] [PubMed] [Google Scholar]
- Pancholy SB, Palamaner Subash Shantha G, Romagnoli E, Kedev S, Bernat I, Rao SV, Jolly S, Bertrand OF, Patel TM. Impact of access site choice on outcomes of patients with cardiogenic shock undergoing percutaneous coronary intervention: A systematic review and meta-analysis. Am Heart J. 2015;170:353–61. doi: 10.1016/j.ahj.2015.05.001. [DOI] [PubMed] [Google Scholar]
- Guedeney P, Thiele H, Kerneis M, Barthelemy O, Baumann S, Sandri M, de Waha-Thiele S, Fuernau G, Rouanet S, Piek JJ, Landmesser U, Hauguel-Moreau M, Zeitouni M, Silvain J, Lattuca B, Windecker S, Collet JP, Desch S, Zeymer U, Montalescot G, Akin I CULPRIT-SHOCK Investigators. 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. doi: 10.1016/j.ahj.2020.04.014. [DOI] [PubMed] [Google Scholar]
- Tehrani BN, Damluji AA, Sherwood MW, Rosner C, Truesdell AG, Epps KC, Howard E, Barnett SD, Raja A, deFilippi CR, Murphy CE, O'Connor CM, Batchelor WB. Transradial access in acute myocardial infarction complicated by cardiogenic shock: Stratified analysis by shock severity. Catheter Cardiovasc Interv. 2021;97:1354–66. doi: 10.1002/ccd.29098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tousek P, Rokyta R, Tesarova J, Pudil R, Belohlavek J, Stasek J, Rohac F, Widimský P. 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. doi: 10.3109/17482941.2011.567282. [DOI] [PubMed] [Google Scholar]
- Gorog DA, Price S, Sibbing D, Baumbach A, Capodanno D, Gigante B, Halvorsen S, Huber K, Lettino M, Leonardi S, Morais J, Rubboli A, Siller-Matula JM, Storey RF, Vranckx P, Rocca B. Antithrombotic therapy in patients with acute coronary syndrome complicated by cardiogenic shock or out-of-hospital cardiac arrest: a Joint Position Paper from the European Society of Cardiology (ESC) Working Group on Thrombosis, in association with the Acute Cardiovascular Care Association (ACCA) and European Association of Percutaneous Cardiovascular Interventions (EAPCI). Eur Heart J Cardiovasc Pharmacother. 2021;7:125–40. doi: 10.1093/ehjcvp/pvaa009. [DOI] [PubMed] [Google Scholar]
- De Backer D, Biston P, Devriendt J, Madl C, Chochrad D, Aldecoa C, Brasseur A, Defrance P, Gottignies P, Vincent J-L. Comparison of dopamine and norepinephrine in the treatment of shock. N Engl J Med. 2010;362:779–789. doi: 10.1056/NEJMoa0907118. [DOI] [PubMed] [Google Scholar]
- Levy B, Clere-Jehl R, Legras A, Morichau-Beauchant T, Leone M, Frederique G, Quenot J-P, Kimmoun A, Cariou A, Lassus J, Harjola V-P, Meziani F, Louis G, Rossignol P, Duarte K, Girerd N, Mebazaa A, Vignon P, Mattei M, Thivilier C, Perez P, Auchet T, Fritz C, Boisrame-Helme J, Mercier E, Garot D, Perny J, Gette S, Hammad E, Vigne C, Dargent A, Andreu P, Guiot P. Epinephrine versus norepinephrine for cardiogenic shock after acute myocardial infarction. J Am Coll Cardiol. 2018;72:173–82. doi: 10.1016/j.jacc.2018.04.051. [DOI] [PubMed] [Google Scholar]
- Asfar P, Meziani F, Hamel JF, Grelon F, Mégarbane B, Anguel N, Mira JP, Dequin PF, Gergaud S, Weiss N, Legay F, Le Tulzo Y, Conrad M, Robert R, Gonzalez F, Guitton C, Tamion F, Tonnelier JM, Guezennec P, van der Linden T, Vieillard-Baron A, Mariotte E, Pradel G, Lesieur O, Ricard JD, Hervé F, Du Cheyron D, Guerin C, Mercat A, Teboul JL, Radermacher P SEPSISPAM Investigators. High versus low blood-pressure target in patients with septic shock. N Engl J Med. 2014;370:1583–93. doi: 10.1056/NEJMoa1312173. [DOI] [PubMed] [Google Scholar]
- Ameloot K, Jakkula P, Hästbacka J, Reinikainen M, Pettilä V, Loisa P, Tiainen M, Bendel S, Birkelund T, Belmans A, Palmers PJ, Bogaerts E, Lemmens R, De Deyne C, Ferdinande B, Dupont M, Janssens S, Dens J, Skrifvars MB. Optimum Blood Pressure in Patients With Shock After Acute Myocardial Infarction and Cardiac Arrest. J Am Coll Cardiol. 2020;76:812–24. doi: 10.1016/j.jacc.2020.06.043. [DOI] [PubMed] [Google Scholar]
- Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JG, Coats AJ, Falk V, González-Juanatey JR, Harjola VP, Jankowska EA, Jessup M, Linde C, Nihoyannopoulos P, Parissis JT, Pieske B, Riley JP, Rosano GM, Ruilope LM, Ruschitzka F, Rutten FH, van der Meer P ESC Scientific Document Group. 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)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016;37:2129–200. doi: 10.1093/eurheartj/ehw128. [DOI] [PubMed] [Google Scholar]
- Fuhrmann JT, Schmeisser A, Schulze MR, Wunderlich C, Schoen SP, Rauwolf T, Weinbrenner C, Strasser RH. Levosimendan is superior to enoximone in refractory cardiogenic shock complicating acute myocardial infarction. Crit Care Med. 2008;36:2257–66. doi: 10.1097/CCM.0b013e3181809846. [DOI] [PubMed] [Google Scholar]
- Cholley B, Caruba T, Grosjean S, Amour J, Ouattara A, Villacorta J, Miguet B, Guinet P, Lévy F, Squara P, Aït Hamou N, Carillion A, Boyer J, Boughenou MF, Rosier S, Robin E, Radutoiu M, Durand M, Guidon C, Desebbe O, Charles-Nelson A, Menasche P, Rozec B, Girard C, Fellahi JL, Pirracchio R, Chatellier G. Effect of Levosimendan on Low Cardiac Output Syndrome in Patients With Low Ejection Fraction Undergoing Coronary Artery Bypass Grafting With Cardiopulmonary Bypass: The LICORN Randomized Clinical Trial. JAMA. 2017;318:548–56. doi: 10.1001/jama.2017.9973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gordon AC, Perkins GD, Singer M, McAuley DF, Orme RML, Santhakumaran S, Mason AJ, Cross M, Al-Beidh F, Best-Lane J, Brealey D, Nutt CL, McNamee JJ, Reschreiter H, Breen A, Liu KD, Ashby D. Levosimendan for the Prevention of Acute Organ Dysfunction in Sepsis. N Engl J Med. 2016;375:1638–48. doi: 10.1056/NEJMoa1609409. [DOI] [PubMed] [Google Scholar]
- Landoni G, Lomivorotov VV, Alvaro G, Lobreglio R, Pisano A, Guarracino F, Calabrò MG, Grigoryev EV, Likhvantsev VV, Salgado-Filho MF, Bianchi A, Pasyuga VV, Baiocchi M, Pappalardo F, Monaco F, Boboshko VA, Abubakirov MN, Amantea B, Lembo R, Brazzi L, Verniero L, Bertini P, Scandroglio AM, Bove T, Belletti A, Michienzi MG, Shukevich DL, Zabelina TS, Bellomo R, Zangrillo A CHEETAH Study Group. Levosimendan for Hemodynamic Support after Cardiac Surgery. N Engl J Med. 2017;376:2021–31. doi: 10.1056/NEJMoa1616325. [DOI] [PubMed] [Google Scholar]
- Mehta RH, Leimberger JD, van Diepen S, Meza J, Wang A, Jankowich R, Harrison RW, Hay D, Fremes S, Duncan A, Soltesz EG, Luber J, Park S, Argenziano M, Murphy E, Marcel R, Kalavrouziotis D, Nagpal D, Bozinovski J, Toller W, Heringlake M, Goodman SG, Levy JH, Harrington RA, Anstrom KJ, Alexander JH LEVO-CTS Investigators. Levosimendan in Patients with Left Ventricular Dysfunction Undergoing Cardiac Surgery. N Engl J Med. 2017;376:2032–42. doi: 10.1056/NEJMoa1616218. [DOI] [PubMed] [Google Scholar]
- Gaudry S, Hajage D, Schortgen F, Martin-Lefevre L, Pons B, Boulet E, Boyer A, Chevrel G, Lerolle N, Carpentier D, de Prost N, Lautrette A, Bretagnol A, Mayaux J, Nseir S, Mégarbane B, Thirion M, Forel JM, Maizel J, Yonis H, Markowicz P, Thiery G, Tubach F, Ricard JD, Dreyfuss D AKIKI Study Group. Initiation Strategies for Renal-Replacement Therapy in the Intensive Care Unit. N Engl J Med. 2016;375:122–33. doi: 10.1056/NEJMoa1603017. [DOI] [PubMed] [Google Scholar]
- STARRT-AKI Investigators; Canadian Critical Care Trials Group; Australian and New Zealand Intensive Care Society Clinical Trials Group; United Kingdom Critical Care Research Group; Canadian Nephrology Trials Network; Irish Critical Care Trials Group. Timing of Initiation of Renal-Replacement Therapy in Acute Kidney Injury. N Engl J Med. 2020;383:240–51. doi: 10.1056/NEJMoa2000741. [DOI] [PubMed] [Google Scholar]
- Fuernau G. Lactate and other biomarkers as treatment target in cardiogenic shock. Curr Opin Crit Care. 2019;25:403–9. doi: 10.1097/MCC.0000000000000628. [DOI] [PubMed] [Google Scholar]
- Thiele H, Jobs A, Ouweneel DM, Henriques JPS, Seyfarth M, Desch S, Eitel I, Pöss J, Fuernau G, de Waha S. Percutaneous short-term active mechanical support devices in cardiogenic shock: A systematic review and collaborative meta-analysis of randomized trials. Eur Heart J. 2017;38:3523–31. doi: 10.1093/eurheartj/ehx363. [DOI] [PubMed] [Google Scholar]
- Freund A, Jobs A, Lurz P, Feistritzer HJ, de Waha-Thiele S, Meyer-Saraei R, Montalescot G, Huber K, Noc M, Windecker S, Zeymer U, Ouarrak T, Schneider S, Thiele H, Desch S. Frequency and Impact of Bleeding on Outcome in Patients With Cardiogenic Shock. JACC Cardiovasc Interv. 2020;13:1182–93. doi: 10.1016/j.jcin.2020.02.042. [DOI] [PubMed] [Google Scholar]
- Cheng R, Hachamovitch R, Kittleson M, Patel J, Arabia F, Moriguchi J, Esmailian F, Azarbal B. Complications of extracorporeal membrane oxygenation for treatment of cardiogenic shock and cardiac arrest: a meta-analysis of 1,866 adult patients. Ann Thorac Surg. 2014;97:610–6. doi: 10.1016/j.athoracsur.2013.09.008. [DOI] [PubMed] [Google Scholar]
- Fuernau G, Beck J, Desch S, Eitel I, Jung C, Erbs S, Mangner N, Lurz P, Fengler K, Jobs A, Vonthein R, de Waha-Thiele S, Sandri M, Schuler G, Thiele H. Mild Hypothermia in Cardiogenic Shock Complicating Myocardial Infarction. Circulation. 2019;139:448–57. doi: 10.1161/CIRCULATIONAHA.117.032722. [DOI] [PubMed] [Google Scholar]
- Thiele H, Vranckx, Schuler . Cardiogenic Shock. In: Eeckhout E, Serruys PW, Wijns W, Vahanian A, van Sambeek M, De Palma R, editors. The PCR-EAPCI Textbook - Percutaneous Interventional Cardiovascular Medicine. Toulouse, France: Europa Digital & Publishing; 2012. pp. 1–36. [Google Scholar]
- Jung RG, Simard T, Kovach C, Flint K, Don C, Di Santo P, Adamo M, Branca L, Valentini F, Benito-González T, Fernández-Vázquez F, Estevez-Loureiro R, Berardini A, Conti N, Rapezzi C, Biagini E, Parlow S, Shorr R, Levi A, Manovel A, Cardenal-Piris R, Diaz Fernandez J, Shuvy M, Haberman D, Sala A, Alkhouli MA, Marini C, Bargagna M, Schiavi D, Denti P, Markovic S, Buzzatti N, Chan V, Hynes M, Mesana T, Labinaz M, Pappalardo F, Taramasso M, Hibbert B. Transcatheter Mitral Valve Repair in Cardiogenic Shock and Mitral Regurgitation: A Patient-Level, Multicenter Analysis. JACC Cardiovasc Interv. 2021;14:1–11. doi: 10.1016/j.jcin.2020.08.037. [DOI] [PubMed] [Google Scholar]
- Thiele H, Zeymer U, Neumann FJ, Ferenc M, Olbrich HG, Hausleiter J, de Waha A, Richardt G, Hennersdorf M, Empen K, Fuernau G, Desch S, Eitel I, Hambrecht R, Lauer B, Böhm M, Ebelt H, Schneider S, Werdan K, Schuler G Intraaortic Balloon Pump in cardiogenic shock II (IABP-SHOCK II) trial investigators. 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. doi: 10.1016/S0140-6736(13)61783-3. [DOI] [PubMed] [Google Scholar]
- Thiele H, Zeymer U, Thelemann N, Neumann FJ, Hausleiter J, Abdel-Wahab M, Meyer-Saraei R, Fuernau G, Eitel I, Hambrecht R, Böhm M, Werdan K, Felix SB, Hennersdorf M, Schneider S, Ouarrak T, Desch S, de Waha-Thiele S IABPSHOCK II Trial (Intraaortic Balloon Pump in Cardiogenic Shock II) Investigators. Intraaortic Balloon Pump in Cardiogenic Shock Complicating Acute Myocardial Infarction: Long-Term 6-Year Outcome of the Randomized IABP-SHOCK II Trial. Circulation. 2018 doi: 10.1161/CIRCULATIONAHA.118.038201. [DOI] [PubMed] [Google Scholar]
- Shah M, Patnaik S, Patel B, Ram P, Garg L, Agarwal M, Agrawal S, Arora S, Patel N, Wald J, Jorde UP. Trends in mechanical circulatory support use and hospital mortality among patients with acute myocardial infarction and non-infarction related cardiogenic shock in the United States. Clin Res Cardiol. 2018;107:287–303. doi: 10.1007/s00392-017-1182-2. [DOI] [PubMed] [Google Scholar]
- Backhaus T, Fach A, Schmucker J, Fiehn E, Garstka D, Stehmeier J, Hambrecht R, Wienbergen H. Management and predictors of outcome in unselected patients with cardiogenic shock complicating acute ST-segment elevation myocardial infarction: results from the Bremen STEMI Registry. Clin Res Cardiol. 2018;107:371–9. doi: 10.1007/s00392-017-1192-0. [DOI] [PubMed] [Google Scholar]
- Kjaergaard J, Møller JE, Hassager C. Mechanical circulatory support for decompensated heart failure: the last remaining indication for intra-aortic balloon pump? EuroIntervention. 2019;15:571–3. doi: 10.4244/EIJV15I7A104. [DOI] [PubMed] [Google Scholar]
- Becher PM, Schrage B, Sinning CR, Schmack B, Fluschnik N, Schwarzl M, Waldeyer C, Lindner D, Seiffert M, Neumann JT, Bernhardt AM, Zeymer U, Thiele H, Reichenspurner H, Blankenberg S, Twerenbold R, Westermann D. Venoarterial Extracorporeal Membrane Oxygenation for Cardiopulmonary Support. Circulation. 2018;138:2298–300. doi: 10.1161/CIRCULATIONAHA.118.036691. [DOI] [PubMed] [Google Scholar]
- Karagiannidis C, Brodie D, Strassmann S, Stoelben E, Philipp A, Bein T, Müller T, Windisch W. Extracorporeal membrane oxygenation: evolving epidemiology and mortality. Intensive Care Med. 2016;42:889–96. doi: 10.1007/s00134-016-4273-z. [DOI] [PubMed] [Google Scholar]
- Schäfer A, Werner N, Westenfeld R, Moller JE, Schulze PC, Karatolios K, Pappalardo F, Maly J, Staudacher D, Lebreton G, Delmas C, Hunziker P, Fritzenwanger M, Napp LC, Ferrari M, Tarantini G. Clinical scenarios for use of transvalvular microaxial pumps in acute heart failure and cardiogenic shock- A European experienced users working group opinion. Int J Cardiol. 2019;291:96–104. doi: 10.1016/j.ijcard.2019.05.044. [DOI] [PubMed] [Google Scholar]
- Bochaton T, Huot L, Elbaz M, Delmas C, Aissaoui N, Farhat F, Mewton N, Bonnefoy E IMPELLA-STIC investigators. Mechanical circulatory support with the Impella® LP5.0 pump and an intra-aortic balloon pump for cardiogenic shock in acute myocardial infarction: The IMPELLA-STIC randomized study. Arch Cardiovasc Dis. 2020;113:237–43. doi: 10.1016/j.acvd.2019.10.005. [DOI] [PubMed] [Google Scholar]
- Schrage B, Ibrahim K, Loehn T, Werner N, Sinning JM, Pappalardo F, Pieri M, Skurk C, Lauten A, Landmesser U, Westenfeld R, Horn P, Pauschinger M, Eckner D, Twerenbold R, Nordbeck P, Salinger T, Abel P, Empen K, Busch MC, Felix SB, Sieweke JT, Moller JE, Pareek N, Hill J, Maccarthy P, Bergmann MW, Henriques JPS, Mobius-Winkler S, Schulze PC, Ouarrak T, Zeymer U, Schneider S, Blankenberg S, Thiele H, Schäfer A, Westermann D. Impella Support for Acute Myocardial Infarction Complicated by Cardiogenic Shock. Circulation. 2019;139:1249–58. doi: 10.1161/CIRCULATIONAHA.118.036614. [DOI] [PubMed] [Google Scholar]
- Dhruva SS, Ross JS, Mortazavi BJ, Hurley NC, Krumholz HM, Curtis JP, Berkowitz A, Masoudi FA, Messenger JC, Parzynski CS, Ngufor C, Girotra S, Amin AP, Shah ND, Desai NR. 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. doi: 10.1001/jama.2020.0254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amin AP, Spertus JA, Curtis JP, Desai N, Masoudi FA, Bach RG, McNeely C, Al-Badarin F, House JA, Kulkarni H, Rao SV. 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. doi: 10.1161/CIRCULATIONAHA.119.044007. [DOI] [PubMed] [Google Scholar]
- Udesen NJ, Moller JE, Lindholm MG, Eiskjaer H, Schäfer A, Werner N, Holmvang L, Terkelsen CJ, Jensen LO, Junker A, Schmidt H, Wachtell K, Thiele H, Engstrøm T, Hassager C DanGer Shock investigators. Rationale and design of DanGer shock: Danish-German cardiogenic shock trial. Am Heart J. 2019;214:60–8. doi: 10.1016/j.ahj.2019.04.019. [DOI] [PubMed] [Google Scholar]
- Ouweneel DM, Schotborgh JV, Limpens J, Sjauw KD, Engstrom AE, Lagrand WK, Cherpanath TGV, Driessen AHG, de Mol B, Henriques JPS. Extracorporeal life support during cardiac arrest and cardiogenic shock: a systematic review and meta-analysis. Intensive Care Med. 2016;42:1922–34. doi: 10.1007/s00134-016-4536-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brunner S, Guenther SPW, Lackermair K, Peterss S, Orban M, Boulesteix AL, Michel S, Hausleiter J, Massberg S, Hagl C. Extracorporeal Life Support in Cardiogenic Shock Complicating Acute Myocardial Infarction. J Am Coll Cardiol. 2019;73:2355–7. doi: 10.1016/j.jacc.2019.02.044. [DOI] [PubMed] [Google Scholar]
- Thiele H, Freund A, Gimenez MR, de Waha-Thiele S, Akin I, Pöss J, Feistritzer HJ, Fuernau G, Graf T, Nef H, Hamm C, Böhm M, Lauten A, Schulze PC, Voigt I, Nordbeck P, Felix SB, Abel P, Baldus S, Laufs U, Lenk K, Landmesser U, Skurk C, Pieske B, Tschöpe C, Hennersdorf M, Wengenmayer T, Preusch M, Maier LS, Jung C, Kelm M, Clemmensen P, Westermann D, Seidler T, Schieffer B, Rassaf T, Mahabadi AA, Vasa-Nicotera M, Meincke F, Seyfarth M, Kersten A, Rottbauer W, Boekstegers P, Muellenbach R, Dengler T, Kadel C, Schempf B, Karagiannidis C, Hopf HB, Lehmann R, Bufe A, Baumanns S, Öner A, Linke A, Sedding D, Ferrari M, Bruch L, Goldmann B, John S, Möllmann H, Franz J, Lapp H, Lauten P, Noc M, Goslar T, Oerlecke I, Ouarrak T, Schneider S, Desch S, Zeymer U ECLS-SHOCK Investigators. Extracorporeal life support in patients with acute myocardial infarction complicated by cardiogenic shock - Design and rationale of the ECLS-SHOCK trial. Am Heart J. 2021;234:1–11. doi: 10.1016/j.ahj.2021.01.002. [DOI] [PubMed] [Google Scholar]
- Banning AS, Adriaenssens T, Berry C, Bogaerts K, Erglis A, Distelmaier K, Guagliumi G, Haine S, Kastrati A, Massberg S, Orban M, Myrmel T, Vuylsteke A, Alfonso F, Van de Werf F, Verheugt F, Flather M, Sabate M, Vrints C, Gershlick AH Collaborators. Veno-arterial extracorporeal membrane oxygenation (ECMO) in patients with cardiogenic shock: rationale and design of the randomised, multicentre, open-label EURO SHOCK trial. EuroIntervention. 2021;16:e1227–36. doi: 10.4244/EIJ-D-20-01076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schrage B, Becher PM, Bernhardt A, Bezerra H, Blankenberg S, Brunner S, Colson P, Cudemus Deseda G, Dabboura S, Eckner D, Eden M, Eitel I, Frank D, Frey N, Funamoto M, Gossling A, Graf T, Hagl C, Kirchhof P, Kupka D, Landmesser U, Lipinski J, Lopes M, Majunke N, Maniuc O, McGrath D, Mobius-Winkler S, Morrow DA, Mourad M, Noel C, Nordbeck P, Orban M, Pappalardo F, Patel SM, Pauschinger M, Pazzanese V, Reichenspurner H, Sandri M, Schulze PC, Schwinger RHG, Sinning JM, Aksoy A, Skurk C, Szczanowicz L, Thiele H, Tietz F, Varshney A, Wechsler L, Westermann D. Left Ventricular Unloading is Associated with Lower Mortality in Cardiogenic Shock Patients Treated with Veno-Arterial Extracorporeal Membrane Oxygenation: Results From An International, Multicenter Cohort Study. Circulation. 2020;142:2095–106. doi: 10.1161/CIRCULATIONAHA.120.048792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russo JJ, Aleksova N, Pitcher I, Couture E, Parlow S, Faraz M, Visintini S, Simard T, Di Santo P, Mathew R, So DY, Takeda K, Garan AR, Karmpaliotis D, Takayama H, Kirtane AJ, Hibbert B. Left ventricular unloading during extracorporeal membrane oxygenation in patients with cardiogenic shock. J Am Coll Cardiol. 2019;73:654–62. doi: 10.1016/j.jacc.2018.10.085. [DOI] [PubMed] [Google Scholar]
- Kowalewski M, Malvindi PG, Zieliński K, Martucci G, Słomka A, Suwalski P, Lorusso R, Meani P, Arcadipane A, Pilato M, Raffa GM. Left Ventricle Unloading with Veno-Arterial Extracorporeal Membrane Oxygenation for Cardiogenic Shock. Systematic Review and Meta-Analysis. J Clin Med. 2020;9:1039. doi: 10.3390/jcm9041039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lüsebrink E, Orban M, Kupka D, Scherer C, Hagl C, Zimmer S, Luedike P, Thiele H, Westermann D, Massberg S, Schäfer A, Orban M. Prevention and treatment of pulmonary congestion in patients undergoing venoarterial extracorporeal membrane oxygenation for cardiogenic shock. Eur Heart J. 2020;41:3753–61. doi: 10.1093/eurheartj/ehaa547. [DOI] [PubMed] [Google Scholar]
- O’Neill WW, Schreiber T, Wohns DH, Rihal C, Naidu SS, Civitello AB, Dixon SR, Massaro JM, Maini B, Ohman EM. The current use of Impella 2.5 in acute myocardial infarction complicated by cardiogenic shock: results from the USpella Registry. J Interv Cardiol. 2014;27:1–11. doi: 10.1111/joic.12080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Basir MB, Schreiber TL, Grines CL, Dixon SR, Moses JW, Maini BS, Khandelwal AK, Ohman EM, O’Neill WW. Effect of Early Initiation of Mechanical Circulatory Support on Survival In Cardiogenic Shock. Am J Cardiol. 2017;119:845–51. doi: 10.1016/j.amjcard.2016.11.037. [DOI] [PubMed] [Google Scholar]
- Basir MB, Schreiber T, Dixon S, Alaswad K, Patel K, Almany S, Khandelwal A, Hanson I, George A, Ashbrook M, Blank N, Abdelsalam M, Sareen N, Timmis SBH, O'Neill WW. Feasibility of early mechanical circulatory support in acute myocardial infarction complicated by cardiogenic shock: The Detroit cardiogenic shock initiative. Catheter Cardiovasc Interv. 2018;91:454–61. doi: 10.1002/ccd.27427. [DOI] [PubMed] [Google Scholar]
- Basir MB, Kapur NK, Patel K, Salam MA, Schreiber T, Kaki A, Hanson I, Almany S, Timmis S, Dixon S, Kolski B, Todd J, Senter S, Marso S, Lasorda D, Wilkins C, Lalonde T, Attallah A, Larkin T, Dupont A, Marshall J, Patel N, Overly T, Green M, Tehrani B, Truesdell AG, Sharma R, Akhtar Y, McRae T, 3rd, O'Neill B, Finley J, Rahman A, Foster M, Askari R, Goldsweig A, Martin S, Bharadwaj A, Khuddus M, Caputo C, Korpas D, Cawich I, McAllister D, Blank N, Alraies MC, Fisher R, Khandelwal A, Alaswad K, Lemor A, Johnson T, Hacala M, O'Neill WW National Cardiogenic Shock Initiative Investigators. Improved Outcomes Associated with the use of Shock Protocols: Updates from the National Cardiogenic Shock Initiative. Catheter Cardiovasc Interv. 2019;93:1173–83. doi: 10.1002/ccd.28307. [DOI] [PubMed] [Google Scholar]
- Huang CC, Hsu JC, Wu YW, Ke SR, Huang JH, Chiu KM, Liao PC. Implementation of extracorporeal membrane oxygenation before primary percutaneous coronary intervention may improve the survival of patients with ST-segment elevation myocardial infarction and refractory cardiogenic shock. Int J Cardiol. 2018;269:45–50. doi: 10.1016/j.ijcard.2018.07.023. [DOI] [PubMed] [Google Scholar]
- Lemor A, Basir MB, Patel K, Kolski B, Kaki A, Kapur NK, Riley R, Finley J, Goldsweig A, Aronow HD, Belford PM, Tehrani B, Truesdell AG, Lasorda D, Bharadwaj A, Hanson I, Lalonde T, Gorgis S, O'Neill W National Cardiogenic Shock Initiative Investigators. Multivessel Versus Culprit-Vessel Percutaneous Coronary Intervention in Cardiogenic Shock. JACC Cardiovasc Interv. 2020;13:1171–8. doi: 10.1016/j.jcin.2020.03.012. [DOI] [PubMed] [Google Scholar]
- Iborra-Egea O, Rueda F, García-García C, Borràs E, Sabidó E, Bayés-Genís A. Molecular signature of cardiogenic shock. Eur Heart J. 2020;41:3839–48. doi: 10.1093/eurheartj/ehz783. [DOI] [PubMed] [Google Scholar]
- Kraut JA, Madias NE. Lactic acidosis. N Engl J Med. 2014;371:2309–19. doi: 10.1056/NEJMra1309483. [DOI] [PubMed] [Google Scholar]