Вы находитесь на странице: 1из 12

REVIEWS

Management of refractory cardiogenic


shock
Alex Reyentovich1, Maya H. Barghash1 and Judith S. Hochman2
Abstract | Cardiogenic shock is a life-threatening condition that occurs in response to reduced
cardiac output in the presence of adequate intravascular volume and results in tissue hypoxia.
Cardiogenic shock has several underlying aetiologies, with the most common being acute
myocardial infarction (AMI). Refractory cardiogenic shock presents as persistent tissue
hypoperfusion despite administration of adequate doses of two vasoactive medications and
treatment of the underlying aetiology. Investigators of the SHOCK trial reported a long-term
mortality benefit of emergency revascularization for shock complicating AMI. Since the
publication of the SHOCK trial and subsequent guideline recommendations, the increase in
community-based use of percutaneous coronary intervention for this condition has resulted
in a significant decline in mortality. Despite these successes in the past 15 years, mortality still
remains exceptionally high, particularly in patients with refractory cardiogenic shock. In this
Review, we discuss the aetiology and pathophysiology of cardiogenic shock and summarize the
data on the available therapeutics and their limitations. Although new mechanical circulatory
support devices have been shown to improve haemodynamic variables in patients with shock
complicating AMI, they did not improve clinical outcomes and are associated with high costs
and complications.

Cardiogenic shock describes a physiological state of shock5. The clinical criteria for cardiogenic shock used
end-organ hypoperfusion characterized by reduced in the trial included hypotension (defined as systolic
cardiac output in the presence of adequate intravascu- blood pressure (SBP) <90 mmHg for at least 30 min, or
lar volume1. The overall incidence of cardiogenic shock mean arterial pressure 30 mmHg lower than baseline,
has not changed from 2001 to 2014, although in 2014 a or the need for pharmacological or intra-aortic balloon
higher proportion of patients presented with cardiogenic pump (IABP) support to maintain SBP >90 mm Hg) and
shock on admission (1.9% in 2001 versus 2.7% in 2014, end-organ hypoperfusion (defined as cool extremities,
P = <0.01) and fewer developed cardiogenic shock dur- oliguria with urine output of <30 ml/h, altered mental
ing hospitalization (4.8% in 2001 versus 2.1% in 2014, status, serum lactate >2.0 mmol/l, and clinical signs of
P = 0.01)2. Cardiogenic shock has a wide spectrum of pulmonary congestion)6. Haemodynamic criteria, when
presentation, ranging from preshock to refractory central monitoring was available, included cardiac index
cardio­genic shock, and patients with refractory cardio- ≤2.2 l/min/m2 and adequate or elevated left ventricular
genic shock have the worst prognosis3,4. At present, there (LV) filling pressures (that is, PCWP >15 mmHg 1,2,7).
1
Division of Cardiology, is no agreed consensus on the criteria that define cardio­ In the IABP-SHOCK II trial8, the investigators defined
New York University–Langone
Medical Center, 530 First
genic shock, nor is there a well-established severity persistent cardiogenic shock complicating myocardial
Avenue, New York, scale. Clinical recognition of signs of preshock (that is, infarction (MI) as hypoperfusion with SBP <100 mmHg
New York 10016, USA. hypoperfusion without hypotension) is critical for early (despite treatment with ≥7 mcg/kg/min of dopamine or
2
Department of Medicine, intervention. In the SHOCK trial registry 5, patients with equivalent dose of noradrenaline), low cardiac index
Cardiovascular Clinical
nonhypotensive cardiogenic shock had high systemic (<2.2 l/min/m2 if measured off IABP, or <2.5 l/min/m2 if
Research Center, 530 First
Avenue, New York, New York vascular resistance and stable blood pressure levels, but measured on IABP), and PCWP >15 mmHg (despite
10016, USA. low cardiac indices, low ejection fraction, and elevated establishing a patent infarct-related artery). In a trial
Correspondence to A.R. pulmonary capillary wedge pressure (PCWP), similar to utilizing mechanical circulatory support (MCS),
alex.reyentovich@nyumc.org those with classic cardiogenic shock. Inhospital mortal- severe refractory cardiogenic shock was defined as SBP
doi:10.1038/nrcardio.2016.96 ity in patients with nonhypotensive cardiogenic shock <90 mmHg, cardiac index <2.0 l/min/m2, and evidence
Published online 30 June 2016 was lower than in those with hypotensive cardiogenic of end-organ failure despite IABP or pressor support 9.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points have reported that cardiogenic shock occurs within 24 h
(early shock) after MI symptom onset in 50–75% of
• Cardiogenic shock is characterized by acute hypoperfusion and end-organ patients (including those with mechanical complica-
dysfunction owing to reduced cardiac output, and is commonly caused by acute tions), and within the following 3 days in the remainder
myocardial infarction (AMI) with left ventricular dysfunction of patients5,9,11,12.
• Emergent revascularization is the only therapy that has been shown to reduce Cardiogenic shock can also occur secondary to myo-
mortality in patients with cardiogenic shock complicating AMI carditis, an inflammatory condition of the heart muscle
• Refractory cardiogenic shock can be defined as ongoing evidence of tissue that manifests commonly as a cardiac complication of
hypoperfusion despite administration of adequate doses of two vasoactive viral infection13. While most patients show only mild
medications and treatment of the underlying aetiology
symptoms, a small subset of patients develop a severe
• Refractory cardiogenic shock carries a poor prognosis, with an inhospital mortality of form of myocarditis — known as fulminant myo­carditis
~50% despite pharmacological and mechanical circulatory support
— that results in heart failure and cardiogenic shock
• The use of mechanical circulatory support devices for cardiogenic shock is increasing, character­ized by severe haemodynamic compromise
but there is currently no evidence showing that they improve clinical outcomes
requiring high-dose vasopressor and MCS. The factors
• Novel therapeutics and robust randomized trial data are needed to address the that determine whether a patient presents with fulminant
persistently high mortality in patients with refractory cardiogenic shock
or nonfulminant myocarditis are not known, as the clin-
ical course of myocarditis has little correlation with the
degree of lymphocytic infiltration, necrosis, or fibrosis14.
In this Review, we provide a brief overview of the Patients presenting with fulminant myocarditis are more
epidemiology, aetiology, and pathophysiology of cardio­ likely to have a fever or flu-like symptoms than those with
genic shock. Diagnostic strategies will also be high- the nonfulminant form of the disease, and are more likely
lighted, and the current therapeutic strategies used to to present with sudden onset of symptoms, in addition
manage and treat patients with refractory cardiogenic to severe global LV dysfunction with normal LV dimen-
shock will be discussed. Although much is known about sions15. Both giant cell myocarditis and active lymphocytic
this condition, mortality remains very high, emphasizing myocarditis can lead to cardiogenic shock13. Like ACS,
the need for further research. myocarditis is characterized by elevated troponin levels
and electrocardiographic changes such as ST‑segment
Aetiology eleva­tion, T‑wave inversions, ST‑segment depressions,
Cardiogenic shock can be caused by an acute cardiac con- and pathological Q waves. A diagnosis of acute myo­
dition or a systemic illness that triggers a chronic cardiac carditis should be considered in young patients p ­ resenting
condition associated with minimal cardiac reserve. Acute with ACS without traditional coronary risk factors14,16.
MI (AMI), unstable angina, postcardiotomy syndrome, Postcardiotomy cardiogenic shock complicates 2–6%
valvular heart disease, myocardial disease (such as myo- of all cases of cardiothoracic surgery 17,18 and is character­
carditis), LV outflow obstruction in hypertrophic cardio­ ized by development of low cardiac output and hypo­
myopathy, stress-induced cardiomyopathy, pericardial perfusion in the early postoperative period19. Several
tamponade, congenital lesions, and mechanical injury to factors predispose patients to developing postcardiotomy
the heart have all been implicated in the pathogenesis of low output syndrome, including LV ejection fraction
cardiogenic shock (BOX 1). <20%, reoperative or emergency surgery, age >70 years,
Acute coronary syndrome (ACS) with or without diabetes mellitus, triple-vessel or left main disease, and
ST-segment elevation is the most common cause of recent MI20. Mortality for postcardiotomy cardiogenic
cardio­genic shock, accounting for up to 80% of all cases10. shock is 50–80%19. Several centres have used temporary
Among all cases of cardiogenic shock complicating AMI surgically implanted extracorporeal centrifugal pumps to
in the SHOCK trial and registry5,6, 78.5% were attributable treat postcardiotomy shock21,22.
to predominant LV failure. Among these patients, 58.8% RV shock can occur after MI, postcardiotomy syn-
presented with anterior MI, whereas 34.4% presented drome, heart transplantation, or LV assist device (LVAD)
with MI in the inferior wall without anterior involvement. placement, or can be a complication of chronic heart fail-
Of the patients with primary inferior wall MI leading to ure, pulmonary embolism, or myocarditis. Isolated RV
cardiogenic shock, 38.3% had a history of MI. Other non- failure occurs in ~3% of patients presenting with cardio-
primary LV failure-related causes of cardio­genic shock genic shock complicating AMI23, and these patients tend
complicating AMI include isolated right ventricu­lar (RV) to be younger, and are more likely to have an inferior
shock and mechanical complications such as acute severe MI and a single-vessel right coronary artery occlusion
mitral regurgitation (caused by papillary muscle rupture than patients with cardiogenic shock owing primarily
or dysfunction), ventricular s­ eptal rupture, free wall rup- to LV failure. RV shock also tends to present sooner
ture, cardiac tamponade, a history of severe valvular heart after MI diagnosis in these patients24. Despite younger
disease, excessive β­ ‑­blockade or Ca2+ channel-blockade, age and inferior location of MI, patients with RV shock
or procedural complications5. The majority of patients have similarly high inhospital mortality to those with LV
who develop cardio­genic shock as a result of MI do not shock (53.1% versus 60.8%; P = 0.296)23. Most patients
present with cardiogenic shock at the time of initial med­ with acute ischaemic RV dysfunction display spontane-
ical contact or hospital­ization. In the SHOCK trial6, the ous haemodynamic improvement within 3–10 days, and
median time from symptom onset of MI to cardiogenic recovery of RV function within 3–12 months, regardless
shock owing to LV failure was 5.5 h. Several studies of patency of the infarct-related artery 24–27.

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Refractory cardiogenic shock can be a manifestation owing to RV infarction presents as hypotension with
of any of the aforementioned acute aetiologies of cardio­ an i­ nferior wall MI, elevated jugular venous pressure,
genic shock or a manifestation of acute decompen­ and no evidence of pulmonary congestion on exam or
sation of chronic heart failure. The relative incidence of chest radio­graphy. An electrocardiogram is helpful for
refractory cardiogenic shock attributed to the various diagnosing this particular aetiology of shock33. Anterior
­aetiologies has not yet been described. MI with LV pump failure as a cause of cardiogenic
shock is likely in patients with ST‑segment elevation
Pathophysiology in the pre­cordial leads. In the case of a first inferior
Regardless of the initiating cause of cardiogenic shock, ST‑segment elevation MI (STEMI) as the cause of
the heart’s inability to deliver adequate cardiac output
leads to tissue hypoperfusion. Cardiac dysfunction
resulting from cardiogenic shock is usually caused by Box 1 | Causes of cardiogenic shock
a large MI, and the degree of myocardial necrosis is
an important predictor of risk of cardiogenic shock28. Acute myocardial infarction
Myocardial dysfunction leads to a reduction in stroke Left ventricular failure
volume and cardiac output, consequently increas- • Large infarction
ing ventricular diastolic pressure and wall stress, all • Small/moderate infarction with
of which further reduce coronary perfusion pressure. -- pre-existing dysfunction
Furthermore, LV dysfunction and ischaemia increase -- extensive ischaemia
diastolic stiffness, which elevates left atrial pressure, • Global ischaemia
leading to pulmonary congestion, hypoxia, and worsen- Right ventricular failure
ing ischaemia. Myocardial perfusion is further reduced • Large infarction
by the presence of hypotension and tachycardia12,29. • Small/moderate infarction with
Compensatory mechanisms are activated dur- -- pre-existing dysfunction
ing cardiogenic shock to increase sympathetic tone, -- pulmonary hypertension
which increases heart rate and contractility, and to Mechanical complications
stimulate the renin–angiotensin–aldosterone system, • Acute mitral regurgitation with
which leads to fluid retention, increased preload, and -- rupture of a papillary muscle
vaso­constriction to maintain systemic blood pressure. -- rupture of chordae tendinae
An increase in systemic inflammatory response is often -- severe papillary muscle dysfunction
a consequence of a large infarction and prolonged hypo­ • Ventricular septal defect caused by rupture of the
perfusion. Activation of the inflammatory cascade leads interventricular septum
to release and activation of inducible nitric oxide syn- • Left ventricular free wall rupture
thase, resulting in further vaso­dilatation and worsen­ing • Pericardial tamponade owing to rupture of the left
hypotension and hypoperfusion4. Furthermore, activa- ventricular free wall or haemorrhagic pericardial
tion of this cascade results in the paradoxically low sys- effusion
temic vascular resistance observed in the SHOCK trial6 Concomitant conditions causing mixed aetiology
and might contribute to the development of refractory • Haemorrhage
cardiogenic shock (FIG. 1). The presence of concomi­ • Infection
tant low systemic vascular resistance and systemic • Excess negative inotropic or vasodilator medications
inflammatory response syndrome with cardiogenic • Sustained bradyarrhythmia or tachyarrhythmia
shock is associated with the development of sepsis and • Hyperglycaemia or ketoacidosis
poor outcomes30–32.
Other conditions
Diagnosis End-stage cardiomyopathy
The classic clinical syndrome of cardiogenic shock Myocarditis
owing to LV failure is characterized by systemic hypo- Septic shock with severe myocardial depression
tension, SBP <90 mmHg or the need for high-dose ino- Left ventricular outflow tract obstruction
tropic support to maintain SBP between 90–100 mmHg, • Aortic stenosis
and symptoms or signs of organ hypoperfusion and • Hypertrophic obstructive cardiomyopathy
pulmonary congestion. Notably, up to 25% of patients Obstruction to left ventricular filling
presenting with cardiogenic shock owing to LV failure • Mitral stenosis
in the setting of AMI do not present with the classic • Left atrial myxoma
symptoms, instead displaying isolated hypoperfusion Acute mitral regurgitation (chordal rupture)
in the absence of pulmonary congestion, despite high Acute aortic insufficiency
PCWP12. On clinical examination, cardiogenic shock Myocardial contusion
normally presents as distant heart sounds, but might Postcardiotomy shock
also present with third or fourth heart sounds and
Global ischaemia
a systolic murmur, which should raise suspicion for a
Stress-induced cardiomyopathy
mechanical complication such as mitral regurgita-
Cardiac tamponade
tion or ventricular septal rupture33. Meanwhile, shock

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

cardiogenic shock, the electrocardiogram should show Invasive haemodynamic assessment using pulmo-
marked ST‑segment elevation in the inferior leads with nary artery catheterization is not recommended for
anterior ST‑segment depressions reflecting posterior routine use, but is useful to confirm and characterize
infarction. RV infarction can complicate acute inferior suspected shock, and is often used to manage refrac-
MI and can be denoted by precordial ST‑segment ele- tory cardiogenic shock. For patients with cardiogenic
vation predomi­nantly in V1–V2, as well as ST‑segment shock owing predominantly to LV failure, right heart
eleva­tion in the right-sided leads. In general, cardio- catheterization can confirm low cardiac output (cardiac
genic shock owing to a first inferior MI should immedi­ index <2.2 l/min/m2) and high filling pressures (PCWP
ately prompt echocardio­graphic screening to detect ≥15 mmHg)35,32. For other aetiologies of cardio­genic
papillary muscle rupture with acute mitral regurgitation shock complicating AMI, such as ventricular septal
or ­ventricular septal rupture34,35. ­rupture, a step‑up in oxygen saturation can be seen
Echocardiography is essential for the evaluation of from the right atrium to ventricle. Furthermore, in acute
RV and LV function, and assessment of mechanical severe mitral regurgitation, pulmonary artery wedge
complications, including free wall rupture with cardiac CV waves (large systolic waves seen in atrial tracing
tamponade, acute mitral regurgitation, and ventricular indicating increased atrial filling) are often 10 mmHg
septal rupture. Left atrial pressure estimation and car- above mean PCWP, and in RV shock, disproportion-
diac output measured using echocardiography can aid in ately high right-sided filling pressures and Kussmaul’s
the timely diagnosis of cardiogenic shock. A restrictive sign (a paradox­ical rise in jugular venous pressure upon
filling pattern on echocardiography, defined as a mitral inspiration) can be observed23. Emergency coronary
inflow deceleration time of <140 ms, was seen in 60.9% angiography in the setting of cardiogenic shock com-
of patients in the SHOCK trial5, and was predictive of plicating MI is used to determine the distribution and
both a lower ejection fraction and PCWP ≥20 mmHg. extent of coronary artery obstruction and guide either
No association was observed between a restrictive filling percutaneous or surgical revascularization36.
pattern and mortality 34.
Prognosis
Various clinical and haemodynamic criteria can be used
Myocardial injury to develop prognostic markers that provide a frame-
work to understand predictors of refractory cardiogenic
shock. The most common cause of death after cardio-
↓ Cardiac output genic shock is pump failure, which often occurs within
↓ Stroke volume
days after the event 37. Two scoring systems based on
↑ NOS multivariate models for cardiogenic shock severity have
been devised based on data from the SHOCK trial38: the
↑ NO stage 1 risk score based on clinical data, and the stage 2
↑ Peroxynitrite
Hypotension risk score that incorporates haemodynamic data. In the
stage 1 model, eight clinical risk factors were identified
as predictors of inhospital mortality: advanced age, shock
Vasodilatation ↑ Inflammatory on admission, clinical evidence of end-organ hypoper-
↓ SVR cytokines fusion, anoxic brain damage, decreasing SBP, previous
CABG surgery, noninferior MI, and creatinine levels
>1.9 mg/dl. Mortality associated with this disease stage
ranged from 22% to 88%, depending on score category 38.
↓ Systemic ↓ Coronary
perfusion perfusion pressure For the stage 2 model, risk factors included advanced
Systemic
inflammation age, end-organ hypoperfusion, anoxic brain damage,
decreasing stroke work, and LV ejection fraction <28%.
Worsening
myocardial The effect of early revascularization did not vary by risk
function category, suggesting that all patients will derive benefit
from early revascular­ization38. In the multicentre, ran-
domized TRIUMPH study 39, predictors of mortality
End-organ in patients with persistent vasodepressor-dependent
dysfunction
cardio­genic shock following AMI despite a patent infarct-­
related artery included treatment with a high number of
vasopressors, decreased SBP, and low creatinine clear-
Death ance40. The observational CardShock study 41, conducted
between 2010 and 2015 to identify prognostic factors for
Figure 1 | The downward spiral of refractory cardiogenicNature shock.Reviews
The downward
| Cardiology both ACS and non-ACS aetiologies of cardiogenic shock,
spiral of refractory cardiogenic shock results from severe cardiac dysfunction, most often
due to myocardial injury, leading to ongoing systemic and coronary hypoperfusion. determined that advanced age, a history of MI or CABG
A commonly observed systemic inflammatory response might lead to vasodilatation surgery, altered mental status, low LV ejection frac-
and further contribute to ongoing cardiac dysfunction and end-organ insult. tion, low estimated glomerular filtration rate, and high
Cardiogenic shock will inevitably lead to death if the cycle of damage is not interrupted. blood lactate levels were independently associated with
NO, nitric oxide; NOS, nitric oxide synthase; SVR, systemic vascular resistance. increased inhospital mortality 41.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The prognostic and management implications results in increased likelihood of survival. A critical
of RV involvement in cardiogenic shock in condi- timeframe of treatment must also be defined for patients
tions other than RV MI have not been established. with cardiogenic shock, after which the consequences of
Haemodynamic variables that were used to predict prolonged end-organ insult will inevitably lead to death.
RV failure after dur­able LVAD implantation (cen- This timeframe will be particularly relevant for patients
tral venous pressure (CVP) >10 mmHg, CVP/PCWP with refractory cardiogenic shock, who have ongoing
>0.63, pulmonary artery pulsatility index <2.0, and RV hypoperfusion despite addressing all reversible causes
stroke work index <450 mmHg/ml/m2) were evaluated of shock. The remainder of this Review will focus on the
in a patient cohort with cardiogenic shock and AMI management of patients with AMI who develop cardio-
using data from the SHOCK trial and registry 5,6,42. RV genic shock owing to LV failure, although certain prin-
dysfunction was found to be present in 37% and 36% ciples and therapies (such as inotropes and mechanical
of patients in the trial and the registry, respectively 42. circulatory support) can be applied to a broader range
Future studies are needed to establish the utility of of patients with cardiogenic shock.
these variables as ­indicators of RV function in clinical
management decisions. Emergency revascularization
In addition to clinical and haemodynamic ­factors, Emergency cardiac catheterization and revasculariza-
echocardiographic criteria, including severity of mitral tion are guideline-recommended (class I, level of evi-
regurgitation and reduction of LV ejection fraction dence B) for patients with ACS36,47,48. In the SHOCK
are also independently associated with survival in trial6, 302 patients with STEMI and cardiogenic shock
patients with cardiogenic shock. Importantly, a sur- owing to LV dysfunction were randomized within 36 h
vival benefit from early revascularization was noted of MI onset to either emergency revascularization
across the spectrum of LV ejection fractions and mitral or to i­nitial medical stabilization for ≥54 h, followed
regurgitation severity 43. by cardiac catheterization and revascularization as
Of all causes of cardiogenic shock in patients clinically indicated. The majority of patients in both
with MI in the SHOCK registry 5, ventricular septal treatment groups were treated with IABP and ino-
rupture was associated with the highest inhospital tropes or vasopressors. The average cardiac index was
mortal­ity (87%)44. In an observational study involving 1.8 l/min/m2 and nearly one-third of patients suffered
117 patients with refractory cardiogenic shock, 68% of cardiac arrest before random­ization. Angiography in
all cases of shock were attributable to ischaemic cardio­ patients with cardio­genic shock owing to AMI often
myopathy, predomin­antly AMI. The remaining cases revealed multivessel disease, with left main disease
were attribut­able to nonischaemic aetiologies includ- in 23% of patients and triple-­vessel disease in 64%3,49.
ing myocarditis, valvular disease, orthotopic heart While the primary end point of all-cause mortality at
transplant rejection, peripartum cardiomyopathy, 30 days was not different between an invasive strat-
­sarcoidosis, and Takotsubo syndrome9. egy of emergent revascularization and a conservative
Mortality from cardiogenic shock complicating AMI strategy of initial medical stabil­ization (46.7% versus
has fallen over the past decades with increasing use of 56.0%; P = 0.11), at the 6‑month follow‑up, mortality
reperfusion therapy, particularly percutaneous coronary was significantly lower with revascular­ization (50.3%
intervention (PCI). In the US Worcester Mass Heart versus 63.1%; P = 0.027). This mortality benefit rep-
Attack Study 45, 76% of patients who developed cardio- resented a large treatment effect, with the number
genic shock died in the hospital between 1975 and 1990 needed to treat of eight to save one life6, and persisted
(compared with 16.5% who did not develop cardio­ up to 11 years (average 6 years follow‑up; 32.8% ver-
genic  shock), consistent with 81% mortality for sus 19.6%; P = 0.03)50. Among patients random­ized
­cardiogenic shock described by Killip and Kimball in to emergency revascularization in the SHOCK trial6,
1967 (REF. 46). Between 2003 and 2005, 45.4% of patients those treated with CABG surgery (~40%) had a higher
with AMI who developed cardiogenic shock died dur- prevalence of diabetes and more extensive coronary
ing hospitalization, compared with 7.3% of those who disease (three-vessel and left main coronary artery dis-
did not develop cardiogenic shock45. Not surprisingly, ease) than those treated with PCI. However, survival
­inhospital mortality of cardiogenic shock was higher rates at 30 days and 1 year were similar between these
among elderly patients, with 35.7% mortality in patients revascular­ization modalities. Emergency CABG surgery
aged 65–74 and 64.7% in patients aged >85 years45. should be considered in patients with multivessel coro-
nary disease, and should be performed instead of PCI in
Management of cardiogenic shock patients with mech­anical complications of AMI, such as
The initial goal in the management of cardiogenic shock ventricular septal rupture or papillary muscle rupture8.
is to identify and address all reversible causes. While Since the publication of the SHOCK trial and
ACS accounts for the majority of cases of cardiogenic updated guideline recommendations for early
shock, alternative aetiologies must be rapidly identified revascular­ization in cardiogenic shock complicating
if coronary thrombosis is not present. An emphasis has MI, the use of revascular­ization for the management
been placed on a ‘golden hour’ when managing many of cardiogenic shock has increased47. This observed
clinical conditions, including trauma, STEMI, and acute increase is primarily attributable to an increased rate
stroke. The golden hour refers to a time period in which of PCI, as the total number of CABG surgeries per-
medical management that addresses reversible causes formed did not increase. Despite the marked decrease

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Percent migration Total annual hospital impact of these agents are limited, findings from the SOAP II
10% $254,010,000
randomized trial53 suggest that noradrenaline is prefer­
able to dopamine for initial management. In total,
25% $635,025,000
1,679 patients with circulatory shock of varying aetiolo-
50% $1,270,050,000 gies (280 patients had cardiogenic shock) were assigned
100% $2,540,100,000 to initial therapy with either dopamine or noradrena-
line. Within the subgroup of patients with cardiogenic
$3,000,000,000
Projected incremental hospital cost shock, the rate of death at 28 days was higher with dopa-
$2,500,000,000
w/IABP to pVAD migration mine than with noradrenaline, although this finding
must be viewed in the context of an overall nonsignifi-
$2,000,000,000 cant difference in mortality and lack of heterogeneity of
treatment effect. An overall higher rate of arrhythmias
$1,500,000,000 — predomi­nantly atrial fibrillation — in patients treated
with dopamine was also observed in the cardiogenic
$1,000,000,000
shock subgroup53. In general, efforts should be made
$500,000,000 to minimize the number of drugs used, as well as their
dose and dur­ation, owing to the risk of arrhythmia and
$– vasopressor-­mediated end-organ insult. A vasopressor
10% 25% 50% 100%
migration migration migration migration
(typically noradrenaline) is usually initiated in patients
with severe hypotension (SBP <80 mm Hg or mean
Figure 2 | An analysis of potential incremental
Nature cost of
Reviews | Cardiology arterial pressure <60 mm Hg). The use of dobutamine
shifting from using intra-aortic balloon pumps (IABP) alone is limited to patients with a low cardiac index,
to percutaneous left ventricular assist devices (pVAD). high PCWP, and a relatively stable SBP (80–90 mmHg).
The data show an incremental increase in cost of
Patients in a low output state who do not have hypo-
$33,957,839 for the hospital system in the USA associated
with use of pVAD in patients with cardiogenic shock tension can be treated with vaso­dilators, including
undergoing percutaneous coronary intervention, without intravenous nitroglycerin or nitroprusside, initiated
an associated improvement in mortality, reduced hospital in low doses. In general, a third agent probably has no
length of stay, or hospital readmission. Reprinted from additional benefit if there is evidence of hypoperfusion
Shah, A. et al. Clinical and economic effectiveness of despite therapeutic doses of two vasoactive agents.
percutaneous ventricular assist devices for high-risk
patients undergoing percutaneous coronary intervention. Percutaneous and surgical MCS devices
J. Invasive Cardiol. 27, 148–154 (2015), with permission MCS devices can maintain organ perfusion, reduce
from HMP Communications. cardiac filling pressures, reduce LV volume and wall
stress, decrease myocardial oxygen consumption, and
augment coronary perfusion54. The first clinically used
in overall mortality over time attributable to increased MCS device, the IABP, was developed in the 1960s
use of PCI, as noted above, mortality in patients with when mortality from cardiogenic shock complicating
cardiogenic shock managed with PCI has not fallen over MI was exceptionally high46 (~80%), before the advent
time. In 2013, the large CathPCI Registry 47 reported of life-saving reperfusion. On the basis of observa-
a significant rise in inhospital mortality in patients tional evidence showing that IABP improves haemo­
treated with PCI from 27.6% in 2005–2006 to 30.6% dynamics and systemic perfusion55, IABP became the
in 2011–2013. Refractory cardio­genic shock might stan­dard of care in most tertiary centres in the USA
possibly be the result of incomplete revascularization. despite a lack of randomized trial evidence demon-
CABG surgery has remained i­ nfrequently used despite strating its effect on mortality. Newer percutaneous
the high preva­lence of multi­vessel disease in patients and surgically implanted devices have since been
who develop cardiogenic shock and the potential of developed to provide greater haemodynamic support
CABG surgery to provide complete revascularization47. to address the cohort of patients with persistently high
A multicentre, prospective, observational study con- mortality owing to refractory cardiogenic shock. Again,
ducted from 1998 to 2010 demonstrated that patients although no clinical trial evidence exists showing the
receiving culprit-only PCI when presenting with cardio- benefit of these devices in reducing mortality, their use
genic shock had signifi­cantly worse 6‑month survival is increasing on the basis of superior haemodynamic
than patients receiving multivessel PCI (43.9% versus support 56. This increase in the use of novel percutane-
20.4%, P = 0.008)51. The benefit of culprit versus multi­ ous support devices, despite lack of demonstrable clin-
vessel PCI for the treatment of cardiogenic shock com- ical benefit, has a potentially large effect on health-care
plicating MI is the s­ ubject of an ongoing r­ andomized expenditure (FIG. 2).
clinical trial52. Temporary MCS devices are suitable for candidates
with refractory cardiogenic shock despite optimization
Pharmacological circulatory support of volume status, preload, afterload, and the use of ino-
Sympathomimetic inotropic and vasopressor agents tropic or vasopressor support. The use of temporary
are the first-line drugs for patients who develop cardio- MCS devices in this patient cohort is also based on
genic shock. Although the data comparing the efficacy the likelihood of recovery of organ function, and after

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

consideration of more advanced heart failure therapies54. reported59. Although the IABP is widely available, the
The decision as to which device to use is predicated on IABP Shock Trial60 demonstrated that IABP treatment
understanding the potential haemodynamic benefits conferred negli­gible haemodynamic benefit compared
of each device, as well as knowledge of their various with medical therapy alone60. Optimal haemodynamic
complications and limitations (TABLE 1). Uncommon benefit of an IABP depends on its position in the aorta,
conditions, such as acute fulminant myocarditis or the blood displacement volume, the balloon diameter
peripartum cardiomyopathy, are associated with a high in relation to aortic diameter, timing of balloon infla-
likelihood of recovery of myocardial function and are tion in diastole and deflation in systole, intrinsic heart
often well-suited for temporary MCS. Patients can be rate, systemic vascular resistance, and blood pressure54.
gradually weaned from MCS as ventricular function For the device to be effective, there must be a minimum
recovers, without the need for long-term mechanical level of LV function and electrical stability 37.
support or heart transplantation. For the larger cohort In 2009, a meta-analysis of several randomized
of patients with cardiogenic shock post‑MI with irre- ­trials and cohort studies reported minimal evidence-­
versible myocardial necrosis or acute decompensation based support for widespread use of IABP therapy in
of chronic systolic heart failure, the candidacy of these patients with AMI complicated by cardiogenic shock61.
patients for long-term mechanical support or heart Although no benefit was observed in the meta-­analysis
transplantation should be assessed when considering of randomized trials, the meta-analysis of cohort studies
whether a t­ emporary MCS device is suitable. demonstrated that the use of IABP with thrombolysis
was associated with an 18% reduction in mortality 61.
Intra-aortic balloon pump. From its development in The subsequent randomized IABP-SHOCK II trial8
the late 1960s, the IABP became the standard of care in evaluated the efficacy of IABP support in 600 patients
patients with cardiogenic shock. In the SHOCK trial6, with cardio­g enic shock complicating AMI, all of
85% of patients with cardiogenic shock were treated whom received early revascularization (predomi-
with an IABP. The IABP can decrease myocardial nantly PCI). No differences in mortality or complica-
oxygen consumption, increase coronary artery perfu- tion rate were detected between patients treated with
sion, enhance cardiac output by augmenting diastolic IABP and those not treated with IABP8. At present,
arterial pressure and coronary blood flow during the an IABP for cardio­genic shock complicating MI is
balloon inflation phase, and increase LV stroke vol- given a class IIa, level of evidence B recommendation
ume by lower­ing LV afterload57,58. Although preclinical in the 2013 AHA/ACC STEMI practice guidelines, a
reports have suggested that early (prerevascularization) class IIb recommendation in the ESC STEMI practice
implementation of mechanical support can reduce guidelines36,62–65, and a class III r­ ecommendation in the
infarct size, no confirmatory data in patients have been ESC non-STEMI guidelines48.

Table 1 | Temporary mechanical circulatory support devices


Device IABP Impella 2.5 Impella CP Impella 5 VA ECMO TandemHeart CentriMag Impella RP
features PTVA System
Pump Pneumatic Axial flow Axial flow Axial flow Centrifugal Centrifugal Centrifugal Axial flow
mechanism
Cannula 8 Fr 13 Fr 14 Fr 23 Fr • 18‑21 Fr • 21 Fr inflow • 32 Fr venous 9 Fr
inflow • 15–17 Fr • 22 Fr arterial
• 15–22 Fr outflow
outflow
Insertion Percutaneous; Percutaneous; Percutaneous; Surgical Percutaneous Percutaneous; Surgical; RVAD: Percutaneous;
descending femoral femoral cutdown: and surgical; inflow via inflow via RA or femoral
aorta via artery artery femoral inflow via femoral vein; RV, outflow via vein across
femoral retrograde retrograde artery femoral vein, outflow via PA; LVAD: inflow pulmonic
artery across aortic across aortic retrograde outflow via femoral artery via LA and LV, valve
valve valve across femoral artery outflow via aorta
aortic valve
Haemodynamic 0–1 l/min 2.5 l/min 3–4 l/min 5 l/min >4.5 l/min 4 l/min 5–10 l/min >2.5 l/min RV
support Biventricular Biventricular
Limb ischaemia + ++ ++ ++ +++ +++ - +

Haemolysis + ++ ++ ++ ++ ++ - ++
Afterload Reduced Neutral Neutral Neutral Increased Increased Neutral Neutral
PCWP Slightly Slightly Slightly Slightly Variable Reduced Reduced Neutral
reduced reduced reduced reduced or slightly
increased
Fr, French; IABP, intra-aortic balloon pump; LA, left atrium; LV, left ventricle; LVAD, left ventricular assist device; PA, pulmonary artery; PCWP, pulmonary capillary
wedge pressure; PTVA, percutaneous transseptal ventricular assist; RA, right atrium; RCT, randomized controlled trial; RV, right ventricle; RVAD, right ventricular
assist device; VA ECMO, veno-arterial extracorporeal membrane oxygenation.

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Rapid reversal of systemic hypoperfusion with IABP in infarct size after reperfusion with Impella 2.5 sup-
is associated with improved prognosis
100 port 68, no randomized clinical trial data demonstrating
Log-rank P <0.001, Wilcoxon P <0.001 the benefits of its use in the management of cardiogenic
90
NCRH (n = 117) shock are available. In a randomized trial comparing
80
CRH (n = 68) Impella 2.5 with IABP placement in patients with
cardiogenic shock, haemodynamics variables were
All-cause mortality (%)

70
significantly improved with Impella support, but no
60
difference in 30‑day mortality was detected (45% in
50 the IABP group versus 43% in the Impella 2.5 group);
40 of note, this study was not powered to detect differences
30
between mortality 69. In addition, no difference in major
bleeding was observed, but patients who were treated
20
with an Impella 2.5 had significantly more haemolysis
10 in the first 24 h (P <0.05 between treatment groups at
0 time points <24 h), with no increase in the requirement
0 2 4 6 8 10 12 for packed red blood cells and fresh-frozen plasma.
Time since randomization (months) One patient in the Impella 2.5 group had acute limb
Figure 3 | Rapid reversal of systemic hypoperfusion with an intra-aortic balloon ­ischaemia that required surgery 69.
pump (IABP) is associated with improved prognosis. Complete Nature Reviews
reversal Cardiology
of |systemic The TandemHeart PTVA System (CardiacAssist,
hypoperfusion 30 min after placement of an IABP is independently associated with Inc., USA) is a percutaneous ventricular assist device
markedly lower 1‑year mortality in the SHOCK trial, as compared with patients without that can be used for short-term circulatory support.
complete reversal. CRH, complete reversal systemic hypoperfusion; NCRH, The device can be inserted in the cardiac catheterization
non-complete reversal systemic hypoperfusion. Reprinted from Ramanathan, K. et al.
laboratory using a transseptal puncture, with a venous
Rapid complete reversal of systemic hypoperfusion after intra-aortic balloon pump
inflow ­cannula inserted into the left atrium, or it can
counterpulsation and survival in cardiogenic shock complicating acute myocardial
infarction. Am. Heart J. 162, 268–275 (2011) with permission from Elsevier. be implanted surgically 54. Oxygenated blood is drawn
from the left atrium and returned extracorporeally via
a centrifugal pump to the femoral artery, bypassing
Interestingly, although minimal data are available the left ventricle. LV unloading occurs by redirecting
demonstrating the efficacy of IABP therapy in patients blood from the left atrium to the ileofemoral arterial
with cardiogenic shock, lack of response to IABP system. The system is capable of delivering flow of up
counter­pulsation in patients with cardiogenic shock to 5.0 l/min, depending on the size of the femoral artery
owing to predominant LV failure post‑MI seems to be cannula. Two small randomized clinical trials comparing
a powerful prognostic marker. Incomplete reversal of IABP to the TandemHeart device in patients with cardio-
systemic hypoperfusion, defined as inability to reverse genic shock showed favourable haemodynamic variables
extremity hypoperfusion, or lack of improvement in with TandemHeart support 70,71. However, complications
mentation after 30 min of 1:1 augmentation with IABP, were more common with use of TandemHeart, includ-
were independently associated with higher inhospital ing severe bleeding (RR 2.35, 95% CI 1.40–3.93) and a
and 1‑year mortality in the SHOCK trial66 (FIG. 3). trend towards more limb ischaemia (RR 2.59, 95% CI
0.75–8.97)72. No differences in mortality were detected
New MCS devices. New temporary MCS devices have between the two treatment groups in a meta-analysis,
been developed for patients with potentially reversible but the studies analysed were not powered to detect
multiorgan failure who demonstrate ongoing hypo­ mortality differences73 (FIG. 4).
perfusion and shock despite pharmacological therapy The Impella 2.5 and TandemHeart percutaneous
and IABP. Peripherally inserted devices include axial flow devices are both resource-intensive and associated with
pumps and left atrial to left femoral artery bypass pumps, complications. Although randomized clinical trials have
which are typically implanted in a cardiac catheterization shown improved haemodynamics with use of these
setting. Veno-arterial extra­corporeal ­membrane oxygen- devices compared with IABP, meta-analyses published
ation (VA ECMO) can either be implanted peripherally in the past two years reported no demonstrable bene-
or centrally (via surgery). Surgically placed temporary fits on outcomes or costs with use of these devices72,74.
extracorporeal centrifugal pumps can ­provide full left, The 2013 ACC/AHA guidelines for the management of
right, or biventricular support. STEMI recommends that alternative LVADs might be
The Impella 2.5 (Abiomed, USA) is a temporary considered in patients with refractory cardiogenic shock
percutaneous LVAD with a nonpulsatile axial flow (class IIb, level of evidence C), although the guidelines
pump that propels blood from the left ventricle into do not specify which devices36. The consensus statement
the ascending aorta through the catheter. The Impella on the use of percutaneous MCS in cardiovascular care
2.5 is inserted percutaneously via the femoral artery. published in 2015 provides a guideline for MCS use and
The device requires adequate RV function or con- offers an expansive summary of the potential haemo-
comitant RV support to maintain LV preload and is dynamic benefits of these therapies54. Importantly, the
contra­indicated in patients with a mechanical aortic authors of this statement acknowledge the need for
valve or LV thrombus54,67. Although preclinical studies regis­tries and randomized clinical trial data to justify the
with a­ nimal models of MI have reported a reduction increase in the use of these devices in clinical practice54.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

VA ECMO or percutaneous cardiopulmonary bypass might not be adequate to reduce ventricular wall stress.
provides full haemodynamic support and might be Theoretically, this high oxygen demand has negative
helpful in biventricular failure, although the use of this consequences on myocardial protection, unless the LV
device is limited to centres with expertise in the perfu- is vented (centrally) or unloaded by concomitant IABP
sion technique. A main advantage of modern VA ECMO or percutaneous LVAD; however, no evidence showing
is its rapid implementation peripherally at the bedside that this practice can improve clinical outcomes exists75.
(outside of the catheterization laboratory or operating The most common complication reported in patients
room) and its capacity to provide therapeutic hypother- from the Extracorporeal Life Support Registry 76 is can-
mia in patients who had a cardiac arrest 54. VA ECMO nula bleeding from the peripherally placed VA ECMO
provides systemic circulatory support with flow velo­city (20.5%) or surgical bleeding in centrally placed VA
that can exceed 6 l/min, depending on cannula size54. ECMO (25.5%). Limb ischaemia is reported in up to
Owing to high myocardial oxygen demand (secondary 20% of patients in some centres, but is only partially
to high filling pressures and volume), VA ECMO alone addressed with the use of distal perfusion catheters77.
Surgically implanted extracorporeal centrifugal
pumps offer temporary support either as a LVAD, RV
LVAD IABP Cardiac index P(heterogeneity) = 0.22 assist device (RVAD), or a biventricular assist device,
mean ± SD mean ± SD mean difference I2 = 34.0%
and can be performed with or without cardiopulmonary
Thiele et al.71 2.3 ± 0.6 1.8 ± 0.4 0.55 (0.23 to 0.87) bypass. A RVAD uses the right atrium as the inflow and
Burkhoff et al.70 2.2 ± 0.6 2.1 ± 0.2 0.16 (–0.14 to 0.46) the main pulmonary artery as outflow, while a LVAD can
Seyfarth et al. 69
2.2 ± 0.6 1.8 ± 0.7 0.36 (–0.16 to 0.88) use multiple sites for inflow cannulation, including the
Pooled 0.35 (0.09 to 0.61) left atrium or the left ventricle via LV apical cannulation
or by advancing the left atrial cannula across the mitral
–2 –1 0 1 2
Favours IABP Favours LVAD valve into the ventricle78. The outflow cannula for the
LVAD is in the ascending aorta. Although these pumps
LVAD IABP Mean arterial pressure P(heterogeneity) = 0.10 are designed for short-term support, several techniques
mean ± SD mean ± SD mean difference I2 = 55.9%
allow for mobilization and prolonged use79. Data from
Thiele et al.71 76 ± 10 70 ± 16 5.5 (–2.9 to 13.9) centres with experience in using surgically implanted
Burkhoff et al.70 91 ± 16 72 ± 12 18.6 (9.4 to 27.9) extracorporeal pumps for management of refractory
Seyfarth et al.69 87 ± 18 71 ± 22 16.0 (0.5 to 31.5) cardio­genic shock (n = 143 patients) showed major
Pooled 12.8 (3.6 to 22.0) bleeding events in 33% of patients and cerebrovascular
accidents in 14%80.
–50 –25 0 25 50
Favours IABP Favours LVAD Although currently no randomized trial data support
the routine use of these more invasive and expensive
LVAD IABP Pulmonary wedge pressure P(heterogeneity) = 0.01
mean ± SD mean ± SD mean difference I2 = 76.6%
therapeutics, VA ECMO and extracorporeal ventricular
assist devices have become widely embraced at several
Thiele et al.71 16 ± 5 22 ± 7 –5.6 (–9.2 to –2.1)
tertiary-care centres. The outcomes data available in the
Burkhoff et al.70 16 ± 4 25 ± 3 –8.4 (–11.0 to –5.8) literature are mostly derived from small single-­centre
Seyfarth et al.69 19 ± 5 20 ± 6 –1.0 (–5.2 to 3.2) studies80,81. One such study investigated the use of extra-
Pooled –5.3 (–9.4 to –1.2) corporeal ventricular assist devices or VA ECMO in
–20 –10 0 10 20 90 patients with refractory cardiogenic shock attribut-
Favours LVAD Favours IABP able to AMI, chronic heart failure, or myocarditis, and
demonstrated a rate of survival to hospital discharge of
LVAD IABP 30-day mortality P(heterogeneity) = 0.83
n/N n/N relative risk I2 = 0% 49%82. How broadly applicable these results are remains
Thiele et al.71 9/21 9/20
unknown, particularly for less-experienced centres.
0.95 (0.48 to 1.90)
Burkhoff et al.70 9/19 5/14 1.33 (0.57 to 3.10) Management of RV shock
Seyfarth et al.69 6/13 6/13 1.00 (0.44 to 2.29) Optimal management of RV shock is aimed at supporting
Pooled 24/53 20/47 1.06 (0.68 to 1.66) the right ventricle and reversing any ischaemia. Medical
0.1 1 10 therapy for RV shock consists of maintaining RV preload
Favours LVAD Favours IABP with adequate volume resuscitation, restoring physio­
Figure 4 | Meta-analysis of randomized trials comparingNature the effect of | Cardiology
Reviews logical rhythm, reducing RV afterload, and enhancing
percutaneous left ventricular assist devices (LVAD) versus the intra-aortic RV contractility with inotropic support33. Treatment with
balloon pump (IABP) on 30‑day mortality and haemodynamics. Despite clear nitrates and morphine should be avoided as they reduce
improvements in haemodynamics with percutaneous LVAD compared with IABP, 30‑day RV preload. Excess fluid administration is also detrimen-
mortality appeared to be similar. However, these trials were not powered to test for tal, as it can lead to marked RV dilatation with septal shift
mortality differences. Random effect models were used for meta-analysis. For the top
into the left ventricle, which diminishes LV filling 33.
three panels, weighted mean differences with 95% confidence intervals are presented on
the right of the figure. For the bottom panel, relative risks with 95% confidence intervals In those patients with refractory RV shock despite
are presented on the right of the figure. Modified from Cheng, M. et al. Percutaneous left medical management and revascularization, treatment
ventricular assist devices versus intra-aortic balloon pump counterpulsation for options include atrial septostomy, temporary MCS, VA
treatment of cardiogenic shock: a meta-analysis of controlled trials. Eur. Heart J. 2009; ECMO, and heart transplantation. Percutaneous RVADs
30 (17): 2102–2108, with permission from Oxford University Press. can also improve haemodynamics during recovery from

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

acute RV failure83,84. The Impella RP System (Abiomed, quality-of‑life measures. A subsequent randomized
USA) is a RVAD that provides peripherally placed trial published in 2009 compared the pulsatile-flow
mechanical circulatory support for up to 14  days HeartMate XVE with the continuous-flow Heartmate II
in patients with refractory RV shock. The pump is (Thoratec Corporation, USA)89. Both devices signifi­
inserted via the femoral vein, into the right atrium, and cantly improved quality of life and functional cap­
through to the pulmonary artery, and can provide flow acity, but the continuous-flow LVAD further improved
of up to 5 l/min at a maximal speed of 32,000 rpm85. probability of stroke-free survival compared with the
The TandemHeart right ventricular support device pulsatile device. Although these two randomized trials
(CardiacAssist, Inc., USA) is a centrifugal flow pump predominantly included patients with chronic systolic
that can be implanted percutaneously via a bifemoral or heart failure, some patients probably also met criteria
right internal jugular approach, surgically by direct can- for cardiogenic shock88,89. The Seventh INTERMACS
nulation of the right atrium and pulmonary artery, or by registry 90 annual report indicated that the proportion
peripheral cannulation of the femoral vein and direct can- of patients considered to fit the profile of cardiogenic
nulation of the pulmonary artery. The use of this device shock at the time of implant with a long-term LVAD or
has been associated with an acute reduction in right heart a total artificial heart was approximately 15%. Patients
filling pressures and improved cardiac index over a 48‑h with cardiogenic shock at the time of implant have worse
period in a wide array of clinical conditions leading to postimplant survival than more stable patients, as well as
RV failure68. Finally, as with acute mechanical LV support increased postoperative length of stay 91.
devices, no available randomized trial data demonstrate
improved clinical outcomes in patients with RV shock Conclusion
with these aforementioned devices for RV support. While the overall outcomes for patients with cardiogenic
shock have improved over the past several decades,
Long-term MCS patients with refractory cardiogenic shock continue to
A clear end point must be established before instituting pose a clinical challenge. Although mechanical support
temporary support, because approximately one-third therapies are available, no randomized trial evidence
of patients who survive temporary support will do so exists to support their widespread use, and outcomes
by receiving a cardiac transplant or surgical ventricular continue to be poor even with the most invasive
assist device. The ability to progress to long-term MCS and resource-intensive therapeutics. Observational and
or cardiac transplant if cardiac function does not recover randomized data have shown improvement in haemo­
is predicated on recovery of end-organ function. Patients dynamic support with the use of temporary MCS, but no
undergoing long-term LVAD implantation in the set- improvement in mortality rates has been reported, even
ting of AMI have similar outcomes to the other patients when studies are pooled in a meta-analysis. Registries
receiving LVAD, despite being more ill before implanta- should be developed to improve the understanding of
tion86. Furthermore, retrospective data suggest that no practice patterns across the spectrum of hospitals (from
differences exist in long-term LVAD outcome between community to quaternary) and to serve as a platform for
early and late ventricular assist device i­mplantation pragmatic trials to answer unresolved questions. In light
after MI87. of the persistently high mortality associated with refrac-
The landmark, randomized REMATCH trial88 was tory cardiogenic shock, the lack of incremental improve-
designed to evaluate the suitability of the HeartMate ment in mortality despite use of PCI, and the high cost of
XVE (vented electric) LVAD (Thoratec Corporation, current treatment patterns, there is a critical need to find
USA) for long-term use in patients who were ineligi- a treatment strategy besides reperfusion or revascular­
ble for cardiac transplantation. The LVAD conferred ization that can improve outcomes for patients with
a clinically meaningful survival benefit and improved cardiogenic shock.

1. Hasdai, D. Cardiogenic shock: diagnosis and 7. Holmes, D. R. et al. GUSTO‑I Investigators. 13. Magnani, J. W. & Dec, G. W. Myocarditis: current
treatment (Humana Press, 2002). Contemporary reperfusion therapy for cardiogenic trends in diagnosis and treatment. Circulation 116,
2. De Luca, L. et al. Temporal trends in the epidemiology, shock: the Gusto‑I trial experience. J. Am. Coll. 876–890 (2006).
management, and outcome of patients with Cardiol. 26, 668–674 (1995). 14. Dec, G. W. et al. Viral myocarditis mimicking acute
cardiogenic shock complicating acute coronary 8. Thiele, H. et al. Intra-aortic balloon support for myocardial infarction. J. Am. Coll. Cardiol. 20, 85–89
syndromes. Eur. J. Heart Fail. 17, 1124–1132 (2015). myocardial infarction with cardiogenic shock. N. Engl. (1992).
3. Dzavik, V. et al. Effects of nitric oxide synthase J. Med. 367, 1287–1296 (2012). 15. Felker, G. M. et al. Echocardiographic findings in
inhibition on hemodynamics and outcome of patients 9. Kar, B., Gregoric, I. D., Basra, S. S., Idelchik, G. M. fulminant and acute myocarditis. J. Am. Coll. Cardiol.
with persistent cardiogenic shock complicating acute & Loyalka, P. The percutaneous ventricular assist 36, 227–232 (2000).
myocardial infarction: a phase II dose-ranging study. device in severe refractory cardiogenic shock. J. Am. 16. Angelini, A. et al. Myocarditis mimicking acute
Eur. Heart J. 28, 1109–1116 (2007). Coll. Cardiol. 57, 688–696 (2011). myocardial infarction: role of endomyocardial biopsy in
4. Kohsaka, S. et al. Systemic inflammatory response 10. Laslett, L. J. et al. The worldwide environment of the differential diagnosis. Heart 84, 245–250 (2000).
syndrome after acute myocardial infarction cardiovascular disease: prevalence, diagnosis, therapy 17. Hasumann, H. et al. Prognosis after implantation
complicated by cardiogenic shock. Arch. Intern. Med. and policy issues. J. Am. Coll. Cardiol. 60, S1–S49 of an intra-aortic balloon pump in cardiac surgery
165, 1643–1650 (2005). (2012). calculated with a new score. Circulation 106,
5. Hochman, J. S. et al. Cardiogenic shock complicating 11. Babaev, A. et al. Trends in management and 203–206 (2002).
acute myocardial infarction — etiologies, outcomes of patients with acute myocardial infarction 18. Torchiana, D. F. et al. Intra-aortic balloon pumping
management, and outcome: a report from the SHOCK complicated by cardiogenic shock. JAMA 294, for cardiac support: trends in practice and outcome,
trial registry. J. Am. Coll. Cardiol. 36, 1063–1070 448–454 (2005). 1968 to 1995. J. Thorac. Cardiovasc. Surg. 113,
(2000). 12. Webb, J. G. et al. Implications of the timing of onset 758–764 (1997).
6. Hochman, J. S. et al. Early revascularization in acute of cardiogenic shock after acute myocardial infarction: 19. Goldstein, D. J. & Oz, M. C. Mechanical support for
myocardial infarction complicated by cardiogenic a report from the SHOCK trial registry. J. Am. Coll. postcardiotomy cardiogenic shock. Semin. Thorac.
shock. N. Engl. J. Med. 341, 625–634 (1999). Cardiol. 36, 1084–1090 (2000). Cardiovasc. Surg. 12, 220–228 (2000).

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

20. Rao, V. et al. Surgery for acquired heart disease: 44. Menon, V. et al. Outcome and profile of ventricular 63. Sanborn, T. A. et al. Impact of thrombolysis,
predictors of low cardiac output syndrome after septal rupture with cardiogenic shock after myocardial intraaortic balloon pump counterpulsation, and their
coronary artery bypass. J. Thorac. Cardiovasc. Surg. infarction: a report from the shock trial registry. J. Am. combination in cardiogenic shock complicating acute
112, 38–51 (1996). Coll. Cardiol. 36, 1110–1116 (2000). myocardial infarction: a report from the SHOCK Trial
21. Alsoufi, B., Rao, V., Tang, A., Maganti, M. 45. Goldberg, R. J., Spencer, F. A., Gore, J. M., Lessard, D. Registry. SHould we emergently revascularize
& Cusimano, R. Risk modeling for ventricular assist & Yarzebski, Y. Thirty-year trends (1975 to 2005) in Occluded Coronaries for cardiogenic shocK?
device support in post-cardiotomy shock. J. Saudi the magnitude of, management of, and hospital death J. Am. Coll. Cardiol. 36, 1123–1129 (2000).
Heart Assoc. 24, 69–72 (2012). rates associated with cardiogenic shock in patients 64. Romeo, A. et al. The outcome of intra-aortic balloon
22. Mohite, P. N. et al. Short-term ventricular assist device with acute myocardial infarction: a population-based pump support in acute myocardial infarction
in post-cardiotomy cardiogenic shock: factors perspective. Circulation 119, 1211–1219 (2009). complicated by cardiogenic shock according to
influencing survival. J. Artif. Organs. 17, 228–235 46. Killip, T. & Kimball, J. T. Treatment of myocardial the type of revascularization: a comprehensive
(2014). infarction in a coronary care unit. A two year meta‑analysis. Am. Heart J. 165, 679 (2013).
23. Jacobs, A. K. et al. Cardiogenic shock caused by right experience with 250 patients. Am. J. Cardiol. 20, 65. Steg, G. P. et al. ESC guidelines for the management of
ventricular infarction: a report from the SHOCK 457–464 (1967). acute myocardial infarction in patients presenting with
registry. J. Am. Coll. Cardiol. 41, 1273–1279 (2003). 47. Wayangankar, S. A. et al. Temporal trends and ST‑segment elevation. Eur. Heart J. 33, 2569–2619
24. Zehender, M. et al. Right ventricular infarction as an outcomes of patients undergoing percutaneous (2012).
independent predictor of prognosis after acute inferior coronary interventions for cardiogenic shock in the 66. Ramanathan, K. et al. Rapid complete reversal of
myocardial infarction. N. Engl. J. Med. 328, 981–988 setting of acute myocardial infarction: a report from systemic hypoperfusion after intra-aortic balloon
(1993). the CathPCI Registry. JACC Cardiovasc. Interv. 9, pump counterpulsation and survival in cardiogenic
25. Bowers, T. R. et al. Effect of reperfusion on biventricular 341–351 (2016). shock complicating an acute myocardial infarction.
function and survival after right ventricular infarction. 48. Roffi, M. et al. 2015 ESC guidelines for the Am. Heart J. 162, 268–275 (2011).
N. Engl. J. Med. 338, 933–940 (1998). management of acute coronary syndromes in patients 67. Ostadal, P. et al. Direct comparison of percutaneous
26. Bowers, T. R. et al. Patterns of coronary compromise presenting without persistent ST‑segment elevation: circulatory support systems in specific hemodynamic
resulting in acute right ventricular ischemic task force for the management of acute coronary conditions in a porcine model. Circ. Arrhythm.
dysfunction. Circulation 106, 1104–1109 (2002). syndromes without persistent ST‑segment elevation of Electrophysiol. 5, 1202–1206 (2012).
27. Dell’Italia, L. J. et al. Hemodynamically important right the European Society of Cardiology (ESC). Eur. Heart J. 68. Kapur, N. K. et al. Mechanical circulatory support for
ventricular infarction: follow up evaluation of right 37, 267–315 (2016). right ventricular failure. JACC Heart Fail. 1, 127–134
ventricular systolic function at rest and during exercise 49. Sanborn, T. A. et al. Correlates of one-year survival in (2013).
with radionuclide ventriculography and respiratory gas patients with cardiogenic shock complicating acute 69. Seyfarth, M. et al. A randomized clinical trial to
exchange. Circulation 75, 996–1003 (1987). myocardial infarction: angiographic findings from the evaluate the safety and efficacy of a percutaneous left
28. Page, D. L., Caulifield, J. B., Kaster, J. A., SHOCK trial. J. Am. Coll. Cardiol. 42, 1373–1379 ventricular assist device versus intra-aortic balloon
DeSanctis, R. W. & Sanders, C. A. Myocardial changes (2003). pumping for treatment of cardiogenic shock caused
associated with cardiogenic shock. N. Engl. J. Med. 50. Hochman, J. S. et al. Early revascularization and by myocardial infarction. J. Am. Coll. Cardiol. 52,
285, 133–137 (1971). long‑term survival in cardiogenic shock complicating 1584–1588 (2008).
29. Hollenberg, S. M., Kavinsky, C. J. & Parrillo, J. E. acute myocardial infarction. JAMA 295, 2511–2515 70. Burkhoff, D., Cohen, H., Brunckhorst, C. & O’Neill, W.
Cardiogenic shock. Ann. Intern. Med. 131, 47–59 (2006). W. A randomized multicenter clinical study to evaluate
(1999). 51. Mylotte, D. et al. Primary percutaneous coronary the safety and efficacy of the TandemHeart
30. Neumann, F. J. et al. Cardiac release of cytokines and intervention in patients with acute myocardial percutaneous ventricular assist device versus
inflammatory response in acute myocardial infarction. infarction, resuscitated cardiac arrest, and conventional therapy with intraaortic balloon pumping
Circulation 92, 748–755 (1995). cardiogenic shock: the role of primary multivessel for treatment of cardiogenic shock. Am. Heart J. 152,
31. Nicholls, S. J. et al. Metabolic profiling of arginine revascularization. JACC Cardiovasc. Interv. 6, 469.e1–469.e8 (2006).
and nitric oxide pathways predicts hemodynamic 115–125 (2013). 71. Thiele, H. et al. Randomized comparison of intra-
abnormalities and mortality in patients with 52. US National Library of Medicine. Clinical-Trials.gov, aortic balloon support with a percutaneous left
cardiogenic shock after acute myocardial infarction. https://clinicaltrials.gov/show/NCT01927549 (2015). ventricular assist device in patients with
Circulation 116, 2315–2324 (2007). 53. De Backer, D. et al. Comparison of dopamine and revascularized acute myocardial infarction
32. Ratshin, R. A., Rackley, C. E. & Russel, R. O. norepinephrine in the treatment of shock. N. Engl. complicated by cardiogenic shock. Eur. Heart J. 26,
Hemodynamic evaluation of left ventricular function J. Med. 362, 779–789 (2010). 1276–1283 (2005).
in shock complicating myocardial infarction. 54. Rihal, C. S. et al. 2015 SCAI/ACC/HFSA/STS clinical 72. Stretch, R., Sauer, C. M., Yuh, D. D. & Bonde, P.
Circulation 45, 127–139 (1972). expert consensus statement on the use of National trends in the utilization of short-term
33. Hochman, J. S. & Ohman, E. M. Cardiogenic shock percutaneous mechanical circulatory support devices mechanical circulatory support: incidence, outcomes,
(AHA Clinical Series, 2009). in cardiovascular care (endorsed by the American and cost analysis. J. Am. Coll. Cardiol. 64,
34. Reynolds, H. R. et al. Restrictive physiology Heart Association, the Cardiological Society of India, 1407–1415 (2014).
in cardiogenic shock: observations from and Sociedad Latino Americana de Cardiologia 73. Cheng, J. M. et al. Percutaneous left ventricular assist
echocardiography. Am. Heart J. 151, 890.e9–890.e15 Intervencion; affirmation of value by the Canadian devices versus intra-aortic balloon pump
(2006). Association of Interventional Cardiology-Association counterpulsation for treatment of cardiogenic shock:
35. Menon, V. et al. The clinical profile of patients with Canadienne de Cardiologie d’intervention). J. Cardiac a meta-analysis of controlled trials. Eur. Heart J. 30,
suspected cardiogenic shock due to predominant left Fail. 21, 499–518 (2015). 2102–2108 (2009).
ventricular failure: a report form the SHOCK trial 55. Scheidt, S. et al. Intra-aortic balloon counterpulsation 74. Shah, A. P. et al. Clinical and economic effectiveness
registry. J. Am. Coll. Cardiol. 36, 1071–1076 (2000). in cardiogenic shock. Report of a co‑operative clinical of percutaneous ventricular assist devices for high-risk
36. O’Gara, P. T. et al. 2013 ACCF/AHA guideline for the trial. N. Engl. J. Med. 288, 979–984 (1973). patients undergoing coronary intervention. J. Invasive
management of ST‑elevation myocardial infarction: 56. Agarwal, S., Sud, K., Martin, J. M. & Menon, V. Trends Cardiol. 27, 148–154 (2015).
a report of the American College of Cardiology in the use of mechanical circulatory support devices 75. Cheng, R. et al. Lack of survival benefit found with use
Foundation/American Heart Association task force in patients presenting with ST‑segment elevation of intraaoartic balloon pump in extracorporeal
on practice guidelines. Circulation 127, 362–425 myocardial infarction. JACC Cardiovasc. Interv. 8, membrane oxygenation: a pooled experience of 1517
(2013). 1772–1774 (2015). patients. J. Invasive Cardiol. 27, 453–458 (2015).
37. Jeger, R. V. et al. Causes of death and 57. Chatterjee, S. & Rosensweig, J. Evaluation of 76. Paden, M. L., Conrad, S. A., Rycus, P. T.
re‑hospitalization in cardiogenic shock. Acute Card. intra‑aortic balloon counterpulsation. J. Thorac & Thiagarajan, R. R. Extracorporeal life support
Care 9, 25–33 (2007). Cardiovasc. Surg. 61, 405–410 (1971). organization registry report 2012. ASAIO J. 59,
38. Sleeper, L. A. et al. A severity scoring system for 58. Kantrowitz, A. et al. Current status of intraaortic 202–210 (2013).
risk assessment of patients with cardiogenic shock: balloon pump and initial clinical experience with aortic 77. Mohite, P. N. et al. Distal limb perfusion: Achilles’ heel
a report form the SHOCK trial and registry. patch mechanical auxiliary ventricle. Transplant. Proc. in peripheral venoarterial extracorporeal membrane
Am. Heart J. 160, 443–450 (2010). 3, 1459–1471 (1971). oxygenation. Artif. Organs 38, 940–944 (2014).
39. The TRIUMPH Investigators et al. Effect of tilarginine 59. Meyns, B., Stolinski, J., Leunens, V., Verbeken, E. 78. Takayama, H. et al. Bridge-to‑decision therapy with
acetate in patients with acute myocardial infarction & Flameng, W. Left ventricular support by a continuous-flow external ventricular assist device
and cardiogenic shock: the TRIUMPH randomized catheter‑mountedaxial flow pump reduces infarct size. in refractory cardiogenic shock of various causes.
controlled trial. JAMA 297, 1657–1666 (2007). J. Am. Coll. Cardiol. 41, 1087–1095 (2003). Circulation 7, 799–806 (2014).
40. Katz, J. N. et al. Predictors of 30‑day mortality in 60. Prondzinsky, R. et al. Hemodynamic effects of intra- 79. Takayama, H., Chen, J. M., Jorde, U. P. & Naka, Y.
patients with refractory cardiogenic shock following aortic balloon counterpulsation in patients with acute Implantation technique of the CentriMag biventricular
acute myocardial infarction despite a patent infarct myocardial infarction complicated by cardiogenic assist device allowing ambulatory rehabilitation.
artery. Am. Heart J. 158, 680–687 (2009). shock: the prospective randomized IABP Shock Trial. Interact. Cardiovasc. Thorac. Surg. 12, 110–111
41. Harjola, V. et al. Clinical picture and risk prediction Shock 37, 378–384 (2012). (2011).
of short-term mortality in cardiogenic shock. 61. Sjauw, K. D. et al. A systematic review and meta- 80. Diez-Villanueva, P. et al. Early treatment of refractory
Eur. J. Heart Fail. 17, 501–509 (2015). analysis of intra-aortic balloon pumpo therapy in cardiogenic shock with percutaneous veno-arterial
42. Kapur, N. K. et al. TCT‑196 The recover right trial ST‑elevation myocardial infarction: should we change ECMO implanted in the cardiac catheterization
criteria for right ventricular failure: an analysis of the guidelines? Eur. Heart J. 30, 459–468 (2009). laboratory. Rev. Esp. Cardiol. (Engl. Ed.) 67,
the SHould we emergently revascularize Occluded 62. Ohman, E. M. et al. Thrombolysis and 1059–1061 (2014).
coronaries for Cardiogenic shock (SHOCK) trial and counterpulsation to improve survival in myocardial 81. Hsu, P. S. et al. Extracorporeal membrane
registry. J. Am. Coll. Cardiol. 66 (15_S), 66 (2015). infarction complicated by hypotension and suspected oxygenation for refractory cardiogenic shock after
43. Picard, M. H. et al. Echocardiographic predictors of cardiogenic shock or heart failure: results of the cardiac surgery: predictors of early mortality and
survival and response to early revascularization in TACTICS trial. J. Thromb. Thrombolysis 19, 33–39 outcome from 51 adult patients. Eur. J. Cardiothorac.
cardiogenic shock. Circulation 107, 279–284 (2003). (2005). Surg. 37, 328–333 (2010).

NATURE REVIEWS | CARDIOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

82. Takayama, H. et al. Clinical outcome of mechanical support for recovery from acute right ventricular 90. Kirklin, J. K. et al. Seventh INTERMACS annual report:
circulatory support for refractory cardiogenic shock failure: clinical outcomes. J. Heart Lung Transplant. 15,000 patients and counting. J. Heart
in the current era. J. Heart Lung Transplant. 32, 33, 794–799 (2014). Lung Transplant. 34, 1495–1504 (2015).
106–111 (2013). 86. Acharya, D. et al. Ventricular assist device in acute 91. Cotts, W. G. et al. Predictors of hospital length of
83. Cheung, A., Freed, D., Hunziker, P. myocardial infarction-findings from INTERMACS. stay after implantation of a left ventricular
& Leprince, P. TCT‑371 first clinical evaluation of a J. Heart Lung Transplant. 34, 4S (2015). assist device: an analysis of the INTERMACS
novel percutaneous right ventricular assist device: 87. Chen, J. M. et al. Improved survival rates support left registry. J. Heart Lung Transplant. 33, 682–688
the Impella RP. J. Am. Coll. Cardiol. 59 (Suppl. 1), ventricular assist device implantation early after (2014).
E872 (2012). myocardial infarction. J. Am. Coll. Cardiol. 33,
84. Atiemo, A. D., Conte, J. V. & Heldman, A. W. 1903–1908 (1999). Author contributions
Resuscitation and recovery from acute right 88. Rose, E. A. et al. Long-term use of a left ventricular All authors researched data for the article, made substantial
ventricular failure using a percutaneous right assist device for end-stage heart failure. N. Engl. contribution to discussion of the content, and wrote,
ventricular assist device. Catheter. Cardiovasc. Interv. J. Med. 345, 1435–1443 (2001). reviewed, and edited the manuscript before submission.
66, 78–82 (2006). 89. Slaughter, M. S. et al. Advanced heart failure treated
85. Cheung, A. W., White, C. W., Davis, M. K. with continuous-flow left ventricular assist device. Competing interests statement
& Freed, D. H. Short-term mechanical circulatory N. Engl. J. Med. 361, 2241–2251 (2009). The authors declare no competing interests.

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrcardio


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Вам также может понравиться