reviewarticle diastolic heart failure in …...systolic heart failure.14 55 93 recent studies...

85
REVIEWARTICLE Diastolic heart failure in anaesthesia and critical care R. Pirracchio 1 , B. Cholley 1 , S. De Hert 3 , A. Cohen Solal 2 and A. Mebazaa 1 * 1 Departments of Anaesthesiology and Critical Care and 2 Cardiology, Lariboisie `re University Hospital, University Paris VII, Paris, France. 3 Department of Anaesthesiology, University Hospital Antwerp, Edegem, Belgium *Corresponding author: De ´partement d’anesthe ´sie-re ´animation, Ho ˆpital Lariboisie `re, 2 rue Ambroise Pare ´, 75010 Paris, France. E-mail: [email protected] Diastolic heart failure is an underestimated pathology with a high risk of acute decompensation during the perioperative period. This article reviews the epidemiology, risk factors, pathophysiology, and treatment of diastolic heart failure. Although frequently underestimated, diastolic heart failure is a common pathology. Diastolic heart failure involves heart failure with preserved left ventricular (LV) function, and LV diastolic dysfunction may account for acute heart failure occurring in critical care situations. Hypertensive crisis, sepsis, and myocardial ischaemia are frequently associated with acute diastolic heart failure. Symptomatic treatment focuses on the reduction in pulmonary conges- tion and the improvement in LV filling. Specific treatment is actually lacking, but encouraging data are emerging concerning the use of renin–angiotensin–aldosterone axis blockers, nitric oxide donors, or, very recently, new agents specifically targeting actin–myosin cross-bridges. Br J Anaesth 2007; 98: 707–21 Keywords: assessment, preanaesthetic; complications, hypertension; complications, pulmonary oedema; heart, inodilators; heart, myocardial function Left ventricular (LV) diastolic dysfunction refers to abnormalities of diastolic distensibility, filling, or relaxation, regardless of whether LV ejection fraction (LVEF) is normal or abnormal and whether the patient is symptomatic or not. 8 If signs and symptoms of congestive heart failure (effort intolerance, dyspnoea, and pulmonary oedema) develop in a patient with normal or near normal systolic function, it is appropriate to classify this situation as dia- stolic heart failure. Chronic diastolic heart failure and decompensated diastolic heart failure are common entities, but their frequency is widely underestimated. Underestimation of diastolic heart failure might be related in part to the difficulties in obtaining the criteria defined by AHA-ACC guidelines, 52 requiring objective evidence using echocardiography. In addition, the poor sensitivity and specificity of echocardiography to recognize diastolic heart failure may aggravate this underestimation. There is increas- ing evidence that clinicians should differentiate systolic from diastolic heart failure since pathophysiology and man- agement may differ greatly between the two entities. 110 Since patients with diastolic heart failure are at high risk of decompensation in the perioperative period or during an ICU stay, 84 anaesthetists should be familiar with the pathophysiology of diastolic heart failure. Epidemiology of diastolic heart failure Between 30% and 50% of patients with chronic heart failure have preserved LVEF. 14 73 81 110 Two recent cohorts of patients hospitalized with decompensated heart failure showed that 35% had a preserved LVEF, 66 and that patients with preserved LVEF were 2–4 yr older than those with impaired ejection fraction and were mainly women (70%). Diastolic heart failure represents ,15% of chronic heart failure in patients younger than 50 yr, and the pro- portion raises to 33% in 50–70 yr olds and to 70% in those aged .70 yr. 120 This prevalence in elderly subjects is related to alterations in the cardiovascular system frequently associated with ageing, namely, coronary artery disease, systemic hypertension, hypertrophic, and infiltrative cardio- myopathies that cause structural changes of the LV leading to chronic deterioration of LV diastolic properties. 3 Diastolic heart failure is a condition that greatly affects patient outcome. Several authors have shown that the read- mission rate and mortality rate were high in diastolic heart failure patients and might be similar to those observed in systolic heart failure. 14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within 6 months after hospital discharge for # The Board of Management and Trustees of the British Journal of Anaesthesia 2007. All rights reserved. For Permissions, please e-mail: [email protected] British Journal of Anaesthesia 98 (6): 707–21 (2007) doi:10.1093/bja/aem098 Advance Access publication April 28, 2007 at KwaZulu-Natal University on January 21, 2011 bja.oxfordjournals.org Downloaded from

Upload: others

Post on 25-May-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

REVIEWARTICLE

Diastolic heart failure in anaesthesia and critical care

R. Pirracchio1, B. Cholley1, S. De Hert3, A. Cohen Solal2 and A. Mebazaa1*

1Departments of Anaesthesiology and Critical Care and 2Cardiology, Lariboisiere University Hospital,

University Paris VII, Paris, France. 3Department of Anaesthesiology, University Hospital Antwerp,

Edegem, Belgium

*Corresponding author: Departement d’anesthesie-reanimation, Hopital Lariboisiere, 2 rue Ambroise Pare,

75010 Paris, France. E-mail: [email protected]

Diastolic heart failure is an underestimated pathology with a high risk of acute decompensation

during the perioperative period. This article reviews the epidemiology, risk factors, pathophysiology,

and treatment of diastolic heart failure. Although frequently underestimated, diastolic heart failure

is a common pathology. Diastolic heart failure involves heart failure with preserved left ventricular

(LV) function, and LV diastolic dysfunction may account for acute heart failure occurring in critical

care situations. Hypertensive crisis, sepsis, and myocardial ischaemia are frequently associated with

acute diastolic heart failure. Symptomatic treatment focuses on the reduction in pulmonary conges-

tion and the improvement in LV filling. Specific treatment is actually lacking, but encouraging data

are emerging concerning the use of renin–angiotensin–aldosterone axis blockers, nitric oxide

donors, or, very recently, new agents specifically targeting actin–myosin cross-bridges.

Br J Anaesth 2007; 98: 707–21

Keywords: assessment, preanaesthetic; complications, hypertension; complications, pulmonary

oedema; heart, inodilators; heart, myocardial function

Left ventricular (LV) diastolic dysfunction refers to

abnormalities of diastolic distensibility, filling, or relaxation,

regardless of whether LV ejection fraction (LVEF) is

normal or abnormal and whether the patient is symptomatic

or not.8 If signs and symptoms of congestive heart failure

(effort intolerance, dyspnoea, and pulmonary oedema)

develop in a patient with normal or near normal systolic

function, it is appropriate to classify this situation as dia-

stolic heart failure. Chronic diastolic heart failure and

decompensated diastolic heart failure are common entities,

but their frequency is widely underestimated.

Underestimation of diastolic heart failure might be related in

part to the difficulties in obtaining the criteria defined by

AHA-ACC guidelines,52 requiring objective evidence using

echocardiography. In addition, the poor sensitivity and

specificity of echocardiography to recognize diastolic heart

failure may aggravate this underestimation. There is increas-

ing evidence that clinicians should differentiate systolic

from diastolic heart failure since pathophysiology and man-

agement may differ greatly between the two entities.110

Since patients with diastolic heart failure are at high

risk of decompensation in the perioperative period or

during an ICU stay,84 anaesthetists should be familiar with

the pathophysiology of diastolic heart failure.

Epidemiology of diastolic heart failure

Between 30% and 50% of patients with chronic heart

failure have preserved LVEF.14 73 81 110 Two recent cohorts

of patients hospitalized with decompensated heart failure

showed that 35% had a preserved LVEF,66 and that patients

with preserved LVEF were 2–4 yr older than those with

impaired ejection fraction and were mainly women

(�70%). Diastolic heart failure represents ,15% of chronic

heart failure in patients younger than 50 yr, and the pro-

portion raises to 33% in 50–70 yr olds and to 70% in those

aged .70 yr.120 This prevalence in elderly subjects is

related to alterations in the cardiovascular system frequently

associated with ageing, namely, coronary artery disease,

systemic hypertension, hypertrophic, and infiltrative cardio-

myopathies that cause structural changes of the LV leading

to chronic deterioration of LV diastolic properties.3

Diastolic heart failure is a condition that greatly affects

patient outcome. Several authors have shown that the read-

mission rate and mortality rate were high in diastolic heart

failure patients and might be similar to those observed in

systolic heart failure.14 55 93 Recent studies reported that

16–30% of patients with diastolic heart failure had to be

readmitted within 6 months after hospital discharge for

# The Board of Management and Trustees of the British Journal of Anaesthesia 2007. All rights reserved. For Permissions, please e-mail: [email protected]

British Journal of Anaesthesia 98 (6): 707–21 (2007)

doi:10.1093/bja/aem098 Advance Access publication April 28, 2007

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 2: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

chronic heart failure symptoms, a figure that is comparable

with the 22% observed in patients with impaired

LVEF.2 95 120 Mortality rates in diastolic heart failure

patients at 1 and 3 yr seem slightly lower than or compar-

able with those in systolic heart failure.2 14 18 118

Pathophysiology of LV diastolic dysfunction

LV diastolic dysfunction can be defined as the inability of

the LV chamber to fill up at low atrial pressures. This dys-

function can result either from an impairment in LV com-

pliance (passive mechanism) or from an alteration in LV

relaxation (active process). Relaxation is usually the first

to alter in LV diastolic dysfunction and relaxation abnorm-

alities can occur abruptly, especially in the context of

anaesthesia or critical care.

Physiology and pathophysiology of LV relaxation

Zile and Brutsaert120 defined relaxation as ‘the time period

during which the myocardium loses its ability to generate

force and shorten and returns to an unstressed length and

force’. From a mechanical point of view, the transition

between systole and diastole was described as the time of

aortic valve closure. However, the transition between con-

traction and relaxation corresponds to the dissociation of

actin–myosin cross-bridges that begins during the early

phase of LV ejection, before aortic valve closure.40

The dissociation of actin–myosin cross-bridges follows the

lowering of the intracellular calcium concentration (Fig. 1).

The transfer of calcium from the cytosole into the sarco-

plasmic reticulum requires energy. Phosphorylation of

phospholamban that is needed to activate the ATPase-

induced calcium sequestration into the sarcoplasmic

reticulum, sodium/calcium exchanger-induced extrusion of

calcium from the cytoplasm, release of calcium from tro-

ponin C, detachment of the actin–myosin cross-bridges,

and return of the sarcomere to its resting length are all

energy-consuming processes. Thus, alterations leading to

diastolic dysfunction may involve phenomena that occur not

only during ‘classic’ diastole, but also earlier in the cardiac

cycle, at the time when intracellular calcium falls.

Functional factors are likely to occur during the peri-

operative period or in the ICU. Affecting LV relaxation,

they may precipitate latent LV diastolic dysfunction.

Myocyte energy imbalance

Since relaxation is an energy-consuming process, it is

adversely affected by myocardial ischaemia. Ischaemia

precludes optimal calcium exchanges between the cytosole

and sarcoplasmic reticulum, and is rapidly associated with

impairment in LV relaxation.100 Sepsis is also likely to

alter myocytes energetic balance and, thus, to alter LV

relaxation.85

Contraction–relaxation coupling

As described earlier, relaxation begins early during

systole40 98 indicating that relaxation and contraction are

Fig 1 Calcium-induced calcium release in cardiac myocytes. After electrical stimulation, calcium influx through the calcium channels (Ica) stimulates

ryanodin receptors (RyR) to release more calcium from the sarcoplasmic reticulum (SR) into the cytosol. Calcium concentration falls (i) by reuptake in the

SR via phospholamban (PLB) stimulation and (ii) by calcium efflux by Naþ/Ca2þ exchange (NCX). The inset summarizes the time course: action potential

precedes calcium influx and the peak of myocyte contraction is seen while intracellular calcium concentration falls. From Bers and Despa12 with permission.

Pirracchio et al.

708

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 3: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

intimately coupled. As a consequence, LV relaxation is

greatly affected by the lack of homogeneity in LV contrac-

tion. Both LV segmental coordination and atrio-ventricular

synchronization are essential to guarantee an efficient

relaxation.13 34 The loss of atrial contraction associated

with atrial fibrillation not only alters LV filling but also

results in a slowing of myocardial relaxation.

Several other factors known to alter contractile function,

including changes in afterload and the use of inotropes,

markedly affect relaxation. However, the effect of preload

variations on relaxation is not clear.

Impact of afterload conditions on relaxation

Afterload dependency

In failing hearts, an increase in afterload induces a delay

in the onset of relaxation and an increase in the time-

constant of isovolumetric relaxation (t, ms).16 34 42 The

afterload-dependence of t has also been shown to be

influenced by the inotropic state. Facing an increased

afterload, beta-adrenoreceptor agonists help to keep t

constant, whereas antagonists (beta-blockers) markedly

increased it.15

To describe better the relationship between load, con-

tractility, and relaxation, the concept of relative load was

defined experimentally as the ratio of peak systolic LV

pressure to peak isovolumetric pressure.38 40 58 The higher

the relative load, the lower the contractile reserve

(Fig. 2A). The contractile reserve quantifies the percentage

force developed by the LV with respect to its maximum

value. The consequences on relaxation of an elevation in

LV afterload can range from a moderate acceleration to a

marked deceleration, depending on the relative load. Up to

a relative load of around 80%, the diastolic decline in LV

pressure accelerates, but above this, LV pressure decline

decelerates. Acceleration of the LV pressure decrease in

response to a load elevation is observed in the normal

heart, whereas slowing of the LV pressure decline is

associated with impaired cardiac function.31 This intro-

duced the concept of afterload reserve which relates to

the capacity of the normal LV to respond to elevation of

afterload without changes in LV end-systolic volume and

LV pressure decline.38 39 58 Ventricles with altered con-

tractile function consistently show a decreased ‘afterload

reserve’.21 22 24 27 31 In such ventricles, even a small after-

load elevation will cause a marked deterioration in LV

relaxation and increase LV systolic and diastolic volumes.

An alternative concept: end-systolic volume dependency

Chemla and colleagues17 recently proposed an alternative

approach based on the suggestion that, at constant heart

rate, relaxation might depend more on LV end-systolic

volume than on afterload, namely LV systolic pressure.

Indeed, recoiling forces are generated when the LV con-

tracts below its equilibrium volume (usually slightly

higher than LV end-systolic volume) and therefore

recoiling forces act during early diastole. Thus, since a

healthy heart is able to respond to increased afterload

without any change in its LV end-systolic volume, relax-

ation remains unaffected. However, in failing dilated ven-

tricles, LV end-systolic volume might exceed the

equilibrium volume, which deprives the LV of recoiling

forces and impairs the rate of isovolumetric relaxation.

Impact of preload conditions

Myocardial relaxation was initially considered not to be

affected by preload conditions.35 However, it has been

suggested that marked preload variation can modify actin–

myosin cross-bridge kinetics.41 Indeed, in a patient with

LV diastolic dysfunction, a leg elevation manoeuvre can

result in a substantial increase in end-diastolic pressure

(Fig. 2B). This case report suggests that LV diastolic dys-

function can be undetected in normo- or hypovolaemic

patient. In contrast, any excess in fluid can induce a dra-

matic increase in LV end-diastolic pressure (LVEDP).

Thus, preload reserve is reduced in patients with altered

LV diastolic properties.

Analysis of diastole by cardiac catheterization

Diastolic heart failure is characterized by an elevated

LVEDP (16–26 mm Hg) in 92% of patients,108 121 whereas

control patients had an average of 10 mm Hg. Using

micromanometer catheters, it is possible to acquire high-

fidelity instantaneous LV pressure curves. Such tools are

used to plot pressure/volume loops and to assess the rate of

LV pressure decline (dP/dtmin) and the time constant of

isovolumetric relaxation (t).

Pressure/volume loops

Systolic and diastolic functions are best described using

the LV pressure/volume relationship, represented graphi-

cally as P/V loops (Fig. 3). In animals, a series of

pressure/volume loops can be measured at baseline and

after changes in preload (by inferior vena cava occlusion).

The latter allow measurements of preload-independent

variables as the end-systolic pressure/volume relationship

and end-diastolic pressure/volume relationship.

In humans, pressure/volume tracings are usually per-

formed only at baseline. In one study,56 pressure/volume

loops were performed at baseline and during a sustained

handgrip manoeuvre (Fig. 4).

Using this mechanical approach, diastole begins at the

closure of the aortic valve and lasts until the closure of the

mitral valve. Diastole can be divided into two phases.

The first corresponds to the LV pressure decline at con-

stant volume, isovolumetric relaxation, which lasts from

closure of the aortic valve to opening of the mitral valve.

The second, auxotonic relaxation, corresponds to LV

chamber filling and lasts until the closure of the mitral

valve. LV filling mainly depends on the pressure gradient

between the left atrium (LA) and LV, which is mainly

Diastolic heart failure in anaesthesia and critical care

709

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 4: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

influenced by passive chamber properties (compliance),

active relaxation, and, at end-diastole, by atrial contraction.

Thus, impairment of LV compliance (decreased LA–LV

pressure gradient), or the loss of atrial contraction, directly

impair diastolic filling. Structural modifications (i.e. myo-

cardial hypertrophy, fibrosis) mainly affect the passive,

late phase of diastole and are more likely to develop

chronically, whereas functional factors (i.e. ischaemia,

sepsis) adversely affect active relaxation during early dia-

stole. In LV diastolic dysfunction, the diastolic portion of

the pressure/volume loop (compliance curve) is shifted to

the left and upward (Fig. 3). Consequently, for a given LV

end-diastolic volume (LVEDV), LVEDP is increased and

may result in pulmonary congestion.

Systolic dysfunction also affects the pressure/volume

loop: the end-systolic pressure/volume slope is shifted

Fig 2 (A) Effect of afterload. Each panel displays two superimposed heartbeats, control (solid line), and isovolumetric (dotted line). The isovolumetric

heartbeat was experimentally obtained in dogs by occluding the ascending aorta during diastole. Relative load is defined as the ratio of baseline to

isovolumetric systolic LV pressure (LVP) expressed as a percentage. Top, isovolumetric pressure elevation is limited (32 mm Hg) and relative load is

79%. Bottom, isovolumetric pressure elevation is fair (56 mm Hg) and relative load is 68%. Relative load is the main determinant of load dependence

of LVP fall. This dependence is related to the timing of the transition from contraction to relaxation. This transition occurs when 81% to 84% of peak

isovolumetric pressure is reached, or the equivalent timing during early ejection. The transition occurs precisely at the time indicated by a vertical line

in both panels. From Gillebert and colleagues40 with permission. (B) Effect of preload. On the upper part, LV (solid lines) and LA (dotted lines)

pressure tracings in a cardiac surgical patient at baseline and after leg elevation; on the lower part, amplification of the diastolic phase. In baseline

conditions, LV filling is already impaired. With leg elevation, diastolic failure develops with further slowing of the myocardial relaxation and a marked

increase in LVEDP, exceeding the left atrial pressure (LAP) resulting in an impaired filling of the LV during diastole.

Pirracchio et al.

710

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 5: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

downward and to the right, indicating a reduction in con-

tractility, whereas end-systolic and end-diastolic LV

volumes are increased, which may also lead to upstream

congestion. Patients with heart failure can have combined

systolic and diastolic dysfunction. In such cases, modest

increases in LVEDV may result in large increases in

LVEDP. Kawaguchi and colleagues56 recently demon-

strated that, in patients admitted with heart failure but with

preserved LVEF, the diastolic portion of the pressure/

volume loops, although apparently normal at rest, altered

on exertion. Indeed, manoeuvres such as sustained iso-

metric handgrip markedly impaired LV diastolic proper-

ties, increased LVEDP, and could reveal diastolic heart

failure (Fig. 4).

Pressure decline analysis

In diastolic heart failure patients, LV pressure decline

analysis reveals a significant increase in the time constant

of isovolumetric relaxation, t.108 121

A recent multicentre, prospective study, using cardiac

catheterization and echocardiography to assess LV dia-

stolic properties in 47 patients with diastolic heart failure

and 10 normal controls,119 demonstrated that insight into

the respective roles of active relaxation and compliance

could be gained using a detailed analysis of the LV dia-

stolic pressure curve. The following measurements are of

particular interest:

(i) t, the time constant of the isovolumetric relaxation;

(ii) Pmin, LV minimal pressure after the opening of the

mitral valve;

(iii) PPre-A, LV pressure just before atrial contraction;

(iv) LVEDP, LV end-diastolic pressure, just after atrial

contraction.

This study showed that in patients with diastolic heart

failure, in contrast to the control subjects, isovolumetric

relaxation was incomplete at the time of Pmin. Thus, t was

prolonged and Pmin increased, resulting in a positive corre-

lation between t and Pmin. Incomplete relaxation

accounted for 7 (1) mm Hg of the measured increase in

Pmin. In these patients, LV compliance was also signifi-

cantly altered with an increase in LVEDP, despite a

reduced LVEDV.

Assessment of LV diastolic function inclinical practice

There are differences between heart failure with reduced

and preserved ejection fraction with regards to symptoms,

physical examination, echocardiographic and ECG

abnormalities, and X-ray findings (Table 1).

Echocardiography

The combination of several ultrasound modalitieses is

useful in the assessment of diastolic function in patients

with possible diastolic heart failure. These modalities

include: 2-D echo, pulsed-wave Doppler, M-mode colour

Doppler, and tissue Doppler imaging. In the presence of

acute pulmonary oedema, echocardiography is suggestive

of diastolic heart failure, if preserved LV systolic function

is associated with indirect signs of elevated LA pressure.

Fig 3 Pressure/volume loop. The solid line represents a normal heart.

The dotted line represents an isolated LV diastolic dysfunction. In patients

with diastolic heart failure, the main alteration in the P/V loop is a shift

in the end-diastolic pressure/volume relationship (EDPVR), whereas the

end-systolic pressure/volume relationship (ESPVR) remains unaltered.

AVO, aortic valve opening; AVC, aortic valve closure; MVO, mitral valve

opening; MVC, mitral valve closure; LVEDP: left ventricular end-diastolic

pressure.

Fig 4 Decompensation of LV diastolic dysfunction during exertion.

Pressure/volume relationship before (dotted line) and after (dark solid

line) sustained isometric handgrip in a patient with chronic class II

NYHA heart failure admitted for rapid-onset pulmonary oedema and

LVEF .50%. Handgrip was used to assess LV diastolic properties in

response to increase systolic blood pressure and LV afterload. In a patient

with normal LV diastolic function, relaxation and filling are unaltered by

increased LV afterload. Handgrip-induced upward shift of the PV loop

induced a parallel increase in the filling curve strongly suggesting that the

LV wall is stiff without relaxation reserve. From Kawaguchi and

colleagues,56 with permission.

Diastolic heart failure in anaesthesia and critical care

711

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 6: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

LV systolic function can be assessed by 2-D echo and

global performance estimated qualitatively (‘eye-balling’)

and, if possible, quantitatively (measurement of ejection

fraction). These measurements should be performed as

soon as possible after the onset of symptoms, as a low

ejection fraction can return to normal within 24–48 h after

treatment of decompensated heart failure. Ejection fraction

values ranging between 0.4 and 0.5 are not strictly

‘normal’, but cannot explain per se the occurrence of

acute pulmonary oedema. Therefore, the criterion gener-

ally required to define ‘preserved LVEF’ is a value .0.5.

However, Petrie and colleagues82 showed that ‘heart

failure with preserved LVEF’ could be associated with

subtle LV systolic dysfunction, yet recognizable using new

measures of LV systolic function (measurement of LV sys-

tolic atrio-ventricular plane displacement). Another study,9

however, concluded that even if ‘subtle abnormalities in

regional systolic function’ might exist in diastolic heart

failure, they are ‘unlikely to be responsible for clinical

signs of heart failure’.

LA pressure cannot be directly measured using echocar-

diography. Pulsed-wave Doppler measures the velocity of

blood at a precise location: the Doppler ‘window’. This

velocity is proportional to the pressure gradient. Thus, if

the Doppler window is placed at the tip of the mitral

valve, the diastolic flow velocity profile will reflect the

pressure gradient between the LA and the LV.4 The mitral

blood flow is composed of an E (early) wave for passive

diastolic filling followed by an A (auricular) wave for

atrial systole. Mitral blood flow profile is affected by LV

relaxation, LV compliance, and LA pressure. Normal dia-

stole is characterized by a predominant E wave (peak and

area under the curve), implying that most of the LV filling

is occurring during the early phase of diastole. Mitral

blood flow abnormalities are of three types:

(i) in mild diastolic dysfunction, only relaxation is

impaired and atrial contraction contributes relatively

more to ventricular filling; thus, peak of A wave

.peak of E wave, with prolonged E wave decelera-

tion time (usually .240 ms);

(ii) in moderate diastolic dysfunction, relaxation is

impaired, LV compliance is decreased, and atrial

pressure increased; a pseudo-normal pattern with a

predominant E wave is observed, but the E wave

deceleration time is shortened;

(iii) in severe diastolic dysfunction, LV compliance is

extremely low; a restrictive pattern is observed with a

high peak E wave velocity, usually more than twice

the peak A wave velocity.

However, Zile and colleagues reported a lack of sensitivity

of the mitral blood flow analysis for the diagnosis of dia-

stolic heart failure. In their study,121 the E/A ratio was

abnormal in only 48% of the patients presenting signs of

heart failure with a normal ejection fraction. The E wave

deceleration time was found to be more sensitive (abnor-

mal in 64% of the patients).

If the Doppler window is located within a pulmonary

vein, the flow velocity will reflect the pressure gradient

between the vein and the LA throughout the cardiac cycle.

A markedly increased LA pressure (.18 mm Hg) will

generate characteristic alterations in the velocity profiles.

Pulmonary vein flow is composed of two waves, one

systolic and one diastolic. The elevation of the LA

pressure impairs atrial filling, and the pulmonary vein

diastolic wave becomes predominant. Nevertheless, pul-

monary vein flow abnormalities cannot discriminate

between systolic and diastolic heart failure.

Tissue Doppler Imaging directly measures myocardial

velocity and allows wall movements to be directly

analysed.28 Tissue Doppler has been validated for the

evaluation of cardiac function.43 67 101 107 The myocardial

portion commonly studied is above the mitral annulus.

Three waveforms are visualized: peak systolic wave, early

diastolic wave (Ea), and end-diastolic wave related to atrial

contraction. Systolic velocities have been shown to be good

predictors of contraction.46 The Ea wave97 is relatively inde-

pendent of loading conditions and is therefore used to

assess LV relaxation.28 A cut-off of 8 cm s21 for Ea

measurement is now widely accepted as a sign of diastolic

dysfunction. The E (early mitral inflow velocity)/Ea ratio is

considered a useful indicator of LV filling pressures.70 71

Although influenced by LVEF, E/Ea measurement per-

formed on the lateral mitral annulus can reliably be used to

evaluate filling pressures in patients presenting with a pre-

served ejection fraction.90 More recently, the delay from

onset of Ea to onset of E has been shown to correlate

strongly with invasively acquired relaxation indices.89

None of these Doppler indices is 100% sensitive or

specific and a combination of indices is usually required to

ascertain high LA pressure.

Natriuretic peptides

Brain natriuretic peptide (BNP) is recognized as a specific

marker of heart failure in patients presenting with acute

dyspnoea.69 Maisel and colleagues62 measured BNP in

1586 patients presenting with acute dyspnoea. Of the 452

patients with a final diagnosis of heart failure, 165

Table 1 Characteristics of heart failure with preserved or reduced LVEF

Altered ejection fraction Preserved ejection fraction

Dyspnoea Chronic Transient mainly

Heart rate Increased Increased

Mitral regurgitation Present Rare

S3/S4 gallop S3.S4 S4 mainly

Rales Present Present

Peripheral oedema Present Rare

Cardiomegaly Constant Inconstant

LV dilatation Nearly constant Absent

ECG abnormal Constant Inconstant

BNP Markedly increased Often mildly increased

Pirracchio et al.

712

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 7: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

(36.5%) had preserved LV function on echocardiography,

whereas 287 (63.5%) had systolic dysfunction. Patients

with non-systolic heart failure had significantly lower BNP

concentrations than those with systolic heart failure (413

vs 821 pg ml21, P,0.001). When comparing patients with

acute diastolic dysfunction with those with non-

cardiogenic dyspnoea, a BNP concentration �100 pg ml2 1

had a sensitivity of 86%, a negative predictive value of

96%, and an accuracy of 75% for detecting abnormal dia-

stolic dysfunction.63 Mildly elevated values of BNP may

not differentiate between systolic and diastolic heart

failure and BNP may be normal in some cases of acute

hypertensive pulmonary oedema in patients with preserved

LV systolic function.60

Diastolic heart failure in clinical practice

Many factors including uncontrolled hypertension, atrial

fibrillation, myocardial ischaemia, anaemia, renal insuffi-

ciency, and non-compliance with treatment may precipitate

overt systolic and diastolic heart failure.106 However,

uncontrolled hypertension is involved in more than 50%

of the cases of acute (decompensated) diastolic heart

failure.36 118 Anaesthetists may have to deal with acute

decompensated diastolic heart failure during the periopera-

tive period, in the ICU or in the emergency department.

Perioperative setting

The perioperative period carries a risk of decompensation

of chronic diastolic heart failure or induction of acute dia-

stolic dysfunction. Therefore, it is important to identify

high-risk patients and situations and drugs likely to

adversely affect LV diastolic function and be able to

prevent and treat acute decompensations.

Preoperative screening should focus on the detection of:

(i) history of diastolic heart failure or structural factors

potentially associated with an impaired LV diastolic

function: LV hypertrophy (except in young athletes)

and atrial arrhythmia affecting LV ‘active filling’;

(ii) factors carrying a higher risk of diastolic heart

failure: female, age more than 70 yr old, history of

untreated hypertension, ischaemic heart disease, or

diabetes mellitus;

(iii) clinical signs of heart failure, especially dyspnoea on

exertion;

(iv) specific measures from echocardiography: LV hyper-

trophy, impairment of diastolic function, preserved

LVEF.

Different guidelines have been published in order to

clarify the definition and the diagnosis of diastolic heart

failure.52 87 111 Vasan and Levy111 produced pragmatic cri-

teria that are widely used in the cardiology. They separated

the diagnostic procedure into three sequential steps: (1)

diagnosis of heart failure, (2) preserved systolic LV

function (LVEF .0.5), and (3) documentation of LV dia-

stolic dysfunction if feasible (Table 2).

Zile and colleagues121 prospectivly compared cardiac

catheterization and echocardiography in patients suspected

of diastolic heart failure (heart failure symptoms and LVEF

.0.5) and concluded that ‘objective measurement of LV

diastolic function is useful to confirm rather than establish

the diagnosis of diastolic heart failure. The diagnosis of

diastolic heart failure can be made without measurement of

parameters that reflect LV diastolic function’.121

On the basis of this, and using the criteria proposed by

Vasan and Levy,111 a specific algorithm can be proposed

for the preoperative risk stratification of LV diastolic func-

tion impairment (Fig. 5). Particular attention should be

paid in patients with potential LV diastolic dysfunction to

avoid a further deterioration of diastolic function,

especially hypovolaemia, tachycardia, and rhythms other

than sinus. For elective surgery, patients with definite dia-

stolic heart failure group would benefit from a cardiologist

opinion and their treatment checked to optimize diastolic

function before surgery.

Perioperative period

During the perioperative period, haemodynamic changes

and anaesthetic agents can adversely affect LV diastolic

function.

Table 2 Definition of definite, probable, and possible DHF.111 SAP, systolic

arterial pressure; DAP, diastolic arterial pressure; BP, blood pressure

Diagnosis Criteria

Clinical Echocardiography

Definite Clinical evidence of heart

failure

Preserved LVEF within 72 h

of the heart failure

eventsþdocumented diastolic

dysfunction

Probable Clinical evidence of heart

failure

Preserved LVEF within 72 h

of the heart failure events but

no documentation of diastolic

dysfunction

Possible Clinical evidence of heart

failure

Preserved LVEF but not at the

time of the heart failure events

without documentation of

diastolic dysfunction

Upgrade from

possible to

probable diastolic

heart failure

SAP .160 mm Hg or

DAP .100 mm Hg during

the episode of heart failure,

which may include the

following:

Concentric LV hypertrophy

without wall-motion

abnormalities

tachyarrhythmia

precipitation of event by

the infusion of a small

amount of i.v. fluid

clinical improvement in

response to therapy

directed at the cause of

diastolic dysfunction

(such as lowering BP,

reducing heart rate, or

restoring the atrial booster

mechanism)

Diastolic heart failure in anaesthesia and critical care

713

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 8: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Haemodynamic changes affecting diastolic time, such

as arrhythmia and myocardial ischaemia, are likely to

decompensate further pre-existing diastolic dysfunction.

Tachycardia shortens diastole and is likely to impair LV

filling. Rhythm disturbances can be precipitated by hypo-

or hyper-kalaemia, anaemia, or hypovolaemia. Treatment

with beta-blockers or non-dihydropyridine calcium-

channels blockers has been proposed to prevent tachycar-

dia and improve LV filling.6 94

Myocardial ischaemia or acute increases in cardiac

loading (volume loading or changes in position) (Fig. 2B)

may result in a significant slowing of myocardial relax-

ation. Myocardial ischaemia may also induce rhythm

disturbances that will further aggravate LV diastolic dys-

function. Thus, prevention of ischaemic episodes should

remain a major objective for anaesthetists dealing with

suspected diastolic heart failure. Beta-blockers still remain

the best drug to achieve a safe reduction in myocardial

oxygen consumption. Indeed, perioperative use of beta-

blockers has been shown to reduce overall mortality due to

cardiac events.64 113 Whether this strategy is still appli-

cable to diastolic heart failure remains to be determined.

Finally, the use of regional or general anaesthesia is still

debated, but no study has found benefit of one technique

over the other.

The effect of anaesthetic agents on LV diastolic proper-

ties has been extensively studied for volatile agents, but

less for i.v. agents (Table 3). The volatile agents, sevoflur-

ane and desflurane, as well as opioids and muscle relax-

ants do not appear to affect LV diastolic properties.

In high-risk diastolic heart failure patients, anaesthetists

should pay particular attention to the choice of monitor-

ing and to avoiding acute perioperative changes in load

conditions, heart rate, and myocardial oxygen balance.

Fig 5 Algorithm for preoperative risk stratification of patients with suspected diastolic heart failure.

Pirracchio et al.

714

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 9: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

There are no good clinical data on the action of anaes-

thetic drugs in LV diastolic dysfunction.

Recovery room, ICU, and emergency room

Hypertensive crisis

Gandhi and colleagues36 compared the echocardiographic

findings on admission and after 2–3 days of treatment in

patients presenting with acute pulmonary oedema as a con-

sequence of severe arterial hypertension. Although transi-

ent LV systolic dysfunction due to the hypertensive crisis

was expected, no difference in LVEF and regional wall

motion was found between the acute episode and after 24

and 72 h of treatment. Around 50% of the patients

admitted with an acute pulmonary oedema had preserved

ejection fraction and 89% of the patients who had a pre-

served ejection fraction after treatment also had no sign of

systolic dysfunction during the acute episode. Similarly, in

patients with an impaired ejection fraction, no differences

were found within the first 72 h, suggesting that acute dia-

stolic failure might also be the major mechanism of

decompensation in patients with baseline systolic dysfunc-

tion. Acute diastolic dysfunction during hypertensive crisis

remains poorly understood. In diastolic heart failure at

rest, it is understandable that a small increase in LVEDV

will lead to a marked elevation in LVEDP. However, this

is less clear in patients without diastolic dysfunction at

rest. It has been proposed that a hypertensive crisis leads

to a marked increase in coronary perfusion pressure and

thus in coronary turgor.114 Nevertheless, it is unlikely that

an increase in coronary blood volume can cause a signifi-

cant increase in wall thickness.

Myocardial ischaemia

Myocardial ischaemia is one of the main mechanisms of

LV diastolic dysfunction in the early postoperative period,

and several factors, including pain-induced sympathetic

activation (tachycardia, hypertension), shivering, anaemia,

hypovolaemia, and hypoxia, may alter myocardial oxygen

balance.

In a rabbit model,80 the early and late haemodynamic con-

sequences of a circumflex artery ligation were analysed by

echocardiography and Doppler. One hour after experimental

infarct, the rabbits exhibited a significant alteration of the

LV filling pattern; decrease in E and A waves, A wave rever-

sal velocities and increase in the mean pulmonary venous

systolic-to-diastolic ratio. Three weeks after coronary lig-

ation, the rabbits still exhibited significant abnormalities in

filling pattern. Stugaard and colleagues100 assessed LV dia-

stolic function in 20 patients during coronary angioplasty

and in eight anaesthetized dogs during experimental coron-

ary occlusion. Diastolic function was explored using

M-mode Doppler, which determines the time difference

between the peak velocity in the apical region and in the

mitral tip. The authors reported a significant increase in time

difference in both patients and dogs, and the time difference

evolution correlated significantly with the variation in the

time constant of isovolumetric relaxation. Pacing tachycar-

dia, volume loading, and vena cava restriction did not sig-

nificantly alter the time difference.

Nitric oxide (NO) metabolism seems to play a controver-

sial role in acute diastolic dysfunction following episodes

of ischaemia–reperfusion. A beneficial role for NO has

been suggested since pre-treatment with cGMP donors or

with NO donors protects myocytes from relaxation failure

in experimental models of hypoxia-reoxygenation.30 92

However, excessive NO production during reperfusion

appears to alter diastolic function due to an excess of

peroxynitrite formation.115

Sepsis

Increasing evidence suggests that both systolic and dia-

stolic functions are affected in severe sepsis and septic

shock.85 We recently showed, using pressure/volume

tracings in anaesthetized endotoxaemic rabbits, that LV

diastolic properties are altered; prolonged relaxation,

decreased LV compliance leading to increased end-

diastolic pressure.

In a transmitral Doppler analysis of 13 patients in septic

shock,54 10 in sepsis without shock, and 33 controls

patients with septic shock and sepsis without shock had a

Table 3 Effects of volatile and i.v. anaesthetic agents on diastolic function.

Experimental models were either animal models29 44 51 65 68 72 75 76 96 109 116

or cellular models. 45 47 – 49 59 88 112 Human models were either healthy33 74 or

patients with underlying cardiac impairment10 20 23 26 50

LV relaxation LV compliance

Enflurane Decrease116

Experimental

model

Impairment51 116

Human No data

Halothane

Experimental

model

Impairment29 47 48 51 75 112 No change,44 109

decrease68 75 116

Human No change,33 impairment50

Isoflurane

Experimental

model

No change,48 49 112

impairment51 75No change75 116

Human No change,74 impairment50

Sevoflurane

Experimental

model

No change48 49 No change116

Human No change,10 20 23 33

impairment26

Nitrous oxide

Experimental

model

Impairment65

Decrease65

Propofol

Experimental

model

No significant change,33 59 72 96

impairment88 Decrease76

Human Impairment,10 23 20 no change37

Ketamine

Experimental

model

Impairment76 Decrease76

Midazolam

Human No change37

Morphine

Experimental

model

No change68

Diastolic heart failure in anaesthesia and critical care

715

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 10: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

significantly altered LV filling pattern in comparison with

controls. More recently, in a study of systolic and diastolic

function using transoesophageal echocardiography and

pulmonary artery catheters in 25 consecutive patients in

septic shock,83 8 of the 25 patients had no regional wall

motion abnormality and a normal LV filling pattern (trans-

mitral E/A waves ratio .1; pulmonary veins systolic/dia-

stolic waves ratio .1); 11 had evidence of abnormal left

auricular filling (systolic/diastolic waves ratio ,1) but

with a preserved systolic function and E/A waves ratio.

According to the investigators, transmitral flow in this

group could be considered as ‘pseudo-normalized’.

Finally, 6 of the 25 patients exhibited both systolic and

diastolic dysfunctions. The authors concluded ‘cardiac

effects of septic shock can be expressed in various

degrees, ranging from a normal pattern, through diastolic

dysfunction up to both poor LV systolic and diastolic func-

tions resulting in combined cardiogenic-septic shock’.

Mechanisms of sepsis-induced systolic and diastolic dys-

functions are complex and described elsewhere.85 Delayed

relaxation and impaired compliance are likely to be related

to nitration of contractile proteins rather than alterations in

calcium homeostasis. We and others showed that calcium

influx is unaltered in papillary muscle and cardiac myo-

cytes from endotoxaemic or septic animals.102 – 104

Increased free radical production, especially peroxynitrite

overproduction, seems to play a major role in the nitration

and therefore the deterioration of protein function in septic

patients.57 86 In the myocardium of patients who died of

septic shock, contractile proteins such as myosine appear

to be specifically nitrated by peroxynitrite.86

The effects of the three most commonly used inotropes

(dobutamine, enoximone, and levosimendan) were recently

studied in normal and endotoxaemic rabbits (personal

data). Together with the improvement in the indexes of

LV systolic function, the inotropes generally improved

relaxation or LV compliance in normal rabbits. In endo-

toxaemic rabbits, however, only levosimendan improved

both relaxation and LV compliance. The effects of two

vasopressors, norepinephrine and vasopressin, were also

studied in endotoxaemic rabbits,32 and for a similar

increase in systolic blood pressure (15%), norepinephrine

induced no change in the index of LV systolic function

dP/dtmax nor in cardiac output whereas vasopressin

induced marked deterioration in these measurements. The

effect of inotropes and vasopressors on LV diastolic func-

tion in septic patients requires further investigation.

Management of diastolic heart failure

Initial management of acute decompensated

diastolic heart failure

The management of acute decompensated diastolic heart

failure is based on a reduction in pulmonary congestion

and a correction of the precipitating factors, such as a

hypertensive crisis, myocardial ischaemia, acute rhythm

disturbances, and sepsis. Specific treatment of precipitating

factors should always be considered: vasodilators for

hypertensive crisis, coronary revascularization, restoration

of sinus rhythm, and haemodynamic optimization in septic

shock.

A hypertensive crisis can be managed by i.v. calcium-

antagonists such as nicardipine or nitrendipine (sublingual

nifedipine is not recommended). High-dose nitrates i.v.

can decrease both preload and afterload. Sodium nitroprus-

side can also produce balanced vasodilatation, but may

result in severe unloading or hypotension in these non-

dilated ventricles. Angiotensin converting enzyme inhibi-

tors are not useful in the acute phase because of their

slower onset of action. Beta-adrenergic blockers or diltia-

zem may be used in acute heart failure related to rapid

atrial fibrillation or severe myocardial ischaemia.

Pulmonary congestion can be reduced by controlling

blood volume or improving LV filling. Because of the

steepness of the LV diastolic pressure/volume relation-

ship, a small decrease in LVEDV can lead to a marked

decrease in LVEDP. The reduction in the blood volume

can be achieved either by nitrates or by diuretics, in

order to reduce venous return and decrease LVEDV.

Nevertheless, diuretics should be carefully considered in

the context of acute hypertensive crisis, as blood volume

is often already decreased by chronic hypertension or the

long-term use of diuretics. As contractile function is pre-

served, the role of sympathomimetic inotropes is limited.

Digoxin is useful only to slow the heart rate in rapid

atrial fibrillation.

Continuous positive airway pressure seems to be effec-

tive in the treatment of diastolic dysfunction.11

Future treatments for acute diastolic heart failure

Future approaches will probably focus on the optimization

of LV relative load at the cardiac level and cardiac myo-

cytes calcium homeostasis at the cellular level.

As mentioned earlier, drugs that may decrease LV rela-

tive load are of interest. The decrease in the relative load

can be obtained either by decreasing cardiac load or by

improving systolic function, or by the combination of

both. Thus, levosimendan may be a candidate for acute

diastolic heart failure treatment as it combines a vasodila-

tor effect, by opening ATP-sensitive Kþ channels, and a

positive inotropic effect, by modulating the interaction

between troponin and calcium.105 Despite a ‘calcium sen-

sitizer’ effect that is expected to worsen diastolic proper-

ties of the heart, levosimendan has been recently shown to

improve LV diastolic properties.77 99

The effect of NO donors on LV diastolic function has

been studied in animals and humans.78 They have been

shown to induce an early relaxation and a decrease in basal

tone both related to a reduction in cardiac myofilament

Pirracchio et al.

716

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 11: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

responsiveness to Ca2þ, within seconds of administration

in vitro. Similarly, intracoronary injections of the NO

donor, sodium nitroprusside, in normal hearts caused an

earlier onset of LV relaxation, a decline in LV minimum

and end-diastolic pressures, an increase in LVEDV, and a

down and rightward shift of the LV diastolic pressure/

volume relationship, within minutes of administration.79

These results are consistent with a direct NO-induced

improvement in diastolic function. A direct beneficial

effect of NO donors on diastolic dysfunction has not been

shown in patients in the acute/early phase of acute dia-

stolic heart failure. Of note, two other vasodilators, urapi-

dil and nicardipine, seem to have no effect on relative load

or directly on the diastolic properties of the LV.25 In

addition, another endogenous cardiovascular mediator,

endothelin, has no effects on diastolic properties in normal

or chronic heart failure patients.61

Long-term management of chronic LV

diastolic dysfunction

Myocardial remodelling over months or years is essential

to restore adequate LV filling conditions. Several

approaches have been proposed to control LV structural

abnormalities. Because the renin–angiotensin–aldosterone

system plays an important role in the development of dia-

stolic heart failure and particularly in myocardial remodel-

ling and fluid retention, angiotensin-converting enzyme

inhibitors, angiotensin receptors antagonists, and aldoster-

one antagonists have been proposed in the treatment of

diastolic heart failure.19 Spironolactone has recently been

shown to limit the evolution of cardiac muscle fibrosis.53 91

A study of enalapril on diastolic heart failure in elderly

patients with prior myocardial infarction7 reported a benefit

in terms of exercise capacity. The CHARM-preserved study

is a multicentre, randomized, double-blinded study compar-

ing in diastolic heart failure the effects of a selective

angiotensin-receptors blocker (candesartan) and placebo.117

This showed a significant reduction in hospitalization rate

after 36 months follow-up in the candesartan group.

Losartan has been shown to improve echocardiography and

exercise tolerance.5

Of note, cardiac resynchronization therapy affects LV

loading conditions and has recently been shown to

improve LV filling.1

Conclusion

Although frequently underestimated, diastolic heart failure

is a common pathology. Diastolic heart failure is recog-

nized as the mechanism involved in heart failure with a

preserved LV function, and LV diastolic dysfunction can

also account for acute heart failure occurring in critical

care situations. Hypertensive crisis, sepsis, and myocardial

ischaemia are frequently associated with acute diastolic

heart failure. Symptomatic treatment focuses on the

reduction in pulmonary congestion and the improvement

in LV filling. Specific treatment is actually lacking, but

encouraging data are emerging concerning the use of

renin–angiotensin–aldosterone axis blockers, NO donors

or very recently new agents specifically targeting actin–

myosin cross-bridges.

AcknowledgementThis work was partially supported by a grant from the Ministere del’Enseignement Superieur et de la Recherche (EA322).

References1 Agacdiken A, Vural A, Ural D, et al. Effect of cardiac resynchro-

nization therapy on left ventricular diastolic filling pattern in

responder and nonresponder patients. Pacing Clin Electrophysiol2005; 28: 654–60

2 Agoston I, Cameron CS, Yao D, Dela Rosa A, Mann DL,Deswal A. Comparison of outcomes of white versus blackpatients hospitalized with heart failure and preserved ejection

fraction. Am J Cardiol 2004; 94: 1003–73 Angeja BG, Grossman W. Evaluation and management of dia-

stolic heart failure. Circulation 2003; 107: 659–634 Appleton CP, Hatle LK, Popp RL. Relation of transmitral flow

velocity patterns to left ventricular diastolic function: newinsights from a combined hemodynamic and Doppler echocar-diographic study. J Am Coll Cardiol 1988; 12: 426–40

5 Araujo AQ, Arteaga E, Ianni BM, Buck PC, Rabello R, Mady C.Effect of Losartan on left ventricular diastolic function in

patients with nonobstructive hypertrophic cardiomyopathy. Am JCardiol 2005; 96: 1563–7

6 Aronow WS, Ahn C, Kronzon I. Effect of propranolol versus nopropranolol on total mortality plus nonfatal myocardial infarc-tion in older patients with prior myocardial infarction, conges-

tive heart failure, and left ventricular ejection fraction . or¼40% treated with diuretics plus angiotensin-converting enzymeinhibitors. Am J Cardiol 1997; 80: 207–9

7 Aronow WS, Kronzon I. Effect of enalapril on congestive heartfailure treated with diuretics in elderly patients with prior myo-

cardial infarction and normal left ventricular ejection fraction.Am J Cardiol 1993; 71: 602–4

8 Aurigemma GP, Gaasch WH. Clinical practice. Diastolic heartfailure. N Engl J Med 2004; 351: 1097–105

9 Aurigemma GP, Zile MR, Gaasch WH. Contractile behavior ofthe left ventricle in diastolic heart failure: with emphasis onregional systolic function. Circulation 2006; 113: 296–304

10 Bein B, Renner J, Caliebe D, et al. Sevoflurane but not propofolpreserves myocardial function during minimally invasive direct

coronary artery bypass surgery. Anesth Analg 2005; 100: 610–6,table of contents

11 Bendjelid K, Schutz N, Suter PM, et al. Does continuous positiveairway pressure by face mask improve patients with acute car-diogenic pulmonary edema due to left ventricular diastolic dys-

function? Chest 2005; 127: 1053–812 Bers DM, Despa S. Cardiac myocytes Ca2þ and Naþ regulation

in normal and failing hearts. J Pharmacol Sci 2006; 100: 315–22

Diastolic heart failure in anaesthesia and critical care

717

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 12: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

13 Betocchi S, Piscione F, Villari B, et al. Effects of inducedasynchrony on left ventricular diastolic function in patients withcoronary artery disease. J Am Coll Cardiol 1993; 21: 1124–31

14 Bhatia RS, Tu JV, Lee DS, et al. Outcome of heart failure with

preserved ejection fraction in a population-based study. N Engl JMed 2006; 355: 260–9

15 Blaustein AS, Gaasch WH. Myocardial relaxation. VI. Effects ofbeta-adrenergic tone and asynchrony on LV relaxation rate. Am J

Physiol 1983; 244: H417–2216 Brutsaert DL, Sys SU. Relaxation and diastole of the heart.

Physiol Rev 1989; 69: 1228–31517 Chemla D, Coirault C, Hebert JL, Lecarpentier Y. Mechanics

of relaxation of the human heart. News Physiol Sci 2000; 15:

78–8318 Chen HH, Lainchbury JG, Senni M, Bailey KR, Redfield MM.

Diastolic heart failure in the community: clinical profile, naturalhistory, therapy, and impact of proposed diagnostic criteria.J Card Fail 2002; 8: 279–87

19 Chinnaiyan KM, Alexander D, McCullough PA. Role of angioten-sin II in the evolution of diastolic heart failure. J Clin Hypertens(Greenwich) 2005; 7: 740–7

20 De Hert SG, Cromheecke S, ten Broecke PW, et al. Effects ofpropofol, desflurane, and sevoflurane on recovery of myocardial

function after coronary surgery in elderly high-risk patients.Anesthesiology 2003; 99: 314–23

21 De Hert SG, Gillebert TC, Ten Broecke PW, Mertens E,Rodrigus IE, Moulijn AC. Contraction–relaxation coupling and

impaired left ventricular performance in coronary surgerypatients. Anesthesiology 1999; 90: 748–57

22 De Hert SG, Gillebert TC, Ten Broecke PW, Moulijn AC.Length-dependent regulation of left ventricular function in cor-onary surgery patients. Anesthesiology 1999; 91: 379–87

23 De Hert SG, ten Broecke PW, Mertens E, et al. Sevoflurane butnot propofol preserves myocardial function in coronary surgerypatients. Anesthesiology 2002; 97: 42–9

24 De Hert SG, ten Broecke PW, Rodrigus IE, Mertens E,Stockman BA, Vermeyen KM. The effects of the pericardium on

length-dependent regulation of left ventricular function in cor-onary artery surgery patients. J Cardiothorac Vasc Anesth 2001;15: 300–5

25 De Hert SG, Van der Linden PJ, Ten Broecke PW, Sermeus LA,Gillebert TC. Effects of nicardipine and urapidil on

length-dependent regulation of myocardial function in coronaryartery surgery patients. J Cardiothorac Vasc Anesth 1999; 13:677–83

26 De Hert SG, Van der Linden PJ, ten Broecke PW, Vermeylen KT,

Rodrigus IE, Stockman BA. Effects of desflurane and sevofluraneon length-dependent regulation of myocardial function in coron-ary surgery patients. Anesthesiology 2001; 95: 357–63

27 De Hert SG, Vander Linden PJ, ten Broecke PW, De Mulder PA,Rodrigus IE, Adriaensen HF. Assessment of length-dependent

regulation of myocardial function in coronary surgery patientsusing transmitral flow velocity patterns. Anesthesiology 2000; 93:374–81

28 Dokainish H. Tissue Doppler imaging in the evaluation of leftventricular diastolic function. Curr Opin Cardiol 2004; 19:

437–4129 Doyle RL, Foex P, Ryder WA, Jones LA. Effects of halothane on

left ventricular relaxation and early diastolic coronary bloodflow in the dog. Anesthesiology 1989; 70: 660–6

30 Draper NJ, Shah AM. Beneficial effects of a nitric oxide donor

on recovery of contractile function following brief hypoxia inisolated rat heart. J Mol Cell Cardiol 1997; 29: 1195–205

31 Eichhorn EJ, Willard JE, Alvarez L, et al. Are contraction andrelaxation coupled in patients with and without congestiveheart failure? Circulation 1992; 85: 2132–9

32 Faivre V, Kaskos H, Callebert J, et al. Cardiac and renal effects

of levosimendan, arginine vasopressin, and norepinephrine inlipopolysaccharide-treated rabbits. Anesthesiology 2005; 103:514–21

33 Filipovic M, Wang J, Michaux I, Hunziker P, Skarvan K,

Seeberger MD. Effects of halothane, sevoflurane and propofolon left ventricular diastolic function in humans during spon-taneous and mechanical ventilation. Br J Anaesth 2005; 94:186–92

34 Gaasch WH, Blaustein AS, Andrias CW, Donahue RP, Avitall B.

Myocardial relaxation. II. Hemodynamic determinants of rate ofleft ventricular isovolumic pressure decline. Am J Physiol 1980;239: H1–6

35 Gaasch WH, Carroll JD, Blaustein AS, Bing OH. Myocardialrelaxation: effects of preload on the time course of isovolu-

metric relaxation. Circulation 1986; 73: 1037–4136 Gandhi SK, Powers JC, Nomeir AM, et al. The pathogenesis of

acute pulmonary edema associated with hypertension. N Engl JMed 2001; 344: 17–22

37 Gare M, Parail A, Milosavljevic D, Kersten JR, Warltier DC,

Pagel PS. Conscious sedation with midazolam or propofol doesnot alter left ventricular diastolic performance in patients withpreexisting diastolic dysfunction: a transmitral and tissueDoppler transthoracic echocardiography study. Anesth Analg

2001; 93: 865–7138 Gillebert TC, Leite-Moreira AF, De Hert SG. The hemodynamic

manifestation of normal myocardial relaxation. A framework forexperimental and clinical evaluation. Acta Cardiol 1997; 52:223–46

39 Gillebert TC, Leite-Moreira AF, De Hert SG. Load dependentdiastolic dysfunction in heart failure. Heart Fail Rev 2000; 5:345–55

40 Gillebert TC, Leite-Moreira AF, De Hert SG. Relaxation-systolicpressure relation. A load-independent assessment of left ventri-

cular contractility. Circulation 1997; 95: 745–5241 Gillebert TC, Raes DF. Preload, length-tension relation, and iso-

metric relaxation in cardiac muscle. Am J Physiol 1994; 267:H1872–9

42 Gillebert TC, Sys SU, Brutsaert DL. Influence of loading pat-

terns on peak length–tension relation and on relaxation incardiac muscle. J Am Coll Cardiol 1989; 13: 483–90

43 Gorcsan J 3rd, Gulati VK, Mandarino WA, Katz WE.Color-coded measures of myocardial velocity throughout the

cardiac cycle by tissue Doppler imaging to quantify regional leftventricular function. Am Heart J 1996; 131: 1203–13

44 Greene ES, Gerson JI. One versus two MAC halothane anesthe-sia does not alter the left ventricular diastolic pressure–volumerelationship. Anesthesiology 1986; 64: 230–7

45 Gueugniaud PY, Hanouz JL, Vivien B, Lecarpentier Y, Coriat P,Riou B. Effects of desflurane in rat myocardium: comparisonwith isoflurane and halothane. Anesthesiology 1997; 87:599–609

46 Gulati VK, Katz WE, Follansbee WP, Gorcsan J, 3rd. Mitral

annular descent velocity by tissue Doppler echocardiography asan index of global left ventricular function. Am J Cardiol 1996;77: 979–84

47 Hanouz JL, Riou B, Massias L, Lecarpentier Y, Coriat P.Interaction of halothane with alpha- and beta-adrenoceptor

stimulations in rat myocardium. Anesthesiology 1997; 86:147–59

Pirracchio et al.

718

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 13: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

48 Hanouz JL, Vivien B, Gueugniaud PY, Lecarpentier Y, Coriat P,Riou B. Comparison of the effects of sevoflurane, isofluraneand halothane on rat myocardium. Br J Anaesth 1998; 80:621–7

49 Hanouz JL, Vivien B, Gueugniaud PY, Lecarpentier Y, Coriat P,Riou B. Interaction of isoflurane and sevoflurane with alpha- andbeta-adrenoceptor stimulations in rat myocardium. Anesthesiology1998; 88: 1249–58

50 Houltz E, Caidahl K, Adin C, Gustavsson T, Ricksten SE. Effectsof halothane and isoflurane on left ventricular diastolic functionduring surgical stress in patients with coronary artery disease.Acta Anaesthesiol Scand 1997; 41: 931–8

51 Humphrey LS, Stinson DC, Humphrey MJ, et al. Volatile anes-

thetic effects on left ventricular relaxation in swine.Anesthesiology 1990; 73: 731–8

52 Hunt SA, Baker DW, Chin MH, et al. ACC/AHA guidelines forthe evaluation and management of chronic heart failure in theadult: executive summary. A report of the American College of

Cardiology/American Heart Association Task Force on PracticeGuidelines (Committee to revise the 1995 Guidelines for theEvaluation and Management of Heart Failure). J Am Coll Cardiol2001; 38: 2101–13

53 Izawa H, Murohara T, Nagata K, et al. Mineralocorticoid recep-

tor antagonism ameliorates left ventricular diastolic dysfunctionand myocardial fibrosis in mildly symptomatic patients with idio-pathic dilated cardiomyopathy: a pilot study. Circulation 2005;112: 2940–5

54 Jafri SM, Lavine S, Field BE, Bahorozian MT, Carlson RW. Leftventricular diastolic function in sepsis. Crit Care Med 1990; 18:709–14

55 Judge KW, Pawitan Y, Caldwell J, Gersh BJ, Kennedy JW.Congestive heart failure symptoms in patients with preserved

left ventricular systolic function: analysis of the CASS registry.J Am Coll Cardiol 1991; 18: 377–82

56 Kawaguchi M, Hay I, Fetics B, Kass DA. Combined ventricularsystolic and arterial stiffening in patients with heart failure andpreserved ejection fraction: implications for systolic and dia-

stolic reserve limitations. Circulation 2003; 107: 714–2057 Lanone S, Mebazaa A, Heymes C, et al. Muscular contractile

failure in septic patients: role of the inducible nitric oxidesynthase pathway. Am J Respir Crit Care Med 2000; 162:2308–15

58 Leite-Moreira AF, Gillebert TC. Nonuniform course of left ven-tricular pressure fall and its regulation by load and contractilestate. Circulation 1994; 90: 2481–91

59 Lejay M, Hanouz JL, Lecarpentier Y, Coriat P, Riou B.

Modifications of the inotropic responses to alpha- andbeta-adrenoceptor stimulation by propofol in rat myocardium.Anesth Analg 1998; 87: 277–83

60 Logeart D, Saudubray C, Beyne P, et al. Comparative value ofDoppler echocardiography and B-type natriuretic peptide assay

in the etiologic diagnosis of acute dyspnea. J Am Coll Cardiol2002; 40: 1794–800

61 MacCarthy PA, Grocott-Mason R, Prendergast BD, Shah AM.Contrasting inotropic effects of endogenous endothelin in thenormal and failing human heart: studies with an intracoronary

ET(A) receptor antagonist. Circulation 2000; 101: 142–762 Maisel AS, Krishnaswamy P, Nowak RM, et al. Rapid measure-

ment of B-type natriuretic peptide in the emergency diagnosisof heart failure. N Engl J Med 2002; 347: 161–7

63 Maisel AS, McCord J, Nowak RM, et al. Bedside B-Type natriure-

tic peptide in the emergency diagnosis of heart failure withreduced or preserved ejection fraction. Results from the

Breathing Not Properly Multinational Study. J Am Coll Cardiol2003; 41: 2010–7

64 Mangano DT, Layug EL, Wallace A, Tateo I. Effect of atenolol onmortality and cardiovascular morbidity after noncardiac surgery.

Multicenter Study of Perioperative Ischemia Research Group.N Engl J Med 1996; 335: 1713–20

65 Marsch SC, Dalmas S, Philbin DM, Ryder WA, Wong LS, Foex P.Effects and interactions of nitrous oxide, myocardial ischemia,

and reperfusion on left ventricular diastolic function. AnesthAnalg 1997; 84: 39–45

66 Masoudi FA, Havranek EP, Smith G, et al. Gender, age, and heartfailure with preserved left ventricular systolic function. J Am CollCardiol 2003; 41: 217–23

67 McDicken WN, Sutherland GR, Moran CM, Gordon LN.Colour Doppler velocity imaging of the myocardium. UltrasoundMed Biol 1992; 18: 651–4

68 Moores WY, Weiskopf RB, Baysinger M, Utley JR. Effects ofhalothane and morphine sulfate on myocardial compliance fol-

lowing total cardiopulmonary bypass. J Thorac Cardiovasc Surg1981; 81: 163–70

69 Morrison LK, Harrison A, Krishnaswamy P, Kazanegra R,Clopton P, Maisel A. Utility of a rapid B-natriuretic peptide assayin differentiating congestive heart failure from lung disease in

patients presenting with dyspnea. J Am Coll Cardiol 2002; 39:202–9

70 Nagueh SF, Middleton KJ, Kopelen HA, Zoghbi WA,Quinones MA. Doppler tissue imaging: a noninvasive tech-

nique for evaluation of left ventricular relaxation and esti-mation of filling pressures. J Am Coll Cardiol 1997; 30:1527–33

71 Ommen SR, Nishimura RA, Appleton CP, et al. Clinical utility ofDoppler echocardiography and tissue Doppler imaging in the

estimation of left ventricular filling pressures: a comparative sim-ultaneous Doppler-catheterization study. Circulation 2000; 102:1788–94

72 Ouattara A, Langeron O, Souktani R, Mouren S, Coriat P, Riou B.Myocardial and coronary effects of propofol in rabbits with com-

pensated cardiac hypertrophy. Anesthesiology 2001; 95: 699–70773 Owan TE, Hodge DO, Herges RM, Jacobsen SJ, Roger VL,

Redfield MM. Trends in prevalence and outcome of heart failurewith preserved ejection fraction. N Engl J Med 2006; 355:251–9

74 Oxorn D, Edelist G, Harrington E, Tsang S. Echocardiographicassessment of left ventricular filling during isoflurane anaesthe-sia. Can J Anaesth 1996; 43: 569–74

75 Pagel PS, Kampine JP, Schmeling WT, Warltier DC. Alteration of

left ventricular diastolic function by desflurane, isoflurane,and halothane in the chronically instrumented dog with auto-nomic nervous system blockade. Anesthesiology 1991; 74:1103–14

76 Pagel PS, Schmeling WT, Kampine JP, Warltier DC. Alteration of

canine left ventricular diastolic function by intravenous anes-thetics in vivo. Ketamine and propofol. Anesthesiology 1992; 76:419–25

77 Parissis JT, Panou F, Farmakis D, et al. Effects of levosimendan onmarkers of left ventricular diastolic function and neurohormonal

activation in patients with advanced heart failure. Am J Cardiol2005; 96: 423–6

78 Paulus WJ, Shah AM. NO and cardiac diastolic function.Cardiovasc Res 1999; 43: 595–606

79 Paulus WJ, Vantrimpont PJ, Shah AM. Paracrine coronary endo-

thelial control of left ventricular function in humans. Circulation1995; 92: 2119–26

Diastolic heart failure in anaesthesia and critical care

719

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 14: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

80 Pennock GD, Yun DD, Agarwal PG, Spooner PH, Goldman S.Echocardiographic changes after myocardial infarction in amodel of left ventricular diastolic dysfunction. Am J Physiol 1997;273: H2018–29

81 Pernenkil R, Vinson JM, Shah AS, Beckham V, Wittenberg C,Rich MW. Course and prognosis in patients . or¼ 70 yearsof age with congestive heart failure and normal versusabnormal left ventricular ejection fraction. Am J Cardiol 1997;

79: 216–982 Petrie MC, Caruana L, Berry C, McMurray JJ. ‘Diastolic heart

failure’ or heart failure caused by subtle left ventricular systolicdysfunction? Heart 2002; 87: 29–31

83 Poelaert J, Declerck C, Vogelaers D, Colardyn F, Visser CA. Left

ventricular systolic and diastolic function in septic shock.Intensive Care Med 1997; 23: 553–60

84 Priebe HJ. The aged cardiovascular risk patient. Br J Anaesth2000; 85: 763–78

85 Rabuel C, Mebazaa A. Septic shock: a heart story since the

1960s. Intensive Care Med 2006; 32: 799–80786 Rabuel C, Renaud E, Brealey D, et al. Human septic myopathy:

induction of cyclooxygenase, heme oxygenase and activation ofthe ubiquitin proteolytic pathway. Anesthesiology 2004; 101:583–90

87 Remme WJ, Swedberg K. Guidelines for the diagnosis andtreatment of chronic heart failure. Eur Heart J 2001; 22:1527–60

88 Riou B, Besse S, Lecarpentier Y, Viars P. In vitro effects of pro-

pofol on rat myocardium. Anesthesiology 1992; 76: 609–1689 Rivas-Gotz C, Khoury DS, Manolios M, Rao L, Kopelen HA,

Nagueh SF. Time interval between onset of mitral inflow andonset of early diastolic velocity by tissue Doppler: a novel indexof left ventricular relaxation: experimental studies and clinical

application. J Am Coll Cardiol 2003; 42: 1463–7090 Rivas-Gotz C, Manolios M, Thohan V, Nagueh SF. Impact of left

ventricular ejection fraction on estimation of left ventricularfilling pressures using tissue Doppler and flow propagation vel-ocity. Am J Cardiol 2003; 91: 780–4

91 Roongsritong C, Sutthiwan P, Bradley J, Simoni J, Power S,Meyerrose GE. Spironolactone improves diastolic function inthe elderly. Clin Cardiol 2005; 28: 484–7

92 Schluter KD, Weber M, Schraven E, Piper HM. NO donorSIN-1 protects against reoxygenation-induced cardiomyocyte

injury by a dual action. Am J Physiol 1994; 267: H1461–693 Setaro JF, Soufer R, Remetz MS, Perlmutter RA, Zaret BL.

Long-term outcome in patients with congestive heart failureand intact systolic left ventricular performance. Am J Cardiol

1992; 69: 1212–694 Setaro JF, Zaret BL, Schulman DS, Black HR, Soufer R.

Usefulness of verapamil for congestive heart failure associatedwith abnormal left ventricular diastolic filling and normalleft ventricular systolic performance. Am J Cardiol 1990; 66:

981–695 Smith GL, Masoudi FA, Vaccarino V, Radford MJ, Krumholz HM.

Outcomes in heart failure patients with preserved ejection frac-tion: mortality, readmission, and functional decline. J Am CollCardiol 2003; 41: 1510–8

96 Sohma A, Foex P, Ryder WA. Regional distensibility, chamberstiffness, and elastic stiffness constant in halothane and propofolanesthesia. J Cardiothorac Vasc Anesth 1993; 7: 188–94

97 Sohn DW, Chai IH, Lee DJ, et al. Assessment of mitral annulusvelocity by Doppler tissue imaging in the evaluation of left

ventricular diastolic function. J Am Coll Cardiol 1997; 30:474–80

98 Solomon SB, Nikolic SD, Frater RW, Yellin EL. Contraction–relaxation coupling: determination of the onset of diastole. Am JPhysiol 1999; 277: H23–7

99 Sonntag S, Sundberg S, Lehtonen LA, Kleber FX. The calcium

sensitizer levosimendan improves the function of stunnedmyocardium after percutaneous transluminal coronary angio-plasty in acute myocardial ischemia. J Am Coll Cardiol 2004; 43:2177–82

100 Stugaard M, Smiseth OA, Risoe C, Ihlen H. Intraventricularearly diastolic filling during acute myocardial ischemia, assess-ment by multigated color m-mode Doppler echocardiography.Circulation 1993; 88: 2705–13

101 Sutherland GR, Stewart MJ, Groundstroem KW, et al. Color

Doppler myocardial imaging: a new technique for theassessment of myocardial function. J Am Soc Echocardiogr 1994; 7:441–58

102 Tavernier B, Garrigue D, Boulle C, Vallet B, Adnet P. Myofilamentcalcium sensitivity is decreased in skinned cardiac fibres of

endotoxin-treated rabbits. Cardiovasc Res 1998; 38: 472–9103 Tavernier B, Li JM, El-Omar MM, et al. Cardiac contractile

impairment associated with increased phosphorylation of tropo-nin I in endotoxemic rats. Faseb J 2001; 15: 294–6

104 Tavernier B, Mebazaa A, Mateo P, Sys S, Ventura-Clapier R,

Veksler V. Phosphorylation-dependent alteration in myofilamentCa2þ sensitivity but normal mitochondrial function in septicheart. Am J Respir Crit Care Med 2001; 163: 362–7

105 Toller WG, Stranz C. Levosimendan, a new inotropic and vaso-

dilator agent. Anesthesiology 2006; 104: 556–69106 Tsuyuki RT, McKelvie RS, Arnold JM, et al. Acute precipitants of

congestive heart failure exacerbations. Arch Intern Med 2001;161: 2337–42

107 Uematsu M, Miyatake K, Tanaka N, et al. Myocardial velocity

gradient as a new indicator of regional left ventricular contrac-tion: detection by a two-dimensional tissue Doppler imagingtechnique. J Am Coll Cardiol 1995; 26: 217–23

108 van Heerebeek L, Borbely A, Niessen HW, et al. Myocardialstructure and function differ in systolic and diastolic heart

failure. Circulation 2006; 113: 1966–73109 Van Trigt P, Christian CC, Fagraeus L, et al. The mechanism of

halothane-induced myocardial depression. Altered diastolicmechanics versus impaired contractility. J Thorac Cardiovasc Surg1983; 85: 832–8

110 Vasan RS, Benjamin EJ, Levy D. Prevalence, clinical features andprognosis of diastolic heart failure: an epidemiologic perspec-tive. J Am Coll Cardiol 1995; 26: 1565–74

111 Vasan RS, Levy D. Defining diastolic heart failure: a call for stan-

dardized diagnostic criteria. Circulation 2000; 101: 2118–21112 Vivien B, Hanouz JL, Gueugniaud PY, Lecarpentier Y, Coriat P,

Riou B. Myocardial effects of halothane and isoflurane in ham-sters with hypertrophic cardiomyopathy. Anesthesiology 1997;87: 1406–16

113 Wallace A, Layug B, Tateo I, et al. Prophylactic atenolol reducespostoperative myocardial ischemia. McSPI Research Group.Anesthesiology 1998; 88: 7–17

114 Wexler LF, Grice WN, Huntington M, Plehn JF, Apstein CS.Coronary hypertension and diastolic compliance in isolated

rabbit hearts. Hypertension 1989; 13: 598–606115 Xie YW, Kaminski PM, Wolin MS. Inhibition of rat cardiac

muscle contraction and mitochondrial respiration by endogen-ous peroxynitrite formation during posthypoxic reoxygenation.Circ Res 1998; 82: 891–7

116 Yamada T, Takeda J, Koyama K, Sekiguchi H, Fukushima K,Kawazoe T. Effects of sevoflurane, isoflurane, enflurane, and

Pirracchio et al.

720

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 15: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

halothane on left ventricular diastolic performance in dogs.J Cardiothorac Vasc Anesth 1994; 8: 618–24

117 Yusuf S, Pfeffer MA, Swedberg K, et al. Effects of candesartan inpatients with chronic heart failure and preserved left-ventricular

ejection fraction: the CHARM-Preserved Trial. Lancet 2003;362: 777–81

118 Zannad F, Mebazaa A, Juilliere Y, et al. Clinical profile, contem-porary management and one-year mortality in patients with

severe acute heart failure syndromes: The EFICA study. Eur JHeart Fail 2006; 8: 697–705

119 Zile MR, Baicu CF, Gaasch WH. Diastolic heart failure—abnormalities in active relaxation and passive stiffness of the leftventricle. N Engl J Med 2004; 350: 1953–9

120 Zile MR, Brutsaert DL. New concepts in diastolic dysfunction and

diastolic heart failure: Part I: diagnosis, prognosis, and measure-ments of diastolic function. Circulation 2002; 105: 1387–93

121 Zile MR, Gaasch WH, Carroll JD, et al. Heart failure with anormal ejection fraction: is measurement of diastolic function

necessary to make the diagnosis of diastolic heart failure?Circulation 2001; 104: 779–82

Diastolic heart failure in anaesthesia and critical care

721

at Kw

aZulu-N

atal University on January 21, 2011

bja.oxfordjournals.orgD

ownloaded from

Page 16: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

South African Journal of Anaesthesia & Analgesia • November 2005 117

GUEST EDITORIAL 1

Is a rethink of our approach tohypertension necessary?

IntroductionThe risks and management of hypertensive patients forelective anaesthesia are often debated. There is aperception that patients with untreated or ineffectivelytreated hypertension are at risk, in particular for cardiacand cerebrovascular events.

The change in internal to external diameter of thevasculature results in an altered vascular pressure-flowrelationship in the hypertensive patient. This gives rise tothe exaggerated hypertensive and hypotensive responseseen. The hypotensive tendency is aggravated by thediastolic dysfunction often present. In addition,autoregulation is right shifted, impacting on the lower limitof autoregulation.

Treating patients for their hypertension requires monthsto years to restore the autoregulation towards normal, eventhough the blood pressure may have been be normalized.

Except for patients with significant end-organdysfunction (such as cardiac failure), application of ourknowledge of the pathophysiology of hypertension,combined with skill, should allow the anaesthetist toreconsider some of the current dogma in the managementof the hypertensive patient.

Anaesthetists have differing approaches to the patientwith hypertension scheduled for elective surgery. On manyoccasions, this leads to surgery being cancelled becausethe practitioner is of the opinion that the hypertension isuncontrolled. In this paper, the position projected by theauthors, is that if the pathophysiology of hypertension isclearly understood, the majority of hypertensive patientscan be accepted for anaesthesia, whether blood pressurecontrol is deemed adequate or not. Nonetheless, it is alsoimportant that the anaesthetist appreciates the exceptions,and the reasons for these exceptions.

It must be made clear at the outset that this paper iswritten with the intent to provoke debate and discussion

on this topic. Furthermore, the opinion stated in this articledoes not question existing wisdom that long-termadvantages can be gained from the effective treatment ofchronic hypertension. The discussion that follows thereforeapplies only to the perioperative period.

Anaesthetic problems associated with thehypertensive patientThe hypertensive patient presents the anesthetist withthree main problems:1. Episodes of hypertension;2. Periods of hypotension;3. Concerns regarding (pre-existing or consequent) end-

organ damage, especially the brain, heart, vasculatureand kidneys.

The first two are well known occurrences in patientswith hypertension subject to anesthesia and surgery. Inour view, understanding the pathophysiology shouldenable the anaesthetist to prevent these problems. Wewill also argue that concerns regarding end-organdamage are speculative and or can be effectivelymanaged.

The essential pathophysiology of hypertensionVascular pathophysiology and blood pressure labilityChanges occur both in:• The structure of the peripheral arteries ;• Intravascular blood volume.

These pathophysiological changes are of practicalsignificance as they are central to the understanding ofthe variations in blood pressure that anaesthetist has todeal with.

There are a number of well described changes thatoccur in the large, medium and small arteries andarterioles. Cumulatively this is referred to as medialhypertrophy.1 Medial hypertrophy results in the normalvascular lumen to wall ratio of 1:5.2 decreasing to 1.3 to4.7 in the hypertensive patient.2

The importance of this smaller than normal lumen towall ratio was summarized in experimental work by

Correspondence:

Prof Andre Coetzee

e-mail: [email protected]

A Coetzee, A LevinDepartment of Anaesthesiology and Critical Care, Faculty of Health Sciences, University of Stellenbosch, Tygerberg,Stellenbosch, South Africa

Page 17: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

South African Journal of Anaesthesia & Analgesia • November 2005 118

GUEST EDITORIAL 1

Folkow.3,4 In his studies, controlled perfusion of thevasculature of normotensive and hypertensive ratsresulted in the flow – pressure relationships depicted inFigure 1. From the flow – pressure quotient, which is theinverse of resistance, one can deduce from the graph thatnormotensive rats have a lower vascular resistance whencompared to the hypertensive rat.When Folkow subjected the vasculature to catecholaminestimulation, (Figure 2), the threshold for a response wasthe same in the normal and hypertensive rats. However,once a response was obtained, the slope of the dose-resistance relationship was steeper for the hypertensiverats compared with the normotensive ones.3,5

The following conclusions can be drawn from theseexperiments:• The threshold for obtaining a pressure response

appears to be similar for the hypertensive andnormotensive rat. Therefore, the problem is not thatthe vasculature of hypertensive rats is inherently moresensitive to stimuli.

• However, the problem is that once a response isobtained, the extent of this response is morepronounced in the hypertensive compared with thenormotensive rats.

The interpretation of these results is that there is nodifference in autonomic control of the vasculature inhypertensive and normotensive subjects. However, thepathophysiological problem is that resistance to flow ishigher in hypertensive subjects because of the smallerinternal lumen of the vasculature. These conclusionsapply equally to hypertensive human subjects.5

Blood pressure changes in lieu of the vascularchangesFrom the above results, Folkow and colleagues compiledthe instructive vascular response curves for bothnormotensive and hypertensive subjects (Figure 3).3,4 Theauthors used % shortening of the circumferential arterialmuscles and resistance on the x and y-axes respectively.For the purpose of clinical applicability and clarity, the xand y-axes may be relabeled “vasoconstriction” and“blood pressure” respectively. These extrapolations arejustified because of the relationship between thecircumference of a vessel and its radius (circumference =2πr) and the relationship between pressure, flow andradius described in Pouseuille’s equation.6

Consider, on the one hand, what would happen ifvasoconstriction occurred and the vessel diameter in Fig4 decreased from a1 to a2. The resulting increase inblood pressure would be significantly greater in thehypertensive patient, compared with the normotensiveone.

On the other hand, if the stress is removed, autonomicmediated vasoconstriction will decrease and the internalradius of the vessel will increase. For similar decreases invessel radius (from c1 to c2 on the y axis of Fig. 5), theresultant fall in blood pressure is represented bydecreases from d1 to d2, and from e1 to e2, for thenormotensive and hypertensive patients respectively.Figure 5 therefore illustrates that the extent of the fall inblood pressure, after similar percentage increases in

Fig 1: Flow and pressure relationship in normo (N) and hypertensive (H) ratvasculature. The resistance is higher in the H rat.

Fig 2: Response of the resistance to catecholamine stimulation. The threshold forthe normotensive (N) and hypertensive (H) are the same but the responsethereafter is clearly more in the H vasculature as compared to the N vasculature.

Fig. 3: Vessel diameter versus resistance (or blood pressure) in normotensive(N) and hypertensive (H) rats. For any degree of constriction the blood pressurein H will exceed the pressure in N.

Fig 4: For any particular increase in vasoconstriction (a1 to a2) , the resultantincrease in blood pressure will be significantly more in the H (c1 to c2)compared to the normotensive (N) (form b1 to b2). This graph explains theexcessive pressor response seen in the uncontrolled hypertensive patient.

Page 18: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

South African Journal of Anaesthesia & Analgesia • November 2005 119

GUEST EDITORIAL 1

vessel diameter, will be much greater in hypertensivethan normotensive patients.

On studying Figures 4 and 5, it is easy to appreciate boththe excessive hypertension following a vasoconstrictorystimulus (for example laryngoscopy and intubation) andalso the exaggerated fall in pressure that follows evensmall amounts of vasodilation (for example when theappropriate anaesthetic level for surgery has beenachieved).

On studying Figures 4 and 5, it is also noteworthy that,when the point of maximal vasodilatation is approached, theFolkow relationships for both hypertensive and normotensivepatients approximate each other and are located on the lesssteep part of the vessel radius - pressure curve. Therefore,albeit the resting vessel resistance remains increased inhypertensive patients, the percent blood pressure changewhich occurs when the vessel diameter changes, is lesswhen the patient is operating on the left side of the Folkowcurve.

Another important pathophysiological principle is thatuncontrolled hypertensive patients have a contractedintravascular blood volume.7,8 The effect of this will beaggravated by the well documented diastolic dysfunctionoften seen in the hypertensive patient.9 The single mostimportant defense against a fall in blood pressure, when thestress stimulus is withdrawn, is the status of theintravascular volume.

Hence, the hypertensive patient is not only penalizedby virtue of the vessel diameter dynamics, but preload,the main physiological buffer protecting againsthypotension, is less efficient. Notwithstanding thisproblem, the opinion has often been expressed that,because a patient is hypertensive, fluid loading should beon the conservative side. Such an approach (partially)explains the subsequent hypotension the anaesthetist hasto deal with. In contrast, the aforementionedpathophysiology suggests that a liberal fluid policy shouldbe applied, while simultaneously considering the cardiacfunction of any particular patient. The vast majority ofhypertensive patients have normal or near normal cardiacfunction and can cope with the required 10 – 15%expansion of the blood volume. To effectively expand theintravascular volume, consideration should be given tousing a colloid that is retained within the intravascularspace for a significant period of time.

AutoregulationAutoregulation is defined as the “intrinsic tendency of anorgan or tissue to maintain constant blood flow despitechanges in arterial pressure”.10 The salient feature illustratedin a characteristic normal autoregulation curve is thatbetween certain limits of blood pressure (marked by points aand b in Figure 6), organ blood flow is constant. If bloodpressure decreases below point a, termed the lower limit forautoregulation (LLA), or above point b, the upper limit forautoregulation, flow becomes pressure dependant. Pressuredependant flow has particular physiological implications forthe anaesthetist. To illustrate these implications, one canapply the autoregulation concept to the brain. Normal brainblood flow averages 50 ml/min/100g tissue. Below the lowerlimit of autoregulation, brain blood flow progressivelydecreases. When brain blood flow decreases to 20 - 25 ml/100g/min, electrical activity of the brain ceases.11 Cessationor a decrease of organ function with reductions in their bloodflow and nutrient supply is termed hibernation, a term moreoften used in cardiac pathophysiology. Hibernationrepresents a method whereby organs protect themselvesagainst low oxygen supply scenarios and does notnecessarily equate to tissue damage. Once nutrient flow isimproved, function will be restored. Only when blood flowdecreases to 6 - 15 ml/min/100g brain tissue, will membranedysfunction and neuronal death occur.

The important question that has to be addressed is what isthe mean arterial pressure (MAP) at which organ bloodflow decreases to potentially dangerous levels? Strangaardand colleagues12 studied the effects of decreasing bloodpressure on cerebral blood flow in awake patients. The 3groups they studied comprised normotensive (mean arterialpressure [MAP] 98 ± 10 mm Hg), treated hypertensive(MAP 116 ±18 mm Hg) and untreated hypertensive subjects(MAP 145 ±17 mm Hg). The initial part of their studycomprised lowering blood pressure and noting the pointwhere flow becomes pressure dependant (LLA). Thereafter,they continued lowering blood pressure to the level wherethe patients demonstrated signs of cerebral ischaemia(ischaemic threshold, IT).

A particularly instructive aspect of the Strangaard study isthat, if the LLA is described as a percentage of the pre-testMAP, the LLA occurred at approximately 75% of the baselineMAP. This study also demonstrated that a significant linearrelationship between the MAP and LLA (and IT) existed. The

Fig 5: If vasodilation occurs (from a1 to a2), the extent of the fall in pressurewill be from b1 to b2 for the normotensive (N) as compared to the greater fall(from c1 to c2) in the hypertensive.

Fig 6: Autoregulation in the normotensive (N) and hypertensive (H) patient. TheH curve is right shifted and of significance is the lower limit for autoregulation(LLA) which increases from a to c. The upper limit also increases from b to d.

Page 19: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

South African Journal of Anaesthesia & Analgesia • November 2005 120

GUEST EDITORIAL 1

Strangaard study is valuable in that it provides theanaesthetist with a rough guide as to how much reduction ofthe awake preoperative blood pressure can be toleratedintraoperatively. The important guideline that is created isthat a reduction of blood pressure in excess of 20% ofbaseline should not be tolerated.

Albeit the above guideline is useful, cognizance should betaken of the wide variation in LLA that occurs. Drummond13,in a letter to the editor of Anesthesiology, pointed out thatmean LLA values range between 73 to 91 mm Hg in awakenormotensive patients; however LLA values for individualsvaried between 41 to 113 mm Hg. This wide variation in LLArepresents perhaps one of the limiting factors when dealingwith the concept of autoregulation i.e. the anaesthetist doesnot know the exact limits for any particular patient.

In the hypertensive patient it has been confirmed that thepositions of the coronary and cerebral autoregulation curveare shifted to the right.12,14

The problem that the anaesthetist is therefore presentedwith is that the position of the LLA and slope of the pressuredependant portion of the autoregulation is unknown forindividual patients. In other words, it is not known at whatblood pressure organ blood flow will decrease sufficiently toresult in tissue damage. Hence it is prudent thatanaesthetists elect not to approach the LLA too closely andremain on the flow independent part of the autoregulationcurve.

Changes in pathophysiology with treatmentRat studies indicate that antihypertensive drug therapyeffectively reduces blood pressure. However, from theanaesthetist’s perspective, the reduction of blood pressure isnot the final aim of antihypertensive treatment. What is ofimportance to the anaesthetist is preservation of organ bloodflow. It is therefore of some significance to note that only ifthis therapy is effective in reducing blood pressure and isadministered for sufficient time, will remodeling of thevasculature occur. Chronic antihypertensive treatment (overan extensive period) represents a method of normalizingautoregulation and lessening the risk of organunderperfusion during anaesthesia.

Two studies are instructive in this regard. In the one study,Hoffman15 subjected both spontaneous hypertensive (SHR)and normotensive rats to antihypertensive therapy. In theSHR group that was treated with antihypertensive drugs,blood pressure was effectively reduced to that of the controlgroup after 2 weeks of therapy. At this point, they subjectedboth the treated and untreated animals to hypotensiveepisodes. There was no difference between cerebral bloodflow and oxygen consumption in the treated and untreatedSHR groups. The lesson learned from this study is thatnormalization of blood pressure is not synonymous withnormalization of autoregulation.

It is also noteworthy that in the Hoffman study,autoregulation was not restored by 10 weeks ofantihypertensive therapy. If it is considered that a single ratmonth approximates 37 human months, and if we areallowed the liberty of a direct extrapolation of these timeperiods, it implies that blood pressure must be effectivelycontrolled for years in humans before the anesthetist can rely

on a normal autoregulation curve.Another study16 demonstrated similar results to the

Hoffman study. In this study, a renal artery clip hypertensiverat model was studied. After release of the clip, bloodpressure decreased to normal values within 1 week.However, the exaggerated pressor response seen inhypertensives only normalized 19 weeks after removal of theclip.

This perspective presents anaesthesiologists with thefollowing dilemma. Even if the patient is on treatment forhypertension, we cannot assume that autoregulation isnormalized until months or years have passed after thetherapy was initiated. Therefore, when we are presentedwith patients that have hypertension who have relativelyrecently started treatment, we do not know whichintraoperative blood pressure should be regarded as “safe” inorder to maintain organ blood flow?

Risk to organsLong standing uncontrolled hypertension is a proven threatto the brain, heart, vasculature, kidneys and eyes. However,in the anaesthetist’s perioperative management, the twoorgans that are paramount are the brain and heart. This doesnot imply that the other organs can be managed with acasual attitude, but that these two organs present a greaterpotential to cause acute life threatening crises.

Cardiac riskThe question arises whether hypertension results in anincreased cardiovascular risk? It is interesting that theGoldman cardiac risk index17,18, one of the earliest and bestknown scoring systems developed to determine perioperativecardiovascular risk, did not include hypertension as one of itsparameters. Furthermore, a large study published in 1990also did not consider hypertension as an independent riskfactor in patient undergoing non-cardiac surgery.19 However,Howell and colleagues20 did report an association betweenthe history of hypertension and perioperative cardiovasculardeath. A meta-analysis could not confirm a significantincrease in the odds ratio for perioperative cardiac outcomeand hypertensive disease (OR 1.35 (1.17 – 1.56)).21

At a minimum there is significant doubt whether a clearlink has been established between hypertension andperioperative cardiovascular risk with the proviso that certainaspects of the cardiac risk be managed in an effective manneras is explained below.

The earliest studies from the Oxford Group, lead by Prys-Roberts22,23, warned that the untreated or ineffectivelytreated hypertensive patient is at risk of both excessivepressor response and myocardial ischaemia. They alsoreported that, in these hypertensive patients, theperioperative use of β adrenergic receptor blockers reducedthe incidence of myocardial ischaemia. In the non β-blockedpatients, the incidence of myocardial ischaemia was 38%compared to 4% in those who received practolol. The OxfordGroup’s studies were conducted in a small group of patients,but were reconfirmed in a larger and more recent study.24

Stone and colleagues24 (comparing oxprenolol, acebutolol andlabetolol with no treatment in hypertensives) showed thatuse of β adrenergic blockers in the perioperative period,

Page 20: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

South African Journal of Anaesthesia & Analgesia • November 2005 121

GUEST EDITORIAL 1

reduced the incidence of myocardial ischaemia to much thesame figures as reported in the earlier Prys Roberts study. Oncloser inspection of the Stone data, it is evident that the peakblood pressures recorded during intubation and extubationwere within reasonable clinical proximity of each other(except for a group who received labetolol). The untreatedpatients had mean blood pressures of 125±4 and 127±3mmHg on intubation and extubation respectively. Thiscompares favorably with the 118±4 and 119±3 mmHgrecorded in the patients treated with oxprenolol. However,more consistent differences in heart rate were observed inthe Stone study. Data demonstrated that the group whoreceived β adrenergic blockade had lower heart rates duringperiods of stress compared with non-treated patients. It isreasonable to suggest that the differences in the incidence ofmyocardial ischaemia between the groups were mostprobably the result of the slower heart rate associated with βadrenergic blockade and not the insignificant differences inblood pressure per se.

Why is it that the myocardial ischaemia seen during orafter laryngoscopy and intubation as well as other periods ofstress, appears to be related to increases in heart rate ratherthan increases in blood pressure? The answers may lie, onthe one hand, with the observation that coronary blood flowvaries directly with the perfusion pressure in the presence ofa critical coronary artery stenosis.25 This means that smallincreases in blood pressure will increase or maintain coronaryblood flow. On the other hand, the myocardial ischemiaassociated with periods of stress is most likely the result ofan oxygen delivery problem associated with the decrease indiastolic time that accompanies an increase in heart rate.26

According to this interpretation of the data, the maincardiac risk for the hypertensive patient is an ischaemicinsult. This risk is more closely related to diastolic coronaryperfusion time and not the blood pressure. In thehypertensive patient, provided that the pressor response isnot so great that it causes the left ventricle (LV) to fail, thelogical conclusion is that the major acute perioperativecardiac risk (i.e. myocardial ischaemia) can be managed withβ adrenergic blockers. Therefore, the perceived cardiac risksshould not deter the anaesthetist from subjecting thehypertensive patient to anaesthesia and surgery, whetherthe hypertension is controlled or not.

In fact, the popular practice of using β blockers as anadjuvant to reduce cardiovascular stress, acknowledges theabove logic. Vascular tone is the result of a balance betweenβ

2 and α adrenergic stimuli. If the β

1 and β

2 receptors are

blocked with an appropriate drug, the alpha-receptors areleft, so to speak, relatively unopposed. This explains why inmany patients there may well be a further small increase inblood pressure when α adrenergic blockers are initiated.Hence, treatment of the expected hypertensive pressorresponse with a β adrenergic blocker alone defies logic, but inmost instances, serendipitously brings the ischemic riskunder control by containing the heart rate.

There are 3 other caveats regarding the cardiovascularsystem and anaesthesia in hypertensive patients.1. Assidao and Donegan27 have shown that uncontrolled

preoperative hypertension is associated with an increased

risk for transient ischemic attacks (TIA) in patientsundergoing carotid endarterectomy. This represents one ofthe few studies and scenarios in which the hypertensivepatient has unequivocally been shown to have anincreased perioperative risk.

2. It is important to appreciate that, if left ventricular (LV)afterload is high enough to prevent the LV ejectingeffectively, the blood pressure requires effective treatmentbefore embarking on anaesthesia. This caveat especiallyapplies to patients with marginal or poor LV performance.

3. There is also the fear that the hypotensive episodes willresult in trespassing into the region below the LLM. Thishypotensive risk can be managed in the following ways:a. Careful fluid preloading, taking cognizance of the 15-

200% intravascular fluid deficit,b. The use of the synergistic relationship that exists for

example between opioids and anaesthesia agents. Thissynergistic relationship applies to both the reduction inthe dose of induction agents needed to produce sleep,as well as the reduction in the MAC of inhalationagents.

c. Frequent measurements of blood pressure.d. The availability of pressor agents to maintain blood

pressure if required.

In summary, there is no final and or convincing proof thatblood pressure per se represents the major risk in thehypertensive patients when viewed from a cardiacperspective. The real risk appears to be that of an increase inheart rate and myocardial ischaemia; we have drugs that caneffectively manage this potential problem. Furthermore, thecaveats mentioned emphasize that the approach to thehypertensive patient must be individualized and end-organfunction examined before a decision can be made whether ornot to accept the current blood pressure for the purpose ofanaesthesia.

The BrainAnother organ that is of great concern to the anaesthetistwhen the topic of hypertension is discussed, is the brain. Theconcern is that because perioperative blood pressure canvary significantly in hypertensive patients, this may lead tocerebral injury. Because of the position of the autoregulationcurve, hypotension would cause hypoperfusion of the brain,whereas high blood pressures would rupture the vessels andcause cerebral bleeding.28

Any brain injury (which was caused by excessive pressureand flow variation during anaesthesia) is likely to be evidentshortly (hours rather than days) after surgery.

A review by Kam and Calcroft29 estimates the incidence ofperioperative stroke during general surgery at 0.2 – 0.7%.Nonetheless, this incidence of perioperative stroke ismodified by several factors. Both Kam and Calcroft29 andWong30 emphasize that the incidence of stroke varies withage. The risk of perioperative stroke in males is 0.38% forsubjects over 50, 0.5% over 70, 3.54% over 80 and 3.2% overthe age of 90. The stroke usually occurred well into thepostoperative period, happening on average on the 7thpostoperative day.

Moreover, a prior history of cerebrovascular pathology has

Page 21: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

South African Journal of Anaesthesia & Analgesia • November 2005 122

GUEST EDITORIAL 1

been shown to be associated with a 10 fold increase in theincidence of perioperative stroke.31

The association between hypertension and postoperativestroke has been examined by a limited number of studies. Astudy often referred to is that of Parikh and Cohen.32 Thisretrospective study with matched controls studied 24641patients and reported a perioperative stroke rate of 0.08%.On close inspection of the paper, issues are raised whichbelie the conclusions the authors made i.e. hypertension is arisk for perioperative stroke. Firstly, 6 of their 20 patients thatsuffered a stroke had atrial fibrillation (AF), a major risk factorin itself for perioperative stroke. Secondly, one patient had anepisode of significant hypertension in the postoperativeperiod. A subsequent CT scan revealed the patient hadsuffered a cerebral bleed. This begs the question as towhether the bleed or the hypertension occurred first. Lastly,if both the published stroke rate for the 67 year average agein this study and also the atrial fibrillation group is taken intoaccount, it becomes clear that the conclusions of this study,that hypertension is a risk for perioperative stroke, wereindeed speculative.

Data studying the association of perioperative stroke andhypertension suffers from a problem with the definition ofwhat is meant by the term “perioperative”? It is the opinionof the authors that if blood pressure fluctuations, be it hyper-or hypotension, are indeed the cause for cerebral injury, itshould be present immediately or very soon after theanaesthetic. We are critical of studies that employ, forinstance, a 30-day postoperative evaluation period as areflection of the effects of blood pressure fluctuations duringanesthesia on brain tissue. While it may be acceptable toterm this period “perioperative”, it is probably not acceptableand speculative to attribute a stroke to blood pressurevariation occurring during the procedure.

It is relevant to this discussion that the data from a studyconducted by Hart and Hindman33 be considered. This studyshowed that stroke was associated with hypotension thatoccurred in the postoperative recovery period. There was noassociation between stroke and intraoperative events andthis study emphasizes the necessity for close control of bloodpressure into the post operative period.

The role of AF is repeatedly stressed as one of the majorcauses of postoperative stroke. The data from Hart andHindman32 reveals that in 42% of their cases, stroke wascardiogenic in origin with AF being the most important singlefactor.

Larsen34 found 9 postoperative strokes in 2463 surgicalpatients. However, on close examination of this data, themajority of the strokes occurred after the 5th postoperativeday. In addition, 3 patients had AF and only 4 hadhypertension. Of those 4 patients with hypertension, 2 hadAF. Furthermore, only two patients did not have other knownrisk factors such as previous cerebral incidents and dementia.If the 2 patients with AF in the hypertensive group arediscounted, only two patients who suffered a strokepostoperatively (from day 5 onwards) had hypertension. Also,it is reasonable to ask why the authors considered that strokeon or after the 5th postoperative day was related toanaesthesia. This interpretation gives a different perspectiveof the data as presented by the authors.

In summary, studies looking at the association betweenperioperative stroke and hypertension suffer from manylimitations, and are difficult to interpret. While logicsuggests that tight blood pressure control is to beadvocated perioperatively, at least one study disputes thecontention that intraoperative hemodynamic instabilityincreases perioperative stroke. Furthermore, there is clearevidence that factors other than perioperative bloodpressure fluctuations such as AF, increasing age,postoperative hypotension, uncontrolled hypertension inpatients scheduled for carotid endarterectomy, and priorcerebrovascular incidents increase the risk of perioperativestroke.

A time for change?Decisions to accept hypertensive patients for electivesurgery are frequently non-scientific and based on “gutfeeling” driven opinions. The available guidelines alsoreflect personal preference and the uncertainty.35 Indeveloping a new approach, the following should beconsidered:1. The anaesthetist must understand the pathophysiology

of the variation in blood pressure that is so often seen inthese patients. The initiation of antihypertensivetreatment will result in rapid reductions in bloodpressure. However, the anaesthetist may be misled toaccept the new lower pressure as a reference point. Itmust be remembered that vascular remodeling is stilltaking place and both the pressor response andautoregulation are still that of the hypertensive patient.It probably requires many months or even years ofeffective treatment before it is reasonable to accept thatvascular remodeling has normalized. One must not bemisled by the dangerous practice of colleagues whostart treatment in their rooms and admit the patient oneweek later with a normal blood pressure.

2. Our (extrapolated) knowledge about the change in theautoregulation curve must be meticulously applied. Thismeans that variations of more than 20% in thepreoperative mean arterial pressure should not beallowed.

3. Contingency plans should be made to measure, preventand treat hypotension by restoration of the intravascularvolume and use of α adrenergic agonists. Hypertensiveepisodes can easily be managed firstly by ensuring aproper surgical level of anaesthesia and secondly, withthe use of intravenous vasodilators.

4. It appears that the cardiac risk, is primarily that ofmyocardial ischaemia. To a large extent, heart ratecontrol will contain this problem. However, it must alsobe understood that β adrenergic blockers will notcontain the pressor response to stressful stimuli. Asmentioned, administration of an intravenous vasodilatormay be required to control pressure surges.

5. Literature seems to suggest an increased perioperativerisk of stroke in hypertensive subjects. However,rigorous interrogation of the published data indicatesthat the real risk of stroke appears to be a veryuncertain issue.a. Nonetheless, cognizance must be taken of the fact

Page 22: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

South African Journal of Anaesthesia & Analgesia • November 2005 123

GUEST EDITORIAL 1

that patients with prior cerebral incidents appear tobe at a higher risk of sustaining postoperativecerebral events.

b. Furthermore, carotid artery surgery results in ahigher incidence of cerebral events in patients whohave elevated blood pressure preoperatively. In thissubgroup of patients, the available data suggeststhat preoperative blood pressure control is indeedwise.

6. The blood pressure that the anesthetist should acceptmust be individualized. For instance, patients with araised blood pressure who is either in congestive heartfailure, or is scheduled for carotid endarterectomy, havespecific and understood reasons why the blood pressureneeds to be controlled before embarking on electivesurgery.

In the light of better knowledge of the pathophysiology,appreciation of the above proviso’s, combined withanaesthetic skills and the judicious use of the appropriatedrugs, the often quoted views on hypertension in theperioperative period need to be reconsidered.

References1. Leitschuh M, Chobanian A. Vascular changes in hypertension.

Med Clin North America 1987; 71: 827 – 839.

2. Van Citters RL. Occlusion of lumina in small arterioles during

vasoconstriction. Circ Res 1966; 18: 199 – 204.

3. Folkow B. The hemodynamic consequences of adaptive

structural changes of the resistance vessels in hypertension.

Clinical Sciences 1971; 41: 1 – 12.

4. Folkow B, Grimby G, Thulesius O. Adaptive structural changes

of the vascular walls in hypertension and their relation to the

control of the peripheral resistance. Acta Physiologica

Scandinavica 1956; 44: 255 – 272.

5. Sivertsson R, Olander R. Aspects of the nature of increased

vascular resistance and increased “reactivity” to noradrenaline

in hypertensive subjects. Life Sciences 1968; 7: 1291 – 1297

6. Giancoli D. In : Physics. Principles and Applications. Prentice

Hall , 5th Ed, 1988; 295 and 532.

7. Bauer JH, Brooks CS. Body fluid composition in normal and

hypertensive man. Clinical Science 1982; 62: 43 – 49.

8. Dagnino J, Prys Roberts C. Strategy for patients with

hypertensive heart disease. Clin Anesth 1989; 3: 261.

9. Pagel P, Grossman W, Haering JM, Warltier DC. Left ventricular

diastolic function in the normal and diseased heart.

Anesthesiology 1993; 79: 836 - 854.

10. Dorland’s Illustrated Medical Dictionary. 25th edition. WB

Saunders, Philadelphia, London, Toronto. 11.

11. Drummond J, Sharpiro H. Cerebral physiology In: Anesthesia .

Ed RD Miller 4th Ed, Churchill Livingstone, New York 1994;

711.

12. Strangaard S, Oleson J, Skinhoj E et al. Autoregulation of brain

circulation in severe arterial hypertension. Br Med J 1973; 1:

507 - 509.

13. Drummond J. The lower limit of autoregulation. Time to revise

our thinking ? Anesthesiology 1997; 86: 1431 – 1432.

14. Polese A, DeCesare N, Montorsi P et al Upward shift of the

lower range of coronary flow autoregulation in hypertensive

patients with hypertrophy of the left ventricle. Circulation

1991; 83: 845 - 853.

15. Hoffman WE, Miletich DJ, Albrecht RF. The influence of

antihypertensive therapy on cerebral autoregulation in aged

hypertensive rats. Stroke 1982; 13: 701 - 705.

16. Lundgren Y. Regression of cardiovascular changes after

reversal of experimental hypertension in rats. Acta Physiol

Scand 1974; 91: 275 - 283.

17. Goldman L, Caldera D, Nussbaum SR et al. Multifactorial index

of cardiac risk in non-cardiac surgical procedures. N Eng J Med

1977; 297: 845 - 850.

18. Goldman L, Caldera D. Risk of general anesthesia and elective

operation in the hypertensive patient. Anesthesiology 1979; 50:

285 - 292.

19. Mangano D, Browner W, Hollenberg M et al. Association of

preoperative myocardial ischemia with cardiac morbidity and

mortality in men undergoing non-cardiac surgery. N Eng J Med

1990; 323: 1781 - 1787.

20. Howell SJ, Hemming AE, Allman KG et al. Predictors of

postoperative myocardial ischemia. The role of intercurrent

arterial hypertension and other cardiovascular risk factors.

Anaesthesia 1996; 57: 1781.

21. Howell SJ, Sear J, Foex P. Hypertension, the hypertensive heart

disease and perioperative cardiac risk. Br J Anaesth 2004; 92:

570 - 583.

22. Prys Roberts C, Foex P, Biro G et al. Studies of anaesthesia in

relation to hypertension. V Adrenergic beta receptor blockade.

Br J Anaesth 1973; 45: 671 – 681.

23. Prys Roberts C, Meloche R, Foex P. Studies of anaesthesia in

relation to hypertension. I Cardiovascular responses of treated

and untreated patients. Br J Anaesth 1971;43: 122- 137.

24. Stone JG, Foex P, Sear J et al. Myocardial ischemia in untreated

hypertensive patient: Effect of a single small oral dose of beta

adrenergic blocking agent. Anesthesiology 1988; 68: 495 - 500.

25. Coetzee A, Foex P, Ryder A. Coronary blood flow during

variation in blood pressure. S Afr Med J 1985; 68; 15.

26. Coetzee A, Coetzee G. Beta-adrenergic blockade during

ischemia. J Cardiothorac Vasc Anesth 2002; 16: 670 – 679.

27. Assidao C, Donegan JH. Factors associated with perioperative

complications during carotid endarterectomy. Anesth and

Analg 1982; 61: 631.

28. Mayham WG, Faraci FM, Heistad D. Disruption of the blood

brain barrier in the cerebrum and brain stem during acute

hypertension. Am J Physiol 1986; 251: H 1171 – H 1175.

29. Kam PCA, Calcroft RM. Perioperative stroke in general surgical

patients. Anaesthesia 1997; 52: 879 – 883.

30. Wong D. Perioperative stroke: general surgery, carotid disease

and carotid end-arterectomy. Can J Anaesth 1991; 38: 347 –

373.

31. Landecasper J, Metz EJ, Coghill TH et al. Perioperative stroke

risk in 173 consecutive patients with a past history of stroke.

Arch Surg 1990; 125: 986 – 989.

32. Parikh S, Cohen JR. Perioperative stroke after general surgical

procedures. NY State J Med 1993; 93; 162 – 165.

33. Hart R, Hindman B. Mechanisms of perioperative cerebral

infarction. Stroke 1982; 13: 766 - 773.

34. Larsen SF, Zaric D, Boysen G. Postoperative cerebrovascular

accidents in general surgery. Acta Anaesthesiol Scand

1988; 32: 698 -701.

35. Miller pp 937. In: Anesthesia . Ed RD Miller, 4th Ed, Churchill

Livingstone, New York, 1994; 937.

Page 23: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Hypertension, hypertensive heart disease andperioperative cardiac risk²

S. J. Howell1*, J. W. Sear2 and P. FoeÈx2

1Academic Unit of Anaesthesia, University of Leeds, Leeds General In®rmary, Leeds LS1 3EX, UK.2Nuf®eld Department of Anaesthetics, University of Oxford, John Radcliffe Hospital, Headley Way,

Headington, Oxford OX3 9DU, UK

*Corresponding author. E-mail: [email protected]

The evidence for an association between hypertensive disease, elevated admission arterial pres-

sure, and perioperative cardiac outcome is reviewed. A systematic review and meta-analysis of

30 observational studies demonstrated an odds ratio for the association between hypertensive

disease and perioperative cardiac outcomes of 1.35 (1.17±1.56). This association is statistically

but not clinically signi®cant. There is little evidence for an association between admission arter-

ial pressures of less than 180 mm Hg systolic or 110 mm Hg diastolic and perioperative compli-

cations. The position is less clear in patients with admission arterial pressures above this level.

Such patients are more prone to perioperative ischaemia, arrhythmias, and cardiovascular labi-

lity, but there is no clear evidence that deferring anaesthesia and surgery in such patients

reduces perioperative risk. We recommend that anaesthesia and surgery should not be can-

celled on the grounds of elevated preoperative arterial pressure. The intraoperative arterial

pressure should be maintained within 20% of the best estimate of preoperative arterial pres-

sure, especially in patients with markedly elevated preoperative pressures. As a result, atten-

tion should be paid to the presence of target organ damage, such as coronary artery disease,

and this should be taken into account in preoperative risk evaluation. The anaesthetist should

be aware of the potential errors in arterial pressure measurements and the impact of white

coat hypertension on them. A number of measurements of arterial pressure, obtained by com-

petent staff (ideally nursing staff), may be required to obtain an estimate of the `true' preopera-

tive arterial pressure.

Br J Anaesth 2004; 92: 570±83

Keywords: arterial pressure; complications, hypertension; risk, perioperative

The importance of hypertension

The association between elevated arterial pressure and

cardiovascular disease is unequivocally established and well

known to doctors and the general public. The risk of

cardiovascular events in the general population increases

steadily with increases in arterial pressure. The individuals

at greatest risk of suffering a cardiovascular event because

of hypertension are those with the highest arterial pressures.

However, mild to moderate hypertension is more common

than severe hypertension, and much of the population

burden of disease because of hypertension may be attributed

to moderate rather than severe hypertension. This is

illustrated in Figure 1, which documents the association

between systolic hypertension and deaths as a result of

coronary artery disease.74 The highest risk of death is seen

in patients with systolic arterial pressures of greater than

180 mm Hg. However, the greatest number of excess deaths

(calculated as the difference between the number of deaths

that would be expected from coronary artery disease on the

basis of the rate in the group with a systolic arterial pressure

of less than 110 mm Hg and the number of deaths actually

recorded) is seen in the largest group of subjects. That is,

those with systolic arterial pressures of between 140 and 149

mm Hg. Hence, medical guidelines for the treatment of

hypertension emphasize the treatment of mild to moderate

hypertension. The British Hypertension Society Guidelines

on the management of hypertension use a threshold of 140/

90 mm Hg for the initiation of treatment.63 This review will

REVIEW ARTICLE

²This article is accompanied by Editorial II.

British Journal of Anaesthesia 92 (4): 570±83 (2004)

DOI: 10.1093/bja/aeh091

Ó The Board of Management and Trustees of the British Journal of Anaesthesia 2004

Page 24: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

suggest that, while these guidelines are appropriate for the

medical management of hypertension, the cut-offs that they

use are overly demanding for the management of hyperten-

sion in the perioperative setting.

Historical background

Sprague ®rst identi®ed an association between hypertension

and perioperative cardiac risk in 1929. He described a series

of 75 hypertensive patients of whom one-third died in the

perioperative period; 12 of these had cardiovascular com-

plications.71

The introduction of antihypertensive drugs led to con-

cerns that patients on such drugs might be at increased risk

of perioperative cardiac lability. In 1966, Dingle recom-

mended that patients presenting for anaesthesia and surgery

should, if possible, undergo autonomic testing before their

operation. This would give some indication of their risk of

cardiac lability and whether or not their antihypertensive

therapy should be continued.14 These recommendations

were overtaken by the work of Prys-Roberts and colleagues,

who published a series of studies on the interaction between

hypertension and anaesthesia. The ®rst of these studies

examined a small group of 34 patients undergoing anaes-

thesia and elective surgery.62 Fifteen of the patients were

classi®ed as normotensive, although by current standards all

of their control patients would now be considered

hypertensive. The remainder of the patients were classi®ed

as treated or untreated hypertensives. By current standards,

these patients would probably be considered to have severe

hypertension as several were reported as having systolic

arterial pressures of 220±230 mm Hg. The patients under-

went intensive haemodynamic monitoring. The authors

reported that the untreated hypertensive patients had a

greater decrease in arterial pressure at induction of anaes-

thesia and that they were more prone to intraoperative

myocardial ischaemia. There were no adverse events

reported in either the control or hypertensive groups.

On the basis of these ®ndings the authors recommended

that, where possible, hypertensive patients should have

anaesthesia and surgery deferred to allow their hypertension

to be treated. This recommendation led to a major change in

anaesthetic practice, and to the modern perception that

where possible untreated hypertensives should not be

subjected to elective anaesthesia and surgery without ®rst

treating their arterial pressure. However, these recommen-

dations should be applied with some caution. The percep-

tion of what constitutes hypertension has changed

considerably since these studies were undertaken. Arterial

pressures that in the early 1970s would have been

considered acceptable are today consistent with levels of

hypertension where treatment is obligatory. As already

stated, all of the control patients in the study by Prys-

Roberts and colleagues would now be considered to be

hypertensive. The recommendations of Prys-Roberts and

colleagues therefore need to be reconsidered in the light of

the modern views of hypertension and its management.

The classi®cation of hypertension

It has been indicated above that raised arterial pressure is

associated with a continuum of risk, with greatest risk

associated with the highest arterial pressures. For the

purposes of analysis, discussion and treatment recommenda-

tions, it is necessary to grade and classify raised arterial

pressure in some way or another. This may be done implicitly

by de®ning treatment thresholds, as in the British

Hypertension Society guidelines, or explicitly, by dividing

arterial pressure into bands of increasingly severe hyperten-

sion, as in the classi®cation of the Sixth Joint National

Committee on the Detection, Evaluation and Treatment of

High Blood Pressure (JNC VI).35 However, it has been

pointed out that the differences between classi®cations can

have major implications for estimating the prevalence of

hypertension and the number of people in a population who

may require treatment. The World Hypertension Society/

International Society of Hypertension (WHO/ISH) guide-

lines set lower thresholds than those advocated by either the

British Hypertension Society or JNC VI. Acceptance of the

WHO/ISH thresholds results in 45% of the population as a

whole and 60% of the adult population being classi®ed as

hypertensive.51 It is important to remember the ultimate

goals in the treatment of hypertension. These are the

reduction of the risk of cardiovascular events for the

individual patient and in the population as a whole.

Hypertension is only one of a number of risk factors for

cardiovascular disease and a number of guidelines, includ-

ing those issued by the British Hypertension Society,

advocate treatment not on the basis of arterial pressure

alone but according to the overall estimate of cardiovascular

risk (Fig. 2).63

Fig 1 The effect of systolic pressure at entry to MRFIT (Multiple Risk

Factor Intervention Trial) on the relative risk of death because of

coronary artery disease. The number of excess deaths is calculated

relative to the number of deaths because of coronary artery disease that

would be expected from the death rate in the baseline group, that is those

patients with a systolic arterial pressure of less than 110 mm Hg.74

Hypertension and perioperative risk

571

Page 25: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

For the purposes of this review, the classi®cation of

hypertension described in JNC VI will be followed

(Table 1).35 This classi®cation is based solely on arterial

pressure readings and does not take into account other risk

factors, although the importance of taking these into account

when deciding on treatment is highlighted. It de®nes bands

for both systolic and diastolic pressure. Where a patient's

systolic and diastolic arterial pressures fall into two different

categories, the higher category is selected. It offers a graded

classi®cation with six bands of arterial pressure and

acknowledges that levels of arterial pressure above optimal

pressures of less than 120/80 mm Hg carry some increased

risk, while not leading us to classify a large proportion of the

population as hypertensive. The anaesthetist is often called

upon to take a view on whether or not a given level of

arterial pressure is clinically important. The JNC VI

classi®cation allows us to identify the place of an individual

patient's arterial pressure on a scale of increasing severity. It

does, however, have some limitations when applied to the

patient presenting for surgery. The most important is that the

classi®cation of hypertension is based on the average of two

or more readings of arterial pressure taken at two or more

visits after initial screening. However, in current British

Fig 2 The British Hypertension Society Guidelines. (CHD = Coronary Heart Disease.) (Reproduced with permission from reference 63.)

Table 1 The classi®cation of hypertension proposed by the Sixth National

Committee on the Detection, Evaluation and Treatment of High Blood

Pressure35

Category Systolic arterial pressure Diastolic arterial pressure(mm Hg) (mm Hg)

Optimal <120 <80

Normal 120±129 80±84

High Normal 130±139 85±89

Hypertension

Stage 1 140±159 90±99

Stage 2 160±179 100±109

Stage 3 >180 >110

Howell et al.

572

Page 26: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

practice, it is uncommon for the anaesthetist to have the

bene®t of arterial pressure readings taken on a number of

recent occasions.

De®ning the questions

The perioperative management of hypertensive patients is a

complex issue that can be divided into a number of different

questions.

1. Is having a diagnosis of hypertension of itself

associated with increased perioperative risk, regardless of

the arterial pressure at the time of admission to hospital for

surgery?

2. Is elevated arterial pressure at the time of admission for

surgery associated with increased perioperative cardiac

risk?

3. What is the importance, if any, of poorly controlled

hypertension in the perioperative setting? Is there any

interaction between elevated admission arterial pressure and

being diagnosed with hypertensive disease previously such

that this increases perioperative risk?

4. Does the treatment of elevated admission arterial

pressure before surgery reduce perioperative cardiac risk?

For the purposes of this review the term `hypertensive

patient' refers to anyone who has been labelled a

hypertensive: that is, someone for whom interventions to

lower persistently raised arterial pressure would be appro-

priate, or someone who is already on treatment for

hypertension. Raised arterial pressure will be described as

such.

The core of this review will be an examination of the

available observational studies that address the ®rst three

questions and a discussion of the issues surrounding the

interpretation of these studies. Related issues including

arterial pressure measurement, white coat hypertension, the

use of ambulatory arterial pressure monitoring, and

perioperative arterial pressure lability will also be discussed.

Recommendations will be offered for the perioperative

management of hypertensive patients, although these are

based only on observational data. It should be stated at the

outset that the authors know of no randomized controlled

trial that addresses the ®nal question. The practice of

deferring elective surgery to allow poorly controlled arterial

pressure to be treated is solely based on the perception that

such elevated pressure is associated with increased

perioperative risk, and therefore reducing the arterial

pressure must be a good thing to do. There is no level one

evidence to support this approach.76

Hypertensive disease and anaesthesia

This section presents a meta-analysis of observational

studies examining the association between hypertension

and perioperative cardiac risk in patients undergoing non-

cardiac surgery. Papers were identi®ed as relevant to the

association between hypertension and perioperative

cardiovascular outcome if published between 1971 and the

end of 2001. The former date was chosen as the lower cut-

off year, because this was the year in which the paper

`Studies of anaesthesia in relation to hypertension. I.

Cardiovascular responses of treated and untreated patients'

was published.62 The MEDLINE database was interrogated

using the following combinations of search terms: {anaes-

thesia OR anesthesia} AND cardiac risk; {anaesthesia OR

anesthesia} AND cardiovascular risk; hypertension AND

postoperative complication AND adult NOT animal; hyper-

tension AND intraoperative complication AND adult NOT

animal; arterial pressure AND postoperative complication

AND adult NOT animal; arterial pressure AND intraopera-

tive complication AND adult NOT animal; preoperative risk

strati®cation.

All searches were limited to articles in English. The

abstracts of the papers identi®ed were scanned `on-line' to

identify relevant papers. The reference lists of those papers

that were identi®ed for inclusion in the meta-analysis were

also scanned to identify further relevant studies.

The papers identi®ed from these searches were read in

full. Those that included data concerning the association

between hypertensive disease and perioperative cardiovas-

cular complications were identi®ed. Reports were included

if they examined outcomes considered to be major

cardiovascular complications occurring up to 30 days after

anaesthesia and surgery. Major cardiovascular complica-

tions were considered to be cardiovascular death, myocar-

dial infarction, new or more severe angina, heart failure,

life-threatening arrhythmias, and cerebrovascular accident.

Several studies examined `minor' complications such as

perioperative bradycardia and tachycardia, and periopera-

tive hypotension and hypertension, and more serious

complications. Where it was impossible to separate infor-

mation on major complications from data on all complica-

tions, both major and minor, the study was excluded.

Studies that reported the association between hypertension

and perioperative myocardial ischaemia detected on Holter

monitoring but did not contain data on the association

between hypertension and clinically evident events were

excluded.

For a study to be included, it had to be possible to derive

from the report the crude odds ratio for the association

between hypertension and perioperative cardiovascular

complications, together with the variance of that odds

ratio. The ideal would have been to include the adjusted

odds ratios in which allowance had been made for the effect

of other confounding variables. In most instances, this was

not available.

A number of relevant studies were not primarily designed

to examine hypertension or other perioperative cardiovas-

cular risk factors, but were studies of diagnostic tests for

preoperative cardiovascular assessment or (in one case) of

the value of actively warming the patient during surgery.

We have included those studies where the report of the study

Hypertension and perioperative risk

573

Page 27: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

included relevant data on the association between hyper-

tension and perioperative cardiovascular complications.

A number of studies examining stroke after carotid

endarterectomy have been excluded, as it is argued that

these studies examined a particular complication in an

exceptional population, and the ®ndings from such studies

may not generalize to patients undergoing other types of

surgery.

The main focus of this meta-analysis was the association

between hypertensive disease and perioperative complica-

tions, rather than any association between admission arterial

pressure and such complications. Consequently, studies that

de®ned hypertension solely in terms of the level of

admission arterial pressure were excluded. Studies were

included where the de®nition of hypertension was not given

in the report. For example, in the `Multi-Center Study of

General Anesthesia', the anaesthetist was asked to indicate

if the patient was hypertensive or not, but the de®nition of

hypertension used is not given.20

A total of 4691 citations were identi®ed from the

MEDLINE database. From these, 128 potentially relevant

studies were identi®ed from 126 reports. (The full list of 126

citations can be viewed in the version of this review

published on the British Journal of Anaesthesia website at

http://bja.oupjournals.org/.) For these 128 studies, the full

reports were obtained and read in detail. Ninety-eight

studies described in 97 reports were excluded from further

analysis.

In 80 studies, including the two studies described in a

single paper, an effect estimate for the association between

hypertension and cardiac complications was not given and

could not be derived from the publication. Three studies

were excluded because they appeared to include patients

who had been examined in another study already included in

the meta-analysis. In each case, only one of the pair of

papers concerned was included in the meta-analysis.39 41 58

In six of the excluded studies, hypertension was de®ned in

terms of the arterial pressure alone with no reference to

hypertensive disease. In two studies, hypertension was

de®ned as either an elevated admission arterial pressure or a

history of treatment with antihypertensive medications and

no indication was given of which patients fell into each

category. In three of the excluded studies, no distinction was

made between major cardiovascular complications such as

perioperative myocardial infarction and minor complica-

tions such as intraoperative bradycardia. One study was

excluded because preoperative coronary artery by-pass

grafting and perioperative cardiac complications were

Table 2 Studies included in the meta-analysis of hypertension and anaesthesia. Note that the odds ratios for the study by Sprung and colleagues72 and the

studies by Howell and colleagues33 34 were derived from paired data

Year First author Number ofpatients

Number of hypertensive patients Number of normotensive patients Odds ratio(95% CI)

Withcomplications

Withoutcomplications

Withcomplications

Withoutcomplications

1978 Steen75 587 17 164 19 387 2.1 (1.1±3.8)

1979 Riles67 683 10 339 6 328 1.6 (0.5±4.5)

1981 Von Knorring81 214 10 27 27 150 2.1 (0.9±4.7)

1983 Rao I64 364 9 121 2 232 8.6 (1.8±40.6)

1983 Rao II64 733 3 315 1 414 3.9 (0.4±38.1)

1986 Foster21 1600 66 432 119 983 1.3 (0.9±1.7)

1987 Larsen41 2609 18 443 50 2098 1.7 (1.0±3.0)

1989 Eagle16 200 18 106 12 64 0.9 (0.4±2.0)

1990 Lette44 60 4 21 5 30 1.1 (0.3±4.8)

1990 Shah69 688 25 325 16 323 1.6 (0.8±3.0)

1992 Lette43 360 12 171 22 155 0.5 (0.2±1.0)

1992 Pasternack54 385 11 197 8 169 1.2 (0.5±3.0)

1993 Ashton3 835 7 368 8 452 1.1 (0.4±3.0)

1993 Eichelberger18 75 3 53 2 17 0.5 (0.1±3.10)

1994 Poldermans58 131 4 49 11 67 0.5 (0.2±1.7)

1994 Baron5 457 50 168 36 203 1.7 (1.0±2.7)

1995 Gillespie26 213 17 128 5 63 1.7 (0.5±4.7)

1995 Koutelou40 106 4 51 1 50 3.9 (0.4±36.3)

1995 Ombrellaro53 266 25 176 13 52 0.6 (0.3±1.3)

1995 Sicari70 136 6 57 3 65 2.3 (0.6±9.5)

1996 Varma78 108 23 76 3 6 0.6 (0.1±2.6)

1996 Kontos39 87 5 48 9 25 0.3 (0.1±0.9)

1996 Stratmann77 140 7 74 4 55 1.3 (0.4±4.7)

1997 Frank22 300 11 199 1 89 4.9 (0.7±38.7)

1998 Badner4 323 13 169 5 136 2.1 (0.7±6.2)

1998 Howell34 230 54 31 61 84 2.4 (1.3±4.7)

1999 Howell33 146 16 19 57 54 1.3 (0.6±2.8)

1999 Heiba30 101 8 41 9 43 0.9 (0.3±2.6)

2000 Sprung72 214 86 84 21 23 1.1 (0.6±2.1)

2001 Boersma6 1320 23 560 22 715 1.3 (0.7±2.4)

Totals 13 671 565 5012 567 7532 1.3 (1.1±1.5)

Howell et al.

574

Page 28: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

grouped together as one outcome. Separate information was

not given on the association between hypertension and

perioperative complications. One study included data on

676 operations in 617 patients. No information was given on

which patients underwent more than one procedure. It was

felt that using data from this study could lead to an

underestimate of the variance of the odds ratio for hyper-

tension and the study was excluded from the meta-analysis.

(The full list of excluded studies can be viewed in the

electronic version of this paper.)

Initially, it was planned to restrict this review and meta-

analysis to patients undergoing general anaesthesia. It

rapidly became clear that this was not practical. In those

papers containing useful data that also gave information

on the type of anaesthesia used a signi®cant proportion

of patients received regional or local anaesthe-

sia.3 5 26 53 64 67 69 75 78 81 Many papers, that did not indicate

the type of anaesthesia used, included patients some of

whom were likely to have been managed with local regional

anaesthesia, for example those undergoing carotid endarter-

ectomy or lower limb revascularization.

Thirty studies were included in the ®nal meta-analysis

(Table 2).3±6 16 18 21 22 26 30 33 34 39±41 43 44 53 54 58 64 67 69 70 72

75 77 78 81 These studies were published between 1978 and

2001 and include 12 995 patients. The analysis included two

separate studies by Rao and colleagues, both described in

the same paper.64 One was a retrospective study of 364

patients anaesthetized between June 1973 and June 1976,

and the other a prospective study of 733 patients

anaesthetized between July 1977 and June 1982.

A ®xed effects meta-analysis of the crude odds ratios

from these studies for the association between hypertension

and cardiovascular complications was performed using the

`meta' command of Stata v7.0. The Forrest plot of the data

is shown in Figure 3. The pooled odds ratio from this

analysis was 1.35 (1.17±1.56) P<0.001. However, the test

for heterogeneity also achieved statistical signi®cance

(Q=44.76, 29 df, P=0.031). (Heterogeneity represents the

extent or magnitude of differences in treatment or exposure

effects between different studies.)52 The source of this

heterogeneity was sought through a number of sensitivity

analyses grouping the data by year of study (for example

1978±1990 and 1991±2001), and by type of surgery. These

analyses yielded little impact on the odds ratio and there

remained considerable heterogeneity within the subgroups

studied. Thus, the odds ratio of 1.31, while statistically

signi®cant, is small and must be interpreted with consider-

able caution in this meta-analysis of heterogeneous

observational studies, with no correction for confounding.

In the context of a low perioperative event rate, this small

odds ratio probably represents a clinically insigni®cant

association between pre-existing hypertension and peri-

operative cardiac risk.

Pre-existing hypertension and target organdamage

The association between hypertension and end or target

organ damage is well established. Hypertension is inextric-

ably linked to ischaemic heart disease and heart failure, to

Fig 3 Forrest plot for a meta-analysis of the risk of perioperative cardiovascular complications in hypertensive and normotensive patients. The x-axis

is on a logarithmic scale. The solid vertical line lies at unity, the dotted vertical line at the combined estimate. For each study, the point estimate of

the odds ratio is indicated by the box; the size of the box indicates the size of the study and the weight given to the study. The error bars indicate the

95% con®dence intervals for each point estimate.

Hypertension and perioperative risk

575

Page 29: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

cerebrovascular disease and to renal impairment. Both the

British Hypertension Society Guidelines and the World

Health Organisation-International Society of Hypertension

Guidelines for the management of arterial hypertension

highlight the importance of co-existing disease and

cardiovascular risk factors in setting treatment thresh-

olds.12 63 In both sets of guidelines, the presence of target

organ damage and other cardiovascular risk factors lead to

lower treatment thresholds for raised arterial pressure.

Similar considerations apply in the perioperative setting.

Many reviews have highlighted the associations between

the sequelae of hypertension, such as heart failure and

ischaemic heart disease, and perioperative complica-

tions.9 15 The meta-analysis of observational studies de-

scribed above suggested an association between a diagnosis

of hypertension and increased perioperative cardiac risk.

However, the odds ratio was small and the conclusion must

be treated with some circumspection in the light of

heterogeneity of the studies examined. This is not to

dismiss the role of clinically evident target organ damage in

increasing perioperative risk. A recent study by Lee and

colleagues identi®ed ischaemic heart disease, heart failure,

and renal failure as risk factors for perioperative cardiac

complications.42 In assessing perioperative risk of major

cardiovascular complications, pre-existing hypertension per

se may be of limited importance, but this does not grant the

anaesthetist a licence to ignore the target organ damage

caused by hypertension. Such damage may carry signi®cant

risk and its importance should be assessed using guidelines

such as those referenced above.9 15

Admission arterial pressure andperioperative cardiac risk

The best-designed study in this area is that of Goldman and

Caldera.27 This examined a sub-population of patients who

were studied in the production of the Goldman Risk Index.

Patients were divided into ®ve groups. These were:

normotensive patients, patients treated with diuretics,

treated hypertensives whose arterial pressure was con-

trolled, patients who were hypertensive despite treatment,

and untreated hypertensives. No signi®cant differences in

perioperative cardiac risk were found between the hyper-

tensive patients and the remaining groups. However,

although extremely well designed, this study lacked the

statistical power to either con®rm or refute an association

between hypertensive heart disease and perioperative car-

diac risk. Other studies, such as those by Cooperman,

Eerola, and Steen examined admission arterial pressure as

one of a number of variables that may contribute to

perioperative cardiac risk. However, these studies either

lacked the statistical power to effectively examine hyper-

tension as a risk factor, or gave only limited information on

the impact of hypertension in the ®nal report of the

study.13 17 75

None of the studies described above examined admission

arterial pressure as a continuous variable. All have taken a

speci®c cut-off for arterial pressure. The only studies of

which the authors are aware that have examined arterial

pressure as a continuous variable are those by Howell and

colleagues.33 34 The ®rst was a retrospective case controlled

study which examined patients who died of a cardiac cause

within 30 days of anaesthesia and elective surgery and a

matched controlled population who underwent the same

operations but who did not die.34 There were no signi®cant

differences between admission systolic and diastolic pres-

sures between the cases and the controls (Fig. 4). The second

was a similar study of emergency surgery; again there were

no signi®cant differences between the arterial pressures of

the cases and the controls, although in this case there was a

tendency for the survivors to have higher admission arterial

pressure than those patients who died.33 While both of these

studies suggest that there is no association between admis-

sion arterial pressure and perioperative cardiac risk, they are

both limited by the fact that most of the patients studied had

Stage 1 or Stage 2 hypertension. Few patients with Stage 3

hypertension were studied.

There is very little evidence on which to base the

perioperative management of patients who present for

surgery with admission arterial pressures consistent with

Stage 3 hypertension. Perhaps the best data come from the

original study by Prys-Roberts and colleagues.62 In this

Fig 4 Box and whisker plot of admission systolic and diastolic pressures

of cases and controls undergoing elective surgery. The boxes indicate the

median values and 25th and 75th centiles. The upper whisker is given by

the data point closest to (75th percentile+1.5 (interquartile range)). The

lower whisker is given by the data point closest to (25th percentile ± 1.5

(interquartile range)). The cases are patients who died of a cardiovascular

cause within 30 days of anaesthesia and surgery; the controls are patients

matched for type of surgery, age, and sex who did not die of a

cardiovascular cause in the perioperative period. There were no

signi®cant differences between the admission systolic or diastolic arterial

pressures of the cases and the controls. (Reproduced with permission

from reference 34.)

Howell et al.

576

Page 30: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

study most, if not all, of the hypertensive patients had

arterial pressures consistent with Stage 3 hypertension. As

already discussed, this study demonstrated increased

cardiovascular lability and an increased risk of perioperative

myocardial ischaemia in patients with poorly controlled

hypertension. It was too small to determine if there was an

increased incidence of cardiac events in this population.

The evidence from medical studies suggests that patients

with Stage 3 hypertension are at signi®cantly increased risk

of target organ damage, whether or not this is clinically

evident. For example, Stamler and colleagues, and Liao and

colleagues demonstrated a steadily increasing incidence of

ECG abnormalities in this population.45 73 There is certainly

evidence to support a steadily increasing incidence of

postoperative myocardial ischaemia with increasing admis-

sion systolic arterial pressure.31 Many patients with admis-

sion arterial pressures consistent with Stage 3 hypertension

will have isolated systolic hypertension. There is evidence

from the Framingham population of signi®cantly increased

cardiovascular risk in this population. Recent analyses

suggest that systolic pressure and pulse pressure are more

reliable indicators of cardiovascular risk than diastolic

pressure.25 On the basis of these data, we suggest that it is

appropriate to defer anaesthesia and surgery where possible

in patients with admission arterial pressures consistent with

Stage 3 hypertension, especially if there is evidence of

target organ damage. However, it must be borne in mind

that this recommendation is made on the basis of evidence

of risk in medical patients rather than data on perioperative

risk. Studies of perioperative risk in patients with Stage 3

hypertension are required.

Isolated systolic hypertension

The steady increase in arterial pressure with age in the

Western population is well known. An analysis of data from

the Framingham population by Franklin and colleagues has

added detail to this picture.23 They describe a steady

increase in systolic arterial pressure starting in childhood

and continuing throughout adult life. In contrast, diastolic

pressure rises in early adult life and then stabilizes or

declines in the ®fth and sixth decade of life. There is a

steady rise in pulse pressure throughout adult life and the

rate of rise increases after the age of 50 yr. It is against this

background that the phenomenon of isolated systolic

hypertension has been recognized; that is the situation in

which the diastolic arterial pressure is normal, but the

systolic arterial pressure is elevated. As might be expected,

isolated systolic hypertension accounts for the majority of

hypertension in patients over 50 yr old. In the NHANES III

data, Franklin and colleagues found that 80% of the subjects

aged over 50 yr who had hypertension had isolated systolic

hypertension.24 As a corollary of this, pulse pressure had

been found to be strongly associated with cardiovascular

risk.25 The bene®ts of treating isolated systolic hypertension

are now clearly established.29

While the studies of hypertension undertaken by Prys-

Roberts and colleagues used the then standard de®nition of

hypertension of a diastolic arterial pressure of greater than

95 mm Hg, it is clear from their publications that many of

their patients had severe systolic hypertension.59±62 Most

later studies that have included an examination of the

association between admission arterial pressure and peri-

operative complications have focused on older patients: for

example, in the study by Cooperman and colleagues the

average age of the patients was 61 yr; in the study by Eerola

and colleagues, 69 of the 111 patients studied were over 60

yr old; and in the study of myocardial re-infarction by Steen

and colleagues, 361 of the 466 patients studied were aged 60

yr or over.13 17 75 It is likely that the majority of poorly

controlled hypertensives in these studies had isolated

systolic hypertension.

A recent study by Aronson and colleagues examined the

association between isolated systolic hypertension and

cardiovascular complications in patients undergoing cardiac

surgery and these data are worth rehearsing here. This was a

prospective study of over 2000 patients in 24 centres

undergoing elective cardiac surgery. Patients were classi®ed

as having normal preoperative arterial pressure, isolated

systolic hypertension (systolic arterial pressure greater than

140 mm Hg), diastolic hypertension (diastolic arterial

pressure greater than 90 mm Hg) or a combination of

these. After adjusting for other risk factors, isolated systolic

hypertension was associated with a small but statistically

signi®cant increase in the likelihood of perioperative

morbidity (odds ratio 1.3, 95% con®dence interval 1.1±

1.6).2 The mean systolic arterial pressure of the patients

with isolated systolic hypertension is not given, although as

the average age of the patients was 65 yr, it is tempting to

speculate that it was considerably greater than 140 mm Hg.

It is clear that many of the patients who present for

surgery and have arterial pressures consistent with Stage 3

hypertension will be elderly patients with isolated systolic

hypertension. There are few if any studies that explicitly

examine the impact of isolated systolic hypertension on

outcome from non-cardiac surgery and, as with Stage 3

hypertension, work in this area is required. However, the

®ndings of the study by Aronson and colleagues and the

work of Franklin and colleagues on the Framingham

population do little to reassure the anaesthetist.

White coat hypertension

So-called white coat hypertension is directly relevant to

anaesthetic practice. It is formally de®ned as a persistently

elevated clinic arterial pressure in combination with a

normal ambulatory arterial pressure.56 Various different

arterial pressure thresholds have been used to de®ne white

coat hypertension in different studies, leading to con¯icting

data as to the prognosis of this condition.36 Recently, criteria

have been agreed and are now widely accepted. These

de®ne white coat hypertension as an of®ce arterial pressure

Hypertension and perioperative risk

577

Page 31: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

of 140/90 mm Hg or greater in the presence of an average

daytime reading of less than 135/85 mm Hg.50 56 Pickering

and colleagues highlight the dif®culties in comparing

different studies in this area, but suggest that the majority

of the data indicate a benign prognosis for white coat

hypertension.56 Kaplan identi®es four prospective studies of

the prognosis of hypertension that support this

view.36 38 55 65 80 The largest of these, by Verdecchia and

colleagues, followed up 1522 hypertensive subjects for 10

yr. Using a conservative de®nition of white coat hyperten-

sion, based on an ambulatory arterial pressure of less than

130/90 mm Hg, this study found the rate of cardiovascular

events in white coat hypertension (0.67/100 patient yr) to be

little different to that seen in normotensives.80 It should be

noted that when more liberal de®nitions of white coat

hypertension, with a higher ambulatory arterial pressure,

were used, the cardiovascular event rate in patients with

white coat hypertension was close to that of `true'

hypertensives. These data suggest that patients who present

for elective surgery with admission arterial pressures

consistent with Stage 3 hypertension that then settle to

levels consistent with normotension may be at less risk of

cardiovascular complications than patients with sustained

hypertension. However, extrapolating from data derived

from a long-term study conducted in the medical setting to

draw conclusions about patients undergoing surgery is a

leap, and any conclusions drawn have to be treated with

some caution.

The various guidelines on the management of hyperten-

sion all indicate that the arterial pressure should be measured

on a number of occasions over a period of weeks before the

diagnosis of hypertension is con®rmed. It is rare for the

anaesthetist to have this luxury and often a decision has to be

made on perioperative management on the basis of two or

three readings taken over a period of hours.

Both doctors and nurses may produce an initial elevation

in arterial pressure when they visit a patient, but the effect is

greater for doctors than for nurses. This is impressively

illustrated by data from Mancia and colleagues. They

studied 30 subjects who underwent a 24-h intra-arterial

recording after 5±7 days in hospital. During the intra-arterial

recording period the arterial pressure was additionally

measured at different times using a sphygmomanometer by

a male doctor and a female nurse, half of the subjects being

randomized to see the doctor ®rst, and the other half the

nurse. When the doctor took the ®rst reading, the arterial

pressure rose by an average of 22/14 mm Hg. The rises

when the ®rst arterial pressure was taken by a nurse were

only half as great. The arterial pressure usually returned to

near baseline after 10 min when the reading was taken by a

nurse, but this was not the case when the pressure was taken

by a doctor (Fig. 5).47 It is clear from their data that, in many

surgical patients, the admission arterial pressure will not

equate to the patient's usual arterial pressure. If a member of

the medical staff ®nds the patient's arterial pressure to be

elevated, this should be con®rmed by a nurse with

appropriate training.

Cardiovascular lability

Patients diagnosed as `hypertensive' have a reputation for

displaying increased cardiovascular lability during anaes-

thesia. There is certainly a pathophysiological basis for such

behaviour. Established hypertension is associated with an

increased systemic vascular resistance.36 The systemic

vasodilatation associated with anaesthesia might well be

expected to have profound effects on arterial pressure in

such patients. Prys-Roberts and colleagues, and Goldman

and Caldera both demonstrated that induction of anaesthesia

is associated with a decrease in arterial pressure to a similar

nadir in both hypertensive and normotensive patients.27 62

However, because hypertensive patients in these studies

generally had a higher pre-induction arterial pressure,

absolute decrease in arterial pressure in these patients was

greater. For many anaesthetists, however, cardiovascular

lability implies something more than the decrease in arterial

pressure seen at induction of anaesthesia. It suggests swings

in arterial pressure over a wide range of values, graphically

described by Longnecker as `Alpine Anesthesia'.46 Prys-

Fig 5 Comparison of systolic arterial pressure changes in 30 subjects (10

normotensive and 20 hypertensive) when subjects were visited by doctor

(solid line) and a nurse (dashed line) and three arterial pressure readings

taken with a sphygmomanometer at the 2nd, 5th, and 8th minute of the

visit. All arterial pressures values shown in the graph were obtained from

a transducer attached to an intra-arterial cannula. Values were recorded

by a mini-tape recorder and were not visible to the visiting doctor or

nurse. Baseline arterial pressures were obtained from the intra-arterial

cannula readings taken 4 min before the visit of the doctor or nurse. The

y-axis shows the peak deviation from this baseline arterial pressure and

the arterial pressures 5 and 10 min later. Values shown are mean (SEM).47

(Modi®ed with permission from reference 47.)

Howell et al.

578

Page 32: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Roberts and colleagues established that poorly controlled

hypertensive patients have a more vigorous cardiovascular

response to laryngoscopy and intubation than do normoten-

sives or well controlled hypertensives.61 Studies by

Charleson and colleagues documented swings in arterial

pressure in hypertensive and diabetic patients.7 8 However,

their work makes no comparison between hypertensive and

normotensive patients in respect to their cardiovascular

behaviour, and neither con®rms nor refutes the suggestion

that hypertensives are particularly prone to cardiovascular

instability. Chung and colleagues examined the association

between pre-existing medical conditions and adverse events

in 17 638 patients undergoing day-case surgery.11 They

identi®ed an association between pre-existing hypertension

and intraoperative cardiovascular events. No major compli-

cations, such as death or perioperative infarction, are

reported and the majority of these cardiovascular events

were episodes of hypertension, although there were also

instances of hypotension and arrhythmia. In the large

`Multicenter Study of General Anesthesia', it was noted that

hypertensive patients were more likely to require interven-

tions for perioperative hypertension.19 However, the de®n-

ition of hypertension used in this study was not given in the

report. Taken together, the weight of the evidence is that

patients with hypertension may be expected to suffer a

greater decrease in arterial pressure at induction of anaes-

thesia than normotensive patients, although the arterial

pressure probably decreases to a similar nadir in both patient

groups. The work of Prys-Roberts and colleagues supports a

more vigorous response to noxious stimuli in these patients.

The ®ndings of Chung and colleagues indicate that patients

with pre-existing hypertension frequently have high arterial

pressures during the intraoperative period.11

The clinical impact of wide variations in arterial pressure

is dif®cult to quantify, not least because most anaesthetists

would not be prepared to leave large changes in arterial

pressure untreated for more than a short period of time.

Charleson and colleagues reported that, within a high-risk

group of hypertensive patients and diabetic patients under-

going elective non-cardiac surgery, those with more than 1 h

of a decrease in mean arterial pressure of greater than/equal

to 20 mm Hg and those with less than 1 h of a decrease in

arterial pressure of greater than/equal to 20 mm Hg and

more than 15 min of an increase in arterial pressure of

greater than/equal to 20 mm Hg were at greatest risk of

complications.8 In so far as we can tell, the use of vasoactive

drugs was allowed in the perioperative period. One has to

ask if, in the patients who had wide excursions of arterial

pressure, a decision was made not to treat these changes in

arterial pressure or if the changes in arterial pressure were

refractory to treatment because of ongoing perioperative

cardiovascular complications. The association between

intraoperative myocardial ischaemia and haemodynamic

changes is certainly not clear-cut. In a study of 100 patients

who either had, or were at risk for, coronary artery disease,

intraoperative ischaemic episodes were preceded by acute

increases in arterial pressure in only 15% of episodes and by

acute decreases in only 8% of episodes.48 A recent paper by

Reich and colleagues has described an association between

intraoperative hypertension and tachycardia and adverse

outcome in protracted surgery.66 It was by no means clear,

however, that this was a causal association.

Perioperative management of patients withhypertension or raised arterial pressure

The meta-analysis presented above suggests association

between a diagnosis of hypertension and increased peri-

operative cardiac risk. However, the odds ratio for the effect

of hypertension was small and the conclusion must be

treated with some circumspection in the light of hetero-

geneity of the studies examined. There is evidence from

many studies that conditions that may represent target organ

damage as a result of hypertension contribute to periopera-

tive cardiac risk. A study by Lee and colleagues identi®ed

ischaemic heart disease, heart failure, and renal failure as

risk factors for perioperative cardiac complications.42 It

would seem sensible to suggest that anaesthetists should pay

more heed to the presence of signi®cant target organ

damage than to a diagnosis of hypertension per se.

With regard to the management of surgical patients with

elevated admission arterial pressure, there are few substan-

tive guidelines over which patients should be cancelled to

allow treatment before surgery or the duration of such

treatment before proceeding with surgery. The American

Heart Association/American College of Cardiology (ACC/

AHA) guidelines comment that hypertension (Stages 1 and

2) is not an independent risk factor for perioperative

cardiovascular complications.15 However, they suggest that

Stage 3 hypertension (SAP >180 mm Hg and/or DAP >110

mm Hg) should be controlled before surgery.15 To quote the

guidelines:

`In many instances establishment of an effective treat-

ment regimen can be achieved over several days to weeks of

preoperative outpatient management. If surgery is more

urgent, rapid acting agents can be administered to allow

effective control in a matter of minutes or hours. Beta-

blockers appear to be particularly attractive agents.

Continuation of preoperative antihypertensive treatment

through the perioperative period is critical.'

The observational data presented in this review support

the recommendations for Stages 1 and 2 hypertension. The

AHA/ACC recommendations for Stage 3 hypertension are

not supported by substantial data relating exclusively to

patients with arterial pressures greater than 180/110 mm Hg.

The best perioperative management of these patients

remains unclear. The options available to the anaesthetist

are: to ignore the elevated arterial pressure and to continue

with anaesthesia and surgery; to institute acute treatment to

control the arterial pressure; or to defer surgery for a period

of weeks to allow the arterial pressure to be controlled.

Hypertension and perioperative risk

579

Page 33: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

High arterial pressures are associated with high levels of

after load and cardiac work. This may predispose to

myocardial ischaemia and infarction, especially in the

presence of coronary artery disease and left ventricular

hypertrophy, and therefore simply ignoring markedly ele-

vated arterial pressure may not be appropriate. However,

there is evidence that very rapid control of arterial pressure

with drugs such as sublingual nifedipine is associated with

increased morbidity and mortality.79 Taken together, these

concerns pose the dilemma that markedly raised arterial

pressures and wide excursions of arterial pressure should be

avoided in the perioperative period, but that dramatic acute

reductions in arterial pressure may also be fraught with risk.

Observational data lend weight to these concerns. The

work of Charleson and colleagues suggests that excursions

in mean arterial pressure of greater than 20% in patients

with hypertension and or diabetes are associated with

perioperative complications.8 The work of Gould and

colleagues indicates that marked perioperative reductions

in arterial pressure may be associated with reduced

splanchnic blood ¯ow even in the `well ®lled' patient.28

The best course of action for the anaesthetist would seem to

be to defer surgery to allow the arterial pressure to be

treated. However, there are no trial data to suggest that this

strategy reduces perioperative risk and this advice takes no

account of the many issues and problems associated with

cancelling an operation within 24 h of surgery. Also, if

surgery is deferred to allow the arterial pressure to be

treated, it is unclear for how long treatment should be given

before the patient returns to have his or her operation.

Weksler and colleagues reported recently the results of a

clinical trial in which patients were brought to a waiting

room in the operating theatre suite, sedated with midazolam,

and had their diastolic pressures measured whilst awaiting

surgery.82 989 patients whose diastolic arterial pressure was

between 110 and 130 mm Hg immediately before surgery

were entered into the trial. 589 patients were randomized to

receive nifedipine 10 mg administered intranasally, while

400 patients were randomized to have their surgery

postponed. Those patients in whom surgery was deferred

remained in hospital until the diastolic arterial pressure was

below 110 mm Hg for at least 3 consecutive days. The

frequency of perioperative hypotension and hypertension

was similar in the two groups, as was the incidence of

tachyarrhythmias and bradyarrhythmias. There were no

neurological or cardiovascular complications in either

group. This study has a number of weaknesses. It was not

blinded, it ran over a 9-yr period, during which many other

aspects of patient management could have changed, and

systolic hypertension was not studied. However, it offers no

support for deferring anaesthesia and surgery to allow the

arterial pressure to be treated.

We suggest that, if the patient is considered ®t for surgery

in other respects, their operation should not be deferred

simply on account of an elevated admission arterial

pressure. If the arterial pressure is consistently elevated to

levels of 180 mm Hg systolic or greater or 110 mm Hg

diastolic or greater, surgery may proceed, but care should be

taken to ensure perioperative cardiovascular stability.

Invasive arterial pressure monitoring is indicated for

major procedures, and the arterial pressure should be

actively managed to prevent excursions of the mean arterial

pressure of greater than 20% from baseline. Monitoring

should continue into the postoperative period until it is clear

that the patient is cardiovascularly stable. It may be

appropriate to manage the patient in a high dependency

area in the immediate postoperative period. In those patients

in whom there is no contraindication, perioperative beta-

adrenergic block may be of value. These drugs are known to

reduce perioperative myocardial ischaemia and cardiovas-

cular complications in high-risk patients.49 57 They carry the

additional merit of not producing marked arterial pressure

reductions in normotensive subjects. It should be pointed

out that Boersma and colleagues have produced observa-

tional data that support the widespread use of perioperative

beta-adrenergic blockade, but that the available data from

randomized controlled trials only provide clear support for

their use in high-risk patients with demonstrable new wall

motion abnormalities on dobutamine stress echocardio-

graphy.6 32 Clinical trial data to support the use of

perioperative beta-adrenergic block in other patients with

cardiac disease are awaited. There may be a place for other

sympatholytic therapies such as alpha-2 agonists or thoracic

epidural block. The pharmacology and use of the alpha-2

agonists has been reviewed by Khan and colleagues.37 A

meta-analysis by Rogers and colleagues suggested that

neuroaxial block does offer protection from perioperative

myocardial injury.68 The validity of this ®nding has been

challenged and the current position remains unclear.1

Although Weksler and colleagues reported no problems

with intranasal nifedipine administered to 589 patients

immediately before surgery, we are unable to recommend

its use because of the concerns expressed by Varon and

colleagues.79 82

In making clinical judgements about perioperative man-

agement, white coat hypertension is an ever-present prob-

lem. If the preoperative arterial pressure is giving cause for

concern, several further readings should be obtained by

someone who is competent to do so. It seems indefensible to

defer planned surgery on the basis of a single arterial

pressure reading. In view of the vigorous alerting reaction

that can be produced by a visit from a doctor, readings

obtained by an experienced nurse may be invaluable. If at all

possible, the patient's family doctor should be contacted and

enquiry made about arterial pressure readings obtained in

the family practitioner's of®ce. It must be a source of

irritation for the patient and family doctor for surgery to be

deferred and the patient be sent back for treatment of their

arterial pressure when the they have been on carefully

monitored treatment for months or years and the arterial

pressure is known to be well controlled.

Howell et al.

580

Page 34: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Addendum

During the preparation of this review, there has been

published the Seventh Report of the Joint National

Committee on prevention, detection, evaluation and treat-

ment of high arterial pressure (2003). This recognizes that

the risk of cardiovascular disease begins at a pressure of

115/75 mm Hg and doubles with each increment of 20/10

mm Hg. Individuals with a systolic arterial pressure of 120±

139 mm Hg or diastolic arterial pressure of 80±89 mm Hg

should be considered as pre-hypertensive. The JNC VII

classi®es arterial pressure in adults as: normal: systolic

arterial pressure greater than 120 mm Hg and diastolic

arterial pressure less than 80 mm Hg; pre-hypertension:

systolic arterial pressure 120±139 mm Hg or diastolic

arterial pressure 80±89 mm Hg; Stage I hypertension:

systolic arterial pressure 140±159 mm Hg or diastolic

arterial pressure 90±99 mm Hg; Stage II hypertension:

systolic arterial pressure greater than 160 mm Hg or

diastolic arterial pressure greater than 100 mm Hg.

As in previous publications from the JNC, there are no

recommendations or guidelines for the perioperative care of

the hypertensive patient.10

Longer version of this paper

A longer version of this paper can be found in British

Journal of Anaesthesia on-line as supplementary data.

References1 Which anaesthetic technique? Bandolier 2001; 86: 5

2 Aronson S, Boisvert D, Lapp W. Isolated systolic hypertension isassociated with adverse outcomes from coronary artery bypassgrafting surgery. Anesth Analg 2002; 94: 1079±84

3 Ashton CM, Petersen NJ, Wray NP, et al. The incidence ofperioperative myocardial infarction in men undergoingnoncardiac surgery. Ann Intern Med 1993; 118: 504±10

4 Badner NH, Knill RL, Brown JE, Novick TV, Gelb AW.Myocardial infarction after noncardiac surgery. Anesthesiology1998; 88: 572±8

5 Baron JF, Mundler O, Bertrand M, et al. Dipyridamole-thalliumscintigraphy and gated radionuclide angiography to assess cardiacrisk before abdominal aortic surgery. N Engl J Med 1994; 330:663±9

6 Boersma E, Poldermans D, Bax JJ, et al. Predictors of cardiacevents after major vascular surgery: role of clinicalcharacteristics, dobutamine echocardiography, and beta-blocker therapy. JAMA 2001; 285: 1865±73

7 Charlson ME, MacKenzie CR, Gold JP, et al. The preoperativeand intraoperative hemodynamic predictors of postoperativemyocardial infarction or ischemia in patients undergoingnoncardiac surgery. Ann Surg 1989; 210: 637±48

8 Charlson ME, MacKenzie CR, Gold JP, Ales KL, Topkins M,Shires GT. Intraoperative blood pressure. What patterns identifypatients at risk for postoperative complications? Ann Surg 1990;212: 567±80

9 Chassot PG, Delabays A, Spahn DR. Preoperative evaluation ofpatients with, or at risk of, coronary artery disease undergoingnon-cardiac surgery. Br J Anaesth 2002; 89: 747±59

10 Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report ofthe Joint National Committee on Prevention, Detection,

Evaluation, and Treatment of High Blood Pressure: the JNC 7

report. JAMA 2003; 289: 2560±72

11 Chung F, Mezei G, Tong D. Pre-existing medical conditions as

predictors of adverse events in day-case surgery. Br J Anaesth

1999; 83: 262±70

12 Committee of World Health Organisation-International Society

of Hypertension. 1999 World Health Organization-International

Society of Hypertension (WHO-ISH). Guidelines for the

Management of Hypertension. J Hypertens 1999; 17: 151±83

13 Cooperman M, P¯ug B, Martin EW, jr, Evans WE. Cardiovascular

risk factors in patients with peripheral vascular disease. Surgery

1978; 84: 505±9

14 Dingle HR. Antihypertensive drugs and anaesthesia. Anaesthesia

1966; 21: 151±72

15 Eagle KA, Berger PB, Calkins H, et al. ACC/AHA guideline update

for perioperative cardiovascular evaluation for noncardiacsurgery. A report of the American College of Cardiology/

American Heart Association Task Force on Practice Guidelines

(Committee to Update the 1996 Guidelines on Perioperative

Cardiovascular Evaluation for Noncardiac Surgery). 2002.

American College of Cardiology Web site. Available at http./

www.acc.org/clinical/guidelines/perio/driIndex.htm

16 Eagle KA, Coley CM, Newell JB, et al. Combining clinical and

thallium data optimizes preoperative assessment of cardiac risk

before major vascular surgery. Ann Intern Med 1989; 110: 859±66

17 Eerola M, Eerola R, Kaukinen S, Kaukinen L. Risk factors in

surgical patients with veri®ed preoperative myocardial infarction.

Acta Anesthesiol Scand 1980; 24: 219±23

18 Eichelberger JP, Schwarz KQ, Black ER, Green RM, Ouriel K.

Predictive value of dobutamine echocardiography just before

noncardiac vascular surgery. Am J Cardiol 1993; 72: 602±7

19 Forrest JB, Rehder K, Cahalan MK, Goldsmith CH. Multicenter

study of general anesthesia. III. Predictors of severe

perioperative adverse outcomes. Anesthesiology 1992; 76: 3±15

20 Forrest JB, Rehder K, Goldsmith CH, et al. Multicenter study of

general anesthesia. I. Design and patient demography.

Anesthesiology 1990; 72: 252±61

21 Foster ED, Davis KB, Carpenter JA, Abele S, Fray D. Risk of

noncardiac operation in patients with de®ned coronary disease:

the Coronary Artery Surgery Study (CASS) registry experience.

Ann Thorac Surg 1986; 41: 42±50

22 Frank SM, Fleisher LA, Breslow MJ, et al. Perioperative

maintenance of normothermia reduces the incidence of

morbid cardiac events. A randomized clinical trial. JAMA 1997;

277: 1127±34

23 Franklin SS, Gustin W 4th, Wong ND, et al. Hemodynamic

patterns of age-related changes in blood pressure. The

Framingham Heart Study. Circulation 1997; 96: 308±15

24 Franklin SS, Jacobs MJ, Wong ND, L'Italien GJ, Lapuerta P.

Predominance of isolated systolic hypertension among middle-

aged and elderly US hypertensives: analysis based on National

Health and Nutrition Examination Survey (NHANES) III.Hypertension 2001; 37: 869±74

25 Franklin SS, Khan SA, Wong ND, Larson MG, Levy D. Is pulse

pressure useful in predicting risk for coronary heart disease? The

Framingham heart study. Circulation 1999; 100: 354±60

26 Gillespie DL, LaMorte WW, Josephs LG, Schneider T, Floch NR,

Menzoian JO. Characteristics of patients at risk for perioperative

myocardial infarction after infrainguinal bypass surgery: an

exploratory study. Ann Vasc Surg 1995; 9: 155±62

27 Goldman L, Caldera DL. Risks of general anesthesia and elective

Hypertension and perioperative risk

581

Page 35: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

operation in the hypertensive patient. Anesthesiology 1979; 50:285±92

28 Gould TH, Grace K, Thorne G, Thomas M. Effect of thoracicepidural anaesthesia on colonic blood ¯ow. Br J Anaesth 2002; 89:446±51

29 Group for Collaborative Research: Systolic Hypertension in theElderly Program. Prevention of stroke by antihypertensive drugtreatment in older persons with isolated systolic hypertension.Final results of the Systolic Hypertension in the Elderly Program(SHEP). JAMA 1991; 265: 3255±64

30 Heiba SI, Jacobson AF, Shattuc S, Ferreira MJ, Sharma PN,Cerqueira MD. The additive values of left ventricular functionand extent of myocardium at risk to dipyridamole perfusionimaging for optimal risk strati®cation prior to vascular surgery.Nuclear Med Commun 1999; 20: 887±94

31 Howell SJ, Hemming AE, Allman KG, Glover L, Sear JW, Foex P.Predictors of postoperative myocardial ischaemia. The role ofintercurrent arterial hypertension and other cardiovascular riskfactors. Anaesthesia 1997; 52: 107±11

32 Howell SJ, Sear JW, Foex P. Peri-operative beta-blockade: auseful treatment that should be greeted with cautiousenthusiasm. Br J Anaesth 2001; 86: 161±4

33 Howell SJ, Sear JW, Sear YM, Yeates D, Goldacre M, Foex P.Risk factors for cardiovascular death within 30 days afteranaesthesia and urgent or emergency surgery: a nested case-control study. Br J Anaesth 1999; 82: 679±84

34 Howell SJ, Sear YM, Yeates D, Goldacre M, Sear JW, Foex P.Risk factors for cardiovascular death after elective surgery undergeneral anaesthesia. Br J Anaesth 1998; 80: 14±9

35 Joint National Committee on prevention detection, evaluation,and treatment of high blood pressure. The sixth report of theJoint National Committee on prevention, detection, evaluation,and treatment of high blood pressure. Arch Intern Med 1997; 157:2413±46

36 Kaplan NM. Clinical Hypertension, 8th edn. Philadelphia: LippincottWilliams and Wilkins, 2002; 35±36, 92±96

37 Khan ZP, Ferguson CN, Jones RM. Alpha-2 and imidazolinereceptor agonists. Their pharmacology and therapeutic role.Anaesthesia 1999; 54: 146±65

38 Khattar RS, Senior R, Lahiri A. Cardiovascular outcome in white-coat versus sustained mild hypertension: a 10-year follow-upstudy. Circulation 1998; 98: 1892±7

39 Kontos MC, Brath LK, Akosah KO, Mohanty PK. Cardiaccomplications in noncardiac surgery: relative value of restingtwo-dimensional echocardiography and dipyridamole thalliumimaging. Am Heart J 1996; 132: 559±66

40 Koutelou MG, Asimacopoulos PJ, Mahmarian JJ, Kimball KT,Verani MS. Preoperative risk strati®cation by adenosine thallium201 single-photon emission computed tomography in patientsundergoing vascular surgery. J Nuclear Cardiol 1995; 2: 389±94

41 Larsen SF, Olesen KH, Jacobsen E, et al. Prediction of cardiac riskin non-cardiac surgery. Eur Heart J 1987; 8: 179±85

42 Lee TH, Marcantonio ER, Mangione CM, et al. Derivation andprospective validation of a simple index for prediction of cardiacrisk of major noncardiac surgery. Circulation 1999; 100: 1043±9

43 Lette J, Waters D, Bernier H, et al. Preoperative and long-termcardiac risk assessment. Predictive value of 23 clinical descriptors,7 multivariate scoring systems, and quantitative dipyridamoleimaging in 360 patients. Ann Surg 1992; 216: 192±204

44 Lette J, Waters D, Lassonde J, et al. Postoperative myocardialinfarction and cardiac death. Predictive value of dipyridamole-thallium imaging and ®ve clinical scoring systems based onmultifactorial analysis. Ann Surg 1990; 211: 84±90

45 Liao YL, Liu KA, Dyer A, et al. Major and minor electro-

cardiographic abnormalities and risk of death from coronaryheart disease, cardiovascular diseases and all causes in men and

women. J Am Coll Cardiol 1988; 12: 1494±500

46 Longnecker DE. Alpine anesthesia: can pretreatment with

clonidine decrease the peaks and valleys? Anesthesiology 1987;

67: 1±2

47 Mancia G, Parati G, Pomidossi G, Grassi G, Casadei R,

Zanchetti A. Alerting reaction and rise in blood pressure

during measurement by physician and nurse. Hypertension 1987;

9: 209±15

48 Mangano DT, Hollenberg M, Fegert G, et al. Perioperative

myocardial ischemia in patients undergoing noncardiac surgeryÐ

I: Incidence and severity during the 4 day perioperative period.

The Study of Perioperative Ischemia (SPI) Research Group. J Am

Coll Cardiol 1991; 17: 843±50

49 Mangano DT, Layug EL, Wallace A, Tateo I. Effect of atenolol on

mortality and cardiovascular morbidity after noncardiac surgery.

Multicenter Study of Perioperative Ischemia Research Group.

N Engl J Med 1996; 335: 1713±20

50 O'Brien E, Coats A, Owens P, et al. Use and interpretation of

ambulatory blood pressure monitoring: recommendations of the

British hypertension society. BMJ 2000; 320: 1128±34

51 O'Brien E, Staessen JA. Critical appraisal of the JNC VI, WHO/

ISH and BHS guidelines for essential hypertension. Expert Opin

Pharmacother 2000; 1: 675±82

52 O'Connell D, Glasziou P, Hill S, Sarunac J, Lowe J, Henry H.

Results of clinical trials and systematic reviews: to whom do they

apply? In: Stevens A, Abrams K, Brazier J, Fitzpatrick R, Lilford R,

eds. The Advanced Handbook of Methods in Evidence Based

Healthcare. London: Sage Publications, 2001; 56±72

53 Ombrellaro MP, Dieter RA, 3rd, Freeman M, Stevens SL,

Goldman MH. Role of dipyridamole myocardial scintigraphy in

carotid artery surgery. J Am Coll Surg 1995; 181: 451±8

54 Pasternack PF, Grossi EA, Baumann FG, et al. Silent myocardial

ischemia monitoring predicts late as well as perioperative cardiac

events in patients undergoing vascular surgery. J Vasc Surg 1992;16: 171±9

55 Perloff D, Sokolow M. Ambulatory blood pressure: mortality and

morbidity. J Hyperten Suppl 1991; 9: S31±3

56 Pickering TG, Coats A, Mallion JM, Mancia G, Verdecchia P.

Blood pressure monitoring. Task force V: white-coat hyper-

tension. Blood Press Monit 1999; 4: 333±41

57 Poldermans D, Boersma E, Bax JJ, et al. The effect of bisoprolol

on perioperative mortality and myocardial infarction in high-risk

patients undergoing vascular surgery. Dutch Echocardiographic

Cardiac Risk Evaluation Applying Stress Echocardiography Study

Group. N Engl J Med 1999; 341: 1789±94

58 Poldermans D, Fioretti PM, Boersma E, et al. Dobutamine-

atropine stress echocardiography in elderly patients unable to

perform an exercise test. Hemodynamic characteristics, safety,

and prognostic value. Arch Intern Med 1994; 154: 2681±6

59 Prys-Roberts C, Foex P, Biro GP, Roberts JG. Studies of

anaesthesia in relation to hypertension. V. Adrenergic beta-

receptor blockade. Br J Anaesth 1973; 45: 671±81

60 Prys-Roberts C, Foex P, Greene LT, Waterhouse TD. Studies of

anaesthesia in relation to hypertension. IV. The effects of

arti®cial ventilation on the circulation and pulmonary gas

exchanges. Br J Anaesth 1972; 44: 335±49

61 Prys-Roberts C, Greene LT, Meloche R, Foex P. Studies of

anaesthesia in relation to hypertension. II. Haemodynamic

consequences of induction and endotracheal intubation. Br J

Anaesth 1971; 43: 531±47

62 Prys-Roberts C, Meloche R, Foex P. Studies of anaesthesia in

Howell et al.

582

Page 36: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

relation to hypertension. I. Cardiovascular responses of treatedand untreated patients. Br J Anaesth 1971; 43: 122±37

63 Ramsay LE, Williams B, Johnston GD, et al. British HypertensionSociety guidelines for hypertension management 1999: summary.BMJ 1999; 319: 630±5

64 Rao TL, Jacobs KH, El Etr AA. Reinfarction following anesthesiain patients with myocardial infarction. Anesthesiology 1983; 59:499±505

65 Redon J, Campos C, Narciso ML, Rodicio JL, Pascual JM, RuilopeLM. Prognostic value of ambulatory blood pressure monitoringin refractory hypertension: a prospective study. Hypertension1998; 31: 712±8

66 Reich DL, Bennett-Guerrero E, Bodian CA, Hossain S,Winfree W, Krol M. Intraoperative tachycardia and hyper-tension are independently associated with adverse outcome innoncardiac surgery of long duration. Anesth Analg 2002; 95: 273±7

67 Riles TS, Kopelman I, Imparato AM. Myocardial infarctionfollowing carotid endarterectomy: a review of 683 operations.Surgery 1979; 85: 249±52

68 Rodgers A, Walker N, Schug S, et al. Reduction of postoperativemortality and morbidity with epidural or spinal anaesthesia:results from overview of randomised trials. BMJ 2000; 321: 1493

69 Shah KB, Kleinman BS, Rao TL, Jacobs HK, Mestan K,Schaafsma M. Angina and other risk factors in patients withcardiac diseases undergoing noncardiac operations. Anesth Analg1990; 70: 240±7

70 Sicari R, Picano E, Lusa AM, et al. The value of dipyridamoleechocardiography in risk strati®cation before vascular surgery. Amulticenter study. The EPIC (Echo Persantine InternationalStudy) Group±Subproject: risk strati®cation before majorvascular surgery. Eur Heart J 1995; 16: 842±7

71 Sprague HB. The heart in surgery. An analysis of the results ofsurgery on cardiac patients during the past ten years at theMassachusetts General Hospital. Surg Gynecol Obstet 1929; 49:54±8

72 Sprung J, Abdelmalak B, Gottlieb A, et al. Analysis of risk factors

for myocardial infarction and cardiac mortality after major

vascular surgery. Anesthesiology 2000; 93: 129±40

73 Stamler J, Dyer AR, Shekelle RB, Neaton J, Stamler R.

Relationship of baseline major risk factors to coronary and all-

cause mortality, and to longevity: ®ndings from long-term follow-

up of Chicago cohorts. Cardiology 1993; 82: 191±222

74 Stamler J, Stamler R, Neaton JD. Blood pressure, systolic and

diastolic, and cardiovascular risks. US population data. Arch Intern

Med 1993; 153: 598±615

75 Steen PA, Tinker JH, Tarhan S. Myocardial reinfarction after

anesthesia and surgery. JAMA 1978; 239: 2566±70

76 Stevens A, Abrams K. Consensus, reviews and meta-analysis. In:

Stevens A, Abrams K, Brazier J, Fitzpatrick R, Lilford R, eds.

Methods in Evidence Based Healthcare. London: Sage, 2001; 367±9

77 Stratmann HG, Younis LT, Wittry MD, Amato M, Mark AL,

Miller DD. Dipyridamole technetium 99m sestamibi myocardial

tomography for preoperative cardiac risk strati®cation before

major or minor nonvascular surgery. Am Heart J 1996; 132: 536±

41

78 Varma MK, Puri GD, Chari P, Verma JS, Kohli KK. Perioperative

myocardial infarction in coronary artery disease patients and `at-

risk' for coronary artery disease patients undergoing non-cardiac

surgery. Natl Med J India 1996; 9: 214±7

79 Varon J, Marik PE. The diagnosis and management of

hypertensive crises. Chest 2000; 118: 214±27

80 Verdecchia P, Schillaci G, Borgioni C, Ciucci A, Porcellati C.

White-coat hypertension. Lancet 1996; 348: 1444±5

81 vonKnorring J. Postoperative myocardial infarction: a pros-

pective study in a risk group of surgical patients. Surgery 1981;

90: 55±60

82 Weksler N, Klein M, Szendro G, et al. The dilemma of immediate

preoperative hypertension: to treat and operate, or to postpone

surgery? J Clin Anesth 2003; 15: 179±83

Hypertension and perioperative risk

583

Page 37: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Hypertensive Crises*Challenges and Management

Paul E. Marik, MD, FCCP; and Joseph Varon, MD, FCCP

Hypertension affects > 65 million people in the United States and is one of the leading causes ofdeath. One to two percent of patients with hypertension have acute elevations of BP that requireurgent medical treatment. Depending on the degree of BP elevation and presence of end-organdamage, severe hypertension can be defined as either a hypertensive emergency or a hyperten-sive urgency. A hypertensive emergency is associated with acute end-organ damage and requiresimmediate treatment with a titratable short-acting IV antihypertensive agent. Severe hyperten-sion without acute end-organ damage is referred to as a hypertensive urgency and is usuallytreated with oral antihypertensive agents. This article reviews definitions, current concepts,common misconceptions, and pitfalls in the diagnosis and management of patients with acutelyelevated BP as well as special clinical situations in which BP must be controlled.

(CHEST 2007; 131:1949–1962)

Key words: aortic dissection; �-blockers; calcium-channel blockers; clevidipine; eclampsia; fenoldopam; hypertension;hypertensive crises; hypertensive encephalopathy; labetalol; nicardipine; nitroprusside; pre-eclampsia; pregnancy

Abbreviations: ACE � angiotensin-converting enzyme; APH � acute postoperative hypertension; DBP � diastolicBP; FDA � Food and Drug Administration; JNC � Joint National Committee; MAP � mean arterial pressure;SBP � systolic BP

H ypertension is one of the most common chronicmedical conditions in the United States, affect-

ing close to 30% of the population � 20 years old.1While chronic hypertension is an established riskfactor for cardiovascular, cerebrovascular, and renaldisease, acute elevations in BP can result in acuteend-organ damage with significant morbidity. Hy-pertensive emergencies and hypertensive urgencies(see definitions below) are commonly encounteredby a wide variety of clinicians. Prompt recognition,evaluation, and appropriate treatment of these con-

ditions are crucial to prevent permanent end-organdamage. This article reviews our current understand-ing of hypertensive crises, the common misconcep-tions and pitfalls in its diagnosis and management, aswell as pharmacotherapy and special situations thatclinicians may encounter.

Definitions

The classification and approach to hypertensionundergoes periodic review by the Joint NationalCommittee (JNC) on Prevention, Detection, Evalu-ation, and Treatment of High Blood Pressure, withthe most recent report (JNC 7) having been releasedin 2003 (Table 1).2 Although not specifically ad-dressed in the JNC 7 report, patients with a systolicBP (SBP) � 179 mm Hg or a diastolic BP (DBP)� 109 mm Hg are usually considered to be having a“hypertensive crisis.” The 1993 report3 of the JNCproposed an operational classification of hyperten-sive crisis as either “hypertensive emergencies” or“hypertensive urgencies.” This classification remainsuseful today. Severe elevations in BP were classified

*From the Department of Pulmonary and Critical Care (Dr.Marik), Thomas Jefferson University, Philadelphia, PA; andDepartment of Acute and Continuing Care (Dr. Varon), TheUniversity of Texas Health Science Center at Houston, Houston,TX.The authors have no conflicts of interest to disclose.Manuscript received October 11, 2006; revision accepted January23, 2007.Reproduction of this article is prohibited without written permissionfrom the American College of Chest Physicians (www.chestjournal.org/misc/reprints.shtml).Correspondence to: Paul E. Marik, MD, FCCP, 834 Walnut St,Suite 650, Philadelphia, PA 19107; e-mail: [email protected]: 10.1378/chest.06-2490

CHEST Postgraduate Education CornerCONTEMPORARY REVIEWS IN CRITICAL CARE MEDICINE

www.chestjournal.org CHEST / 131 / 6 / JUNE, 2007 1949

Page 38: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

as hypertensive emergencies in the presence of acuteend-organ damage, or as hypertensive urgencies inthe absence of acute target-organ involvement. Dis-tinguishing hypertensive urgencies from emergen-cies is important in formulating a therapeutic plan.Patients with hypertensive urgency should have theirBP reduced within 24 to 48 h, whereas patients withhypertensive emergency should have their BP low-ered immediately, although not to “normal” levels.The term malignant hypertension has been used todescribe a syndrome characterized by elevated BPaccompanied by encephalopathy or acute nephrop-athy.4 This term, however, has been removed fromNational and International Blood Pressure Controlguidelines and is best referred to as a hypertensiveemergency.

Epidemiology

Hypertensive emergencies were first described byVolhard and Fahr5 in 1914, who saw patients withsevere hypertension accompanied by signs of vascu-lar injury to the heart, brain, retina, and kidney. Thissyndrome had a rapidly fatal course, ending in heartattack, renal failure, or stroke. It was not, however,until 1939 when the first large study6 of the naturalhistory of hypertensive emergencies was published.The results of this seminal article by Keith andcolleagues6 revealed that untreated hypertensiveemergencies had a 1-year mortality rate of 79%, witha median survival of 10.5 months. Prior to theintroduction of antihypertensive medications, ap-proximately 7% of hypertensive patients had a hy-pertensive emergency.7 Currently, it is estimatedthat 1 to 2% of patients with hypertension will havea hypertensive emergency at some time in theirlife.8,9

In the United States, hypertensive emergenciescontinue to be quite common, and the epidemiologyof this disorder parallels the distribution of essentialhypertension, being higher among the elderly andAfrican Americans, with men being affected twotimes more frequently than women.10,11 Despite thedevelopment of increasingly effective antihyperten-sive treatments over the past 4 decades, the inci-dence of hypertensive emergencies has increased.12

The vast majority of patients presenting with ahypertensive emergency to an emergency depart-ment have a previous diagnosis of hypertension andhave been prescribed antihypertensive agents.10,13

However, in many of these patients BP control priorto presentation was inadequate.13 The lack of aprimary care physician, as well as the failure toadhere to prescribed antihypertensive regimens havebeen associated with the development of a hyperten-sive emergency.14,15 In some studies,15 � 50% ofpatients presenting to an emergency departmentwith a hypertensive emergency were not adherentwith their antihypertensive medication regimen inthe preceding week. In both major metropolitanareas and smaller communities, illicit drug use hasbeen reported14 to be a major risk factor for thedevelopment of hypertensive emergency.

Pathophysiology

Acute severe hypertension can develop de novo orcan complicate underlying essential or secondaryhypertension. The factors leading to the severe andrapid elevation of BP in patients with hypertensivecrises are poorly understood. The rapidity of onsetsuggests a triggering factor superimposed on preex-isting hypertension. Hypertensive crisis is thought tobe initiated by an abrupt increase in systemic vascu-lar resistance likely related to humoral vasoconstric-tors.16,17 The subsequent increase in BP generatesmechanical stress and endothelial injury leading toincreased permeability, activation of the coagulationcascade and platelets, and deposition of fibrin. Withsevere elevations of BP, endothelial injury and fi-brinoid necrosis of the arterioles ensue.16,17 Thisprocess results in ischemia and the release of addi-tional vasoactive mediators generating a vicious cycleof ongoing injury. The renin-angiotensin system isoften activated, leading to further vasoconstrictionand the production of proinflammatory cytokinessuch as interleukin-6.18,19 The volume depletion thatresults from pressure natriuresis further simulatesthe release of vasoconstrictor substances from thekidney. These collective mechanisms can culminatein end-organ hypoperfusion, ischemia and dysfunc-tion that manifests as a hypertensive emergency.

Clinical Presentation

Most patients have persistent BP elevation foryears before they manifest a hypertensive emer-gency. The clinical manifestations of hypertensiveemergency are directly related to the particularend-organ dysfunction that has occurred (Table 2).

Table 1—JNC 7 BP Categorization*

BP Class SBP, mm Hg DBP, mm Hg

Normal � 120 � 80Prehypertension 121–139 80–89Stage I 140–159 90–99Stage II � 160 � 100

*From Chobanian et al.2

1950 Postgraduate Education Corner

Page 39: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

The signs and symptoms therefore vary from patientto patient. Zampaglione and colleagues20 reportedthat the most frequent presenting signs in patientswith hypertensive emergencies were chest pain(27%), dyspnea (22%), and neurologic deficits(21%). No particular BP threshold has been associ-ated with the development of a hypertensive emer-gency. However, organ dysfunction is uncommonwith a DBP � 130 mm Hg (except in children andpregnancy).21 The absolute level of BP may not be asimportant as the rate of increase. For example, inpatients with long-standing hypertension, a SBP of200 mm Hg or a DBP up to 150 mm Hg may be welltolerated without the development of hypertensiveencephalopathy; whereas in children and pregnantwomen, encephalopathy may develop with a DBP ofonly 100 mm Hg.22

Initial Evaluation

Patients with hypertensive emergency usuallypresent for evaluation as a result of a new symptomcomplex related to their elevated BP. Patient triageand physician evaluation should proceed expedi-tiously to prevent ongoing end-organ damage. Afocused medical history that includes the use of anyprescribed or over-the-counter medications shouldbe obtained. If the patient is known to have hyper-tension, their hypertensive history, previous control,current antihypertensive medications with dosing,adherence with their medication regimen, and thetime from last dose are important facts to acquireprior to initiating treatment. Inquiry into the use ofrecreational drugs (amphetamines, cocaine, phen-cyclidine) or monoamine oxidase inhibitors shouldbe made. Confirmation of the BP should be obtainedby a physician in both arms using an appropriate-sizeBP cuff. The appropriate-size cuff is particularlyimportant because the use of a cuff too small for thearm has been shown to artificially elevate BP read-ings in obese patients.23,24

The physical examination should attempt to iden-tify evidence of end-organ damage by assessing

pulses in all extremities, auscultating the lungs forevidence of pulmonary edema, the heart for mur-murs or gallops, the renal arteries for bruits, andperforming a focused neurologic and fundoscopicexamination. Headache and altered level of con-sciousness are the usual manifestations of hyperten-sive encephalopathy.25,26 Focal neurologic findings,especially lateralizing signs, are uncommon in hyper-tensive encephalopathy, being more suggestive of acerebrovascular accident. Subarachnoid hemorrhageshould be considered in patients with a sudden onsetof a severe headache. The ocular examination mayshow evidence of advanced retinopathy with arterio-lar changes, exudates, hemorrhages, or papilledemaassisting in the identification of hypertensive en-cephalopathy. Cardiac evaluation should aim to iden-tify angina or myocardial infarction with the focus onclarifying any atypical symptoms such as dyspnea,cough, or fatigue that may be overlooked.10,27 On thebasis of this evaluation, the clinician should be ableto distinguish between a hypertensive emergency oran urgency and to formulate the subsequent plan forfurther diagnostic tests and treatment.

If the clinical picture is consistent with aorticdissection (severe chest pain, unequal pulses, wid-ened mediastinum), a contrast CT scan or MRI ofthe chest should be obtained promptly to rule outaortic dissection. Although transesophageal echocar-diography has excellent sensitivity and specificity foraortic dissection, this study should not be performeduntil adequate blood control has been achieved. Inpatients presenting with pulmonary edema, it isimportant to obtain an echocardiogram to distinguishbetween diastolic dysfunction, transient systolic dys-function, or mitral regurgitation.28 Many patients,particularly the elderly, have a normal ejection frac-tion, and in such patients heart failure is due toisolated diastolic dysfunction.28 The managementthese patients differs from those patients with pre-dominant systolic dysfunction and those with tran-sient mitral regurgitation (Table 3).

Initial Management of BP

The majority of patients in whom severe hyper-tension (SBP � 160 mm Hg, DBP � 110 mm Hg) isidentified on initial evaluation will have no evidenceof end-organ damage and thus have a hypertensiveurgency. Since no acute end-organ damage ispresent, these patients may present for evaluation ofanother complaint, and the elevated BP may repre-sent an acute recognition of chronic hypertension. Inthese patients, utilizing oral medications to lower theBP gradually over 24 to 48 h is the best approach tomanagement. Rapid reduction of BP may be associ-ated with significant morbidity in hypertensive ur-

Table 2—Clinical Manifestation of HypertensiveEmergencies*

Hypertensive encephalopathyAcute aortic dissectionAcute myocardial infarctionAcute coronary syndromePulmonary edema with respiratory failureSevere pre-eclampsia, HELLP syndrome, eclampsiaAcute renal failureMicroangiopathic hemolytic anemiaAPH

*HELLP � hemolysis, elevated liver enzymes, low platelets.

www.chestjournal.org CHEST / 131 / 6 / JUNE, 2007 1951

Page 40: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

gency due to a rightward shift in the pressure/flowautoregulatory curve in critical arterial beds (cere-bral, coronary, renal).29 Rapid correction of severelyelevated BP below the autoregulatory range of thesevascular beds can result in marked reduction inperfusion causing ischemia and infarction. There-fore, although the BP must be reduced in thesepatients, it must be lowered in a slow and controlledfashion to prevent organ hypoperfusion.

Altered autoregulation also occurs in patients withhypertensive emergency, and since end-organ dam-age is present already, rapid and excessive correctionof the BP can further reduce perfusion and propa-gate further injury. Therefore, patients with a hyper-tensive emergency are best managed with a contin-uous infusion of a short-acting, titratableantihypertensive agent. Due to unpredictable phar-macodynamics, the sublingual and IM route shouldbe avoided. Patients with a hypertensive emergencyshould be managed in an ICU with close monitoring.For those patients with the most severe clinicalmanifestations or with the most labile BP, intra-arterial BP monitoring may be prudent. There are avariety of rapid-acting IV agents that are available foruse in patients with hypertensive emergency, and theagent of choice depends on which manifestation ofend-organ damage is present and the available mon-itored setting (Table 3). Rapid-acting IV agentsshould not be used outside of an ICUs monitoredsetting to prevent precipitous falls of BP that mayhave significant morbidity or mortality. The imme-diate goal is to reduce DBP by 10 to 15% or toapproximately 110 mm Hg over a period of 30 to 60min. In patients with aortic dissection, the BP shouldbe reduced rapidly (within 5 to 10 min), targeting aSBP of � 120 mm Hg and mean arterial pressure(MAP) � 80 mm Hg.30,31 Once there is stable BPcontrol with IV agents and end-organ damage has

ceased, oral therapy can be initiated as the IV agentsare slowly titrated down. An important considerationprior to initiating IV therapy is to assess the patient’svolume status. Due to pressure natriuresis, patientswith hypertensive emergencies may be volume de-pleted, and restoration of intravascular volume withIV saline solution will serve to restore organ perfu-sion and prevent a precipitous fall in BP whenantihypertensive regimens are initiated.

Pharmacologic Agents Used in theTreatment of Hypertensive Emergencies

A number of drugs are available for the manage-ment of hypertensive emergency. The agent ofchoice in any particular situation will depend on theclinical presentation (Table 3). The preferred agentsinclude labetalol, esmolol, nicardipine, and fenoldo-pam. Phentolamine and trimethaphan camsylate areless commonly used today; however, they may beuseful in particular situations such as catecholamine-induced hypertensive crises (ie, pheochromocy-toma). Sodium nitroprusside may be used in patientswith acute pulmonary edema and/or severe leftventricular dysfunction and in patients with aorticdissection.32 Oral and sublingual nifedipine are po-tentially dangerous in patients with hypertensiveemergencies and are not recommend. Clonidine andangiotensin-converting enzyme (ACE) inhibitors arelong acting and poorly titratable; however, theseagents may be useful in the management of hyper-tensive urgencies. ACE inhibitors are contraindi-cated in pregnancy.33,34 Clevidipine is a relativelynew agent under investigation for the managementof postoperative hypertension and hypertensiveemergencies.35 At this time, clevidipine is not avail-able in the United States for use outside of clinical

Table 3—Recommended Antihypertensive Agents for Hypertensive Crises

Conditions Preferred Antihypertensive Agents

Acute pulmonary edema/systolicdysfunction

Nicardipine, fenoldopam, or nitroprusside in combination with nitroglycerin and a loop diuretic

Acute pulmonary edema/diastolicdysfunction

Esmolol, metoprolol, labetalol, or verapamil in combination with low-dose nitroglycerin and a loopdiuretic

Acute myocardial ischemia Labetalol or esmolol in combination with nitroglycerinHypertensive encephalopathy Nicardipine, labetalol, or fenoldopamAcute aortic dissection Labetalol or combination of nicardipine and esmolol or combination of nitroprusside with either

esmolol or IV metoprololPre-eclampsia, eclampsia Labetalol or nicardipineAcute renal failure/microangiopathic

anemiaNicardipine or fenoldopam

Sympathetic crisis/cocaine overdose Verapamil, diltiazem, or nicardipine in combination with a benzodiazepineAPH Esmolol, nicardipine, or labetalolAcute ischemic stroke/intracerebral

bleedNicardipine, labetalol, or fenoldopam

1952 Postgraduate Education Corner

Page 41: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

trials. The recommended IV antihypertensive agentsare reviewed briefly below. Dosage and adverseeffects of commonly used parenteral antihyperten-sive medications are listed in Table 4.

Labetalol

Labetalol is a combined selective �1-adrenergicand nonselective �-adrenergic receptor blocker withan �- to �-blocking ratio of 1:7.36 Labetalol ismetabolized by the liver to form an inactive glucuro-nide conjugate.37 The hypotensive effect of labetalolbegins within 2 to 5 min after its IV administration,reaching a peak at 5 to 15 min following administra-tion, and lasting for about 2 to 4 h.37,38 Due to its�-blocking effects, the heart rate is either main-tained or slightly reduced. Unlike pure �-adrenergicblocking agents that decrease cardiac output, labe-talol maintains cardiac output.39 Labetalol reducesthe systemic vascular resistance without reducingtotal peripheral blood flow. In addition, the cerebral,renal, and coronary blood flow are maintained.39–42

This agent has been used in the setting of pregnancy-induced hypertensive crisis because little placentaltransfer occurs mainly due to the negligible lipidsolubility of the drug.39

Labetalol may be administered as loading dose of20 mg, followed by repeated incremental doses of 20to 80 mg at 10-min intervals until the desired BP isachieved. Alternatively, after the initial loading dose,an infusion commencing at 1 to 2 mg/min andtitrated up to until the desired hypotensive effect isachieved is particularly effective. Bolus injections of1 to 2 mg/kg have been reported to produce precip-itous falls in BP and should therefore be avoided.43

Nicardipine

Nicardipine is a second-generation dihydropyri-dine derivative calcium-channel blocker with highvascular selectivity and strong cerebral and coronaryvasodilatory activity. The onset of action of IV nicar-dipine is from 5 to 15 min, with a duration of actionof 4 to 6 h. IV nicardipine has been shown to reduceboth cardiac and cerebral ischemia.44 The nicardi-pine dosage is independent of the patient’s weight,with an initial infusion rate of 5 mg/h, increasing by2.5 mg/h every 5 min to a maximum of 15 mg/h untilthe desired BP reduction is achieved.21 A usefultherapeutic benefit of nicardipine is that the agenthas been demonstrated to increase both stroke vol-ume and coronary blood flow with a favorable effecton myocardial oxygen balance.44–48 This property isuseful in patients with coronary artery disease andsystolic heart failure.

Esmolol

Esmolol is an ultrashort-acting cardioselective,�-adrenergic blocking agent.49–51 The onset of actionof this agent is within 60 s, with a duration of actionof 10 to 20 min.49–51 The metabolism of esmolol isvia rapid hydrolysis of ester linkages by RBC ester-ases and is not dependant on renal or hepaticfunction. Due to its pharmacokinetic properties,some authors21 consider it an “ideal �-adrenergicblocker” for use in critically ill patients. This agent isavailable for IV use both as a bolus and as aninfusion. Esmolol is particularly useful in severepostoperative hypertension.52–58 Esmolol is a suit-able agent in situations in which cardiac output,heart rate, and BP are increased. Typically, the drug

Table 4—Dosage and Adverse Effects of Commonly Used Parenteral Antihypertensive Medications

Agents Dosage Adverse Effects

Enalaprilat 1.25 mg over 5 min every 4 to 6 h, titrate by 1.25-mgincrements at 12- to 24-h intervals to maximum of5 mg q6h

Variable response, potential hypotension in high reninstates, headache, dizziness

Esmolol 500 �g/kg loading dose over 1 min, infusion at 25 to 50 �g/kg/min, increased by 25 �g/kg/min every 10 to 20 min tomaximum of 300 �g/kg/min

Nausea, flushing, first-degree heart block, infusion site pain

Fenoldopam 0.1 �g/kg/min initial dose, 0.05 to 0.1 �g/kg/min incrementsto maximum of 1.6 �g/kg/min

Nausea, headache, flushing

Labetalol 20-mg initial bolus, 20- to 80-mg repeat boluses or startinfusion at 2 mg/min with maximum 24-h dose of 300 mg

Hypotension, dizziness, nausea/vomiting, paresthesias, scalptingling, bronchospasm

Nicardipine 5 mg/h, increase at 2.5 mg/h increments every 5 min tomaximum of 15 mg/h

Headache, dizziness, flushing, nausea, edema, tachycardia

Nitroglycerin 5 �g/min, titrated by 5 �g/min every 5 to 10 min tomaximum of 60 �g/min

Headache, dizziness, tachyphylaxis

Nitroprusside 0.5 �g/kg/min, increase to maximum of 2 �g/kg/min toavoid toxicity

Thiocyanate and cyanide toxicity, headache,nausea/vomiting, muscle spasm, flushing

Phentolamine 1- to 5-mg boluses, maximum 15-mg dose Flushing, tachycardia, dizziness, nausea/vomiting

www.chestjournal.org CHEST / 131 / 6 / JUNE, 2007 1953

Page 42: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

is administered as a 0.5 to 1 mg/kg loading dose over1 min, followed by an infusion starting at 50 �g/kg/min and increasing up to 300 �g/kg/min as neces-sary.

Fenoldopam

Fenoldopam is unique among the parenteral BPagents because it mediates peripheral vasodilation byacting on peripheral dopamine-1 receptors.Fenoldopam is rapidly and extensively metabolizedby conjugation in the liver, without participation ofcytochrome P-450 enzymes. The onset of action iswithin 5 min, with the maximal response beingachieved by 15 min.59–61 The duration of action isfrom 30 to 60 min, with the pressure graduallyreturning to pretreatment values without reboundonce the infusion is stopped.59–61 No adverse effectshave been reported.59 An initial starting dose of 0.1�g/kg/min is recommended. Fenoldopam improvescreatinine clearance, urine flow rates, and sodiumexcretion in severely hypertensive patients with bothnormal and impaired renal function.62–64 The use offenoldopam as a prophylactic agent to prevent con-trast-induced nephropathy has been disappoint-ing.65,66

Nitroprusside

Sodium nitroprusside is an arterial and venousvasodilator that decreases both afterload and pre-load.67,68 Nitroprusside decreases cerebral bloodflow while increasing intracranial pressure, effectsthat are particularly disadvantageous in patients withhypertensive encephalopathy or following a cerebro-vascular accident.69–72 In patients with coronaryartery disease, a significant reduction in regionalblood flow (coronary steal) can occur.73 In a largerandomized, placebo-controlled trial,74 nitroprussidewas shown to increase mortality when infused in theearly hours after acute myocardial infarction (mor-tality at 13 weeks, 24.2% vs 12.7%). Nitroprusside isa very potent agent, with an onset of action ofseconds, a duration of action of 1 to 2 min, and aplasma half-life of 3 to 4 min.67 Due to its potency,rapidity of action, and the development of tachyphy-laxis, we recommend intraarterial BP monitoring. Inaddition, sodium nitroprusside requires special han-dling to prevent its degradation by light. Thesefactors limit the use of this drug.15

Nitroprusside contains 44% cyanide by weight.75

Cyanide is released nonenzymatically from nitro-prusside, the amount generated being dependent onthe dose of nitroprusside administered. Cyanide ismetabolized in the liver to thiocyanate.75 Thiosulfateis required for this reaction.75,76 Thiocyanate is 100times less toxic than cyanide. The thiocyanate gen-

erated is excreted largely through the kidneys. Cya-nide removal therefore requires adequate liver func-tion, adequate renal function, and adequatebioavailability of thiosulfate. Nitroprusside maytherefore cause cytotoxicity due to the release ofcyanide with interference of cellular respiration.77,78

Cyanide toxicity has been documented to result in“unexplained cardiac arrest,” coma, encephalopathy,convulsions, and irreversible focal neurologic abnor-malities.68,79 The current methods of monitoring forcyanide toxicity are insensitive. Metabolic acidosis isusually a preterminal event. In addition, a rise inserum thiocyanate levels is a late event and notdirectly related to cyanide toxicity. RBC cyanideconcentrations (although not widely available) maybe a more reliable method of monitoring for cyanidetoxicity.75 An RBC cyanide concentration � 40nmol/mL results in detectable metabolic changes.Levels � 200 nmol/L are associated with severeclinical symptoms, and levels � 400 nmol/mL areconsidered lethal.75 Data suggest that nitroprussideinfusion rates � 4 �g/kg/min, for as little as 2 to 3 hmay lead to cyanide levels in the toxic range.75 Therecommended doses of nitroprusside of up to 10�g/kg/min results in cyanide formation at a fargreater rate than human beings can detoxify. Sodiumnitroprusside has also been demonstrated to causecytotoxicity through the release of nitric oxide, withhydroxyl radical and peroxynitrite generation leadingto lipid peroxidation.77,80–82

Considering the potential for severe toxicity withnitroprusside, this drug should only be used whenother IV antihypertensive agents are not availableand then only in specific clinical circumstances andin patients with normal renal and hepatic function.68

The duration of treatment should be as short aspossible, and the infusion rate should not be � 2�g/kg/min. An infusion of thiosulfate should be usedin patients receiving higher dosages (4 to 10 �g/kg/min) of nitroprusside.76

Clevidipine

Clevidipine is third-generation dihydropyridinecalcium-channel blocker that has been developed foruse in clinical settings in which tight BP control iscrucial.83 Clevidipine is an ultrashort-acting selectivearteriolar vasodilator.84,85 Clevidipine acts by selec-tively inhibiting the influx of extracellular calciumthrough the L-type channel, relaxing smooth muscleof small arteries, and reducing peripheral vascularresistance.86 Similar to esmolol, it is rapidly metab-olized by RBC esterases; thus, its metabolism is notaffected by renal or hepatic function. Clevidipinereduces BP by a direct and selective effect onarterioles, thereby reducing afterload without affect-

1954 Postgraduate Education Corner

Page 43: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

ing cardiac filling pressures or causing reflex tachy-cardia.35 Stroke volume and cardiac output usuallyincrease. Moreover, clevidipine has been shown toprotect against ischemia/reperfusion injury in ananimal model of myocardial ischemia and to main-tain renal function and splanchnic blood flow.87–89

Several small trials90,91 have shown clevidipine tobe very effective in the control of postoperativehypertension. Although no studies have investigatedthe role of this drug in hypertensive emergencies, itsprofile makes it a potentially ideal drug for thisindication. At this time, clevidipine is not available inthe United States for use outside of clinical trials.

Nifedipine, nitroglycerin, and hydralazine are notrecommended in the management of hypertensiveemergencies. The basis of these recommendationsare discussed below.

Nifedipine

Nifedipine has been widely used via oral or sub-lingual administration in the management of hyper-tensive emergencies, severe hypertension associatedwith chronic renal failure, postoperative hyperten-sion, and pregnancy-induced hypertension. Althoughnifedipine has been administered via the sublingualroute, the drug is poorly soluble and is not absorbedthrough the buccal mucosa. It is however rapidlyabsorbed from the GI tract after the capsule isbroken/dissolved.92 This mode of administration hasnot been approved by the US Food and DrugAdministration (FDA). A significant decrease in BPis usually observed 5 to 10 min after nifedipineadministration, with a peak effect from 30 to 60 min,and a duration of action of approximately 6 to 8 h.93

Sudden uncontrolled and severe reductions in BPaccompanying the administration of nifedipine mayprecipitate cerebral, renal, and myocardial ischemicevents that have been associated with fatal out-comes.94 Elderly hypertensive patients with under-lying organ impairment and structural vascular dis-ease are more vulnerable to the rapid anduncontrolled reduction in arterial pressure. Giventhe seriousness of the reported adverse events andthe lack of any clinical documentation attesting to abenefit, the use of nifedipine capsules for hyperten-sive emergencies and “pseudoemergencies” shouldbe abandoned. The Cardiorenal Advisory Committeeof the FDA has concluded that the practice of admin-istering sublingual/oral nifedipine should be aban-doned because this agent is not safe nor efficacious.95

Nitroglycerin, Hydralazine, and Diuretics

Nitroglycerin is a potent venodilator and only athigh doses affects arterial tone.96 It causes hypoten-

sion and reflex tachycardia, which are exacerbated bythe volume depletion characteristic of hypertensiveemergencies. Nitroglycerin reduces BP by reducingpreload and cardiac output; undesirable effects inpatients with compromised cerebral and renal per-fusion. Low-dose administration (approximately 60mg/min) may, however, be used as an adjunct to IVantihypertensive therapy in patients with hyperten-sive emergencies associated with acute coronarysyndromes or acute pulmonary edema.

Hydralazine is a direct-acting vasodilator. Follow-ing IM or IV administration, there is an initial latentperiod of 5 to 15 min followed by a progressive andoften precipitous fall in BP that can last up to12 h.97,98 Although the circulating half-life of hydral-azine is only approximately 3 h, the half-time of itseffect on BP is approximately 10 h.99,100 Because ofthe prolonged and unpredictable antihypertensiveeffects of hydralazine and the inability to effectivelytitrate its hypotensive effect, it is best avoided in themanagement of hypertensive crises.

Volume depletion is common in patients withhypertensive emergencies, and the administration ofa diuretic together with a hypertensive agent canlead to a precipitous drop in BP. Diuretics should beavoided unless specifically indicated for volumeoverload, as occurs in renal parenchymal disease orcoexisting pulmonary edema.

Special Conditions

Acute Aortic Dissection

Aortic dissection should be considered a likelydiagnostic possibility in patients presenting to theemergency department with acute chest pain andelevated BP. Left untreated, approximately threefourths of patients with type A dissection (ascendingaorta) die within 2 weeks of an acute episode, butwith successful therapy the 5-year survival rate is75%.30,101 Hence, timely recognition of this diseaseentity coupled with urgent and appropriate manage-ment is the key to a successful outcome in themajority of these patients. It is important to recog-nize that the propagation of the dissection is depen-dent not only on the elevation of the BP itself butalso on the velocity of left ventricular ejec-tion.30,31,101–103

A vasodilator alone is not ideal in the treatment ofacute aortic dissection because this can promotereflex tachycardia, increase aortic ejection velocity,and promote dissection propagation. The combina-tion of a �-adrenergic antagonist and vasodilator isthe standard approach to treatment.30,31 Esmolol isthe �-adrenergic antagonist of choice with metopro-lol as a suitable alternative.104,105 Although nitroprus-

www.chestjournal.org CHEST / 131 / 6 / JUNE, 2007 1955

Page 44: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

side has traditionally been used as the vasodilator ofchoice, nicardipine or fenoldopam are less toxic,equally effective alternatives.105,106 All patients withaortic dissection require cardiovascular surgical con-sultation to determine if surgical management isnecessary. Unless significant medical comorbiditiesare present, surgery is indicated for all patients withtype A dissection.107,108 Patients with type B dissec-tions and distal aortic dissections can be managedwith aggressive BP control because outcomes havebeen shown to be the same with either medical orsurgical treatment unless complications such as leak,rupture, or impaired flow to vital organs super-vene.30,31,103

Cerebrovascular Accidents

The vast majority of patients with cerebral isch-emia present with acutely elevated BP regardless ofthe subtype of infarct or preexisting hyperten-sion.109,110 The BP elevation decreases spontane-ously over time. The elevated BP is not a manifes-tation of a hypertensive emergency but rather aprotective physiologic response to maintain cerebralperfusion pressure to the vascular territory affectedby ischemia. Lowering the BP in patients withischemic strokes may reduce cerebral blood flow,which because of impaired autoregulation, may re-sult in further ischemic injury. The common practiceof “normalizing” the BP following a cerebrovascularaccident is potentially dangerous. It should be notedthat the Intravenous Nimodipine West EuropeanTrial for acute stroke was stopped because of in-creased neurologic deterioration in the treatmentgroup, which the investigators111,112 attributed to theeffects of hypotension.

The American Stroke Association and the Euro-pean Stroke Initiative guidelines113,114 recommendwithholding antihypertensive therapy for acute isch-emic stroke unless there is planned thrombolysis,evidence of concomitant noncerebral acute organdamage, or if the BP is excessively high, arbitrarilychosen as a SBP � 220 mm Hg or a DBP � 120 mmHg based on the upper limit of normal autoregula-tion. In these patients, the aim is to reduce thepressure by not more than 10 to 15% in the first 24 h.Semplicini and colleagues115 demonstrated that ahigh initial BP was associated with a better neuro-logic outcome following an acute ischemic stroke.These authors115 suggests that hypertension may beprotective during an acute ischemic stroke and thatlowering the BP may be potentially harmful. Indeed,pharmacologic elevation of BP in patients with isch-emic stroke is a promising investigational approach.Small studies116–118 of patients treated with vaso-pressors and plasma expanders have demonstrated

short-term neurologic improvement in 20 to 40% ofpatients without any adverse effects. These protocolsgenerally call for raising the MAP by 20% or to 130to 140 mm Hg while keeping the SBP � 200 mmHg. It is thought that patients with fluctuatingdeficits, proximal large vessel stenosis or occlusion,or large areas of MRI diffusion-perfusion mismatchare most likely to respond to induced hypertension.In patients receiving thrombolytic therapy, antihy-pertensive therapy is required for SBP � 185 mmHg or DBP � 110 mm Hg, with a targeted SBP of180 mm Hg and a DBP of 105 mm Hg.113,119,120 Thecurrent American Heart Association guidelines119

recommend the use of labetalol or nicardipine if theSBP is � 220 mm Hg or the DBP is from 121 to 140mm Hg, and nitroprusside for a DBP � 140 mm Hg.For the reasons outlined above, we believe nitro-prusside to be a poor choice in patients with intra-cranial pathology. The Acute Candesartan CilexetilTherapy in Stroke Survivors study121 demonstrated areduction in 12-month mortality and the number ofvascular events in patients with a SBP � 200 mm Hgor a DBP � 110 mm Hg who were treated with anangiotensin type 1 receptor blockade (candesartancilexetil) immediately after an ischemic stroke. Themechanism(s) by which the angiotensin type 1 re-ceptor blocker exerted its beneficial effects is un-clear, as the BP profiles were nearly identical in thetreatment and placebo groups. Additional studies arerequired to confirm the benefit of angiotensin type 1receptor blockers in patients with ischemic stroke.

In patients with intracerebral hematomas, there isalmost always a rise in intracranial pressure withreflex systemic hypertension. There is no evidencethat hypertension provokes further bleeding in pa-tients with intracranial hemorrhage. However, aprecipitous fall in systemic BP will compromisecerebral perfusion. The controlled lowering of theBP is currently recommended only when the SBP is� 200 mm Hg, the DBP is � 110 mm Hg, or theMAP is � 130 mm Hg.122–124 A study125 has demon-strated that the rapid decline of BP within the first24 h after presentation of an intracranial hemorrhagewas associated with increased mortality; the rate ofdecline in BP was independently associated withincreased mortality. Nicardipine has been demon-strated to be an effective agent for the control of BPin patients with intracerebral hemorrhage.126

Preeclampsia and Eclampsia

Hypertension is one of the most common medicaldisorders affecting pregnancy. It complicates 12% ofpregnancies and is responsible for 18% of maternaldeaths in the United States.127 The presentation of apatient with pregnancy-induced hypertension may

1956 Postgraduate Education Corner

Page 45: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

range from a mild to a life-threatening diseaseprocess.128 Initial therapy of preeclampsia includesvolume expansion, magnesium sulfate (MgSO4) forseizure prophylaxis and BP control.129–131 Delivery isthe definitive treatment for preeclampsia andeclampsia.

Magnesium sulfate is usually administered as aloading dose of 4 to 6 g in 100 mL 5% dextrose in 1/4normal saline solution over 15 to 20 min, followed bya constant infusion of 1 to 2 g/h of MgSO4 dependingon urine output and deep tendon reflexes, which arechecked on an hourly basis. The next step in themanagement of preeclampsia is to reduce the BP toa safe range being diligent to avoid significant hypo-tension. The objective of treating severe hyperten-sion is to prevent intracerebral hemorrhage andcardiac failure without compromising cerebral per-fusion or jeopardizing uteroplacental blood flow,which is already reduced in many women withpreeclampsia.128 Studies132–134 of women with mildpreeclampsia have shown no benefit to antihyperten-sive therapy (labetalol or calcium-channel blockers)and suggested that antihypertensive therapy mayincrease the risk of intrauterine growth retardation.Antihypertensive therapy is therefore administeredprimarily to prevent complications in the mother.The Working Group Report on High Blood Pressurein Pregnancy135 recommends initiation of antihyper-tensive therapy for a DBP � 105 mm Hg. Further-more, most authorities and the current guidelinesfrom the American College of Obstetricians andGynecologists128,135–138 recommend keeping SBPfrom 140 to 160 mm Hg and DBP from 90 to 105mm Hg. This recommendation is supported by astudy139 that demonstrated that SBP � 160 mm Hgwas the most important factor associated with acerebrovascular accident in patients with severepreeclampsia and eclampsia. This would suggest thatSBP from 155 to 160 mm Hg should be the primarytrigger to initiate antihypertensive therapy in a pa-tient with severe preeclampsia or eclampsia.139,140 Itshould be noted that patients with preeclampsia/eclampsia may have a very labile BP; this facttogether with the narrow target BP range dictate thatthese patients be closely monitored in an ICU,preferably with an arterial catheter. Intracerebralhemorrhage is a devastating complication in thesepatients that can be avoided by scrupulous attentionto BP control.

No antihypertensive medication is specifically ap-proved by the FDA for use in pregnant women.Hydralazine has been recommended as the drug ofchoice to treat severe preeclampsia and eclampsiasince the early 1970s.141 However, hydralazine has anumber of properties that make it unsuitable for thisindication. Its side effects (such as headache, nausea,

and vomiting) are common and mimic symptoms ofdeteriorating preeclampsia. Most importantly, how-ever, it has a delayed onset of action, an unpredict-able hypotensive effect, and a prolonged duration ofaction. These properties may result in a precipitoushypotensive overshoot compromising both maternalcerebral blood flow and uteroplacental blood flow.Indeed, in a metaanalysis published by Magee andcolleagues,142 hydralazine was associated with anincreased risk of maternal hypotension that wasassociated with an excess of cesarean sections, pla-cental abruptions, and low Apgar scores. Based onthe available data, we suggest that hydralazine not beused as first-line treatment of severe hypertension inpregnancy. Similarly, sublingual or oral nifedipineshould be avoided in this setting. Our preference isIV labetalol or nicardipine, which are easier to titrateand have a more predictable dose response thanhydralazine. Both agents appear to be safe andeffective in hypertensive pregnant patients.143–149

Nitroprusside and ACE inhibitors are contraindi-cated in pregnant patients.

Sympathetic Crises

The most commonly encountered sympatheticcrises are related to the recreational use of sympa-thomimetic drugs such as cocaine, amphetamine, orphencyclidine. Rarely, these crises may be seen withpheochromocytoma, patients receiving a monoamineoxidase inhibitor who ingest a tyramine-containingfood, or patients who abruptly stop antihypertensivemedications such as clonidine or �-adrenergic antag-onists.

In the clinical situations characterized by sympa-thetic overstimulation, �-adrenergic antagonistsshould be avoided to prevent vascular �-receptorantagonism resulting in unopposed �-adrenergic ac-tivity and potential increase in BP. In fact, in co-caine-induced hypertensive emergency, the use of�-adrenergic blockade can increase coronary vaso-constriction, fail to control heart rate, increase BP,and decrease survival.150–152 Interestingly, althoughlabetalol is traditionally considered the ideal agentdue to its �- and �-adrenergic antagonism, experi-mental studies153–157 do not support its use in thisclinical setting. BP control is best achieved withnicardipine, fenoldopam, or verapamil in combina-tion with a benzodiazepine.152,158,159 Phentolamine isan alternative agent.160

Acute Postoperative Hypertension

Acute postoperative hypertension (APH) has beendefined as a significant elevation in BP during theimmediate postoperative period that may lead toserious neurologic, cardiovascular, or surgical-site

www.chestjournal.org CHEST / 131 / 6 / JUNE, 2007 1957

Page 46: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

complications and that requires urgent manage-ment.161 Despite the widespread and long-standingrecognition of APH, there is no agreement in theliterature on a more precise quantitative defini-tion.161–163 APH has an early onset, being observedwithin 2 h after surgery in most cases and is typicallyof short duration, with most patients requiring treat-ment for � 6 h. Postoperative complications of APHmay include hemorrhagic stroke, cerebral ischemia,encephalopathy, myocardial ischemia, myocardial in-farction, cardiac arrhythmia, congestive cardiac fail-ure with pulmonary edema, failure of vascular anas-tomoses, and bleeding at the surgical site. AlthoughAPH may occur following any major surgery, it ismost commonly associated with cardiothoracic, vas-cular, head and neck, and neurosurgical procedures.The pathophysiologic mechanism underlying APH isuncertain and may vary with the surgical procedureand other factors. However, the final common path-way leading to hypertension appears to be activationof the sympathetic nervous system, as evidenced byelevated plasma catecholamine concentrations inpatients with APH.17 The primary hemodynamicalteration observed in APH is an increase in afterloadwith an increase in SBP and DBP with or withouttachycardia.

There is no consensus concerning the treatmentthreshold for the management of noncardiac surgerypatients with APH. Treatment is frequently a bed-side decision by the anesthesiologist or surgeon thattakes into consideration the baseline BP, concomi-tant disease, and the perceived risk of complications.In contrast, in cardiac surgery patients, treatment isrecommended for a BP � 140/90 or a MAP of atleast 105 mm Hg.161–163 Pain and anxiety are com-mon contributors to BP elevations and should betreated before administration of antihypertensivetherapy. Other potentially reversible causes of APHinclude hypothermia with shivering, hypoxemia, hy-percarbia, and bladder distension. Short-term ad-ministration of a short-acting IV agent is recom-mended when there is no identifiable treatable causeof hypertension. Labetalol, esmolol, nicardipine, andclevidipine have proven effective in the managementof APH.90,91,161,164–168

Conclusions

Patients with hypertensive emergencies requirethe immediate reduction of the elevated BP toprevent and arrest progressive end-organ damage.The best clinical setting to achieve this BP control isin the ICU, with the use of titratable IV hypotensiveagents. There are several antihypertensive agentsavailable including esmolol, nicardipine, labetalol,

and fenoldopam. While sodium nitroprusside is arapid-acting and potent antihypertensive agent, itmay be associated with significant toxicity and shouldtherefore be used in select circumstances at a dosenot to exceed 2 �g/kg/min. The appropriate thera-peutic approach of each patient will depend on theclinical presentation of the patient. Agents such asnifedipine and hydralazine should be abandonedbecause these agents are associated with significanttoxicities and/or side effect profile.

ACKNOWLEDGMENT: The author acknowledges the assis-tance of Susan Baik, RN, for her helpful critique and editorialassistance with this article.

References1 Hajjar I, Kotchen TA. Trends in prevalence, awareness,

treatment, and control of hypertension in the United States,1988–2000. JAMA 2003; 290:199–206

2 Chobanian AV, Bakris GL, Black HR, et al. The seventhReport of the Joint National Committee on Prevention,Detection, Evaluation, and Treatment of High Blood Pres-sure: the JNC 7 report. JAMA 2003; 289:2560–2572

3 The fifth report of the Joint National Committee on Detec-tion, Evaluation, and Treatment of High Blood Pressure.Arch Intern Med 1993; 153:154–183

4 Joint National Committee for the Detection, Evaluation andTreatment of High Blood Pressure: the 1984 report. ArchIntern Med 1984; 114:1045–1057

5 Volhard F, Fahr T. Die brightsche Nierenkranbeit: Klinik,Pathologie und Atlas. Berlin, Germany: Springer, 1914

6 Keith NM, Wagener HP, Barker NW. Some different typesof essential hypertension: their course and prognosis. Am JMed Sci 1939; 197:332–343

7 Laragh J. Laragh’s lessons in pathophysiology and clinicalpearls for treating hypertension. Am J Hypertens 2001;14:837–854

8 McRae RPJ, Liebson PR. Hypertensive crisis. Med ClinNorth Am 1986; 70:749–767

9 Vidt DG. Current concepts in treatment of hypertensiveemergencies. Am Heart J 1986; 111:220–225

10 Bennett NM, Shea S. Hypertensive emergency: case crite-ria, sociodemographic profile, and previous care of 100cases. Am J Public Health 1988; 78:636–640

11 Potter JF. Malignant hypertension in the elderly. Q J Med1995; 88:641–647

12 Vital and health statistics: detailed diagnoses and proceduresfor patients discharged from short-stay hospitals: UnitedStates, 1983–1990. Hyattsville, MD: National Center forHealth Statistics, 1997

13 Tisdale JE, Huang MB, Borzak S, et al. Risk factors forhypertensive crisis: importance of out-patient blood pres-sure control. Fam Pract 2004; 21:420–424

14 Shea S, Misra D, Ehrlich MH, et al. Predisposing factors forsevere, uncontrolled hypertension in an inner-city minoritypopulation. N Engl J Med 1992; 327:776–781

15 Tumlin JA, Dunbar LM, Oparil S, et al. Fenoldopam, adopamine agonist, for hypertensive emergency: a multi-center randomized trial: fenoldopam Study Group. AcadEmerg Med 2000; 7:653–662

16 Ault MJ, Ellrodt AG. Pathophysiological events leading tothe end-organ effects of acute hypertension. Am J EmergMed 1985; 3:10–15

17 Wallach R, Karp RB, Reves JG, et al. Pathogenesis of

1958 Postgraduate Education Corner

Page 47: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

paroxysmal hypertension developing during and after coro-nary bypass surgery: a study of hemodynamic and humoralfactors. Am J Cardiol 1980; 46:559–565

18 Funakoshi Y, Ichiki T, Ito K, et al. Induction of interleukin-6expression by angiotensin II in rat vascular smooth musclecells. Hypertension 1999; 34:118–125

19 Han Y, Runge MS, Brasier AR. Angiotensin II inducesinterleukin-6 transcription in vascular smooth muscle cellsthrough pleiotropic activation of nuclear factor-� B tran-scription factors. Circ Res 1999; 84:695–703

20 Zampaglione B, Pascale C, Marchisio M, et al. Hypertensiveurgencies and emergencies: prevalence and clinical presen-tation. Hypertension 1996; 27:144–147

21 Varon J, Marik PE. The diagnosis and management ofhypertensive crises. Chest 2000; 118:214–227

22 Rey E, LeLorier J, Burgess E, et al. Report of the CanadianHypertension Society Consensus Conference: 3. Pharmaco-logic treatment of hypertensive disorders in pregnancy. CanMed Assoc J 1997; 157:1245–1254

23 Graves JW. Prevalence of blood pressure cuff sizes in areferral practice of 430 consecutive adult hypertensives.Blood Press Monit 2001; 6:17–20

24 Linfors EW, Feussner JR, Blessing CL, et al. Spurioushypertension in the obese patient: effect of sphygmoma-nometer cuff size on prevalence of hypertension. ArchIntern Med 1984; 144:1482–1485

25 Hickler RB. “Hypertensive emergency”: a useful diagnosticcategory. Am J Public Health 1988; 78:623–624

26 Garcia JYJ, Vidt DG. Current management of hypertensiveemergencies. Drugs 1987; 34:263–278

27 Fromm RE, Varon J, Gibbs L. Congestive heart failure andpulmonary edema for the emergency physician. J EmergMed 1995; 13:71–87

28 Gandhi SK, Powers JC, Nomeir AM, et al. The pathogenesisof acute pulmonary edema associated with hypertension.N Engl J Med 2001; 344:17–22

29 Strandgaard S, Olesen J, Skinhoj E, et al. Autoregulation ofbrain circulation in severe arterial hypertension. BMJ 1973;1:507–510

30 Khan IA, Nair CK. Clinical, diagnostic, and managementperspectives of aortic dissection. Chest 2002; 122:311–328

31 Estrera AL, Miller CC III, Safi HJ, et al. Outcomes ofmedical management of acute type B aortic dissection.Circulation 2006; 114:I384–I389

32 Khot UN, Novaro GM, Popovic ZB, et al. Nitroprusside incritically ill patients with left ventricular dysfunction andaortic stenosis. N Engl J Med 2003; 348:1756–1763

33 DiPette DJ, Ferraro JC, Evans RR, et al. Enalaprilat, anintravenous angiotensin-converting enzyme inhibitor, in hy-pertensive crises. Clin Pharmacol Ther 1985; 38:199–204

34 Hirschl MM, Binder M, Bur A, et al. Impact of therenin-angiotensin-aldosterone system on blood pressure re-sponse to intravenous enalaprilat in patients with hyperten-sive crises. J Hum Hypertens 1997; 11:177–183

35 Nordlander M, Bjorkman JA, Regard HCG, et al. Pharma-cokinetics and hemodynamic effects of an ultrashort-actingcalcium antagonist [abstract]. Br J Anaesth 1996; 76 (suppl):A24

36 Lund-Johansen P. Pharmacology of combined �-�-block-ade: II. Haemodynamic effects of labetalol. Drugs 1984;28(suppl 2):35–50

37 Kanot J, Allonen H, Kleimola T, et al. Pharmacokinetics oflabetalol in healthy volunteers. Int J Clin Pharmacol TherToxicol 1981; 19:41–44

38 Goldberg ME, Clark S, Joseph J, et al. Nicardipine versusplacebo for the treatment of postoperative hypertension. AmHeart J 1990; 119:446–450

39 Pearce CJ, Wallin JD. Labetalol and other agents that blockboth �- and �-adrenergic receptors. Cleve Clin J Med 1994;61:59–69

40 Wallin JD. Adrenoreceptors and renal function. J ClinHyperten 1985; 1:171–178

41 Marx PG, Reid DS. Labetalol infusion in acute myocardialinfarction with systemic hypertension. Br J Clin Pharmacol1979; 8:233S–238S

42 Olsen KS, Svendsen LB, Larsen FS, et al. Effect of labetalolon cerebral blood flow, oxygen metabolism and autoregula-tion in healthy humans. Br J Anaesth 1995; 75:51–54

43 Rosei EA, Trust PM, Brown JJ. Intravenous labetalol insevere hypertension. Lancet 1975; 2:1093–1094

44 Schillinger D. Nifedipine in hypertensive emergencies: aprospective study. J Emerg Med 1987; 5:463–473

45 Lambert CR, Hill JA, Feldman RL, et al. Effects ofnicardipine on exercise- and pacing-induced myocardialischemia in angina pectoris. Am J Cardiol 1987; 60:471–476

46 Lambert CR, Hill JA, Nichols WW, et al. Coronary andsystemic hemodynamic effects of nicardipine. Am J Cardiol1985; 55:652–656

47 Vincent JL, Berlot G, Preiser JC, et al. Intravenous nicardi-pine in the treatment of postoperative arterial hypertension.J Cardiothorac Vasc Anesth 1997; 11:160–164

48 Lambert CR, Hill JA, Feldman RL, et al. Effects ofnicardipine on left ventricular function and energetics inman. Int J Cardiol 1986; 10:237–250

49 Gray RJ. Managing critically ill patients with esmolol: anultra short-acting �-adrenergic blocker. Chest 1988; 93:398–403

50 Lowenthal DT, Porter RS, Saris SD, et al. Clinical pharma-cology, pharmacodynamics and interactions with esmolol.Am J Cardiol 1985; 56:14F–18F

51 Reynolds RD, Gorczynski RJ, Quon CY. Pharmacology andpharmacokinetics of esmolol. J Clin Pharmacol 1986;26(suppl):A3–A14

52 Balser JR, Martinez EA, Winters BD. et al. �-Adrenergicblockade accelerates conversion of postoperative supraven-tricular tachyarrhythmias. Anesthesiol 1998; 89:1052–1059

53 Platia EV, Michelson EL, Porterfield JK, et al. Esmololversus verapamil in the acute treatment of atrial fibrillationor atrial flutter. Am J Cardiol 1989; 63:925–929

54 Stumpf JL. Drug therapy of hypertensive crises. Clin Pharm1988; 7:582–591

55 Smerling A, Gersony WM. Esmolol for severe hypertensionfollowing repair of aortic coarctation. Crit Care Med 1990;18:1288–1290

56 Gray RJ, Bateman TM, Czer LS, et al. Use of esmolol inhypertension after cardiac surgery. Am J Cardiol 1985;56:49F–56F

57 Gray RJ, Bateman TM, Czer LS, et al. Comparison ofesmolol and nitroprusside for acute post-cardiac surgicalhypertension. Am J Cardiol 1987; 59:887–891

58 Muzzi DA, Black S, Losasso TJ, et al. Labetalol and esmololin the control of hypertension after intracranial surgery.Anesth Analg 1990; 70:68–71

59 Bodmann KF, Troster S, Clemens R, et al. Hemodynamicprofile of intravenous fenoldopam in patients with hyperten-sive crisis. Clin Investig 1993; 72:60–64

60 Munger MA, Rutherford WF, Anderson L, et al. Assessmentof intravenous fenoldopam mesylate in the management ofsevere systemic hypertension. Crit Care Med 1990; 18:502–504

61 White WB, Radford MJ, Gonzalez FM, et al. Selectivedopamine-1 agonist therapy in severe hypertension: effectsof intravenous fenoldopam. J Am Coll Cardiol 1988; 11:1118–1123

www.chestjournal.org CHEST / 131 / 6 / JUNE, 2007 1959

Page 48: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

62 Shusterman NH, Elliott WJ, White WB. Fenoldopam, butnot nitroprusside, improves renal function in severely hy-pertensive patients with impaired renal function. Am J Med1993; 95:161–168

63 Elliott WJ, Weber RR, Nelson KS, et al. Renal and hemo-dynamic effects of intravenous fenoldopam versus nitroprus-side in severe hypertension. Circulation 1990; 81:970–977

64 White WB, Halley SE. Comparative renal effects of intra-venous administration of fenoldopam mesylate and sodiumnitroprusside in patients with severe hypertension. ArchIntern Med 1989; 149:870–874

65 Ng TM, Shurmur SW, Silver M, et al. Comparison ofN-acetylcysteine and fenoldopam for preventing contrast-induced nephropathy (CAFCIN). Int J Cardiol 2006; 109:322–328

66 Pannu N, Wiebe N, Tonelli M. Prophylaxis strategies forcontrast-induced nephropathy. JAMA 2006; 295:2765–2779

67 Friederich JA, Butterworth JF. Sodium nitroprusside:twenty years and counting. Anesth Analg 1995; 81:152–162

68 Robin ED, McCauley R. Nitroprusside-related cyanide poi-soning: time (long past due. for urgent, effective interven-tions: chest 1992; 102:1842–1845

69 Hartmann A, Buttinger C, Rommel T, et al. Alteration ofintracranial pressure, cerebral blood flow, autoregulationand carbon dioxide-reactivity by hypotensive agents in ba-boons with intracranial hypertension. Neurochirurgia 1989;32:37–43

70 Kondo T, Brock M, Bach H. Effect of intra-arterial sodiumnitroprusside on intracranial pressure and cerebral autoreg-ulation. Jpn Heart J 1984; 25:231–237

71 Griswold WR, Reznik V, Mendoza SA. Nitroprusside-in-duced intracranial hypertension [letter]. JAMA 1981; 246:2679–2680

72 Anile C, Zanghi F, Bracali A, et al. Sodium nitroprusside andintracranial pressure. Acta Neurochir 1981; 58:203–211

73 Mann T, Cohn PF, Holman LB, et al. Effect of nitroprussideon regional myocardial blood flow in coronary artery disease:results in 25 patients and comparison with nitroglycerin.Circulation 1978; 57:732–738

74 Cohn JN, Franciosa JA, Francis GS, et al. Effect of short-term infusion of sodium nitroprusside on mortality rate inacute myocardial infarction complicated by left ventricularfailure: results of a Veterans Administration cooperativestudy. N Engl J Med 1982; 306:1129–1135

75 Pasch T, Schulz V, Hoppenshauser G. Nitroprusside-in-duced formation of cyanide and its detoxication with thio-sulphate during deliberate hypotension. J Cardiovasc Phar-macol 1983; 5:77–85

76 Hall VA, Guest JM. Sodium nitroprusside-induced cyanideintoxication and prevention with sodium thiosulphate pro-phylaxis. Am J Crit Care 1992; 2:19–27

77 Niknahad H, O’Brien PJ. Involvement of nitric oxide innitroprusside-induced hepatocyte cytotoxicity. BiochemPharmacol 1996; 51:1031–1039

78 Izumi Y, Benz AM, Clifford DB, et al. Neurotoxic effects ofsodium nitroprusside in rat hippocampal slices. Exp Neurol1993; 121:14–23

79 Vesey CJ, Cole PV, Simpson PJ. Cyanide and thiocyanateconcentrations following sodium nitroprusside infusion inman. Br J Anaesth 1976; 48:651–659

80 Nakamura Y, Yasuda M, Fujimori H, et al. Cytotoxic effectof sodium nitroprusside on PC12 cells. Chemosphere 1997;34:317–324

81 Gobbel GT, Chan TY, Chan PH. Nitric oxide- and superox-ide-mediated toxicity in cerebral endothelial cells. J Phar-macol Exp Ther 1997; 282:1600–1607

82 Rauhala P, Khaldi A, Mohanakumar KP, et al. Apparent role

of hydroxyl radicals in oxidative brain injury induced bysodium nitroprusside. Free Radic Biol Med 1998; 24:1065–1073

83 Rodriguez G, Varon J. Clevidipine: a unique agent for thecritical care practitioner. Crit Care Shock 2006; 9:9–15

84 Bailey JM, Lu W, Levy JH, et al. Clevidipine in adult cardiacsurgical patients: a dose-finding study. Anesthesiol 2002;96:1086–1094

85 Ericsson H, Fakt C, Jolin-Mellgard A, et al. Clinical andpharmacokinetic results with a new ultrashort-acting cal-cium antagonist, clevidipine, following gradually increasingintravenous doses to healthy volunteers. Br J Clin Pharmacol1999; 47:531–538

86 Ericsson H, Tholander B, RegardH CG. In vitro hydrolysisrate and protein binding of clevidipine, a new ultrashort-acting calcium antagonist metabolised by esterases, in dif-ferent animal species and man. Eur J Pharmaceut Sci 1999;8:29–37

87 Segawa D, Sjoquist PO, Wang QD, et al. Time-dependentcardioprotection with calcium antagonism and experimentalstudies with clevidipine in ischemic-reperfused pig hearts:part II. J Cardiovasc Pharmacol 2002; 40:339–345

88 Segawa D, Sjoquist PO, Wang QD, et al. Calcium antagonistprotects the myocardium from reperfusion injury by inter-fering with mechanisms directly related to reperfusion: anexperimental study with the ultrashort-acting calcium antag-onist clevidipine. J Cardiovasc Pharmacol 2000; 36:338–343

89 Stephens CT, Jandhyala BS. Effects of fenoldopam, adopamine D-1 agonist, and clevidipine, a calcium channelantagonist, in acute renal failure in anesthetized rats. ClinExp Hypertension 2002; 24:301–313

90 Powroznyk AV, Vuylsteke A, Naughton C, et al. Comparisonof clevidipine with sodium nitroprusside in the control ofblood pressure after coronary artery surgery. Eur J Anaes-thesiol 2003; 20:697–703

91 Kieler-Jensen N, Jolin-Mellgard A, Nordlander M, et al.Coronary and systemic hemodynamic effects of clevidipine,an ultra-short-acting calcium antagonist, for treatment ofhypertension after coronary artery surgery. Acta Anaesthe-siol Scand 2000; 44:186–193

92 van Harten J, Burggraaf K, Danhof M, et al. Negligiblesublingual absorption of nifedipine. Lancet 1987; 2:1363–1365

93 Huysmans FT, Sluiter HE, Thien TA, et al. Acute treatmentof hypertensive crisis with nifedipine. Br J Clin Pharmacol1983; 16:725–727

94 Grossman E, Messerli FH, Grodzicki T, et al. Should amoratorium be placed on sublingual nifedipine capsulesgiven for hypertensive emergencies and pseudoemergen-cies? JAMA 1996; 276:1328–1331

95 Levy JH. Treatment of perioperative hypertension. Anesthe-siol Clin North Am 1999; 17:569–570

96 Bussmann WD, Kenedi P, von Mengden HJ, et al. Compar-ison of nitroglycerin with nifedipine in patients with hyper-tensive crisis or severe hypertension. Clin Investig 1992;70:1085–1088

97 Schroeder HA. Effects on hypertension of sulfhydryl andhydrazine compounds. J Clin Invest 1951; 30:672–673

98 Shepherd AM, Ludden TM, McNay JL, et al. Hydralazinekinetics after single and repeated oral doses. Clin PharmacolTher 1980; 28:804–811

99 O’Malley K, Segal JL, Israili ZH, et al. Duration of hydral-azine action in hypertension. Clin Pharmacol Ther 1975;18:581–586

100 Ludden TM, Shepherd AM, McNay JL, et al. Hydralazinekinetics in hypertensive patients after intravenous adminis-tration. Clin Pharmacol Ther 1980; 28:736–742

1960 Postgraduate Education Corner

Page 49: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

101 Kouchoukos NT, Dougenis D. Surgery of the thoracic aorta.N Engl J Med 1997; 336:1876–1888

102 Cohn LH. Aortic dissection: new aspects of diagnosis andtreatment. Hosp Pract 1994; 29:47–56

103 Chen K, Varon J, Wenker OC, et al. Acute thoracic aorticdissection: the basics. J Emerg Med 1997; 15:859–867

104 O’Connor B, Luntley JB. Acute dissection of the thoracicaorta: esmolol is safer than and as effective as labetalol[letter]. BMJ 1995; 310:875

105 Hoshino T, Ohmae M, Sakai A. Spontaneous resolution of adissection of the descending aorta after medical treatmentwith a � blocker and a calcium antagonist. Br Heart J 1987;58:82–84

106 Iguchi A, Tabayashi K. Outcome of medically treated Stan-ford type B aortic dissection. Jpn Circ J 1998; 62:102–105

107 Pitt MP, Bonser RS. The natural history of thoracic aorticaneurysm disease: an overview. J Card Surg 1997; 12:270–278

108 Borst HG, Laas J. Surgical treatment of thoracic aorticaneurysms. Adv Card Surg 1993; 4:47–87

109 Wallace JD, Levy LL. Blood pressure after stroke. JAMA1981; 246:2177–2180

110 Britton M, Carlsson A, de Faire U. Blood pressure course inpatients with acute stroke and matched controls. Stroke1986; 17:861–864

111 Wahlgren NG, MacMohon DG, De Keyser J, et al. TheIntravenous Nimodipine West Europen Trial (INWEST) ofnimodipine in the treatment of acute ischemic stroke.Cerebrovasc Dis 1994; 4:204–210

112 Ahmed N, Nasman P, Wahlgren NG. Effect of intravenousnimodipine on blood pressure and outcome after acutestroke. Stroke 2000; 31:1250–1255

113 Adams HP Jr, Adams RJ, Brott T, et al. Guidelines for theearly management of patients with ischemic stroke: a scien-tific statement from the Stroke Council of the AmericanStroke Association. Stroke 2003; 34:1056–1083

114 The European Stroke Initiative Executive Committee andthe EUSI Writing Committee. European Stroke Initiativerecommendations for stroke management: update 2003.Cerebrovasc Dis 2003; 16:311–337

115 Semplicini A, Maresca A, Boscolo G, et al. Hypertension inacute ischemic stroke: a compensatory mechanism or anadditional damaging factor? Arch Intern Med 2003; 163:211–216

116 Rordorf G, Koroshetz WJ, Ezzeddine MA, et al. A pilotstudy of drug-induced hypertension for treatment of acutestroke. Neurology 2001; 56:1210–1213

117 Rordorf G, Cramer SC, Efird JT, et al. Pharmacologicalelevation of blood pressure in acute stroke: clinical effectsand safety. Stroke 1997; 28:2133–2138

118 Hillis AE, Ulatowski JA, Barker PB, et al. A pilot random-ized trial of induced blood pressure elevation: effects onfunction and focal perfusion in acute and subacute stroke.Cerebrovasc Dis 2003; 16:236–246

119 Adams H, Adams R, Del ZG, et al. Guidelines for the earlymanagement of patients with ischemic stroke: 2005 guide-lines update; a scientific statement from the Stroke Councilof the American Heart Association/American Stroke Associ-ation. Stroke 2005; 36:916–923

120 NINDS rtPA Stroke Study Group. Tissue plasminogenactivator for acute ischemic stroke. N Engl J Med 1995;333:1581–1587

121 Schrader J, Luders S, Kulschewski A, et al. The ACCESSstudy: evaluation of acute candesartan cilexetil therapy instroke survivors. Stroke 2003; 34:1699–1703

122 Lavin P. Management of hypertension in patients with acutestroke. Arch Intern Med 1986; 146:66–68

123 Hirschl MM. Guidelines for the drug treatment of hyper-tensive crises. Drugs 1995; 50:991–1000

124 O’Connell J, Gray C. Treating hypertension after stroke.BMJ 1994; 308:1523–1524

125 Qureshi AI, Bliwise DL, Bliwise NG, et al. Rate of 24-hourblood pressure decline and mortality after spontaneousintracerebral hemorrhage: a retrospective analysis with arandom effects regression model. Crit Care Med 1999;27:480–485

126 Qureshi AI, Harris-Lane P, Kirmani JF, et al. Treatment ofacute hypertension in patients with intracerebral hemor-rhage using American Heart Association guidelines. CritCare Med 2006; 34:1975–1980

127 Koonin LM, MacKay AP, Berg CJ, et al. Pregnancy-relatedmortality surveillance–United States, 1987–1990. MorbMortal Wkly Rep 1997; CDC Surveillance Summaries 46:17–36

128 Sibai BM. Diagnosis, prevention, and management ofeclampsia. Obstet Gynecol 2005; 105:402–410

129 Belfort MA, Anthony J, Saade GR, et al. A comparison ofmagnesium sulfate and nimodipine for the prevention ofeclampsia. N Engl J Med 2003; 348:304–311

130 Coetzee EJ, Dommisse J, Anthony J. A randomised con-trolled trial of intravenous magnesium sulphate versus pla-cebo in the management of women with severe pre-eclamp-sia. Br J Obstet Gynaecol 1998; 105:300–303

131 Lucas MJ, Leveno KJ, Cunningham FG. A comparison ofmagnesium sulfate with phenytoin for the prevention ofeclampsia. N Engl J Med 1995; 333:201–205

132 Sibai BM, Gonzalez AR, Mabie WC, et al. A comparison oflabetalol plus hospitalization versus hospitalization alone inthe management of preeclampsia remote from term. ObstetGynecol 1987; 70:323–327

133 Sibai BM, Barton JR, Akl S, et al. A randomized prospectivecomparison of nifedipine and bed rest versus bed rest alonein the management of preeclampsia remote from term. Am JObstet Gynecol 1992; 167:879–884

134 von DP, Ornstein MP, Bull SB, et al. Fall in mean arterialpressure and fetal growth restriction in pregnancy hyperten-sion: a meta-analysis. Lancet 2000; 355:87–92

135 Gifford RW, August PA, Cunningham G. Report of theNational High Blood Pressure Education Program WorkingGroup on high blood pressure in pregnancy. Am J ObstetGynecol 2000; 183:S1–22

136 National High Blood Pressure Education Program WorkingGroup on High Blood Pressure in Pregnancy. Report of theNational High Blood Pressure Education Program Workinggroup on high blood pressure in pregnancy. Am J ObstetGynecol 2000; 183:S1–S22

137 Diagnosis and management of preeclampsia and eclampsia:ACOG Practice Bulletin No. 33. American College ofObstetricians and Gynecologists. Obstet Gynecol 2002; 99:159–167

138 Sibai BM. Diagnosis, controversies, and management of thesyndrome of hemolysis, elevated liver enzymes, and lowplatelet count. Obstet Gynecol 2004; 103:981–991

139 Martin JN Jr, Thigpen BD, Moore RC, et al. Stroke andsevere preeclampsia and eclampsia: a paradigm shift focus-ing on systolic blood pressure. Obstet Gynecol 2005; 105:246–254

140 Cunningham FG. Severe preeclampsia and eclampsia: sys-tolic hypertension is also important. Obstet Gynecol 2005;105:237–238

141 Hellman LM, Pritchard JA. Williams obstetrics, 14th ed.New York, NY: Appleton-Century-Crofts, 1971

142 Magee LA, Cham C, Waterman EJ, et al. Hydralazine for

www.chestjournal.org CHEST / 131 / 6 / JUNE, 2007 1961

Page 50: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

treatment of severe hypertension in pregnancy: meta-anal-ysis. BMJ 2003; 327:955–960

143 Pickles CJ, Broughton PF, Symonds EM. A randomisedplacebo controlled trial of labetalol in the treatment of mildto moderate pregnancy induced hypertension. Br J ObstetGynaecol 1992; 99:964–968

144 Pickles CJ, Symonds EM, Pipkin FB. The fetal outcome ina randomized double-blind controlled trial of labetalol ver-sus placebo in pregnancy-induced hypertension. Br J ObstetGynaecol 1989; 96:38–43

145 Mabie WC, Gonzalez AR, Sibai BM, et al. A comparativetrial of labetalol and hydralazine in the acute management ofsevere hypertension complicating pregnancy. Obstet Gy-necol 1987; 70:328–333

146 Jannet D, Carbonne B, Sebban E, et al. Nicardipine versusmetoprolol in the treatment of hypertension during preg-nancy: a randomized comparative trial. Obstet Gynecol1994; 84:354–359

147 Carbonne B, Jannet D, Touboul C, et al. Nicardipinetreatment of hypertension during pregnancy. Obstet Gy-necol 1993; 81:908–914

148 Hanff LM, Vulto AG, Bartels PA, et al. Intravenous use ofthe calcium-channel blocker nicardipine as second-linetreatment in severe, early-onset pre-eclamptic patients.J Hypertension 2005; 23:2319–2326

149 Elatrous S, Nouira S, Ouanes BL, et al. Short-term treat-ment of severe hypertension of pregnancy: prospectivecomparison of nicardipine and labetalol. Intensive Care Med2002; 28:1281–1286

150 Lange RA, Cigarroa RG, Flores ED, et al. Potentiation ofcocaine-induced coronary vasoconstriction by �-adrenergicblockade. Ann Intern Med 1990; 112:897–903

151 Pitts WR, Lange RA, Cigarroa JE, et al. Cocaine-inducedmyocardial ischemia and infarction: pathophysiology, recog-nition, and management. Prog Cardiovasc Dis 1997; 40:65–76

152 Hollander JE. The management of cocaine-associated myo-cardial ischemia. N Engl J Med 1995; 333:1267–1272

153 Gay GR, Loper KA. The use of labetalol in the managementof cocaine crisis. Ann Emerg Med 1988; 17:282–283

154 Dusenberry SJ, Hicks MJ, Mariani PJ. Labetalol treatmentof cocaine toxicity [letter]. Ann Emerg Med 1987; 16:235

155 Spivey WH, Schoffstall JM, Kirkpatrick R, et al. Comparisonof labetalol, diazepam, and haloperidol for the treatment of

cocaine toxicity in a swine mode [abstract]. Ann Emerg Med1990; 19:467–468

156 Catravas JD, Waters IW. Acute cocaine intoxication in theconscious dog: studies on the mechanism of lethality. J Phar-macol Exp Ther 1981; 217:350–356

157 Boehrer JD, Moliterno DJ, Willard JE, et al. Influence oflabetalol on cocaine-induced coronary vasoconstriction inhumans. Am J Med 1993; 94:608–610

158 Negus BH, Willard JE, Hillis LD, et al. Alleviation ofcocaine-induced coronary vasoconstriction with intravenousverapamil. Am J Cardiol 1994; 73:510–513

159 Moore NA, Rees G, Sanger G, et al. Effect of L-typecalcium channel modulators on stimulant-induced hyperac-tivity. Neuropharmacology 1993; 32:719–720

160 Hollander JE, Carter WA, Hoffman RS. Use of phentol-amine for cocaine-induced myocardial ischemia [letter].N Engl J Med 1992; 327:361

161 Haas CE, LeBlanc JM, Haas CE, et al. Acute postoperativehypertension: a review of therapeutic options. Am J HealthSystem Pharm 2004; 61:1661–1673

162 Cheung AT. Exploring an optimum intra/postoperative man-agement strategy for acute hypertension in the cardiacsurgery patient. J Card Surg 2006; 21(suppl 1):S8–S14

163 Weant KA, Flynn JD, Smith KM. Postoperative hyperten-sion. Orthopedics 2004; 27:1159–1161

164 Kwak YL, Oh YJ, Bang SO, et al. Comparison of the effectsof nicardipine and sodium nitroprusside for control ofincreased blood pressure after coronary artery bypass graftsurgery. J Int Med Res 2004; 32:342–350

165 Halpern NA, Sladen RN, Goldberg JS, et al. Nicardipineinfusion for postoperative hypertension after surgery of thehead and neck. Crit Care Med 1990; 18:950–955

166 Halpern NA, Alicea M, Krakoff LR, et al. Postoperativehypertension: a prospective, placebo-controlled, random-ized, double-blind trial, with intravenous nicardipine hydro-chloride. Angiology 1990; 41:992–1004

167 Halpern NA, Goldberg M, Neely C, et al. Postoperativehypertension: a multicenter, prospective, randomized com-parison between intravenous nicardipine and sodium nitro-prusside. Crit Care Med 1992; 20:1637–1643

168 Wiest D. Esmolol: a review of its therapeutic efficacy andpharmacokinetic characteristics. Clin Pharmacokinet 1995;28:190–202

1962 Postgraduate Education Corner

Page 51: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Review Article

Perioperative Management of Chronic Heart Failure

Leanne Groban, MD*

John Butterworth, MD†

Heart failure (HF) is one of the few cardiac conditions that is increasing. Despite abetter understanding of how hormones and other signaling systems underlie thepathophysiology, and despite improved outcomes from pharmacologic therapy, manyHF patients receive no effective treatment. Patients with HF commonly require medicaldiagnosis and management in operating rooms and critical care units; thus anesthesi-ologists are obliged to remain up-to-date both with advances in outpatient (chronic)medical management and with inpatient treatments for acute exacerbations of HF.Accordingly, we reviewed angiotensin-converting enzyme inhibitors, angiotensinreceptor blockers, �-adrenergic receptor blockers, and aldosterone antagonists becausethese drugs prolong life and are included in current clinical practice guidelines fortreating patients with chronic HF. We also reviewed the implications of chronic HF forpatients undergoing surgery and anesthesia and discuss how best to provide intensivetreatment for acute exacerbations of symptoms, such as might be caused by excessiveintravascular volume, inappropriate drug “holidays,” or worsening of the underlyingcardiac disease.(Anesth Analg 2006;103:557–75)

Chronic heart failure (HF), a clinical syndrome inwhich abnormalities of ventricular function and neu-rohormonal regulation lead to pulmonary venouscongestion, exercise intolerance, and decreased lifeexpectancy, remains the one major cardiovascular(CV) disorder that has increased both in incidence andprevalence in recent years (1). Chronic HF affectsnearly five million persons in the United States, whereroughly 550,000 new cases are diagnosed annually.Currently, 1% of those 50–59 yr of age and 10% ofthose older than 80 yr have HF (2). Thus HF isprimarily a disease of the elderly, and its prevalencewill likely increase two- to threefold over the nextdecade as the median age of world populations in-creases (3). The increasingly prolonged survival ofpatients with various CV disorders that culminate inleft ventricular dysfunction (e.g., acute mortality aftermyocardial infarction has declined) adds to the HFepidemic. Treatment of HF costs the United States anestimated $38 billion annually (4), and it contributes to

approximately 250,000 deaths per year (5). Given therapid evolution of standard therapy and the frequencywith which chronic HF patients present to the operat-ing room and intensive care unit, anesthesiologists areobliged to know contemporary “best practices” tomake appropriate diagnostic and treatment choicesand appropriate judgments about the need for cardiacconsultations.

We review the medical management of chronic HF,focusing on the results of large-scale, randomizedclinical trials and on consensus guidelines publishedby the American College of Cardiology (ACC), theAmerican Heart Association (AHA), the Heart FailureSociety of America, and the European Society ofCardiology (6–8). These trials and guidelines empha-size chronic HF associated with left ventricular sys-tolic dysfunction; nevertheless, we will also discussHF with preserved systolic function (or diastolic HF).We also review the management of acutely decom-pensated HF. Finally, this review does not focus onthe acute HF that appears for the first time during orafter cardiac surgery, as the mechanisms and treat-ments of this condition are quite different fromchronic HF.

HF CLASSIFICATIONThe updated ACC/AHA guidelines for evaluating

and managing HF include a new, four-stage classifi-cation system emphasizing the progression of thedisease(Fig. 1). The new guidelines include patientswith “preclinical” stages of HF with the hope ofslowing (and perhaps reversing) progression of dis-ease. The staging system is meant to complement, notreplace, the widely used New York Heart Association(NYHA) classification, which is organized according

From the *Department of Anesthesiology, Wake Forest UniversitySchool of Medicine, Winston-Salem, North Carolina; †Departmentof Anesthesia, Indiana University School of Medicine, Indianapolis,Indiana.

Accepted for publication April 25, 2006.Supported, in part, by grant AG022598-01 from the National

Institute on Aging, the Dennis W. Jahnigen Career Developmentand Paul Beeson Scholars Award (K08-AG026764-01) and a MerckGeriatric Cardiology Award to Dr. Groban.

No reprints of this article will be available.Address correspondence to Leanne Groban, MD, Department of

Anesthesiology, Wake Forest University School of Medicine, Medi-cal Center Boulevard, Winston-Salem, NC 27157-1009. Addresse-mail to [email protected].

Copyright © 2006 International Anesthesia Research SocietyDOI: 10.1213/01.ane.0000226099.60493.d9

Vol. 103, No. 3, September 2006 557

Page 52: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

to severity of symptoms. The latter remains usefulbecause severity of symptoms has a robust correlationwith survival and quality of life (9). The ACC/AHAclassification system recognizes the progressive courseof HF and identifies those at risk, reinforcing the impor-tance of neurohormonal antagonism in an attempt toarrest disease progression.

HF may result from coronary artery disease, hyper-tension, valvular heart disease, or any of a long list ofcardiomyopathies, in which progression of the under-lying disease produces symptomatic or asymptomaticleft ventricular dysfunction (specifically, left ventricu-lar systolic dysfunction for the purposes of this dis-cussion). The neurohormonal responses to impairedcardiac performance (sympathetic stimulation, salt andwater retention, and vasoconstriction) are initiallyadaptive but over time become maladaptive, resultingin pulmonary congestion and excessive afterload. Theend result is a vicious cycle of increased and inefficientcardiac energy expenditure and worsened pump func-tion and tissue perfusion. The renal and peripheralcirculatory consequences to the neurohormonal re-sponses in HF provided the theoretical basis fortreatment with diuretics, vasodilators, and positiveinotropes (10,11).

Our understanding of treatment for patients withchronic HF changed in the 1990s when clinical trialsshowed that angiotensin-converting enzyme (ACE) in-hibitors (12,13) and angiotensin receptor blockers (ARB)(14,15), but not most other vasodilators (16), prolonged

survival. Similarly, certain �-adrenergic blockers, despitenegative inotropic effects, were found to improve mor-bidity and mortality in adequately powered, random-ized, clinical trials (17–19). More recently, small dosesof aldosterone antagonists added to conventionaltherapy for HF have reduced mortality in patientswith severe HF (20,21). These accumulated outcomeresults support evidence from basic investigationsshowing that angiotensin II is a growth factor as wellas a vasoconstrictor and that antagonizing this media-tor accomplishes more than mere hemodynamic im-provement (22). Growth factor actions of angiotensinII have been confirmed in models of inflammation,cancer, metabolic syndrome, and atherosclerosis. In arecent clinical study, increased angiotensin II receptordensity was associated with increased tumor angio-genesis and poorer survival in patients with ovariancancer (23). Overall, these data have promoted a shiftin focus from renal and circulatory processes towardcardiac remodeling as the underlying mechanism ofprogression in HF (24). Although it is not entirely clearhow the hemodynamic and neurohormonal factorsinteract to cause maladaptive cardiac remodelingand progression to HF, there is evidence to suggestthat increased energy expenditure, increased wallstress, altered calcium regulatory function of thesarcoplasmic reticulum, altered cardiac gene expres-sion, increased oxidative stress, myocyte necrosis,and apoptosis are involved (25) (Fig. 2).

Figure 1. Reprinted with permission from Hunt SA et al. ACC/AHA Guidelines for the Evaluation and Management ofChronic Heart Failure in the Adult: An Executive Summary. J Heart Lung Transplant 2003;21:189–203.

558 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 53: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

With these underlying pathophysiologic conceptsin mind, we will review the major classes of drugsthat are used to treat HF, emphasizing those drugsthat improve survival in chronic HF. However, eventhough the descriptions of each drug class are inde-pendent, clinical management of real patients willoften require regimens that include multiple drugs.This will become obvious in the descriptions of recentclinical trials where “conventional therapy” will in-clude 3 classes of drugs with the presence or absenceof the “newest” drug being the only variable understudy.

INHIBITORS OF THE RENIN-ANGIOTENSIN SYSTEMACE Inhibitors

Mechanism(s) of ActionACE inhibitors inhibit the protease that cleaves the

decapeptide angiotensin-I to form the octapeptideangiotensin-II. Because ACE also metabolizes brady-kinin, ACE inhibitors increase circulating and tissueconcentrations of bradykinin, which is thought tounderlie the side effects of these drugs, includingcough and angioedema (Fig. 3). ACE inhibitors haveseveral useful actions in chronic HF. They are potent

vasodilators because they decrease concentrations ofangiotensin-II and norepinephrine and increase con-centrations of bradykinin, nitric oxide (NO), and pros-tacyclin (26). They reduce the secretion of aldosteroneand antidiuretic hormone, thereby reducing salt andwater reabsorption by the kidney, and they promotebinding of angiotensin-I to receptors on nerve termi-nals, reducing norepinephrine release from sympa-thetic nerves. Within tissue, ACE inhibitors limit theproduction of angiotensin-II, attenuating angiotensin-II-mediated ventricular and vascular remodeling.

Clinical EvidenceThe abundant evidence supporting the benefits

from use of ACE inhibitors in chronic HF patients issummarized in Table 1. Initially, ACE inhibitors wereevaluated for treatment of symptomatic HF (theseclinical trials went by the acronyms of SOLVD,V-HeFT, and CONSENSUS). Patients with NYHAClass II–IV HF treated with ACE inhibitors had 16% to31% reduced risk of mortality. In later trials, ACEinhibitors also improved outcome for asymptomaticpatients with left ventricular systolic dysfunction in thefollowing categories: patients with ejection fractions (EF)

Figure 2. Pathophysiology of chronic heart failure. Remodeling stimuli, such as wall stress from either pressure overload orvolume overload, contribute to neurohormonal activation, oxidative stress, and cytokine activation, which lead to a complexseries of local effects including myocyte loss and alterations in the cardiac extracellular matrix, intracellular calciumregulation, and gene expression. In addition to the local events, systemic effects leading to LV dilation, myocyte hypertrophy,and LV remodeling contribute to the progression of heart failure. IL, interleukin; LV, left ventricle, TNF, tumor necrosis factor;SR sarcoplasmic reticulum; MHC, myosin heavy chain. Adapted with permission from Drexler H, Hasenfuss G. Physiologyof the normal and failing heart. In: Crawford MH, DiMarco JP, Paulus WJ, eds. Cardiology, 2nd ed. Philadelphia: Mosby(Elsevier), 2004.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 559

Page 54: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

�35% resulting from cardiomyopathy (27), patientswith EF �40% 2 wk after myocardial infarction (28),and patients presenting within the first 24 h of myo-cardial infarction regardless of EF (29). Results fromthe Heart Outcomes Prevention Evaluation (HOPE)study have further expanded the indications for ACEinhibitors to include prevention of new onset HF inasymptomatic, high-risk patients (30). In this trial ofpatients with either diabetes or peripheral vasculardisease and an additional atherosclerotic risk factor(but without HF or systolic dysfunction), ramipril(10 mg/day) reduced the new occurrence of HF by23% over a mean 4.5-yr treatment interval.

Together, these data expand the use of ACE inhibi-tors as first-line preventive therapy for a broad spec-trum of patients, including those with left ventricularsystolic dysfunction with or without symptoms (ClassB–D) and those high-risk patients with vascular dis-ease and/or diabetes, in addition to patients with thetraditional risk factors for coronary artery disease(Class A). Interestingly, retrospective data from theSOLVD and V-HeFT (Vasodilator Heart Failure trials)suggest that renin-angiotensin-aldosterone system(RAAS) inhibition, particularly with ACE inhibitors,may not be as effective in the African-American HFpatient as in Caucasians. In SOLVD, there was noethnic difference in the efficacy of enalapril for reduc-ing mortality and preventing the development of HF,but enalapril was more effective for Caucasians inreducing hospitalizations. Moreover, in V-HeFT-II,enalapril was more effective than the combination ofisosorbide dinitrate and hydralazine for Caucasians inreducing mortality but not in African-Americans. Datafrom the A-HeFT (African American Heart FailureTrial) showed a survival benefit in African AmericanHF patients treated with BiDil (n � 518), a fixed-dose

combination of isosorbide dinitrate (60 up to 120 mg)and hydralazine (112.5 up to 225 mg) in 3 divideddoses versus placebo (n � 532), added to standardbackground RAS blockade (31).

Nevertheless, one should only cautiously initiateACE inhibitor therapy in patients with low initialblood pressures (BP) (systolic blood pressures �80mm Hg), marked renal dysfunction (serum creatininelevels �3.0 mg/dL), serum potassium concentrations�5.5 mMol/L, renal artery stenosis, or left ventricularoutflow tract obstruction. ACE inhibitors are contra-indicated in patients who are pregnant, have a historyof porphyria, are in cardiogenic shock, or who havehad a severe reaction (e.g., angioedema or anuric renalfailure) to members of this drug class. As noted earlier(Fig. 1 and Table 1), ACE inhibitors are administeredboth to slow the progression of clinical HF throughACE inhibitor-mediated vasodilatory action and toinhibit the cellular mechanisms responsible for pro-gression of HF.

Perioperative ImplicationsAnesthetic drugs, surgical procedures, patient po-

sitioning on the operating table, and blood loss influ-ence the RAAS and the sympathetic nervous system(SNS) (32). Controversy remains whether chronic ACEinhibitor therapy should be continued or withdrawnpreoperatively. Patients treated with ACE-I are prone tohypotension with induction and maintenance of generalanesthesia, most likely as a result of intravascularvolume deficits and the inability of angiotensin-II tocounterbalance the usual anesthetic effects on the SNS(including increased venous pooling of blood, reducedcardiac output, and reduced arterial BP) (33–35). Ryck-waert and Colson (34) report a 22% incidence of severehypotension in patients who received ACE-I until the

Figure 3. Schematic of the renin-angiotensin-aldosterone system and site of action ofangiotensin-converting enzyme (ACE) in-hibitors (one slash), angiotensin receptorblockers (dotted slash), and aldosterone re-ceptor blockers (double slash). Adaptedwith permission from McMurray JJV, Pfef-fer MA, Swedberg K, Dzau VJ. Which in-hibitor of the renin-angiotensin systemshould be used in chronic heart failure andacute myocardial infarction? Circulation2004;110:3281–8.

560 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 55: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

day of surgery. Instability of BP and heart rate afterinduction of anesthesia was much the same in patientsreceiving chronic ACE inhibitor therapy regardless ofwhether there was left ventricular systolic dysfunc-tion. There are multiple case reports about hypoten-sion in patients treated with ACE inhibitors who arealso receiving general, spinal, epidural, or combinedgeneral-epidural techniques (36), and it remains un-clear whether any specific anesthetic technique ismore or less likely than others to show adverseinteractions in patients. Long-term ACE inhibitortreatment does not exaggerate the BP decrease as-sociated with spinal anesthesia, perhaps because

vasopressin and norepinephrine concentrations re-main sufficient to compensate for the inhibitedRAAS (37). Although temporary withdrawal of ACEinhibitors may prevent or attenuate intraoperativehypotension and hypovolemia, the recovery of RAScontrol on BP may be at the expense of impairedregional circulation. Boldt et al. (38) showed, in 88randomized cardiac surgical patients, that adminis-tration of IV enalapril after anesthetic inductionuntil commencement of cardiopulmonary bypass(CPB) resulted in lower levels of cardiac enzymerelease than clonidine, enoximone, or placebo. Periop-erative ACE-I administration may also protect the

Table 1. Selected Clinical Trials of Angiotensin-Converting Enzyme Inhibitors in Heart Failure (HF)

Trial Drug Population OutcomeHeart Failure

SOLVD (treat)* Enalapril NYHA Class II–III (EF�35%), Stage C

� Lower incidence of deathor hospitalization for HF inenalapril group vs. placebo(48% vs. 57%)

V-Heft II (153) Enalapril vs.hydralazine/isosorbide dinitrate

NYHA II–IV, EF �45%;Stage C

� At 2 yr, there was adecreased mortality withenalapril vs. hydralazine/isosorbide group (18% vs.28.2%). Mortality benefitfrom reduction in suddencardiac death.

CONSENSUS† Enalapril NYHA IV, Stage D � 31% decrease in mortalitywith enalapril vs. placebo� 50% decrease inprogressive HF death inenalapril vs. placebo

Asymptomatic LV Dysfunction

SOLVD(prevent)‡

Enalapril NYHA I, EF �35%, Stage B � At 37 months, combinedend-point of death or HFwas lower in enalapril groupcompared with placebo (30%vs. 39%). Fewerhospitalizations for enalaprilgroup (21% vs. 25% forplacebo).

SAVE (28) Captopril Post-MI, EF �40%, Stage B � 22% reduction inmortality with or withoutHF� 25% reduction in rate ofnonfatal MI

GISSI-3 Lisinopril, captopril Acute MI (with 36 h ofsymptoms), Stage B

� 7% reduction in mortalityat 30 days vs. placebo

ISIS-4 (29)

Asymptomatic High-Risk

HOPE (30) Ramipril History of DM, PVD, andcoronary risk factors,Stage A

� Significant reduction inmortality, major vascularevents, and development ofHF. Incidence of HF 9% vs.11.5% (placebo). Incidence ofMI, stroke, or CV-relateddeath 14% vs. 17.8% placebo.

Reprinted from Royster RL, Butterworth J, Groban L, Slaughter TF, Zvara DA. Cardiovascular pharmacology. In: Kaplan JA, ed., Cardiac Anesthesia, 5th ed. Philadelphia: WB Saunders (Elsevier),In Press.*Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. The SOLVD Investigators. N Engl J Med 1991;325:293–302.†Effects of enalapril on mortality in severe congestive heart failure. Results of the Cooperative North Scandinavian Enalapril Survival Study (CONSENSUS). N Engl J Med 1987;316:1429–35.‡Effect of enalapril on mortality and the development of heart failure in asymptomatic patients with reduced left ventricular ejection fractions. The SOLVD Investigators. N Engl J Med1992;327:685–91.NYHA � New York Heart Association; EF � ejection fraction; MI � myocardial infarction; DM � diabetes mellitus; PVD � peripheral vascular disease; CV � cardiovascular.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 561

Page 56: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

kidney. Aortocoronary bypass patients pretreatedwith captopril starting 2 days before surgery hadbetter preserved renal plasma flow and glomerularfiltration rate during CPB compared with placebo-treated patients (39). Moreover, patients undergoingaortic-abdominal surgery pretreated with a singledose of enalapril before anesthetic induction had asmaller reduction in cardiac output and glomerularfiltration rate with aortic clamping and a significantlygreater creatinine clearance on the first postoperativeday compared with the placebo group (40). Whetherthe organ-protective benefits of ACE inhibitors justifytheir routine prophylactic administration to patients atrisk requires further study.

There is overwhelming evidence of the benefit ofRAS modulation in nonsurgical settings; however, theevidence supporting continuing these medications un-til the time of surgery is less complete and lessconvincing (41–43). Nevertheless, although we recog-nize the potential for hypotension on induction ofanesthesia in patients chronically treated with an ACEinhibitor, on balance, we nevertheless suggest thatanesthesiologists continue the medication. Such briefepisodes of hypotension can usually be treated withmodest doses of sympathomimetics (e.g., ephedrine)or � adrenergic agonists (e.g., phenylephrine) andcareful expansion of intravascular volume (39). In therare event of hypotension refractory to these interven-tions either during noncardiac surgery or on weaningfrom CPB during cardiac surgery (44,45), the pre-sumed ACE-I-induced decrease in catecholamine re-sponsiveness can be managed with either small bo-luses (1–2 U) or an infusion of arginine vasopressin(4–6 U/h) (46). In the case of severe systemic hypoten-sion (presumed secondary to reduced renin secretion)during spinal or epidural anesthesia, � adrenergic stimu-lation with epinephrine (0.5–1 �g/min) may also beconsidered (47). Certainly, the problem may often beprevented by incremental administration of inductiondrugs and/or by selecting drugs less likely to causehypotension (e.g., ketamine or etomidate).

Because amiodarone can increase the incidence ofhypotension when combined with ACE inhibitors inanesthetized patients (48) and because ACE inhibitorscombined with aprotinin may lead to a greater pro-pensity for renal insufficiency after CPB (49)—werecognize that this may be a primary aprotinin effect(50)—hypovolemia and hypotension may pose anincreased risk to these patients. If one chooses todiscontinue ACE inhibitors preoperatively (e.g., for anasymptomatic HF patient without hypertension),there is no risk of rebound or other circulatory com-plications (51). The long-term benefits of ACE inhibi-tor therapy (e.g., on ventricular remodeling) will likelynot be harmed by brief drug holidays. However, incardiac surgery, Pigott et al. (52) found no reduction inthe incidence of hypotension on induction of anesthe-sia or in the need for vasoconstrictors after CPB whenACE inhibitors were omitted before surgery.

ARBsMechanism of ActionPlasma concentrations of angiotensin-II and aldoste-

rone may increase during chronic ACE inhibitor therapybecause of accumulation of substrate (angiotensin I) orbecause of increased production through non-ACE de-pendent pathways such as chymase. Moreover, non-ACE-generated angiotensin-II within the myocardiumcontributes to left ventricular remodeling and HFprogression through AT1 receptor effects. SelectiveAT1-blockers prevent angiotensin-II from directlycausing vasoconstriction, sodium retention, and re-lease of norepinephrine (Fig. 3). They also delay orprevent left ventricular hypertrophy and interstitialfibrosis (53). Angiotensin type-2 receptors (AT-2) andtheir actions, including NO release and vasodilation,remain unaffected by AT-1 receptor blockade (Fig. 3).The putative counter-regulatory role of AT-2 receptorsignaling in the heart (anti-growth and anti-fibroticeffects) and other effects that inhibit cell proliferate orpromote apoptosis are of questionable clinical impor-tance in the overall regulation of the RAS in HF (27).

Clinical EvidenceOutcome benefit from ARBs was first suggested in

the ELITE I trial, which showed, as a secondaryend-point, a significantly reduced risk of suddendeath with losartan (4.8%) compared with captopril(8.7%) (54), despite there being no between-groupdifferences in the primary end-points: renal dysfunc-tion or hypotension. The follow-up ELITE II trial(Table 2) had greater statistical power than ELITE I,but failed to confirm that losartan was superior tocaptopril in reducing mortality in older patients withHF (55). Moreover, in subgroup analyses, the ELITE IItrial patients receiving preexisting �-adrenergic block-ers tended to have less favorable outcomes withlosartan, as opposed to captopril. Two more trials,Valsartan in Heart Failure (Val-HeFT) and Candesar-tan in Heart Failure Assessment in Reduction ofMortality (CHARM), tested the hypothesis that ARBsplus conventional therapy (including �-adrenergicblockers, ACE inhibitors, and diuretics) for symptom-atic HF would provide additional clinical benefit. TheVal-HeFT study supports the use of valsartan inpatients with chronic HF who are intolerant to ACEinhibitors. However, among those patients who re-ceived ACE inhibitors and �-adrenergic blockers (93%of their patient population) there was a trend towardan increased risk of death or hospitalization whenvalsartan was added to standard treatment (14). Onthe other hand, the CHARM-Added trial (56) showedsafety with regard to use of candesartan in combina-tion with ACE inhibitors and �-adrenergic blockers(15% relative risk reduction in CV-related mortality orhospitalization). In patients intolerant to ACE (Alter-native group), the relative risk reduction in mortalityor hospitalization was 23%. Patients with left ventric-ular EF �40% not receiving ACE inhibition (Preserved

562 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 57: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

group) showed no difference in CV mortality and onlya small reduction in HF hospitalizations (57,58). In theCHARM-overall trial, candesartan use significantlyreduced both CV-related death and hospitalizations(relative risk reduction for CV death was 16%) (15).

Taken together, these studies show that ARBs aresuitable alternatives to ACE inhibitors for the treat-ment of patients with symptomatic HF when there areside effects to ACE inhibitors (e.g., persistent cough-ing, angioedema, hyperkalemia, or worsening renaldysfunction) or persistent hypertension despite ACEinhibitors and �-adrenergic blockers. The availableevidence is not convincing that patients with HFbenefit from the addition of ARBs to standard therapywith ACE-I and �-adrenergic blockers.

Perioperative ImplicationsAs is true with ACE inhibitors, patients chronically

treated with ARBs appear more prone to hypotensionwith induction of anesthesia (51,59,60) and are, wepresume, also more likely to require vasoconstrictorsduring and after separation from CPB (44) than pa-tients receiving other antihypertensive drugs and,perhaps, even compared with patients receiving ACEinhibitors (61). Moreover, patients receiving ARBs areless responsive to conventional vasopressors such asephedrine and phenylephrine (59), in part because ofan attenuated adrenergic responsiveness (62). Omis-sion of the ARB on the day of surgery will not likely

improve CV stability because these drugs have aprolonged duration of action (63,64); however, after adrug-free interval of at least 24 h, patients will havesignificantly fewer episodes of hypotension than thosewho continue to receive their ARB therapy (65). Va-sopressin and vasopressin analogs will treat intraop-erative hypotension refractory to conventional drugsin ARB-treated, anesthetized patients (46,66–68). Hy-potension during anesthetic induction may be accom-panied by bradycardia, particularly when vagotonicdrugs are used (e.g., sufentanil). Accordingly, we andothers advocate administering a prophylactic dose ofglycopyrrolate (0.2 mg) to elderly patients taking anARB on a chronic basis (69).

Aldosterone Receptor AntagonistsMechanisms of ActionAside from the “traditional” effects of mineralocor-

ticoid receptor blockade (natriuresis, diuresis, andpotassium retention) (70), beneficial nonrenal effectsof aldosterone antagonism include decreased myocar-dial collagen formation (71), increased myocardialnorepinephrine uptake, and decreased circulating nor-epinephrine levels (71), normalization of baroreceptorfunction, increased heart rate variability (72), reducedendothelial dysfunction, and increased basal vascularNO bioactivity (Fig. 3) (73).

Table 2. Angiotensin Receptor Blocker Trial in Heart Failure (HF)

Trial Drug Population OutcomeELITE II (55) Losartan 50 mg every day

Age �60 yr• Losartan was not better thancaptopril

NYHA II–IV • No difference in mortalityEF �40% • Losartan was better tolerated

• Losartan � beta blocker had worseoutcome

Val-HeFT (14) Valsartan 160 mg twice daily, orplacebo plus open label ACE-I(93%) NYHA II–IV • No difference in all-cause mortality

EF �40%• 13% significant difference incombined morbidity/mortality• HF hospital decrease 27%• Most benefit observed in ACE-Iintolerant patients (7% of studygroup) with 45% reduction incombined primary end-points

CHARM Candesartan 32 mg every day vs.placebo with or without open labelACE-I NYHA II–IV

Added (56) EF �40% • 15% relative risk reduction in all-cause mortality

Preserved (58) EF �40% • Mild reduction in HF-relatedhospitalizations

Alternative (57) EF �40% • 23% relative risk reduction in HF-related mortality or hospitalizations

Overall (15) ACE-Iintolerant

• Significant difference in all-causemortality

Reprinted from Royster RL, Butterworth J, Groban L, Slaughter TF, Zvara DA. Cardiovascular pharmacology. In: Kaplan JA, ed., Cardiac Anesthesia, 5th ed. Philadelphia: WB Saunders (Elsevier),In Press.ACE � angiotensin-converting enzyme; NYHA � New York Heart Association; EF � ejection fraction.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 563

Page 58: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Aldosterone receptor antagonists cause conservationof potassium, and hyperkalemia is a long-recognizedcomplication (74–76).

Clinical EvidenceTwo large clinical trials have demonstrated im-

proved outcomes with aldosterone receptor antago-nists in chronic HF. The Randomized AldactoneEvaluation Study (RALES) (20), including more than1600 symptomatic HF (e.g., Stage C, NYHA 3–4)patients, showed reduced mortality with spironolac-tone (26 mg/day) in combination with standardtherapy (ACE inhibitor, loop diuretic, and in somecases digoxin and/or �-adrenergic blocker). Regard-less of age, gender, or HF etiology, the treatmentgroup experienced a 30% reduction in all-cause mor-tality and in CV mortality compared with standardtherapy. Because �-adrenergic blockers were usedinconsistently in the RALES study (10%–20%), therelative place of aldosterone antagonists in contempo-rary management of HF remained unclear. Moreover,in reports subsequent to the RALES study there was amarked increase in hospital admissions and deathsrelated to hyperkalemia associated with spironolac-tone (74). Some of the concerns were addressed in theEplerenone Post-acute Myocardial Infarction HeartFailure Efficacy and Survival Study (EPHESUS).Eplerenone is a new aldosterone antagonist that lackssome of spironolactone’s common side effects (mostnotably gynecomastia) (77). The study was conductedin more than 6600 patients with symptomatic HF 3 to14 days post-myocardial infarction, and showedthat eplerenone (25 to 50 mg/daily), in combinationwith ACE inhibitor, loop diuretic, and �-adrenergicblocker, significantly reduced all cause mortality,death from CV causes, and hospitalization for CVevents (21). Large-scale randomized controlled trialsin class A and B HF patients are lacking. Pilot datashow that aldosterone inhibition improves endothelialfunction (73), exercise tolerance, and EF (78) andattenuates collagen formation. Laboratory studieshave shown that even in the presence of ACE inhibi-tors and AT-1 receptor blockade, activation of aldoste-rone synthetase (“aldosterone escape”) in the heartand vasculature leads to myocardial hypertrophy,myocardial and vascular collagen production, endo-thelial dysfunction, and increased sodium retention(79,80). Laboratory and clinical data suggest that aldo-sterone antagonists may provide organ protection(71,81). Thus, we suspect that the indications foraldosterone receptor blockers may be widened toinclude patients with asymptomatic systolic left ven-tricular dysfunction.

Perioperative ImplicationsAlthough we doubt that doses of aldosterone recep-

tor antagonists used for HF treatment contribute toanesthetic-induced hypotension, there is no doubt that

these drugs may cause life-threatening hyperkalemia.The risk of hyperkalemia is increased when aldoste-rone antagonists are administered in combinationwith other RAS blockers, preexisting renal insuffi-ciency, diabetes, or anemia (74–76). Thus, intraopera-tive measurement of serum potassium would seemprudent when these conditions are present, particu-larly in the event of red cell transfusion.

Beta-Adrenergic Receptor AntagonistsMechanism of ActionIn chronic HF, the beneficial effects of �-adrenergic

receptor blockade include improved systolic functionand myocardial energetics and reversal of pathologicremodeling. A shift in substrate utilization from freefatty acids to glucose (a more efficient fuel duringmyocardial ischemia) may partly explain the im-proved energetics and mechanics (82). �-blockadealso tends to offset the effects of neurohumoralactivation, a central feature of both chronic HF and ofmajor surgery with general anesthesia (83,84). Chronicneurohumoral activation leads to adverse cardiac re-modeling. Heart rate, a major determinant of myocar-dial oxygen consumption, is reduced by �1-receptorblockade.

Systolic dysfunction of individual myocytes is as-sociated with up-regulation of gene expression fornatriuretic peptides and fetal-like �-myosin heavy chainand increased expression of the cardiac sarcoplasmic-endoplasmic reticulum calcium uptake pump (SERCA2)and �-myosin heavy chain (the more efficient, faster,adult isoform) (85). �-blockade reverses these changes ingene expression and concurrently improves left ventric-ular function (86).

�-adrenergic blockade may also limit the disturbedexcitation-contraction coupling and predisposition toventricular arrhythmias associated with HF. In animalmodels of HF, the increases in L-type calcium currentsand in cytosolic calcium concentrations that occur inresponse to �-adrenergic surges often result in ven-tricular arrhythmias and sudden death (87). Theseeffects are likely the result of excess activation ofintracellular �-adrenergic mediated pathways viacAMP and protein kinase A (PKA), ultimately leadingto an “excessive phosphorylation” state.

As noted earlier, the SNS is chronically activated inthe failing heart with reduced cardiac output. In thissetting, excitation-contraction coupling becomesmaladaptive because of “leaky” Ca2� from the sarco-plasmic reticulum (SR) (Fig. 4). Protein kinase A (PKA)-hyperphosphorylated RYR2 channels cause a diastolicSR Ca2� leak that, together with reduced SERCA2-mediated SR Ca2� uptake (resulting from PKA-hyperphosphorylated phospholamban that inhibitsSERCA2a), depletes SR Ca2� and leads to contractiledysfunction of cardiac muscle (88). This depletion of SRCa2� stores may explain, in part, the reduced contractil-ity and the predisposition to ventricular arrhythmias of

564 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 59: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

cardiac muscle in HF (89). Interestingly, studies in ani-mal models of HF show that chronic �-blockade mayreverse the PKA hyperphosphorylated state and restorenormal structure and function of the RYR2 Ca2� releasechannel (90). In studies after human cardiac transplan-tation, �-adrenergic blockade restored RYR2 function,phosphorylation, and levels of the binding proteinstoward baseline, improving ventricular compliance andresponses to �-adrenergic agonists (91). Besides normal-izing the calcium leak, the benefit of �-adrenergic block-ade in HF patients may also include a decrease incalcium-dependent apoptosis and a metabolic effect pro-moting improved work efficiency (92). In a dog model ofHF, �-adrenergic blockade also reduced myocyte apo-ptosis (93). Thus, chronic �-adrenergic blockade reduces

the harmful effects of excessive SNS stimulation of theheart and reverses left ventricular remodeling.

Clinical EvidenceFor many years, �-adrenergic blockers were rarely

given to patients with HF because of the perceivedrisk of decompensation from their negative inotropiceffects. However, data from both human and animalstudies have shown that �-adrenergic blockers im-prove energetics and ventricular function and reversepathologic remodeling. Although these beneficial ef-fects may take 3 mo or more to manifest, they havetranslated into improved outcomes (reduced deathsand hospitalizations) in patients with HF. This ap-pears NOT to be a drug class effect, as not all�-adrenergic blockers improve outcomes in HF. The

Figure 4. Effects of inotropic therapy on intracellular calcium handling in cardiac myocytes. Many sarcolemmal receptorsaffect calcium handling in cardiac myocytes. Agonists through G proteins increase adenyl cyclase (AC) activity, which resultsin cAMP production. This results in activation of protein kinase A (PKA), which leads to phosphorylation of L-type calciumchannels, phospholamban, and troponin I, ultimately leading to a greater release of calcium from the sarcoplasmic reticulum(SR) and faster relaxation. Digoxin inhibits Na�/K� ATPase pump, which increases intracellular Na�. This results in increasein intracellular Ca2� via Na�/Ca2� exchanger, which leads to enhanced Ca2� loading of SR and increase in Ca2� release.Phosphodiesterase inhibitors (PDEI) block breakdown of cAMP, which increases its intracellular level and activates PKA.Calcium sensitizers, e.g., levosimendan, increase sensitivity of myofilaments to Ca2�, enhancing myofilament activation forany concentration of Ca2�. PDE � phosphodiesterase; PKC � protein kinase C. Reprinted with permission from StevensonLW. Clinical use of inotropic therapy for heart failure: looking backward or forward? Part I: Inotropic infusions duringhospitalization. Circulation 2003;108:367–72. (Fig. 1, page 369).

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 565

Page 60: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

available randomized trials show that metoprololCR/XL, bisoprolol, and carvedilol (in conjunctionwith ACE inhibitors) reduce morbidity (hospitaliza-tions) in symptomatic, Stage C and D (not in cardio-genic shock) HF patients (NYHA II–IV class) (Table 3)(18,19,94,95). Although �-adrenergic blocker therapyis recommended for asymptomatic HF patients (StageA and B), there is no evidence from randomized trialsto support this apparently widespread practice (96). Inclinical trials, �-adrenergic blockers are initiated insmall doses and increased progressively as toleratedby the patient. The goal in clinical management is toadminister the doses shown to be effective for pro-longing life in clinical trials, not to decrease heart rateby an arbitrary increment or to an arbitrary targetvalue.

�-adrenergic blockers are classified as being first-,second-, or third-generation based on specific pharma-cologic properties. First-generation drugs, such aspropranolol and timolol, block both �1 and �2 adre-noreceptors, are considered nonselective, have noancillary properties, and are not recommended for HFpatients. Second-generation drugs, such as metopro-lol, bisoprolol, and atenolol, are relatively specific forthe �1 adrenoreceptor subtype but lack additionalmechanisms of CV activity. Third-generation drugs,such as bucindolol, carvedilol, and labetalol, blockboth �1 and �2 adrenoreceptors and have vasodilatoryand other ancillary properties. Specifically, labetaloland carvedilol produce vasodilation by �1 adrenore-ceptor antagonism, whereas bucindolol produces mildvasodilation through a cyclic guanosine monophos-phate (cGMP)-mediated mechanism. Carvedilol in-creases insulin sensitivity (97) and has antioxidanteffects (98) and an affinity for �3 adrenoreceptors(99,100). �3 adrenoreceptors are up-regulated in HF, andtheir activation is thought to decrease contractilitythrough a NO and cGMP-mediated pathway (101–104).Although it is not clear whether these ancillary prop-erties of the third-generation �-adrenergic blocker,carvedilol, translate into better outcomes as comparedwith second generation drugs, findings from theCarvedilol or Metoprolol European Trial (COMET)

suggest that carvedilol may be more effective thanother �-adrenergic blockers. COMET compared carve-dilol (25 mg twice daily) to metoprolol tartrate (50 mgtwice daily) in symptomatic patients with EF �35%and demonstrated that carvedilol reduced the risk ofdeath relative to metoprolol (all-cause mortality riskreduction: 17%; P � 0.0017 and CV death risk reduc-tion: 20%; P � 0.00004) (105). The superiority ofcarvedilol over metoprolol could reflect the impor-tance of carvedilol’s ancillary effects, pharmacody-namic (e.g., half-life) differences or other, as yetunknown, differences (106). The COMET study didNOT use the long-acting metoprolol CR-XL butinstead used “conventional” metoprolol. The BESTtrial showed that not all �-adrenergic blockers im-prove outcome in HF. Bucindolol showed no benefitcompared to placebo (95). Whether the selective �1-specific drugs bisoprolol and metoprolol CR/XL exertsimilar clinical benefits to carvedilol remains unclear.Nonetheless, based on the results of the COMETstudy, carvedilol is preferred to conventional meto-prolol, but not metoprolol CR/XL, for HF treatment.

Most guidelines now include long-term �-adrenergicblockade for Stage B–D HF patients, except for patientswith continuing decompensation (e.g., requiring IV ino-tropes or vasodilators), to limit disease progression andreduce mortality (Fig. 1, Table 4). Despite concerns aboutinhibition of hypoglycemic symptoms, �-adrenergicblockers are advocated for diabetics with HF. There isstrong evidence that the benefits from triple therapyincluding �-adrenergic blockers, ACE-I, and aldosteroneantagonists are additive (Fig. 5) (107).

Perioperative ImplicationsAlthough there is a lack of large clinical trials of

RAAS modulation in the perioperative period, such isnot the case for �-adrenergic blockers. Randomizedclinical trials show that these drugs should be given toprevent ischemic events and arrhythmias in high-riskcardiac patients with ischemia, arrhythmias, or hyper-tension or a history of these conditions and to patientswith ischemia in perioperative testing submitted fornoncardiac surgery (particularly vascular surgery)

Table 3. Large-Scale Placebo-Controlled Mortality Trials of Beta Blockade in Heart Failure (HF)

Patients(n)

TargetDose (mg)

Effect on all cause

Trial Drug HF Severity Mortality HospitalizationUS Carvedilol (94) Carvedilol NYHA II–III 1094 6.25–50 bid 265% 227%CIBIS-II (18) Bisoprolol EF �35; NYHA III–IV 2647 10 qd 234% 220%MERIT-HF* Metoprolol CR/XL EF �40; NYHA II–IV 3991 200 qd 234% 218%BEST (95) Bucindolol EF �35; NYHA III–IV 2708 50–100 bid NS 2 8%COPERNICUS† Carvedilol EF �25; NYHA IV 2289 25 bid 235% 220%Reprinted from Royster RL, Butterworth J, Groban L, Slaughter TF, Zvara DA. Cardiovascular pharmacology. In: Kaplan JA, ed., Cardiac Anesthesia, 5th ed. Philadelphia: WB Saunders (Elsevier),2006:213–80.*Effects of controlled-release metoprolol on total mortality, hospitalization, and well-being in patients with heart failure: the Metoprolol CR/XL Randomized Intervention Trial in congestive heartfailure (MERIT-HF). MERIT-HF Study Group. JAMA 2000;283:1295–302.†Effect of carvedilol on the morbidity of patients with severe chronic heart failure; results of the carvedilol prospective randomized cumulative survival (COPERNICUS) study. Circulation2002;106:2194–9.NYHA � New York Heart Association; EF � ejection fraction.

566 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 61: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

(108). Also, �-adrenergic blockers are indicated forthe treatment of perioperative hypertension, isch-emia, and arrhythmias identified preoperativelyand previously untreated (108). Perioperative�-adrenergic blocker therapy in high-risk patients isunderused (109). Nevertheless, whether �-adrenergicblockers should be newly initiated before surgerysolely for management of HF remains highly specula-tive. Withdrawal of �-adrenergic blocker therapy frompatients who have received it chronically may beparticularly dangerous (108). Recent data also suggestthat while initiating �-adrenergic blocker therapy maybe highly advantageous for some surgical patientpopulations, it may be considerably less advantageous(perhaps even deleterious) for other patient popula-tions (110).

Adjunctive DrugsIn addition to ACE inhibitors and �-adrenergic

blockers, diuretics and digoxin are often prescribedfor patients with left ventricular systolic dysfunctionand symptomatic HF. Diuretics provide rapid symp-tomatic relief of HF in the acute setting, and theymaximize the benefits from ACE-I and �-adrenergicblockers, which are dependent on minimization of

excessive intravascular volume (16). Moreover, olderhypertensive patients who use diuretics in combina-tion with �-adrenergic blockers have lower meanpulse pressures as compared with those patients re-ceiving �-adrenergic blockers alone (111). Widenedpulse pressure is an independent predictor of adverseCV outcomes in older persons (112). Diuretics con-tinue to have a role in the outpatient management ofHF in conjunction with ACE-I, �-adrenergic blockers,and (in some cases) aldosterone antagonists eventhough no randomized controlled trials have demon-strated a survival benefit from diuretics in HF. Impor-tantly, hospitalization or death from worsening HFwere significantly more frequent in HF patients re-ceiving non-potassium-sparing diuretics than thosenot receiving diuretics (relative risk � 1.31, 95%confidence interval, 1.09–1.57) in a large post hocreview of data from SOLVD (Studies of Left Ventric-ular Dysfunction) patients (113).

Digoxin continues to be useful for patients withHF and left ventricular systolic dysfunction whoremain symptomatic despite receiving ACE inhibi-tor, �-adrenergic blocker, and diuretic. Digoxin is theonly positive inotropic drug that does not increase

Table 4. Diastolic Heart Failure Management

Goal Management Strategy Drugs/Recommended DosesReduce the congestive state Salt restriction �2 g of sodium/day

Diuretics (avoid reductions in CO) Furosemide, 10–120 mgHydrochlorothiazide, 12.5–25 mg

ACE inhibitors Enalapril, 2.5–40 mgLisinopril, 10–40 mg

Angiotensin II-receptor blockers Candesartan, 4–32 mgLosartan, 25–100 mg

Target underlying causeControl hypertension Antihypertensive drugs (�130/80) Beta blockers, ACE inhibitors,

AII receptor blockersaccording to publishedguidelines

Restore sinus rhythm Cardioversion of atrial fibrillationAV-sequential pacing

Prevent tachycardia Beta-adrenergic blockers, calciumchannel blockers

Prevent/treat ischemia Morphine, nitrates, oxygen, aspirinangioplasty or revascularization?

Atenolol, 12.5–100 mg;

Metoprolol 25–100 mg;Diltiazem, 120–540 mg

Treat aortic stenosis Aortic valve replacementTarget underlying

mechanisms(theoretical)

Promote regression ofhypertropy and preventmyocardial fibrosis

Renin-angiotensin axis blockade Enalapril, 2.5–40 mg

Lisinopril, 10–40 mgCaptopril, 25–150 mgCandesartan, 4–32 mgLosartan, 50–100 mgSpironolactone, 25–75 mgEplerenone, 25–50 mg

Reprinted from Royster RL, Butterworth J, Groban L, Slaughter TF, Zvara DA. Cardiovascular pharmacology. In: Kaplan JA, ed., Cardiac Anesthesia, 5th ed. Philadelphia: WB Saunders (Elsevier),In Press.CO � cardiac output; ACE � angiotensin-converting enzyme; AV � atrial-ventricular.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 567

Page 62: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

mortality in chronic HF (Fig. 4). The Digitalis Investi-gators Group (DIG) trial (114), enrolling more than6500 patients with an average follow-up of 37 mo,showed that digoxin reduced the incidence of HFexacerbations but had no effect on survival. Patientswith mildly symptomatic chronic HF, who wererandomized to digoxin withdrawal (PROVED andRADIANCE trials), had an increased likelihood of anacute exacerbation compared with those who contin-ued to receive digoxin (115,116). On the other hand,doubling the dose of digoxin from 0.125 to 0.25 mg dailyprovided no significant benefit in terms of exercisetolerance or ventricular function, suggesting that dosesof digoxin should be kept small (117).

Perioperative ImplicationsChronic treatment with diuretics can lead to hypo-

volemia and an imbalance of electrolytes, particularlyhypokalemia. These side effects are most common inthe elderly (118). Chronic diuretic therapy can lead tohypotension and arrhythmias (119) during anesthesia.However, chronic use of diuretics for the managementof HF has not been associated with perioperative CVdeath (within 30 days of surgery) in emergency andurgent surgical patients (120). In contrast, complica-tions of digoxin therapy can be life-threatening andoften difficult to diagnose and treat, given digoxin’snarrow therapeutic index. Aggravating conditionsthat predispose to digoxin toxicity include hypomag-nesemia, hypercalcemia, and hypokalemia, all ofwhich may occur during the perioperative period.Treatment of digoxin toxicity, which often manifestsas nausea, arrhythmias, and visual symptoms consistsof correcting any underlying electrolyte imbalances,administering antiarrhythmic drugs (most commonlyphenytoin), and in refractory cases, commercially pre-pared antibodies to digoxin (e.g., digoxin-specific

Fab (Digibind; Glaxo-SmithKline, Research TrianglePark, NC), a mixture of antidigoxin Fab fragmentsprepared from sheep sera). Despite continuing con-cerns about digoxin toxicity, perioperative discon-tinuation of digoxin therapy remains controversial.After adjustment for the confounding effect of HF,Sear et al. (120) report that digoxin therapy wasassociated with an increased cardiac risk in urgent andemergent surgical patients. Given that rate andrhythm control and positive inotropy can be achievedwith other drugs with shorter half-lives and lesstoxicity, we tend to discontinue digoxin in elderlysurgical patients where age-related alterations in drugdistribution and excretion may make toxicity increas-ingly likely (121).

PHARMACOLOGIC TREATMENT OF DIASTOLIC HFAbnormal diastolic ventricular function is present

in nearly all patients with symptomatic HF (122). Asmany as one in three patients presenting with clinicalsigns of chronic HF have a normal or near-normal EF(�40%). Although the prognosis of patients withisolated diastolic HF is better than for those withsystolic HF (5%–8% versus 10%–15% annual mortal-ity), the complication rate is the same (123). The 1-yrreadmission rate for patients with isolated diastolicHF approaches 50% (124).

Large randomized trials have led to the treatmentguidelines for systolic HF; however, there are few com-pleted randomized, double-blind, placebo-controlled,multicenter trials performed in patients with diastolicHF. The CHARM-Preserved Trial (58) data of 3023patients indicate that treatment with the ARB cande-sartan reduces hospitalization rates but does not altermortality in patients with diastolic HF. Findings fromthe I-PRESERVE (Irbesartan in HF with preserved

Figure 5. Lives saved over 2 yr. Effect of triple therapy with an angiotensin-converting enzyme (ACE) inhibitor, �-adrenergicblocker, and aldosterone antagonist. Based on SOLVD, MERIT, and EPHESUS (for NYHA II) and CONSENSUS�COPERNICUS, and RALES (for NYHA III/IV). Reprinted with permission from Cleland JG, Coletta AP, Nikitin N, Louis A,Clark A. Update of clinical trials from the American College of Cardiology 2003. EPHESUS, SPORTIF-III, ASCOT,COMPANION, UK-PACE and T-wave alternans. Eur J Heart Fail 2003;5:391–8.

568 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 63: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

systolic function) (125) trial of more than 4000 subjectswill likely provide conclusive data regarding theprimary end-point of death and the role of ARBblockade in the management of diastolic HF. Datafrom the Seniors trial (126) of nebivolol in 2128 HFpatients, of whom 752 had diastolic HF (EF defined as�35%), suggest that �-adrenergic blockade is equallybeneficial in patients with diastolic as with systolicHF. Preliminary findings from continuing studiessuggest that aldosterone antagonists may also im-prove exercise tolerance and quality of life in patientswith diastolic HF (127,128). However, until validationfrom adequately powered, randomized controlled tri-als becomes available, the contemporary treatment ofchronic diastolic HF remains empiric (Table 4).

MANAGEMENT OF ACUTE EXACERBATIONSOF CHRONIC HF

Patients with chronic HF may experience episodesof acutely decompensated HF, heralded by the classicsymptoms of dyspnea or fatigue. These patients willrequire all the standard medications as outlined inprevious sections (except for perhaps holding ACE-Iwhen systolic BP �80 mm Hg), and may also requireinfusions of vasodilators or positive inotropic drugs(129) (Table 5).

IV vasodilators have long been used to treat thesymptoms of low cardiac output in patients withdecompensated chronic HF. In general, vasodilatorsreduce ventricular filling pressures and systemic vas-cular resistance while increasing stroke volume andcardiac output. Nitroglycerin is commonly used for

this purpose and has been studied in numerous clini-cal trials (129). In addition, recombinant brain natri-uretic peptide (BNP) has received regulatory approvalas a drug (nesiritide), indicated for patients with acuteHF and dyspnea. Nesiritide binds to A- and B-typenatriuretic peptide receptors on endothelial and vas-cular smooth muscle cells. It produces venous andarterial dilation, with subsequent reductions in pre-load and afterload, through increasing cGMP. Nesirit-ide does not increase heart rate, and has no effect oncardiac inotropy. Nesiritide exerts diuretic and natri-uretic effects and causes coronary vasodilation. It hasa rapid onset of action with a distribution half-life of 2min and a terminal elimination half-life of 18 min.Onset of the drug’s effects is later than would bepredicted based on its pharmacokinetic parameters.For example, with an initial loading dose and mainte-nance infusion, only 60% of the reduction in pulmo-nary wedge pressure that will be measured at 3 h isachieved 15 min after the bolus dose (130). Clinicaleffects have also been observed to persist longer thanwould be anticipated (based on drug levels) after thedrug is discontinued.

Nesiritide is metabolized by three mechanisms:endocytotic internalization by its surface receptor,hydrolysis by neutral endopeptidase, and renal excre-tion (minor role) (131). When initiated in the periop-erative setting (e.g., post-CPB), a starting infusiondose of 0.005 �g � kg�1 � min�1, without a bolus, isrecommended to avoid hypotension in patients withincreased filling pressures and low systemic vascularresistance (�800 dyne � s � cm�5) who might also bereceiving ACE-I and �-adrenergic blockers. Studies

Table 5. Management of Decompensated Heart Failure

Class Drugs Infusions Side EffectsDiuretics Furosemide 5–20 mg/h Hyponatremia, hypokalemia,

hypomagnesemia; predisposespatients to toxicity of cardiacglycosides

Bumetanide 2–5 mg slow infusionVasodilators Nitroglycerin 10–200 �g/min Hypotension, nausea, headaches,

tachycardia, tolerance

Nitroprusside 0.1–5 �g � kg�1 � min�1Hypotension, nausea, headaches,

thiocyanate poisoningInotropes Dobutamine 2.5–15 �g � kg�1 � min�1

load, 50 �g/kg(eliminate load ifsystolic blood pressure�100 mm Hg) 0.375–0.75 �g � kg�1 � min�1

Tolerance (beta receptor down-regulation) hypotension,supraventricular and ventriculartachyarrhythmias

MilrinoneLevosimendan* 12 �g/kg over 10 min

(load); 0.1 �g �kg�1 � min�1

Dose-related increase in heart rate

Vasodilator/Diuretic/Natriuretic

Nesiritide 2 �g/kg load; 0.01�g � kg�1 � min�1

0.005–0.03 �g �kg�1 � min�1 (preferredin perioperative setting)

Hypotension, nausea

*Used for treatment of acute, decompensated heart failure in Europe.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 569

Page 64: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

have shown that nesiritide reduces symptoms of acutedecompensated HF similarly to nitroglycerin, includ-ing a reduction of pulmonary artery pressure, withoutdevelopment of acute tolerance (132). In early studies,patients receiving nesiritide experienced fewer ad-verse events than those receiving nitroglycerin (133).Compared with dobutamine, nesiritide was associatedwith fewer instances of ventricular tachycardia orcardiac arrest (134). In the ADHERE registry (135) ofmore than 65,000 episodes of acute decompensatedHF, treatment with either nesiritide or a vasodilatorwas associated with a 0.59 odds ratio for mortalitycompared with either milrinone or dobutamine. Re-cent data, however, suggest that not only may nesirit-ide not offer a compelling safety advantage, it mayalso be associated with an increased incidence ofadverse side effects, including renal failure and mor-tality, when administered to patients with acutelydecompensated chronic HF (136,137). These publica-tions prompted the Food and Drug Administration toconvene an expert panel, which made several recom-mendations, including that nesiritide be used only forhospitalized patients with acute decompensated HFand that the drug not be used to enhance diuresis or to“protect” the kidneys (138).

Clinical trials showed that chronic treatment withpositive inotropes such as inamrinone and milrinoneled to increased mortality (139–141). Nevertheless,positive inotropic drugs, principally dobutamine ormilrinone, have long been used to treat decompen-sated HF (Fig. 4). There is a lack of data supportingtheir discretionary administration (142), e.g., on amonthly schedule to patients awaiting cardiac trans-plantation to avoid the need for ventricular assistdevices. Levosimendan, a new cAMP-independentpositive inotrope, may prove to have no negativeoutcome effects when used to treat acute decompen-sation of chronic HF (143). Levosimendan acts byincreasing myocyte sensitivity to calcium via stabiliz-ing the calcium-bound conformation of troponin C(Fig. 5). Levosimendan also opens KATP channels invascular smooth muscle inducing vasodilation and incardiac muscle, where these channels may protectagainst ischemia (144). When compared with dobut-amine, levosimendan reduced 1-mo mortality (andreduced mortality compared with placebo at 14 days(145). Calcium sensitivity is increased during systolewithout causing calcium overload during diastole.This results in enhanced inotropic performance andpreserved diastolic performance.

When drug treatment proves unsuccessful, HFpatients may require invasive therapy, includingventricular assist devices, resynchronization withbiventricular pacing, coronary bypass with or with-out surgical remodeling, or even cardiac orthotopictransplantation (146). These important modalities arebeyond the scope of this review.

Current Clinical PracticeDiagnosisFor most patients, the diagnosis of HF will have

been made long before they arrive for surgery orintensive care. Current guidelines provide a helpfulframework by which primary care physicians andcardiologists can make the appropriate diagnoses andfollow the disease process over time (6). On the otherhand, how does a perioperative physician determinequickly, conveniently, and inexpensively whether adyspneic patient’s symptoms are the result of new orworsening HF, lung disease, or a combination of thetwo? Clearly the issue can be settled using the medicalhistory and physical examination, electrocardiogram,echocardiogram, chest radiograph, and consultationwith either a pulmonary medicine specialist or cardi-ologist. On the other hand, measurements of BNP inblood are widely used to help triage patients present-ing with acute dyspnea (147). Taken together withphysical examination and history, if the BNP is �100pg/mL, then HF is highly unlikely; negative predictivevalue, 90%, and if the BNP level is �500 pg/mL, then HFis highly likely, positive predictive value is 90%. For BNPlevels of 100–500 pg/mL, one must consider whether thebaseline is increased as a result of advanced age, under-lying stable left ventricular dysfunction, right ventricularfailure secondary to pulmonary hypertension or acutepulmonary embolism (148,149).

TreatmentCurrent guidelines begin pharmacotherapy of HF

with primary prevention of left ventricular dysfunc-tion (6,7) (Fig. 1). Because hypertension and coronaryartery disease are leading primary causes of leftventricular dysfunction, adequate control of bothhypertension (according to the Joint NationalCommittee-7 guidelines) (150) and hypercholesterol-emia has been endorsed after encouraging results inprevention trials (151). ACE inhibitors, and possibly�-adrenergic blockers, should be initiated in diabetic,hypertensive, and hypercholesterolemia patients(AHA/ACC, Stage A HF) who are at increased risk forCV events, despite normal contractile function, toreduce the onset of new HF (HOPE trial) (30). Inpatients with asymptomatic left ventricular dysfunc-tion (EF � 40%) (Stage B), treatment with ACE inhibi-tors and �-adrenergic blockers can blunt the diseaseprogression. In the symptomatic HF patient (Stage C),diuretics are titrated to relieve symptoms of pulmo-nary congestion and peripheral edema and to re-store a normal state of intravascular volume (152).ACE inhibitors and �-adrenergic blockers are rec-ommended to blunt disease progression. Althoughdigoxin has no effect on patient survival, it may beconsidered in Stage C if the patient remains symptom-atic despite adequate doses of ACE inhibitors anddiuretics. An alternative for patients (particularlyAfrican-American patients) with systolic dysfunction

570 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 65: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

and contraindications, intolerance, or unresponsive-ness to ACE inhibitors or ARBs is isosorbide dinitratethree times a day in combination with hydralazinethree times a day (153) or BiDil (fixed dose combina-tion of isosorbide dinitrate and hydralazine hydro-chloride (A-HeFT trial) (31). The use of an NO donor(isosorbide) in HF heralds the new suggestion that“NO balance” may be important in the pathophysiol-ogy of HF (154).

In general, the primary treatment objectives forStages A–C HF are: 1) improved quality of life, 2)reduced morbidity, and 3) reduced mortality. At thistime, the most important way to improve long-termoutcome is through inhibiting disease progression bycounteracting neurohormonal effects. Pharmacologictherapy in patients with severe, decompensated HF(Stage D) is based on hemodynamic status. Symptom-atic treatment with diuretics, vasodilators, and, in pallia-tive circumstances, IV inotropic infusions is added to“standard” treatment. Finally, some of these patientsmay require device therapies or surgical procedures,such as cardiac transplants.

What is an anesthesiologist to do when faced witha patient with Stage D or decompensated Stage C HFwho requires emergency surgery? If tracheal intuba-tion and positive pressure ventilation are needed tomanage pulmonary edema, then there is little reasonto select a regional anesthesia technique. When fea-sible (this will be rare because these patients oftencannot lie flat on the operating table), regional nerveblock techniques, rather than general anesthesia orneuroaxial block techniques, may avoid intraoperativecrystalloid infusions. There is no evidence basis bywhich to select either an induction or a maintenanceanesthetic drug in these patients. We have successfullyused most IV induction drugs in these patients (in-cluding thiopental, propofol, ketamine, etomidate, mi-dazolam, and diazepam) and have seen no obviousreason to recommend any one of them over the others.Similarly, while many authors advocate maintaininganesthesia in these very sick patients using benzodi-azepines and opioids, our usual practice is to maintainanesthesia with inhaled anesthetics. We find intraop-erative fluid and medical management considerablymore challenging than anesthetic choice in these pa-tients. Accordingly, when HF patients must undergomajor surgery, we suggest invasive arterial BP moni-toring and transesophageal echocardiography (TEE)to help guide intraoperative decision-making. TEE isespecially useful in diagnosing whether hypotensiveepisodes are the result of inadequate circulating bloodvolume, worsening ventricular function, or arterialvasodilation (155–157). Pulmonary artery cathetershave long been used in these patients for this purpose;if TEE is not available, pulmonary artery cathetersmay be a useful, if controversial, alternative (158).

Large volumes of blood, colloid, or crystalloidshould be used to treat hypotension in HF patientsonly when there is a reasonable suspicion that true

hypovolemia is present. This advice may be evenmore important for patients receiving spinal or epi-dural anesthesia (in the latter case there seems to be aneven greater tendency to use IV fluid/colloid/bloodrather than vasoactive drugs to treat hypotension).Patients receiving loop diuretics on an outpatient basismay prove refractory to the usual IV doses of furo-semide and continuous infusions of furosemide (20mg/h) or nesiritide (0.005–0.01 �g � kg�1 � min�1)may be needed. Finally, transfusion for perioperativeanemia in a hemodynamically stable patient with ahistory of HF (e.g., stage C) must be approached withgreater caution than usual. It is easy to produceintravascular volume overload in these patients (159).

When we consider our aging patient population inwhich prolonged survival with hypertension and/orcoronary artery disease is expected and the better HFtreatment strategies now available to them, we con-clude that anesthesiologists will encounter an increas-ing number of patients with either a predisposition toHF (stages A and B) or a history of HF (stages C andD). Thus, knowledge of the evolving pharmacologicstrategies for the management of chronic HF is essen-tial both for patient care and for our continued cred-ibility as perioperative physicians.

APPENDIXAbbreviations for Trials and Registries: ADHERE:

Acute Decompensated Heart Failure National Regis-try, A-HeFT: African-American Heart Failure Trial,ATLAS: Assessment of treatment with lisinopril andsurvival, BEST: �-Blocker evaluation of survivalstudy, CHARM: Candesartan in heart failure assess-ment in reduction of mortality, CIBIS: Cardiac insuf-ficiency bisoprolol study, COMET: Carvedilol ormetoprolol European trial, CONSENSUS: CooperativeNorth Scandinavian Enalapril Survival Study,COPERNICUS: Carvedilol prospective randomizedcumulative survival, DIG: Digitalis InvestigationGroup, ELITE: Evaluation of losartan in the elderly,EPHESUS: Eplerenone post-MI heart failure efficacyand survival study, GISSI: Gruppo Italiano per loStudio della Sopravvivenza nell’Infarto MiocardicoInvestigators, HOPE: Heart outcomes preventionevaluation, I-PRESERVE: Irbesartan in heart failurewith preserved systolic function, ISIS: Internationalstudy of infarct survival, MERIT-HF: MetoprololCR/XL randomized intervention trial in congestiveheart failure, PROVED: Prospective randomizedstudy of ventricular function and efficacy of digoxin,RALES: Randomized aldactone evaluation study,RADIANCE: Randomized assessment of digoxin orinhibitors of the angiotensin-converting enzyme,SAVE: Survival and ventricular enlargement, SOLVD:Study of left ventricular dysfunction, US CARVE-DILOL: United States carvedilol, V-Heft: Veteran’sAdministration Heart Failure Trial, Val-HeFT: Valsar-tan in heart failure trial.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 571

Page 66: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

REFERENCES

1. American Heart Association 2002 DT. Heart Disease andStroke Statistics-2003. Dallas: American Heart Association.Available at: www.americanheart.org.

2. Ho KK, Pinsky JL, Kannel WB, Levy D. The epidemiology ofheart failure: the Framingham Study. J Am Coll Cardiol1993;22(4 Suppl A):6A–13.

3. Eriksson H. Heart failure: a growing public health problem.J Intern Med 1995;237:135–41.

4. O’Connell JB, Bristow MR. Economic impact of heart failure inthe United States: time for a different approach. J Heart LungTransplant 1994;13:S107–12.

5. Massie BM, Shah NB. Evolving trends in the epidemiologicfactors of heart failure: rationale for preventive strategies andcomprehensive disease management. Am Heart J 1997;133:703–12.

6. Hunt SA. American College of Cardiology; American HeartAssociation Task Force on Practice Guidelines (Writing Com-mittee to Update the 2001 Guidelines for the Evaluation andManagement of Heart Failure). ACC/AHA 2005 guidelineupdate for the diagnosis and management of chronic heartfailure in the adult: a report of the American College ofCardiology/American Heart Association Task Force on Prac-tice Guidelines (Writing Committee to Update the 2001 Guide-lines for the Evaluation and Management of Heart Failure).J Am Coll Cardiol 2005;46:e1–82.

7. Swedberg K, Cleland J, Dargie H, et al. Task Force for theDiagnosis and Treatment of Chronic Heart Failure of theEuropean Society of Cardiology. Guidelines for the diagnosisand treatment of chronic heart failure: executive summary(update 2005): The Task Force for the Diagnosis and Treatmentof Chronic Heart Failure of the European Society of Cardiol-ogy. Eur Heart J 2005;26:1115–40.

8. Nieminen MS, Bohm M, Cowie MR, et al. ESC Committee forPractice Guideline (CPG). Executive summary of the guide-lines on the diagnosis and treatment of acute heart failure: theTask Force on Acute Heart Failure of the Eur Society ofCardiology. Eur Heart J 2005;26:384–416.

9. al-Kaade S, Hauptman PJ. Health-related quality of life mea-surement in heart failure: challenges for the new millennium.J Card Fail 2001;7:194–201.

10. Mehta J, Pepine CJ, Conti CR. Haemodynamic effects ofhydralazine and of hydralazine plus glyceryl trinitrate paste inheart failure. Br Heart J 1978;40:845–50.

11. LeJemtel TH, Keung E, Ribner HS, et al. Sustained beneficialeffects of oral amrinone on cardiac and renal function inpatients with severe congestive heart failure. Am J Cardiol1980;45:123–9.

12. Garg R, Yusuf S. Overview of randomized trials of angiotensin-converting enzyme inhibitors on morality and morbidity inpatients with heart failure. JAMA 1995;273:1450–6.

13. Flather MD, Yusuf S, Kober L, et al. Long-term ACE inhibitortherapy in patients with heart failure or left ventricular dys-function: a systematic overview of data from individual pa-tients. ACE inhibitor Myocardial Infarction CollaborativeGroup. Lancet 2000;355:1575–81.

14. Cohn JN, Tognoni G. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure. N EnglJ Med 2001;345:1667–75.

15. Pfeffer MA, Swedberg K, Granger CB, et al. Effects of cande-sartan on mortality and morbidity in patients with chronicheart failure: the CHARM-Overall programme. Lancet 2003;362:759–66.

16. Consensus recommendations for the management of chronicheart failure: on behalf of the membership of the advisorycouncil to improve outcomes nationwide in heart failure. AmJ Cardiol 1999;83(Suppl 2A):1A–38.

17. Packer M, Coats AJ, Fowler MB, et al. Effect of carvedilol onsurvival in severe chronic heart failure. N Engl J Med2001;344:1651–8.

18. The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): arandomised trial. Lancet 1999;353:9–13.

19. Effect of metroprolol CR/XL in chronic heart failure: Metopro-lol CR/XL Randomised Intervention Trial in Congestive HeartFailure (MERIT-HF) Lancet 1999;353:2001–7.

20. Pitt B, Zannad F, Remme WJ, et al. The effect of spironolactoneon morbidity and mortality in patients with severe heart

failure. Randomized Aldactone Evaluation Study Investiga-tors. N Engl J Med 1999;341:709–17.

21. Pitt B, Remme W, Zannad F, et al. Eplerenone, a selectivealdosterone blocker, in patients with left ventricular dys-function after myocardial infarction. N Engl J Med 2003;348:1309–21.

22. Katz AM. Angiotensin II. hemodynamic regulator or growthfactor? J Mol Cell Cardiol 1990;22:739–747.

23. Ino K, Shibata K, Kajiyama H, et al. Angiotensin II type 1receptor expression in ovarian cancer and its correlation withtumour angiogenesis and patient survival. Br J Cancer2006;94:552–60.

24. Cohn JN, Ferrari R, Sharpe N. Cardiac remodeling–conceptsand clinical implications: a consensus paper from an interna-tional forum on cardiac remodeling. J Am Coll Cardiol2000;35:569–82.

25. Opie LH, Commerford PJ, Gersh BJ, Pfeffer MA. Controversiesin ventricular remodelling. Lancet 2006;367:356–67.

26. Varin R, Mulder P, Tamion F, et al. Improvement of endothe-lial function by chronic angiotensin-converting enzyme inhibi-tion in heart failure: role of nitric oxide, prostanoids, oxidantstress, and bradykinin. Circulation 2000;102:351–6.

27. Dell’Italia LJ, Sabri A. Activation of the renin-angiotensinsystem in hypertrophy and heart failure. In Mann DL, ed.Heart Failure, a companion to Braunwald’s Heart disease, 1sted. Philadelphia: WB Saunders, 2004:129–43.

28. Pfeffer MA, Braunwald E, Moye LA, et al. Effect of captopril onmortality and morbidity in patients with left ventricular dys-function after myocardial infarction. Results of the survivaland ventricular enlargement trial. The SAVE investigators.N Engl J Med 1992;327:669–77.

29. ISIS-4: a randomised factorial trial assessing early oral capto-pril, oral mononitrate, and intravenous magnesium sulphate in58,050 patients with suspected acute myocardial infarction.ISIS-4 (Fourth International Study of Infarct Survival) Collabo-rative Group. Lancet 1995;345:669–85.

30. Arnold JM, Yusuf S, Young J, et al. Prevention of heart failurein patients in the heart outcomes prevention evaluation(HOPE) study. Circulation 2003;107:1284–90.

31. Taylor AL, Ziesche S, Yancy C, et al. Combination of isosorbidedinitrate and hydralazine in blacks with heart failure. N EnglJ Med 2004;351:2049–57.

32. Colson P, Ryckwaert F, Coriat P. Renin angiotensin systemantagonists and anesthesia. Anesth Analg 1999;89:1143–55.

33. Colson P, Saussine M, Seguin JR, et al. Hemodynamic effects ofanesthesia in patients chronically treated with angiotensin-converting enzyme inhibitors. Anesth Analg 1992;74:805–8.

34. Ryckwaert F, Colson P. Hemodynamic effects of anesthesia inpatients with ischemic heart failure chronically treated withangiotensin-converting enzyme inhibitors. Anesth Analg 1997;84:945–9.

35. Coriat P, Richer C, Douraki T, et al. Influence of chronicangiotensin-converting enzyme inhibition on anesthetic induc-tion. Anesthesiology 1994;81:299–307.

36. Larsen JK, Nielsen MB, Jespersen TW. Angiotensin-convertingenzyme inhibitors and anesthesia [in Danish]. Ugeskr Laeger1996;158:6081–4.

37. Hohne C, Meier L, Boemke W, Kaczmarczyk G. ACE inhibitiondoes not exaggerate the blood pressure decrease in the earlyphase of spinal anaesthesia. Acta Anaesthesiol Scand2003;47:891–6.

38. Boldt J, Rothe G, Schindler E, et al. Can clonidine, enoximone,and enalaprilat help to protect the myocardium against isch-aemia in cardiac surgery? Heart 1996;76:207–13.

39. Colson P, Ribstein J, Mimran A, et al. Effect of angiotensinconverting enzyme inhibition on blood pressure and renalfunction during open heart surgery. Anesthesiology 1990;72:23–7.

40. Licker M, Bednarkiewicz M, Neidhart P, et al. Preoperativeinhibition of angiotensin-converting enzyme improves sys-temic and renal haemodynamic changes during aortic abdomi-nal surgery. Br J Anaesth 1996;76:632–9.

41. Prys-Roberts C. Hypertension and anesthesia-fifty years on.Anesthesiology 1979;50:281–4.

42. Goldman L, Caldera DL. Risks of general anesthesia andelective operation in the hypertensive patient. Anesthesiology1979;50:285–92.

572 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 67: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

43. Howell SJ, Hemming AE, Allman KG, et al. Predictors ofpostoperative myocardial ischaemia: the role of intercurrentarterial hypertension and other cardiovascular risk factors.Anaesthesia 1997;52:107–11.

44. Tuman KJ, McCarthy RJ, O’Connor CJ, et al. Angiotensin-converting enzyme inhibitors increase vasoconstrictor require-ments after cardiopulmonary bypass. Anesth Analg 1995;80:473–9.

45. Thaker U, Geary V, Chalmers P, Sheikh F. Low systemicvascular resistance during cardiac surgery: case reports, briefreview, and management with angiotensin II. J CardiothoracAnesth 1990;4:360–3.

46. Boccara G, Ouattara A, Godet G, et al. Terlipressin versusnorepinephrine to correct refractory arterial hypotension aftergeneral anesthesia in patients chronically treated with renin-angiotensin system inhibitors. Anesthesiology 2003;98:1338–44.

47. Hopf HB, Schlaghecke R, Peters J. Sympathetic neural blockadeby thoracic epidural anesthesia suppresses renin release in re-sponse to arterial hypotension. Anesthesiology 1994;80:992–9.

48. Mackay JH, Walker IA, Bethune DW. Amiodarone and anaes-thesia: concurrent therapy with ACE inhibitors: an additionalcause for concern? Can J Anaesth 1991; 38:687.

49. Kincaid EH, Ashburn DA, Hoyle JR. Does the combination ofaprotinin and angiotensin-converting enzyme inhibitor causerenal failure after cardiac surgery?. Ann Thorac Surg 2005;80:1388–93.

50. Mangano DT, Tudor IC, Dietzel C. Multicenter study ofperioperative ischemia research group; ischemia research andeducation foundation. The risk associated with aprotinin incardiac surgery. N Engl J Med 2006;354:353–65.

51. Comfere T, Sprung J, Kumar MM, et al. Angiotensin systeminhibitors in a general surgical population. Anesth Analg2005;100:636–44.

52. Pigott DW, Nagle C, Allman K, et al. Effect of omitting regularACE inhibitor medication before cardiac surgery on haemody-namic variables and vasoactive drug requirements. Br J An-aesth 1999;83:715–20.

53. Burnier M, Brunner HR. Angiotensin II receptor antagonists.Lancet 2000;355:637–45.

54. Pitt B, Segal R, Martinez FA, et al. Randomised trial of losartanversus captopril in patients over 65 with heart failure (Evalu-ation of Losartan in the Elderly Study, ELITE). Lancet1997;349:747–52.

55. Pitt B, Poole-Wilson PA, Segal R, et al. Effect of losartancompared with captopril on mortality in patients with symp-tomatic heart failure: randomised trial–the Losartan HeartFailure Survival Study ELITE II. Lancet 2000;355:1582–7.

56. McMurray JJ, Ostergren J, Swedberg K, et al. Effects of candesar-tan in patients with chronic heart failure and reduced left ven-tricular systolic function taking angiotensin-converting-enzymeinhibitors: the CHARM-Added trial. Lancet 2003;362:767–71.

57. Granger CB, McMurray JJ, Yusuf S, et al. Effects of candesartanin patients with chronic heart failure and reduced left ventric-ular systolic function intolerant to angiotensin-converting-enzyme inhibitors: the CHARM-Alternative trial. Lancet 2003;362:772–6.

58. Yusuf S, Pfeffer MA, Swedberg K, et al. Effects of candesartanin patients with chronic heart failure and preserved leftventricular ejection fraction: the CHARM-Preserved Trial. Lan-cet 2003;362:777–81.

59. Brabant SM, Bertrand M, Eyraud D, et al. The hemodynamiceffects of anesthetic induction in vascular surgical patientschronically treated with angiotensin II receptor antagonists.Anesth Analg 1999;89:1388–92.

60. Bertrand M, Godet G, Meersschaert K, et al. Should theangiotensin II antagonists be discontinued before surgery?Anesth Analg 2001;92:26–30.

61. Licker M, Morel DR. Inhibitors of the renin angiotensin system:implications for the anaesthesiologist. Curr Opin Anaesthesiol1998;11:321–6.

62. Licker M, Neidhart P, Lustenberger S, et al. Long-termangiotensin-converting enzyme inhibitor treatment attenuatesadrenergic responsiveness without altering hemodynamic con-trol in patients undergoing cardiac surgery. Anesthesiology1996;84:789–800.

63. Mancia G, Dell’Oro R, Turri C, Grassi G. Comparison ofangiotensin II receptor blockers: impact of missed doses of

candesartan cilexetil and losartan in systemic hypertension.Am J Cardiol 1999;84:28S–34.

64. MacDonald JM. Anaesthesia and angiotensin II receptor antag-onist. Anaesthesia 2000;55:1038.

65. Bertrand M, Godet G, Meersschaert K, et al. Should theangiotensin II antagonists be discontinued before surgery?Anesth Analg 2001;92:26–30.

66. Morelli A, Tritapepe L, Rocco M, et al. Terlipressin versusnorepinephrine to counteract anesthesia-induced hypotensionin patients treated with renin-angiotensin system inhibitors:effects on systemic and regional hemodynamics. Anesthesiol-ogy 2005;102:12–9.

67. Eyraud D, Brabant S, Nathalie D, et al. Treatment of intraop-erative refractory hypotension with terlipressin in patientschronically treated with an antagonist of the renin-angiotensinsystem. Anesth Analg 1999;88:980–4.

68. Meersschaert K, Brun L, Gourdin M, et al. Terlipressin-ephedrineversus ephedrine to treat hypotension at the induction of anes-thesia in patients chronically treated with angiotensin converting-enzyme inhibitors: a prospective, randomized, double-blinded,crossover study. Anesth Analg 2002;94:835–40.

69. Skues MA, Richards MJ, Jarvis AP, Prys-Roberts C. Preinduc-tion atropine or glycopyrrolate and hemodynamic changesassociated with induction and maintenance of anesthesia withpropofol and alfentanil. Anesth Analg 1989;69:386–90.

70. Weber KT. Aldosterone in congestive heart failure. New EnglJ Med 2001;345:1689–97.

71. Zannad F, Alla F, Dousset B, et al. Limitation of excessiveextracellular matrix turnover may contribute to survival ben-efit of spironolactone therapy in patients with congestive heartfailure: insights from the randomized aldactone evaluationstudy (RALES). RALES Investigators. Circulation 2000;102:2700–6.

72. Yee KM, Pringle SD, Struthers AD. Circadian variation in theeffects of aldosterone blockade on heart rate variability and QTdispersion in congestive heart failure. J Am Coll Cardiol2001;37:1800–7.

73. Farquharson CA, Struthers AD. Spironolactone increases nitricoxide bioactivity, improves endothelial vasodilator dysfunc-tion, and suppresses vascular angiotensin I/angiotensin IIconversion in patients with chronic heart failure. Circulation2000;101:594–7.

74. Juurlink DN, Mamdani MM, Lee DS, et al. Rates of hyperka-lemia after publication of the Randomized Aldactone Evalua-tion Study. N Engl J Med 2004;351:543–51.

75. Anton C, Cox AR, Watson RD, Ferner RE. The safety ofspironolactone treatment in patients with heart failure. J ClinPharm Ther 2003;28:285–7.

76. Schepkens H, Vanholder R, Billiouw JM, Lameire N. Life-threatening hyperkalemia during combined therapy withangiotensin-converting enzyme inhibitors and spironolactone:an analysis of 25 cases. Am J Med 2001;110:438–41.

77. Brown NJ. Eplerenone: cardiovascular protection. Circulation2003;107:2512–8.

78. Cicoira M, Zanolla L, Rossi A, et al. Long-term, dose-dependenteffects of spironolactone on left ventricular function and exercisetolerance in patients with chronic heart failure. J Am Coll Cardiol2002;40:304–10.

79. Weber KT. Aldosterone and spironolactone in heart failure. NEngl J Med 1999;341:753–5.

80. Pitt B. “Escape” of aldosterone production in patients with leftventricular dysfunction treated with an angiotensin convertingenzyme inhibitor: implications for therapy. Cardiovasc DrugsTher 1995;9:145–9.

81. Zhou X, Ono H, Ono Y, Frohlich ED. Aldosterone antagonismameliorates proteinuria and nephrosclerosis independent ofglomerular dynamics in l-NAME/SHR model. Am J Nephrol2004;24:242–9.

82. Wallhaus TR, Taylor M, DeGrado TR, et al. Myocardial freefatty acid and glucose use after carvedilol treatment in patientswith congestive heart failure. Circulation 2001;103:2441–6.

83. Takeda Y, Fukutomi T, Suzuki S, et al. Effects of carvedilol onplasma B-type natriuretic peptide concentration and symp-toms in patients with heart failure and preserved ejectionfraction. Am J Cardiol 2004;94:448–53.

84. Borgdorff PJ, Ionescu TI, Houweling PL, Knape JT. Large-doseintrathecal sufentanil prevents the hormonal stress response

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 573

Page 68: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

during major abdominal surgery: a comparison with intrave-nous sufentanil in a prospective randomized trial. AnesthAnalg 2004;99:1114–20.

85. Eichhorn EJ, Bristow MR. Antagonism of �-adrenergic recep-tors in heart failure. In: Mann DL, ed. Heart failure: a compan-ion to Braunwald’s heart disease, 1st ed. Philadelphia: WBSaunders, 2004:619–36.

86. Lowes BD, Gilbert EM, Abraham WT, et al. Myocardial geneexpression in dilated cardiomyopathy treated with beta-blocking agents. N Engl J Med 2002;346:1357–65.

87. Pogwizd SM, Schlotthauer K, Li L, Yuan W, Bers DM.Arrhythmogenesis and contractile dysfunction in heart fail-ure: roles of sodium-calcium exchange, inward rectifierpotassium current, and residual beta-adrenergic responsive-ness. Circ Res 2001;88:1159–67.

88. Marks AR, Reiken S, Marx SO. Progression of heart failure: isprotein kinase a hyperphosphorylation of the ryanodine recep-tor a contributing factor? Circulation 2002;105:272–5.

89. Schlotthauer K, Bers DM. Sarcoplasmic reticulum Ca(2�)release causes myocyte depolarization: underlying mechanismand threshold for triggered action potentials. Circ Res 2000;87:774–80.

90. Doi M, Yano M, Kobayashi S, et al. Propranolol prevents thedevelopment of heart failure by restoring FKBP12.6-mediatedstabilization of ryanodine receptor. Circulation 2002;105:1374–9.

91. Terracciano CM, Hardy J, Birks EJ, et al. Clinical recovery fromend-stage heart failure using left ventricular assist device andpharmacological therapy correlates with increased sarcoplas-mic reticulum calcium content but not with regression ofcellular hypertrophy. Circulation 2004;109:2263–5.

92. Beanlands RS, Nahmias C, Gordon E, et al. The effects ofbeta(1)-blockade on oxidative metabolism and the metaboliccost of ventricular work in patients with left ventriculardysfunction: a double-blind, placebo-controlled, positron-emission tomography study. Circulation 2000;102:2070–5.

93. Sabbah HN, Sharov VG, Gupta RC, et al. Chronic therapy withmetoprolol attenuates cardiomyocyte apoptosis in dogs withheart failure. J Am Coll Cardiol 2000;36:1698–705.

94. Packer M, Bristow MR, Cohn JN, et al. The effect of carvedilolon morbidity and mortality in patients with chronic heartfailure. U.S. Carvedilol Heart Failure Study Group. N EnglJ Med 1996;334:1349–55.

95. Beta-Blocker Evaluation of Survival Trial Investigators. A trialof the beta-blocker bucindolol in patients with advancedchronic heart failure. N Engl J Med 2001;344:1659–67.

96. Hunt SA, Baker DW, Chin MH, et al. ACC/AHA guidelinesfor the evaluation and management of chronic heart failure inthe adult: executive summary. A report of the AmericanCollege of Cardiology/American Heart Association Task Forceon Practice Guidelines (Committee to revise the 1995 guide-lines for the Evaluation and Management of Heart Failure).J Am Coll Cardiol 2001;38:2101–13.

97. Jacob S, Rett K, Wicklmayr M, et al. Differential effect ofchronic treatment with two beta-blocking agents on insulinsensitivity: the carvedilol-metoprolol study. J Hypertens1996;14:489–94.

98. Dulin B, Abraham WT. Pharmacology of carvedilol. Am J Car-diol 2004;93:3B–6.

99. Hoffmann CW, Leitz MR, Oberdorf-Maass S, et al. Compara-tive pharmacology of human beta-adrenergic receptor sub-types–characterization of stably transfected receptors in CHOcells. Naunyn Schmiedebergs Arch Pharmacol 2004;369:151–9.

100. Moniotte S, Balligand JL. Potential use of beta(3)-adrenoceptorantagonists in heart failure therapy. Cardiovasc Drug Rev2002;20:19–26.

101. Cheng HJ, Zhang ZS, Onishi K, et al. Upregulation of func-tional beta(3)-adrenergic receptor in the failing canine myocar-dium. Circ Res 2001;89:599–606.

102. Morimoto A, Hasegawa H, Cheng HJ, et al. Endogenousbeta3-adrenoreceptor activation contributes to left ventricularand cardiomyocyte dysfunction in heart failure. Am J PhysiolHeart Circ Physiol 2004;286:H2425–33.

103. Moniotte S, Kobzik L, Feron O, et al. Upregulation of beta(3)-adrenoceptors and altered contractile response to inotropicamines in human failing myocardium. Circulation 2001;103:1649–55.

104. Gauthier C, Leblais V, Kobzik L, et al. The negative inotropiceffect of beta3-adrenoceptor stimulation is mediated by activa-tion of a nitric oxide synthase pathway in human ventricle.J Clin Invest 1998;102:1377–84.

105. Poole-Wilson PA, Swedberg K, Cleland JG, et al. Carvedilol orMetoprolol Eur Trial Investigators. Comparison of carvediloland metoprolol on clinical outcomes in patients with chronicheart failure in the Carvedilol or Metoprolol Eur Trial(COMET): randomised controlled trial. Lancet 2003;362:7–13.

106. Di Lenarda A, Sabbadini G, Sinagra G. Do pharmacologicaldifferences among beta-blockers affect their clinical efficacy inheart failure? Cardiovasc Drugs Ther 2004;18:91–3.

107. Braunstein JB, Anderson GF, Gerstenblith G, et al. Noncardiaccomorbidity increases preventable hospitalizations and mor-tality among Medicare beneficiaries with chronic heart failure.J Am Coll Cardiol 2003;42:1226–33.

108. Eagle KA, Berger PB, Calkins H, et al. ACC/AHA guidelineupdate for perioperative cardiovascular evaluation for noncar-diac surgery—executive summary: a report of the AmericanCollege of Cardiology/American Heart Association Task Forceon Practice Guidelines (Committee to Update the 1996 Guide-lines on Perioperative Cardiovascular Evaluation for Noncar-diac Surgery). J Am Coll Cardiol 2002;39:542–53.

109. Warltier DC. Beta-adrenergic-blocking drugs: incredibly use-ful, incredibly underutilized. Anesthesiology 1998;88:2–5.

110. Lindenauer PK, Pekow P, Wang K, et al. Perioperative beta-blocker therapy and mortality after major noncardiac surgery.N Engl J Med 2005;353:349–61.

111. Chang JJ, Luchsinger JA, Shea S. Antihypertensive medicationclass and pulse pressure in the elderly: analysis based on thethird National Health and Nutrition Examination Survey. AmJ Med 2003;115:536–42.

112. Domanski MJ, Davis BR, Pfeffer MA, et al. Isolated systolichypertension : prognostic information provided by pulse pres-sure. Hypertension 1999;34:375–80.

113. Domanski M, Norman J, Pitt B, et al. Studies of left ventriculardysfunction: diuretic use, progressive heart failure, and deathin patients in the studies of left ventricular dysfunction(SOLVD). J Am Coll Cardiol 2003;42:705–8.

114. The effect of digoxin on mortality and morbidity in patientswith heart failure. N Engl J Med 1997;336:525–33.

115. Adams KF Jr., Gheorghiade M, Uretsky BF, et al. Patients withmild heart failure worsen during withdrawal from digoxintherapy. J Am Coll Cardiol 1997;30:42–8.

116. Rahimtoola SH. Digitalis therapy for patients in clinical heartfailure. Circulation 2004;109:2942–6.

117. Slatton ML, Irani WN, Hall SA, et al. Does digoxin provideadditional hemodynamic and autonomic benefit at higherdoses in patients with mild to moderate heart failure andnormal sinus rhythm? J Am Coll Cardiol 1997;29:1206–13.

118. Allison SP, Lobo DN. Fluid and electrolytes in the elderly. CurrOpin Clin Nutr Metab Care 2004;7:27–33.

119. Cooper HA, Dries DL, Davis CE, et al. Diuretics and risk ofarrhythmic death in patients with left ventricular dysfunction.Circulation 1999;100:1311–5.

120. Sear JW, Howell SJ, Sear YM, et al. Intercurrent drug therapyand perioperative cardiovascular mortality in elective andurgent/emergency surgical patients. Br J Anaesth 2001;86:506–12.

121. El-Salawy SM, Lowenthal DT, Ippagunta S, Bhinder F. Clinicalpharmacology and physiology conference: digoxin toxicity inthe elderly. Int Urol Nephrol 2005;37:665–8.

122. Brucks S, Little WC, Chao T, et al. Contribution of leftventricular diastolic dysfunction to heart failure regardless ofejection fraction. Am J Cardiol 2005;95:603–6.

123. Aurigemma GP, Gaasch WH. Clinical practice: diastolic heartfailure. N Engl J Med 2004;351:1097–1105.

124. Zile MR, Brutsaert DL. New concepts in diastolic dysfunction anddiastolic heart failure: Part I: diagnosis, prognosis, and measure-ments of diastolic function. Circulation 2002;105:1387–93.

125. Carson P, Massie BM, McKelvie R, et al. The irbesartan in heartfailure with preserved systolic function (I-PRESERVE) trial:rationale and design. J Card Fail 2005;11:576–85.

126. Flather MD, Shibata MC, Coats AJ, et al. Randomized trial todetermine the effect of nebivolol on mortality and cardiovas-cular hospital admission in elderly patients with heart failure(SENIORS). Eur Heart J 2005;26:215–25.

574 Heart Failure and Pharmacotherapies ANESTHESIA & ANALGESIA

Page 69: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

127. Daniel KR, Wells GL, Fray B, et al. The effect of spironolactoneon exercise tolerance and quality of life in elderly women withdiastolic heart failure. Am J Geriatr Cardiol 2003;12:131.

128. Roongsritong C, Sutthiwan P, Bradley J, et al. Spironolactoneimproves diastolic function in the elderly. Clin Cardiol 2005;28:484–7.

129. DiDomenico RJ, Park HY, Southworth MR, et al. Guidelines foracute decompensated heart failure treatment. Ann Pharmaco-ther 2004;38:649–60.

130. Cheng JW. Nesiritide: review of clinical pharmacology androle in heart failure management. Heart Dis 2002;4:199–203.

131. Iyengar S, Feldman DS, Trupp R, Abraham WT. Nesiritide forthe treatment of congestive heart failure. Expert Opin Pharma-cother 2004;5:901–7.

132. Elkayam U, Akhter MW, Singh H, et al. Comparison of effectson left ventricular filling pressure of intravenous nesiritide andhigh-dose nitroglycerin in patients with decompensated heartfailure. Am J Cardiol 2004;93:237–40.

133. Publication Committee for the VMAC Investigators (Vasodila-tation in the Management of Acute CHF). Intravenous nesirit-ide vs nitroglycerin for treatment of decompensated congestiveheart failure: a randomized controlled trial. JAMA 2002;287:1531–40.

134. Burger AJ, Horton DP, LeJemtel T, et al. Effect of nesiritide(B-type natriuretic peptide) and dobutamine on ventriculararrhythmias in the treatment of patients with acutely decom-pensated congestive heart failure: the PRECEDENT study. AmHeart J 2002;144:1102–8.

135. Abraham WT, Adams KF, Fonarow GC, et al. ADHEREScientific Advisory Committee and Investigators; ADHEREStudy Group. In-hospital mortality in patients with acutedecompensated heart failure requiring intravenous vasoactivemedications: an analysis from the Acute Decompensated HeartFailure National Registry (ADHERE). J Am Coll Cardiol2005;46:57–64.

136. Sackner-Bernstein JD, Kowalski M, Fox M, Aaronson K.Short-term risk of death after treatment with nesiritide fordecompensated heart failure: a pooled analysis of random-ized controlled trials. JAMA 2005;293:1900–5.

137. Sackner-Bernstein JD, Skopicki HA, Aaronson KD. Risk of wors-ening renal function with nesiritide in patients with acutelydecompensated heart failure. Circulation 2005;111:1487–91.

138. MedWatch – Natrecor® (nesiritide) Dear Doctor Letter. Avail-able at: www.fda.gov/medwatch/safety/2005/natrecor2_hcp.pdf. Accessed February 17, 2006.

139. Packer M, Carver JR, Rodeheffer RJ, et al. Effect of oralmilrinone on mortality in severe chronic heart failure. ThePROMISE Study Research Group. N Engl J Med 1991;325:1468–75.

140. Massie B, Bourassa M, DiBianco R, et al. Long-term oraladministration of amrinone for congestive heart failure: lackof efficacy in a multicenter controlled trial. Circulation1985;71:963–71.

141. Abraham WT, Adams KF, Fonarow GC, et al. In-hospitalmortality in patients with acute decompensated heart failurerequiring intravenous vasoactive medications: an analysisfrom the Acute Decompensated Heart Failure National Regis-try (ADHERE). J Am Coll Cardiol 2005;46:57–64.

142. Stevenson LW. Clinical use of inotropic therapy for heartfailure: looking backward or forward? Part I: Inotropic infu-sions during hospitalization. Circulation 2003;108:367–372.

143. Follath F, Cleland JG, Just H, et al. Efficacy and safety ofintravenous levosimendan compared with dobutamine in se-vere low-output heart failure (the LIDO study): a randomiseddouble-blind trial. Lancet 2002;360:196–202.

144. Kersten JR, Montgomery MW, Pagel PS, Warltier DC. Levosi-mendan, a new positive inotropic drug, decreases myocardialinfarct size via activation of KATP channels. Anesth Analg2000;90:5–11.

145. Moiseyev VS, Poder P, Andrejevs N, et al. Safety and effi-cacy of a novel calcium sensitizer, levosimendan, in patientswith left ventricular failure due to an acute myocardialinfarction: a randomized, placebo-controlled, double-blindstudy (RUSSLAN). Eur Heart J 2002;23:1422–32.

146. Renlund DG. Building a bridge to heart transplantation.N Engl J Med 2004;351:849–51.

147. Wang CS, FitzGerald JM, Schulzer M, et al. Does this dyspneicpatient in the emergency department have congestive heartfailure? JAMA 2005;294:1944–56.

148. McCullough PA, Nowak RM, McCord J, et al. B-type natri-uretic peptide and clinical judgment in emergency diagnosis ofheart failure: analysis from Breathing Not Properly (BNP)Multinational Study. Circulation 2002;106:416–22.

149. Silver MA, Maisel A, Yancy CW, et al. BNP Consensus Panel2004: A clinical approach for the diagnostic, prognostic, screen-ing, treatment monitoring, and therapeutic roles of natriureticpeptides in cardiovascular diseases. Congest Heart Fail2004;10(5 Suppl 3):1–30.

150. Chobanian AV, Bakris GL, Black HR, et al. The seventh reportof the joint national committee on prevention, detection, evalu-ation, and treatment of high blood pressure: the JNC 7 report.JAMA 2003;289:2560–72.

151. Shephard J, Cobbe SM, Ford I, et al. Prevention of coronaryartery disease with pravastatin in men with hypercholesterol-emia. West of Scotland Coronary Prevention Study Group.N Engl J Med 1995;333:1301–7.

152. Brater DC. Diuretic therapy. N Engl J Med 1998;339:387–95.153. Cohn JN, Johnson G, Ziesche S, et al. A comparison of enalapril

with hydralazine-isosorbide dinitrate in the treatment ofchronic congestive heart failure. N Engl J Med 1991;325:303–10.

154. Tang WH, Francis GS. The year in heart failure. J Am CollCardiol 2005;46:2125–33.

155. Colreavy FB, Donovan K, Lee KY, Weekes J. Transesophagealechocardiography in critically ill patients. Crit Care Med2002;30:989–96.

156. Tousignant CP, Walsh F, Mazer CD. The use of transesopha-geal echocardiography for preload assessment in critically illpatients. Anesth Analg 2000;90:351–5.

157. Practice guidelines for perioperative transesophageal echocar-diography. A report by the American Society of Anesthesiolo-gists and the Society of Cardiovascular Anesthesiologists TaskForce on Transesophageal Echocardiography. Anesthesiology1996;84:986–1006.

158. Robin ED. Death by pulmonary artery flow-directed catheter:time for a moratorium? Chest 1987;92:727–31.

159. Felker GM, Adams KF Jr., Gattis WA, O’Connor CM. Anemiaas a risk factor and therapeutic target in heart failure. J Am CollCardiol 2004;44:959–66.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 575

Page 70: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Seminar

www.thelancet.com Vol 368 August 19, 2006 687

Peripartum cardiomyopathyKaren Sliwa, James Fett, Uri Elkayam

Peripartum cardiomyopathy (PPCM) is a disorder in which initial left ventricular systolic dysfunction and symptoms of heart failure occur between the late stages of pregnancy and the early postpartum period. It is common in some countries and rare in others. The causes and pathogenesis are poorly understood. Molecular markers of an infl ammatory process are found in most patients. Clinical presentation includes usual signs and symptoms of heart failure, and unusual presentations relating to thromboembolism. Clinicians should consider PPCM in any peripartum patient with unexplained disease. Conventional heart failure treatment includes use of diuretics, β blockers, and angiotensin-converting enzyme inhibitors. Eff ective treatment reduces mortality rates and increases the number of women who fully recover left ventricular systolic function. Outcomes for subsequent pregnancy after PPCM are better in women who have fi rst fully recovered heart function. Areas for future research include immune system dysfunction, the role of viruses, non-conventional treatments such as immunosuppression, immunoadsorption, apheresis, antiviral treatment, suppression of proinfl ammatory cytokines, and strategies for control and prevention.

Peripartum cardiomyopathy (PPCM) is a disorder of unknown cause in which initial left ventricular systolic dysfunction and symptoms of heart failure occur between the last month of pregnancy and the fi rst 5 months postpartum.1,2 Elkayam and colleagues3 described an identical clinical condition that appeared earlier in pregnancy; these women (n=23) were diagnosed with “pregnancy-associated” cardiomyopathy at 17–36 weeks of gestation, and did not diff er clinically from women with the usual presentation of PPCM (n=100). Some reports of PPCM4–6 also include women who presented with fi rst heart failure in the sixth month postpartum. All these conditions might be part of the same clinical entity, with an expanded time interval for onset of heart failure.

An essential element in diagnosis of PPCM is the demonstration of left ventricular systolic dysfunction. Because echocardiography was not available at the time of Demakis’ original description of the condition,1 specifi c echocardiographic measures of left ventricular systolic dysfunction have been proposed as additional criteria.7 These measures include an ejection fraction of less than 45%, fractional shortening of less than 30%, or both, and end-diastolic dimension of greater than 2·7 cm/m² body surface-area. The need for echo cardiographic confi rmation of left ventricular systolic dysfunction often presents a diffi culty in developing countries where the necessary technology is not available. However, even in settings of poverty, echocardiographic strategies have been shown to be possible and practical.8,9

PPCM remains a diagnosis of exclusion. No additional specifi c criteria have been identifi ed to allow distinction between a peripartum patient with new onset heart failure and left ventricular systolic dysfunction as PPCM and another form of dilated cardiomyopathy. Therefore, all other causes of dilated cardiomyopathy with heart failure must be systematically excluded before accepting the designation of PPCM. Recent observations from Haiti10 suggest that a latent form of PPCM without clinical symptoms might exist. The investigators identifi ed four clinically normal postpartum women with asymptomatic systolic dysfunction on echocardiography, who sub-

sequently either developed clinically detectable dilated cardiomyopathy, or improved and completely recovered heart function.

Epidemiology Although it seems likely that women of reproductive age all over the world have some risk of developing PPCM, good data about incidence are unavailable because so few population-based registries exist. Recent reports suggest an estimated incidence of one case per 299 livebirths in Haiti,11 one case per 1000 livebirths in South Africa,12 and one case per 2289 livebirths13 to one case per 4000 livebirths in the USA.2 These more recent data from the USA suggest a higher incidence than reported in earlier studies.14,15 The reasons for this variation in incidence between countries remain unknown.

Cases of PPCM that adhere to the stated diagnostic criteria probably represent a similar disease, despite geographical variation in contributing factors. The remarkable propensity for recovery of left ventricular function observed in the diverse locations provides further evidence for a similar disease process. It should not be surprising that varying genetic pools and diverse environmental factors play parts of varying importance

Lancet 2006; 368: 687–93

Soweto Cardiovascular Research Unit, Department of Cardiology, Chris-Hani-Baragwanath Hospital, University of the Witwatersrand, P O Bertsham 2013, Johannesburg, South Africa (K Sliwa MD); Department of Adult Medicine, Hôpital Albert Schweitzer, Deschapelles, Haiti (J Fett MD); and Division of Cardiovascular Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA (U Elkayam MD)

Correspondence to: Prof Karen [email protected]

Search strategy and selection criteria

We aimed to summarise the results of a MEDLINE search (January, 1960, to January, 2006) on the current understanding of the epidemiology, aetiology, clinical profi le, and management of PPCM. We used the search terms “peripartum cardiomyopathy”, “postpartum cardiomyopathy”, “heart failure in pregnancy”, and “heart failure postpartum”. We also searched the reference lists of articles identifi ed by this strategy and selected those we judged relevant. Several review articles and book chapters were included because they provided comprehensive overviews. We identifi ed areas in which knowledge is defi cient and where molecular research eff orts should be directed in seeking to understand the basic pathogenesis of PPCM.

Page 71: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Seminar

688 www.thelancet.com Vol 368 August 19, 2006

in diff erent areas. However, cultural practices in some areas such as heavy intake of lake salt and body heating in clay beds, might precipitate pregnancy-associated heart failure that does not always fi t the diagnostic criteria for PPCM.16–20

Although not clearly delineated, suggested risk factors associated with PPCM have included age, gravidity or parity, African origin, toxaemia or hypertension of pregnancy, use of tocolytics, and twin pregnancy. The incidence of these presumed risk factors reported in the three largest series of patients with PPCM3,8,9,11,21 and in Demakis and colleague’s1,22 original report of the condition are shown in the table.

Although PPCM is thought to be more prevalent in the upper and lower extremes of childbearing age, and in older women of high parity,12,23 it is important to note that 24–37% of cases may occur in young primigravid patients.1,3,9,11,12,21,22 The largest prospectively-identifi ed case series reports, from Haiti11 and South Africa,21 did not show a disproportionate role for older age, multiparity, and long-term use of tocolytic agents in the development

of PPCM. The association with race might be confounded by the observed increased frequency of PPCM in women of lower socioeconomic status.

Aetiology The cause and mechanism of pathogenesis of PPCM remain unknown, and many hypotheses have been proposed (fi gure 1).24–29 Early suggestions that nutritional disorders, such as defi ciencies in selenium and other micronutrients, might contribute could not be confi rmed in studies of Haitian patients with PPCM,27

although unidentifi ed nutritional factors might exist.Because of immune-system changes related to

pregnancy, associations with autoimmune mechanisms and infl ammation have been studied since the 1970s. Melvin and colleagues30 proposed myocarditis as the cause for PPCM and reported a dense lymphocyte infi ltrate with variable amounts of myocyte oedema, necrosis, and fi brosis in right ventricular biopsy specimens. Treatment with prednisone and azathioprine resulted in clinical improvement and loss of infl ammatory infi ltrate on repeated biopsies in the three patients studied. Sanderson and colleagues,31 and subsequently Midei and colleagues,32 emphasised the association between myocarditis and development of PPCM. Although their fi ndings were intriguing, they failed to establish a causal link. Additionally, Rizeq and colleagues33 found an infl ammatory component in less than 10% of biopsy samples from patients with PPCM, a proportion similar to that found in age-and-sex-matched patients with idiopathic dilated cardiomyopathy. A decade later, Felker and colleagues34 confi rmed that the absence or presence of infl ammation on endomyocardial biopsy tissue did not predict outcome in patients with PPCM.

A viral trigger for the development of PPCM has been postulated and investigated in several studies. Bultmann and colleagues35 identifi ed viral genomic material in endomyocardial biopsy tissue from patients with PPCM; however, the same incidence and types of viral positivity were noted in controls. The fi ndings of Kuhl and colleagues25 in idiopathic dilated cardiomyopathy are

Haiti, 20058,11

(n=98)*South Africa, 20059,21 (n=100)*

USA, 20053 (n=100)†

USA, 19711,22 (n=27)‡

Age (years) 31·8 (8·1, 16–51) 31·6 (6·6, 18–45) 30·7 (6·4, 16–43) 14 patients >30, 13 patients <30

Gravidity 4·3 (1–10) 3 (1–7) 2·6 (1–10) 8 patients (29%) G1–2, 19 patients (71%) >G3

Primigravidas 24 (24·5%) 20 (20%) 37 (37%) 8 (29%)§

Hypertension/toxaemia 4 (4%) 2 (2%) 43 (43%) 6 (22%)

Use of tocolytics 0 9 (9%) 19 (19%) 0

African descent 98 (100%) 100 (100%) 19 (19%) 25 (93%)

Twin pregnancy 6 (6%) 6 (6%) 13 (13%) 2 (7%)

Mortality 15 (15·3%) 15 (15%) 9 (9%) 11 (40·7%)

For Haiti, South Africa, and USA (2005), data are mean (SD, range) for age and mean (range) for gravidity.*Single centre prospective study. †Multicentre retrospective study, including survey questionnaire. ‡Single centre retrospective study. §Report included gravida 1–2 together as a group.

Table: Comparison of potential risk factors and mortality rates in PPCM

Activation of protectivecardiac myocytesignalling pathways

Normal Normal

Normalpregnancy

Pregnancy associatedhypertrophy

• Viral antigen persistence• Stress activated cytokines• Autoimmune responses• Genetic factors• Micronutrients• Microchimerism• Increased myocyte apoptosis• Excessive prolactin production

PPCM

Irreversibledysfunction

Figure 1: Proposed factors investigated contributing to the pathogenesis for PPCMReferences for factors listed: protective signalling pathway,24 viral antigen persistence,25 stress-activated cytokines,9,20 autoimmunity,26 micronutrients,27 microchimerism,26 myocyte apoptosis,9,21,28 prolactin.24,29

Page 72: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Seminar

www.thelancet.com Vol 368 August 19, 2006 689

more convincing evidence for the role of virus as a trigger for cardiomyopathy. The investigators demonstrated the presence of viral genomic material in endomyocardial biopsy tissue, including enterovirus (coxsackievirus), parvovirus B19, adenovirus, and herpesvirus. They also showed that clinical improvement of left ventricular systolic function was associated with viral clearing. If future results show that viruses play a part in the development of PPCM (and some forms of idiopathic dilated cardiomyopathy), endomyocardial biopsy could become increasingly important in PPCM and unexplained cardiomyopathy, especially in patients who do not improve with conventional treatment in the early weeks after diagnosis.36,37

Ansari and colleagues26 investigated the role of fetal microchimerism (fetal cells in maternal blood during and after pregnancy) in patients with PPCM. In a small sample of patients, the amount of male chromosomal DNA in maternal plasma was signifi cantly greater in patients with PPCM than in control mothers without PPCM during the third trimester of pregnancy, at term, and in the fi rst week postpartum. The lower concentrations of this foreign protein could possibly contribute to tolerance of the fetus, permitting successful completion of pregnancy; increased levels could theoretically lead to the initiation of autoimmune disease, including an autoimmune myocarditis.

Findings from South Africa9,21 and Haiti26,38 lend support to the hypothesis that immune activation contributes to the pathogenesis of PPCM. Sliwa and colleagues9,21 identifi ed increased concentrations in plasma of the infl ammatory cytokine tumour necrosis factor α (TNFα), C-reactive protein, and a plasma marker of apoptosis, Fas/Apo-1, in a large population of newly diagnosed patients with PPCM. Baseline concentrations of C-reactive protein correlated positively with baseline left ventricular end-diastolic and end-systolic diameters and inversely with left ventricular ejection fraction. Concentrations of Fas/Apo-1 in plasma were signifi cantly higher in patients with PPCM than in healthy controls, and were a predictor of mortality. Concentrations of C-reactive protein in plasma can vary substantially between diff erent ethnic groups, and as much as 40% of this variation is genetically determined.39 Hypothetically, an increase in the intensity of an infl ammatory response could be one of the many factors contributing to development of PPCM. The importance of raised high-sensitivity C-reactive protein in plasma of patients with new and evolving PPCM10,40 merits additional evaluation.

A high incidence of postpartum mortality in mice with a cardiac-tissue-specifi c signal transducer and activator of transcription 3 (STAT 3) knockout of has been reported.24 Before death, female mutant mice presented with symptoms of heart failure, reduced cardiac function, and apoptosis. Data from this study suggested that the cardiac-myocyte-specifi c STAT3 pathway is necessary for protection of the heart from postpartum stress, and that

prolactin cleavage is crucially involved in the pathogenesis of PPCM. Additionally, data from another mouse model of PPCM suggest that apoptosis of cardiomyocytes has a causal role in PPCM, and that pharmacological inhibition of apoptosis by a caspase inhibitor might off er novel therapeutic strategies.28 The fi nding of raised Fas/Apo-1 in women with PPCM9 provides additional evidence for a role of apoptosis.

Warraich and colleagues38 investigated the eff ect and clinical relevance of PPCM on humoral immunity and evaluated the immunoglobulins (class G and subclasses G1, G2, G3) against cardiac myosin in 47 patients with PPCM from South Africa, Mozambique, and Haiti. The immunoglobulin profi les were similar in the three regions and were markedly and non-selectively present (fi gure 2).

Although various causes for PPCM have been proposed, including abnormal hormonal regulation, the role of innate and adaptive immune systems, and the participation of autoantibodies, progenitor dendritic cells, T and B lymphocytes, cytokines, and chemokines,29,38,41–48 so far no cause has been clearly identifi ed. It is likely that the aetiology is multifactorial, and that the search for a cause is hampered by the rarity of this condition in the developed countries, where more research funds are available, by the lack of adherence to diagnostic guidelines, and by the heterogeneity of the populations studied.

Clinical presentationThe most common presentation of PPCM is with symptoms and signs of systolic heart failure.1,8,9,12 Clinical examination of 97 patients seen in South Africa12 over 4 years revealed a displaced hypodynamic apical impulse in 72% and gallop rhythm in 92% of the patients. Functional mitral regurgitation was present in 43%. Left ventricular hypertrophy by electrocardiographic voltage criteria was present in 66% and ST-T wave abnormalities in 96% of the patients. Additional symptoms and signs include

Plasma cardiac myosin autoantibodies

0

10

20

30

40

50

60

70

80

90

South Africa(n=15)

Mozambique(n=9)

Haiti(n=23)

Control (n=15)

IgG1

Prop

ortio

n po

sitiv

e (%

)

IgG2IgG3

Figure 2: Comparison of immunoglobulin profi les in patients with PPCM from South Africa, Haiti, and Mozambique versus 15 healthy mothers from South Africa Data from Warraich and colleagues.38

Page 73: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Seminar

690 www.thelancet.com Vol 368 August 19, 2006

dependent oedema, dyspnoea on exertion, orthopnoea, paroxysmal nocturnal dyspnoea, persistent cough, abdominal discomfort secondary to passive congestion of the liver and other organs, precordial pain, and palpitations. In the later stages postural hypotension may be prominent, refl ecting low cardiac output and low blood pressure.

Early signs and symptoms of heart failure can be obscured by pregnancy, because often the patient considers them to be a normal part of pregnancy. New York Heart Association (NYHA) cardiac functional classifi cation varies from NYHA I to IV, although most frequently initial presentation is with NYHA III and IV. Sudden cardiac arrest might occur in a situation where cardiopulmonary resuscitation is neither available nor successful.49 Delayed diagnosis can be associated with increased morbidity and mortality. Clinicians should think of PPCM in any peripartum patient with unexplained disease. A broad index of suspicion is important, since unusual and non-heart failure presentations are also possible, as illustrated by the following evidence.

Left ventricular thrombus is common in PPCM patients with a left ventricular ejection fraction of less than 0·35.9 With progression of disease, four-chamber dilation may be seen, with thrombus formation in the left atrium and right ventricle. Peripheral embolisation then becomes possible to any part of the body, including arterial occlusion of the lower extremities with compromised circulation,8,50,51 cerebral embolism with hemiplegia, aphasia, and incoordination,8,52 mesenteric artery occlusion with an acute abdomen syndrome secondary to bowel infarction,8

and acute myocardial infarction secondary to coronary artery embolism.53,54 Haemoptysis may be the presenting feature of co-existing pulmonary embolus, for which the patients also have an increased risk.12

A recent report55 identifi ed a 5-weeks postpartum 35-year-old mother who progressively deteriorated with acute hepatic failure and was being considered for liver transplant. An echocardiogram identifi ed dilated cardiomyopathy as the reason for heart failure with subsequent passive congestion of the liver and severe hepatic failure. Appropriate treatment led to survival and recovery from liver failure as well as complete recovery of left ventricular systolic function.

Management and prognosisThe medical management of patients with PPCM is similar to that for other forms of heart failure, and has been reviewed in detail.56 Treatment aims to reduce afterload and preload, and to increase contractility. Angiotensin-converting enzyme (ACE) inhibitors are usually used to reduce afterload by vasodilation if PPCM occurs after pregnancy. Because of potential toxic eff ects on the fetus, hydralazine (with or without nitrates) replaces ACE inhibitors during pregnancy. β blockers are used, since high heart rate, arhythmmias, and sudden death often occur in patients with PPCM. Digitalis, an inotropic agent, is also safe during pregnancy and may help to

maximise contractility and rate control, but has to be closely monitored since excessive digoxin concentrations in serum have been associated with worse outcomes in women.57,58 Women in general, and pregnant women in particular, may be more sensitive than men to its eff ect. Diuretics are also safe and are used to reduce preload and relieve symptoms. Because of the high incidence of thrombo embolism in these patients, the use of heparin is also considered necessary, followed by warfarin in those with left ventricular ejection fractions of less than about 35%.

Although PPCM shares many features with other forms of non-ischaemic cardiomyopathy, an important distinction is that women with PPCM have a higher rate of spontaneous recovery of ventricular function. However, in a single centre prospective study of 100 South African patients with the condition, 15% died, and only 23% recovered normal left ventricular function after 6 months of treatment, despite optimal medical therapy with ACE inhibitors and β blockers.21 In a longer follow-up study of prospectively identifi ed patients in Haiti over 5 years, the mortality rate was also 15%, and 29 of 92 (31·5 %) recovered normal left ventricular function.11 Continuing improvement was observed in the second and third years after diagnosis, confi rming that the recovery phase is not limited to the fi rst 6–12 months.

Subsequent monitoring depends on response to treatment, and includes a follow-up echocardiogram in the fi rst several weeks to confi rm improvement of left ventricular systolic function. After that, an echocardiogram is indicated about every 6 months until recovery is confi rmed or a plateau is reached. The best time to discontinue ACE-inhibitors or β blockers is unknown; however, one or both of these medications should be continued for at least 1 year. Where resources exist, left ventricular assist devices and heart transplantation may be used if necessary.59, 60

Limited studies have been done with the use of immunosuppressive drugs such as azathioprine and steroids, and have shown mixed results. Use of these agents should be reserved pending further assessments, and perhaps should be restricted to patients with biopsy-proven lymphocytic myocarditis in the absence of viral particles. Recent studies in some patients with idiopathic dilated cardiomyopathy suggest a prominent role for cardiotrophic viruses.25,37 As noted previously, only one investigation to date has identifi ed viral genomic material in endomyocardial tissue from patients with PPCM.35 Hence, PCR testing for a range of cardiomyo tropic viruses could become important in the investigation of patients who do not improve in the early weeks following diagnosis.

Aside from their haemodynamic benefi ts, the angio-tensin-converting enzyme inhibitors, β blockers, and angiotensin-receptor blockers may have an additional benefi t to dampen an over-active immune system that plays a role in the basic pathophysiology of PPCM.61–63

Page 74: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Seminar

www.thelancet.com Vol 368 August 19, 2006 691

Given the potential infl ammatory nature of PPCM there may also be a role for other immunomodulatory therapy. A prospective study of 59 consecutive women with PPCM reported a signifi cant reduction in the infl ammatory marker TNFα and improved outcome in patients receiving the immunomodulating agent pentoxifylline in addition to conventional therapy.64

Long-term prognosisVery few studies have been done to investigate the long-term survival and recovery outcomes in patients with PPCM. Felkner and colleagues34 assessed the survival of patients with initially unexplained cardiomyopathy referred for endomyocardial biopsy Johns Hopkins hospital. Patients with PPCM seemed to have a better prognosis than those with other forms of cardiomyopathy. However, patients who died soon after diagnosis were probably not included in that study. Fett and co-workers,11

with over 5 years of echocardiographic observations, noted continuing improvement in cardiac function well beyond the initial 6–12 months after diagnosis (fi gure 3).

Subsequent pregnancy and long term outcomeOne of the greatest concerns of PPCM patients is the safety of additional pregnancies. Since many patients with PPCM develop the disease during or shortly after their fi rst pregnancy, it is especially important to be able to provide them with information about relapse. In a retrospective study, Elkayam and colleagues65 found that subsequent pregnancy in women with PPCM was associated with signifi cant decrease in left ventricular function that resulted in clinical deterioration and even death. Symptoms of heart failure occurred in 21% of those who entered the subsequent pregnancy with normal left ventricular systolic function and 44% of those who entered the subsequent pregnancy with abnormal left ventricular systolic function. However, all deaths (three of 44, 7%) occurred in the group who entered the subsequent pregnancy with abnormal systolic function. Eventual recovery of left ventricular systolic function occurred more frequently in women who had an ejection fraction of greater than 30% at original diagnosis of PPCM.

Serial studies of left ventricular function and TNFα in prospectively-studied African patients with subsequent pregnancy after PPCM showed that deterioration of left ventricular function occurred postpartum to the subsequent pregnancy in fi ve of six patients and was accompanied by raised concentrations of TNFα in plasma.66

In a study67 of 15 prospectively-identifi ed Haitian patients with a subsequent pregnancy after PPCM, seven patients tolerated the subsequent pregnancy without worsening heart failure whereas eight had a relapse. Of those who relapsed, only one regained normal left ventricular systolic function during the 2-year follow-up after the subsequent pregnancy. All those who did not relapse went on to recover normal left ventricular systolic

function. The longer follow-up allowed identifi cation of both late recovery and late deterioration, again emphasising that recovery of heart function is not limited to the fi rst 6–12 months after diagnosis or relapse.

Family-planning counselling is an important aspect of the care of patients after a diagnosis of PPCM. Subsequent pregnancy after a diagnosis of PPCM carries higher risk of relapse if left ventricular systolic function is not fully recovered fi rst; and even with full recovery some additional risk of relapse remains.65

Implications for researchReliable population-based information about incidence and prevalence of PPCM is essential to the development of national and international health policies for prevention and control of this condition. These should include a genetic epidemiological design to estimate the contribution of genetic susceptibility factors to this condition. While investigation continues on the molecular basis of PPCM, controlled trials are also needed for potential new treatments—including apheresis, immunoadsorption, immunosuppression, and antiviral agents—all of which might have a role, as suggested by initial reports from research in idiopathic dilated cardiomyopathy.68–71 The promising results64 that have been obtained with pentoxifylline in a non-randomised trial need to be tested in large multicentre randomised trials with the power to assess mortality and morbidity benefi ts.72

Confl ict of interest statementWe declare that we have no confl ict of interest.

References1 Demakis JG, Rahimtoola SH. Peripartum cardiomyopathy.

Circulation 1971: 44: 964–68.2 Pearson GD, Veille JC, Rahimtoola S, et al. Peripartum

cardiomyopathy. National Heart, Lung, and Blood Institute and Offi ce of Rare Diseases (National Institutes of Health) Workshop recommendations and review. JAMA 2000; 283: 1183–88.

0

0·1

0·2

0·3

0·4

0·5

0·6

0·7

10 2 3 4 5

Time from diagnosis (years)

Recovered (26 of 92)

Mea

n LV

EF

Not recovered (66 of 92)

Figure 3: Echocardiographic data for left ventricular ejection fraction (LVEF) for 92 patients with peripartum cardiomyopathy followed up at Albert-Schweitzer Hospital, HaitiFrom Fett and colleagues11 with permission from Mayo Clinic Proceedings.

Page 75: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Seminar

692 www.thelancet.com Vol 368 August 19, 2006

3 Elkayam U, Akhter MW, Singh HS, et al. Pregnancy-associated cardiomyopathy: clinical characteristics and a comparison between early and late presentation. Circulation 2005; 111: 2050–55.

4 Ravikishore AG, Kaul UA, Sethi KK, Khalilullah M. Peripartum cardiomypathy: prognostic variables at initial evaluation. Int J Cardiol 1991; 32: 377–80.

5 Rizeq MN, Rickenbacher PR, Fowler MB, Billingham ME. Incidence of myocarditis in peripartum cardiomyopathy. Am J Cardiol 1994; 74: 474–77.

6 O’Connell JB, Costanzo-Nordin MR, Subramanian R, et al. Peripartum cardiomyopathy: clinical, hemodynamic, histologic and prognostic characteristics. J Am Coll Cardiol 1986; 8: 52–56.

7 Hibbard JU, Lindheimer M, Lang RM. A modifi ed defi nition for peripartum cardiomyopathy and prognosis based on echocardiography. Obstet Gynecol 1999; 94: 311–16.

8 Fett JD, Carraway RD, Dowell DL, King ME, Pierre R. Periptum cardiomyopathy in the Hospital Albert Schweitzer District of Haiti. Am J Obstet Gynecol 2002; 186: 1005–10.

9 Sliwa K, Skudicky D, Bergemann A, Candy G, Puren A, Sareli P. Peripartum cardiomyopathy: analysis of clinical outcome, left ventricular function, plasma levels of cytokines and Fas/Apo-1. J Am Coll Cardiol 2000; 35: 701–05.

10 Fett JD, Christie LG, Carraway RD, Sundstrom JB, Ansari AA, Murphy JG. Unrecognized peripartum cardiomyopathy in Haitian women. Int J Gynaecol Obstet 2005; 90: 161–66.

11 Fett JD, Christie LG, Carraway RD, Murphy JG. Five-year prospective study of the incidence and prognosis of peripartum cardiomyopathy at a single institution. Mayo Proceed 2005; 80: 1602–06

12 Desai D, Moodley J, Naidoo D. Peripartum cardiomyopathy: experiences at King Edward VIII Hospital, Durban, South Africa and a review of the literature. Trop Doct 1995; 25: 118–23.

13 Mielniczuk LM, Williams K, Davis DR, et al. Frequency of peripartum cardiomyopathy. Am J Cardiol 2006; 97: 1765-68.

14 Cunningham FG, Pritchard JA, Hankins GD, Anderson PL, Lucas MJ, Armstrong KF. Peripartum heart failure: idiopathic cardiomyopathy or compounding cardiovascular events? Obstet Gynecol 1986; 67: 157–68.

15 Brown CS, Bertolet BD. Peripartum cardiomyopathy: a comprehensive review. Am J Obstet Gynecol 1988; 178: 409–14.

16 Ford L, Abdullahi A, Anjorin FI, et al. The outcome of peripartum cardiac failure in Zaria, Nigeria. Q JM 1998; 91: 93–103.

17 Danbauchi SS. Echocardiographic features of peripartum cardiac failure: the Zaria syndrome. Trop Doct 2002; 32: 24–27.

18 Kane A, Dia AA, Diop IB, Sarr M, Ba SA, Diouf SM. Peripartum heart failure: the underestimated role of frequent diseases in the Sudan-Sahelian area. Dakar Med 2000; 45: 131–33.

19 Sanderson JE, Adesanya CO, Anjorin FI, Parry EH. Postpartum cardiac failure—heart failure due to volume overload? Am Heart J 1979; 97: 613–21.

20 Cenac A, Djibo A. Postpartum cardiac failure in Sudanese-Sahelian Africa: clinical prevalence in western Niger. Am J Trop Med Hyg 1998; 58: 319–23.

21 Sliwa K, Forster O, Libhaber E, et al. Peripartum cardiomyopathy: infl ammatory markers as predictors of outcome in 100 prospectively studied patients. Eur Heart J 2006; 27: 441–46.

22 Demakis JG, Rahimtoola SH, Sutton GC, et al. Natural course of peripartum cardiomyopathy. Circulation 1971; 44: 1053–60.

23 Homans DC. Peripartum cardiomyopathy. N Engl J Med 1985; 312: 1432–37.

24 Podewski E, Hilfi ker A, Hilfi ker-Kleiner D, Kaminski K, Drexler H. Stat 3 protects female hearts from postpartum cardiomyopathy in the mouse: the potential role of prolactin (abstract 2178). Munich: European Society of Cardiology Meeting, 2004.

25 Kuhl U, Pauschinger M, Seeberg B, et al. Viral persistence in the myocardium is associated with progressive cardiac dysfunction. Circulation 2005; 112: 1965–70.

26 Ansari AA, Fett JD, Carraway RD, Mayne AE, Onlamoon M, Sundstrom JB. Autoimmune mechanisms as the basis for human peripartum cardiomyopathy. Clin Rev Allergy Immunol 2002; 23: 289–312.

27 Fett JD, Sundstrom JB, Ansari AA, Combs GF Jr. Peripartum cardiomyopathy: a selenium disconnection and an autoimmune connection. Int J Cardiol 2002; 86: 311–15.

28 Hayakawa Y, Chandra M, Wenfeng M, et al. Inhibition of cardiac myocyte apoptosis improves cardiac function and abolishes mortality in peripartum cardiomyopathy of Gα(q) transgenic mice. Circulation 2003; 108: 1037-42.

29 Kothari SS. Aetiopathogenesis of peripartum cardiomyopathy: prolactin-selenium interaction. Int J Cardiol 1997; 60: 111–14.

30 Melvin KR, Richarson PJ, Olsen EG, Daly K, Jackson G. Peripartum cardiomyopathy due to myocarditis. N Engl J Med 1982; 307: 731–34.

31 Sanderson JE, Olsen EG, Gatei D. Peripartum heart disease: an endomyocardial biopsy study. Br Heart J 1986; 56: 285–91.

32 Midei MG, DeMent SH, Feldman AM, Hutchins GM, Baighman KL. Peripartum myocarditis and cardiomyopathy. Circulation 1990; 81: 922–28.

33 Rizeq MN, Rickenbacher PR, Fowler MB, Billingham ME. Incidence of myocarditis in peripartum cardiomyopathy. Am J Cardiol 1994; 74: 474–77.

34 Felkner GM, Thompson RE, Hare JM, et al. Underlying causes and long-term survival in patients with initially unexplained cardiomyopathy. N Engl J Med 2000; 342: 1077–84.

35 Bultmann BD, Klingel K, Nabauer M, Wallwiener D, Kandolf R. High prevalence of viral genomes and infl ammation in peripartum cardiomyopathy. Am J Obstet Gyn 2005; 193: 363–65.

36 Ardehali H, Kasper EK, Baughman KL. Diagnostic approach to the patient with cardiomyopathy: whom to biopsy. Am Heart J 2005; 149: 7–12.

37 Zimmermann O, Kochs M, Zwaka TP, et al. Myocardial biopsy based classifi cation and treatment in patients with dilated cardiomyopathy. Int J Cardiol 2005; 104: 92–100.

38 Warraich RS, Fett JD, Damasceno A, et al. Impact of pregnancy related heart failure on humoral immunity: clinical relevance of G3-subclass immunoglobulinss in peripartum cardiomyopathy. Am Heart J 2005; 150: 263–69.

39 Albert M, Glynn R, Buring J, Ridker PM. C-reactive protein levels among women of various ethnic groups living in the United States (from the women health study). Am J Cardiol 2004; 93: 1238–42.

40 Cenac A, Tourmen Y, Adehossi E, Couchouron N, Djibo A, Abgrall JF. The duo low plasma NT-PRO-BRAIN natriuretic peptide and C-reactive protein indicates a complete remission of peripartum cardiomyopathy. Int J Cardiol 2006; 108: 269–70.

41 Fairweather D, Yusung S, Frisancho S, et al. IL-12 receptor beta 1 and Toll-like receptor 4 increase IL-1 beta- and IL-18-associated myocarditis and coxsackievirus replication. J Immunol 2003; 170: 4731–37.

42 Huber SA. Cellular autoimmunity in myocarditis. In: Cooper LT, ed. Myocarditis: from bench to bedside. Totowa: Humana Press, 2003: 55–76.

43 Eriksson U, Ricci R, Hunziker L, et al. Dendritic cell-induced autoimmune heart failure requires cooperation between adaptive and innate immunity. Nat Med 2003; 9: 1484–90.

44 Maisch B, Arsen D, Ristic D. Humoral immune response in viral myocarditis. In: Cooper LT, ed. Myocarditis: from bench to bedside. Totowa: Humana Press, 2003: 77–108.

45 Mason JW. Viral latency: a link between myocarditis and dilated cardiomyopathy? J Mol Cell Cardiol 2002; 34: 695–98.

46 Ansari AA, Neckelmann N, Wang YC, Gravanis MB, Sell KW, Herskowitz A. Immunologic dialogue between cardiac myocytes, endothelial cells, and mononuclear cells. Clin Immunol Immunopathol 1993; 68: 208–14.

47 Feldman AM, McNamara DM. Myocarditis. N Eng J Med 2000; 343: 1388–98.

48 Yagoro A, Tada H, Hidaka Y, et al. Postpartum onset of acute heart failure possibly due to postpartum autoimmune myocarditis. A report of three cases. J Intern Med 1999; 245: 199–203.

49 Diao M, Diop IB, Kane A, et al. Electrocardiographic recording of long duration (Holter) of 24 hours during idiopathic cardiomyopathy of the peripartum. Arch Mal Coeur 2004; 97: 25–30.

50 Isezua SA, Njoku CH, Airede L, Yagoob I, Musa AA, Bello O. Case report: acute limb ischaemia and gangrene associated with peripartum cardiomyopathy. Niger Postgrad Med J 2005; 12: 237–40.

Page 76: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Seminar

www.thelancet.com Vol 368 August 19, 2006 693

51 Gagne PJ, Newman JB, Muhs BE. Ischemia due to peripartum cardiomyopathy threatening loss of a leg. Cardiol Young 2003; 13: 209–11.

52 Heims AK, Kittner SJ. Pregnancy and stroke. CNS Spectr 2005; 10: 580–87.

53 Box LC, Hanak V, Arciniegas JG. Dual coronary emboli in peripartum cardiomyopathy. Tex Heart Inst J 2004; 31: 442–44.

54 Dickfi eld T, Gagliardi JP, Marcos J, Russell SD. Peripartum cardiomyopathy presenting as an acute myocardial infarction. Mayo Clinic Proc 2002; 77: 500–01.

55 Fussell KM, Awad JA, Ware LB. Case of fulminant hepatic failure due to unrecognized peripartum cardiomyopathy. Crit Care Med 2005; 33: 891–93.

56 Murali S, Baldisseri MR. Peripartum cardiomyopathy. Crit Care Med 2005; 33 (suppl): S340–46.

57 Rathore SS, Wang Y, Krumholz HM. Sex-based diff erences in the eff ect of digoxin for the treatment of heart failure. N Engl J Med 2002; 347: 1403–11.

58 Adams KF Jr, Patterson JH, Gattis WA, et al. Relationship of serum digoxin concentration to mortality and morbidity in women in the digitalis investigation group trial: a retrospective analysis. J Am Coll Cardiol 2005; 46: 497–504.

59 Phillips SD, Warnes CA. Peripartum cardiomyopathy: current therapeutic perspectives. Curr Treat Options Cardiovasc Med 2004; 6: 481–88.

60 Aziz TM, Burgess MI, Acladious NN, et al. Heart transplantation for peripartum cardiomyopathy: a report of three cases and a literature review. Cardiovasc Surg 1999; 7: 565–67.

61 Godsel LM, Leon JS, Engman DM. Angiotensin converting enzyme inhibitors and angiotensin II receptor antagonists in experimental myocarditis. Curr Pharm Des 2003; 9: 723–35.

62 Ohtsuka T, Hamada M, Hiasa G, et al. Eff ect of beta-blockers on circulating levels of infl ammatory and anti-infl ammatory cytokines in patients with dilated cardiomyopathy. J Am Coll Cardiol 2001; 37: 412–17.

63 Pauschinger M, Rutschow W, Chandrasekharan K, et al. Carvedilol improves left ventricular function in murine coxsackievirus-induced active myocarditis associated with reduced myocardial interleukin-1beta and MMP-8 expression and a modulated immune response. Eur J Heart Fail 2005; 7: 444–52.

64 Sliwa K, Skukicky D, Candy G, Bergemann A, Hopley M, Sareli P. The addition of pentoxifylline to conventional therapy improves outcome in patients with peripartum cardiomyopathy. Eur J Heart Fail 2002; 4: 305–09.

65 Elkayam U, Padmini P, Tummala P, et al. Maternal and fetal outcomes of subsequent pregnancies in women with peripartum cardiomyopathy. N Engl J Med 2001; 344: 1567–71.

66 Sliwa K, Forster O, Zhanje F, Candy G, Kachope J, Essop R. Subsequent pregnancy in patients with peripartum cardiomyopathy. Am J Cardiol 2004; 93: 1441–43.

67 Fett JD, Christie LG, Murphy JG. Outcome of subsequent pregnancy after peripartum cardiomyopathy: a case series from Haiti. Ann Int Med 2006; 145: 30–34.

68 Staudt A, Schaper F, Stangl V, et al. Immunohistological changes in dilated cardiomyopathy induced by immunoadsorption therapy and subsequent immunoglobulin substitution. Circulation 2001; 103: 2681–86.

69 Felix SB, Staudt A, Landsberger M, et al. Removal of cardiodepressant antibodies in dilated cardiomyopathy by immunoadsorption. J Am Coll Cardiol 2002; 39: 646–52.

70 Frustaci A, Chimenti C, Calabrese F, Pierone M, Thiene G, Maseri A. Immunosuppressive therapy for active lymphocytic myocarditis: virological and immunologic profi le of responders versus nonresponders. Circulation 2003; 107: 857–63.

71 Cooper LT Jr. The natural history and role of immunoadsorption in dilated cardiomyopathy. J Clin Apher 2005; 19: 1–4.

72 Sliwa K, Damasceno A, Mayosi B. Cardiomyopathy in Africa. Circulation 2005; 112: 3577–83.

Page 77: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

Cardiogenic shock

Simon Topalian, MD; Fredric Ginsberg, MD, FACC; Joseph E. Parrillo, MD, FACC

Cardiogenic shock is a life-threatening emergency andthe most common cause ofdeath in patients hospitalized

with acute myocardial infarction. Studiesof this condition over the past 10 yrs havechanged the approach to cardiogenicshock patients so that there is now anopportunity to improve on the high mor-tality rate of this condition.

Definition

Cardiogenic shock (CS) is defined aspersistent hypotension and tissue hypo-perfusion due to cardiac dysfunction inthe presence of adequate intravascularvolume and left ventricular filling pres-sure. Clinical signs include hypotension,tachycardia, oliguria, cool extremities, andaltered mental status. Hemodynamic find-ings are sustained hypotension with sys-tolic blood pressure �90 mm Hg for �30mins, low cardiac index �2.2 L/min/m2,and elevated pulmonary artery occlusionpressure �15 mm Hg (1).

Incidence and Epidemiology

CS is a major complication of myocar-dial infarction. The incidence of CS hasremained stable over the past 3 decadesdespite advances in diagnostic and ther-apeutic modalities. In an early trial ofthrombolytic therapy for acute myocar-dial infarction, the incidence of CS com-plicating acute myocardial infarction was7.2% (2). In an observational communi-ty-wide study, the incidence of CS aver-aged 7.1% over a 23-yr period from 1975through 1997 (3). In a more recent anal-ysis of the National Registry of Myocar-dial Infarction (NRMI) covering the pe-riod from June 1995 through May 2004,CS developed in 8.6% of patients withacute myocardial infarction (ST-segmentelevation or left bundle branch block)hospitalized in 775 U.S. hospitals withrevascularization capability (4) (Fig. 1).

The prognosis of CS is extremely poor.Mortality rates were reported at 50% to80% in older series (1). In-hospital mor-tality in the SHould we emergently revas-cularize Occluded Coronaries for cardio-genic shocK (SHOCK) Trial Registry was60% (5). In the NRMI data, the overallin-hospital mortality decreased from60.3% in 1995 to 47.9% in 2004 (4).

In the NRMI database, 29% of patientswith CS were in shock as they presentedto hospital, and 71% developed CS afteradmission (4) (Fig. 1). Patients �75 yrsold were slightly more likely to presentwith CS. CS patients were more likely tohave a history of hypertension, dyslipide-mia, and prior coronary angioplasty. In

the SHOCK Trial Registry, the mediantime from onset of myocardial infarctionto shock was 7 hrs (5). Rates of recurrentmyocardial infarction or ischemia precip-itating CS were 9.3% and 19.7%, respec-tively. Infarction location was anterior in55% of cases and in multiple locationsin 50% (5).

CS can occur in the setting of ST-elevation myocardial infarction (STEMI)as well as non-ST-elevation myocardialinfarction (NSTEMI). In the Global Use ofStrategies to Open Occluded CoronaryArteries (GUSTO)-IIb trial, CS developedin 4.2% of STEMI and 2.5% of NSTEMIpatients. In the latter group, CS tended tooccur later after presentation (76.3 hrsvs. 9.6 hrs). NSTEMI patients with CSwere older and had higher rates of diabe-tes mellitus, prior myocardial infarction,heart failure, azotemia, bypass surgery,peripheral vascular disease, and three-vessel coronary disease. In-hospital and30-day mortality rates in STEMI andNSTEMI patients were similar (6, 7).

Patients with CS have extensive coro-nary artery disease. Angiographic datafrom the SHOCK Trial Registry revealedthat 53.4% of patients had three-vesseldisease and 15.5% had significant leftmain stenosis (8).

Etiology

Many conditions may lead to CS (Ta-ble 1). However, left ventricular failuredue to extensive acute myocardial infarc-tion remains the most common cause. In

From Cooper University Hospital, Camden, NJ (ST,JEP); Robert Wood Johnson Medical School at Cam-den, University of Medicine and Dentistry of New Jer-sey (FG, JEP); and Cooper Heart Institute (JEP).

Dr. Parrillo has disclosed that he holds consultan-cies with GlaxoSmithKline, Ortho Biotech, ArtesianTherapeutics, and Arginex. The remaining authorshave not disclosed any potential conflicts of interest.

For information regarding this article, E-mail: [email protected]

Copyright © 2007 by the Society of Critical CareMedicine and Lippincott Williams & Wilkins

DOI: 10.1097/01.CCM.0000296268.57993.90

Cardiogenic shock is the most common cause of death inpatients hospitalized with acute myocardial infarction and isassociated with a poor prognosis. More than 75% of cases aredue to extensive left ventricular infarction and ventricular failure.Other causes include right ventricular infarction and papillarymuscle rupture with acute severe mitral regurgitation. Activationof neurohormonal systems and the systemic inflammatory re-sponse worsens shock. To improve outcomes, cardiogenic shockneeds to be recognized early in its course and its cause needs tobe diagnosed rapidly. Treatment strategies using intra-aortic bal-

loon counterpulsation and emergency revascularization by per-cutaneous coronary interventions or coronary bypass surgeryhave been shown to improve outcomes. To decrease the incidenceof cardiogenic shock, public education regarding early presenta-tion to hospital in the course of acute chest pain is important.Emergency medical transport systems may need to take patientswith complicated acute myocardial infarction to hospitals withthe capability to perform urgent revascularization. (Crit Care Med2008; 36[Suppl.]:S66–S74)

S66 Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 78: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

the SHOCK Trial Registry, left ventricu-lar failure accounted for 78.5% of cases ofCS complicating acute myocardial infarc-tion. Acute severe mitral regurgitationaccounted for 6.9%, ventricular septalrupture 3.9%, isolated right ventricularinfarction 2.8%, and tamponade or car-diac rupture 1.4% (5). Other causes, suchas coexistent severe valvular heart dis-ease, excess �-blocker or calcium chan-nel blocker therapy, severe dilated cardio-myopathy, recent hemorrhage, or cardiac

catheterization laboratory complication,accounted for 6.7%.

Pathophysiology

Most commonly, CS occurs after amassive and extensive myocardial infarc-tion or severe myocardial ischemia lead-ing to impaired left ventricular function,reduced systolic contractility, and de-creased cardiac output and arterial bloodpressure. Coronary perfusion will de-crease and further compromise coronaryreserve. Compensatory neurohormonalresponses take place, which include acti-vation of the sympathetic and renin-angiotensin systems, leading to systemicvasoconstriction, tachycardia, and fluidretention. These mechanisms are mal-adaptive and worsen myocardial isch-emia. Thus, “ischemia begets ischemia,”leading to a progressive downward spiralof worsening ischemia, progressive dete-rioration of myocardial function, andworsening shock (1) (Fig. 2).

At the cellular level, inadequate oxy-gen delivery to myocytes affects cellularadenosine triphosphate production. Im-mediately, energy metabolism shifts fromaerobic to anaerobic glycolysis with re-sultant lactic acid production. Intracellu-lar ionic calcium rises and intracellularsodium competes to expel calcium (9). Ifhypoperfusion and ischemia are severe,as occurs in CS, myonecrosis ensues withmitochondrial swelling and subsequentplasma membrane disruption.

This ischemic cascade results in meta-bolic and biochemical alterations, whichlead to left ventricular diastolic dysfunctionfrom impaired myocardial relaxation anddecreased compliance. This leads to in-creased left ventricular filling pressures,manifesting as pulmonary congestion andedema. This in turn increases wall stress

and further compromises coronary perfu-sion. Echocardiographic data from theSHOCK Trial showed that restrictive leftventricular filling, as assessed by Dopplermitral inflow deceleration time, waspresent in most patients (60.9%) (10). Thisrestrictive pattern predicted pulmonary ar-tery occlusion pressures �20 mm Hg.

New evidence has emerged that hasled to expansion of the CS paradigm (11).Wide variations in left ventricular ejec-tion fraction, left ventricular size, andsystemic vascular resistance in patientswith CS suggest that pathophysiologicmechanisms of CS may vary among pa-tients. About one fifth of patients with CScomplicating myocardial infarction in theSHOCK Trial had clinical evidence of asystemic inflammatory response syn-drome, marked by fever, leukocytosis,and low systemic vascular resistance (12).Attempts to inhibit this inflammatory re-sponse focused on nitric oxide (NO), anendogenous vasodilator, which is pro-duced by nitric oxide synthase (NOS). In-ducible NOS (iNOS) is expressed atpathologic levels in many cells, especiallymyocytes, and has many deleterious ef-fects (11). High levels of iNOS are asso-ciated with left ventricular dysfunction(13). Early work with a nonisoform-specific NOS inhibitor, N-monomethyl-L-arginine (L-NMMA), had promising ef-fects on the hemodynamics of patientswith CS, including significantly increas-ing urine output and mean arterial bloodpressure (14). A mortality benefit wasseen in a small group of CS patientstreated with this NOS inhibitor (15) andled to larger randomized studies. How-ever, the Tilarginine Acetate InjectionRandomized International Study in Un-stable AMI Patients/Cardiogenic Shock(TRIUMPH) Trial, a phase III randomizedtrial to study the benefit of the NOS in-hibitor L-NMMA in patients with cardio-genic shock complicating AMI, did notshow any benefit (16).

Approach to the Patient WithCardiogenic Shock

The most important aspects of the ini-tial care of the patient with cardiogenicshock are recognizing the condition earlyin its course and understanding its cause.Rapid assessment of the history, physicalexamination, and chest radiograph ismandatory, recognizing the signs ofheart failure, pulmonary edema (some-times with clear lung fields on examina-

Figure 1. Frequency of cardiogenic shock among patients in the National Registry of MyocardialInfarction. Includes total shock at hospital presentation and shock that develops after hospitalpresentation as a clinical event. For hospitals with open-heart surgery and percutaneous coronaryintervention capability, the ST-elevation myocardial infarction population was 293,633. Data arethrough May 2004. Reproduced with permission from Babaev A, Frederick PD, Paste DJ, et al: Trendsin management and outcomes of patients with acute myocardial infarction complicated by cardiogenicshock. JAMA 2005; 294:448–454.

Table 1. Causes of cardiogenic shock

Acute myocardial infarctionPump failure

Large infarctionSmaller infarction with preexisting left

ventricular dysfunctionInfarction extensionSevere recurrent ischemiaInfarction expansion

Mechanical complicationsAcute mitral regurgitation caused by

papillary muscle ruptureVentricular septal defectFree-wall rupturePericardial tamponade

Right ventricular infarctionOther conditions

End-stage cardiomyopathyMyocarditisMyocardial contusionProlonged cardiopulmonary bypassSeptic shock with severe myocardial

depressionLeft ventricular outflow tract obstruction

Aortic stenosisHypertropic obstructive cardiomyopathy

Obstruction to left ventricular fillingMitral stenosisLeft atrial myxoma

Acute mitral regurgitation (chordal rupture)Acute aortic insufficiencyAcute massive pulmonary embolismAcute stress cardiomyopathyPheochromocytoma

S67Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 79: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

tion), and tissue hypoperfusion, manifest-ing as low blood pressure, rapid heartrate, agitation, confusion, oliguria, cya-nosis, and cool and clammy skin. Appre-ciating the electrocardiographic signs ofacute myocardial ischemia, myocardialinfarction, new left bundle branch block,and arrhythmias is critical. Rapid echo-cardiographic assessment is key. Theecho-Doppler study will assess regionaland global left ventricular function, rightventricular size and function, the pres-ence of mitral regurgitation and othervalve abnormalities, pericardial effusion,and possible septal rupture. Echocardio-graphic assessment of left ventricularejection fraction was found to be similarto ejection fraction as assessed by leftventriculography and can be used as thesole initial evaluation of systolic left ven-tricular function in patients with CS (17).Differential diagnoses to consider in pa-

tients with CS include hemorrhage, sep-sis, aortic dissection, and massive pulmo-nary embolism.

Patients must be assessed regardingthe need for sedation, intubation, andmechanical ventilation in order to cor-rect hypoxemia and reduce the work ofbreathing (1, 18, 19). Initial medicaltherapy includes intravenous fluid chal-lenge for patients with significant hy-potension, if there is no evidence forpulmonary edema or significant eleva-tion of jugular venous pressure. If CSis due to acute myocardial infarctionor ischemia, emergency cardiac cathe-terization and revascularization needto take place in patients believed suit-able for this approach (discussed subse-quently).

The use of an intra-arterial catheter ishelpful in managing patients in shock(20). Pulmonary artery catheterization

can assist in the precise measurement ofvolume status, left and right ventricularfilling pressures, and cardiac output. It isalso valuable in diagnosing right ventric-ular infarction and the mechanical com-plications of acute myocardial infarction(Table 2). Hemodynamic measurementscan help guide fluid management and theuse of inotropic agents and vasopressors.The American College of Cardiology/American Heart Association (ACC/AHA)guidelines list pulmonary artery catheter-ization as a class I recommendation inpatients with hypotension not respondingto fluid administration or when mechan-ical complications of myocardial infarc-tion are suspected and echocardiographyis not available. Pulmonary artery cathe-terization is a class IIa recommendationfor patients in CS who have persistentsigns of hypoperfusion and in patientsreceiving inotropic and vasopressor drugs(20, 21).

The goal of the initial medical ther-apy of CS is to maintain arterial pres-sure adequate for tissue perfusion. Ini-tially, dopamine is the drug of choicebecause it acts as an inotrope as well asa vasopressor. Intravenous norepineph-rine is a more potent vasoconstrictor,with somewhat less effect on heart rate,and should be used in patients withmore severe hypotension. These drugsincrease heart rate and systemic vascu-lar resistance and thus increase myo-cardial oxygen demand, and they mayaggravate ischemia and lead to cardiacarrhythmias. Doses should be adjustedto the lowest levels that improve tissueperfusion (1, 18, 20).

Dobutamine, an inotrope with arterialdilator properties, can be used in patientswith less severe hypotension or can becombined with vasopressors to improvecardiac output (18). There also have beenreports of the effective use of vasopressinin patients with CS (22). Intravenous di-uretics are used in patients with pulmo-nary edema and elevated pulmonary ar-tery occlusion pressure. Aspirin shouldbe given to patients with acute myocar-dial infarction. Intravenous amiodaronecan be given for patients with severe ar-rhythmia. The use of �-blockers and ni-trates should be avoided in the acutephase (20).

Some patients will demonstratesigns of tissue hypoperfusion with sys-tolic blood pressure �90 mm Hg. Thishas been termed nonhypotensive car-diogenic shock or preshock. In the

Figure 2. Classic shock paradigm, as illustrated by S. Hollenberg, is shown in black. The influence ofthe inflammatory response syndrome initiated by a large myocardial infarction is illustrated in gray.LVEDP, left ventricular end-diastolic pressure; NO, nitric oxide; iNOS, inducible NO synthase; SVR,systemic vascular resistance. Reproduced with permission from Hochman JS: Cardiogenic shockcomplicating acute myocardial infarction: Expanding the paradigm. Circulation 2003; 107:2998 –3002.

Table 2. Hemodynamic profiles

Left ventricular shock High PAOP, low CO, high SVRRight ventricular shock High RA, RA/PAOP �0.8, exaggerated RA y descent, RV square root signMitral regurgitation Large PAOP V waveVentricular septal defect Large PAOP V wave, oxygen saturation step-up (�5%) from RA to RVPericardial tamponade Equalization of diastolic pressures �20 mm Hg

PAOP, pulmonary artery occlusion pressure; CO, cardiac output; SVR, systemic vascular resistance;RA, right atrial; RV, right ventricular.

S68 Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 80: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

SHOCK Trial Registry, these patientsdemonstrated the hemodynamic profileof elevated pulmonary artery occlusionpressure with low cardiac index andhigh systemic vascular resistance. Thehospital mortality rate in this group ofpatients was 43%, very high but lowerthan in patients diagnosed with full-blown cardiogenic shock. The mortalityin patients with preshock compareswith a mortality rate of 26% in patientswith hypotension without signs of hy-poperfusion. This preshock state needsto be recognized early so that appro-priate, urgent diagnostic and ther-apeutic measures can be prescribed (23)(Fig. 3).

Intra-Aortic BalloonCounterpulsation (IABP)

IABP is very useful to support patientswith CS. This device will increase coro-nary blood flow, decrease left ventricularafterload, and decrease left ventricularend-diastolic pressure without increasingoxygen demand (1, 19, 24). Cardiac out-put is increased only modestly, and thisdevice does not provide total circulatorysupport (24).

The efficacy of IABP in acute myo-cardial infarction complicated by CShas not been evaluated in a randomizedcontrolled trial. In nonrandomized tri-als, the use of IABP is associated withdecreased mortality, but the use of this

device is consistently associated withmore frequent use of revascularizationtherapies and other aggressive support-ive measures (25). Its use was not asso-ciated with improved survival in patientsundergoing primary percutaneous cor-onary intervention (PCI) in the NRMIdatabase.

Complications of IABP include bleed-ing, thrombocytopenia, hemolysis, legischemia, aortic dissection, femoral ar-tery injury, thromboembolism, and sep-sis. The complication rate has been re-duced with percutaneous insertion andwith smaller pumps (26). Complicationrates have been reported at 10% to 30%,with major complication rates of 2.5% to

Figure 3. Emergency management of complicated ST-segment elevation myocardial infarction. American College of Cardiology/American Heart AssociationPractice Guidelines 2004. BP, blood pressure; IV, intravenous; MI, myocardial. Adapted with permission from Anbe DT, Armstrong PW, Bates ER, et al:ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction: A report of the American College of Cardiology/AmericanHeart Association Task Force on Practice Guidelines (Committee to Revise the 1999 Guidelines for the Management of Patients With Acute MyocardialInfarction). ACC/AHA Practice Guidelines 2004. http://www.acc.org/clinical/guidelines/stemi/index.pdf. Accessed January 2007.

S69Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 81: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

3.0% (25, 26). Patients usually must beanticoagulated during IABP use. Contra-indications to the use of IABP includesevere aortic insufficiency, severe periph-eral vascular disease, and aortic aneu-rysm (18).

Current ACC/AHA guidelines list IABPas a class I recommendation for patientswith low cardiac output states, hypoten-sion, and CS not responding quickly toother measures (20). In the NRMI data-base, IABP was used in 39% of cases withCS, and the frequency of IABP use did notchange over the 10-yr period 1995–2004(4). In the Benchmark CounterpulsationOutcomes Registry, CS accounted for27% of the use of IABP and was associ-ated with a mortality rate of 38.7% (26).IABP is also recommended for use in pa-tients with acute myocardial infarctioncomplicated by severe mitral regurgita-tion or ventricular septal rupture, beforeurgent cardiac surgery, as well as in pa-tients with right ventricular infarctionnot responding to fluid challenge andinotropic therapy.

In summary, the use of IABP in CSimproves the short-term hemodynamicprofile and may allow for improvement infunction of ischemic myocardium. Itlikely does not improve outcomes unlessit is combined with definitive coronaryrevascularization (25).

Emergency Revascularization

Many observations and nonrandom-ized studies have shown that the majorityof patients with acute myocardial infarc-tion who develop CS do so hours afteronset of infarction, often after hospital

presentation (2, 4, 5, 27). Shock that hasa delayed onset results from infarctionextension, reocclusion of a previouslypatent coronary artery, recurrent isch-emia, or decompensation of left ventric-ular function in the noninfarcted zonebecause of metabolic derangements (1).The most successful strategy to success-fully treat acute myocardial infarction israpid restoration of flow in the infarct-related artery (28), and primary coronaryangioplasty results in better outcomesthan fibrinolytic therapy (29). Many re-ports have suggested that early mechan-ical revascularization with either PCI orcoronary artery bypass graft surgery(CABG) is associated with survival bene-fit. The landmark SHOCK trial prospec-tively randomized 302 patients with CSdue to acute myocardial infarction andleft ventricular failure to emergency earlyrevascularization (ERV) with PCI orCABG vs. initial medical stabilization(IMS) with drug therapy and IABP. PCIaccounted for 64% of revascularizationand CABG was performed in 36%. The30-day mortality rate, the primary out-come measured, was lower in the earlyrevascularization group (47% vs. 56%),which was not statistically significant.However, late mortality rates were signif-icantly improved in patients who receivedERV (30). At 6 months, 1 yr, and 6 yrs, astatistically significant absolute survivaldifference of 13% was seen in patientswho received ERV (31, 32). At 6 yrs, over-all survival was 32.8% in the ERV groupand 19.6% in the IMS group. In patientswho survived hospitalization, the 6-yrsurvival rate in patients with ERV was62.4% vs. 44.4% in IMS patients. (Fig. 4)There was no significant difference in

long-term survival between patientstreated with PCI or CABG. Quality of lifemeasures were also better in patients whohad received ERV (33).

Unfortunately, invasive procedurescontinue to be underused in patients withCS (4, 5) In the NRMI, primary PCI useincreased from 27.4% in 1995 to only54.4% in 2004 (4 yrs after the SHOCKtrial was published). CABG was used inonly 3.2% of patients. This increased useof PCI for CS was likely translated into amortality benefit (60.3% mortality in1995, declining to 47.9% in 2004) (4).Patients who were transferred to hospi-tals with revascularization capability alsohad better outcome with ERV (34). Thisbenefit from early mechanical revascular-ization was incorporated in the ACC/AHApractice guidelines. Both PCI and CABGare class I recommendations for patients�75 yrs old with STEMI or acute myo-cardial infarction with left bundle branchblock, who develop shock within 36 hrs ofonset of myocardial infarction (20, 35).

Patients �75 yrs old had worse out-comes with ERV in the SHOCK trial com-pared with IMS. However, the number ofpatients treated with ERV in this agegroup was relatively small (30–32). Anal-ysis from the SHOCK registry showedthat elderly patients were treated less ag-gressively, and their in-hospital mortalitywas higher compared with younger pa-tients (76% vs. 55%). However, outcomesin elderly patients in the registry whowere treated with ERV were better thanin patients who were not revascularized(36). Clearly, patient selection plays a keyrole. Elderly patients with good func-tional status, shorter duration of shock,

Figure 4. Among all patients, the survival rates in the early revascularization (ERV) and initial medical stabilization (IMS) groups, respectively, were 41.4%vs. 28.3% at 3 yrs and 32.8% vs. 19.6% at 6 yrs. With exclusion of eight patients with aortic dissection, tamponade, or severe mitral regurgitation identifiedshortly after randomization, the survival curves remained significantly different (p � .02), with a 14.0% absolute difference at 6 yrs. Among hospitalsurvivors, the survival rates in the ERV and IMS groups, respectively, were 78.8% vs. 64.3% at 3 yrs and 62.4% vs. 44.4% at 6 yrs. Reproduced withpermission from Hochman JS, Sleeper LA, Webb JG, et al: Early revascularization and long-term survival in cardiogenic shock complicating acutemyocardial infarction. JAMA 2006: 295:2511–2515.

S70 Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 82: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

and absence of serious medical comor-bidities should be carefully selected foraggressive therapies, including revascu-larization (21).

Fibrinolytic Therapy inCardiogenic Shock

Urgent transfer to the cardiac cathe-terization laboratory for coronary angiog-raphy and emergency coronary interven-tion has largely supplanted thrombolytictherapy as first-line treatment for pa-tients with acute myocardial infarctionand CS. Fibrinolytics are not as effectiveas accomplishing reperfusion in STEMIwith CS as in patients with STEMI with-out cardiogenic shock (1, 18). This ishypothesized to be due to decreased pen-etration of the coronary thrombus by fi-brinolytics in hypotensive patients, agreater incidence of coronary artery re-occlusion after thrombolysis, and longertimes required to achieve coronary pa-tency (1, 18, 19). In an overview of fi-brinolytic trials, analysis of patients in CSshowed nonsignificant reductions inmortality with fibrinolytic therapy. Sub-group analyses in individual trials haveshown no effect on mortality (18). In theNRMI database, IABP combined withthrombolytic therapy was associated witha significantly lower mortality rate (49%)compared with thrombolytic therapyalone (67%) (25). In the SHOCK TrialRegistry, the addition of IABP to throm-bolytic therapy decreased mortality sig-nificantly from 63% with thrombolysis

alone to 47% with thrombolysis andIABP. Thrombolytic therapy was associ-ated with a lower mortality than in pa-tients who did not receive any reperfu-sion therapy (37). However, mortality inCS patients treated with fibrinolytictherapy combined with IABP was stillhigher than in patients who receivedrevascularization with PCI or coronarybypass surgery.

The development of CS after fibrino-lytic therapy is more likely to occur inolder patients, patients with Killip classII–III heart failure on presentation, andpatients with higher heart rate andlower blood pressure on admission (38).When thrombolytic therapy was givento patients with acute myocardial in-farction before hospital arrival, duringambulance transport, the incidence ofsubsequent CS was lowered from 11.5%to 6.8% (39).

Current guidelines have relegatedthrombolytic therapy for CS as a class Irecommendation only in patients withSTEMI who are unsuitable for invasivetherapy with PCI or bypass surgery(20). In the NMRI observational data-base, the use of thrombolytic therapyfor acute myocardial infarction and CSdeclined from 19.9% to 5.6% of cases inthe years 1995–2004 (4). The strategy ofadministering fibrinolytics with orwithout IABP is recommended if pa-tients present to a hospital that doesnot have a catheterization laboratory orwhen there will be unavoidable delaysin transport to the catheterization lab-oratory (1).

Mechanical Complications ofAcute Myocardial Infarction(Figure 5)

Right Ventricular Infarction. Rightventricular myocardial infarction (RVI)can accompany acute inferior wall myo-cardial infarction and lead to cardiogenicshock. It is estimated that 10% to 15% ofinferior wall myocardial infarctions arecomplicated by hemodynamically signifi-cant RVI (1, 20). RVI accounted for 2.8%of cases of CS in the SHOCK Trial Reg-istry (5). Acute right ventricular dysfunc-tion and right ventricular failure lead todecreased left ventricular preload, de-creased cardiac output, and cardiogenicshock. Right ventricular dilation andshift of the intraventricular septum to-ward the left ventricle can further com-promise left ventricular function andworsen cardiogenic shock.

RVI results from occlusion of the rightcoronary artery proximal to the origin ofthe right ventricular branches. In the erabefore PCI for acute myocardial infarc-tion, in-hospital mortality from RVI wasaround 7.1%, less than the mortality ratefor anterior myocardial infarction buthigher than that for inferior myocardialinfarction without RVI (40). RVI was alsoan independent predictor of 6-monthmortality and was associated with higherrates of CS and sustained ventricular ar-rhythmia.

The diagnosis of RVI should bestrongly considered in patients whopresent with acute inferior wall myocar-dial infarction with hypotension, clearlung fields, and elevated jugular venouspressure or a positive Kussmaul’s sign.Diagnostic findings on electrocardiograminclude �1-mm ST-segment elevation inV1 and in the right precordial lead V4R,although these findings may be transient.Emergency transthoracic echocardio-gram will show a dilated, hypocontractileright ventricle and may show bulging ofthe septum into the left ventricle. Rightheart catheterization will demonstrate amean right atrial pressure �10 mm Hgand right atrial pressure �80% of pulmo-nary artery occlusion pressure. Cardiacindex will be low.

Initial therapy for hypotensive pa-tients with RVI is a fluid challenge ifjugular venous pressure is not elevated.Up to 1 L of intravenous saline should beinfused, which may correct hypotension.Larger volumes may cause significantright ventricular dilation and impair leftventricular output. Inotropic medica-

Figure 5. Mechanical complications of ST-elevation myocardial infarction. Phy Exam, physicalexamination; PA, pulmonary artery; MI, myocardial infarction; JVD, jugular venous distention; EMD,electromechanical dissociation; Peric., pericardial; Diast Press Equal., diastolic pressure equalization;Regurg., regurgitation; PCW, pulmonary artery occlusion pressure. Reproduced with permission fromAnbe DT, Armstrong PW, Bates ER, et al: ACC/AHA guidelines for the management of patients withST-elevation myocardial infarction: A report of the American College of Cardiology/American HeartAssociation Task Force on Practice Guidelines (Committee to Revise the 1999 Guidelines for theManagement of Patients With Acute Myocardial Infarction). ACC/AHA Practice Guidelines 2004.http://www.acc.org/clinical/guidelines/stemi/index.pdf. Accessed January 2007.

S71Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 83: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

tions and IABP are useful in patients whodo not respond to fluid challenge (20).IABP helps to decrease wall stress andincrease coronary perfusion pressure (1).Bradycardia and heart block should becorrected, and atrioventricular sequentialpacing may be necessary to maintainatrioventricular synchrony and effectivecardiac output.

Most patients with RVI will have spon-taneous recovery of right ventricularfunction, but this may occur slowly andmay be incomplete. Urgent revasculariza-tion with PCI is now the cornerstone oftherapy. In a study of 53 patients withacute inferior wall myocardial infarctioncomplicated by RVI, all 53 patients un-derwent emergency PCI (41). The rightcoronary artery was the culprit vessel inall cases. Seventy-seven percent of pa-tients had successful intervention andreperfusion, and these patients demon-strated recovery of right ventricular func-tion, decreased right heart pressures, andreduction in right ventricular size within1 hr, with 95% recovery of normal rightventricular function in 3–5 days. In-hospital mortality was 2.4%. In the 12patients with unsuccessful reperfusion,right ventricular dysfunction persisted at24 hrs and only improved slowly duringhospitalization. Ten of these 12 patientsrequired support with inotropic drugs orIABP, and in-hospital mortality was58.3%. Emergency revascularization ef-forts in these patients is now a class Irecommendation in ACC/AHA guidelinesfor the treatment of acute myocardial in-farction. If coronary bypass surgery is be-lieved to be needed for multivessel dis-ease and significant right ventriculardysfunction is present, then coronary by-pass should be delayed for 4 wks, if pos-sible, to allow for recovery of right ven-tricular function (20).

Acute Severe Mitral Regurgitation.Acute severe mitral regurgitation, due toinfarction and rupture of the head of apapillary muscle, is an uncommon causeof cardiogenic shock. In the SHOCK TrialRegistry, 6.9% of 1,190 patients withcardiogenic shock had acute severe mi-tral regurgitation (5, 42). Acute severemitral regurgitation was more likely tocomplicate inferior and/or posterior myo-cardial infarction (87% of cases of acutemitral regurgitation) than anterior myo-cardial infarction (34%). Acute severe mi-tral regurgitation usually occurs withinthe first 24 hrs of acute myocardial in-farction or may present at days 3–5. Thediagnosis of acute severe mitral regurgi-

tation should be suspected in patientswho present with acute pulmonaryedema complicating acute myocardial in-farction, especially inferior wall myocar-dial infarction. The murmur of mitralregurgitation may be loud but also maybe relatively unimpressive due to high leftatrial pressures and a lower left ventricular/left atrial systolic gradient. Diagnosis ismade by urgent echocardiography.

Acute severe mitral regurgitation isassociated with a high in-hospital mortal-ity. In the SHOCK Trial Registry, patientswho underwent urgent mitral valve sur-gery had a 40% mortality rate. Patientswho did not receive surgery had a 71%mortality rate. The overall 55% hospitalmortality rate was not different from theregistry cohort with cardiogenic shockdue to left ventricular failure (42).

In patients with acute severe mitralregurgitation, early diagnosis and aggres-sive support with inotropic drugs, IABP,and vasodilators (if blood pressure allows)are vital in appropriately selected pa-tients. The ACC/AHA guidelines list ur-gent cardiac surgical repair as a class Irecommendation (20).

Postinfarction Ventricular SeptalRupture. Rupture of the intraventricularseptum (VSR) can complicate acute myo-cardial infarction and lead to CS. Thisoften occurs in the first 24 hrs of infarc-tion. This condition occurred in �1% ofpatients with STEMI in the GUSTO Istudy (20, 21) and accounted for 3.9% ofthe cases of CS in the SHOCK Trial Reg-istry (15, 31). VSR may complicate eitheranterior or inferior wall STEMI. Patientswill present with shock and pulmonaryedema, and a loud holosystolic murmuris present on physical examination. Diag-nosis is made by urgent Doppler echocar-diography. A pattern of right ventricularvolume overload is seen, with Dopplerevidence of left-to-right shunting at ven-tricular level. The septal rupture may bevisualized. Right heart catheterizationwill show higher oxygen saturations inthe pulmonary artery than in the rightatrium due to left-to-right shunting.However, when the diagnosis of VSR ismade by echocardiography, performanceof right heart catheterization should notdelay surgical therapy.

Urgent surgical repair of VSR is a classI recommendation in the ACC/AHAguidelines. However, this condition is as-sociated with a very high mortality ratewith surgical or medical therapy. Waitingfor several days before surgery in order tolet healing occur is not recommended

because many patients will die duringthis waiting period. In the SHOCK TrialRegistry, 31 of 55 patients with VSR un-derwent surgery, with a mortality of 81%.Only 4% of patients survived without sur-gery (43). In the GUSTO I trial, 30-daymortality was 73.8% (21). Another seriesof 76 patients with VSR who underwentsurgery reported that 79% of patients pre-sented with CS, and 30-day postoperativemortality was 49% for these patients (44).

Left Ventricular Free Wall Ruptureand Cardiac Tamponade. Left ventricularfree wall rupture is an uncommon, lethalcomplication of acute myocardial infarc-tion. It is estimated to occur in 1% to 6%of patients with acute myocardial infarc-tion (20), although the true incidence isdifficult to ascertain as the majority ofpatients will die immediately after rup-ture from electromechanical dissocia-tion. However, possibly 30% of free wallrupture may be spontaneously sealed offby elevated intrapericardial pressure fromhemopericardium, and these patientsmay present with CS complicating acutemyocardial infarction. The incidence offree wall rupture has decreased in the eraof thrombolytic and primary angioplastytherapy for acute myocardial infarction.The diagnosis is made by emergencyechocardiography, which shows a signif-icant-sized pericardial effusion, eitherloculated or diffuse, and may show thearea of rupture.

In the SHOCK Trial Registry, 1.4% ofcases of CS were caused by free wall rup-ture and usually occurred in the first 24hrs of infarction (2, 45). Twenty-one of 28patients underwent surgery, with a 62%mortality rate. Six patients underwentpericardiocentesis only, and three sur-vived. Another series listed operativemortality rates of 24% to 52% (46).

Ventricular Assist Devices(VADs)

VADs have been used in small series ofhighly selected patients with CS refrac-tory to IABP and reperfusion strategies.The use of VADs should be considered inpatients with very low cardiac output,�1.2 L/min/m2 (19). Newer VADs can beinserted percutaneously. The Tandem-Heart percutaneous VAD, inserted in thecatheterization laboratory, uses a cathe-ter directed into the left atrium via atransseptal puncture, unloading the leftatrium and left ventricle. Blood is thenpumped into a 15- to 17-Fr catheter in-serted in the femoral artery, producing

S72 Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 84: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

flows of 3.5–4.0 L/min. One report de-scribed 11 patients with acute myocardialinfarction and CS refractory to inotropicand IABP therapy who received percuta-neous VADs. These patients were sup-ported for an average of 89 hrs, withmean cardiac index during VAD therapyof 2.6 L/min/m2. It was concluded fromthis small series that percutaneous VADscould be effective as a bridge to implantedVADs or cardiac transplant therapy, witha low incidence of adverse effects. Thisdevice cannot be used in patients withright ventricular failure or severe periph-eral vascular disease (24, 47).

Implanted VADs have also been usedin patients with CS. In 49 patients treatedwith this device, 78% were successfullybridged to transplant. VADs were placedat an average of 6 days after myocardialinfarction, and patients were supportedfor a mean time of 56 days until trans-plant. In-hospital mortality was only 33%in this group of patients (48). These pa-tients had a 31% incidence in the needfor dialysis and had a relatively high rateof infectious complications. Another se-ries of 18 patients with CS received sup-port with extracorporeal membrane oxy-genation and/or VAD and cardiactransplant therapy. In these highly se-lected patients, hospital mortality was33% and 5-yr survival was 30% (49).

Prevention of CS

Since most patients who develop CSafter acute myocardial infarction do so atsome time after presentation to hospital,there is an opportunity to prevent theoccurrence of CS in individual patients,to reduce the overall incidence of CS, andto significantly improve outcomes in pa-tients who develop this potentially lethalcomplication. The first step in preventionof CS involves educating the public, en-couraging patients to present to hospitalvery early after the development of sig-nificant chest pain or other symptoms ofacute myocardial infarction. Patientsshould be directed to call the emergencyambulance system, so that transport toan emergency department can occurquickly. In addition, hospital transportsystems need to be developed to allowpatients with STEMI to be transporteddirectly to hospitals with PCI and coro-nary surgery capabilities, avoiding delaysinherent when acute myocardial infarc-tion patients are admitted to hospitalswithout these facilities and then emer-gently transferred to tertiary centers. If

patients receive thrombolytic therapy foracute myocardial infarction in hospitalswithout revascularization capabilities,signs of failed thrombolysis should berecognized early, and these patientsshould be transferred quickly to tertiarycenters with revascularization capabili-ties.

In patients hospitalized with acutemyocardial infarction, prompt recogni-tion of the preshock state or the earlysigns of CS is vital. Once the diagnosis ofCS is made, rapid evaluation and institu-tion of supportive medical therapy, inser-tion of the IABP, and emergency revascu-larization need to be performed. Admissionof patients to hospitals with revascular-ization capabilities, or even directly tothe catheterization laboratory, has led tothe greater utilization of these lifesavingtherapies with improved mortality ratesin several nonrandomized studies (30, 31,50). It is unlikely that further large ran-domized controlled trials in patients withCS will be carried out. It is now necessaryto improve systems of care in order totranslate the benefits of early revascular-ization to wide implementation.

REFERENCES

1. Hollenberg SM, Kavinsky CJ, Parrillo JE:Cardiogenic shock. Ann Intern Med 1999;131:47–59

2. Holmes DR Jr, Bates ER, Kleiman NS, et al:Contemporary reperfusion therapy for car-diogenic shock after myocardial infarction:The GUSTO-I trial experience. J Am Coll Car-diol 1995; 26:668–674

3. Goldberg RJ, Samad NA, Yarzebski U, et al:Temporal trends in cardiogenic shock com-plicating acute myocardial infarction. N EnglJ Med 1999; 340:1162–1168

4. Babaev A, Frederick PD, Paste DJ, et al:Trends in management and outcomes of pa-tients with acute myocardial infarction com-plicated by cardiogenic shock. JAMA 2005;294:448–454

5. Hochman JS, Buller CE, Sleeper LA, et al:Cardiogenic shock complicating acute myo-cardial infarction—Etiologies, managementand outcome: A report from the SHOCKTrial Registry. J Am Coll Cardiol 2000; 36:1063–1070

6. Holmes DR Jr, Berger PB, Hochman JS, et al:Cardiogenic shock in patients with acuteischemic syndromes with and without ST-segment elevation. Circulation 1999; 100:2067–2073

7. Jacobs AK, Franch JK, Col J, et al: Cardio-genic shock with non-ST segment elevationmyocardial infarction: A report from theSHOCK Trial Registry. J Am Coll Cardiol2000; 36:1091–1096

8. Chiu Wong S, Sanborn T, Sleeper LA, et al:

Angiographic findings and clinical correlatesin patients with cardiogenic shock compli-cating acute myocardial infarction: A reportfrom the SHOCK Trial Registry. J Am CollCardiol 2000; 36:1077–1083

9. Kloner RA, Bolli R, Marban E, et al: Medicaland cellular implication of stunning, hiber-nation, and preconditioning: An NHLBIworkshop. Circulation 1998; 97:1848–1867

10. Reynolds HR, Anand SK, Fox JM, et al: Re-strictive physiology shock: Observationsfrom echocardiography. Am Heart J 2006;151:890.e9–890.e15

11. Hochman JS: Cardiogenic shock complicat-ing acute myocardial infarction: Expandingthe paradigm. Circulation 2003; 107:2998–3002

12. Kohsaka S, Menon V, Lowe AM, et al: Sys-temic inflammatory response syndrome afteracute myocardial infarction complicated bycardiogenic shock. Arch Intern Med 2005;165:1643–1650

13. Feng O, Lu X, Jones DL, et al: Increasedinducible nitric oxide synthase expressioncontributes to myocardial dysfunction andhigher mortality after myocardial infarctionin mice. Circulation 2001; 104:700–704

14. Cotter G, Kaluski E, Blatt A, et al: L-MNMA(a nitric oxide synthase inhibitor) is effectivein the treatment of cardiogenic shock. Cir-culation 2000; 101:1359–1361

15. Cotter G, Kaluski E, Milo O, et al: LINCS:L-NAME (a nitric oxide synthase inhibitor inthe treatment of refractory shock: A prospec-tive randomized study. Eur Heart J 2003;23:1287–1295

16. TRIUMPH Investigators, Alexander JH, Reyn-olds HR, et al: Effect of tilarginine acetate inpatients with acute myocardial infarctionand cardiogenic shock: The TRIUMPH ran-domized controlled trial. JAMA 2007; 297:1657–1666

17. Berkowitz MJ, Picard MH, Harkness S, et al:Echocardiographic and angiographic corre-lations in patients with cardiogenic shocksecondary to acute myocardial infarction.Am J Cardiol 2006; 98:1004–1008

18. Ellis TC, Lev E, Yazbek NF, et al: Therapeuticstrategies for cardiogenic shock, 2006. CurrTreat Options Cardiovasc Med 2006; 8:79–94

19. Duvernoy CS, Bates ER: Management of car-diogenic shock attributable to acute myocar-dial infarction in the reperfusion era. J In-tensive Care Med 2005; 4:188–198

20. Anbe DT, Armstrong PW, Bates ER, et al:ACC/AHA guidelines for the management ofpatients with ST-elevation myocardial infarc-tion: A report of the American College ofCardiology/American Heart Association TaskForce on Practice Guidelines (Committee toRevise the 1999 Guidelines for the Manage-ment of Patients With Acute Myocardial In-farction). ACC/AHA Practice Guidelines2004. http://www.acc.org/clinical/guidelines/stemi/index.pdf. Accessed January 2007.

21. Menon V, Hochman JS: Management of car-diogenic shock complicating acute myocar-dial infarction. Heart 2003; 88:531–537

S73Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)

Page 85: REVIEWARTICLE Diastolic heart failure in …...systolic heart failure.14 55 93 Recent studies reported that 16–30% of patients with diastolic heart failure had to be readmitted within

22. Mann HJ, Nolan PE Jr: Update on the man-agement of cardiogenic shock. Curr OpinCrit Care 2006; 12:431–436

23. Menon V, Slater JN, White HD, et al: Acutemyocardial infarction complicated by sys-temic hypoperfusion without hypotension:Report of the SHCOK Trial Registry. Am J Med2000; 108:374–380

24. Lee MS, Makkar RR: Percutaneous left ven-tricular support devices. Cardiol Clin 2006;24:265–275

25. Trost JC, Hillis LD: Intra-aortic ballooncounterpulsation. Am J Cardiol 2006; 97:1391–1398

26. Stone GW, Ohman M, Miller MF, et al: Con-temporary utilization and outcomes of intra-aortic balloon counterpulsation in acutemyocardial infarction. J Am Coll Cardiol2003; 41:1940–1945

27. Webb JG, Sleeper LA, Buller CE, et al: Impli-cations of timing of onset of cardiogenicshock after acute myocardial infarction: Areport from the SHOCK trial registry. J AmColl Cardiol 2000; 36:1084–1090

28. The effects of tissue plasminogen activator,streptokinase, or both on coronary arterypatency, ventricular function, and survivalafter acute myocardial infarction. TheGUSTO Angiographic Investigators. N EnglJ Med 1993; 329:1615–1522

29. Keeley EC, Boura JA, Grines CL, et al: Pri-mary angioplasty versus intravenous throm-bolytic therapy for acute myocardial infarc-tion: A quantitative review of 23 randomizedtrials. Lancet 2003; 361:13–20

30. Hochman JS, Sleeper LA, Webb JG, et al:Early revascularization in acute myocardialinfarction complicated by cardiogenic shock.N Engl J Med 1999; 341:625–634

31. Hochman JS, Sleeper LA, White HD, et al:One-year survival following early revascular-ization for cardiogenic shock. JAMA 2001;285:190–192

32. Hochman JS, Sleeper LA, Webb JG, et al:Early revascularization and long-term sur-vival in cardiogenic shock complicatingacute myocardial infarction. JAMA 2006; 295:2511–2515

33. Ohman EM, Chang PP: Improving quality of

life after cardiogenic shock: Do more revas-cularization. J Am Coll Cardiol 2005; 46:274–276

34. Jeger RV, Tseng CH, Hochman JS, et al:Inter-hospital transfer for early revascular-ization in patients with ST-elevation myocar-dial infarction complicated by cardiogenicshock—A report from SHOCK trial and reg-istry. Am Heart J 2006; 152:686–692

35. Eagle KA, Guyton RA, Davidoff R, et al: ACC/AHA 2004 guideline update for coronary ar-tery bypass graft surgery: A report of theAmerican College of Cardiology/AmericanHeart Association Task Force on PracticeGuidelines (Committee to Update the 1999Guidelines for Coronary Artery bypass GraftSurgery). Circulation 2004; 100:e340

36. Dzavik V, Sleeper LA, Cocke TP, et al: Earlyrevascularization is associated with improvedsurvival in elderly patients with acute myo-cardial infarction complicated by cardiogenicshock: A report from the SHOCK trial regis-try. Eur Heart J 2003; 24:828–837

37. Sanborn TA, Sleeper LA, Bates ER, et al:Impact of thrombolysis, intra-aortic balloonpump counterpulsation, and their combina-tion in cardiogenic shock complicating acutemyocardial infarction: A report from theSHOCK Trial Registry. J Am Coll Cardiol2000; 36:1123–1129

38. Hasdai D, Califf RM, Thompson TD, et al:Predictors of cardiogenic shock after throm-bolytic therapy for acute myocardial infarc-tion. J Am Coll Cardiol 2000; 35:136–143

39. Bjorklund E, Stenestrand U, Lindback J, etal: Pre-hospital thrombolysis delivered byparamedics is associated with reduced timedelay and mortality in ambulance-trans-ported real-life patients with ST-elevationmyocardial infarction. Eur Heart J 2006; 27:1146–1152

40. Mehta SR, Eikelboom JW, Natarajan MK, etal: Impact of right ventricular involvementon mortality and morbidity in patients withinferior myocardial infarction. J Am CollCardiol 2001; 37:37–43

41. Bowers TR, O’Neill WW, Grines C, et al: Ef-fect of reperfusion on biventricular function

and survival after right ventricular infarc-tion. N Engl J Med 1998; 338:933–940

42. Thompson CR, Buller CE, Sleeper LA, et al:Cardiogenic shock due to acute severe mitralregurgitation complicating acute myocardialinfarction: A report from the SHOCK TrialRegistry. J Am Coll Cardiol 2000; 36:1104–1109

43. Menon V, Webb JG, Hillis LD, et al: Outcomeand profile of ventricular septal rupture withcardiogenic shock after myocardial infarc-tion: A report from the SHOCK Trial Regis-try. J Am Coll Cardiol 2000; 36:1110–1116

44. Killen DA, Piehler JM, Borkon AM, et al:Early repair of postinfarction ventricular sep-tal rupture. Ann Thorac Surg 1997; 63:138–142

45. Slater J, Brown RJ, Antonelli TA, et al: Car-diogenic shock due to cardiac free-wall rup-ture or tamponade after acute myocardialinfarction: A report from the SHOCK TrialRegistry. J Am Coll Cardiol 2000; 36:1117–1122

46. Reardon MJ, Carr CL, Diamond A, et al: Isch-emic left ventricular free wall rupture: Pre-diction, diagnosis, and treatment. Ann Tho-rac Surg 1997; 64:1509–1313

47. Kar B, Adkins LE, Civitello AB, et al: Clinicalexperience with the TandemHeart percuta-neous ventricular assist device. Tex HeartInst J 2006; 33:111–115

48. Leshnower BG, Gleason TG, O’Hara ML, etal: Safety and efficacy of left ventricular assistdevice support in postmyocardial infarctioncardiogenic shock. Ann Thorac Surg 2006;81:1365–1371

49. Tayara W, Starling RC, Yamani MH, et al:Improved survival after acute myocardial in-farction complicated by cardiogenic shockwith circulatory support and transplantation:Comparing aggressive intervention with con-servative treatment. J Heart Lung Trans-plant 2006; 25:504–509

50. Barbash IM, Behar S, Battler A, et al: Man-agement and outcome of cardiogenic shockcomplicating acute myocardial infarction inhospitals with and without on-site catheter-ization facilities. Heart 2001; 86:145–149

S74 Crit Care Med 2008 Vol. 36, No. 1 (Suppl.)