You are on page 1of 23

DI SEASE FOCUS

The Management of Acute Pulmonary Arterial Hypertension


Gan Hui-li
Cardiac Surgery Department, Beijing Anzhen Hospital, Capital Medical University (BAZHCMU), Beijing Institute of Heart, Lung, and Blood Vessel
Disease, Beijing 100029, China
Keywords
Pulmonary arterial hypertension (PAH); Right
heart failure.
Correspondence
Dr. Gan Hui-li, Cardiac Surgery Department,
Beijing Anzhen Hospital, Capital Medical
University (BAZHCMU), Beijing Institute of
Heart, Lung, and Blood Vessel Disease,
Beijing 100029, China.
Tel.: 86-10-64456885;
Fax: 86-10-62244207;
E-mail: ganhuili@hotmail.com
doi: 10.1111/j.1755-5922.2009.00095.x
Acute pulmonary arterial hypertension (PAH), which may complicate the
course of many complex disorders, is always underdiagnosed and its treat-
ment frequently begins only after serious complications have developed. Acute
PAH is distinctive because they differ in their clinical presentation, diagnostic
ndings, and response to treatment from chronic PAH. The acute PAH may
take either the form of acute onset of chronic PAH or acute PAH or surgery-
related PAH. Signicant pathophysiologic differences existed between acute
and chronic PAH. Therapy of acute PAH should generally be aimed at acutely
relieving right ventricular (RV) pressure overload and preventing RV dysfunc-
tion. There are three classes of drugs targeting the correction of abnormalities
in endothelial dysfunction, which have been approved recently for the treat-
ment of PAH: (1) prostanoids; (2) endothelin receptor antagonists; and (3)
phosphodiesterase-5 inhibitors. The efcacy and safety of these compounds
have been conrmed in uncontrolled studies in patients with PAH. Intra-
venous epoprostenol is suggested to serve as the rst-line treatment for the
most severe patients. In the other situations, the rst-line therapy may include
bosentan, sildenal, or a prostacyclin analogue. Recent advances in the man-
agement of PAH have markedly improved prognosis.
Pulmonary arterial hypertension (PAH) is a progressive,
symptomatic, and ultimately fatal disorder for which sub-
stantial advances in treatment have been made during
the past decade [1]. Despite advances in the manage-
ment of PAH, the mortality rate remains excessive. Acute
PAH, which may lead to refractory systemic arterial hy-
potension, severe hypoxemia, right ventricular (RV) dys-
function and failure, and ultimately result in cardiogenic
and/or obstructive shock and death have always been ne-
glected until serious complications have developed. Ef-
fective management requires timely recognition and ac-
curate diagnosis of the disorder and appropriate selection
among therapeutic alternatives. Despite progress in treat-
ment, obstacles remain that impede the achievement of
optimal outcomes [2]. The pathophysiology, monitoring,
and management of the acute pulmonary hypertension
are reviewed in this article to highlight the importance
and due management of it. The article also reviews es-
tablished approaches to evaluation and treatment, with
emphasis on the appropriate application of calcium chan-
nel blockers (CCBs), prostacyclin analogues, endothe-
lin receptor antagonists (ETRAs), and phosphodiesterase
5 inhibitors.
Pathophysiology and Pathogenesis
of Acute PAH
PAH is characterized by elevated pulmonary arterial pres-
sure (PAP) and secondary RV failure. The denition of
PAH used in clinical trials has been a mean pulmonary ar-
terial pressure (mPAP) >25 mmHg at rest or >30 mmHg
with exercise, a pulmonary capillary wedge pressure
(PCWP) of <15 mmHg, and a pulmonary vascular resis-
tance (PVR) of >3 Wood units, which has been attributed
to the criteria for idiopathic PAH established by the Na-
tional Institutes of Health Registry on Primary Pulmonary
Hypertension (PPH) [3,4]. It is considered to be severe
if mPAP >50 mmHg; of moderate severity, if mPAP 30
50 mmHg or mild, if mPAP <30 mmHg. Right heart
failure is the most important consequence of PAH and
remains a frequent cause of death in patients with PAH.
Increased RV afterload leads to RV dilatation in most
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 153
The Management of Acute Pulmonary Hypertension G. Hui-li
acute cases of PAH when there is not enough time for
adaptive mechanisms (e.g., RV hypertrophy) to develop.
Thus, RV dilatation or RV hypertrophy are the main fea-
tures of the acute and chronic PAH, respectively.
The mechanisms of arterial PAH include vascu-
lar thromboembolism, endothelial dysfunction, hy-
poxic vasoconstriction, pulmonary vascular remodeling,
smooth muscle proliferation with or without neointimal
formation, and in situ thrombosis [4]. Endothelial dys-
function with an imbalance between vasodilation and
vasoconstriction, and between apoptosis and prolifera-
tion is thought to play a pivotal role in the develop-
ment of chronic progressive PAH. Hypoxemic pulmonary
vasoconstriction is an important determinant of arterial
PAH in patients with respiratory disorders [4]. In many
types of PAH, production of endogenous vasodilators (ni-
tric oxide [NO] and prostacyclin) is impaired and produc-
tion of vasoconstrictors [5] (endothelin-1) is increased
[4]. This forms the pathophysiologic basis of common
treatment strategies for arterial PAH, which attempt to
achieve balance in key molecular pathways by increasing
available NO and prostacyclin, or reducing the effects of
endothelin-1.
Acute arterial PAH is characterized by a sudden in-
crease in PAP. Mechanical obstruction with subsequent
vasoconstriction characterizes acute PAH in pulmonary
embolism (PE). In acute lung injury (ALI)/acute res-
piratory distress syndrome (ARDS), both hypoxic pul-
monary vasoconstriction and accumulation of intravas-
cular brin and cellular debris contribute to subsequent
vascular obliteration and PAH [6]. Endotoxin plays a sig-
nicant role in the development of PAH during sepsis.
Multiple animal studies have shown that endotoxin can
cause not only systemic hypotension, but also pulmonary
biphasic hypertension, along with a decrease in compli-
ance and an increase in resistance of respiratory system
[7]. Endotoxin-dependent hemodynamic and respiratory
effects are mediated by excessive release of inamma-
tory mediators and imbalances in production of NO,
prostanoids, and endothelin-1 (ET-1) [8]. PAH in endo-
toxemia is characterized by constriction of proximal pul-
monary arteries during the early phase followed by de-
creased compliance of the distal pulmonary vasculature
[9].
Neurohormonal activation is an important factor in
both acute and chronic RV failure. The consequence of
sympathetic hyperactivity is an increase in PVR with
impedance of ow, causing RV strain that impairs lling
and causes RV volume and pressure overload. The RV di-
lates (and eventually hypertrophy can develop in chronic
gradual pulmonary hypertension), encroaching on the
left ventricle and decreasing preload, cardiac output, and
coronary perfusion. Increased RV wall stress results in RV
ischemia [10]. Tricuspid regurgitation develops as a result
of RV dysfunction and portends a poor prognosis [11].
RV systolic dysfunction, severe tricuspid regurgitation,
arrhythmias, and left ventricular dysfunction caused by
ventricular interdependence may contribute to low car-
diac output and hypotension in patients with pulmonary
hypertension. Regardless of the underlying cause of PAH,
the nal common pathway for hemodynamic deteriora-
tion and death is cor pulmonale and RV failure. Among
other clinically important adverse effects of right heart
failure is the development of systemic venous hyperten-
sion leading to concomitant visceral organ congestion and
dysfunction such as renal function impairment. The nor-
mal RV can acutely adapt to high ow, but is not able
to tolerate any but very short acute high pressure load
[12]. The normal RV cannot acutely increase and sustain
mean PAP >40 mmHg for more than a brief period of
time [13].
The Causes of Arterial Pulmonary
Hypertension
Multifactorial impairment of the physiologic balance can
lead to vasoconstriction, vascular smooth muscle cell and
endothelial cell proliferation/brosis, inammation, re-
modeling, and in situ thrombosis. These are the likely
mechanisms that lead to narrowing of the vessel followed
by progressive increase in PVR and the clinical manifes-
tations of PAH. The etiology of acute PAH is different
from what of chronic (or persistent) PAH, but the patho-
genesis of acute PAH is the same as of chronic (or per-
sistent) PAH, which also includes endothelial injury in
the pulmonary arterioles, vasoconstriction caused by re-
duced production of endogenous NO by the pulmonary
endothelium, and thrombosis [14]. Subsequently, major
goal of the therapy is to avoid acute pulmonary vasocon-
striction, halt the progression of vascular remodeling, and
reverse the early vascular remodeling if possible.
Almost all diseases manifested as PAH can cause right
heart failure: either as an acute condition, if the PAH de-
velops acutely (as with sepsis/acute lung injury (ALI),
PE, cardiac surgery, drug induced, etc.), or as a chronic
condition, if arterial pulmonary hypertension is mainly
chronic (as in chronic obstructive pulmonary disease
(COPD), interstitial lung disease , sleep disorder breath-
ing, chronic hypoventilation, PAH, chronic thromboem-
bolic pulmonary hypertension(CTEPH), portopulmonary
hypertension, etc.). PAH associated with various noncar-
diac etiological diseases (connective-tissue diseases, por-
tal hypertension, HIV infection, consumption of anorectic
agents) account for approximately 50% of cases of PAH.
154 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
The acute PAH may take either the form of acute onset of
chronic PAH or acute PAH or surgery-related PAH.
Acute Onset of Chronic Arterial PAH
Patients with preexisting PAH are particularly vulnerable
to acute illnesses, which commonly result in rapid dete-
rioration and even death [15]. Besides idiopathic PAH,
interstitial lung diseases, COPD, chronic hypoventilation
and sleep disorder breathing, portopulmonary hyperten-
sion and chronic pulmonary thromboembolism are al-
ways accompany with chronic arterial PAH. On the ba-
sis of above-mentioned morbidities, multiple noncardiac
predisposing factors can precipitate acute PAH onset and
right heart failure, even leading to adverse outcome.
These predisposing factors belong to several categories.
In the pulmonary category, there are chronic hypoxia,
and restrictive lung disease (with or w/o COPD), and de-
creased DLCO, and decreased pulmonary compensatory
capacity. In the infection category, these are using of im-
munosuppressive, and disease modifying drugs for un-
derlying disease, and malnutrition and overall decreased
functional reserve. Patients with chronic PAH can rapidly
deteriorate and usually die from progressive RV failure
(49%), progressive respiratory failure (18%), or sudden
cardiac death (17%).
Acute PAH
Massive PE, sepsis, and ALI are the main causes of acute
arterial PAH in the adult patient population [16]. The on-
set of acute right heart failure as a complication of ALI
and its more severe form ARDS is more gradual than in
patients with massive PE. It usually occurs at least 48 h
after the beginning of respiratory support [15]. Evalua-
tion of RV function by transesophageal echocardiography
(TEE) in a group of 75 ARDS patients submitted to pro-
tective ventilation demonstrated 25% incidence of acute
RV dysfuncion, with detrimental hemodynamic conse-
quences associated with tachycardia. However, those
changes in heart function were reversible in patients who
recovered and did not increase mortality [15]. Although
the initial magnitude of pulmonary hypertension was not
an indicator of mortality, PAP further increased in non-
survivors, but not in survivors when followed for 7 days
[17]. Thus, development of PAH in ARDS patients seems
to be a sign of poor prognosis. In a cohort of 352 ARDS
patients, both mortality rate and incidence of right heart
failure were related to the level of plateau pressure during
mechanical ventilation. An interaction between plateau
pressure and right heart failure was observed whereas
odds ratio of dying for an increase in plateau pressure
(from 1826 to 2735 cm H
2
O) in patients without right
heart failure was 1.05 (P = 0.635), in contrast to odds
ratio of 3.32 (P < 0.034) in patients with right heart fail-
ure [18]. The implementation of low tidal volume ven-
tilation in patients with ARDS has signicantly lowered
not only mortality (down to 32%), but also incidence of
acute right heart failure in this patient population [15].
In addition to being the major risk factor for ARDS devel-
opment, sepsis itself can sometimes lead to severe acute
arterial pulmonary hypertension [19].
In massive acute pulmonary embolism (APE), the in-
crease of PVR depends on the thrombosis load and poten-
tially on the pulmonary bed contraction caused by neu-
rohormonal reaction. Plasma endothelin concentrations
assessed on admission are not elevated in patients with
APE and it does not play as important role in acute phase
of increase of pressure in pulmonary arteries as in chronic
pulmonary hypertension [20].
Development of ALI in the critically ill is associated
with an array of abnormal interactions between the heart
and lungs. Of these abnormalities, increased PVR is com-
mon and seems to indicate a worse prognosis than when
PVR is normal. Increased pulmonary artery pressure,
which follows ALI in humans, has been attributed to
many factors. Early in ALI, PAH is secondary to an imbal-
ance between the release of vasoactive mediators derived
from arachidonic acid, endothelium-derived relaxing fac-
tor, and other metabolites. As ALI progresses, the com-
bination of mechanical obstruction and severe regional
hypoxic pulmonary artery vasoconstriction probably be-
comes the main factor responsible for PAH. In addition
to these elements, in situ and peripherally derived throm-
boembolism can be seen in ALI, owing to diverse distur-
bances in the coagulation and brinolytic processes. The
result is increased workload of the right ventricle, which
is caused by increased afterload and may induce hemo-
dynamic disturbances that culminate in overt RV failure.
However, epidemiologic studies have demonstrated that
death following ALI is more often the result of respira-
tory failure or sepsis. The absence of effective therapy
for PAH in ALI might be explained by the pathophysi-
ological and clinical course of the disease. A reasonable
conclusion from the contributing elements cited above
is that PAH complicating sepsis and trauma is simply a
marker of the gravity of the systemic insult that leads
to the development of ALI and probably not a separate
process [21].
Acute pulmonary hypertension (PH) is a character-
istic feature of the ARDS. The magnitude of PH has
been shown to correlate with the severity of lung in-
jury in patients with ARDS independently of the severity
of associated hypoxemia and has an adverse prognostic
signicance. Early in the histopathological evolution of
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 155
The Management of Acute Pulmonary Hypertension G. Hui-li
ARDS, pulmonary vasoconstriction, thromboembolism,
and interstitial edema contribute to the development of
PH, although pulmonary vascular remodeling probably
occurs eventually [22]. Many patients with severe ALI
do not respond to NO inhalational therapy with allevi-
ation of PAH and hypoxemia, so this treatment remains
controversial [23].
During Gram-negative sepsis, endotoxin lipopolysac-
charide (LPS) may activate host-inammatory responses,
resulting in the systemic inammatory response syn-
drome and the adult respiratory distress syndrome. In
cell culture systems, LPS activation of cellular responses
may be potentiated by plasma proteins. In the isolated
perfused rabbit lung, LPS administration markedly in-
creases the pulmonary hypertensive response to subse-
quent administration of platelet-activating factor (PAF).
Components of plasmapossibly LPS binding protein,
and soluble CD14potentiate the priming effect of en-
dotoxin, resulting in an augmented pulmonary hyper-
tensive response to PAF. Thus, plasma proteins decrease
the threshold at which endotoxin primes the lung and
may have a critical role in the pathogenesis of endotoxin-
induced ALI [24]. ALI is a common complication of gram-
negative sepsis. Pulmonary hypertension and increased
lung vascular permeability are central features of lung in-
jury following experimental bacteremia. PAF is a promi-
nent proinammatory mediator during bacterial sepsis.
E. coli bacteremia rapidly induces pulmonary hyperten-
sion stimulated by PAF and mediated at least in part by
endothelin-1 and neutrophil activation and sequestration
in the lung. Microvascular injury with leak is also medi-
ated by PAF during E. coli bacteremia, but the time course
of resultant hypoxemia and hemoconcentration is slower
than that of pulmonary hypertension. The contribution
of hypoxic vasoconstriction in exacerbating pulmonary
hypertension in gram-negative sepsis is probably a late
phenomenon [25].
In sepsis-caused acute PAH, studies demonstrated that
the early acute rise in PAP is caused by thromboxane.
The late sustained pulmonary hypertension of endotox-
emia, on the other hand, appears to be mediated by
endothelin. BMS182874, an endothelin receptor antag-
onist, blocks the effects of exogenously administered en-
dothelins in chronically instrumented awake sheep. Pre-
treatment with BMS182874 signicantly attenuated the
early endotoxin-induced acute increase in PAP and com-
pletely blocked the late sustained pulmonary hyperten-
sion, while having no affect on the increases in throm-
boxane levels. BMS182874 shifts the dose response curve
for U46619, a prostaglandin H2 analogue, to the right.
BMS182874, in addition to functioning as an endothe-
liumreceptor antagonist, appears to counteract the action
of thromboxane at the receptor level [26].
Surgery or Intervention-Related PAH
Some surgical interventions, in particular vascular, car-
diac, and thoracic surgery may cause acute elevation of
PAP either during the surgery or shortly after the inter-
vention has been completed. This is particularly danger-
ous in patients with preexisting PAH, since even short
periods of RV pressure overload can lead to profound
decompensation with all negative hemodynamic conse-
quences. Preexisting PAH is one of the major risk fac-
tors for morbidity and mortality in cardiothoracic surgery
patients [27]. PAH is a major determinant of perioper-
ative morbidity and mortality in special situations such
as heart and lung transplantation, pneumonectomy, and
ventricular assist device placement [28]. It is believed
that an elevated PAP during and after surgery develops
secondary-to-acute left heart failure/dysfunction or can
be a consequence of pulmonary parenchymal and en-
dothelial injury with activation of the systemic and pul-
monary inammatory response to cardiopulmonary by-
pass (CPB) circulation and/or ischemia reperfusion [29].
Protamine-mediated acute PAH and RV failure in the pe-
rioperative period are common (1.78%) complications of
CPB circulation during open heart operations [30]. PAH
can also develop later as a result of ARDS [31] or other
complications (Sepsis, PE, etc.) not directly related to ei-
ther surgery or anesthesia. Intraoperative management
should include prevention of exacerbating factors such as
hypoxemia, hypercarbia, acidosis, hypothermia, hyperv-
olemia, and increased intrathoracic pressure; monitoring
and optimizing RV function; and treatment with selective
pulmonary vasodilators.
Patients with increased (PVR) may experience acute
pulmonary hypertension after heart transplantation. PAP
or a transpulmonary gradient (TPG) >12 mmHg, is an es-
tablished risk factor for mortality in heart transplantation.
An increased PVR or an increased TPG is a risk factor for
increased 3-day and 3-month mortality after heart trans-
plantation (HTx). The reversibility of increased PVR or
TPGunder pharmacologic testing is supposed to indicate a
decreased probability of RV failure/death after transplan-
tation. PAPs greater than 50 mmHg, PVR greater than
6 Woods units, and TPG greater than 15 mmHg that are
unresponsive to optimal vasodilators are contraindica-
tions to orthotopic heart transplantation. Therapies used
to reduce PVR in the cardiac catheterization laboratory
include high-ow oxygen; sublingual nitroglycerin; and
intravenous inotropic agents, vasodilators, and selective
pulmonary vasodilators. Systemic hypotension may be
an undesirable side effect of vasodilators. Inhaled agents
such as NO and prostacyclin are specic to the pulmonary
vasculature and reduce PVR without causing systemic hy-
potension. All pharmacological therapies used to optimize
156 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
pulmonary hemodynamics before transplantation can be
used during transplantation in patients who are at high
risk for acute RV failure and death after orthotopic heart
transplantation because of elevated pulmonary hemody-
namic values. Use of larger donor hearts for patients with
elevated PVR and referral for heartlung transplantation
are potential treatment options [32].
Acute PAH occurring after CPB can be a cause of
postoperative morbidity and mortality. Endogenous en-
dothelin is a mediator of acute pulmonary hyperten-
sion occurring after cardiopulmonary bypass. Bosentan,
a mixed endothelin antagonist completely prevented pul-
monary hypertension after CPB and may, therefore, have
therapeutic applications in the management of patients
following cardiac surgery [33]. Procedures using car-
diopulmonary bypass and aortic cross-clamping are as-
sociated with a variable degree of ischemia/reperfusion
of the lungs, leading to acute pulmonary hyperten-
sion (PHT). Pulmonary selectivity was demonstrated with
sildenal analog, because there were no similar changes
in systemic vascular resistance (SVR) and no signi-
cant changes in systemic hemodynamics. Sildenal ana-
log (UK343664), a PDE5 inhibitor, shows a promising
role for managing the PVR increases that occur following
CPB [34].
Absolute ethyl alcohol is used with increased frequency
in the treatment of conditions such as percutaneous ab-
lation of unresectable hepatic tumor, sclerotherapy of
esophageal varices, ventricular septal ablation, and intra-
venous embolization of arteriovenous malformation. The
postulated mechanism is severe pulmonary vasoconstric-
tion due to the sudden passage of absolute ethyl alco-
hol into pulmonary circulation, causing acute pulmonary
hypertention and RV strain. In patients at risk of sud-
den intravascular absorption of absolute ethyl alcohol,
an increased level of awareness for acute PAH, together
with prompt treatment of PAH with Sildenal analogue
or with a low dose of an NO-donor drug, will minimize
the resk of cardiovascular decompensation and may be
the treatment of such catastrophe [35].
Diagnosis and Monitoring of Patients
with Acute PAH
Effective management requires timely recognition and
accurate diagnosis of the disorder and appropriate selec-
tion among therapeutic alternatives. Since acute PAH are
a life-threatening disorder, elevated pressure and resis-
tance in the pulmonary vessels lead to progressive right
heart failure which results in functional limitations and
ultimately the death of most patients. Thus, the monitor-
ing of RV function is of great importance. The assessment
of PVR plays an important role in the diagnosis and man-
agement of PAH.
The broad availability of Doppler echocardiography has
helped with (earlier) detection of PAH. Doppler echocar-
diography has become a popular screening tool for the
diagnosis of PAH and in some clinical trials has been the
only measure of the severity of the PAH in response to
treatment [36]. Echocardiography is the initial investiga-
tion of choice for noninvasive detection of PAH but mea-
surement of PAP and cardiac output (CO) during right
heart catheterization is mandatory to conrm the diag-
nosis. So PAH is usually recognized when the patient de-
velops obvious signs of progressive RV failure, and during
hemodynamic monitoring by echocardiogram or a pul-
monary artery catheterization. In many cases, PAH re-
mains undiagnosed and its treatment begins only after
serious complications have developed. Timely assessment
of PAP, CO, and PVR with the help of pulmonary artery
catheterization and/or echocardiography is crucial for op-
timal management of patients with pulmonary hyperten-
sion.
One major disadvantage of the use of echocardio-
graphy as the diagnostic tool for evaluation of pul-
monary hemodynamics is the lack of adequate assess-
ment of transpulmonary blood ow and pulmonary
venous pressures. Therefore, right-sided heart catheter-
ization remains the current gold standard for diagno-
sis and vasoreactivity testing [37]. As the gold standard
for the diagnosis of pulmonary hypertension [38], pul-
monary artery (PA) catheterization could provide direct
measurement of the PAP. Besides direct measurement
of the pulmonary hemodynamic parameters, it also pro-
vides useful information regarding response to vasodila-
tor therapy. Analysis of mixed venous oxygen saturations
during passage of the PA catheter on its way through
the cardiac chambers can allow diagnosis of intracardiac
shunts. A PCWP measurement reects left ventricular
end-diastolic (lling) pressure. PCWP <15 mmHg rule
out left ventricular, valvular, and pulmonary venous dis-
eases as possible causes of the pulmonary hypertension
[25]. In critical care settings, PA catheterization and mon-
itoring is highly desirable in patients with severe PAH and
in patients with progressive heart failure [39]. It is impor-
tant to emphasize that the thresholds required for diag-
nosis of severe chronic PAH are incompatible with life in
patients with acute PAH and usually reect worsening of
preexisting chronic PAH.
Echocardiography is an important tool for diagnosing
and determining the degree and clinical signicance of
PAH in critically ill patients [4,14].
It can noninvasively visualize cardiac anatomy and
certain intracardiac shunts and valvular abnormalities,
estimate right atrial and PAP, determine the severity of
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 157
The Management of Acute Pulmonary Hypertension G. Hui-li
right and left ventricular dysfunction, wall motion abnor-
malities, and reveal other potential causes of PAH. In the
absence of pulmonary outow obstruction, pulmonary
artery systolic pressure is equivalent to RV systolic pres-
sure (RVSP), which can be calculated from measured
systolic regurgitant tricuspid ow velocity and estimated
right atrial pressure. Echocardiographic signs of signi-
cant PAH include RV dilation (D-shaped right ventricle)
and its hypertrophy (in sustained cases), septal dyskine-
sia, and bowing into the left ventricle during late sys-
tole to early diastole, RV hypokinesis, tricuspid regur-
gitation, right atrial enlargement, and a dilated inferior
vena cava [18,28,40]. Increased RV size combined with
increased outow resistance and reduced ejection frac-
tion have been also described in acute right heart fail-
ure [18]. A specic pattern of RV dysfunction in APE
has been characterized by a severe hypokinesia of the RV
mid-free wall, with normal contractions of the apical seg-
ment [41]. TTE results were poorly predictive of the risk
of death in patients who developed acute right heart fail-
ure [9]. However, denition of the acute right heart fail-
ure as RV end-diastolic area/left ventricular end-diastolic
area (RVEDA/LVEDA) ratio in the long axis greater than
0.6 associated with septal dyskinesia in the short axis
could be used to stratify the severity of the condition
[16,42]. TEE is more accurate and sensitive in critically
ill patients than TTE, especially in acute diseases such as
PE when acute pulmonary hypertension is highly sus-
pected [43,44]. Determination of pulmonary artery sys-
tolic pressure by Doppler echocardiography (based on the
pressure gradient between the right ventricle and right
atrium) or by right heart catheterization is useful in eval-
uating the severity and prognosis of cardiac disease [45].
PVR is an important hemodynamic variable in the man-
agement of patients with acute PAH. Doppler echocar-
diography could provide Doppler variables of time-
velocity integral of RV outow pulmonary artery ow
and peak tricuspid regurgitant pressure gradient (TRPG).
The TRPG/TVI ratio provides a reliable estimation of PVR
over a wide range in patients with PAH with various un-
derlying causes. [46] The noninvasive index of systolic
pulmonary arterial pressure (SPAP) to heart rate (HR)
times the RV outow tract time-velocity integral (TVI
[RVOT]) (SPAP/[HR TVI [RVOT]) provides clinically
useful estimations of PVR in PAH. The index of SPAP/
(HR TVI [RVOT]) provides useful estimations of PVRI
in patients with PAH [47].
Recent advances in magnetic resonance imaging (MRI)
technology have led to the development of techniques for
noninvasive assessment of cardiovascular structure and
function, including hemodynamic parameters in the pul-
monary circulation, which are superior in their identi-
cation of RV morphologic changes. These advantages
make cardiac MRI an attractive modality for following up
and providing prognoses in patients with PAH. Over the
coming decade, it can be anticipated that continued im-
provements in MRI image acquisition, spatial and tem-
poral resolution, and analytical techniques will result in
improved understanding of PAH pathophysiology, diag-
nosis, and prognostic variables, and will supplement, and
may even replace, some of the invasive procedures cur-
rently applied routinely to the evaluation of PAH [48].
Cardiac MRI has several advantages over other imaging
methods. The use of cine acquisition techniques allows
precise description of characteristic volumetric and func-
tional variables, such as RV volumes, muscle mass, stroke
volume, ejection fraction, or cardiac output. Impaired RV
contractility and function have also been assessed using
measures like ventricular septal bowing and pressure
volume loops. MRI investigations have been performed
to monitor medical treatment, and the improvement in
well-established prognostic factors, such as the 6-min
walk, were correlated with measures of RV function.
Flow-derived parameters of the pulmonary arteries (such
as peak velocity, acceleration time and volume, or pul-
monary owprole) are available using velocity-encoded
imaging, and may detect early signs of remodeling. Addi-
tionally, magnetic resonance angiography is a promising
new tool to visualize pulmonary perfusion and to diag-
nose CTEPH [49].
Biomarker May Serve as Tools for the Diagnosis
of Acute PAH
The most useful biochemical biomarker available to
date may be brain natriuretic peptide (BNP) (and/or
N-terminal prohormone brain natriuretic peptide), which
can inform on the pressure load on the right ventricle and
has prognostic value [50,51]. A reduction in circulating
BNP levels should accompany the response to an effective
treatment. There is increasing recognition of the impor-
tance of maintaining RV function in pulmonary hyper-
tension, but the primary goal is to retard and regress the
pulmonary vascularpathology. As such, BNP levels give
limited information about the response of patients with
pulmonary hypertension to treatment. Elevated BNP is
an important prognostic indicator and correlates strongly
with PVR, cardiac output, and functional status in pa-
tients with PAH [52]. A high level of plasma BNP, and
in particular, a further increase in plasma BNP during
follow-up, may have a strong, independent association
with increased mortality rates in patients with PAH [53].
However, the signicance of measuring BNP level in pa-
tients with PAH in the acute setting remains to be de-
ned. N-terminal pro-BNP (NT-proBNP) is a byproduct
of the BNP that was shown to be of prognostic value in
158 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
pulmonary hypertension (PH). The role of NT-proBNP in
PH has to be determined, especially under the inuence
of renal impairment that might lead to an accumulation
of the peptide, and may be a sign of increased mortality
per se. However, NT-proBNP could be superior to BNP as
a survival parameter in PH because it integrates hemody-
namic impairment and renal insufciency, which serves
as a sign of increased mortality per se [54].
Serum troponin may be elevated in patients with pul-
monary hypertension and has been associated with RV
overdistension and/or ischemia. Troponin I leak due to
acute RV strain from PE has been well studied, and may
predict mortality [55,56].
Management of Acute PAH
Recently published data addressing certain molecular
mechanisms for pathogenesis of PAH have led to the suc-
cessful therapeutic interventions. This review will focus
on the common and critical molecular pathways includ-
ing genetic basis of the development of PAH that on the
whole may be new targets for therapeutic interventions.
For the acute PAH, conventional therapy includes right
uid management, ventilation management and nonspe-
cic drugs (oxygen, warfarin, diuretics). The following
three classes of drugs targeting the correction of abnor-
malities in endothelial dysfunction have been approved
recently for the treatment of PAH: (1) prostanoids; (2) en-
dothelin receptor antagonists; and (3) phosphodiesterase-
5 inhibitors. The efcacy and safety of these compounds
have been conrmed in uncontrolled studies in patients
with PAH. Intravenous epoprostenol is the rst-line treat-
ment for the most severe patients. In the other situa-
tions, the rst-line therapy may include bosentan, silde-
nal, or a prostacyclin analogue. Recent advances in the
management of PAH have markedly improved progno-
sis. The endothelin-1 receptor antagonist bosentan, the
phosphodiesterase-5 inhibitor sildenal, and prostanoids
have been shown to improve symptoms, exercise capac-
ity, and hemodynamics. Intravenous prostacyclin (PGI
2
)
is the rst-line treatment for the most severely affected
patients [57]. The evolution of therapy from vasodilators
to antiproliferative agents reects the advancement in the
understanding of the mechanisms mediating PAH [58].
RV failure may result from a newly developed disease
(e.g., as a consequence of ARDS or of severe pulmonary
embolism) or of already present pulmonary hyperten-
sion (PHT). There is as yet no generally recognized def-
inition of acute or chronic RV failure. The particular clin-
ical picture and the associated hemodynamics determine
this condition. RV failure in the course of PHT represents
a great challenge in clinical and intensive care practice.
Once the vicious circle of right heart failure is reached an
optimal balance has to be found between preload and af-
terload. In addition to optimizing blood volume, positive
inotropic drugs (e.g., dobutamine) are available to main-
tain systemic blood pressure. Furthermore, an increase
in RV contractility by inodilators is aimed at. The cen-
tral goal in the treatment of right heart failure as part
of PHT is to lower PVR and thus decrease RV afterload.
However, it is very difcult to break the vicious circle in-
volved in the acute right heart syndrome; it must be the
primary aim of treatment to recognize as early as possi-
ble any worsening of PHT and prevent acute right heart
failure. Lung transplantation or surgical atrioseptostomy
may represent possible ultimate therapeutic options for
patients with PHT [59].
General Management Principles
Right uid management is the basis for recovery of acute
PAH. In patients with PAH, RV dysfunction/failure can
reduce LV lling and lead to cardiogenic shock. Patients
with cardiogenic shock secondary to RV dysfunction usu-
ally have a very high (>20 mmHg) RV lling pressure
[60]. In addition to decreased RV contractility and car-
diac output, RV dilatation can further limit LV lling via
ventricular interdependence, which causes shifting of the
interventricular septum toward the LV cavity. The chal-
lenge in uid management in those patients is to nd
the optimal RV preload necessary to avoid the detrimen-
tal effects of ventricular interdependence on LV function.
Hemodynamic monitoring of patients with RV failure due
to acute RV myocardial infarction showed that cardiac
and stroke indexes increased and the RV reached its max-
imum stroke work index when the lling pressure was
1014 mmHg. These values may be regarded as the opti-
mal level of RV lling pressure in patients with RV infarc-
tion [61]. There are no data on optimal RV lling pressure
in patients with RV dysfunction secondary to acute PAH.
Right ventilation management is another important
basis for recovery of acute PAH. It was found that con-
trolled ventilation altered RV function primarily by in-
creasing RV afterload during the lung ination period
[62]. Transpulmonary pressure (and related tidal vol-
ume), but not airway pressure itself, was the main de-
terminant factor of RV afterload during mechanical ven-
tilation [63]. This supports a low-volume strategy in
ARDS, recommended as a protective measure for lung
parenchyma, which might also represent a protective
measure for the RV and pulmonary circulation [55]. Fre-
quency of acute right heart failure in ARDS patients de-
clined from 61% to 25% over the last 1530 years, which
could be explained in part by fundamental alterations in
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 159
The Management of Acute Pulmonary Hypertension G. Hui-li
respiratory support and implementation low tidal volume
ventilation [15]. Lower incidence of acute right heart fail-
ure in ARDS patients was associated with lower (less than
2730 mmHg) plateau pressure [20].
RV systolic function was generally negatively affected
by end-expiratory pressure (PEEP) in ARDS patients un-
dergoing mechanical ventilation. In those patients PEEP
titration signicantly affected RV outow impedance, the
lowest values of which was associated with the achieved
better total quasi-static lung compliance (calculated by
dividing tidal volume by the difference between plateau
and end-expiratory airway pressures) [64]. This suggests
that lung hyperination along with either inadequate or
excessive PEEP can signicantly reduce RV systolic func-
tion and cardiac output. On the other hand, in an ex-
perimental study on healthy animals, the open lung con-
cept ventilation resulted in signicantly improved lung
aeration with no negative effect on RV afterload or LV
afterload. This is possibly explained by a loss of hypoxic
pulmonary vasoconstriction due to alveolar recruitment.
The reductions in the CO and in the mean PAP were the
consequences of a reduced preload [65]. A clinical study
in patients after cardiac surgery found no evidence that
ventilation according to the open lung concept affects RV
afterload [66].
Hypercapnia has been shown to induce pulmonary hy-
pertension in animal models. Study on healthy volun-
teers revealed that human pulmonary vascular responses
to hypercapnia and hypocapnia consist, respectively, of
constriction and dilatation that take 1.52 h to reach a
steady level. The time courses for recovery in eucapnia
are similar. Hypercapnia generated a rise in cardiac out-
put by changing heart rate; hypocapnia produced a fall in
cardiac output by changing stroke volume. The nding of
marked vasodilatation in response to hypocapnia demon-
strates that there is normally a substantial vascular tone
in the human pulmonary circulation [67].
Treatment with 100% oxygen is a selective pulmonary
vasodilator in patients with sustained pulmonary hyper-
tension, regardless of primary diagnosis, baseline oxy-
genation, or RV function [68]. In patients with ALI vas-
cular response to oxygen was different and administra-
tion of 100% O2 worsened intrapulmonary shunt possi-
bly secondary to collapse of unstable alveolar units with
very low ventilation-perfusion (V A/Q) ratios. This is in
contrast to administration of 100% O2 to patients with
COPD, in whom only the dispersion of the blood ow
distribution was changed, suggesting release of hypoxic
pulmonary vasoconstriction [69]. Optimal supplemental
oxygen management is an integral component of pul-
monary hypertension therapy [70].
Patients with chronic pulmonary hypertension usually
have multiple predisposing factors, which make them
more vulnerable to acute conditions and explain de-
creased ability to survive acute illness [14]. Tighter moni-
toring combined with early diagnosis and appropriate in-
tervention increases the chance of survival. Treatment
strategies for such patients should be generally directed
at rapid reversal of the precipitating condition as well
as at relieving RV pressure overload and maintaining
systemic pressure for coronary and end-organ perfu-
sion. Unlike patients with acute pulmonary hyperten-
sion [13], patients who develop acute pulmonary hy-
pertension on top of preexisting pulmonary hyperten-
sion can generate higher PAP and also require higher
RV preload (because of RV hypertrophy). Therapy for
acute massive PE with associated hemodynamic instabil-
ity or cardiogenic/obstructive shock aims at urgently re-
lieving mechanical obstruction of the pulmonary circu-
lation by either pharmacologic thrombolytic or by surgi-
cal or catheter thrombectomy (or mechanical clot disrup-
tion) [46,71,72]. Patients with massive PE but without
hemodynamic instability are less likely to benet from
thrombolytic therapy. The time the resolution of RV di-
latation, which usually takes between 10 and 20 days,
is hastened by thrombolytic therapy [16]. The therapy of
acute pulmonary hypertension in patients with sepsis and
ARDS is generally aimed at optimizing gas exchange and
minimizing ventilator-induced lung injury. Inhaled pul-
monary vasodilators are sometimes used in order to pre-
vent or relieve acute right heart failure.
Pulmonary Vasodilators
A number of vascular mediators have been implicated
in the pathophysiology of PAH, including prostacyclin,
thromboxane A2, endothelin-1, and NO [73]. Various
treatments approved by the United States Food and Drug
Administration for the management of PAH target three
of the many pathways implicated in the development of
PAH: prostacyclin-, endothelin-1 (ET-1), and nitric oxide-
mediated pathways.
Over the last decade, treatment of PPH has gained
major progress through medication such as analogue
of prostacyclin (Epoprostenol [74]) and dual antagonist
of endothelin receptor (Bosentan [75]), or inhibitors of
phosphodiesterase (Milrinone and Sildenaphil). All of
these medicines have proven highly clinically effective.
Pulmonary emb basis H of non-cardiac etiologyervention such
as Inhaled Nitric oxide (iNO) is a potent vasodilator that
dilates pulmonary vasculature in ventilated lung areas,
thereby improving ventilation/perfusion (V/Q)match and
oxygenation, and reducing PAP [76] and RV oxygen
demand [77]. This improves cardiac performance with-
out altering RV contractility [78] and cardiac output in
160 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
hemodynamically stable patients with a variety of causes
of pulmonary hypertension [79,80]. Multiple studies on
utilization of the iNO in ARDS patients showed improved
oxygenation for 2448 h, variable improvement in MPAP
[81] and no signicant impact on the duration of ven-
tilatory support or mortality in this patient population
[82,83]. A combination of iNO and other interventions,
such as positive end-expiratory pressure and prone posi-
tioning, yielded benecial and additive effects on arterial
oxygenation [63]. iNO has been successfully used and as-
sociated with improved outcomes in critically ill postop-
erative patients, who developed severe PAH or RV failure
[84].
Because iNO is a selective pulmonary vasodilator with-
out signicant effects on systemic blood pressure, reduc-
tions in PAP is the result of the pulmonary vasodila-
tory response. A combination treatment of iNO with
other pulmonary vasodilators (milrinone, sildenal, etc.)
has been used in management of acute pulmonary hy-
pertension in different conditions. [85,86] iNO treat-
ment is associated with multiple side effects including
methemoglobinemia [56,87] and NO
2
formation [68].
Abrupt withdrawal of nitric oxide has been associated
with rebound pulmonary hypertension, signicant drop
in PaO
2,
and life-threatening hemodynamic deterioration
[68,88]. Close monitoring and gradual discontinuation
are important to prevent and detect rebound pulmonary
hypertension.
Prostaglandin E
1
and prostacyclin both produce signif-
icant pulmonary vasodilatation and can lower PVR. Both
possess antithrombotic, antiproliferative, and antiinam-
matory properties. In patients with chronic severe PAH,
intravenous prostacyclin is highly effective and associ-
ated with a signicant survival benet [89,90]. Prosta-
cyclin, subcutaneous or intravenous treprostinil, intra-
venous PGE1, and other prostanoids have relatively long
half-lives and can signicantly affect systemic hemody-
namics. This feature limits their use in critically ill or
hemodynamically unstable patients. PGI
2
in combination
with norepinephrine and dopamine was effective in the
treatment of protamine-mediated acute PAH and RV fail-
ure in the setting of open heart surgery [25]. Prosta-
cyclin and its analogs (iloprost and treprostinil) in in-
haled forms are as effective as intravenous forms in pa-
tients with chronic PAH [91]. Inhaled prosaglandins have
been used to treat severe sustained pulmonary hyperten-
sion and intractable hypoxia, but were associated with
systemic hypotension [92]. Inhaled iloprost effectively
decreased PAP and improved RV performance immedi-
ately after separation from CPB [93]. Inhaled iloprost
was effective in the treatment of acute RV dysfunction
in the setting of preexisting PAH in heart transplant re-
cipients during weaning from CPB circulation and was
not associated with signicant systemic side-effects. De-
spite the observation that RV ejection fraction increased
on inhaled NO, but not with PGI
2
, both inhaled NO
and PGI2 aerosol showed benecial effects on RV perfor-
mance and may prove helpful in the treatment of acute
PAH [47]. Inhaled prostacyclin showed different effect on
oxygenation and pulmonary hemodynamics in patients
presenting with primary pulmonary ARDS (reduction
in PaO2/FIO2) compared with extrapulmonary cause of
ARDS (increase in PaO2/FIO2 along with a decrease in
mean pulmonary artery pressure) [94]. In a compara-
tive trial in patients with PAH, both inhaled and infused
prostacyclin had profound effects on pulmonary hemody-
namics, almost equipotently reducing PVR, whereas only
inhaled prostacyclin signicantly reduced PAP [95]. In-
halation of iloprost in patients with severe PH associated
with lung brosis resulted in the same degree of reduc-
tion of PVR as intravenous prostacyclin, only inhaled ilo-
prost preserved gas exchange [96].
PGI
2
is a recommended rst-line therapy for unsta-
ble patients in NYHA functional class IV [97,98,99]. How
prostacyclin improves cardiac output in right-heart fail-
ure in conjunction with pulmonary hypertension has
been evaluated in a dog model of acute afterload-induced
RV failure [67]. In this model, prostacyclin improved
right ventriculoarterial coupling and increased cardiac
output by decreasing pulmonary arterial resistance, be-
cause of vasodilating effects, without a detectable effect
on contractility. It is likely that clinical RV failure in hu-
man PAH might be due to aggravated right ventriculoar-
terial decoupling, and eventually a decrease in right ven-
tricle contractility. These observations provide a rationale
for inotropic interventions added to prostacyclin therapy
in patients with PAH who present with RV decompensa-
tion [100].
Continuous intravenous epoprostenol improves exer-
cise capacity, hemodynamics, and survival in IPAH and
is the preferred treatment option for the most critically
ill patients. Treprostinil, a prostanoid, may be delivered
via either continuous intravenous or subcutaneous in-
fusion. Iloprost is a prostanoid delivered by an adaptive
aerosolized device 6 times daily.
Phosphodiesterase inhibitor Sildenal is a selective in-
hibitor of phosphodiesterase type 5 with sustained pul-
monary vasodilatatory effect. Sildenal lowers PVR and
PAP and increases CO in patients with different forms
of chronic PAH [101,102] including patients with PAH
secondary to congestive heart failure [103]. This effect
is mainly achieved by balancing pulmonary and sys-
temic vasodilation. In patients, after coronary artery by-
pass graft surgery, sildenal may improve myocardial
perfusion, reduce platelet activation, and potentially re-
duce early graft occlusion [104]. In the chronic setting, it
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 161
The Management of Acute Pulmonary Hypertension G. Hui-li
is highly effective both alone and in combination with
other pulmonary vasodilators: epoprostenol [105], ilo-
prost [106,107], BNP [108] and iNO [89]. Although
mostly used in patients with chronic PAH and in hemo-
dynamically stable patients sildenal possesses multiple
features that could be effective in treatment of critically ill
patients with severe RV dysfunction related to secondary
PAH [109]. Its ability to augment and prolong the hemo-
dynamic effects of other pulmonary vasodilators has been
successfully used to minimize rebound pulmonary hyper-
tension after iNO discontinuation [110,111,112], wean-
ing from intravenous vasodilators in patients after car-
diac surgery [105] as well as in chronic PAH therapy
[113]. Experimental evaluation of the inhaled and in-
travenous sildenal administration showed similar ef-
fects on pulmonary endothelium-dependent relaxation,
overall hemodynamic prole, and oxygenation after car-
diopulmonary bypass. Both administration routes pre-
vented a signicant increase in mPAP without severe sys-
temic hypotension [114]. Its chronic administration has
been shown to improve exercise capacity, World Health
Organization functional class, and hemodynamics in pa-
tients with symptomatic PAH. Sudden cessation of silde-
nal monotherapy, in patients with PAH, carries with it
a signicant and unpredictable risk of rapid clinical de-
terioration. It is recommended that if sildenal needs to
be ceased, it would be more prudent to consider concur-
rent vasodilator therapy before the gradual cessation of
sildenal [115]. Pilot study suggested benecial effects in
favor of sildenal in several secondary endpoints at both
3 months and 12 months [116].
CCBs were introduced for the treatment of hyperten-
sion in the 1980s [117]. CCBs have been used in hemo-
dynamically stable patients with PAH who demonstrated
pulmonary vascular response to acute vasodilator chal-
lenge [118]. There are no studies of CCBs in critically ill
patients with acute PAH. Acute administration of nifedip-
ine did not cause pulmonary vasodilation, and in contrast
led to increased RVEDP and decreased RV contractility
[46]. Prolonged half-life and negative inotropic effects,
which may precipitate fatal worsening of RV failure, limit
the use of CCBs in treatment of acute pulmonary hyper-
tension. Treatment with high doses of CCBs has been
shown to have a sustained benecial effect in a very
small subset of patients with severe idiopathic PAH who
demonstrated an acute fall in PAP in response to a pul-
monary vasodilator [119]. The empirical use of CCBs is
discouraged because of the risks of systemic hypotension
and impaired right-sided heart function [120]. Conse-
quently, the current recommendations for the treatment
of PAH propose that the acute response of the pulmonary
circulation to a pulmonary vasodilator should be used as
the basis for selecting patients for high-dose CCB treat-
ment [121]. A retrospective analysis of 557 patients with
IPAH showed that only 13% of patients displayed vasore-
activity, as dened by a decrease of more than 20% in
both mPAP and PVR. Of the patients displaying vasore-
activity, only about half beneted from treatment with
long-term CCBs. Patients who beneted from these med-
ications tended to have had a more profound short-acting
vasodilator response during invasive testing [122].
ETRAs basic research in vascular biology has impli-
cated endothelin-1 (ET-1) and its receptors (ETA and
ETB) in diverse preclinical models of PAH, and ET-1
has been shown to contribute signicantly to PAH in
human patients [123]. The introduction of ETRAs to
clinical medicine has substantially expanded therapeu-
tic approach toward severe PAH [124]. Bench-to-bedside
scientic research has shown that endothelin-1 (ET-1)
is overexpressed in several forms of pulmonary vascular
disease and may play an important pathogenetic role in
the development and progression of PAH. Endothelin re-
ceptor antagonism has emerged as an important thera-
peutic approach in PAH.
Oral ETRAs improved exercise capacity, functional sta-
tus, pulmonary hemodynamics, and delayed the time
to clinical worsening in several randomized placebo-
controlled trials. Two ETRAs are currently approved by
the US Food and Drug Administration: bosentan, a dual
ETRAs for patients with class III and IV PAH, and am-
brisentan, selective ETRAs for patients with class II and III
PAH. Sitaxsentan, another selective ETRAs, has been ap-
proved in Europe, Canada, and Australia [125]. ET recep-
tors (ET[A] and ET[B]) have different densities and dis-
tributions throughout the body and are dynamically reg-
ulated, such that blockade of ET(A) and ET(B) receptors
may have different results in normal versus pathological
conditions. Although differences in biological effects can
be found in studies of isolated cells, blood vessels and ani-
mal models, clinical treatment studies have not identied
clear differences in efcacy among the various ETRAs.
The main differences appear to be in safety proles, with
a greater frequency of serum liver function abnormali-
ties occurring with the available dual ET(A)/ET(B) antag-
onist, and possibly higher rates of peripheral edema noted
with selective ET(A) agents [126].
Bosentan, a dual endothelin receptor antagonist, is in-
dicated for the treatment of patients with PAH. Following
oral administration, Bosentan attains peak plasma con-
centrations after approximately 3 h. The terminal half-
life after oral administration is 5.4 h and is unchanged
at steady state. Steady-state concentrations are achieved
within 35 days after multiple-dose administration, when
plasma concentrations are decreased by about 50% be-
cause of a 2-fold increase in clearance, probably due to in-
duction of metabolizing enzymes. The pharmacokinetics
162 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
of bosentan is dose proportional up to 600 mg (single
dose) and 500 mg/day (multiple doses). The pharmacoki-
netics of bosentan in pediatric PAH patients are compa-
rable to those in healthy subjects, whereas adult PAH
patients show a 2-fold increased exposure. Severe re-
nal impairment (creatinine clearance 1530 mL/min) and
mild hepatic impairment (ChildPugh class A) do not
have a clinically relevant inuence on the pharmacoki-
netics of bosentan. Bosentan should generally be avoided
in patients with moderate or severe hepatic impairment
and/or elevated liver aminotransferases. In healthy sub-
jects, bosentan doses >300 mg increase plasma levels
of endothelin-1. In a pharmacokinetic-pharmacodynamic
study in PAH patients, the hemodynamic effects lagged
the plasma concentrations of bosentan [127]. An open-
label study suggests a benecial effect of bosentan ther-
apy not only on pulmonary hemodynamics, but also on
quality of life and exercise capacity for patients with se-
vere CTEPH [128].
Sitaxsentan is the rst oral ETRA with high selectivity
for the endothelin-A (ET [A]) receptor to be approved for
clinical use by regulatory agencies in Europe for the treat-
ment of PAH. Sitaxsentan therapy appears safe and efca-
cious for patients with PAH; reductions in mortality and
the risk for clinical worsening events provide support for
durability of efcacy at 1 year [129]. Clinical trials have
shown it to be well tolerated and to improve exercise tol-
erance, functional class, and pulmonary hemodynamics
in PAH, results which appear to be at least as good as
those for the mixed ETRA bosentan. Importantly, com-
pared to bosentan, Sitaxsentan has a lower incidence of
liver toxicity and no interaction with sildenal [130].
Ambrisentan is the second selective endothelin-A re-
ceptor antagonist to be licensed in Europe, and the rst
in the United States, for the management of PAH. It has
been shown to be clinically effective in improving ex-
ercise tolerance and functional class. Furthermore, am-
brisentan is well tolerated and associated with low rates
of liver toxicity and minimal interactions with other
medicines commonly used to treat PAH. Overall, current
data support a role for ambrisentan in the management
of PAH. However, the results of longer-term follow-up
studies are still required to fully assess efcacy and safety
[131]. Ambrisentan has been shown to be an effective
ETRAs in patients with PAH, at the same time, a signi-
cant advantage of ambrisentan is the lack of any clinically
important drug interactions with warfarin and sildenal,
which are frequently used by patients being treated for
PAH [132].
Platelet-derived growth factor (PDGF) has the abil-
ity to induce the proliferation and migration of smooth
muscle cells and broblasts, and PDGF and its recep-
tors are overexpressed in human and experimental PAH
[133]. Novel therapeutic agents, such as imatinib me-
sylate, inhibit several tyrosine kinases, including PDGF
receptors and . Imatinib has been demonstrated to
reverse pulmonary vascular remodeling in animal mod-
els of pulmonary hypertension. Four cases of clinical and
hemodynamic improvements have also been reported in
human PAH[134,135,136]. Concerns have arisen about
the cardiac safety of tyrosine kinase inhibitors, especially
in patients with preexisting cardiac conditions [137,138].
Safety and efcacy of tyrosine kinase inhibitors are cur-
rently being evaluated in multicenter randomized trials.
Imatinib, as a selective antagonist of the platelet-derived
growth factor receptor, was effective in the case of a sin-
gle patient with severe treatment-refractory familial PAH
[139], suggesting that such antiproliferative drugs may
reverse pulmonary vascular remodeling, thus alleviating
PAH.
A signicant venodilator effect of nitroglycerine de-
creases RV preload leading to adverse consequences in
patients with right heart failure. Inhaled nitroglycerin
may be a safer therapeutic option leading to a signicant
reduction in both mean PAP and PVR in patients after
mitral valve operations without reducing systemic mean
arterial pressure [140]. Intravenous adenosine is a pul-
monary vasodilator with a very short half-life (610 sec-
onds) and can be effective for short term lowering PVR
[76]. In the setting of acute PAH, adenosine infusion may
help lower PAP without lowering systemic arterial pres-
sure and reverse the clinical state of shock by achieving
pulmonary vasodilatation [141]. However, higher doses
(70 mg/kg per min to 100 mg/kg per min) cause systemic
vasodilatation [142]. Adenosine is used to treat persis-
tent pulmonary hypertension of the newborn [143,144].
Treatment with the use of vasoactive intestinal peptide
also may elicit benecial effects in PAH and warrants fur-
ther investigation [145].
Inotropic Agents
Critically ill patients with associated acute pulmonary
hypertension frequently develop profound and refrac-
tory systemic arterial hypotension. In cases with severe
acute arterial pulmonary hypertension (e.g., massive PE),
relieving the acute obstruction (either mechanically or
with pulmonary vasodilators) of the pulmonary circula-
tion can successfully resolve systemic arterial hypoten-
sion. In acute pulmonary hypertension associated with
decreased cardiac contractility and/or SVR, the use of
vasopressors with or without pulmonary vasodilators is
necessary to maintain coronary and end-organ perfu-
sion. Progression of RV failure should be always con-
sidered in the management plan. Cases of severe acute
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 163
The Management of Acute Pulmonary Hypertension G. Hui-li
PAH combined with heart failure and systemic arterial
hypotension require tight hemodynamic monitoring and
aggressive treatment with combinations of pulmonary
vasodilators, inotropic agents, and systemic arterial vaso-
constrictors. The choice of vasopressor and inotropes in
patients with acute pulmonary hypertension should take
into consideration their effects on PVR and cardiac output
when used alone or in combinations with other agents,
and must be individualized based on individual patient
response.
Dobutamine is predominantly a
1
-adrenergic agonist,
with weak 2 activity, and
1
selective activity, although
it is used clinically in cases of cardiogenic shock for its
1
inotropic effect in increasing heart contractility and car-
diac output. At doses of 15 g/kg per min dobutamine
decreased PVR, lowered MAP and slightly increased car-
diac output [146]; at doses of 510 g/kg per min,
dobutamine caused signicant tachycardia and systemic
hypotension without improving PVR [142,147]. Admin-
istration of dobutamine augments myocardial contractil-
ity and reduces left ventricular afterload. It can cause
systemic hypotension in some patients because of pe-
ripheral vasodilatory effects, which may require use of
norepinephrine or other peripheral vasoconstrictors to
maintain appropriate systemic perfusion pressures. In
combination with inhaled NO dobutamine administra-
tion had additive effects on pulmonary circulation [137],
increased cardiac performance, and improved oxygena-
tion [148] with no effect on systemic hemodynamics.
In an animal model of acute RV failure secondary to
acute PAH, dobutamine was superior to norepinephrine
in improving RV function by optimizing pulmonary va-
sodilation, decreasing PA resistance and elastance, and
increasing RV contractility [143]. An arterial catheter
should be placed as soon as possible in patients with sep-
tic shock. Vasopressors are indicated to maintain mean
arterial pressure of <65 mmHg, both during and fol-
lowing adequate uid resuscitation. Norepinephrine or
dopamine is the vasopressor of choice in the treatment
of septic shock. Norepinephrine may be combined with
dobutamine when cardiac output is being measured.
Epinephrine, phenylephrine, and vasopressin are not rec-
ommended as rst-line agents in the treatment of sep-
tic shock. Vasopressin may be considered for salvage
therapy. Low-dose dopamine is not recommended for
the purpose of renal protection. Dobutamine is recom-
mended as the agent of choice to increase cardiac output
but should not be used for the purpose of increasing car-
diac output above physiologic levels [149].
Norepinephrine acts as a drug that will increase blood
pressure by its prominent increasing effects on the vas-
cular tone from -adrenergic receptor activation. The re-
sulting increase in vascular resistance triggers a compen-
satory reex that overcomes its direct stimulatory effects
on the heart, called the baroreceptor reex, which results
in a drop in heart rate called reex bradycardia. Nore-
pinephrine has signicant inotropic effects and produces
vasoconstriction. It is widely used in critical care settings
to treat hemodynamically unstable patients. In addition
to positive effects on cardiac output and systemic arte-
rial pressure, norepinephrine increases PVR and wors-
ens PAH [150]. However, norepeinephrine is superior
to phenylephrine in restoring systemic arterial pressure,
decreasing PVR, augmenting RV myocardial blood ow
and improving cardiac output in animal model of acute
PE [151]. Alfa-adrenergic stimulation can cause a dis-
proportionate rise in PVR [152], which is implicated in
the development of acute PAH in critical illnesses. Be-
sides disproportional increase in PVR, phenylephrine also
causes bradycardia [153] with detrimental consequences
on pulmonary and systemic hemodynamic. The effects
of phenylephrine and norepinephrine in the treatment
of systemic hypotension were evaluated in patients with
chronic PAH who developed systemic hypotension fol-
lowing induction of anesthesia. In contrast to phenyle-
phrine, norepinephrine decreased the ratio of PAP to
systemic blood pressure without a change in cardiac in-
dex. Thus, norepinephrine has been considered to be
preferable to phenylephrine for the treatment of hy-
potension in patients with chronic PAH [145]. There
are growing data on successful combination of nore-
pinephrine and selective pulmonary vasodilators in the
treatment of patients with acute and chronic pulmonary
hypertension [154]. In a small animal study of sepsis-
induced pulmonary hypertension, epinephrine infusion
increased SVR and cardiac output and lowered PVR
[155].
Dopamine is a sympathomimetic catecholamine that
exhibits alpha adrenergic, beta adrenergic, and dopamin-
ergic agonism. Dopamine produces dose-dependent
dopaminergic, beta- and alpha- adrenergic effects on car-
diac output and vascular tone. Low dose: 25 g/kg
per min. Little effect was seen on heart rate or car-
diac output. Increased blood ow accompanied by in-
creased urine output. Intermediate doses: 515 g/kg per
min. An increase in cardiac contractility and cardiac out-
put results in increased normal blood ow and heart
rate. High dose: 15 g/kg per min. Alpha-adrenergic ef-
fects begin to dominate: increased systemic and PVR.
Decrease in normal perfusion. In patients with chronic
PAH dopamine, infusion led to increased heart rate,
mPAP, aortic mean pressure, and cardiac index with con-
comitant fall of SVR [156]. Administration of dopamine,
similar to epinephrine, is associated with high risk of
tachyarrhythmia, with potentially fatal consequences in
patients with severe pulmonary hypertension [146].
164 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
Isoproterenols effects on the cardiovascular system re-
late to its actions on cardiac 1 receptors and 2 recep-
tors on skeletal muscle arterioles. Isoproterenol has pos-
itive inotropic and chronotropic effects on the heart. In
skeletal muscle arterioles, it produces vasodilatation. Its
inotropic and chronotropic effects elevate systolic blood
pressure, while its vasodilatory effects tend to lower dias-
tolic blood pressure. Isoproterenol has positive inotropic
and chronotropic effect, which in therapeutic doses in-
creases cardiac output and produces pulmonary and pe-
ripheral vasodilation. As a pulmonary vasodilator, isopro-
terenol is one of the preferred inotropic agents in heart
transplantation patients with elevated PVR. The dosage of
isoproterenol should be reduced gradually because PVR
may return quickly to elevated baseline levels after dis-
continuation of this drug [148]. In animals with acute
PAH, administration of isoproterenol did not reduce PAP,
and instead produced signicant tachycardia and was as-
sociated with arrhythmias [157].
Vasopressin is derived from a preprohormone precur-
sor that is synthesized in the hypothalamus and stored
in vesicles at the posterior pituitary. Vasopressin is an en-
dogenous peptide hormone with weak nonadrenergic va-
sopressor effect on the systemic vasculature and an ability
to produce NO-mediated selective pulmonary vasodila-
tion [158]. In healthy animals, a linear relationship was
observed between vasopressin levels and systemic vascu-
lar resistance. However, vasopressin did not affect PVR
or any vascular compliance [159]. It was very effective
(at dose of 0.1 U/min) in treating refractory low SVR hy-
potension concomitant with pulmonary hypertension in
postoperative patients [160]. Experimental data on use of
vasopressin in acute pulmonary hypertension are contro-
versial: In one setting of acute pulmonary vasoconstric-
tion vasopressin infusion produced signicant pulmonary
vasodilation [161]. Use of high dose of vasopressin (1.16
units/kg per hour) in another animal model of acute pul-
monary hypertension led to increased PVR and decreased
cardiac output with a decrease in RV contractility, leading
the authors to cautioning against its use when RV func-
tion is compromised [162].
Surgical Intervention
Patients with advanced RV failure who have not bene-
ted from pharmacologic treatment and who have arte-
rial oxygen saturations within an acceptable range should
be considered for percutaneous balloon atrial septostomy
[163,164]. Atrial septostomy has been developed as an
alternative/bridge treatment and applied in patients with
lack of response to medical therapy in the absence of
other surgical treatment options. It has a substantial mor-
bidity and mortality in critically ill patients with severe
RV failure [165]. With growing experience, procedure-
related death rates have been reduced to 5.4%, and the
most suitable patient group has been identied among
patients with a mean right atrial pressure between 10 and
20 mmHg [166]. Acute right heart failure after orthotopic
heart transplantation was successfully managed by de-
compression of the RV through the patent foramen ovale
of the donor heart and inhalation of iloprost [167]. Both
pericardiectomy and creation of atrial septal defects have
been used in extreme cases of acute RV failure secondary
to acute MI [168]. Decompression of the RV through the
septostomy may potentially be an effective alternative in
the management of severe acute pulmonary hyperten-
sion. The defect could be subsequently closed using a
transcatheter septal occlusion device, after the patients
condition has been stabilized. It has been observed that
patients with PPH who have a patent foramen ovale have
a better survival rate than patients with an intact sep-
tum. In an effort to create the more favorable condition
of intraatrial communication, the technique of atrial sep-
tostomy for congenital cardiac defects such as transposi-
tion of the great arteries has been applied to patients with
severe pulmonary hypertension and right heart failure.
This also has been utilized as a bridge to lung transplan-
tation for patients with severe PPH [169].
Mechanical Support Devices
Cardiac surgical patients who do not respond to medical
therapy may be candidates for mechanical support with
a RV assist device. In cases of acute right heart failure af-
ter heart transplantation, mechanical circulatory support
systems, such as RV assist devices, extracorporeal mem-
brane oxygenation, femoral vein-to-femoral artery roller,
or centrifugal pumps, may facilitate hemodynamic stabil-
ity until the transplanted heart has recovered, or until
a new heart has been found for retransplantation [170].
They possibly could be successfully applied in other cases
of potentially reversible acute PAH and right heart fail-
ure. Intraaortic balloon counterpulsation (IABP) has long
been the mainstay of mechanical therapy for cardiogenic
shock. Not every patient has a hemodynamic response
to IABP [188]. However, in patients with acute PAH
and RV failure associated with systemic hypotension, it
could improve coronary and peripheral perfusion and
augment LV performance with an acute decrease in af-
terload [171]. Severe pulmonary hypertension refractory
to medical treatment is a contraindication to orthotopic
heart transplantation in most centers. A study supports
that LVAD support and continuous nonpulsatile mechan-
ical unloading of the left ventricle can reverse medically
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 165
The Management of Acute Pulmonary Hypertension G. Hui-li
unresponsive pulmonary hypertension and render pa-
tients eligible for orthotopic heart transplantation [172].
Mechanical support using an implantable LVAD is a very
efcient approach with an acceptable risk to treat se-
vere pulmonary hypertension in end-stage heart failure
patients before heart transplantation. Adequate reduc-
tion of PVR can be expected within 36 months. Sub-
sequent heart transplantation is associated with a good
outcome [173]. Severely elevated PVR is a relative con-
traindication to orthotopic heart transplantation. A po-
tential novel strategy to reverse elevated PVR may be
implantation of a chronic left ventricular assist device
(LVAD) with subsequent left ventricular unloading. Pa-
tient underwent placement of biventricular assist devices
and subsequently could experience a marked reduction
of PVR, ultimately enabling successful heart transplanta-
tion [174].
Fixed pulmonary hypertension in cardiac transplant
candidates can be lowered using LVADs. The posttrans-
plant survival of these patients is uncertain as pulmonary
hypertension may reappear, possibly affecting posttrans-
plant survival. LVAD therapy lowers xed pulmonary
hypertension in cardiac transplant candidates with xed
pulmonary hypertension. Thereafter, long-term post-
transplant survival is comparable to cardiac transplant re-
cipients without pulmonary hypertension [175]. Elevated
PVR in heart transplant candidates can be reduced using
a LVAD, and LVAD is proposed to be the treatment of
choice for candidates with PH. Pretransplant LVAD ther-
apy reduces an elevated PVR in heart transplant recipi-
ents, but there was no statistically signicant difference
in posttransplant survival in patients with PH with, or
without LVAD therapy [176]. Acute RV failure after car-
diac surgery occurring in the rst postoperative hours is
associated with a bad prognosis. We have used a centrifu-
gal pump either for left, right, or biventricular assistance.
However, the use of this device for pure RV assistance is
rare. We report a 30-year-old female undergoing a mi-
tral valve replacement and a 42-year-old male undergo-
ing a cardiac transplantation, who had a successful RV
assistance using a centrifugal pump, due to a failing right
ventricle, as the result of insufcient myocardial protec-
tion and severe pulmonary hypertension. These two cases
illustrate the value of the mechanical ventricular assist
device for the treatment of right heart failure [177].
The Management of Renal Impairment
Due to PAH
Renal blood ow is the main factor determining GFR
glomerular ltration rate in patients with cardiac dys-
function. Venous congestion, characterized by an in-
creased Right Atrial Pressure, adjusted for RBF is also re-
lated to GFR. Treatment to preserve GFR should not only
focus on improvement of renal perfusion, but also on de-
creasing venous congestion [178].
Combined Therapy Approach
Combination therapy using drugs with different mecha-
nisms of action to maximize clinical benet is an emerg-
ing therapeutic option in PAH [179,180,181,182]. The
choice of drug depends on a variety of factors including
accessibility, approval status, and patients preferences
[183]. The use of combinations of targeted oral thera-
pies for the treatment of PAH is becoming increasingly
commonplace, and benets of early diagnosis and treat-
ment of PAH recently have become apparent [184,185].
The situations and corresponding treatment combina-
tions could be but not limit to as follows:
(1) Protamine-mediated acute PAH and RV failure in the
setting of open heart surgery. PGI
2
in combination with
norepinephrine and dopamine was effective [26].
(2) Acute and chronic pulmonary hypertension and sys-
temic hypotension. Combination of norepinephrine and
selective pulmonary vasodilators should be adopted.
(3) Severe pulmonary hypertension leading to impaired
RV function presented in ARDS. The combination of iNO
and intravenously administered prostacyclin (i.v. PGI2)
might be more useful than either drug alone [186].
(4) Severe pulmonary hypertension. iNO therapy alone
or in combination with a PDE5 inhibtor could be a thera-
peutic alternative [187].
(5) Long-term treatment of PAH and CTEPH patients.
The combination of sildenal and inhaled treprostinil was
well tolerated and induced additive, pulmonary selective
vasodilatation in pulmonary hypertension patients [188].
(6) In combination with iNO, milrinone produced addi-
tive pulmonary vasodilatation in animal model of acute
pulmonary hypertension. A combination of milrinone
and sildenal led to more effective pulmonary vasodi-
lation and increased RV contractility, without additional
systemic vasodilatation in an animal model. Cardiac out-
put and RV performance were signicantly improved
after milrinone or both drugs combined, but not with
sildenal alone.
(7) The combination of PDE3 inhibitor and PDE4 in-
hibitor: Milrinone and sildenal are effective pulmonary
vasodilators, with independent action and additive ef-
fect. Both drugs combined, achieved a better hemody-
namic prole, with greater pulmonary vasodilatation and
increased contractility but without additional systemic
vasodilatation. The systemic hemodynamic prole (sys-
temic vasodilation, cardiac output, RV dP/dT) is improved
with milrinone but not with sildenal [189].
166 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
(8) Bosentan has not been studied in acute care settings.
Currently, the only possible implication of Bosentan in
critically ill patients would be weaning or conversion
from inhaled or intravenous pulmonary vasodilators to
oral medication (Bosentan), but ET blockers are prob-
ably not a good option for acute PAH due to ARDS.
Some study investigating the combination of Bosen-
tan and prostacyclin analogue showed clinical improve-
ment. Additional Bosentan therapy may also reduce
the epoprostenol dose and therefore decrease its side
effects.
(9) PAH patients with NYHA IIIV symptoms. A non-
randomized, uncontrolled trial in reported the effective-
ness of bosentan monotherapy followed by the addition
of sildenal and/or inhaled iloprost [190].
(10) Acute PAH after heart transplantation: Superim-
posed acute RV dysfunction in the setting of preexisting
pulmonary hypertension is a nearly fatal complication af-
ter heart transplantation. The optimal treatment modal-
ity remains a matter of debate. Recently, sildenal citrate,
in combination with iNO, has gained popularity and was
proved safe and effective in the treatment of acute PAH
after heart transplant [191].
Suggestions of Interventions
for Acute PAH
No current treatment approach to PAH provides a cure.
Rather, treatment goals are to reduce PVR, PAP, and
symptoms and to increase patient activity and longevity.
With an increasing number of pharmacologic treatment
options available, the decision regarding when and how
to use such treatment have taken on added complexity
and importance. Although head-to-head comparative tri-
als of available medications are not available, most ex-
perienced investigators and clinicians agree that patients
with more advanced PAH should be treated initially with
parenteral prostacyclin analogues, especially in the set-
ting of RV failure. PDE5 inhibitors or ETRAs are rea-
sonable initial therapies for patients who have mild-to-
moderate PAH (i.e., WHO functional class II or III) or for
patients who are not candidates for more invasive ther-
apies. Conversely, intravenous therapies with a prosta-
cyclin analogue should be considered as rst-line ther-
apy for patients with severe symptoms (WHO functional
class IV) or for patients whose disease progresses on less
invasive therapy. Use of inhaled iloprost and subcuta-
neously administered treprostinil allows for the admin-
istration of an effective prostacyclin analogue without
some of the risks associated with continuous intravenous
infusion. However, when inhaled or delivered subcuta-
neously, these agents appear to be less efcacious than
when delivered via the parenteral route and may not be
well tolerated by the patient. Interventional procedures
such as atrial septostomy and lung transplantation are in-
dicated in patients with advanced NYHA class III and IV
symptoms and refractory to available medical treatment.
Patients who respond to an acute trial of a vasodila-
tor may be treated with oral CCB, whereas oral therapies
such as sildenal and bosentan have been effective in pa-
tients with mild-to-moderate symptoms. Infusions of the
prostacyclin analogues epoprostenol and treprostinil ap-
pear to be the treatment of choice for moderate-to-severe
PAH, and agents with alternate routes of delivery such as
inhaled iloprost may be advantageous in adjunctive roles.
Future trials that focus on the long-term effects of cur-
rently available agents, as well as on combination ther-
apy, are needed [192].
In patients at risk of sudden intravascular absorption of
absolute ethyl alcohol, an increased level of awareness for
acute PAH, together with prompt treatment of PAH with
Sildenal analogue or with a low dose of an NO-donor
drug, will minimize the risk of cardiovascular decompen-
sation and may be the treatment of such catastrophe.
Conict of Interest
The authors have no conict of interest.
References
1. Barber` a JA, Escribano P, Morales P, G omez MA, Oribe
M, Martnez A, Rom an A, Segovia J, Santos F, Subirana
MT. Sociedad Espa nola de Neumologa y Ciruga
Tor acica; Sociedad Espa nola de Cardiologa. [Standards
of care in pulmonary hypertension. Consensus
statement of the Spanish Society of Pulmonology and
Thoracic Surgery (SEPAR) and the Spanish Society of
Cardiology (SEC)]. Rev Esp Cardiol 2008;61:170184.
2. McGoon MD, Kane GC. Pulmonary hypertension:
Diagnosis and management. Mayo Clin Proc
2009;84:191207.
3. McLaughlin VV, Archer SL, Badesch DB, et al. ACCF
(AHA 2009 Expert Consensus Document on Pulmonary
Hypertension: A Report of the American College of
Cardiology Foundation Task Force on Clinical Expert
Consensus Documents. J Am Coll Cardiol
2009;53:15731619.
4. Rich S, Dantzker DR, Ayres SM, Bergofsky EH,
Brundage BH, Detre KM, Fishman AP, Goldring RM,
Groves BM, Koerner SK. Primary pulmonary
hypertension: A national prospective study. Ann Intern
Med 1987;107:216223.
5. Humbert M, Morrell NW, Archer SL, Stenmark KR,
MacLean MR, Lang IM, Christman BW, Weir EK,
Eickelberg O, Voelkel NF, et al. Cellular and molecular
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 167
The Management of Acute Pulmonary Hypertension G. Hui-li
pathobiology of pulmonary arterial hypertension. J Am
Coll Cardiol 2004;43(12 Suppl S):13S24S.
6. Moloney ED, Evans TW. Pathophysiology and
pharmacological treatment of pulmonary hypertension
in acute respiratory distress syndrome. Eur Respir J
2003;21:720727.
7. Albertini M, Ciminaghi B, Mazzola S, Clement MG.
Improvement of respiratory function by bosentan during
endotoxic shock in the pig. Prostaglandins Leukot Essent
Fatty Acids 2001;65:103108.
8. Wort SJ, Evans TW. The role of the endothelium in
modulating vascular control in sepsis and related
conditions. Br Med Bull 1999;55:3048.
9. Lambermont B, Kolh P, Detry O, Gerard P, Marcelle R,
DOrio V. Analysis of endotoxin effects on the intact
pulmonary circulation. Cardiovasc Res 1999;41:
275281.
10. Via G, Braschi A. Pathophysiology of severe pulmonary
hypertension in the critically ill patient. Minerva
Anestesiol 2004;70:233237.
11. Nath J, Foster E, Heidenreich PA. Impact of tricuspid
regurgitation on long-term survival. J Am Coll Cardiol
2004;43:405409.
12. Blaise G, Langleben D, Hubert B. Pulmonary arterial
hypertension: Pathophysiology and anesthetic approach.
Anesthesiology 2003;99:14151432.
13. Chin KM, Kim NH, Rubin LJ. The right ventricle in
pulmonary hypertension. Coron Artery Dis
2005;16:1318.
14. Gaine SP, Rubin LJ. Primary pulmonary hypertension.
Lancet 1998;352:719725.
15. Hoeper MM, Gali e N, Murali S, Olschewski H, Rubenre
M, Robbins IM, Farber HW, McLaughlin V, Shapiro S,
Pepke-Zaba J, et al. Outcome after cardiopulmonary
resuscitation in patients with pulmonary arterial
hypertension. Am J Respir Crit Care Med
2002;165:341344.
16. Vieillard-Baron A, Schmitt JM, Augarde R, Fellahi JL,
Prin S, Page B, Beauchet A, Jardin F. Acute cor
pulmonale in acute respiratory distress syndrome
submitted to protective ventilation: Incidence, clinical
implications, and prognosis. Crit Care Med
2001;29:15511555.
17. Leeman M. Pulmonary hypertension in acute
respiratory distress syndrome. Monaldi Arch Chest Dis
1999;54:146149.
18. Jardin F, Vieillard-Baron A. Is there a safe plateau
pressure in ARDS? The right heart only knows. Intensive
Care Med 2007;33:444447.
19. Sibbald WJ, Paterson NA, Holliday RL, Anderson RA,
Lobb TR, Duff JH. Pulmonary hypertension in sepsis:
Measurement by the pulmonary arterial
diastolic-pulmonary wedge pressure gradient and the
inuence of passive and active factors. Chest
1978;73:583591.
20. Kostrubiec M, Pedowska-Woszek J, Ciurzy nski M,
Bienias P, Pacho S, Piaskowska M, Pruszczyk P.
Endothelin is not elevated in acute pulmonary
embolism. Thromb Res 2009. [Epub ahead of print].
21. Romand JA, Donald FA, Suter PM. Cardiopulmonary
interactions in acute lung injury: Clinical and prognostic
importance of pulmonary hypertension. w Horiz
1994;2:457462.
22. Moloney ED, Evans TW. Pathophysiology and
pharmacological treatment of pulmonary hypertension
in acute respiratory distress syndrome. Eur Respir J
2003;21:720727.
23. Luhr O, Nathorst-Westfelt U, Lundin S, Wickerts CJ,
Stiernstr om H, Berggren L, Aardal S, Johansson LA,
Stenqvist O, Rudberg U, et al. A retrospective analysis of
nitric oxide inhalation in patients with severe acute lung
injury in Sweden and Norway 19911994. Acta
Anaesthesiol Scand 1997;41:12381246.
24. Goldsmith JA, Kavanagh BP, Pearl RG. Plasma
potentiates the priming effects of endotoxin on platelet
activating factor-induced pulmonary hypertension in
the rabbit lung. Anesth Analg 1996;83:242246.
25. Clavijo LC, Carter MB, Matheson PJ, Wills-Frank LA,
Wilson MA, Wead WB, Garrison RN. Platelet-activating
factor and bacteremia-induced pulmonary hypertension.
J Surg Res 2000;88:173180.
26. Snapper JR, Thabes JS, Lefferts PL, Lu W. Role of
endothelin in endotoxin-induced sustained pulmonary
hypertension in sheep. Am J Respir Crit Care Med
1998;157:8188.
27. Bernstein AD, Parsonnet V. Bedside estimation of risk as
an aid for decision-making in cardiac surgery. Ann
Thorac Surg 2000;69:823828.
28. Subramaniam K, Yared JP. Management of pulmonary
hypertension in the operating room. Semin Cardiothorac
Vasc Anesth 2007;11:119136.
29. Wynne R, Botti M. Postoperative pulmonary
dysfunction in adults after cardiac surgery with
cardiopulmonary bypass: Clinical signicance and
implications for practice. Am J Crit Care
2004;13:384393.
30. Ocal A, Kiris I, Erdinc M, Peker O, Yavuz T, Ibrisim E.
Efciency of prostacyclin in the treatment of
protamine-mediated right ventricular failure and acute
pulmonary hypertension. Tohoku J Exp Med
2005;207:5158.
31. Milot J, Perron J, Lacasse Y, L etourneau L, Cartier PC,
Maltais F. Incidence and predictors of ARDS after cardiac
surgery. Chest 2001;119:884888.
32. Coe PF. Managing pulmonary hypertension in heart
transplantation: meeting the challenge. Crit Care Nurse
2000;20:2228, quiz 2930.
33. Jean-Pierre C, S ebastien R, Medhi S, Bruno S, Patricia D,
Yves B, Paul Michel M, Jean-Pierre V. Acute pulmonary
hypertension after cardiopulmonary bypass in pig: The
168 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
role of endogenous endothelin. Eur J Cardiothorac Surg
1999;15:346352.
34. Urdaneta F, Lobato EB, Beaver T, Muehlschlegel JD,
Kirby DS, Klodell C, Sidi A. Treating pulmonary
hypertension post cardiopulmonary bypass in pigs:
Milrinone vs. sildenal analog. Perfusion
2008;23:117125.
35. Sidi A, Naik B, Urdaneta F, Muehlschegel JD, Kirby DS,
Lobato EB. Treatment of ethanol-induced acute
pulmonary hypertension and right ventricular
dysfunction in pigs, by Sildenal analogue(UK343-664)
or nitroglycerin. Annals of Cardiac Anaesthesia
2008;112:96103.
36. Kothari SS, Duggal B. Chronic oral sildenal therapy in
severe pulmonary artery hypertension. Indian Heart J
2002;54:404409.
37. Galie N, Torbicki A, Barst R, Dartevelle P, Haworth S,
Higenbottam T, Olschewski H, Peacock A, Pietra G,
Rubin LJ, et al. Guidelines on diagnosis and treatment of
pulmonary arterial hypertension: The Task Force on
Diagnosis and Treatment of Pulmonary Arterial
Hypertension of the European Society of Cardiology.
EurHeart J 2004;25:22432278.
38. Chemla D, Castelain V, Herv e P, Lecarpentier Y,
Brimioulle S. Haemodynamic evaluation of pulmonary
hypertension. Eur Respir J 2002;20:13141331.
39. Fincke R, Hochman JS, Lowe AM, Menon V, Slater JN,
Webb JG, LeJemtel TH, Cotter G, SHOCK Investigators.
Cardiac power is the strongest hemodynamic correlate
of mortality in cardiogenic shock: A report from the
SHOCK trial registry. J Am Coll Cardiol 2004;44:340348.
40. Bossone E, Bodini BD, Mazza A, Allegra L. Pulmonary
arterial hypertension: The key role of echocardiography.
Chest 2005;127:18361843.
41. McConnell MV, Solomon SD, Rayan ME, Come PC,
Goldhaber SZ, Lee RT. Regional right ventricular
dysfunction detected by echocardiography in acute
pulmonary embolism. Am J Cardiol 1996;78:469473.
42. Engelke C, Rummeny E, Marten K. Acute pulmonary
embolism: Prediction of cor pulmonale and short-term
patient survival from assessment of cardiac dimensions
in routine multidetector-row CT. Rofo
2006;178:9991006.
43. Karski JM. Transesophageal echocardiography in the
intensive care unit. Semin Cardiothorac Vasc Anesth
2006;10:162166.
44. Schneider C, Schwemmer U, Kredel M, Frommer M,
Wurmb T, Greim C, Roewer N, Brederlau J. Handheld
vs. conventional transesophageal echocardiography in
non-cardiac surgical intensive care unit patients.
Ultraschall Med 2008;29:531534.
45. Cotrim C, Miranda R, Loureiro MJ, Almeida S, Lopes L,
Almeida AR, Fernandes R, Sim oes O, Cordeiro A,
Cordeiro P, et al. Echocardiography during treadmill
exercise testing for evaluation of pulmonary artery
systolic pressure: Advantages of the method. Rev Port
Cardiol 2008;27:453461.
46. Kouzu H, Nakatani S, Kyotani S, Kanzaki H, Nakanishi
N, Kitakaze M. Noninvasive estimation of pulmonary
vascular resistance by Doppler echocardiography in
patients with pulmonary arterial hypertension. Am J
Cardiol 2009;103:872876. Epub 2009 Jan 24.
47. Haddad F, Zamanian R, Beraud AS, Schnittger I,
Feinstein J, Peterson T, Yang P, Doyle R, Rosenthal D. A
novel non-invasive method of estimating pulmonary
vascular resistance in patients with pulmonary arterial
hypertension. J Am Soc Echocardiogr 2009. [Epub ahead
of print]
48. Benza R, Biederman R, Murali S, Gupta H. Role of
cardiac magnetic resonance imaging in the management
of patients with pulmonary arterial hypertension. J Am
Coll Cardiol 2008;52:16831692.
49. Kovacs G, Reiter G, Reiter U, Rienm uller R, Peacock A,
Olschewski H. The emerging role of magnetic resonance
imaging in the diagnosis and management of pulmonary
hypertension. Respiration 2008;76:458470. Epub 2008
Nov 12.
50. Nagaya N, Nishikimi T, Okano Y, Uematsu M, Satoh T,
Kyotani S, Kuribayashi S, Hamada S, Kakishita M,
Nakanishi N, et al. Plasma brain natriuretic peptide
levels increase in proportion to the extent of right
ventricular dysfunction in pulmonary hypertension.
J Am Coll Cardiol 1998;31:202208.
51. Nagaya N, Nishikimi T, Uematsu M, Satoh T, Kyotani S,
Sakamaki F, Kakishita M, Fukushima K, Okano Y,
Nakanishi N, et al. Plasma brain natriuretic peptide as a
prognostic indicator in patients with primary pulmonary
hypertension. Circulation 2000;102:865870.
52. Leuchte HH, Holzapfel M, Baumgartner RA, Ding I,
Neurohr C, Vogeser M, Kolbe T, Schwaiblmair M, Behr
J. Clinical signicance of brain natriuretic peptide in
primary pulmonary hypertension. J Am Coll Cardiol
2004;43:764770.
53. Nagaya N, Nishikimi T, Uematsu M, Satoh T, Kyotani S,
Sakamaki F, Kakishita M, Fukushima K, Okano Y,
Nakanishi N, et al. Plasma brain natriuretic peptide as a
prognostic indicator in patients with primary pulmonary
hypertension. Circulation 2000;102:865870.
54. Leuchte HH, El Nounou M, Tuerpe JC, Hartmann B,
Baumgartner RA, Vogeser M, Muehling O, Behr J.
N-terminal pro-brain natriuretic peptide and renal
insufciency as predictors of mortality in pulmonary
hypertension. Chest 2007;131:402409.
55. La Vecchia L, Ottani F, Favero L, Spadaro GL, Rubboli A,
Boanno C, Mezzena G, Fontanelli A, Jaffe AS. Increased
cardiac troponin I on admission predicts in-hospital
mortality in acute pulmonary embolism. Heart
2004;90:633637.
56. Mehta NJ, Jani K, Khan IA. Clinical usefulness and
prognostic value of elevated cardiac troponin I levels in
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 169
The Management of Acute Pulmonary Hypertension G. Hui-li
acute pulmonary embolism. Am Heart J
2003;145:821825.
57. Sanchez O, Mari e E, Lerolle U, Wermert D, Isra el-Biet D,
Meyer G. Pulmonary arterial hypertension in women.
Rev Mal Respir 2008;25:451460.
58. Jas X, Sitbon O, Savale L, Montani D, Humbert M,
Simonneau G. Pulmonary arterial hypertension. Rev Prat
2008;58:19972010.
59. Desole S, K ahler CM. Decompensated pulmonary
hypertension. Dtsch Med Wochenschr 2008;133(Suppl
6):S215S218. Epub 2008 Sep 23.
60. Jacobs AK, Leopold JA, Bates E, Mendes LA, Sleeper LA,
White H, Davidoff R, Boland J, Modur S, Forman R, et
al. Cardiogenic shock caused by right ventricular
infarction: A report from the SHOCK registry. J Am Coll
Cardiol 2003;41:12731279.
61. Berisha S, Kastrati A, Goda A, Popa Y. Optimal value of
lling pressure in the right side of the heart in acute
right ventricular infarction. Br Heart J 1990;63:98102.
62. Jardin F, Delorme G, Hardy A, Auvert B, Beauchet A,
Bourdarias JP. Reevaluation of hemodynamic
consequences of positive pressure ventilation: Emphasis
on cyclic right ventricular afterloading by mechanical
lung ination. Anesthesiology 1990;72:966970.
63. Vieillard-Baron A, Loubieres Y, Schmitt JM, Page B,
Dubourg O, Jardin F. Cyclic changes in right ventricular
output impedance during mechanical ventilation. J Appl
Physiol 1999;87:16441650.
64. Schmitt JM, Vieillard-Baron A, Augarde R, Prin S, Page
B, Jardin F. Positive end-expiratory pressure titration in
acute respiratory distress syndrome patients: Impact on
right ventricular outow impedance evaluated by
pulmonary artery Doppler ow velocity measurements.
Crit Care Med 2001;29:11541158.
65. Miranda DR, Klompe L, Cademartiri F, Haitsma JJ,
Palumbo A, Takkenberg JJ, Lachmann B, Bogers AJ,
Gommers D. The effect of open lung ventilation on right
ventricular and left ventricular function in lung-lavaged
pigs. Crit Care 2006;10:R86.
66. Reis Miranda D, Gommers D, Struijs A, Meeder H,
Schepp R, Hop W, Bogers A, Klein J, Lachmann B. The
open lung concept: Effects on right ventricular afterload
after cardiac surgery. Br J Anaesth 2004;93:327332.
67. Balanos GM, Talbot NP, Dorrington KL, Robbins PA.
Human pulmonary vascular response to 4 h of
hypercapnia and hypocapnia measured using Doppler
echocardiography. J Appl Physiol 2003;94:15431551.
68. Roberts DH, Lepore JJ, Maroo A, Semigran MJ, Ginns
LC. Oxygen therapy improves cardiac index and
pulmonary vascular resistance in patients with
pulmonary hypertension. Chest 2001;120:15471555.
69. Santos C, Ferrer M, Roca J, Torres A, Hern andez C,
Rodriguez-Roisin R. Pulmonary gas exchange response
to oxygen breathing in acute lung injury. Am J Respir Crit
Care Med 2000;161:2631.
70. Zamanian RT, Haddad F, Doyle RL, Weinacker AB.
Management strategies for patients with pulmonary
hypertension in the intensive care unit. Crit Care Med
2007;35:20372050.
71. Monagle P, Chan A, Massicotte P, Chalmers E,
Michelson AD. Antithrombotic therapy for venous
thromboembolic disease: The Seventh ACCP Conference
on Antithrombotic and Thrombolytic Therapy. Chest
2004;126(3 Suppl):645S687S.
72. Yoshida M, Inoue I, Kawagoe T, Ishihara M, Shimatani
Y, Kurisu S, Kusano KF, Ohe T. Novel percutaneous
catheter thrombectomy in acute massive pulmonary
embolism: Rotational bidirectional thrombectomy
(ROBOT). Catheter Cardiovasc Interv 2006;68:112117.
73. Humbert M, Morrell NW, Archer SL, et al. Cellular and
molecular pathobiology of pulmonary arterial
hypertension. J Am Coll Cardiol 2004;43(Suppl
S):13S24S.
74. Bresser P, Fedullo PF, Auger WR, Channick RN, Robbins
IM, Kerr KM, Jamieson SW, Rubin LJ. Continuous
intravenous epoprostenol for chronic thromboembolic
pulmonary hypertension. Eur Respir J 2004;23:595600.
75. Ulrich S, Speich R, Domenighetti G, Geiser T, Aubert JD,
Rochat T, Huber L, Treder U, Fischler M. Bosentan
therapy for chronic thromboembolic pulmonary
hypertension. A national open label study assessing the
effect of Bosentan on haemodynamics, exercise capacity,
quality of life, safety and tolerability in patients with
chronic thromboembolic pulmonary hypertension
(BOCTEPH-Study). Swiss Med Wkly 2007;137:
573580.
76. Pepke-Zaba J, Higenbottam TW, Dinh-Xuan AT, Stone
D, Wallwork J. Inhaled nitric oxide as a cause of
selective pulmonary vasodilatation in pulmonary
hypertension. Lancet 1991;338:11731174.
77. Zwissler B, Welte M., Messmer K. Effects of inhaled
prostacyclin as compared with inhaled nitric oxide on
right ventricular performance in hypoxic pulmonary
vasoconstriction. J Cardiothorac Vasc Anesth
1995;9:283289.
78. Cockrill BA, Kacmarek RM, Fifer MA, Bigatello LM,
Ginns LC, Zapol WM, Semigran MJ. Comparison of the
effects of nitric oxide, nitroprusside, and nifedipine on
hemodynamics and right ventricular contractility in
patients with chronic pulmonary hypertension. Chest
2001;119:128136.
79. Bhorade S, Christenson J, Oconnor M, Lavoie A,
Pohlman A, Hall JB. Response to inhaled nitric oxide in
patients with acute right heart syndrome. Am J Respir
Crit Care Med 1999;159:571579.
80. Fierobe L, Brunet F, Dhainaut JF, Monchi M, Belghith
M, Mira JP, Dallava-Santucci J, Dinh-Xuan AT. Effect of
inhaled nitric oxide on right ventricular function in
adult respiratory distress syndrome. Am J Respir Crit Care
Med 1995;151:14141419.
170 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
81. McIntyre RC Jr, Moore FA, Moore EE, Piedalue F,
Haenel JS, Fullerton DA. Inhaled nitric oxide variably
improves oxygenation and pulmonary hypertension in
patients with acute respiratory distress syndrome.
J Trauma 1995;39:418425.
82. Taylor RW, Zimmerman JL, Dellinger RP, Straube RC,
Criner GJ, Davis K Jr, Kelly KM, Smith TC, Small RJ,
Inhaled Nitric Oxide in ARDS Study Group. Low-dose
inhaled nitric oxide in patients with acute lung injury: A
randomized controlled trial. Jama 2004;291:16031609.
83. Kaisers U, Busch T, Deja M, Donaubauer B, Falke KJ.
Selective pulmonary vasodilation in acute respiratory
distress syndrome. Crit Care Med 2003;31(4
Suppl):S337S342.
84. Della Rocca G, Coccia C. Nitric oxide in thoracic surgery.
Minerva Anestesiol 2005;71:313318.
85. George I, Xydas S, Topkara VK, Ferdinando C, Barnwell
EC, Gableman L, Sladen RN, Naka Y, Oz MC. Clinical
indication for use and outcomes after inhaled nitric
oxide therapy. Ann Thorac Surg 2006;82:21612169.
86. Deb B, Bradford K, Pearl RG. Additive effects of inhaled
nitric oxide and intravenous milrinone in experimental
pulmonary hypertension. Crit Care Med
2000;28:795799.
87. Troncy E, Francoeur M, Blaise G. Inhaled nitric oxide:
clinical applications, indications, and toxicology. Can J
Anaesth 1997;44:973988.
88. Christenson J, Lavoie A, OConnor M, Bhorade S,
Pohlman A, Hall JB. The incidence and pathogenesis of
cardiopulmonary deterioration after abrupt withdrawal
of inhaled nitric oxide. Am J Respir Crit Care Med
2000;161:14431449.
89. McLaughlin VV, Shillington A, Rich S. Survival in
primary pulmonary hypertension: The impact of
epoprostenol therapy. Circulation 2002;106:14771482.
90. McLaughlin VV, Genthner DE, Panella MM, Rich S.
Reduction in pulmonary vascular resistance with
long-term epoprostenol (prostacyclin) therapy in
primary pulmonary hypertension. N Engl J Med
1998;338:273277.
91. Gomberg-Maitland M., Olschewski H. Prostacyclin
therapies for the treatment of pulmonary arterial
hypertension. Eur Respir J 2008;31:891901.
92. Hach e M, Denault AY, B elisle S, Couture P, Babin D,
T etrault F, Guimond JG. Inhaled prostacyclin (PGI2) is
an effective addition to the treatment of pulmonary
hypertension and hypoxia in the operating room and
intensive care unit. Can J Anaesth 2001;48:924929.
93. Theodoraki K, Rellia P, Thanopoulos A, Tsourelis L,
Zarkalis D, Sfyrakis P, Antoniou T. Inhaled iloprost
controls pulmonary hypertension after cardiopulmonary
bypass. Can J Anaesth 2002;49:963967.
94. Domenighetti G, Stricker H, Waldispuehl B. Nebulized
prostacyclin (PGI2) in acute respiratory distress
syndrome: impact of primary (pulmonary injury) and
secondary (extrapulmonary injury) disease on gas
exchange response. Crit Care Med 2001;29:5762.
95. Olschewski H, Walmrath D, Schermuly R, Ghofrani A,
Grimminger F, Seeger W. Aerosolized prostacyclin and
iloprost in severe pulmonaryhypertension. Ann Intern
Med 1996;124:820824.
96. Olschewski H, Ghofrani HA, Walmrath D, Schermuly R,
Temmesfeld-Wollbruck B, Grimminger F, Seeger W.
Inhaled prostacyclin and iloprost in severe pulmonary
hypertension secondary to lung brosis. Am J RespirCrit
Care Med 1999;160:600607.
97. Badesch DB, Abman SH, Simonneau G, Rubin LJ,
McLaughlin VV. Medical therapy for pulmonary arterial
hypertension: Updated ACCP evidence-based clinical
practice guidelines. Chest 2007;131:19171928.
98. Badesch DB, Abman SH, Ahearn GS, Barst RJ, McCrory
DC, Simonneau G, McLaughlin VV. Medical therapy for
pulmonary arte- rial hypertension: ACCP
evidence-based clinical practice guidelines. Chest
2004;126:35S62S.
99. Humbert M, Sitbon O, Simonneau G. Treatment of
pulmonary arterial hypertension. N Engl J Med
2004;351:14251436.
100. Kerbaul F, Brimioulle S, Rondelet B, Dewachter C,
Hubloue I, Naeije R. How prostacyclin improves cardiac
output in right heart failure in conjunction with
pulmonary hypertension. Am J Respir Crit Care Med
2007;175:846850.
101. Michelakis E, Tymchak W, Lien D, Webster L,
Hashimoto K, Archer S. Oral sildenal is an effective and
specic pulmonary vasodilator in patients with
pulmonary arterial hypertension: comparison with
inhaled nitric oxide. Circulation 2002;105:23982403.
102. Mikhail GW, Prasad SK, Li W, Rogers P, Chester AH,
Bayne S, Stephens D, Khan M, Gibbs JS, Evans TW,
et al. Clinical and haemodynamic effects of sildenal in
pulmonary hypertension: acute and mid-term effects.
Eur Heart J 2004;25:431436.
103. Lepore JJ, Maroo A, Bigatello LM, Dec GW, Zapol WM,
Bloch KD, Semigran MJ. Hemodynamic effects of
sildenal in patients with congestive heart failure and
pulmonary hypertension: Combined administration
with inhaled nitric oxide. Chest 2005;127:16471653.
104. Fung E, Fiscus RR, Yim AP, Angelini GD, Ari AA. The
potential use of type-5 phosphodiesterase inhibitors in
coronary artery bypass graft surgery. Chest
2005;128:30653073.
105. Stiebellehner L, Petkov V, Vonbank K, Funk G, Schenk
P, Ziesche R, Block LH. Long-term treatment with oral
sildenal in addition to continuous IV epoprostenol in
patients with pulmonary arterial hypertension. Chest
2003;123:12931295.
106. Ghofrani HA, Wiedemann R, Rose F, Olschewski H,
Schermuly RT, Weissmann N, Seeger W, Grimminger F.
Combination therapy with oral sildenal and inhaled
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 171
The Management of Acute Pulmonary Hypertension G. Hui-li
iloprost for severe pulmonary hypertension. Ann Intern
Med 2002;136:515522.
107. Ghofrani HA, Rose F, Schermuly RT, Olschewski H,
Wiedemann R, Kreckel A, Weissmann N, Ghofrani S,
Enke B, Seeger W, et al. Oral sildenal as long-term
adjunct therapy to inhaled iloprost in severe pulmonary
arterial hypertension. J Am Coll Cardiol 2003;42:158164.
108. Klinger JR, Thaker S, Houtchens J, Preston IR, Hill NS,
Farber HW. Pulmonary hemodynamic responses to
brain natriuretic peptide and sildenal in patients with
pulmonary arterial hypertension. Chest
2006;129:417425.
109. Ng J, Finney SJ, Shulman R, Bellingan GJ, Singer M,
Glynne PA. Treatment of pulmonary hypertension in
the general adult intensive care unit: A role for oral
sildenal? Br J Anaesth 2005;94:774777.
110. Trachte AL, Lobato EB, Urdaneta F, Hess PJ, Klodell CT,
Martin TD, Staples ED, Beaver TM. Oral sildenal
reduces pulmonary hypertension after cardiac surgery.
Ann Thorac Surg 2005;79:194197; discussion 194197.
111. Namachivayam P, Theilen U, Butt WW, Cooper SM,
Penny DJ, Shekerdemian LS. Sildenal prevents
rebound pulmonary hypertension after withdrawal of
nitric oxide in children. Am J Respir Crit Care Med
2006;174:10421047.
112. Atz AM, Wessel DL. Sildenal ameliorates effects of
inhaled nitric oxide withdrawal. Anesthesiology
1999;91:307310.
113. Johnson RF, Loyd JE, Mullican AL, Fink CA, Robbins
IM. Long-term follow-up after conversion from
intravenous epoprostenol to oral therapy with bosentan
or sildenal in 13 patients with pulmonary arterial
hypertension. J Heart Lung Transplant 2007;26:363369.
114. Aubin MC, Laurendeau S, Mommerot A, Lamarche Y,
Denault A, Carrier M, Perrault LP. Differential effects of
inhaled and intravenous sildenal in the prevention of
the pulmonary endothelial dysfunction due to
cardiopulmonary bypass. J Cardiovasc Pharmacol
2008;51:1117.
115. Keogh AM, Jabbour A, Hayward CS, Macdonald PS.
Clinical deterioration after sildenal cessation in patients
with pulmonary hypertension. Vasc Health Risk Manag
2008;4:11111113.
116. Suntharalingam J, Treacy CM, Doughty NJ, Goldsmith
K, Soon E, Toshner MR, Sheares KK, Hughes R, Morrell
NW, Pepke-Zaba J. Long-term use of sildenal in
inoperable chronic thromboembolic pulmonary
hypertension. Chest 2008;134:229236. Epub 2008
Feb 8.
117. Grossman E, Messerli FH. Calcium antagonists. Prog
Cardiovasc Dis 2004;47:3457.
118. Badesch DB, Abman SH, Simonneau G, Rubin LJ,
McLaughlin VV. Medical therapy for pulmonary arterial
hypertension: updated ACCP evidence-based clinical
practice guidelines. Chest 2007;131:19171928.
119. Rich S, Kaufmann E, Levy PS. The effect of high doses of
calciumchannel blockers on survival in primary
pulmonary hypertension. N Engl J Med 1992;327:7681.
120. Weir EK, Rubin LJ, Ayres SM, Bergofsky EH, Brundage
BH, Detre KM, Elliott CG, Fishman AP, Goldring RM,
Groves BM. The acute administration of vasodilators in
primary pulmonary hypertension: experience from the
National Institutes of Health Registry on Primary
Pulmonary Hypertension. Am Rev Respir Dis
1989;140:16231630.
121. Badesch DB, Abman SH, Ahearn GS, Barst RJ, McCrory
DC, Simonneau G, McLaughlin VV. Medical therapy for
pulmonary arterial hypertension: ACCP evidence-based
clinical practice guidelines. Chest 2004;126:35S62S.
122. Sitbon O, Humbert M, Jas X, Ioos V, Hamid AM,
Provencher S, Garcia G, Parent F, Herv e P, Simonneau
G. Long-term response to calcium channel blockers in
idiopathic pulmonary arterial hypertension. Circulation
2005;111:31053111. Epub 2005 Jun 6.
123. Bresser P, Pepke-Zaba J, Jas X, Humbert M, Hoeper
MM. Medical therapies for chronic thromboembolic
pulmonary hypertension: An evolving treatment
paradigm. Proc Am Thorac Soc 2006;3:594600.
124. Abman SH. Role of endothelin receptor antagonists in
the treatment of pulmonary arterial hypertension. Annu
Rev Med 2008. [Epub ahead of print]
125. Steiner MK, Preston IR. Optimizing endothelin receptor
antagonist use in the management of pulmonary arterial
hypertension. Vasc Health Risk Manag 2008;4:943952
126. Trow TK, Taichman DB. Endothelin receptor blockade in
the management of pulmonary arterial hypertension:
Selective and dual antagonism. Respir Med 2009. [Epub
ahead of print]
127. Dingemanse J, van Giersbergen PL. Clinical
pharmacology of bosentan, a dual endothelin receptor
antagonist. Clin Pharmacokinet 2004;43:10891115.
128. Ulrich S, Speich R, Domenighetti G, Geiser T, Aubert JD,
Rochat T, Huber L, Treder U, Fischler M. Bosentan
therapy for chronic thromboembolic pulmonary
hypertension. A national open label study assessing the
effect of Bosentan on haemodynamics, exercise capacity,
quality of life, safety and tolerability in patients with
chronic thromboembolic pulmonary hypertension
(BOCTEPH-Study). Swiss Med Wkly 2007;137:573580.
129. Benza RL, Barst RJ, Galie N, Frost A, Girgis RE, Highland
KB, Strange C, Black CM, Badesch DB, Rubin L, et al.
Sitaxsentan for the treatment of pulmonary arterial
hypertension: a 1-year, prospective, open-label
observation of outcome and survival. Chest
2008;134:775782. Epub 2008 Jul 14.
130. MacIntyre IM, Dhaun N, Goddard J, Webb DJ.
Sitaxsentan sodium for pulmonary hypertension. Drugs
Today (Barc) 2008;44:585600.
131. Macintyre IM, Dhaun N, Goddard J, Webb DJ.
Ambrisentan and its role in the management of
172 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
pulmonary arterial hypertension. Drugs Today (Barc)
2008;44:875885.
132. Hrometz SL, Shields KM. Role of ambrisentan in the
management of pulmonary hypertension. Ann
Pharmacother 2008;42:16531659. Epub 2008 Oct 28.
133. Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S,
Savai R, Roth M, Sydykov A, Lai YJ, Weissmann N,
Seeger W, et al. Reversal of experimental pulmonary
hypertension by PDGF inhibition. J Clin Invest
2005;115:28112821.
134. Ghofrani HA, Seeger W, Grimminger F. Imatinib for the
treatment of pulmonary arterial hypertension. N Engl J
Med 2005;353:14121413.
135. Patterson KC, Weissmann A, Ahmadi T, Farber HW.
Imatinib mesylate in the treatment of refractory
idiopathic pulmonary arterial hypertension. Ann Intern
Med 2006;145:152153.
136. Souza R, Sitbon O, Parent F, Simonneau G, Humbert M.
Long termimatinib treatment in pulmonary arterial
hypertension. Thorax 2006;61:736.
137. Chu TF, Rupnick MA, Kerkela R, Dallabrida SM,
Zurakowski D, Nguyen L, Woulfe K, Pravda E, Cassiola
F, Desai J, et al. Cardiotoxicity associated with tyrosine
kinase inhibitor sunitinib. Lancet 2007;370:
20112019.
138. Atallah E, Durand JB, Kantarjian H, Cortes J. Congestive
heart failureis a rare event in patients receiving imatinib
therapy. Blood 2007;110:12331237.
139. Ghofrani HA, Seeger W, Grimminger F. Imatinib for the
treatment of pulmonary arterial hypertension. N Engl J
Med 2005;353:14121413.
140. Yurtseven N, Karaca P, Kaplan M, Ozkul V, Tuygun AK,
Aksoy T, Canik S, Kopman E. Effect of nitroglycerin
inhalation on patients with pulmonary hypertension
undergoing mitral valve replacement surgery.
Anesthesiology 2003;99:855858.
141. Fullerton DA, Jaggers J, Jones SD, Brown JM, McIntyre
RC Jr. Adenosine for refractory pulmonary
hypertension. Ann Thorac Surg 1996;62:874877.
142. Stobierska-Dzierzek B, Awad H, Michler RE. The
evolving management of acute right-sided heart failure
in cardiac transplant recipients. J Am Coll Cardiol
2001;38:923931.
143. Ng C, Franklin O, Vaidya M, Pierce C, Petros A.
Adenosine infusion for the management of persistent
pulmonary hypertension of the newborn. Pediatr Crit
Care Med 2004;5:1013.
144. Motti A, Tissot C, Rimensberger PC, Prina-Rousso A,
Aggoun Y, Berner M, Beghetti M, da Cruz E.
Intravenous adenosine for refractory pulmonary
hypertension in a low-weight premature newborn: a
potential new drug for rescue therapy. Pediatr Crit Care
Med 2006;7:380382.
145. Petkov V, Mosgoeller W, Ziesche R, et al. Vasoactive
intestinal peptide as a new drug for treatment of primary
pulmonary hypertension. J Clin Invest 2003;111:
13391346.
146. Bradford KK, Deb B, Pearl RG. Combination therapy
with inhaled nitric oxide and intravenous dobutamine
during pulmonary hypertension in the rabbit.
J Cardiovasc Pharmacol 2000;36:146151.
147. Kerbaul F, Rondelet B, Motte S, Fesler P, Hubloue I,
Ewalenko P, Naeije R, Brimioulle S. Effects of
norepinephrine and dobutamine on pressure
load-induced right ventricular failure. Crit Care Med
2004;32:10351040.
148. Vizza CD, Rocca GD, Roma AD, Iacoboni C, Pierconti F,
Venuta F, Rendina E, Schmid G, Pietropaoli P, Fedele F.
Acute hemodynamic effects of inhaled nitric oxide,
dobutamine and a combination of the two in patients
with mild to moderate secondary pulmonary
hypertension. Crit Care 2001;5:355361.
149. Beale RJ, Hollenberg SM, Vincent JL, Parrillo JE.
Vasopressor and inotropic support in septic shock: an
evidence-based review. Crit Care Med 2004;32(11
Suppl):S455S465.
150. Kwak YL, Lee CS, Park YH, Hong YW. The effect of
phenylephrine and norepinephrine in patients with
chronic pulmonary hypertension. Anaesthesia
2002;57:914.
151. Hirsch LJ, Rooney MW, Wat SS, Kleinmann B, Mathru
M. Norepinephrine and phenylephrine effects on right
ventricular function in experimental canine pulmonary
embolism. Chest 1991;100:796801.
152. Brimioulle S, Vachi ery JL, Brichant JF, Delcroix M,
Lejeune P, Naeije R. Sympathetic modulation of hypoxic
pulmonary vasoconstriction in intact dogs. Cardiovasc Res
1997;34:384392.
153. Tisdale JE, Patel RV, Webb CR, Borzak S, Zarowitz BJ.
Proarrhythmic effects of intravenous vasopressors. Ann
Pharmacother 1995;29:269281.
154. Tritapepe L, Voci P, Cogliati AA, Pasotti E, Papalia U,
Menichetti A. Successful weaning from
cardiopulmonary bypass with central venous
prostaglandin E1 and left atrial norepinephrine infusion
in patients with acute pulmonary hypertension. Crit Care
Med 1999;27:21802183.
155. Meadow WL, Rudinsky BF, Strates E. Selective elevation
of systemic blood pressure by epinephrine during
sepsis-induced pulmonary hypertension in piglets.
Pediatr Res 1986;20:872875.
156. Holloway EL, Polumbo RA, Harrison DC. Acute
circulatory effects of dopamine in patients with
pulmonary hypertension. Br Heart J 1975;
37:482485.
157. Prielipp RC, McLean R, Rosenthal MH, Pearl RG.
Hemodynamic proles of prostaglandin E1,
isoproterenol, prostacyclin, and nifedipine in
experimental porcine pulmonary hypertension. Crit Care
Med 1991;19:6067.
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 173
The Management of Acute Pulmonary Hypertension G. Hui-li
158. Evora PR, Pearson PJ, Schaff HV. Arginine vasopressin
induces endothelium-dependent vasodilatation of the
pulmonary artery. V1-receptor-mediated production of
nitric oxide. Chest 1993;103:12411245.
159. Wallace AW, Tunin CM, Shoukas AA. Effects of
vasopressin on pulmonary and systemic vascular
mechanics. Am J Physiol 1989;257(4 Pt 2):H1228H1234.
160. Tayama E, Ueda T, Shojima T, Akasu K, Oda T,
Fukunaga S, Akashi H, Aoyagi S. Arginine vasopressin is
an ideal drug after cardiac surgery for the management
of low systemic vascular resistant hypotension
concomitant with pulmonary hypertension. Interact
Cardiovasc Thorac Surg 2007;6:715719.
161. Trempy GA, Nyhan DP, Murray PA. Pulmonary
vasoregulation by arginine vasopressin in conscious,
halothane-anesthetized, and pentobarbital-anesthetized
dogs with increased vasomotor tone. Anesthesiology
1994;81:632640.
162. Leather HA, Segers P, Berends N, Vandermeersch E,
Wouters PF. Effects of vasopressin on right ventricular
function in an experimental model of acute pulmonary
hypertension. Crit Care Med 2002;30:25482552.
163. Sandoval J, Gaspar J, Pulido T, Bautista E,
Martnez-Guerra ML, Zeballos M, Palomar A, G omez A.
Graded balloon dilation atrial septostomy in severe
primary pulmonary hypertension. J Am Coll Cardiol
1998;32:297304.
164. Reichenberger F, Pepke-Zaba J, McNeil K, Parameshwar
J, Shapiro LM. Atrial septostomy in the treatment of
severe pulmonary arterial hypertension. Thorax
2003;58:797800.
165. Sandoval J, Rothman A, Pulido T. Atrial septostomy for
pulmonary hypertension. Clin Chest Med
2001;22:547560.
166. Klepetko W, Mayer E, Sandoval J, Trulock EP, Vachiery
JL, Dartevelle P, Pepke-Zaba J, Jamieson SW, Lang I,
Corris P. Interventional and surgical modalities of
treatment for pulmonary arterial hypertension. J Am Coll
Cardiol 2004;43(12 Suppl S):73S80S.
167. Ozdogan ME, Erer D, Iriz E, Oktar GL, Kula S,
Buyukates M. Right-to-left shunt through a patent
foramen ovale left open in the management of acute
right heart failure after heart transplantation. J Heart
Lung Transplant 2008;27:135137.
168. Reynolds HR, Hochman JS. Cardiogenic shock: Current
concepts and improving outcomes. Circulation
2008;117:686697.
169. Palevsky HI. Therapeutic options for severe pulmonary
hypertension. Clin Chest Med 1997;18:595609.
170. Aubert S, Leprince P, Bonnet N, Barreda T, Ouattara A,
Varnous S, Pavie A, Gandjbakhch I. Limited mechanical
circulatory support following orthotopic heart
transplantation. Interact Cardiovasc Thorac Surg
2006;5:8889.
171. Arafa OE, Geiran OR, Andersen K, Fosse E, Simonsen S,
Svennevig JL. Intraaortic balloon pumping for
predominantly right ventricular failure after heart
transplantation. Ann Thorac Surg 2000;70:15871593.
172. Etz CD, Welp HA, Tjan TD, Hoffmeier A, Weigang E,
Scheld HH, Schmid C. Medically refractory pulmonary
hypertension: Treatment with nonpulsatile left
ventricular assist devices. Ann Thorac Surg
2007;83:16971705.
173. Martin J, Siegenthaler MP, Friesewinkel O, Fader T, van
de Loo A, Trummer G, Berchtold-Herz M, Beyersdorf F.
Implantable left ventricular assist device for treatment of
pulmonary hypertension in candidates for orthotopic
heart transplantation-a preliminary study. Eur J
Cardiothorac Surg 2004;25:971977.
174. Petrofski JA, Hoopes CW, Bashore TM, Russell SD,
Milano CA. Mechanical ventricular support lowers
pulmonary vascular resistance in a patient with
congential heart disease. Ann Thorac Surg
2003;75:10051007.
175. Zimpfer D, Zrunek P, Sandner S, Schima H, Grimm M,
Zuckermann A, Wolner E, Wieselthaler G.
Post-transplant survival after lowering xed pulmonary
hypertension using left ventricular assist devices. Eur J
Cardiothorac Surg 2007;31:698702. Epub 2007 Feb 7.
176. Liden H, Haraldsson A, Ricksten SE, Kjellman U,
Wiklund L. Does pretransplant left ventricular assist
device therapy improve results after heart
transplantation in patients with elevated pulmonary
vascular resistance? Eur J Cardiothorac Surg 2009. [Epub
ahead of print].
177. Pedemonte VO, Ar anguiz Santander E, Torres HH,
Merello NL, Vera PA, Daz NR, Kapl an MJ. Right
ventricular assistance with a centrifugal pump. Report of
two cases. Rev Med Chil 2008;136:359366. Epub 2008
Jun 3.
178. Damman K, Navis G, Smilde TD, Voors AA, Van Der Bij
W, van Veldhuisen DJ, Hillege HL. Decreased cardiac
output, venous congestion and the association with
renal impairment in patients with cardiac dysfunction.
Eur J Heart Fail 2007;9:872878. Epub 2007 Jun 22
179. Macchia A, Marchioli R, Marsi R, Scarano M, Levantesi
G, Tavazzi L, Tognoni G. A meta-analysis of trials of
pulmonary hypertension: A clinical condition looking
for drugs and research methodology. Am Heart J
2007;153:10371047.
180. Badesch DB, Abman SH, Simonneau G, Rubin LJ,
McLaughlin VV. Medical therapy for pulmonary arterial
hypertension: Updated ACCP evidence-based clinical
practice guidelines. Chest 2007;131:19171928.
181. Humbert M, Sitbon O, Simonneau G. Treatment of
pulmonary arterial hypertension. N Engl J Med
2004;351:14251436.
182. McLaughlin VV, Oudiz RJ, Frost A, Tapson VF, Murali S,
Channick RN, Badesch DB, Barst RJ, Hsu HH, Rubin LJ.
Randomized study ofadding inhaled iloprost to existing
174 Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd
G. Hui-li The Management of Acute Pulmonary Hypertension
bosentan in pulmonary arterial hypertension. Am J
Respir Crit Care Med 2006;174:12571263.
183. Damps-Konsta nska I, Konsta nski Z, Jassem E.
[Treatment of pulmonary hypertension]. Wiad Lek
2007;60:545549.
184. Lunze K, Gilbert N, Mebus S, et al. First experience with
an oral combination therapy using bosentan and
sildenal for pulmonary arterial hypertension. Eur J Clin
Invest 2006;36(Suppl):3238.
185. Gali ` e N, Rubin LJ, Hoeper M, Jansa P, Al-Hiti H, Meyer
G, Chiossi E, Kusic-Pajic A, Simonneau G. Treatment of
patients with mildly symptomatic pulmonary arterial
hypertension with bosentan: results of the EARLY study.
Lancet 2008;371:20932100.
186. Kuhlen R, Walbert E, Fr ankel P, Thaden S, Behrendt W,
Rossaint R. Combination of inhaled nitric oxide and
intravenous prostacyclin for successful treatment of
severe pulmonary hypertension in a patient with acute
respiratory distress syndrome. Intensive Care Med
1999;25:752754.
187. P erez-Pe nate GM, Juli a-Serd` a G, Ojeda-Betancort N,
Garca-Quintana A, Pulido-Duque J, Rodrguez-P erez A,
Cabrera-Navarro P, G omez-S anchez MA. Long-term
inhaled nitric oxide plus phosphodiesterase 5 inhibitors
for severe pulmonary hypertension. J Heart Lung
Transplant 2008;27:13261332. Epub 2008 Oct 26.
188. Voswinckel R, Reichenberger F, Enke B, Kreckel A,
Krick S, Gall H, Schermuly RT, Grimminger F, Rubin LJ,
Olschewski H, et al. Acute effects of the combination of
sildenal and inhaled treprostinil on haemodynamics
and gas exchange in pulmonary hypertension.
Pulm Pharmacol Ther 2008;21:824832. Epub 2008
Jul 9.
189. Lobato EB, Beaver T, Muehlschlegel J, Kirby DS, Klodell
C, Sidi A. Treatment with phosphodiesterase inhibitors
type III and V: Milrinone and sildenal is an effective
combination during thromboxane-induced acute
pulmonary hypertension. Br J Anaesth 2006;96:317322.
Epub 2006 Jan 27.
190. Hoeper MM, Markevych I, Spiekerkotter E, Welte T,
Niedermeyer J. Goal-oriented treatment and
combination therapy for pulmonary arterial
hypertension. Eur Respir J 2005;26:858863.
191. De Santo LS, Mastroianni C, Romano G, Amarelli C,
Marra C, Maiello C, Galdieri N, Della Corte A, Cotrufo
M, Caianiello G. Role of sildenal in acute
posttransplant right ventricular dysfunction: Successful
experience in 13 consecutive patients. Transplant Proc
2008;40:20152018.
192. Benedict N, Seybert A, Mathier MA. Evidence-based
pharmacologic management of pulmonary arterial
hypertension. Clin Ther 2007;29:21342153.
Cardiovascular Therapeutics 29 (2011) 153175 c 2010 Blackwell Publishing Ltd 175

You might also like