today’s and tomorrow’s imaging and circulating biomarkers ... · 2) pulmonary hypertension with...

31
REVIEW Today’s and tomorrow’s imaging and circulating biomarkers for pulmonary arterial hypertension Marjorie Barrier Jolyane Meloche Maria Helena Jacob Audrey Courboulin Steeve Provencher Se ´bastien Bonnet Received: 17 October 2011 / Revised: 18 February 2012 / Accepted: 20 February 2012 / Published online: 25 March 2012 Ó Springer Basel AG 2012 Abstract The pathobiology of pulmonary arterial hypertension (PAH) involves a remodeling process in distal pulmonary arteries, as well as vasoconstriction and in situ thrombosis, leading to an increase in pulmonary vascular resistance, right heart failure and death. Its etiology may be idiopathic, but PAH is also frequently associated with underlying conditions such as connective tissue diseases. During the past decade, more than welcome novel therapies have been developed and are in development, including those increasingly targeting the remodeling process. These therapeutic options modestly increase the patients’ long- term survival, now approaching 60% at 5 years. However, non-invasive tools for confirming PAH diagnosis, and assessing disease severity and response to therapy, are tragically lacking and would help to select the best treat- ment. After exclusion of other causes of pulmonary hypertension, a final diagnosis still relies on right heart catheterization, an invasive technique which cannot be repeated as often as an optimal follow-up might require. Similarly, other techniques and biomarkers used for assessing disease severity and response to treatment gen- erally lack specificity and have significant limitations. In this review, imaging as well as current and future circu- lating biomarkers for diagnosis, prognosis, and follow-up are discussed. Keywords Pulmonary arterial hypertension Biomarkers Imaging Circulating biomarkers Abbreviations 5-HT Serotonin or 5-hydroxytryptamine BMP Bone morphogenetic protein BMPRII BMP receptor 2 BNP Brain natriuretic peptide cGMP Cyclic guanosine 3 0 5 0 -monophosphate CT Computed tomography CTEPH Chronic thromboembolic pulmonary hypertension DCE Delayed contrast enhancement Echo Echocardiography EGF Epidermal growth factor eNOS Endothelial nitric oxide synthase ET-1/3 Endothelin 1 or 3 FC Functional class HDL-C High-density lipoprotein cholesterol IL-6 Interleukin 6 iPAH Idiopathic PAH LIGHT Lymphotoxin-like inducible protein LV Left ventricle miR-X MicroRNA-X miRNA Micro RNA MP Microparticles MRI Magnetic resonance imaging NFAT Nuclear factor of activated T cells NO Nitric oxide NT-proBNP N-terminal pro-brain natriuretic peptide OPN Osteopontin PA Pulmonary arteries PAAT Pulmonary artery acceleration time PAH Pulmonary arterial hypertension M. Barrier J. Meloche M. H. Jacob A. Courboulin S. Provencher S. Bonnet (&) Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Que ´bec, 2725 Chemin Ste-Foy, Que ´bec, QC G1V 4G5, Canada e-mail: [email protected] Cell. Mol. Life Sci. (2012) 69:2805–2831 DOI 10.1007/s00018-012-0950-4 Cellular and Molecular Life Sciences 123

Upload: others

Post on 18-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

REVIEW

Today’s and tomorrow’s imaging and circulating biomarkersfor pulmonary arterial hypertension

Marjorie Barrier • Jolyane Meloche • Maria Helena Jacob •

Audrey Courboulin • Steeve Provencher • Sebastien Bonnet

Received: 17 October 2011 / Revised: 18 February 2012 / Accepted: 20 February 2012 / Published online: 25 March 2012

� Springer Basel AG 2012

Abstract The pathobiology of pulmonary arterial

hypertension (PAH) involves a remodeling process in distal

pulmonary arteries, as well as vasoconstriction and in situ

thrombosis, leading to an increase in pulmonary vascular

resistance, right heart failure and death. Its etiology may be

idiopathic, but PAH is also frequently associated with

underlying conditions such as connective tissue diseases.

During the past decade, more than welcome novel therapies

have been developed and are in development, including

those increasingly targeting the remodeling process. These

therapeutic options modestly increase the patients’ long-

term survival, now approaching 60% at 5 years. However,

non-invasive tools for confirming PAH diagnosis, and

assessing disease severity and response to therapy, are

tragically lacking and would help to select the best treat-

ment. After exclusion of other causes of pulmonary

hypertension, a final diagnosis still relies on right heart

catheterization, an invasive technique which cannot be

repeated as often as an optimal follow-up might require.

Similarly, other techniques and biomarkers used for

assessing disease severity and response to treatment gen-

erally lack specificity and have significant limitations. In

this review, imaging as well as current and future circu-

lating biomarkers for diagnosis, prognosis, and follow-up

are discussed.

Keywords Pulmonary arterial hypertension �Biomarkers � Imaging � Circulating biomarkers

Abbreviations

5-HT Serotonin or 5-hydroxytryptamine

BMP Bone morphogenetic protein

BMPRII BMP receptor 2

BNP Brain natriuretic peptide

cGMP Cyclic guanosine 3050-monophosphate

CT Computed tomography

CTEPH Chronic thromboembolic pulmonary

hypertension

DCE Delayed contrast enhancement

Echo Echocardiography

EGF Epidermal growth factor

eNOS Endothelial nitric oxide synthase

ET-1/3 Endothelin 1 or 3

FC Functional class

HDL-C High-density lipoprotein cholesterol

IL-6 Interleukin 6

iPAH Idiopathic PAH

LIGHT Lymphotoxin-like inducible protein

LV Left ventricle

miR-X MicroRNA-X

miRNA Micro RNA

MP Microparticles

MRI Magnetic resonance imaging

NFAT Nuclear factor of activated T cells

NO Nitric oxide

NT-proBNP N-terminal pro-brain natriuretic peptide

OPN Osteopontin

PA Pulmonary arteries

PAAT Pulmonary artery acceleration time

PAH Pulmonary arterial hypertension

M. Barrier � J. Meloche � M. H. Jacob � A. Courboulin �S. Provencher � S. Bonnet (&)

Pulmonary Hypertension Research Group, Centre de Recherche

de l’Institut Universitaire de Cardiologie et de Pneumologie de

Quebec, 2725 Chemin Ste-Foy, Quebec, QC G1V 4G5, Canada

e-mail: [email protected]

Cell. Mol. Life Sci. (2012) 69:2805–2831

DOI 10.1007/s00018-012-0950-4 Cellular and Molecular Life Sciences

123

Page 2: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

PAP Pulmonary arterial pressure

PASMC Pulmonary artery smooth muscle cell

PDE5 Phosphodiesterase 5

PDGF Platelet-derived growth factor

PECAM Platelet endithelial cell adhesion molecule

PET Position emission tomography

PH Pulmonary hypertension

Pim1 Proviral integration site for Moloney murine

leukemia 1

PPAR Peroxisome proliferator activated receptor

(gamma c or alpha a)

Q Pefusion

RDW Red blood cell distribution width

RHC Right heart catheterization

ROS Reactive oxygen species

RV Right ventricle

RVH Right ventricle hypertrophy

SPECT Single photon emission computed

tomography

SOD Superoxyde dismutase

STAT3 Signal transducer and activator of

transcription 3

TAPSE Tricuspid annular plane systolic excursion

TGFb Tranforming growth factor beta

TXA2 Thromboxane A2

V Ventilation

vWF Plasma von Willebrand factor

WHO World Health Organization

Introduction

Pulmonary hypertension (PH) includes several pathologies.

A new classification of PH designated five distinct cate-

gories differing in their pathophysiology, clinical

presentation, diagnostic findings, and response to treatment

(Table 1) [1]. Amongst these, pulmonary arterial hyper-

tension (PAH) is characterized by pulmonary vascular

remodeling featured by vasoconstriction, remodeling of the

small pulmonary artery (PA) wall, and in situ thrombosis

[2]. PAH is clinically characterized by progressively

increasing pulmonary vascular resistance eventually lead-

ing to right ventricle (RV) hypertrophy (RVH), failure and

death [3, 4]. PAH may be idiopathic or associated with

various conditions (Table 1) [5].

Biomarkers are obviously an essential missing element

for diagnosing PAH [1, 6]. In fact, the average delay before

diagnosing PAH patients is 2.8 years, a period during

which the disease often worsens [7]. The long current delay

is explained by the lack of specificity of PAH symptoms. In

fact, dyspnea is the cardinal symptom of PAH and is

generally attributed to other pulmonary diseases such as

asthma and chronic obstructive pulmonary disease, or to

depression and anxiety disorders, before being associated with

PAH. Moreover, the prevalence of PAH is estimated at 60 per

million, which makes it rare and even more difficult to diag-

nose, since some physicians are not familiar with PAH [8].

Thus, new diagnostic tools should ideally reduce this delay in

order to treat this pathology at its earliest stages. Indeed, PAH

is diagnosed by an exclusion principle to strike out the other

PH groups. Among the PAH group (group 1), iPAH is a

diagnostic of exclusion since no known cause is related to the

disease, as it is idiopathic (Fig. 1).

Fortunately, major improvements have been made in the

treatment of PAH in the past decade. Despite these novel

therapies, most patients still display persistent exercise

intolerance, and the long-term prognosis remains poor [4,

9–12]. A goal-oriented therapy has been proposed to

improve long-term outcomes of PAH patients [13].

Table 1 Classification of pulmonary hypertension (PH)

1) Pulmonary arterial hypertension (PAH)

Idiopathic (iPAH)

Heritable

Related with collagen vascular disease (e.g., systemic sclerosis),

congenital left-to-right shunt, HIV infection, use of drugs or toxins,

portal hypertension or other causes (e.g., scleroderma)

Pulmonary hypertension of the newborn

Pulmonary veino-occlusive disease

Pulmonary capillary hemangiomatosis

2) Pulmonary hypertension with left-sided heart disease

Systolic dysfunction

Diastolic dysfunction

Valvular disease

3) Pulmonary hypertension associated with lung diseases and/or

hypoxemia

Chronic obstructive pulmonary disease (COPD)

Interstitial lung disease

Sleep-disordered breathing, alveolar hypoventilation

Chronic exposure to high altitude

Developmental lung abnormalities

4) Pulmonary hypertension due to chronic thrombotic and/or embolic

disease

Chronic thromboembolic pulmonary hypertension (CTEPH) in

proximal or distal pulmonary arteries

Embolization of other matter, such as tumor, parasites or foreign

material

5) Pulmonary hypertension with unclear multifactorial mechanisms

Hematologic disorders (i.e. myeloproliferative disorders,

splenectomy)

Systemic disorder (i.e. sarcoidosis)

Metabolic disorder (i.e. glycogen storage disease)

Other causes like compression of pulmonary vessels, chronic renal

failure

HIV Human immunodeficiency virus

2806 M. Barrier et al.

123

Page 3: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

However, this proposal has emphasized the lack of a con-

sistent protocol for evaluating the course of the disease.

Indeed, establishing such goals is hampered by the gap in

the evidence-based assessment of PAH treatment efficacy.

Only limited tools are currently available for predicting the

long-term outcome in PAH. An accurate biomarker should

guide the escalation of pharmacotherapy given the risk/

benefit ratio and the timing of referral for lung/heart

transplantation, i.e. the ultimate tool for saving patients

from RV failure. This involves the capacity to evaluate the

improvement or degradation of the patient’s condition. In

such circumstances, referral for life-saving interventions

such as lung transplantation would not be uselessly delayed

in the significant portion of patients who are unresponsive

to new therapies [14].

Current understanding of PAH pathophysiology

Pulmonary arterial hypertension is a vascular disease that is

largely restricted to small PA. Many abnormalities con-

tribute to this syndrome of obstructed, constricted small

PA. This includes abnormalities in blood concentration of a

few growth factors, neurotransmitters and cytokines,

namely increases in platelet-derived growth factor (PDGF),

serotonin, interleukin-6 (IL-6), and endothelin-1 (ET-1).

Nonetheless, these factors may not be reliable biomarkers

since they are increased by stress, inflammation, and other

common assaults on body integrity, and therefore, may not

be specific to PAH. The media in PAH also exhibits an

increased activation of the NFAT (nuclear factor of acti-

vated T cells) transcription factor, leading to increased

Ca2?-dependent PA smooth muscle cell (PASMC) prolif-

eration and decreased mitochondrial-dependent apoptosis

[15]. There is also a metabolic disorder due in part to an

impairment of mitochondrial metabolism known as the

Warburg effect, which is defined as a decrease in glucose

oxidation and an increase in glycolysis [16]. This phe-

nomenon is well described in cancer, and mounting

evidence supports the fact that PAH also displays a War-

burg effect [16]. Finally, the adventitia is infiltrated with

inflammatory cells and exhibits metalloprotease activation

[17]. Despite recent advances in the therapy of PAH,

mortality rates remain high (40% at 5 years) [18]. Over the

last 5 years, many investigators (including our laboratory)

Suspected pulmonary hypertension

Dyspnea on exertion, syncope, other predisposition to PH (family history, sleep apnea, …)

Echocardiography

Normal echocardiography with high clinical suspicion

Elevated right ventricle systolic pressure and RV hypertrophy

Imaging testing•Pulmonary function testing

•Chest radiography•Ventilation-perfusion scan

•CT-scan•Magnetic resonance imaging

Confirmation by right heart catheterization(mPAP1 > 25mmHg)

Pulmonary arterial hypertension

(group 1)

Pulmonary hypertension associated with another disease

(group 2, 3, 4 or 5)

Serologic testing•Complete blood count

•Human immunodeficiencyvirus testing

•Circulating biomarker evaluation (BNP, troponin T, uric acid)

Other•Liver function test•Six-minute walked

distance test•Sleep study may be

considered

No evidence of underlying disease of

pulmonary hypertension

Evidence of underlying disease of pulmonary

hypertension

Confirmation by right heart catheterization(mPAP1 > 25mmHg)

Fig. 1 Pulmonary hypertension

diagnostic algorithm. This

represents the usual algorithm

followed by patients with

suspected pulmonary

hypertension to diagnose the

class of pulmonary hypertension

in order to better adapt the

treatment

Today’s and tomorrow’s imaging and circulating biomarkers 2807

123

Page 4: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

have endeavored to understand the mechanisms for

enhanced PASMC proliferation and decreased apoptosis,

which account for distal PA remodeling. Indeed, the lung

peripheral vascular bed is increasingly the target of novel

treatment strategies, which aim at restoring physiological

vascular tone and at reducing the remodeling process [19].

Many pathways are implicated in PAH development since

this pathology is a combination of many factors such as

PASMC deregulation (enhanced proliferation and migra-

tion, resistance to apoptosis), abnormal endothelial cells

(causing endothelial barrier disruption and plexiform

lesions), and inflammation as well as increased elastolytic

activity. Many factors have been shown to be implicated in

PAH pathogenesis. Among others, RhoA/ROCK, BMPRII

(bone morphgenetic protein receptor II) dysfunction,

STAT3 (signal transducer and activator of transcription-3),

and NFATc2 upregulation seem to play a critical role in

PASMC proliferation. The issue is that neither STAT3, an

enhancer of NFAT expression, nor NFAT itself can be

targeted, because of their critical function in a plethora of

physiological processes, such as the immune response.

More recently, our group has demonstrated that NFAT

expression is STAT3-dependent while its activation relies

on the oncogene protein Pim1 (proviral integration site for

Moloney murine leukemia) [20]. Inflammatory factors such

as TGF-b, IL-6, and BMP can stimulate PASMC prolif-

eration [21]. Modification in BMPRII levels can trigger a

large amount of signaling pathways. For example, tran-

scription factor PPARc (peroxisome proliferator activated

receptor gamma) is decreased by BMPRII dysfunction and

Rabinovich et al. remarkably showed that PPARc is

implicated in PASMC proliferation and migration, but also

in endothelial cells dysfunction contributing to PAH

pathobiology [22–24]. MicroRNA (miRNA) are also

implicated in the sustainability of the disease, Courboulin

et al. have indeed identified miR-204 as implicated in PAH.

Other microRNA, such as miR-21, miR-34c, and miR-133,

are implicated in right ventricular failure and, thus, may

also play a role in PAH [25, 26]. These factors, proteins,

and pathways are not yet used as biomarkers nor thera-

peutic targets, but this shows that the past decade brought

knowledge in understanding PAH in a way to potentially

treat it.

In this review, currently available biomarkers will be

presented together with their limitations and more recent

improvements.

Primary tests

The first signs of PAH are observed by physical exami-

nation, including an increase in jugular venous distension,

a holosystolic murmur of tricuspid valve regurgitation and

a loud P2. Hepatomegaly, peripheral edema, ascites, and

cool extremities are eventually observed when RV fail-

ure is more severe. Electrocardiography is usually

performed at this stage, highlighting a right axis devia-

tion, RV hypertrophy, and peaked P-waves. Moreover,

nocturnal hypoxemia occurs in [75% of PAH patients,

independently of the occurrence of apneas or hypopneas

[27]. Isolated alteration in diffusing lung capacity for

carbon monoxide is traditionally seen on pulmonary

function tests, although pulmonary function may be

normal or exhibits mild obstructive/restive disorder in

PAH patients [28, 29]. When cardiopulmonary exercise

testing is performed, PAH patients generally display

altered cardiac response and gas exchange abnormalities

[30]. However, these abnormalities are generally seen

when right heart failure is already present and lack

specificity.

New York Heart Association/World Health

Organization functional class

The New York Heart Association and World Health

Organization (WHO) functional classes (FC) are based on

symptoms of dyspnea with degree of exercise intolerance

[31], and help to classify PAH severity. PAH severity is

most commonly assessed objectively with the use of WHO

functional class combined with exercise capacity and RV

function. Various studies have shown that FC and exercise

capacity as assessed by the cardiopulmonary exercise test

[32–34] or the 6-min walk test [34–37] have good dis-

criminating properties and predict survival in PAH [35].

However, caution must be exerted when using the func-

tional classification as an endpoint since the assignment of

patients to different classes is subject to investigator and

patient bias, even if they have held up in multivariate

analysis [31, 35, 38, 39].

Right heart catheterization (RHC)

Right heart catheterization is the gold standard for the

diagnosis of PH. It is to date the only direct proof of PH.

PH is defined as a mean pulmonary artery pressure (PAP)

at rest [25 mmHg (Fig. 1). The indication for RHC relies

on the estimated systolic PAP on echocardiography, the

presence of additional echocardiographic findings sugges-

tive of PH and the presence of symptoms or risks factors

for PH [40]. Crucial information such as PAP (systolic,

diastolic and mean) values, right arterial and ventricular

pressures, PA wedge pressure, pulmonary and systemic

vascular resistance, cardiac output and cardiac index and

mixed venous oxygen saturation can all be measured by

2808 M. Barrier et al.

123

Page 5: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

catheterization [41]. RHC is essential for therapy selec-

tion since it provides important data on pulmonary

circulation vasoreactivity [41], and provides important

information about disease severity and response to ther-

apy. Nonetheless, the technique is invasive, although the

incidence of complications in experienced centers is small

[42–44].

Imaging

Imaging represents a powerful tool for assessing lung

peripheral vascular bed, as well as heart function and

morphology. Indeed, although PAH patients suffer from a

lung disease, functional capacity and prognosis is inti-

mately related to heart function [45]. Thus, it is not only

critical to be able to observe improvements at the lung

level but also to visualize and analyze RV functions.

Actually, the complex and unique shape and the contractile

pattern of the RV make it more difficult to assess [46].

Different imaging techniques are now available to assess

lungs as well as the RV. The development of new non-

invasive biomarkers would be of great interest at diagnosis,

but also for follow-up.

Echocardiography

Echocardiography is a key screening tool in the diagnostic

algorithm of PAH (Fig. 1). In comparison with invasive

measurements, it has the advantages of being safe, easily

and rapidly performed, portable, widely available and thus

well suited for screening, despite the fact that it is con-

sidered performer-dependent [47]. After PH is diagnosed

by RHC, disease monitoring by echocardiography is done

every 3–6 months.

Baseline measurements

Doppler echo is performed on patients with suspected PAH

to assess several parameters such as heart function and

shape, as well as pulmonary and cardiac hemodynamics.

Actually, echo allows the assessment of right and left

heart chamber sizes, and PAH patients display an enlarged

right atrial and RV size and RVH, instead of a crescent-

shaped RV. Moreover, it is also possible to see pericardial

effusions and diastolic septal shift [48], which have prog-

nostic values for PAH patients [48, 49]. Echo also allows

exclusion of other types of PH, including valvular dys-

function, congestive heart failure and congenital heart

disease.

Furthermore, RVSP can be estimated by tricuspid

regurgitation using Doppler, right arterial pressure and

pulmonary artery acceleration time (PAAT). PAAT is

decreased in PAH patients and known to correlate with

disease severity. mPAP can also be approximated using

Doppler function, with a correlation varying from 0.57 to

0.93 compared to PAP measured by RHC. PAP measured

by echo also allows the calculation of other echo-derived

parameters (e.g., aorta diameter, pulmonary velocity, heart

rate), and the calculation of PVR, which is well correlated

to disease severity [50]. LV eccentricity index has also

been shown to have some predictive value on PAH patients

outcome [8, 48].

Some findings have also highlighted the prognostic

values of the Tei-Index and the tricuspid annular plane

systolic excursion (TAPSE) [51]. The Tei-Index, already

used to evaluate the performance of the myocardium, has

proved useful in the evaluation of ventricular function.

Tricuspid annular displacement has also shown good

results in the prognosis of PH patients [52]. Furthermore,

systolic displacement of the tricuspid annulus towards the

RV apex is referred as TAPSE, correlates with RV ejection

fraction, and powerfully reflects RV function. A decreased

TAPSE portends a poor prognosis in patients with dilated

cardiomyopathy [53–55], and Forfia et al. [56] showed that

there is also a correlation between TAPSE and PAH

prognosis (survival) in PAH patients. The latter authors

have also demonstrated a good correlation between TAPSE

values and mPAP, but an even better one with PVR. Fur-

thermore, TAPSE is highly reproducible, unlike mPAP

estimated by echocardiography. Nonetheless, further stud-

ies have to be performed to confirm these results.

Follow-up values

Echocardiography is also a good method to monitor ther-

apeutic efficiency on RV function as it is a critical element

of PAH prognosis and since it can be visualized on echo.

Indeed, echo is also used to assess the efficiency of treat-

ment as a follow-up technique. PAH deterioration is

accelerated in PAH patients with RV dysfunction [4, 57].

Indeed, this imaging technique assesses parameters such as

ventricular and septal morphologies, and maximal velocity

of tricuspid regurgitation jets. All these parameters are

significantly different between the intravenous epoprost-

enol (a prostacyclin analogous) and control groups [58].

Moreover, in another randomized controlled trial, a sig-

nificant difference in changes in ventricular morphology,

i.e. the minimum diameter of the inferior vena cava and

Doppler measurement (RV ejection time and mitral valve

peak velocity) in the patients taking bosentan (a endothelin

receptor antagonist) was observed [8, 58], indicating that

echo can be used as a follow-up technique.

Echo also represents a useful tool in the early diagnosis

of PH. Indeed, first-degree relatives of PAH patients should

undergo routine echo every 3–5 years to ensure that no

Today’s and tomorrow’s imaging and circulating biomarkers 2809

123

Page 6: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

hereditary PAH is developing, or if symptoms manifest

themselves in order to get a diagnosis as quickly as pos-

sible if they develop PAH [41, 43, 59]. Echo is also useful

in screening other at-risk population including scleroderma

and HIV-positive patients [41, 60, 61].

Limitations and future developments

The main issue with this technique is its poor inter-obser-

ver agreement, and, due to its 2-D assessment, heart shape

based on certain assumptions. Moreover, the ultrasound

window can be limited not only because of high body mass

index (BMI), lung fibrosis, or emphysema but also due to

the sternum position of the patient [52]. Like other imaging

techniques [magnetic resonance imaging (MRI), computed

tomography (CT) scan], echo is constantly improving and

has been shown to be accurate in assessing RV function

[62]. Nonetheless, despite all its disadvantages, echo is

likely to remain the most prominent tool for screening PAH

and for follow-up, at least for the time being. This is

explained by its availability and, despite its lack of preci-

sion, this imaging technique has nonetheless amply proven

its reliability for screening. Furthermore, this imaging tool

undergoes technical improvement.

The RV has a very complicated shape, which is hard to

assess by echo due to 2-D limitations. Some authors

believe that assessing LV compression yields more reliable

results than RVH [52]. The 3-D echocardiography is in

progress, which is of a great interest since it would

decrease the requirements for geometric assumptions and

provide more accurate estimations of LV and RV volumes

and ejection fractions [63, 64]. New improvements are also

expected concerning insights into RV and LV structure,

function and interdependence [8, 65].

Recently, a novel application for Doppler echocardiog-

raphy permits RV strain analysis, a very promising

development [66]. Indeed, in a recent study in 30 PAH

patients, Filusch et al. have shown that RV systolic strain

and strain rate were significantly altered and correlated

with levels of N-terminal pro-brain natriuretic peptide (NT-

proBNP, a serum biomarker) and reduced 6MWD com-

pared to matched control. Moreover, reduced strains also

correlated with both mPAP and PVR in these PAH patients

[51]. Thus, detection of the strains is able to discriminate

between the different WHO FC and predict in which class

patients will fall. Results from RV strain analysis are also

related to invasive pulmonary hemodynamic parameters

such as PVR, mPAP and cardiac outpout, emphasizing its

prognostic value [47, 67].

Unfortunately, some of the parameters discussed above

may sometimes show lack of specificity. This explains why

RHC must be made on patient with normal echo-derived

RVSP but with high suspicion of PAH, to ensure whether

or not this patient suffers PAH. Indeed, RHC remains the

only diagnostic measure for PAH, and is usually used to

confirm the presence of PAH. Nonetheless, echo is a pre-

cious tool in screening patients with PAH-like symptoms,

to at least exclude pulmonary pathologies, if not being a

PAH diagnosis tool itself.

Radionuclide imagery

This type of imaging technique includes single photon

emission computed tomography (SPECT), positron emis-

sion tomography (PET), scintigraphy, and magnetic

resonance spectroscopy imaging [67]. A few decades ago,

before the emergence of CT and MRI, radionuclide

imaging was commonly used [47]. The presence of per-

fusion (Q) abnormalities using perfusion scintigraphy has

already been shown in late PAH. However, in addition to

impairment in perfusion, there is also a concomitant

impairment in ventilation (V) as seen by planar scintigra-

phy [68].

Using SPECT, which is more sensitive than planar

scintigraphy for assessing focal V–Q disturbances, the V/Q

SPECT-derived ratio can be measured and provides topo-

graphic V–Q distribution and an objective quantification of

cross-sectional lung V–Q imbalance with a correlation of

lung morphology on CT. Some groups have been able to

find V impairment resulting in V–Q mismatch in patients

with mild PAH. As indicated by the significant correlation

observed between the standard deviation of the entire lung

V/Q ratios and the mean PAP in these patients, the lung

pathophysiology causing V–Q imbalance may be more

advanced, as shown by persistently elevated PAP in iPAH

[68]. However, the main indication for lung V/Q scan

remains to date the exclusion of chronic thromboembolism

as the cause of PH [69].

Using PET scan, Wong et al. demonstrated that RV

myocardial blood flow is increased in iPAH, whereas high

O2 extraction fraction was associated with poorer NYHA

class and more severe RV failure. This may explain why an

additional demand to the heart may lead to cardiac ische-

mia and RV failure [70].

Another interesting aspect is the Warburg effect since it

is a hallmark of both cancer and PAH [16]. This patho-

logical phenomenon is characterized by an imbalance

between oxidative phosphorylation and glycolysis, pro-

moting glycolysis and lactate production. This also

involves increased glucose uptake compared to non-

pathological tissues. The PET scan is already well developed

and commonly used in cancer to assess the development of

a Warburg effect and response to treatment, by correlating

that effect with tumor severity and aggressiveness. Thus,

this technique may be thought as also being of some

prognostic value in PAH management. Indeed, using PET,

2810 M. Barrier et al.

123

Page 7: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

Abikhzer et al. [71] have reported that RV, which is usually

invisible in PET, could be imaged in a PAH patient due to

increased glucose uptake. This might represent a future

approach to assess RVH severity. Moreover, Marshboom

et al. have recently demonstrated that this technique may

also be possible as assessed the PAH lungs, as it also

displays Warburg effect [72].

New tools are also emerging, such as radioligand

binding of phosphodiesterase 5 (PDE5) [73, 74] and ET-1

[75], which are currently being evaluated in animal models.

Indeed, these receptors are already targeted in the patho-

physiology of PAH, and the hypothesis that their level is

related to the evolution of patients’ state is under scrutiny.

Thus, it might be interesting to verify the relevance of such

radioligands by quantifying the changes in the expression

of PDE5 and ET-1 as biomarkers in patients. These end-

points will obviously require further studies and will need

to be validated more extensively in PAH animal models

before going through human trials.

Magnetic resonance spectroscopy imaging has much

contributed to the diagnosis and prognosis of cancer

through the study of the metabolic changes occurring in

tumors. Thus, this technique might also bring a contribu-

tion to the field of PAH. Moreover, this imaging tool

supplements the use of MRI in cancer in order to facilitate

the diagnosis and to decide upon a proper therapeutic

strategy. Hence, it might well be possible to apply such a

combination of techniques to PAH [76].

CT scan

The gating system for the lung and heart (based on the elec-

trocardiogram gating system) has been a major improvement

in CT, allowing to overcome artifacts due to respiration and

heartbeat. This allowed the functional assessment of the heart

in addition to lung parameters. Indeed, it is believed that RV

remodeling follows PA remodeling. Thus, it might be of

crucial interest to assess PA remodeling before the effect can

be visualized in the right heart as a result of disease progres-

sion. Moreover, assessing lung remodeling would allow

assessing the response to treatment in distal PA where the

pathophysiological process actually takes place.

Baseline measurements

Some investigators have studied the main PA diameter in

relationship to mean PAP. Unfortunately, its correlation

with mean PAP varies from weak to strong even when

corrected for the size of the aorta, the thoracic vertebrae or

the arterio-bronchial segment in more than three lobes [77,

78], and for the BMI [79]. Moreover, a progressive dila-

tation of the main PA overtime is frequently discrepant

with PAH severity [80].

More importantly, images obtained by CT may allow

specialists to visualize characteristics that help in the dif-

ferential diagnosis of PAH. For example, the combination

of mosaic lung attenuations, a marked variation in the size

of segmental vessels, peripheral parenchymal opacities as

well as dilatation of bronchial and non-bronchial systemic

arteries is common in chronic thromboembolic pulmonary

hypertension (CTEPH) (73%) but is rare in PAH (14%),

which can provide a differential diagnosis between these

two pathologies [77]. Similarly, the CT scan allows the

exclusion of severe parenchymal abnormalities responsible

for PH (e.g., pulmonary fibrosis), and can identify other

possible causes of PH (congenital malformations, coronary

heart disease or cardiac dysfunction) [81]. Finally, it may

document pericardial effusions, which is directly related to

PAH severity [77].

More recently, data from electrocardiographically-gated

multidirectional CT studies have shown that functional

parameters such as right PA dispensability, systolic–dia-

stolic RV outflow tract dimensions, and diastolic wall

thickness are measurable with good inter-observer agree-

ment, and can reliably be used as criteria for PAH

diagnosis [77]. In a recent study, Revel et al. [82] found

that the most reliable parameter among all electro-

cardiographically-gated CT parameters evaluated for

identifying PAH patients is right PA wall distensibility.

This measure also correlates with mean PAP [71]. The

right ventricle/left ventricle ratio, using a gated or non-

gated multi-directed CT reflects RV function if acute pul-

monary embolism. Whether these results can be extended

to PAH patients remains unknown [83].

Devaraj et al. [78] have tested a composite index using

the CT-derived ratio of the diameter of the ascending aorta

and echo-derived RV systolic pressure. The combination of

these two parameters from two different imaging tech-

niques showed a relationship with mean PAP and was a

better predictor of PH than either measurement alone. That

index has been found to increase specificity to 100% with

respect to RHC-derived diagnosis compared to the criterion

of main PA diameter alone [84]. Indeed, a combination of

the (CT-derived main PA diameter)/(ascending aorta) ratio

and echo-derived RV systolic pressure has been found to

better correlate with RHC-derived mean PAP than either

single measurement [78]. A simultaneous use of parame-

ters derived from CT and echo also represents a more

informative combination since CT provides anatomical

data whereas echo identifies their functional implications.

Limitations and future developments

A major limitation of CT-scan is X-ray exposure, which is

problematic when multiple follow-up measures are neces-

sary. Nevertheless, major improvements are currently

Today’s and tomorrow’s imaging and circulating biomarkers 2811

123

Page 8: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

made to limit X-ray levels and the exposure time required

for a given scan quality [85]. Moreover, the contrast agents

used may exhibit significant nephrotoxicity and be aller-

genic in some patients (e.g., iodine). Claustrophobia is a

more mundane limitation that can be taken into account.

New investigations based on technological improve-

ments are constantly being undertaken to circumvent

limitations related to radionuclide imaging, the technique

replaced by CT-scan, but also related to CT-scan itself.

Dual-energy CT-perfusion imaging is a new technology

that decreases X-ray exposure with results comparable to

perfusion scintigraphy. That technique can provide CT

pulmonary angiograms, high-resolution morphologic ima-

ges, and spatially matched perfusion images in the very

same scan, with the same radiation dose that is sustained in

a classic CT pulmonary angiogram. Nevertheless, a few

technical issues remain to be addressed [86, 87]. Another

novel technique is area-detector CT, which will probably

soon be developed in the lung, although radiation exposure

remains significant. The CT-scan is important as it allows

the exclusion of underlying pathologies related to PH.

Furthermore, new types of image acquisition allow the

collecting of increasing amounts of data in a single scan-

ning session.

MRI

MRI has represented a major advance in the assessment of

lung and cardiac anatomy and function. Compared to echo,

MRI provides a higher spatial resolution and exhibits better

inter-observer reproducibility [88], and one of its major

advantages is that planes can be moved, and due to the 3-D

view, no approximation is required. Thus, cardiac MRI is

now considered as the gold standard for the detailed study

of the RV and is currently used as an endpoint in the

multinational European Union-funded Framework 6

EURO-MR project for PH, which will provide an impor-

tant opportunity to more definitely establish its utilization

as an endpoint [89]. Moreover, recent improvements of

hardware and software currently enables semi-automated

myocardial blood border definition, reducing intra- and

inter-observer variability in ventricular volume measure-

ments and decreasing the time spent on analysis [8, 52, 90–

95]. Finally, technical improvements of cine acquisition

have enabled the analysis of dynamic visualization of

several heartbeats (as cine images) with a significant

decrease in breath-holding [46], which is frequently prob-

lematic in PAH patients who usually complain of dyspnea.

Baseline measurements

Cardiac MRI is most commonly used to assess end-systolic

and end-diastolic RV and LV volumes and the resulting

RV ejection fraction, stroke volume and cardiac output.

Cardiac output can also be assessed by cine phase contrast

methods measuring volumetric flow in the main PA. Not

surprisingly, the RV end-diastolic and end-systolic vol-

umes have been shown to be significantly increased,

whereas stroke volume and cardiac output are decreased in

PAH compared to controls. Moreover, the RV ejection

fraction has been described as being impaired by *50% in

PAH patients [52]. MRI also allows the assessment of RV

hypertrophy, including (1) RV mass, which is increased

two- to threefold in PAH [52], (2) the Fulton ratio (RV

mass/LV mass with septum), which is increased by 80% in

PH patients, and (3) end-systolic RV wall thickness. As RV

failure progresses, LV end-diastolic volume and peak LV

filling rate are impaired in PAH patients and may be assessed

with cardiac MRI [52, 96]. In order to assess cardiac function

under stress conditions, some authors have proposed cardiac

MRI using dobutamine, a b1-adrenergic agonist [97], or

during exercise using a MR-compatible ergometer [98].

These experiments documented the incapacity of PAH

patients to increase stroke volume from rest to exercise.

Delayed contrast enhancement (DCE) has been found to

be frequent in PAH patients, especially when associated

with connective tissue disease [99]. DCE is mainly

observed at the RV intersection points and in intraven-

tricular septum area. Although DCE may be associated

with myocardial ischemia, it is more consistent with

myocardial fibrosis. The extent of DCE in the myocardium

has been shown to be inversely related to measures of RV

functions [100]. Finally, PAH patients have been shown to

display an interventricular asynchrony caused by a longer

RV systolic contraction using tagged MRI [46]. Cardiac

MRI is also of value to exclude other causes of PH such as

congenital heart disease of LV impairment (e.g., diastolic

dysfunction, constrictive pericarditis).

MRI can also be used to assess the pulmonary circula-

tion. MRI-measured proximal PA pulsatility, a marker of

PA stiffness predicts mortality in PAH patients [101].

However, the correlation between PA distensibility and

RHC-derived mean PAP remains controversial [101, 102].

Furthermore, 2-D MR angiograms have been reported to

differentiate CTEPH from PAH with a sensitivity of 92%

compared to a V/Q scan [103]. Similarly, Nikolaou et al.

[104] have been able to differentiate CTEPH from PAH

with an impressive accuracy of 90%, using contrast

enhanced perfusion MRI and MR angiograms with parallel

imaging techniques [104]. Moreover, PAH patients have

significantly decreased pulmonary blood flow and pro-

longed mean time transit in the whole lung, as documented

by 3-D dynamic contrast-enhanced perfusion MRI [104].

Nevertheless, CT angiograms are still believed to be

superior to MRI angiograms for visualizing vascular

abnormalities.

2812 M. Barrier et al.

123

Page 9: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

Some groups have also suggested MRI-guided cathe-

terization and real-time MRI-guided catheterization allow

the assessment of an impressive list of direct and calculated

parameters, namely the construction of RV pressure vol-

ume loops, RV afterload, myocardial contractility, pump

function, and RV–PA coupling, which are all useful to

assess PAH severity and response to therapy [46, 105].

Using both RHC and cardiac MRI, some have estimated

RV power. Indeed, RV mechanical efficiency is lower in

NYHA FC III than II, thus correlating with PAH severity.

Decreasing mechanical efficiency of the RV is a charac-

teristic of deterioration of RV functions in iPAH. This team

has also assessed glucose uptake by PET scan. However,

the reduction of RV mechanical efficiency is not due to a

metabolic shift to glucose oxidation, as assessed by PET-

scan [106].

Unfortunately, the relationship between MRI measures

and pulmonary hemodynamics is less straightforward.

Previous studies have suggested that PAP correlates with

RV mass [52], Fulton ratio [52], RV wall thickness, PAAT

[52], pulmonary blood flow transit time [107–109], and

septal curvature (r = 0.77). If leftward ventricular septal

bowing is observed, systolic PAP may be expected to be

[67 mmHg [46]. Among different MRI-derived parame-

ters, namely PA areas, PA stain, average velocity, peak

velocity, acceleration time, and ejection time, it has been

determined that average velocity best correlates with mean

PAP reference. This may imply that RHC might lose its

prominence in the future [46]. However, significant RV and

septal thickening (as seen in long-standing PAH), low left

ventricular systolic pressure may also influence the

importance of the septal curvature [52]. Similar correla-

tions have been suggested for pulmonary vascular

resistance [110]. However, these conclusions should be

considered with caution as these correlations were not

observed by other teams [111].

Followup values

The accuracy and reproducibility of cardiac MRI in

assessing cardiac functions and morphology makes it a

powerful tool for treatment follow-up [90]. Indeed, changes

in MRI-derived measurements are relevant in PAH. For

instance, a progressive dilation of RV, and a decrease in

LV systolic volume and RV stroke volume as detected at a

1-year follow-up by MRI have been correlated with a

worse prognosis in PAH [52, 96]. Changes in RV stroke

volume overtime also correlated with changes in exercise

capacity [112]. Changes in myocardial perfusion, RV mass,

and interventricular septal shift were also observed with

therapy [8, 46, 113, 114].

Limitations

The main limitation of MRI is imposed by the core phys-

ical principle of this technique, i.e. its incompatibility with

metal compounds such as the delivery pump for continuous

parenteral therapies commonly used in PAH [8]. Never-

theless, some teams have extended patients’ tubulure,

allowing the pump to be left outside the MRI room [8].

Moreover, MRI remains expensive, not widely available,

and time-consuming compared to CT scan [90]. MRI is

also problematic for claustrophobic patients (i.e. 2.3% of

patients subjected to MRI), although it can easily be cir-

cumvented by light sedation and the development of a

short-bore machine reducing the noise and the close-space

feeling [115, 116]. As for other imaging technics, body

movements, as well as respiratory and cardiac motions may

result in artifacts. Fortunately, technical improvements in

acquisition speed have reduced breath-holding and scan

times [52]. Finally, end-stage renal disease has been

associated with cases of nephrogenic systemic fibrosis

when gadolinium-containing contrast agents are used [90].

Due to its availability and ease of use, echo will probably

remain the tool of choice for first-tier screening and

detection, whereas MRI will be used subsequently to assess

lung and heart functions [52].

Conclusion

Imaging techniques are improving very fast, and, alone or

in combination, allow the measuring of an increasing

number of clinically relevant parameters both at baseline

and during follow-up. Although RHC is still the gold

standard to establish a diagnosis of PAH, novel imaging

tools are coming progressively closer to the same diag-

nostic quality. While some standardization of procedures is

required to reduce inter-observer and inter-site variability,

imaging techniques are very useful to support a diagnosis

of PAH diagnosis, to exclude other causes of PH, and to

objectively assess disease severity.

Circulating biomarkers

In parallel with imaging techniques, circulating biomarkers

are intensively explored by several groups. Ideally, blood

and urine biomarkers that (1) become abnormally elevated/

decreased in early or severe/end-stage disease (depending

on the biomarker), (2) are independent of indirect conse-

quences of PAH (e.g., renal and left ventricle function); and

(3) parallel the progression of the disease or the favorable

response to therapeutic interventions. Unfortunately, no such

Today’s and tomorrow’s imaging and circulating biomarkers 2813

123

Page 10: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

biomarker has yet been identified. Nonetheless, some of

those presented below show some interest and despite the

fact that no perfect biomarker is yet available, a combi-

nation of several biomarkers might be sufficiently accurate

for diagnostic or prognostic purposes.

Cardiac-derived biomarkers

Brain natriuretic peptides (BNP and NT-proBNP)

Plasma brain natriuretic peptide (BNP) and its biologically

inactive N-terminal fragment, NT-proBNP [117], are car-

diac hormones secreted by the cardiac myocytes and are

used as ventricular dysfunction biomarkers [118]. In PAH,

BNP and NT-proBNP, levels increase due to enhanced

synthesis by the RV and reflect the RV structure and

function [117, 119, 120]. Both forms appear to predict

survival in idiopathic PAH (iPAH) [117, 119, 121, 122] as

well as other types of PH [123, 124]. Furthermore, natri-

uretic peptide levels parallel changes in pulmonary

hemodynamics overtime [253]. However, natriuretic pep-

tides are not a specific PAH biomarker since they are

largely influenced by renal function, fluid retention and

diuretics, situations frequently encountered in PAH [125,

126].

Troponin T

Cardiac troponins are regulatory proteins that can be

detected by highly sensitive assays in peripheral blood after

their release, as a result of cardiac myocyte membrane

disruption. Torbicki et al. have evaluated the prognostic

value of that biochemical parameter in patients with severe

PH [127]. Detectable troponin T was associated with a

higher heart rate and NT-proBNP levels, a shorter 6-min

walk distance, and a poor prognosis. Indeed, 63% of

patients with detectable levels of cardiac troponin T died

during the 2-year follow-up. Although troponin T is a

marker of disease severity, and of prognostic value, it rises

only in end-stage disease [38, 127] and may be confounded

by left heart disease and renal impairment [128–130].

Osteopontin

Osteopontin (OPN), being a cytokine, is related to the

inflammatory process. Circulating OPN has been shown as

an independent predictor of survival in iPAH patients, and

correlates with severity [131]. Indeed, OPN is significantly

higher in iPAH patients compared to healthy subjects. In a

retrospective study, baseline level of OPN has been shown

to correlate with 6-min walk distance, mean PAP, and FC.

However, the origin of OPN production is still unclear. It is

hypothesized to be produced by the RV but this still has to

be confirmed [131]. Moreover, its biomarker value has to

be confirmed in larger studies.

Endothelium-derived biomarkers

cGMP

Cyclic guanosine 30,50-monophosphate (cGMP) is pro-

duced by the activation of guanylate cyclase and is

considered as an intracellular second messenger of natri-

uretic peptides, bradykinin and nitric oxide (NO) [132].

cGMP levels are significantly elevated in PAH patients’

urine compared to healthy controls, and inversely correlate

with the hemodynamic severity of PAH [133]. Wiedemann

et al. have measured cGMP levels in PAH and PH patients,

and, besides a marked increase in cGMP and ANP levels in

these patients, found that iloprost (a stable prostacyclin

analog) inhalation caused a decrease in cGMP in parallel

with pulmonary vasodilation and hemodynamic improve-

ment [134]. However, due to sizable interindividual

variations in cGMP levels, this biomarker has poor dis-

criminative properties [135].

Microparticles

Circulating microparticles (MP) are submicron membrane

fragments shed from activated or damaged vascular cells,

and released during apoptosis and/or activation of various

cell types [136]. Circulating endothelial MP have been

described as markers of endothelial injury as well as sys-

temic vascular remodeling [137]. Amabile et al. [138] have

shown that levels of circulating endothelial platelet endo-

thelial cell adhesion molecule (PECAM?), VE-cadherin?,

E-selectin?, and leukocyte-derived MP levels were

increased in PH patients compared to healthy individuals.

More specifically, levels of PECAM? and vascular endo-

thelium cadherin in PAH were significantly correlated with

the hemodynamic severity of PAH, suggesting that endo-

thelial MP might be reliable predictors of disease severity

in PH. Corroborating these results, Bakouboula et al. have

shown that MP containing active platelet tissue factor

(thrombokinase) and CD105 (endoglin) were increased in

patients with PAH compared to the control group [139].

Pro-coagulant MP were also linked to PH severity. They

hypothesized that circulating MP in the pulmonary vascu-

lar bed might also contribute to lung injury via diverse

pathways: impaired perfusion, vascular remodeling, leu-

kocyte recruitment, and inflammatory response. MP

containing active platelet tissue factor and CD105 may

possibly be considered as PAH early biomarkers in the near

future provided that other studies confirm the above find-

ings and further characterize these MP and their

involvement in PAH.

2814 M. Barrier et al.

123

Page 11: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

Given these encouraging results, research is still ongo-

ing on MP in animal models to better assess their role and

their potential as biomarkers. In order to characterize cir-

culating MP during hypoxic PAH and to study their effects

on endothelial function, the study of Tual-Chalot et al. has

provided evidence that hypoxic circulating MP induce

endothelial dysfunction in rat aorta and PA by decreasing

NO production [140]. Furthermore, they showed that MP

display tissue specificity.

Platelet-derived MP may influence vascular function via

their involvement in leukocyte adhesion, thrombus for-

mation and interaction with endothelium. The role of

platelet MP in thromboxane A2 (TXA2) production was

examined in rabbit PA [141]. TXA2 is well known to be

raised in atherosclerosis, which is a chronic vasculature

disease in which oxidative stress plays a key role [142].

That study provided evidence that platelet MP act as a

cellular source of TXA2 in rabbit aorta and PA. Yet, this

experimental approach demonstrated the ability of platelet

MP and endothelial cells to participate in the transcellular

conversion of arachidonic acid to the important vasocon-

strictor TXA2. Because TXA2 is an interesting mediator in

hypertension known to be deregulated in human PAH

[143], that work has provided an insight into the role of

platelet MP in vascular tone control.

Plasma von Willebrand factor

Plasma von Willebrand factor (vWF) is a large glycopro-

tein synthesized mainly in endothelial cells. The presence

of dysfunctional endothelial cells in PAH has been sug-

gested to be due to increased proteolysis and vWF release

[144]. Veyradier et al. demonstrated that baseline vWF

levels and vWF proteolysis were increased in ten patients

with severe PAH [144]. Moreover, these alterations were

reversible upon initiation of continuous epoprostenol

infusion, and paralleled the improvement of hemodynamic

measurements. More recently, Kawut and coworkers

showed that increased vWF levels at baseline and follow-

up were associated with worse survival in a cohort of 66

PAH patients [145].

D-dimer

D-dimer is a specific marker of cross-linked fibrin and is

associated with microvascular thrombosis, an important

component of PAH pathophysiology. D-dimer, as mea-

sured with an ELISA method, is increased in PAH patients

and correlates with patients’ functional capacity and

hemodynamic severity in iPAH [146, 147]. On the other

hand, such correlation was not found in PAH associated

with systemic sclerosis [148]. These data may suggest that

microvascular thrombosis might not play an important role

in the pathogenesis of PAH in patients with systemic

sclerosis, or that microvascular thrombosis occurs in the

systemic circulation independently of the pulmonary vas-

cular remodeling in these patients. Moreover, D-dimer

elevation occurs in diverse clinical situations, limiting its

accuracy in PAH.

Cancer-shared PAH biomarkers

Inflammation state

GDF-15 Growth differentiation factor (GDF)-15, which

belongs to the transforming growth factor (TGF)-super-

family of cytokines, was identified as an independent

predictor of long-term mortality in iPAH [149]. That bio-

marker displays a tight correlation with echo-measured RV

systolic pressure NT-proBNP plasma levels in systemic

sclerosis-associated PAH. These results suggest that GDF-

15 may have value as a biomarker, but larger studies on

PAH patients are obviously required at this stage [150,

151].

Endothelin-1/endothelin-3 Endothelin-1 (ET-1) is a pro-

liferative cytokine and a potent endogenous vasoconstrictor

with remodeling properties. Plasma ET-1 levels have been

shown to be increased in PH patients [152–155], and ET-1

protein as well as mRNA expression are enhanced in

endothelial cells of affected vessels [156]. More impor-

tantly, synthetic ET receptor antagonists (e.g., bosentan)

were shown to have beneficial effects in patients with

various forms of PAH [157–159].

Rubens et al. [153] have found that active ET-1 and its

precursor, big ET-1, correlate with disease severity and are

good prognostic markers for PAH patients. However, ET-1

measurement is not easily achieved and encounters tech-

nical difficulties such as the extremely short half-life of the

peptides (a few minutes) and their sensitivity to physio-

logical and pathological factors [160]. Endothelin-3 (ET-3)

is produced in many organs by various cells, including

endothelial cells. Montani et al. [161] have demonstrated

that the ET-1/ET-3 ratio is increased, whereas ET-3 plasma

concentrations are decreased in PAH. ET-1 and ET-3 levels

were correlated with hemodynamic and clinical markers of

disease severity, suggesting that the ET-1/ET-3 ratio might

be a novel prognostic factor in PAH.

LIGHT Otterdal et al. [162] investigated thrombus for-

mation and inflammation in PAH pathogenesis. They

demonstrated that lymphotoxin-like inducible protein that

competes with glycoprotein D for herpes virus entry

mediator on T lymphocytes (LIGHT) serum levels, a

platelet-derived ligand of the tumor necrosis factor super-

family, were increased in PAH compared to control

subjects. Moreover, LIGHT levels are significantly related

Today’s and tomorrow’s imaging and circulating biomarkers 2815

123

Page 12: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

to mortality in PAH patients. These findings suggest that,

since the prothrombotic effects of LIGHT in PAH involve

endothelium-related mechanisms, these effects might be part

of a common terminal pathway involved in PAH pathogene-

sis. However, further evidence is needed to confirm these

hypotheses, and larger trials must be carried out before

LIGHT can be considered as a reliable biomarker [162].

C-reactive protein C-reactive protein (CRP), a marker of

inflammation and tissue damage, has been demonstrated to

be an efficient outcome predictor in PAH. Indeed, its level

is significantly higher in PAH patients compared to control,

independently correlating with severity and predicting

mortality and clinical worsening. Moreover, normalization

of CRP levels with therapy was associated with improve-

ment in functional capacity, cardiac index and long-term

survival [163, 164].

Oncogenes/tumor suppressors

A growing number of cancer hallmarks turn out to be

present in PAH. Our recent review [165] reports the ther-

apeutic value of proto-oncogenes/tumor suppressor

proteins. These therapeutic targets might also have bio-

marker potential. Examples of the activation of oncogenic

pathways in PAH include the increase in cytokines and

growth factors such as epidermal growth factor (EGF),

PDGF, cytokines (e.g., IL-6) [166], or peptide agonists

such as ET-1 [167–170]. These factors can activate bio-

logical pathways such as proliferation, migration, survival

and metabolism switches. Some cytokines are already

considered as cancer biomarkers [171–174]. One might

surmise that a cytokine such as IL-6 may also have value as

a PAH biomarker. The main issue to be addressed to assess

that hypothesis is the lack of specificity of many of these

factors. However, cancer and PAH are easy to differentiate,

given their different symptoms, which would bring more

value in using proto-oncogenes as PAH biomarkers.

Pim1 proto-oncogene A preclinical trial is currently

ongoing to assess the potential of the Pim1 protoconcogene

as a biomarker in PAH. As explained earlier, our current

understanding of PAH physiopathology relies on NFAT

and STAT3 activation. Thus, our group has shown that

STAT3 is responsible for enhancing not only NFAT but

also Pim1 expression [20]. In fact, the latter oncogene is

required for NFAT activation in PAH. This means that, in

PAH, NFAT cannot be activated in the absence of Pim1

and cannot deregulate Ca2? homeostasis and other mech-

anisms responsible for PASMC proliferation and resistance

to apoptosis found in distal PA.

Interestingly, Pim1 is not normally expressed in human

tissues. Although Pim1 is also found in diverse types of

cancer, Paulin et al. showed that Pim1 exhibits high specificity

for the pulmonary vascular remodeling that characterize PAH

[20]. In PAH, Pim1 is markedly overexpressed in the

pulmonary vascular bed. Moreover, the activation of these

pathways is also observed in circulating immune cells,

possibly due to the inflammatory state of the lung. Indeed,

our group has shown that it is possible to measure Pim1

expression in the buffy coat of PAH patients. More impor-

tantly, preliminary data suggest Pim1 blood levels are

elevated in early disease and strongly correlate with the

severity of the disease. Finally, the simplicity of the method-

ology used to measure Pim1 (from blood sample and urine)

supports Pim1 as a promising biomarker in PAH [20].

Other oncogenes/tumor supressors The p27 tumor sup-

pressor is often downregulated in cancers harboring p53

mutations. Interestingly, overexpression of p27 decreases

PASMC proliferation and prevents hypoxia-induced PH in vivo

[175, 176]. Conversely, p53 deficiency in a hypoxia mouse

model is associated with a more severe PAH, an increase in

HIF-1a, and a loss of p21 expression. Furthermore, that tumor

suppressor has been shown to have a protective role in a left-to-

right shunt model, which strengthens its protective effect [165].

Yang et al. aimed to investigate the role of apoptosis in the

PA remodeling of PH secondary to hypoxia and to determine

the relative expression of selected genes [177]. Compared to a

control group, the apoptotic index of the hypoxic group

decreased significantly. Through the methods of in situ

hybridization and RT-PCR, this group found that Bcl-2

expression was increased, whereas Bax was decreased

significantly in the hypoxic group. The alteration in Bcl-2

and Bax expression induced by hypoxia plays an important

role in PA remodeling, which is the main pathological change

found in PH secondary to hypoxia [177].

Current understanding of cancer pathophysiology in

apoptosis resistance is fundamental to develop efficient

treatments, which might also be of help in PAH treatment.

However, a number of studies are required to evaluate

whether these oncogenes/tumor suppressors are quantifi-

able and have biomarker values. Indeed, a compounding

factor is that these proteins are not secreted. Actually,

unlike with cancer where the tumor is usually removed and

in situ hybridization may be performed, there is no direct

way to extract these polypeptides with PAH. Nonetheless,

it may be thought that, as for Pim1, some of these factors

might be found in circulating immune cells and, thus, in

patient’s buffy coat. Moreover, proteomic technology and

future improvements in microsurgery might facilitate the

investigation of oncogenes/tumor suppressors in PAH.

miRNA

Small noncoding miRNA (21–23 nt) are now recognized as

important regulators of gene expression and are involved in

2816 M. Barrier et al.

123

Page 13: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

most physiological and pathological processes. Several

studies are currently assessing whether miRNA expression

in blood samples might be used to identify PAH progres-

sion and to help in its diagnosis. Indeed, miRNA can be

secreted and may thus be found in fluid samples. Further-

more, as already discussed, inflammatory mediators are

important actors in PAH. Our laboratory has demonstrated

that the pathways involving NFAT, STAT3 and miR-204

described in PASMC (see below) may also be found in

inflammatory cells, since altered levels of mRNA and

oncogenes have been demonstrated in PH patients’ buffy

coats [14, 20].

Tumor suppressor miR-204 Our group has characterized

the crucial implication of miR-204 in PAH pathogenesis. In

fact, STAT3 is responsible for the decreased expression of

miR-204, which is itself a STAT3 inhibitor. In PAH, the

increase in circulating factors activates STAT3, and,

therefore, downregulates miR-204. Furthermore, as said

earlier, STAT3 enhances NFAT expression. Thus, via

many pathways previously described [15, 178], NFAT

leads to the proliferative and anti-apoptotic phenotype that

is responsible for the vascular remodeling caused by PAH.

This feedback loop between STAT3 and miR-204 might

explain the resistance to inhibitors of circulating factor

receptors such as imatinib (a tyrosine kinase inhibitor) and

bosentan (an endothelin receptor antagonist). We have

described the therapeutic value of miR-204 restoration,

since nebulization with a miR-204 mimic reverses the

pathology in the monocrotaline-PAH rat model [19]. The

latter study also highlights the biomarker potential of miR-

204. Indeed, the decrease in miR-204 expression is found

specifically in the lung and in PAH models, and correlates

with PAH severity in human buffy coat. Thus, although

these results must be replicated in larger patient cohorts,

miR-204 represents a promising biomarker that also cor-

relates with disease severity.

miR-21 oncogene As previously mentioned, PAH is

characterized by an increase in circulating blood factors. A

noteworthy observation is that TGF and bone morphoge-

netic protein (BMP) stimulation have been shown to

rapidly induce miRNA-21 (miR-21) [179, 180]. The latter

miRNA is significantly increased and correlates with poor

prognosis in lung cancer [181].

Wang et al. [182] described the profile of miRNA

expression in human arteries with arteriosclerosis obliter-

ans. Among them, miR-21 was mainly found in arterial

smooth muscle cells and was increased by [sevenfold in

arteriosclerosis obliterans, which was related to HIF-1aexpression. In cultured human arterial smooth muscle cells,

cell proliferation and migration were significantly

decreased by the inhibition of miR-21. These authors were

also able to confirm that tropomyosin-1 is a target of miR-

21 that is involved in the intracellular effects of miR-21.

The study of Sarkar et al. [183] has investigated the role of

miR-21 in hypoxia-induced PASMC proliferation and

migration. The latter group showed that miR-21 expression

increased by threefold in human PASMC after a 6-h

hypoxia (3% O2) and remained high (twofold) even after a

24-h hypoxia. They further showed that miR-21 is essential

for hypoxia-induced cell migration. Protein expression of

miR-21 target genes, i.e. programmed cell death protein-4,

Sprouty 2, and PPARa, was decreased by hypoxia and in

PASMC overexpressing miR-21 under normoxia, and was

increased in hypoxic cells in which miR-21 had been

knocked down. Their findings suggest that miR-21 plays a

significant role in hypoxia-induced PAMSC proliferation

and migration through the regulation of multiple gene

targets.

Using PAH rat models (hypoxic and monocrotaline),

Caruso et al. demonstrated miR-22 and miR-30 were

downregulated, whereas miR-322 and miR-451 were signif-

icantly upregulated in both PAH models. Interestingly,

miR-21 was selectively and consistently downregulated after

monocrotaline injection, but not under hypoxia. They also

described the downregulation of miR-21 expression in iPAH

human samples [184]. These findings suggest that a

substantial loss of miR-21 is associated with vascular

remodeling in monocrotaline-exposed lungs. The authors

suggested that reduced BMP signaling is possibly related to

miR-21 downregulation in the monocrotaline model, which

might be involved in the alteration of smooth muscle cell

phenotype found in PAH.

Taken together, these studies highlight the importance

of ascertaining the role of miR-21 in the different PAH

models as well as defining the potential effect of miR-21

modulation in disease prevention. Undoubtedly, these data

emphasize the importance of investigating miR-21 in the

pathology of PAH.

miR-210 oncogene miR-210 is the main hypoxia-sensi-

tive miRNA involved in the regulation of the hypoxic

response in tumor cells as well as tumor growth [185]. Its

expression is induced by HIF-1a and is known to protect

cancer cells from hypoxia-induced apoptosis, and to par-

ticipate in mitochondrial impairment and cell cycle

regulation, all features that are also found in PAH [19, 184,

186–188]. Nonetheless, miR-210 expression has not been

found to be altered either in human or PAH animal models.

miR-145 tumor suppressor The biomarker potential of

miR-145 has already been assessed in cancer [189].

miR-145 is the most abundant miRNA in normal vascular

walls, and is significantly decreased in pathological con-

ditions of vascular walls with neointimal lesions and

dedifferentiated vascular smooth muscle cells. Cell dedif-

ferentiation is well known to play a crucial role in the

Today’s and tomorrow’s imaging and circulating biomarkers 2817

123

Page 14: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

pathological vascular remodeling process that occurs in

several vascular pathologies, including PAH. Differentia-

tion can be affected by several stimuli, including growth

factors typically increased in PAH. Furthermore, Cheng

et al. [190] demonstrated that its restoration leads to the

loss of the pathological phenotype in an in vitro model of

vascular smooth muscle cells as well as in an in vivo model

of carotid angioplasty in rats. Hence, it might be very

interesting to assess the biomarker value of miR-145 in

PAH as well to determine whether it is also reduced in

PAH patients’ buffy coat, and whether it correlates with the

severity of the disease.

Conclusion Being already considered as biomarkers in

cancer, miRNA might be potential biomarkers in PAH in

the near future. Since miRNA are very short sequences that

are present at low concentrations in the blood, extraction

inaccuracies as well as the variance of replicate values will

constitute a serious methodological challenge [191]. Stan-

dardization of sample conservation, technical dosage and

extraction are of utmost importance before translating the

profiling of serum miRNA to clinical practice, lest such a

promising and powerful tool be misused.

Metabolic biomarkers

Serum lactate dehydrogenase (LDH) The Warburg effect

is increasingly correlated with aggressiveness and poor

prognosis in cancers [192]. Given that a similar metabolic

disorder is also found in PAH, serum LDH might be a

biomarker, although not specific, of the Warburg effect in

PAH.

Urine metabolomic derived biomarker A few teams are

currently working on the use of metabolomics-derived

biomarkers able to differentiate cancer from healthy

patients, and correlating with severity. These groups have

measured various metabolites in patients’ urine such as

quinolinate, 4-hydroxybenzoate, and gentisate [193]. It

might be of major interest to investigate whether specific

differences are observed in the urine of PAH patients.

Oxidative stress biomarkers and PAH

Reactive oxygen species (ROS) are predominantly impli-

cated in cell damage via the inhibition of protein, lipid and

DNA normal functions. These highly reactive species may

initiate several peroxidative reactions damaging the genetic

cell apparatus, as well as cell membranes, proteins and

lipids. ROS may be produced in the lung tissue in severe

PH patients as a result of tissue hypoxia [194], ischemia

[195] or via inflammatory cascade activation and increased

production of inflammatory cytokines [196, 197]. Oxida-

tive stress occurs when repeated external insults result in

excessive ROS formation, which overwhelms antioxidant

systems, thus creating an altered redox state favors oxi-

dation. Oxidative stress is also considered as a cancer

hallmark, but the present discussion will focus on the role

of ROS markers in PAH.

Increases in oxidative stress and in lipid peroxidation

products are associated with elevated PA pressure in dif-

ferent animal models of PH [198–200]. Altered ROS

generation is believed to play a crucial role in the vascular

responses observed under hypoxic conditions, such as

deregulation of pulmonary vasomotor tone [201–203].

Moreover, some animal models studies have shown that

ROS play a role in pathological RV remodeling. It is well

established that increased PAH causes pressure overload of

the RV, leading to RVH [204, 205]. Increased ROS gen-

eration and altered redox state are some of the critical

features of the transition from hypertrophy to heart failure.

Among various causes of hemodynamic and structural

abnormalities of the decompensating RV are neurohor-

monal signaling (angiotensin-II, ET-1, aldosterone),

natriuretic peptides, and inflammation, but also oxidative

stress (including ROS and reactive nitrogen species) [206].

Uric acid

Uric acid is an endogenous free radical scavenger pro-

tecting cells from ROS and reactive nitrogen species

damage. It has been demonstrated that uric acid levels are

increased in PAH [207]. Voelkel et al. hypothesized that

the site of uric acid production in PAH may be either the

ischemic lung tissue or the ischemic RV, or both [207].

Uric acid serum levels correlate with hemodynamic and

functional parameters [208, 209], and are related to sur-

vival in adult [33, 208] and pediatric [209] PAH. Uric acid

is thus a biological serum marker of disease severity in

PAH. However, uric acid serum levels are influenced by

allopurinol, diuretics and hypoxemia and display signifi-

cant interindividual variability, limiting its accuracy as a

biomarker in PAH.

Isoprostanes

Isoprostanes are products of the peroxidative attack of

membrane lipids and are present at nanomolar concentra-

tions in the blood of normal individuals [210, 211]. In

several disease states, and particularly pulmonary diseases

such as PAH, isoprostane levels are increased [199, 212].

Moreover, given their potent and multiple biological

activities, isoprostanes have been thought to account for

many of the pathophysiological processes found in PAH

(e.g., decrease in endothelial relaxing factors) [213]. A key

question about isoprostane analysis is which isoprostane

should be analyzed among the 64 distinct isoprostanes that

2818 M. Barrier et al.

123

Page 15: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

can be generated from arachidonic acid [214]. The 5- and

15-series F2-isoprostanes are produced in closely equal

amounts in vivo, whereas the 8- and 12-series F2-isopros-

tanes are produced in lower yields [215]. Considering their

relative stability, the availability of inexpensive assays for

quantification, the easy collection of blood and/or urine

samples, and the direct relationship between isoprostane

accumulation and oxidative stress intensity has made these

‘‘markers’’ of oxidative stress optimal biomarkers of lung

disease states [213]. Hence, isoprostanes might also be

potentially useful markers for PAH.

Serotonin (5-HT)

An elevation of circulating peripheral serotonin (5-HT) has

been shown to occur upon PAH development both experi-

mentally and clinically [216, 217]. This vasoactive amine may

also produce ROS [218–220] and can induce protein car-

bonylation in PASMC [221]. Moreover, 5-HT stimulates

PASMC proliferation, PA vasoconstriction and local micro-

thrombosis [222]. In addition, studies have demonstrated that

inhibition of 5-HT receptors and 5-HT transporters is able to

delay PAH development and extend rat survival [223, 224].

The therapeutic value of 5-HT also highlights its interest as a

biomarker. Thus, given the evidence that PAH patients have

increased plasma 5-HT levels, and that this hormone may be

considered as a pro-oxidant, it represents an attractive and

easily assayed biomarker for PAH diagnosis and follow-up.

Derived nitric oxide products

NO may act as a signaling molecule, a toxin, and a pro-oxidant

as well as a potential antioxidant [225]. It has been proposed to

act as a pro-oxidant when reacting with superoxide anion

(O2-), thereby generating peroxynitrite [226], or by itself at

high concentrations [227]. NO is a pulmonary vasodilator

produced and released by the endothelium. Among its main

functions are vascular tone regulation [160], platelet aggre-

gation and inhibition of vascular smooth muscle cells

proliferation. A reduction in available NO occurs in ROS-

overproducing cells. The alteration of eNOS expression has

also been associated with systemic and pulmonary hyperten-

sion [228–230]. In theory, low plasma NO might be a marker

of endothelial dysfunction and possibly PAH. However, NO is

too unstable to be measured in gaseous form in the blood

[160]. However, NO treatment has already been tested,

without much success, but NO-products and NO itself may

help PAH diagnostic and follow-up.

HIF-1

HIF-1, which contributes to oxidative stress, is involved in

the PAH pathogenesis, raising a genuine interest in this

protein. HIF-1 is a transcription factor responsible for

mediating physiological responses to hypoxia, and is found

in a variety of organisms. The HIF-1a subunit, also called

‘‘oxygen sensor’’, may be accumulated under normoxic

conditions as a result of various stimuli such as ROS,

growth factors, oncogenic activation and NO [231, 232].

The effect of NO on HIF-1a expression is reciprocal and

depends on the chemical structure and concentration of the

NO donor [233]. HIF-1 alteration may also indicate oxidant

damage [198], and, thus, both in vitro and in vivo studies

have demonstrated that HIF-1 protein levels are inversely

proportional to O2 levels in cell media [234, 235]. Fur-

thermore, HIF-1 has long been believed to increase ‘‘early-

response gene’’ transcription, which modulates vascular

remodeling in the lung as well as cardiovascular function

[236, 237]. Thus, HIF-1 is a promising candidate as a novel

biomarker and/or as a target for PAH treatment. Moreover,

its pathway actors should be carefully studied in order to

find their biomarker potential.

NADPH oxidase/superoxide dismutase (SOD)

Using monocrotaline PAH model, Redout et al. have

demonstrated that NADPH oxidase and mitochondria-

derived ROS production increase RV failure [204]. Their

findings indicate that mitochondrial SOD-1 and SOD-2

mRNA levels were decreased, and nitrotyrosine staining

confirmed the presence of oxidative stress. Importantly,

Archer et al. have identified mitochondrial SOD-2 meth-

ylation as a potential epigenetic mechanism for PAH in the

Fawn-Hooded rat PAH model [238]. It may be relevant to

investigate whether SOD-2 is also downregulated in human

PAH. Interestingly, SOD-2 epigenetic downregulation

impaired H2O2-mediated redox signaling, activating HIF-

1a and creating an apoptosis-resistant state. Importantly,

SOD-mimetic therapy reversed PAH in vivo, reinforcing

the involvement of HIF-1. The study by Archer et al. also

demonstrated that an increase in SOD-2 can restore mito-

chondrial function, inhibit proliferation and increase

PASMC apoptosis in vitro. This study was the first dem-

onstration of an epigenetic basis for PAH. These studies

demonstrate that SOD-2 might be a powerful tool for PAH

treatment and might have potential diagnostic value in

PAH in the future, directly or via proteins functionally

related to SOD-2.

Conclusion

There is an urgent need for alternative biomarkers that are

safer and more reliable than those that are currently

available. Because ROS may promote vasoconstriction,

vascular smooth muscle cells proliferation and vascular

remodeling in many PH forms, research should be pursued

Today’s and tomorrow’s imaging and circulating biomarkers 2819

123

Page 16: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

towards a thorough understanding of its critical role. There

is mounting evidence of a cross-talk between PAH patho-

genesis and oxidative stress, and monitoring oxidative

stress signs and damage may help to improve PAH diag-

nostic and prognosis. Additionally, these data may provide

a rationale for developing therapeutic strategies in RV

failure due to PAH.

Other biomarkers

A renal-derived biomarker: creatinine

Renal dysfunction is well known to predict mortality in

many cardiovascular diseases. Not surprisingly, Shah et al.

demonstrated that higher serum creatinine levels and

decreased glomerular filtration were associated with

hemodynamic severity and worse survival in PAH, results

that were also reported by other groups [126, 164].

CNS-derived: sympathetic nerve activity

Velez-Roa et al. [239] showed that PAH patients had

increased muscle sympathetic nerve activity compared to

controls, especially in advanced PAH. Moreover, they

demonstrated that sympathetic hyperactivity was partially

chemoreflex-mediated. Similarly, McGowan et al. docu-

mented muscle sympathetic nerve activity burst frequency

was higher in PAH patients and was inversely correlated

to the low-frequency spectral component of heart rate

variability [240]. The authors suggested that reduced heart

rate variability and sympathetic nervous system excitation

in PAH patients might increase the likelihood of sudden

death or accelerate progression to RV failure. This

hypothesis was supported by recent experiments docu-

menting the relationship between muscle sympathetic

nerve activity and disease severity and prognosis [254].

This deserves prospective evaluation in larger cohorts in

order to consolidate the status of heart rate variabil-

ity spectral power and sympathetic hyperactivity as

useful noninvasive surrogate markers of disease severity

in PAH.

Proteomic-derived biomarkers

Improvement in proteomic tools has led to the assessment

of proteomic-derived PAH biomarkers. Indeed, Zhang

et al. have suggested that simultaneous analysis of the

expression of ten protein spots may distinguish patients

with PAH from normal controls [241]. Nine proteins and

their isoforms were significantly different in the serum of

PAH patients relative to controls, including leucine-rich a-

2-glycoprotein, haptoglobin precursor, albumin isoform-2,

transferrin variant, C3 complement, hydroxypyruvate

reductase isoform-1, RAF1, fibrinogen isoform c-A, and

fibrinogen isoform c-B. In addition, significant and

important associations between LRG levels and functional

class FC (r = 0.71, p \ 0.01) and cardiac output (r = -

0.65, p \ 0.01) were shown.

Geraci et al. have also hypothesized that the gene

expression pattern found in PAH patient lung tissue has a

characteristic profile compared to healthy lungs [242].

Using oligonucleotide microarray technology, they char-

acterized gene expression patterns in the lung tissue

obtained from six PAH patients—including two patients

with the familial PAH form—as compared to six matched

healthy patients. All PAH lung tissue samples had a

decreased expression of several genes encoding various

kinases and phosphatases, whereas the expression of sev-

eral oncogenes and genes encoding ion channel proteins

was increased. Alterations in the expression of TGF-breceptor III, BMP2, MAP kinase kinase-5, RACK-1, apo-

lipoprotein C-III, and laminin receptor-1 were found

exclusively in PAH patients. The microarray gene

expression technique is a very useful molecular tool pro-

viding novel pertinent data that may bring a better

understanding of the pathobiology of different PH clinical

phenotypes. Unfortunately, the methodology used in such

studies cannot yet be used in the clinical practice due to its

high cost. Nevertheless, it remains a promising new

approach that should be kept in mind with the incessant

technical improvements in proteomic analysis, and

might also lead to the discovery of more easily assessable

serum or urine biomarkers. Taken together, these results

indicate that proteomic analysis might be helpful in PAH

diagnosis.

Red blood cells distribution width

Red blood cell distribution width (RDW) reflects red blood

cell size variability and is commonly reported in automated

complete blood counts. The relationship between RDW

and PAH was investigated in a prospective study of 162

patients [243]. The authors found RDW to be related to

disease severity and demonstrated that it is independently

associated with mortality in PAH. Moreover, RDW per-

formed better as a prognostic indicator than NT-proBNP

and added important prognostic value over NT-proBNP

measurement and exercise capacity. The authors concluded

that RDW and NT-proBNP might be used in combination

to improve PAH prognosis.

Immune system-derived biomarkers: antibodies

Specific antibodies such as anti-Scl-70 may also be used as

biomarkers to assess the implications of other pathologies

[45]. Around 40% of PAH patients have positive but low

2820 M. Barrier et al.

123

Page 17: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

antinuclear antibody titers [244]. Other antibodies seem to

be promising, such as anti-endothelial cell antibodies [245]

or even antifibroblast antibodies [246]. Indeed, considering

the ease with which antibodies can be tested, it might be of

great interest to assess the presence and level of such

antibodies in the serum of PAH patients. This might rep-

resent easily measurable clinical parameters of potential

usefulness for future diagnosis and prognosis.

Hyponatremia

Growing evidences demonstrate that hyponatremia is

strongly associated with advanced RV dysfunction, poor

survival and WHO FC in PAH [247]. Since it is already

used and it is easy to assess, hyponatremia could represent

a powerful and independent biomarker. However, the

mechanism of hyponatremia remains elusive. Thus, even

though renal dysfunction is frequently seen in PAH patients

often treated with diuretics, Forfia et al. hypothesized that

hyponatremia may be a result of neurohormonal activation

(increased in PAH) possibly caused by more advanced RV

dysfunction [247].

High-density lipoprotein cholesterol (HDL-C)

It is well known that HDL-C is associated with poor

prognostic in cardiovascular diseases, but, interestingly,

decrease in this factor is also associated with higher mor-

tality and clinical worsening and outcome in PAH.

Furthermore, Heresi et al., have demonstrated that a

decrease in HDL-C does not appear to be caused by

underlying cardiovascular risk such as diabete mellitus,

BMI or other cardiovascular risk factors. Moreover, HDL-

C level is even lower in PAH patients than in a ‘‘high-risk

population’’ (patients with coronary disease, systemic

hypertension and diabetes, conditions associated with

lower HDL-C) [248].

Conclusion

All the biomarkers discussed in this part have been sum-

marized in Fig. 2. As shown above, current circulating

biomarkers may be helpful in the diagnosis of PAH, but

most of them lack specificity, and, unfortunately, many

appear only at severe stages of the disease. For these rea-

sons, PAH experts now consider the identification and

validation of a clinically relevant biomarker that is repre-

sentative of PAH physiopathology as a high priority

objective [89, 233, 249–252]. Cancer-related biomarkers

are promising tools but may lead to misdiagnosis. How-

ever, it should be kept in mind that, since symptoms of

cancer and PAH are very different, the use of cancer

biomarkers as PAH biomarkers might well be feasible in

practice. Moreover, the effectiveness of cancer diagnosis is

constantly improving. Likewise, oxidative stress-derived

biomarkers might be of some value for PAH diagnosis and

prognosis purposes. Finally, technological improvements

of proteomic tools may highlight the implication of new

actors in PAH physiopathology and lead to the discovery of

novel biomarkers and therapeutical targets. However,

many of the promising biomarkers already described must

still be assessed in larger patient cohorts to be validated. A

coordination between large PAH treatment centers should

be expected to include the standardization of study proto-

cols and tools in order to increase the number of patient in

such studies, and might significantly contribute to accel-

erate the validation of these biomarkers. In addition, the

combination of different biomarkers might be highly

relevant.

General conclusion

As of today, PAH diagnosis is still an exclusion diagnosis

involving a long period between the discovery of symp-

toms and diagnosis, resulting in a long delay before

initiating any treatment. Unfortunately, current imaging

and circulating biomarkers are only reliable in late stage

disease, limiting their utility for screening at-risk popula-

tions. Hence, improvements are much needed to improve

diagnostic strategies. Nevertheless, imaging biomarkers

represent crucial tools in determining the etiology of PAH,

excluding other types of PH and providing information

regarding RV function, disease severity and response to

therapy. The very rapid improvement in imaging tech-

niques seen in the previous decades strongly suggest that

imaging by CT, MRI and even echo will be further

improved, thereby decreasing the requirement for invasive

techniques such as RHC and allowing an earlier and more

accurate diagnosis. In addition, circulating biomarkers

show promises in PAH. Technical improvements are also

implemented in biochemical engineering, and the detection

of very small amounts of protein is now possible, thus

opening new avenues for circulating biomarkers. More

importantly, studies on biomarkers including oncogenes

and miRNA as well as oxidative stress-related proteins

may increase the understanding of PAH pathophysiology

and identify novel therapeutic targets.

Despite the technical improvements, the prospective

validation of new imaging and circulating biomarkers

relies on multicenter large-scale trials and standardization

of the techniques. While no ‘‘perfect biomarker’’ is likely

to be discovered any time soon, one might surmise that a

combination of biomarkers will together allow earlier

Today’s and tomorrow’s imaging and circulating biomarkers 2821

123

Page 18: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

diagnosis, more accurate prognostication and improved

long-term outcomes in PAH.

Acknowledgments M.H.J. is supported by CAPES (Brazilian

Research Agency) and by Laval University. J.M. is a recipient of a

Graduate Scholarship from the Canadian Institutes of Health Research

CIHR) and A.C. received a graduate scholarship from La Societe

Quebecoise d’Hypertension Arterielle (SQHA). This work was sup-

port by Canada Research Chairs (CRC) and by Canadian Institutes of

Health Research (CIHR) to S.B. We would like to thank Dr. Richard

Poulin for editorial help in processing this manuscript.

References

1. Simonneau G, Galie N, Rubin LJ, Langleben D, Seeger W,

Domenighetti G, Gibbs S, Lebrec D, Speich R, Beghetti M, Rich

S, Fishman A (2004) Clinical classification of pulmonary

hypertension. J Am Coll Cardiol 43(12 Suppl S):5S–12S. doi:

10.1016/j.jacc.2004.02.037

2. Voelkel NF, Tuder RM (1997) Cellular and molecular biology

of vascular smooth muscle cells in pulmonary hypertension.

Pulm Pharmacol Ther 10(5–6):231–241 pii: S1094-5539(98)

90100-6

3. Rubin LJ (1997) Primary pulmonary hypertension. N Engl J

Med 336(2):111–117. doi:10.1056/NEJM199701093360207

4. D’Alonzo GE, Barst RJ, Ayres SM, Bergofsky EH, Brundage

BH, Detre KM, Fishman AP, Goldring RM, Groves BM, Kernis

JT et al (1991) Survival in patients with primary pulmonary

hypertension. Results from a national prospective registry. Ann

Intern Med 115(5):343–349

5. Simonneau G, Robbins IM, Beghetti M, Channick RN, Delcroix

M, Denton CP, Elliott CG, Gaine SP, Gladwin MT, Jing ZC,

Krowka MJ, Langleben D, Nakanishi N, Souza R (2009)

Updated clinical classification of pulmonary hypertension. J Am

Coll Cardiol 54(1 Suppl):S43–54. doi:10.1016/j.jacc.2009.

04.012

6. Humbert M, Nunes H, Sitbon O, Parent F, Herve P, Simonneau

G (2001) Risk factors for pulmonary arterial hypertension. Clin

Chest Med 22(3):459–475

7. Galie N, Torbicki A, Barst R, Dartevelle P, Haworth S, Hi-

genbottam T, Olschewski H, Peacock A, Pietra G, Rubin LJ,

Simonneau G (2005) Guidelines on diagnosis and treatment of

pulmonary arterial hypertension. Rev Esp Cardiol 58(5):523–

566

Fig. 2 Pulmonary endothelium, circulating cells and microparticles

derived biomarkers. A large number of disorders are accountable for

PAH development and sustainability, which are also accountable for

distinct potential biomarkers. Among these, pulmonary artery smooth

muscle cells have a cancer like phenotype (miRNA, Pim1), metabolic

disorders (lactacte dehydrogenase, LDH) and increased oxidative

stress (isoprostane, uric acid). Furthermore, inflammation is

enhanced, inducing production of biomarkers, such as Endothelin

(ET), Interleukin, Englodin (Eng), GDF-15 and LIGHT. These

markers could be assessed in blood samples

2822 M. Barrier et al.

123

Page 19: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

8. McLure LE, Peacock AJ (2007) Imaging of the heart in pul-

monary hypertension. Int J Clin Pract Suppl 156:15–26. doi:

10.1111/j.1742-1241.2007.01496.x

9. Provencher S, Jais X, Yaici A, Sitbon O, Humbert M, Simon-

neau G (2005) Clinical challenges in pulmonary hypertension:

Roger S Mitchell lecture. Chest 128(6 Suppl):622S–628S. doi:

10.1378/chest.128.6_suppl.622S-a

10. Sitbon O, Humbert M, Simonneau G (2002) Primary pulmonary

hypertension: current therapy. Prog Cardiovasc Dis 45(2):115–

128 pii: S0033062002500384

11. McLaughlin VV, Rich S (2004) Pulmonary hypertension. Curr

Probl Cardiol 29(10):575–634. doi:10.1016/j.cpcardiol.2004.

04.001

12. McLaughlin VV (2002) Medical management of primary pul-

monary hypertension. Expert Opin Pharmacother 3(2):159–165.

doi:10.1517/14656566.3.2.159

13. Hoeper MM, Markevych I, Spiekerkoetter E, Welte T, Nieder-

meyer J (2005) Goal-oriented treatment and combination

therapy for pulmonary arterial hypertension. Eur Respir J

26(5):858–863. doi:10.1183/09031936.05.00075305

14. Courboulin A, Paulin R, Giguere NJ, Saksouk N, Perreault T,

Meloche J, Paquet ER, Biardel S, Provencher S, Cote J, Simard

MJ, Bonnet S (2011) Role for miR-204 in human pulmonary

arterial hypertension. J Exp Med. doi:10.1084/jem.20101812

15. Bonnet S, Rochefort G, Sutendra G, Archer SL, Haromy A,

Webster L, Hashimoto K, Bonnet SN, Michelakis ED (2007)

The nuclear factor of activated T cells in pulmonary arterial

hypertension can be therapeutically targeted. Proc Natl Acad Sci

USA 104(27):11418–11423. doi:10.1073/pnas.0610467104

16. Rehman J, Archer SL (2010) A proposed mitochondrial-meta-

bolic mechanism for initiation and maintenance of pulmonary

arterial hypertension in fawn-hooded rats: the Warburg model of

pulmonary arterial hypertension. Adv Exp Med Biol 661:171–

185. doi:10.1007/978-1-60761-500-2_11

17. Zaidi SH, You XM, Ciura S, Husain M, Rabinovitch M (2002)

Overexpression of the serine elastase inhibitor elafin protects

transgenic mice from hypoxic pulmonary hypertension. Circu-

lation 105(4):516–521

18. Archer S, Rich S (2000) Primary pulmonary hypertension: a

vascular biology and translational research ‘‘Work in progress’’.

Circulation 102(22):2781–2791

19. Courboulin A, Paulin R, Giguere NJ, Saksouk N, Perreault T,

Meloche J, Paquet ER, Biardel S, Provencher S, Cote J, Simard

MJ, Bonnet S (2011) Role for miR-204 in human pulmonary

arterial hypertension. J Exp Med 208 (3):535–548. doi:10.1084/

jem.20101812

20. Paulin R, Courboulin A, Meloche J, Mainguy V, Dumas de la

Roque E, Saksouk N, Cote J, Provencher S, Sussman MA, Bonnet S

(2011) Signal transducers and activators of transcription-3/pim1

axis plays a critical role in the pathogenesis of human pulmonary

arterial hypertension. Circulation 123 (11):1205–1215. doi:

10.1161/CIRCULATIONAHA.110.963314

21. Morrell NW, Yang X, Upton PD, Jourdan KB, Morgan N,

Sheares KK, Trembath RC (2001) Altered growth responses of

pulmonary artery smooth muscle cells from patients with pri-

mary pulmonary hypertension to transforming growth factor-

beta(1) and bone morphogenetic proteins. Circulation 104(7):

790–795

22. Hansmann G, Wagner RA, Schellong S, Perez VA, Urashima T,

Wang L, Sheikh AY, Suen RS, Stewart DJ, Rabinovitch M

(2007) Pulmonary arterial hypertension is linked to insulin

resistance and reversed by peroxisome proliferator-activated

receptor-gamma activation. Circulation 115(10):1275–1284.

doi:10.1161/CIRCULATIONAHA.106.663120

23. Hansmann G, de Jesus Perez VA, Alastalo TP, Alvira CM,

Guignabert C, Bekker JM, Schellong S, Urashima T, Wang L,

Morrell NW, Rabinovitch M (2008) An antiproliferative BMP-

2/PPARgamma/apoE axis in human and murine SMCs and its

role in pulmonary hypertension. J Clin Invest 118

(5):1846–1857. doi:10.1172/JCI32503

24. Rabinovitch M (2010) PPARgamma and the pathobiology of

pulmonary arterial hypertension. Adv Exp Med Biol

661:447–458. doi:10.1007/978-1-60761-500-2_29

25. Care A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P,

Bang ML, Segnalini P, Gu Y, Dalton ND, Elia L, Latronico MV,

Hoydal M, Autore C, Russo MA, Dorn GW 2nd, Ellingsen O,

Ruiz-Lozano P, Peterson KL, Croce CM, Peschle C, Condorelli

G (2007) MicroRNA-133 controls cardiac hypertrophy. Nat

Med 13(5):613–618. doi:10.1038/nm1582

26. Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M (2007)

MicroRNAs play an essential role in the development of cardiac

hypertrophy. Circ Res 100(3):416–424. doi:10.1161/01.RES.

0000257913.42552.23

27. Rafanan AL, Golish JA, Dinner DS, Hague LK, Arroliga AC

(2001) Nocturnal hypoxemia is common in primary pulmonary

hypertension. Chest 120(3):894–899

28. Spiekerkoetter E, Fabel H, Hoeper MM (2002) Effects of

inhaled salbutamol in primary pulmonary hypertension. Eur

Respir J 20(3):524–528

29. Sun XG, Hansen JE, Oudiz RJ, Wasserman K (2003) Pulmonary

function in primary pulmonary hypertension. J Am Coll Cardiol

41(6):1028–1035 pii: S0735109702029649

30. Sun XG, Hansen JE, Oudiz RJ, Wasserman K (2001) Exercise

pathophysiology in patients with primary pulmonary hyperten-

sion. Circulation 104(4):429–435

31. Taichman DB, McGoon MD, Harhay MO, Archer-Chicko C,

Sager JS, Murugappan M, Chakinali MM, Palevsky HI, Gallop

R (2009) Wide variation in clinicians’ assessment of New York

Heart Association/World Health Organization functional class in

patients with pulmonary arterial hypertension. Mayo Clin Proc

84(7):586–592. doi:10.4065/84.7.586

32. Rhodes J, Barst RJ, Garofano RP, Thoele DG, Gersony WM

(1991) Hemodynamic correlates of exercise function in patients

with primary pulmonary hypertension. J Am Coll Cardiol

18(7):1738–1744 pii: 0735-1097(91)90513-9

33. Wensel R, Opitz CF, Anker SD, Winkler J, Hoffken G, Kleber

FX, Sharma R, Hummel M, Hetzer R, Ewert R (2002) Assess-

ment of survival in patients with primary pulmonary

hypertension: importance of cardiopulmonary exercise testing.

Circulation 106(3):319–324

34. Groepenhoff H, Vonk-Noordegraaf A, Boonstra A, Spreeu-

wenberg MD, Postmus PE, Bogaard HJ (2008) Exercise testing

to estimate survival in pulmonary hypertension. Med Sci Sports

Exerc 40(10):1725–1732. doi:10.1249/MSS.0b013e31817c92c0

35. Miyamoto S, Nagaya N, Satoh T, Kyotani S, Sakamaki F, FujitaM, Nakanishi N, Miyatake K (2000) Clinical correlates and

prognostic significance of six-minute walk test in patients with

primary pulmonary hypertension. Comparison with cardiopul-

monary exercise testing. Am J Respir Crit Care Med 161(2 Pt

1):487–492

36. Provencher S, Chemla D, Herve P, Sitbon O, Humbert M, Si-

monneau G (2006) Heart rate responses during the 6-minute

walk test in pulmonary arterial hypertension. Eur Respir J

27(1):114–120. doi:10.1183/09031936.06.00042705

37. Lee WT, Peacock AJ, Johnson MK The role of per cent pre-

dicted 6-min walk distance in pulmonary arterial hypertension.

Eur Respir J 36(6):1294–1301. doi:10.1183/09031936.00155009

38. Hoeper MM, Oudiz RJ, Peacock A, Tapson VF, Haworth SG,

Frost AE, Torbicki A (2004) End points and clinical trial designs

in pulmonary arterial hypertension: clinical and regulatory per-

spectives. J Am Coll Cardiol 43(12 Suppl S):48S–55S. doi:

10.1016/j.jacc.2004.02.010

Today’s and tomorrow’s imaging and circulating biomarkers 2823

123

Page 20: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

39. Farber HW, Foreman AJ, Miller DP, McGoon MD REVEAL

Registry: correlation of right heart catheterization and echocar-

diography in patients with pulmonary arterial hypertension.

Congest Heart Fail 17(2):56–64. doi:10.1111/j.1751-7133.

2010.00202.x

40. Galie N, Hoeper MM, Humbert M, Torbicki A, Vachiery JL,

Barbera JA, Beghetti M, Corris P, Gaine S, Gibbs JS, Gomez-

Sanchez MA, Jondeau G, Klepetko W, Opitz C, Peacock A,

Rubin L, Zellweger M, Simonneau G (2009) Guidelines for the

diagnosis and treatment of pulmonary hypertension. Eur Respir

J 34(6):1219–1263. doi:10.1183/09031936.00139009

41. Budev MM, Arroliga AC, Jennings CA (2003) Diagnosis and

evaluation of pulmonary hypertension. Cleve Clin J Med

70(Suppl 1):S9–17

42. Hoeper MM, Lee SH, Voswinckel R, Palazzini M, Jais X,

Marinelli A, Barst RJ, Ghofrani HA, Jing ZC, Opitz C, Seyfarth

HJ, Halank M, McLaughlin V, Oudiz RJ, Ewert R, Wilkens H,

Kluge S, Bremer HC, Baroke E, Rubin LJ (2006) Complications

of right heart catheterization procedures in patients with pul-

monary hypertension in experienced centers. J Am Coll Cardiol

48(12):2546–2552. doi:10.1016/j.jacc.2006.07.061

43. McGoon M, Gutterman D, Steen V, Barst R, McCrory DC,

Fortin TA, Loyd JE (2004) Screening, early detection, and

diagnosis of pulmonary arterial hypertension: ACCP evidence-

based clinical practice guidelines. Chest 126(1 Suppl):14S–34S.

doi:10.1378/chest.126.1_suppl.14S

44. Mahapatra S, Nishimura RA, Sorajja P, Cha S, McGoon MD

(2006) Relationship of pulmonary arterial capacitance and

mortality in idiopathic pulmonary arterial hypertension. J Am

Coll Cardiol 47(4):799–803. doi:10.1016/j.jacc.2005.09.054

45. Schannwell CM, Steiner S, Strauer BE (2007) Diagnostics in

pulmonary hypertension. J Physiol Pharmacol 58 Suppl 5(Pt

2):591–602

46. Benza R, Biederman R, Murali S, Gupta H (2008) Role of

cardiac magnetic resonance imaging in the management of

patients with pulmonary arterial hypertension. J Am Coll Car-

diol 52(21):1683–1692. doi:10.1016/j.jacc.2008.08.033

47. Ramani GV, Gurm G, Dilsizian V, Park MH Noninvasive

assessment of right ventricular function: will there be resurgence

in radionuclide imaging techniques? Curr Cardiol Rep

12(2):162–169. doi:10.1007/s11886-010-0092-y

48. Raymond RJ, Hinderliter AL, Willis PW, Ralph D, Caldwell EJ,

Williams W, Ettinger NA, Hill NS, Summer WR, de Boisblanc

B, Schwartz T, Koch G, Clayton LM, Jobsis MM, Crow JW,

Long W (2002) Echocardiographic predictors of adverse out-

comes in primary pulmonary hypertension. J Am Coll Cardiol

39(7):1214–1219

49. Eysmann SB, Palevsky HI, Reichek N, Hackney K, Douglas PS

(1989) Two-dimensional and Doppler-echocardiographic and

cardiac catheterization correlates of survival in primary pul-

monary hypertension. Circulation 80(2):353–360

50. Abraham J, Abraham TP (2009) The role of echocardiography

in hemodynamic assessment in heart failure. Heart Fail Clin

5(2):191–208. doi:10.1016/j.hfc.2008.11.002

51. Filusch A, Mereles D, Gruenig E, Buss S, Katus HA, Meyer FJ

(2010) Strain and strain rate echocardiography for evaluation of

right ventricular dysfunction in patients with idiopathic pul-

monary arterial hypertension. Clin Res Cardiol 99(8):491–498.

doi:10.1007/s00392-010-0147-5

52. Kovacs G, Reiter G, Reiter U, Rienmuller R, Peacock A, Ols-

chewski H (2008) The emerging role of magnetic resonance

imaging in the diagnosis and management of pulmonary

hypertension. Respiration 76(4):458–470. doi:10.1159/0001

58548

53. Karatasakis GT, Karagounis LA, Kalyvas PA, Manginas A,

Athanassopoulos GD, Aggelakas SA, Cokkinos DV (1998)

Prognostic significance of echocardiographically estimated right

ventricular shortening in advanced heart failure. Am J Cardiol

82(3):329–334

54. Ghio S, Recusani F, Klersy C, Sebastiani R, Laudisa ML,

Campana C, Gavazzi A, Tavazzi L (2000) Prognostic usefulness

of the tricuspid annular plane systolic excursion in patients with

congestive heart failure secondary to idiopathic or ischemic

dilated cardiomyopathy. Am J Cardiol 85(7):837–842

55. Samad BA, Alam M, Jensen-Urstad K (2002) Prognostic impact

of right ventricular involvement as assessed by tricuspid annular

motion in patients with acute myocardial infarction. Am J

Cardiol 90(7):778–781

56. Forfia PR, Fisher MR, Mathai SC, Housten-Harris T, Hemnes AR,

Borlaug BA, Chamera E, Corretti MC, Champion HC, Abraham

TP, Girgis RE, Hassoun PM (2006) Tricuspid annular displacement

predicts survival in pulmonary hypertension. Am J Respir Crit Care

Med 174(9):1034–1041. doi:10.1164/rccm.200604-547OC

57. McLaughlin VV, Presberg KW, Doyle RL, Abman SH, McCr-

ory DC, Fortin T, Ahearn G (2004) Prognosis of pulmonary

arterial hypertension: ACCP evidence-based clinical practice

guidelines. Chest 126(1 Suppl):78S–92S. doi:10.1378/chest.126.

1_suppl.78S

58. Hinderliter AL, Willis PWt, Barst RJ, Rich S, Rubin LJ, Ba-

desch DB, Groves BM, McGoon MD, Tapson VF, Bourge RC,

Brundage BH, Koerner SK, Langleben D, Keller CA, Murali S,

Uretsky BF, Koch G, Li S, Clayton LM, Jobsis MM, Blackburn

SD Jr., Crow JW, Long WA (1997) Effects of long-term infu-

sion of prostacyclin (epoprostenol) on echocardiographic

measures of right ventricular structure and function in primary

pulmonary hypertension. Primary Pulmonary Hypertension

Study Group. Circulation 95(6):1479–1486

59. Grunig E, Weissmann S, Ehlken N, Fijalkowska A, Fischer C,

Fourme T, Galie N, Ghofrani A, Harrison RE, Huez S, Humbert

M, Janssen B, Kober J, Koehler R, Machado RD, Mereles D,

Naeije R, Olschewski H, Provencher S, Reichenberger F, Re-

tailleau K, Rocchi G, Simonneau G, Torbicki A, Trembath R,

Seeger W (2009) Stress Doppler echocardiography in relatives

of patients with idiopathic and familial pulmonary arterial

hypertension: results of a multicenter European analysis of

pulmonary artery pressure response to exercise and hypoxia.

Circulation 119(13):1747–1757. doi:10.1161/CIRCULATION

AHA.108.800938

60. Hachulla E, Gressin V, Guillevin L, Carpentier P, Diot E, Sibilia

J, Kahan A, Cabane J, Frances C, Launay D, Mouthon L,

Allanore Y, Tiev KP, Clerson P, de Groote P, Humbert M

(2005) Early detection of pulmonary arterial hypertension in

systemic sclerosis: a French nationwide prospective multicenter

study. Arthritis Rheum 52(12):3792–3800. doi:10.1002/art.

21433

61. Sitbon O, Lascoux-Combe C, Delfraissy JF, Yeni PG, Raffi F,

De Zuttere D, Gressin V, Clerson P, Sereni D, Simonneau G

(2008) Prevalence of HIV-related pulmonary arterial hyperten-

sion in the current antiretroviral therapy era. Am J Respir Crit

Care Med 177(1):108–113. doi:10.1164/rccm.200704-541OC

62. Vitarelli A, Conde Y, Cimino E, Stellato S, D’Orazio S,

D’Angeli I, Nguyen BL, Padella V, Caranci F, Petroianni A,

D’Antoni L, Terzano C (2006) Assessment of right ventricular

function by strain rate imaging in chronic obstructive pulmonary

disease. Eur Respir J 27(2):268–275. doi:10.1183/09031936.06.

00072005

63. Gopal AS, Chukwu EO, Mihalatos DG, Katz AS, Mathew ST,

Lachmann JS, Toole RS, Schapiro W, Reichek N (2007) Left

ventricular structure and function for postmyocardial infarction

and heart failure risk stratification by three-dimensional echo-

cardiography. J Am Soc Echocardiogr 20(8):949–958. doi:

10.1016/j.echo.2007.01.014

2824 M. Barrier et al.

123

Page 21: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

64. Grapsa J, Gibbs JS, Cabrita IZ, Watson GF, Pavlopoulos H,

Dawson D, Gin-Sing W, Howard LS, Nihoyannopoulos P

(2012) The association of clinical outcome with right atrial and

ventricular remodelling in patients with pulmonary arterial

hypertension: study with real-time three-dimensional echocar-

diography. Eur Heart J Cardiovasc Imaging

65. Apfel HD, Shen Z, Gopal AS, Vangi V, Solowiejczyk D, Alt-

mann K, Barst RJ, Boxt LM, Allan LD, King DL (1996)

Quantitative three dimensional echocardiography in patients

with pulmonary hypertension and compressed left ventricles:

comparison with cross sectional echocardiography and magnetic

resonance imaging. Heart 76(4):350–354

66. Sachdev A, Villarraga HR, Frantz RP, McGoon MD, Hsiao JF,

Maalouf JF, Ammash NM, McCully RB, Miller FA, Pellikka

PA, Oh JK, Kane GC (2011) Right ventricular strain for pre-

diction of survival in patients with pulmonary arterial

hypertension. Chest 139(6):1299–1309

67. Borges AC, Knebel F, Eddicks S, Panda A, Schattke S, Witt C,

Baumann G (2006) Right ventricular function assessed by two-

dimensional strain and tissue Doppler echocardiography in

patients with pulmonary arterial hypertension and effect of

vasodilator therapy. Am J Cardiol 98(4):530–534. doi:

10.1016/j.amjcard.2006.02.060

68. Suga K, Tokuda O, Okada M, Koike M, Iwanaga H, Matsunaga

N (2010) Assessment of cross-sectional lung ventilation-perfu-

sion imbalance in primary and passive pulmonary hypertension

with automated V/Q SPECT. Nucl Med Commun 31(7):

673–681. doi:10.1097/MNM.0b013e328339ea9b

69. Mehta S, Helmersen D, Provencher S, Hirani N, Rubens FD, De

Perrot M, Blostein M, Boutet K, Chandy G, Dennie C, Granton

J, Hernandez P, Hirsch AM, Laframboise K, Levy RD, Lien D,

Martel S, Shoemaker G, Swiston J, Weinkauf J (2010) Diag-

nostic evaluation and management of chronic thromboembolic

pulmonary hypertension: a clinical practice guideline. Can

Respir J 17(6):301–334

70. Wong YY, Raijmakers PG, Knaapen P, Lubberink M, Ruiter G,

Marcus JT, Boonstra A, Lammertsma AA, Westerhof N, van der

Laarse WJ, Vonk-Noordegraaf (2011) A Supine-exercise-

induced oxygen supply to the right myocardium is attenuated in

patients with severe idiopathic pulmonary arterial hypertension.

Heart 97(24):2069–2074. doi:10.1136/heartjnl-2011-300237

71. Abikhzer Y, Probst S, Rush C (2008) Pulmonary hypertension

findings detected by F-18 FDG PET scan. Clin Nucl Med

33(6):405–406. doi:10.1097/RLU.0b013e31817082ad

72. Marsboom G, Wietholt C, Haney CR, Toth PT, Ryan JJ, Mor-

row E, Thenappan T, Bache-Wiig P, Piao L, Paul J, Chen CT,

Archer SL (2012) Lung 18F-fluorodeoxyglucose PET for diag-

nosis and monitoring of pulmonary arterial hypertension. Am J

Respir Crit Care Med. doi:10.1164/rccm.201108-1562OC

73. Jakobsen S, Kodahl GM, Olsen AK, Cumming P (2006) Syn-

thesis, radiolabeling and in vivo evaluation of [11C]RAL-01, a

potential phosphodiesterase 5 radioligand. Nucl Med Biol

33(5):593–597. doi:10.1016/j.nucmedbio.2006.04.006

74. Lewis GD, Semigran MJ (2004) Type 5 phosphodiesterase

inhibition in heart failure and pulmonary hypertension. Curr

Heart Fail Rep 1(4):183–189

75. 4-[18F]Fluorobenzoyl-endothelin-1 (2004). NBK23311

[bookaccession]

76. He Q, Xu RZ, Shkarin P, Pizzorno G, Lee-French CH, Rothman

DL, Shungu DC, Shim H (2003) Magnetic resonance spectro-

scopic imaging of tumor metabolic markers for cancer

diagnosis, metabolic phenotyping, and characterization of tumor

microenvironment. Dis Markers 19(2–3):69–94

77. Grosse C, Grosse A (2010) CT findings in diseases associated

with pulmonary hypertension: a current review. Radiographics

30(7):1753–1777. doi:10.1148/rg.307105710

78. Devaraj A, Wells AU, Meister MG, Corte TJ, Wort SJ, Hansell

DM (2010) Detection of pulmonary hypertension with multi-

detector CT and echocardiography alone and in combination.

Radiology 254(2):609–616. doi:10.1148/radiol.09090548

79. Sanchez O, Revel MP, Couchon S, Meyer G (2007) Imaging of

pulmonary arterial hypertension. Rev Mal Respir 24(2):155–169

pii:MDOI-RMR-02-2007-24-2-0761-8425-101019-200520005

80. Boerrigter B, Mauritz GJ, Marcus JT, Helderman F, Postmus

PE, Westerhof N, Vonk-Noordegraaf A (2010) Progressive

dilatation of the main pulmonary artery is a characteristic of

pulmonary arterial hypertension and is not related to changes in

pressure. Chest 138(6):1395–1401. doi:10.1378/chest.10-0363

81. Nicol ED, Kafka H, Stirrup J, Padley SP, Rubens MB, Kilner PJ,

Gatzoulis MA (2009) A single, comprehensive non-invasive

cardiovascular assessment in pulmonary arterial hypertension:

combined computed tomography pulmonary and coronary

angiography. Int J Cardiol 136(3):278–288. doi:10.1016/j.ijcard.

2008.05.049

82. Revel MP, Faivre JB, Remy-Jardin M, Delannoy-Deken V,

Duhamel A, Remy J (2009) Pulmonary hypertension: ECG-

gated 64-section CT angiographic evaluation of new functional

parameters as diagnostic criteria. Radiology 250(2):558–566.

doi:10.1148/radiol.2502080315

83. Matsuoka S, Washko GR, Dransfield MT, Yamashiro T, San

Jose Estepar R, Diaz A, Silverman EK, Patz S, Hatabu H (2010)

Quantitative CT measurement of cross-sectional area of small

pulmonary vessel in COPD: correlations with emphysema and

airflow limitation. Acad Radiol 17(1):93–99. doi:10.1016/j.acra.

2009.07.022

84. Tan RT, Kuzo R, Goodman LR, Siegel R, Haasler GB, Presberg

KW (1998) Utility of CT scan evaluation for predicting pul-

monary hypertension in patients with parenchymal lung disease.

Medical College of Wisconsin Lung Transplant Group. Chest

113(5):1250–1256

85. d’Agostino AG, Remy-Jardin M, Khalil C, Delannoy-Deken V,

Flohr T, Duhamel A, Remy J (2006) Low-dose ECG-gated

64-slices helical CT angiography of the chest: evaluation of

image quality in 105 patients. Eur Radiol 16(10):2137–2146.

doi:10.1007/s00330-006-0218-1

86. Johnson TR, Krauss B, Sedlmair M, Grasruck M, Bruder H,

Morhard D, Fink C, Weckbach S, Lenhard M, Schmidt B, Flohr

T, Reiser MF, Becker CR (2007) Material differentiation by

dual energy CT: initial experience. Eur Radiol 17(6):1510–1517.

doi:10.1007/s00330-006-0517-6

87. Thieme SF, Becker CR, Hacker M, Nikolaou K, Reiser MF,

Johnson TR (2008) Dual energy CT for the assessment of lung

perfusion–correlation to scintigraphy. Eur J Radiol 68(3):369–

374. doi:10.1016/j.ejrad.2008.07.031

88. Ley S, Kreitner KF, Fink C, Heussel CP, Borst MM, Kauczor

HU (2004) Assessment of pulmonary hypertension by CT and

MR imaging. Eur Radiol 14(3):359–368. doi:10.1007/s00330-

003-2208-x

89. McLaughlin VV, Badesch DB, Delcroix M, Fleming TR, Gaine SP,

Galie N, Gibbs JS, Kim NH, Oudiz RJ, Peacock A, Provencher S,

Sitbon O, Tapson VF, Seeger W (2009) End points and clinical trial

design in pulmonary arterial hypertension. J Am Coll Cardiol 54(1

Suppl):S97–107. doi:10.1016/j.jacc.2009.04.007

90. McLure LE, Peacock AJ (2009) Cardiac magnetic resonance

imaging for the assessment of the heart and pulmonary circu-

lation in pulmonary hypertension. Eur Respir J 33(6):1454–

1466. doi:10.1183/09031936.00139907

91. Boxt LM, Katz J, Kolb T, Czegledy FP, Barst RJ (1992) Direct

quantitation of right and left ventricular volumes with nuclear

magnetic resonance imaging in patients with primary pulmonary

hypertension. J Am Coll Cardiol 19(7):1508–1515 pii: 0735-

1097(92)90611-P

Today’s and tomorrow’s imaging and circulating biomarkers 2825

123

Page 22: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

92. Doherty NE 3rd, Fujita N, Caputo GR, Higgins CB (1992)

Measurement of right ventricular mass in normal and dilated

cardiomyopathic ventricles using cine magnetic resonance

imaging. Am J Cardiol 69(14):1223–1228

93. Katz J, Whang J, Boxt LM, Barst RJ (1993) Estimation of right

ventricular mass in normal subjects and in patients with primary

pulmonary hypertension by nuclear magnetic resonance imag-

ing. J Am Coll Cardiol 21(6):1475–1481 pii: 0735-1097(93)

90327-W

94. Pattynama PM, Willems LN, Smit AH, van der Wall EE, de

Roos A (1992) Early diagnosis of cor pulmonale with MR

imaging of the right ventricle. Radiology 182(2):375–379

95. Suzuki J, Caputo GR, Masui T, Chang JM, O’Sullivan M,

Higgins CB (1991) Assessment of right ventricular diastolic and

systolic function in patients with dilated cardiomyopathy using

cine magnetic resonance imaging. Am Heart J 122(4 Pt 1):

1035–1040

96. van Wolferen SA, Marcus JT, Boonstra A, Marques KM,

Bronzwaer JG, Spreeuwenberg MD, Postmus PE, Vonk-Noo-

rdegraaf A (2007) Prognostic value of right ventricular mass,

volume, and function in idiopathic pulmonary arterial hyper-

tension. Eur Heart J 28(10):1250–1257. doi:10.1093/eurheartj/

ehl477

97. Tulevski II, Lee PL, Groenink M, van der Wall EE, Stoker J,

Pieper PG, Romkes H, Hirsch A, Mulder BJ (2000) Dobuta-

mine-induced increase of right ventricular contractility without

increased stroke volume in adolescent patients with transposi-

tion of the great arteries: evaluation with magnetic resonance

imaging. Int J Card Imaging 16(6):471–478

98. Holverda S, Gan CT, Marcus JT, Postmus PE, Boonstra A,

Vonk-Noordegraaf A (2006) Impaired stroke volume response

to exercise in pulmonary arterial hypertension. J Am Coll Car-

diol 47(8):1732–1733. doi:10.1016/j.jacc.2006.01.048

99. Hachulla AL, Launay D, Gaxotte V, de Groote P, Lamblin N,

Devos P, Hatron PY, Beregi JP, Hachulla E (2009) Cardiac

magnetic resonance imaging in systemic sclerosis: a cross-sec-

tional observational study of 52 patients. Ann Rheum Dis

68(12):1878–1884. doi:10.1136/ard.2008.095836

100. Blyth KG, Groenning BA, Martin TN, Foster JE, Mark PB,

Dargie HJ, Peacock AJ (2005) Contrast enhanced-cardiovascu-

lar magnetic resonance imaging in patients with pulmonary

hypertension. Eur Heart J 26(19):1993–1999. doi:10.1093/

eurheartj/ehi328

101. Gan CT, Lankhaar JW, Westerhof N, Marcus JT, Becker A,

Twisk JW, Boonstra A, Postmus PE, Vonk-Noordegraaf A

(2007) Noninvasively assessed pulmonary artery stiffness pre-

dicts mortality in pulmonary arterial hypertension. Chest

132(6):1906–1912. doi:10.1378/chest.07-1246

102. Jardim C, Rochitte CE, Humbert M, Rubenfeld G, Jasinowo-

dolinski D, Carvalho CR, Souza R (2007) Pulmonary artery

distensibility in pulmonary arterial hypertension: an MRI pilot

study. Eur Respir J 29(3):476–481. doi:10.1183/09031936.0001

6806

103. Bergin CJ, Hauschildt J, Rios G, Belezzuoli EV, Huynh T,

Channick RN (1997) Accuracy of MR angiography compared

with radionuclide scanning in identifying the cause of pulmon-

ary arterial hypertension. Am J Roentgenol 168(6):1549–1555

104. Nikolaou K, Schoenberg SO, Attenberger U, Scheidler J, Die-

trich O, Kuehn B, Rosa F, Huber A, Leuchte H, Baumgartner R,

Behr J, Reiser MF (2005) Pulmonary arterial hypertension:

diagnosis with fast perfusion MR imaging and high-spatial-

resolution MR angiography-preliminary experience. Radiology

236(2):694–703. doi:10.1148/radiol.2361040502

105. Kuehne T, Yilmaz S, Steendijk P, Moore P, Groenink M, Saaed

M, Weber O, Higgins CB, Ewert P, Fleck E, Nagel E, Schulze-

Neick I, Lange P (2004) Magnetic resonance imaging analysis

of right ventricular pressure-volume loops: in vivo validation

and clinical application in patients with pulmonary hyperten-

sion. Circulation 110(14):2010–2016. doi:10.1161/01.CIR.0000

143138.02493.DD

106. Wong YY, Ruiter G, Lubberink M, Raijmakers PG, Knaapen P,

Marcus JT, Boonstra A, Lammertsma AA, Westerhof N, van der

Laarse WJ, Vonk-Noordegraaf A (2011) Right ventricular fail-

ure in idiopathic pulmonary arterial hypertension is associated

with inefficient myocardial oxygen utilization. Circ Heart Fail

4(6):700–706. doi:10.1161/CIRCHEARTFAILURE.111.962381

107. Ohno Y, Koyama H, Nogami M, Takenaka D, Matsumoto S,

Onishi Y, Matsumoto K, Murase K, Sugimura K (2008)

Dynamic perfusion MRI: capability for evaluation of disease

severity and progression of pulmonary arterial hypertension in

patients with connective tissue disease. J Magn Reson Imaging

28(4):887–899. doi:10.1002/jmri.21550

108. Ohno Y, Hatabu H, Murase K, Higashino T, Nogami M, Yos-

hikawa T, Sugimura K (2007) Primary pulmonary hypertension:

3D dynamic perfusion MRI for quantitative analysis of regional

pulmonary perfusion. Am J Roentgenol 188(1):48–56. doi:

10.2214/AJR.05.0135

109. Okajima Y, Ohno Y, Washko GR, Hatabu H (2011) Assessment

of pulmonary hypertension what CT and MRI can provide. Acad

Radiol 18(4):437–453. doi:10.1016/j.acra.2011.01.003

110. Kondo C, Caputo GR, Masui T, Foster E, O’Sullivan M, Stul-

barg MS, Golden J, Catterjee K, Higgins CB (1992) Pulmonary

hypertension: pulmonary flow quantification and flow profile

analysis with velocity-encoded cine MR imaging. Radiology

183(3):751–758

111. Roeleveld RJ, Marcus JT, Boonstra A, Postmus PE, Marques

KM, Bronzwaer JG, Vonk-Noordegraaf A (2005) A comparison

of noninvasive MRI-based methods of estimating pulmonary

artery pressure in pulmonary hypertension. J Magn Reson

Imaging 22(1):67–72. doi:10.1002/jmri.20338

112. Roeleveld RJ, Vonk-Noordegraaf A, Marcus JT, Bronzwaer JG,

Marques KM, Postmus PE, Boonstra A (2004) Effects of epo-

prostenol on right ventricular hypertrophy and dilatation in

pulmonary hypertension. Chest 125(2):572–579

113. Chin KM, Kingman M, de Lemos JA, Warner JJ, Reimold S,

Peshock R, Torres F (2008) Changes in right ventricular struc-

ture and function assessed using cardiac magnetic resonance

imaging in bosentan-treated patients with pulmonary arterial

hypertension. Am J Cardiol 101(11):1669–1672. doi:10.1016/

j.amjcard.2008.01.055

114. Michelakis ED, Tymchak W, Noga M, Webster L, Wu XC, Lien

D, Wang SH, Modry D, Archer SL (2003) Long-term treatment

with oral sildenafil is safe and improves functional capacity and

hemodynamics in patients with pulmonary arterial hypertension.

Circulation 108(17):2066–2069. doi:10.1161/01.CIR.00000995

02.17776.C2

115. Enders J, Zimmermann E, Rief M, Martus P, Klingebiel R,

Asbach P, Klessen C, Diederichs G, Bengner T, Teichgraber U,

Hamm B, Dewey M (2011) Reduction of claustrophobia during

magnetic resonance imaging: methods and design of the

‘‘CLAUSTRO’’ randomized controlled trial. BMC Med Imaging

11:4. doi:10.1186/1471-2342-11-4

116. Kuijpers D, van Dijkman PR, Janssen CH, Vliegenthart R, Zi-

jlstra F, Oudkerk M (2004) Dobutamine stress MRI. Part II. Risk

stratification with dobutamine cardiovascular magnetic reso-

nance in patients suspected of myocardial ischemia. Eur Radiol

14(11):2046–2052. doi:10.1007/s00330-004-2426-x

117. Gan CT, McCann GP, Marcus JT, van Wolferen SA, Twisk JW,

Boonstra A, Postmus PE, Vonk-Noordegraaf A (2006) NT-

proBNP reflects right ventricular structure and function in pul-

monary hypertension. Eur Respir J 28(6):1190–1194. doi:

10.1183/09031936.00016006

2826 M. Barrier et al.

123

Page 23: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

118. Tsutamoto T, Wada A, Maeda K, Hisanaga T, Maeda Y, Fukai

D, Ohnishi M, Sugimoto Y, Kinoshita M (1997) Attenuation of

compensation of endogenous cardiac natriuretic peptide system

in chronic heart failure: prognostic role of plasma brain natri-

uretic peptide concentration in patients with chronic

symptomatic left ventricular dysfunction. Circulation 96(2):

509–516

119. Nagaya N, Nishikimi T, Uematsu M, Satoh T, Kyotani S,

Sakamaki F, Kakishita M, Fukushima K, Okano Y, Nakanishi N,

Miyatake K, Kangawa K (2000) Plasma brain natriuretic peptide

as a prognostic indicator in patients with primary pulmonary

hypertension. Circulation 102(8):865–870

120. Nagaya N, Ando M, Oya H, Ohkita Y, Kyotani S, Sakamaki F,

Nakanishi N (2002) Plasma brain natriuretic peptide as a nonin-

vasive marker for efficacy of pulmonary thromboendarterectomy.

Ann Thorac Surg 74(1):180–184 (discussion 184)

121. Blyth KG, Groenning BA, Mark PB, Martin TN, Foster JE,

Steedman T, Morton JJ, Dargie HJ, Peacock AJ (2007)

NT-proBNP can be used to detect right ventricular systolic

dysfunction in pulmonary hypertension. Eur Respir J 29(4):737–

744. doi:10.1183/09031936.00095606

122. Fijalkowska A, Kurzyna M, Torbicki A, Szewczyk G, Florczyk

M, Pruszczyk P, Szturmowicz M (2006) Serum N-terminal brain

natriuretic peptide as a prognostic parameter in patients with

pulmonary hypertension. Chest 129(5):1313–1321. doi:10.1378/

chest.129.5.1313

123. Andreassen AK, Wergeland R, Simonsen S, Geiran O, Guevara

C, Ueland T (2006) N-terminal pro-B-type natriuretic peptide as

an indicator of disease severity in a heterogeneous group of

patients with chronic precapillary pulmonary hypertension. Am

J Cardiol 98(4):525–529. doi:10.1016/j.amjcard.2006.02.061

124. Giannakoulas G, Dimopoulos K, Bolger AP, Tay EL, Inuzuka R,

Bedard E, Davos C, Swan L, Gatzoulis MA (2010) Usefulness

of natriuretic Peptide levels to predict mortality in adults with

congenital heart disease. Am J Cardiol 105(6):869–873. doi:

10.1016/j.amjcard.2009.11.041

125. Lang CC, Coutie WJ, Struthers AD, Dhillon DP, Winter JH,

Lipworth BJ (1992) Elevated levels of brain natriuretic peptide

in acute hypoxaemic chronic obstructive pulmonary disease.

Clin Sci (Lond) 83(5):529–533

126. Shah SJ, Thenappan T, Rich S, Tian L, Archer SL, Gomberg-

Maitland M (2008) Association of serum creatinine with

abnormal hemodynamics and mortality in pulmonary arterial

hypertension. Circulation 117(19):2475–2483. doi:10.1161/

CIRCULATIONAHA.107.719500

127. Torbicki A, Kurzyna M, Kuca P, Fijalkowska A, Sikora J,

Florczyk M, Pruszczyk P, Burakowski J, Wawrzynska L (2003)

Detectable serum cardiac troponin T as a marker of poor

prognosis among patients with chronic precapillary pulmonary

hypertension. Circulation 108(7):844–848. doi:10.1161/01.CIR.

0000084544.54513.E2

128. Sitbon O, Humbert M, Nunes H, Parent F, Garcia G, Herve P,

Rainisio M, Simonneau G (2002) Long-term intravenous epo-

prostenol infusion in primary pulmonary hypertension:

prognostic factors and survival. J Am Coll Cardiol 40(4):780–

788 pii: 0735109702020120

129. La Vecchia L, Mezzena G, Zanolla L, Paccanaro M, Varotto L,

Bonanno C, Ometto R (2000) Cardiac troponin I as diagnostic

and prognostic marker in severe heart failure. J Heart Lung

Transplant 19(7):644–652 pii: S1053-2498(00)00120-0

130. Sato Y, Yamada T, Taniguchi R, Kataoka K, Sasayama S,

Matsumori A, Takatsu Y (1998) Serum concentration of cardiac

troponin T in patients with cardiomyopathy: a possible mecha-

nism of acute heart failure. Heart 80(2):209–210

131. Lorenzen JM, Nickel N, Kramer R, Golpon H, Westerkamp V,

Olsson KM, Haller H, Hoeper MM (2011) Osteopontin in

patients with idiopathic pulmonary hypertension. Chest

139(5):1010-1017. doi:10.1378/chest.10-1146

132. McDonald LJ, Murad F (1996) Nitric oxide and cyclic GMP

signaling. Proc Soc Exp Biol Med 211(1):1–6

133. Bogdan M, Humbert M, Francoual J, Claise C, Duroux P, Si-

monneau G, Lindenbaum A (1998) Urinary cGMP concentrations

in severe primary pulmonary hypertension. Thorax 53(12):1059–

1062

134. Giaid A, Saleh D (1995) Reduced expression of endothelial

nitric oxide synthase in the lungs of patients with pulmonary

hypertension. N Engl J Med 333(4):214–221. doi:10.1056/

NEJM199507273330403

135. Wiedemann R, Ghofrani HA, Weissmann N, Schermuly R,

Quanz K, Grimminger F, Seeger W, Olschewski H (2001) Atrial

natriuretic peptide in severe primary and nonprimary pulmonary

hypertension: response to iloprost inhalation. J Am Coll Cardiol

38(4):1130–1136 pii: S0735-1097(01)01490-5

136. Boulanger CM, Amabile N, Tedgui A (2006) Circulating mi-

croparticles: a potential prognostic marker for atherosclerotic

vascular disease. Hypertension 48(2):180–186. doi:10.1161/01.

HYP.0000231507.00962.b5

137. Amabile N, Guerin AP, Leroyer A, Mallat Z, Nguyen C,

Boddaert J, London GM, Tedgui A, Boulanger CM (2005)

Circulating endothelial microparticles are associated with

vascular dysfunction in patients with end-stage renal failure.

J Am Soc Nephrol 16(11):3381–3388. doi:10.1681/ASN.20050

50535

138. Amabile N, Heiss C, Real WM, Minasi P, McGlothlin D, Rame

EJ, Grossman W, De Marco T, Yeghiazarians Y (2008) Circu-

lating endothelial microparticle levels predict hemodynamic

severity of pulmonary hypertension. Am J Respir Crit Care Med

177(11):1268–1275. doi:10.1164/rccm.200710-1458OC

139. Bakouboula B, Morel O, Faure A, Zobairi F, Jesel L, Trinh A,

Zupan M, Canuet M, Grunebaum L, Brunette A, Desprez D,

Chabot F, Weitzenblum E, Freyssinet JM, Chaouat A, Toti F

(2008) Procoagulant membrane microparticles correlate with the

severity of pulmonary arterial hypertension. Am J Respir Crit

Care Med 177(5):536–543. doi:10.1164/rccm.200706-840OC

140. Tual-Chalot S, Guibert C, Muller B, Savineau JP, Andrian-

tsitohaina R, Martinez MC (2010) Circulating microparticles

from pulmonary hypertensive rats induce endothelial dysfunc-

tion. Am J Respir Crit Care Med 182(2):261–268. doi:

10.1164/rccm.200909-1347OC

141. Pfister SL (2004) Role of platelet microparticles in the pro-

duction of thromboxane by rabbit pulmonary artery.

Hypertension 43(2):428–433. doi:10.1161/01.HYP.0000110906.

77479.91

142. Dogne JM, Hanson J, Pratico D (2005) Thromboxane, prosta-

cyclin and isoprostanes: therapeutic targets in atherogenesis.

Trends Pharmacol Sci 26(12):639–644. doi:10.1016/j.tips.

2005.10.001

143. Christman BW, McPherson CD, Newman JH, King GA, Ber-

nard GR, Groves BM, Loyd JE (1992) An imbalance between

the excretion of thromboxane and prostacyclin metabolites in

pulmonary hypertension. N Engl J Med 327(2):70–75. doi:

10.1056/NEJM199207093270202

144. Veyradier A, Nishikubo T, Humbert M, Wolf M, Sitbon O,

Simonneau G, Girma JP, Meyer D (2000) Improvement of von

Willebrand factor proteolysis after prostacyclin infusion in

severe pulmonary arterial hypertension. Circulation 102(20):

2460–2462

145. Kawut SM, Horn EM, Berekashvili KK, Widlitz AC, Rosen-

zweig EB, Barst RJ (2005) von Willebrand factor independently

predicts long-term survival in patients with pulmonary arterial

hypertension. Chest 128(4):2355–2362. doi:10.1378/chest.128.

4.2355

Today’s and tomorrow’s imaging and circulating biomarkers 2827

123

Page 24: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

146. Shitrit D, Bendayan D, Bar-Gil-Shitrit A, Huerta M, Rudensky

B, Fink G, Kramer MR (2002) Significance of a plasma D-dimer

test in patients with primary pulmonary hypertension. Chest

122(5):1674–1678

147. Shitrit D, Bendayan D, Rudensky B, Izbicki G, Huerta M, Fink

G, Kramer MR (2002) Elevation of ELISA d-dimer levels in

patients with primary pulmonary hypertension. Respiration

69(4):327–329 pii: 63270

148. Kiatchoosakun S, Ungkasekvinai W, Wonvipaporn C, Tatsa-

navivat P, Foocharoen C, Suwannaroj S, Nanagara R (2007)

D-dimer and pulmonary arterial hypertension in systemic scle-

rosis. J Med Assoc Thai 90(10):2024–2029

149. Nickel N, Kempf T, Tapken H, Tongers J, Laenger F, Lehmann

U, Golpon H, Olsson K, Wilkins MR, Gibbs JS, Hoeper MM,

Wollert KC (2008) Growth differentiation factor-15 in idio-

pathic pulmonary arterial hypertension. Am J Respir Crit Care

Med 178(5):534–541. doi:10.1164/rccm.200802-235OC

150. Meadows CA, Risbano MG, Zhang L, Geraci MW, Tuder RM,

Collier DH, Bull TM Increased expression of growth differen-

tiation factor-15 in systemic sclerosis-associated pulmonary

arterial hypertension. Chest 139 (5):994–1002. doi:10.1378/chest.

10-0302

151. Nickel N, Jonigk D, Kempf T, Bockmeyer CL, Maegel L, Ri-

sche J, Laenger F, Lehmann U, Sauer C, Greer M, Welte T,

Hoeper MM, Golpon HA GDF-15 is abundantly expressed in

plexiform lesions in patients with pulmonary arterial hyperten-

sion and affects proliferation and apoptosis of pulmonary

endothelial cells. Respir Res 12:62. doi:10.1186/1465-9921-

12-62

152. Cacoub P, Dorent R, Nataf P, Carayon A, Riquet M, Noe E,

Piette JC, Godeau P, Gandjbakhch I (1997) Endothelin-1 in the

lungs of patients with pulmonary hypertension. Cardiovasc Res

33(1):196–200 pii: S0008-6363(96)00189-7

153. Rubens C, Ewert R, Halank M, Wensel R, Orzechowski HD,

Schultheiss HP, Hoeffken G (2001) Big endothelin-1 and

endothelin-1 plasma levels are correlated with the severity of

primary pulmonary hypertension. Chest 120(5):1562–1569

154. Stewart DJ, Levy RD, Cernacek P, Langleben D (1991)

Increased plasma endothelin-1 in pulmonary hypertension:

marker or mediator of disease? Ann Intern Med 114(6):464–469

155. Cody RJ, Haas GJ, Binkley PF, Capers Q, Kelley R (1992)

Plasma endothelin correlates with the extent of pulmonary

hypertension in patients with chronic congestive heart failure.

Circulation 85(2):504–509

156. Giaid A, Yanagisawa M, Langleben D, Michel RP, Levy R,

Shennib H, Kimura S, Masaki T, Duguid WP, Stewart DJ (1993)

Expression of endothelin-1 in the lungs of patients with pul-

monary hypertension. N Engl J Med 328(24):1732–1739. doi:

10.1056/NEJM199306173282402

157. Barst RJ, Langleben D, Frost A, Horn EM, Oudiz R, Shapiro S,

McLaughlin V, Hill N, Tapson VF, Robbins IM, Zwicke D,

Duncan B, Dixon RA, Frumkin LR (2004) Sitaxsentan therapy

for pulmonary arterial hypertension. Am J Respir Crit Care Med

169(4):441–447. doi:10.1164/rccm.200307-957OC

158. Rubin LJ, Badesch DB, Barst RJ, Galie N, Black CM, Keogh A,

Pulido T, Frost A, Roux S, Leconte I, Landzberg M, Simonneau

G (2002) Bosentan therapy for pulmonary arterial hypertension.

N Engl J Med 346(12):896–903. doi:10.1056/NEJMoa012212

159. Galie N, Olschewski H, Oudiz RJ, Torres F, Frost A, Ghofrani

HA, Badesch DB, McGoon MD, McLaughlin VV, Roecker EB,

Gerber MJ, Dufton C, Wiens BL, Rubin LJ (2008) Ambrisentan

for the treatment of pulmonary arterial hypertension: results of

the ambrisentan in pulmonary arterial hypertension, randomized,

double-blind, placebo-controlled, multicenter, efficacy (ARIES)

study 1 and 2. Circulation 117(23):3010–3019. doi:

10.1161/CIRCULATIONAHA.107.742510

160. Aubert JD (2005) Biochemical markers in the management of

pulmonary hypertension. Swiss Med Wkly 135(3–4):43–49. doi:

2005/03/smw-10723

161. Montani D, Souza R, Binkert C, Fischli W, Simonneau G,

Clozel M, Humbert M (2007) Endothelin-1/endothelin-3 ratio: a

potential prognostic factor of pulmonary arterial hypertension.

Chest 131(1):101–108. doi:10.1378/chest.06-0682

162. Otterdal K, Andreassen AK, Yndestad A, Oie E, Sandberg WJ,

Dahl CP, Pedersen TM, Ueland T, Gullestad L, Brosstad FR,

Aukrust P, Damas JK (2008) Raised LIGHT levels in pulmonary

arterial hypertension: potential role in thrombus formation. Am J

Respir Crit Care Med 177(2):202–207. doi:10.1164/rccm.2007

03-506OC

163. Quarck R, Nawrot T, Meyns B, Delcroix M (2009) C-reactive

protein: a new predictor of adverse outcome in pulmonary

arterial hypertension. J Am Coll Cardiol 53(14):1211–1218. doi:

10.1016/j.jacc.2008.12.038

164. Sztrymf B, Souza R, Bertoletti L, Jais X, Sitbon O, Price LC,

Simonneau G, Humbert M (2010) Prognostic factors of acute

heart failure in patients with pulmonary arterial hypertension.

Eur Respir J 35(6):1286–1293. doi:10.1183/09031936.00070209

165. Paulin R, Courboulin A, Barrier M, Bonnet S (2011) From

oncoproteins/tumor suppressors to microRNAs, the newest

therapeutic targets for pulmonary arterial hypertension. J Mol

Med (Berl). doi:10.1007/s00109-011-0788-5

166. Soon E, Holmes AM, Treacy CM, Doughty NJ, Southgate L,

Machado RD, Trembath RC, Jennings S, Barker L, Nicklin P,

Walker C, Budd DC, Pepke-Zaba J, Morrell NW Elevated levels

of inflammatory cytokines predict survival in idiopathic

and familial pulmonary arterial hypertension. Circulation

122(9):920–927. doi:10.1161/CIRCULATIONAHA.109.933762

167. Humbert M, Monti G, Fartoukh M, Magnan A, Brenot F, Rain

B, Capron F, Galanaud P, Duroux P, Simonneau G, Emilie D

(1998) Platelet-derived growth factor expression in primary

pulmonary hypertension: comparison of HIV seropositive and

HIV seronegative patients. Eur Respir J 11(3):554–559

168. Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S, Savai R,

Roth M, Sydykov A, Lai YJ, Weissmann N, Seeger W,

Grimminger F (2005) Reversal of experimental pulmonary

hypertension by PDGF inhibition. J Clin Invest 115(10):

2811–2821. doi:10.1172/JCI24838

169. Csiszar A, Labinskyy N, Olson S, Pinto JT, Gupte S, Wu JM, Hu F,

Ballabh P, Podlutsky A, Losonczy G, de Cabo R, Mathew R, Wolin

MS, Ungvari Z (2009) Resveratrol prevents monocrotaline-induced

pulmonary hypertension in rats. Hypertension 54(3):668–675. doi:

10.1161/HYPERTENSIONAHA.109.133397

170. Frasch HF, Marshall C, Marshall BE (1999) Endothelin-1 is

elevated in monocrotaline pulmonary hypertension. Am J

Physiol 276(2 Pt 1):L304–310

171. Gonzalez RM, Daly DS, Tan R, Marks JR, Zangar RC Plasma

Biomarker Profiles Differ Depending on Breast Cancer Subtype

but RANTES Is Consistently Increased. Cancer Epidemiol

Biomarkers Prev 20(7):1543–1551. doi:10.1158/1055-9965.

EPI-10-1248

172. Nastase A, Paslaru L, Niculescu AM, Ionescu M, Dumitrascu T,

Herlea V, Dima S, Gheorghe C, Lazar V, Popescu I (2011)

Prognostic and predictive potential molecular biomarkers in

colon cancer. Chirurgia (Bucur) 106(2):177–185

173. Klebe S, Henderson DW (2011) Early stages of mesothelioma,

screening and biomarkers. Recent Results Cancer Res

189:169–193. doi:10.1007/978-3-642-10862-4_10

174. Tronov VA, Artamonov DN, Abramov ME, Gorbacheva LB

(2011) Cellular markers based on DNA damage and repair

(BER, MMR), expression of MLHI, MSH2, FasR, and cell death

of lymphocytes as predictive parameters for clinical response to

chemotherapy of melanoma. Tsitologiia 53(1):10–16

2828 M. Barrier et al.

123

Page 25: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

175. Fouty BW, Grimison B, Fagan KA, Le Cras TD, Harral JW,

Hoedt-Miller M, Sclafani RA, Rodman DM (2001) p27(Kip1) is

important in modulating pulmonary artery smooth muscle cell

proliferation. Am J Respir Cell Mol Biol 25(5):652–658

176. Yu L, Quinn DA, Garg HG, Hales CA (2005) Cyclin-dependent

kinase inhibitor p27Kip1, but not p21WAF1/Cip1, is required

for inhibition of hypoxia-induced pulmonary hypertension and

remodeling by heparin in mice. Circ Res 97(9):937–945. doi:

10.1161/01.RES.0000188211.83193.1a

177. Yang C, Wang S, Liang T, Wang J, Wang K, Wang B (2001)

The roles of bcl-2 gene family in the pulmonary artery remod-

eling of hypoxia pulmonary hypertension in rats. Chin Med Sci J

16(3):182–184

178. Moudgil R, Michelakis ED, Archer SL (2006) The role of k?

channels in determining pulmonary vascular tone, oxygen

sensing, cell proliferation, and apoptosis: implications in hyp-

oxic pulmonary vasoconstriction and pulmonary arterial

hypertension. Microcirculation 13(8):615–632. doi:10.1080/

10739680600930222

179. Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A Smad

proteins bind a conserved RNA sequence to promote microRNA

maturation by Drosha. Mol Cell 39 (3):373–384. doi:10.1016/

j.molcel.2010.07.011

180. Hatton N, Frech T, Smith B, Sawitzke A, Scholand MB,

Markewitz B Transforming growth factor signalling: a common

pathway in pulmonary arterial hypertension and systemic scle-

rosis. Int J Clin Pract 65 Suppl 172:35–43. doi:10.1111/j.1742-

1241.2011.02726.x

181. Liu XG, Zhu WY, Huang YY, Ma LN, Zhou SQ, Wang YK,

Zeng F, Zhou JH, Zhang YK (2011) High expression of serum

miR-21 and tumor miR-200c associated with poor prognosis in

patients with lung cancer. Med Oncol. doi:10.1007/s12032-011-

9923-y

182. Wang M, Li W, Chang GQ, Ye CS, Ou JS, Li XX, Liu Y,

Cheang TY, Huang XL, Wang SM (2011) MicroRNA-21 Reg-

ulates Vascular Smooth Muscle Cell Function via Targeting

Tropomyosin 1 in Arteriosclerosis Obliterans of Lower

Extremities. Arterioscler Thromb Vasc Biol 31 (9):2044–2053.

doi:10.1161/ATVBAHA.111.229559

183. Sarkar J, Gou D, Turaka P, Viktorova E, Ramchandran R, Raj

JU (2010) MicroRNA-21 plays a role in hypoxia-mediated

pulmonary artery smooth muscle cell proliferation and migra-

tion. Am J Physiol Lung Cell Mol Physiol 299(6):L861–871.

doi:10.1152/ajplung.00201.2010

184. Caruso P, Maclean MR, Khanin R, McClure J, Soon E, South-

wood M, McDonald RA, Greig JA, Robertson KE, Masson R,

Denby L, Dempsie Y, Long L, Morrell NW, Baker AH (2010)

Dynamic changes in lung MicroRNA profiles during the

development of pulmonary hypertension due to chronic hypoxia

and monocrotaline. Arterioscler Thromb Vasc Biol. doi:

10.1161/ATVBAHA.109.202028

185. Huang X, Ding L, Bennewith KL, Tong RT, Welford SM, Ang

KK, Story M, Le QT, Giaccia AJ (2009) Hypoxia-inducible mir-

210 regulates normoxic gene expression involved in tumor ini-

tiation. Mol Cell 35(6):856–867. doi:10.1016/j.molcel.2009.09.

006

186. Fasanaro P, Greco S, Lorenzi M, Pescatori M, Brioschi M,

Kulshreshtha R, Banfi C, Stubbs A, Calin GA, Ivan M, Ca-

pogrossi MC, Martelli F (2009) An integrated approach for

experimental target identification of hypoxia-induced miR-210.

J Biol Chem 284(50):35134–35143. doi:10.1074/jbc.M109.

052779

187. Giannakakis A, Sandaltzopoulos R, Greshock J, Liang S, Huang

J, Hasegawa K, Li C, O’Brien-Jenkins A, Katsaros D, Weber

BL, Simon C, Coukos G, Zhang L (2008) miR-210 links

hypoxia with cell cycle regulation and is deleted in human

epithelial ovarian cancer. Cancer Biol Ther 7(2):255–264

pii:5297

188. Chen Z, Li Y, Zhang H, Huang P, Luthra R (2010) Hypoxia-

regulated microRNA-210 modulates mitochondrial function and

decreases ISCU and COX10 expression. Oncogene 29(30):

4362–4368. doi:10.1038/onc.2010.193

189. Drebber U, Lay M, Wedemeyer I, Vallbohmer D, Bollschweiler

E, Brabender J, Monig SP, Holscher AH, Dienes HP, Odenthal

M (2011) Altered levels of the onco-microRNA 21 and the

tumor-supressor microRNAs 143 and 145 in advanced rectal

cancer indicate successful neoadjuvant chemoradiotherapy. Int J

Oncol 39(2):409–415. doi:10.3892/ijo.2011.1036

190. Cheng Y, Liu X, Yang J, Lin Y, Xu DZ, Lu Q, Deitch EA, Huo

Y, Delphin ES, Zhang C (2009) MicroRNA-145, a novel smooth

muscle cell phenotypic marker and modulator, controls vascular

neointimal lesion formation. Circ Res 105(2):158–166. doi:

10.1161/CIRCRESAHA.109.197517

191. McDonald JS, Milosevic D, Reddi HV, Grebe SK, Algeciras-

Schimnich A (2011) Analysis of Circulating MicroRNA: pre-

analytical and analytical challenges. Clin Chem. doi:10.1373/

clinchem.2010.157198

192. Chen Y, Zhang H, Xu A, Li N, Liu J, Liu C, Lv D, Wu S, Huang

L, Yang S, He D, Xiao X (2006) Elevation of serum l-lactate

dehydrogenase B correlated with the clinical stage of lung

cancer. Lung Cancer 54(1):95–102. doi:10.1016/j.lungcan.2006.

06.014

193. Kim K, Taylor SL, Ganti S, Guo L, Osier MV, Weiss RH (2011)

Urine metabolomic analysis identifies potential biomarkers and

pathogenic pathways in kidney cancer. OMICS 15(5):293–303.

doi:10.1089/omi.2010.0094

194. Hoshikawa Y, Ono S, Suzuki S, Tanita T, Chida M, Song C, Noda

M, Tabata T, Voelkel NF, Fujimura S (2001) Generation of oxi-

dative stress contributes to the development of pulmonary

hypertension induced by hypoxia. J Appl Physiol 90(4):1299–1306

195. Al-Mehdi AB, Zhao G, Dodia C, Tozawa K, Costa K, Muzy-

kantov V, Ross C, Blecha F, Dinauer M, Fisher AB (1998)

Endothelial NADPH oxidase as the source of oxidants in lungs

exposed to ischemia or high K?. Circ Res 83(7):730–737

196. Tuder RM, Chacon M, Alger L, Wang J, Taraseviciene-Stewart

L, Kasahara Y, Cool CD, Bishop AE, Geraci M, Semenza GL,

Yacoub M, Polak JM, Voelkel NF (2001) Expression of angi-

ogenesis-related molecules in plexiform lesions in severe

pulmonary hypertension: evidence for a process of disordered

angiogenesis. J Pathol 195(3):367–374. doi:10.1002/path.953

197. Humbert M, Monti G, Brenot F, Sitbon O, Portier A, Grangeot-

Keros L, Duroux P, Galanaud P, Simonneau G, Emilie D (1995)

Increased interleukin-1 and interleukin-6 serum concentrations

in severe primary pulmonary hypertension. Am J Respir Crit

Care Med 151(5):1628–1631

198. Bowers R, Cool C, Murphy RC, Tuder RM, Hopken MW, Flores

SC, Voelkel NF (2004) Oxidative stress in severe pulmonary

hypertension. Am J Respir Crit Care Med 169(6):764–769. doi:

10.1164/rccm.200301-147OC

199. Cracowski JL, Cracowski C, Bessard G, Pepin JL, Bessard J,

Schwebel C, Stanke-Labesque F, Pison C (2001) Increased lipid

peroxidation in patients with pulmonary hypertension. Am J

Respir Crit Care Med 164(6):1038–1042

200. Kaneko FT, Arroliga AC, Dweik RA, Comhair SA, Laskowski

D, Oppedisano R, Thomassen MJ, Erzurum SC (1998) Bio-

chemical reaction products of nitric oxide as quantitative

markers of primary pulmonary hypertension. Am J Respir Crit

Care Med 158(3):917–923

201. Rhoades RA, Packer CS, Roepke DA, Jin N, Meiss RA (1990)

Reactive oxygen species alter contractile properties of pul-

monary arterial smooth muscle. Can J Physiol Pharmacol

68(12):1581–1589

Today’s and tomorrow’s imaging and circulating biomarkers 2829

123

Page 26: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

202. Demiryurek AT, Wadsworth RM (1999) Superoxide in the

pulmonary circulation. Pharmacol Ther 84(3):355–365

203. Gupte SA, Kaminski PM, Floyd B, Agarwal R, Ali N, Ahmad

M, Edwards J, Wolin MS (2005) Cytosolic NADPH may reg-

ulate differences in basal Nox oxidase-derived superoxide

generation in bovine coronary and pulmonary arteries. Am J

Physiol Heart Circ Physiol 288(1):H13–21. doi:10.1152/ajpheart.

00629.2004

204. Redout EM, Wagner MJ, Zuidwijk MJ, Boer C, Musters RJ, van

Hardeveld C, Paulus WJ, Simonides WS (2007) Right-ventric-

ular failure is associated with increased mitochondrial complex

II activity and production of reactive oxygen species. Cardio-

vasc Res 75(4):770–781. doi:10.1016/j.cardiores.2007.05.012

205. Farahmand F, Hill MF, Singal PK (2004) Antioxidant and oxi-

dative stress changes in experimental cor pulmonale. Mol Cell

Biochem 260(1–2):21–29

206. DeMarco VG, Habibi J, Whaley-Connell AT, Schneider RI,

Heller RL, Bosanquet JP, Hayden MR, Delcour K, Cooper SA,

Andresen BT, Sowers JR, Dellsperger KC (2008) Oxidative

stress contributes to pulmonary hypertension in the transgenic

(mRen2)27 rat. Am J Physiol Heart Circ Physiol 294(6):

H2659–2668. doi:10.1152/ajpheart.00953.2007

207. Voelkel MA, Wynne KM, Badesch DB, Groves BM, Voelkel

NF (2000) Hyperuricemia in severe pulmonary hypertension.

Chest 117(1):19–24

208. Nagaya N, Uematsu M, Satoh T, Kyotani S, Sakamaki F,

Nakanishi N, Yamagishi M, Kunieda T, Miyatake K (1999)

Serum uric acid levels correlate with the severity and the mor-

tality of primary pulmonary hypertension. Am J Respir Crit Care

Med 160(2):487–492

209. Van Albada ME, Loot FG, Fokkema R, Roofthooft MT, Berger

RM (2008) Biological serum markers in the management of

pediatric pulmonary arterial hypertension. Pediatr Res

63(3):321–327. doi:10.1203/PDR.0b013e318163a2e7

210. Davi G, Ciabattoni G, Consoli A, Mezzetti A, Falco A, Santa-

rone S, Pennese E, Vitacolonna E, Bucciarelli T, Costantini F,

Capani F, Patrono C (1999) In vivo formation of 8-iso-prosta-

glandin f2alpha and platelet activation in diabetes mellitus:

effects of improved metabolic control and vitamin E supple-

mentation. Circulation 99(2):224–229

211. Pratico D, Barry OP, Lawson JA, Adiyaman M, Hwang SW,

Khanapure SP, Iuliano L, Rokach J, FitzGerald GA (1998)

IPF2alpha-I: an index of lipid peroxidation in humans. Proc Natl

Acad Sci USA 95(7):3449–3454

212. Jankov RP, Luo X, Cabacungan J, Belcastro R, Frndova H, Lye

SJ, Tanswell AK (2000) Endothelin-1 and O2-mediated pul-

monary hypertension in neonatal rats: a role for products of lipid

peroxidation. Pediatr Res 48(3):289–298

213. Janssen LJ (2008) Isoprostanes and lung vascular pathology. Am

J Respir Cell Mol Biol 39(4):383–389. doi:10.1165/rcmb.

2008-0109TR

214. Cracowski JL, Durand T, Bessard G (2002) Isoprostanes as a

biomarker of lipid peroxidation in humans: physiology, phar-

macology and clinical implications. Trends Pharmacol Sci

23(8):360–366 pii: S0165614702020539

215. Waugh RJ, Morrow JD, Roberts LJ 2nd, Murphy RC (1997)

Identification and relative quantitation of F2-isoprostane re-

gioisomers formed in vivo in the rat. Free Radic Biol Med

23(6):943–954 pii: S0891-5849(97)00133-0

216. Kanai Y, Hori S, Tanaka T, Yasuoka M, Watanabe K, Aikawa

N, Hosoda Y (1993) Role of 5-hydroxytryptamine in the pro-

gression of monocrotaline induced pulmonary hypertension in

rats. Cardiovasc Res 27(9):1619–1623

217. Herve P, Launay JM, Scrobohaci ML, Brenot F, Simonneau G,

Petitpretz P, Poubeau P, Cerrina J, Duroux P, Drouet L (1995)

Increased plasma serotonin in primary pulmonary hypertension.

Am J Med 99(3):249–254 pii: S0002934399801569

218. Lee SL, Wang WW, Fanburg BL (1998) Superoxide as an

intermediate signal for serotonin-induced mitogenesis. Free

Radic Biol Med 24(5):855–858 pii: S0891-5849(97)00359-6

219. Lee SL, Wang WW, Finlay GA, Fanburg BL (1999) Serotonin

stimulates mitogen-activated protein kinase activity through the

formation of superoxide anion. Am J Physiol 277(2 Pt

1):L282–291

220. Bianchi P, Kunduzova O, Masini E, Cambon C, Bani D, Rai-

mondi L, Seguelas MH, Nistri S, Colucci W, Leducq N, Parini A

(2005) Oxidative stress by monoamine oxidase mediates

receptor-independent cardiomyocyte apoptosis by serotonin and

postischemic myocardial injury. Circulation 112(21):

3297–3305. doi:10.1161/CIRCULATIONAHA.104.528133

221. Wong CM, Cheema AK, Zhang L, Suzuki YJ (2008) Protein

carbonylation as a novel mechanism in redox signaling. Circ Res

102(3):310–318. doi:10.1161/CIRCRESAHA.107.159814

222. MacLean MR, Herve P, Eddahibi S, Adnot S (2000)

5-hydroxytryptamine and the pulmonary circulation: receptors,

transporters and relevance to pulmonary arterial hypertension.

Br J Pharmacol 131(2):161–168. doi:10.1038/sj.bjp.0703570

223. Hironaka E, Hongo M, Sakai A, Mawatari E, Terasawa F,

Okumura N, Yamazaki A, Ushiyama Y, Yazaki Y, Kinoshita O

(2003) Serotonin receptor antagonist inhibits monocrotaline-

induced pulmonary hypertension and prolongs survival in rats.

Cardiovasc Res 60(3):692–699 pii: S0008636303006539

224. Guignabert C, Raffestin B, Benferhat R, Raoul W, Zadigue P,

Rideau D, Hamon M, Adnot S, Eddahibi S (2005) Seroto-

nin transporter inhibition prevents and reverses monocrotaline-

induced pulmonary hypertension in rats. Circulation 111(21):

2812–2819. doi:10.1161/CIRCULATIONAHA.104.524926

225. Hummel SG, Fischer AJ, Martin SM, Schafer FQ, Buettner GR

(2006) Nitric oxide as a cellular antioxidant: a little goes a long

way. Free Radic Biol Med 40(3):501–506. doi:10.1016/j.

freeradbiomed.2005.08.047

226. Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA

(1990) Apparent hydroxyl radical production by peroxynitrite:

implications for endothelial injury from nitric oxide and super-

oxide. Proc Natl Acad Sci USA 87(4):1620–1624

227. Joshi MS, Ponthier JL, Lancaster JR Jr (1999) Cellular antiox-

idant and pro-oxidant actions of nitric oxide. Free Radic Biol

Med 27(11–12):1357–1366 pii: S0891-5849(99)00179-3

228. Shesely EG, Maeda N, Kim HS, Desai KM, Krege JH, Laubach

VE, Sherman PA, Sessa WC, Smithies O (1996) Elevated blood

pressures in mice lacking endothelial nitric oxide synthase. Proc

Natl Acad Sci USA 93(23):13176–13181

229. Huang PL, Huang Z, Mashimo H, Bloch KD, Moskowitz MA,

Bevan JA, Fishman MC (1995) Hypertension in mice lacking

the gene for endothelial nitric oxide synthase. Nature

377(6546):239–242. doi:10.1038/377239a0

230. Fagan KA, Tyler RC, Sato K, Fouty BW, Morris KG Jr, Huang

PL, McMurtry IF, Rodman DM (1999) Relative contributions of

endothelial, inducible, and neuronal NOS to tone in the murine

pulmonary circulation. Am J Physiol 277(3 Pt 1):L472–478

231. Chun YS, Kim MS, Park JW (2002) Oxygen-dependent and -

independent regulation of HIF-1alpha. J Korean Med Sci

17(5):581–588 pii: 200210581

232. Semenza G (2002) Signal transduction to hypoxia-inducible

factor 1. Biochem Pharmacol 64(5–6):993–998 pii: S000629520

2011681

233. Rubin L, Simonneau G (2010) Perspective on the optimal end-

points for pulmonary arterial hypertension trials. Curr Opin

Pulm Med 16(Suppl 1):S43–46. doi:10.1097/01.mcp.00003702

10.53379.b3

2830 M. Barrier et al.

123

Page 27: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

234. Jiang BH, Semenza GL, Bauer C, Marti HH (1996) Hypoxia-

inducible factor 1 levels vary exponentially over a physiologi-

cally relevant range of O2 tension. Am J Physiol 271(4 Pt

1):C1172–1180

235. Yu AY, Frid MG, Shimoda LA, Wiener CM, Stenmark K,

Semenza GL (1998) Temporal, spatial, and oxygen-regulated

expression of hypoxia-inducible factor-1 in the lung. Am J

Physiol 275(4 Pt 1):L818–826

236. Tuder RM, Flook BE, Voelkel NF (1995) Increased gene

expression for VEGF and the VEGF receptors KDR/Flk and Flt

in lungs exposed to acute or to chronic hypoxia. Modulation of

gene expression by nitric oxide. J Clin Invest 95(4):1798–1807.

doi:10.1172/JCI117858

237. Gupte SA, Wolin MS (2008) Oxidant and redox signaling in

vascular oxygen sensing: implications for systemic and pul-

monary hypertension. Antioxid Redox Signal 10(6):1137–1152.

doi:10.1089/ars.2007.1995

238. Archer SL, Marsboom G, Kim GH, Zhang HJ, Toth PT,

Svensson EC, Dyck JR, Gomberg-Maitland M, Thebaud B,

Husain AN, Cipriani N, Rehman J (2010) Epigenetic attenuation

of mitochondrial superoxide dismutase 2 in pulmonary arterial

hypertension: a basis for excessive cell proliferation and a new

therapeutic target. Circulation 121(24):2661–2671. doi:

10.1161/CIRCULATIONAHA.109.916098

239. Velez-Roa S, Ciarka A, Najem B, Vachiery JL, Naeije R, van de

Borne P (2004) Increased sympathetic nerve activity in pul-

monary artery hypertension. Circulation 110(10):1308–1312.

doi:10.1161/01.CIR.0000140724.90898.D3

240. McGowan CL, Swiston JS, Notarius CF, Mak S, Morris BL,

Picton PE, Granton JT, Floras JS (2009) Discordance between

microneurographic and heart-rate spectral indices of sympa-

thetic activity in pulmonary arterial hypertension. Heart

95(9):754–758. doi:10.1136/hrt.2008.157115

241. Zhang J, Zhang Y, Li N, Liu Z, Xiong C, Ni X, Pu Y, Hui R, He

J, Pu J (2009) Potential diagnostic biomarkers in serum of idi-

opathic pulmonary arterial hypertension. Respir Med

103(12):1801–1806. doi:10.1016/j.rmed.2009.07.017

242. Geraci MW, Moore M, Gesell T, Yeager ME, Alger L, Golpon

H, Gao B, Loyd JE, Tuder RM, Voelkel NF (2001) Gene

expression patterns in the lungs of patients with primary pul-

monary hypertension: a gene microarray analysis. Circ Res

88(6):555–562

243. Hampole CV, Mehrotra AK, Thenappan T, Gomberg-Maitland

M, Shah SJ (2009) Usefulness of red cell distribution width as a

prognostic marker in pulmonary hypertension. Am J Cardiol

104(6):868–872. doi:10.1016/j.amjcard.2009.05.016

244. Rich S, Kieras K, Hart K, Groves BM, Stobo JD, Brundage BH

(1986) Antinuclear antibodies in primary pulmonary hyperten-

sion. J Am Coll Cardiol 8(6):1307–1311

245. Dib H, Tamby MC, Bussone G, Regent A, Berezne A, Lafine C,

Broussard C, Simonneau G, Guillevin L, Witko-Sarsat V,

Humbert M, Mouthon L Targets of anti-endothelial cell

antibodies in pulmonary hypertension and scleroderma. Eur

Respir J. doi:09031936.00181410 [pii]10.1183/09031936.001

81410

246. Terrier B, Tamby MC, Camoin L, Guilpain P, Berezne A, Ta-

mas N, Broussard C, Hotellier F, Humbert M, Simonneau G,

Guillevin L, Mouthon L (2010) Antifibroblast antibodies from

systemic sclerosis patients bind to {alpha}-enolase and are

associated with interstitial lung disease. Ann Rheum Dis

69(2):428–433. doi:10.1136/ard.2008.104299

247. Forfia PR, Mathai SC, Fisher MR, Housten-Harris T, Hemnes

AR, Champion HC, Girgis RE, Hassoun PM (2008) Hypona-

tremia predicts right heart failure and poor survival in

pulmonary arterial hypertension. Am J Respir Crit Care Med

177(12):1364–1369. doi:10.1164/rccm.200712-1876OC

248. Heresi GA, Aytekin M, Newman J, DiDonato J, Dweik RA

(2010) Plasma levels of high-density lipoprotein cholesterol and

outcomes in pulmonary arterial hypertension. Am J Respir Crit

Care Med 182(5):661–668. doi:10.1164/rccm.201001-0007OC

249. Ghofrani HA, Distler O, Gerhardt F, Gorenflo M, Grunig E,

Haefeli WE, Held M, Hoeper MM, Kahler CM, Kaemmerer H,

Klose H, Kollner V, Kopp B, Mebus S, Meyer A, Miera O,

Pittrow D, Riemekasten G, Rosenkranz S, Schranz D, Vos-

winckel R, Olschewski H (2010) Treatment of pulmonary

arterial hypertension (PAH): recommendations of the Cologne

Consensus Conference. Dtsch Med Wochenschr 135 (Sup-

pl3):S87–101. doi:10.1055/s-0030-1263316

250. Jardim C, Hoette S, Souza R Contemporary issues in pulmonary

hypertension. Eur Respir Rev 19(118):266–271. doi:10.1183/

09059180.00008810

251. Schumann C, Lepper PM, Frank H, Schneiderbauer R, Wibmer

T, Kropf C, Stoiber KM, Rudiger S, Kruska L, Krahn T, Kramer

F (2010) Circulating biomarkers of tissue remodelling in pul-

monary hypertension. Biomarkers 15(6):523–532. doi:10.3109/

1354750X.2010.492431

252. Galie N, Hoeper MM, Humbert M, Torbicki A, Vachiery JL,

Barbera JA, Beghetti M, Corris P, Gaine S, Gibbs JS, Gomez-

Sanchez MA, Jondeau G, Klepetko W, Opitz C, Peacock A,

Rubin L, Zellweger M, Simonneau G (2009) Guidelines for the

diagnosis and treatment of pulmonary hypertension: the Task

Force for the Diagnosis and Treatment of Pulmonary Hyper-

tension of the European Society of Cardiology (ESC) and the

European Respiratory Society (ERS), endorsed by the Interna-

tional Society of Heart and Lung Transplantation (ISHLT). Eur

Heart J 30(20):2493–2537. doi:10.1093/eurheartj/ehp297

253. Leuchte HH, Holzapfel M, Baumgartner RA, Neurohr C,

Vogeser M, Behr J (2005) Characterization of brain natriuretic

peptide in long-term follow-up of pulmonary arterial hyperten-

sion. Chest 128:2368–2374

254. Ciarka A, Doan V, Velez-Roa S, Naeije R, van de Borne P

(2010). Prognostic significance of sympathetic nervous system

activation in pulmonary arterial hypertension. Am J Respir Crit

Care Med 181:1269–1275

Today’s and tomorrow’s imaging and circulating biomarkers 2831

123

Page 28: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

EDITORIAL

Clin. Invest. (2011) 1(3), 363–366New therapeutics for pulmonary arterial hypertension: do gene therapies have translational values?Roxane Paulin1*, Marjorie Barrier1* & Sebastien Bonnet†1,2

Keywords: gene therapy • pulmonary hypertension

Pulmonary arterial hypertension (PAH) is a devastating disease characterized by a marked and sustained elevation of pulmonary vascular resistance (PVR) increasing pulmonary arterial pressure. Progressively, patients will develop right ventricular hypertrophy (RVH) and then right ventricular failure. The pathogenesis of PAH is multifactorial. Endothelial cell (EC) dysfunction is well recognized as an early feature of the disease, leading to an imbalance in production of vasodilatator and vasoconstrictor factors (a decrement in vasodilatators such as prostacyclin and nitric oxide [NO], and a concomitant increase in vasoconstrictors such as thromboxane and endothelin-1) [1]. Over the past several years, therapy has focused on re-establishing this imbalance by, for example, exogenous delivery of vasodilatating prostaglandins and inhaled NO, or by blocking the endothelin axis, which leads to vasodilatation and improves the pulmonary circulation. Unfortunately, some of these treatments have undesired side effects, principally systemic hypotension (epoprostenol) and liver toxicity (sitaxentan, withdrawn from the market in 2010). Moreover, some patients are resistant to these therapies, and even for those who show responses, they failed to totally reverse PAH, unless the patients also received a lung/heart transplant. In fact, the major cause of the elevated PVR is the obstructive vascular remodeling due to an imbalance in the proliferation and apoptosis rates of the pulmonary artery smooth muscle cells (PASMCs) [1], which show a cancer-like behavior. Thus, the sci-entific community has started to focus more and more on the cellular and molecular mechanisms implicated in pulmonary artery remodeling and have started to develop therapies aimed at reversing the proliferative phenotype of PASMCs in the vascular wall [2]. Genomic approaches such as gene expression profiling and sequencing have demonstrated that many genes are aberrantly expressed in PAH [3] and are often involved in the activation of pathways responsible for the pro-proliferative and anti-apoptotic phenotype of PASMCs. Moreover, advances in gene-transfer technologies made the development of ‘gene therapy’ the modification of choice for therapeutic proposes. First designed to restore a genetic disorder or a mutation by transferring a normal copy of the gene, it soon became apparent that the range of targeted diseases could be extended to those showing aberrant gene expression. Therefore, in the last decade, several researches explored gene therapy for PAH.

Major recent advancesCompared with other thoracic malignancies, such as lung cancer, asthma, emphy-sema and cystic fibrosis, PAH appears to be late in gene therapy applications. In fact, the first clinical trial implicating gene transfer in PAH only began in 2010 [4]. Autologous endothelial progenitor cells (EPCs) programmed to overexpress the endothelial NO synthase (eNOS) were administered to patients with iPAH and

1Department of Medicine, Laval University, Quebec City, QC, Canada 2Centre de recherche de L’Hôtel-Dieu de Quebec, 10 rue McMahon, Quebec, Qc, G1R 2J6, Canada †Author for correspondence: Tel.: +1 418 525 4444 Ext. 16350 Fax: +1 418 691 5562 E-mail: [email protected] *Authors contributed equally.

363ISSN 2041-679210.4155/CLI.11.2 © 2011 Future Science Ltd

For reprint orders, please contact [email protected]

Page 29: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

PAH associated with systemic sclerosis. Preliminary data showed modest but significant improvements in 6-min walk test and mean pulmonary arterial pressure. Given the fact that EPC therapy only (nonassociated with gene transfer) in two small-randomized trials in humans showed improvement in 6-min walk test and PVR [5], it is difficult to evaluate the real benefit of this additional gene therapy.

Antiproliferative candidates for gene therapy in PAHGene therapy targeting endothelial dysfunction is cur-rently being studied, but targeting the pro-proliferative phenotype of PASMCs is still to be achieved. Several experiments restoring mRNA levels of genes implicated in the aberrant activation of proliferative pathways in PAH have been performed in the last 5 years in animal models of PAH, and results are really promising.

■ BMPRII gene therapyHeterozygous mutations in BMPRII, a member of the TGF family of receptors, have been identified in many cases of familial and sporadic PAH [6]. In the case where BMPRII is not mutated, its downregulation is often observed in the PAH patients. Adenoviral deliv-ery of vector containing BMPRII gene in pulmonary vascular endothelium of chronic hypoxia-induced PAH (CH-PAH) rats reduced the pulmonary hypertensive responses [7]; whereas BMPRII intratracheal nebuliza-tion of adenoviral gene therapy in the monocrotaline rat model of PAH did not improve pulmonary hypertension despite a good distribution of the gene in the arteriolar network [8]. The heterogeneity of these results suggests that BMPRII may not be the best candidate for gene therapy. Nevertheless, even if BMPRII gene therapy could not be universally applied for PAH patients, it may be beneficial in some cases, or could be effectively coupled with other genes therapies.

■ Kv gene therapyContractility and proliferation of PASMCs is controlled by cytosolic Ca2+ levels, which are largely determined by membrane potential (Em). In fact, Em is depolarized in human and experimental PAH cells due to, at least in part, the decreased expression and function of voltage-gated K+ channels (Kv1.5 and Kv2.1). This ‘K+-channelopathy’ leads to PASMCs depolarization and Ca2+ overload, thus promoting vasoconstriction and PASMCs proliferation. Therefore, targeting and improving expression and function of these channels is thought to be promising. Restoration of Kv channel expression in PAH by aerosol gene therapy using an adenovirus expressing Kv2.1 might actually be beneficial [9]. Furthermore, Kv1.5 expression in established CH-PAH in rats reduces PVR and restores

RVH [10], and KCNA5 gene transfer in human PASMCs increases K+ currents and enhances apoptosis [11], demon-strating that Kv1.5 may serve as an important strategy for preventing the progression of PAH.

■ Survivin gene therapyPulmonary arterial remodeling in PAH might be explained by a pro-proliferative and antiapoptotic phenotype of the PASMCs. Survivin, a member of the inhibitor of apoptosis protein family, was first discov-ered in cancer. It is normally undetectable in healthy adult differentiated tissues, but it is expressed in remod-eled PAs from patients and rats with PAH, highlighting once again this proliferative phenotype. Adenovirus-mediated survivin overexpression induces PAH in rats, underlying an implication of this oncoprotein in PAH, whereas inhalation of a survivin-dominant negative adeno virus reverses established monocrotaline-induced PAH (MCT-PAH) [12] by avoiding PASMC proliferation and, thus, the subsequent PAH.

■ VIP gene therapyAmong its actions, vasoactive intestinal peptide inhibits proliferation of vascular smooth muscle cells. PASMCs treated with adenovirus expressing the VIP gene are less proliferative [13]. It could be interesting to see VIP-based gene therapy trials on animal models.

■ CGRP gene therapyCalcitonin gene-related peptide has also been described to have an antiproliferative effect. Intratracheal injec-tion of adenovirus carrying the pre-proCGRP gene into the lung of hypoxia-induced PAH mice attenuates the increase in PVR, RVH, remodeling and pulmonary pressure [14].

■ Growth factor gene therapyVEGF-A overexpression in MCT rats using cell-based gene transfer prevents PAH [15]. In the same way, HGF gene transfection in MCT rats prevents media wall thickening [16]. These findings imply that VEFA-A and HGF have protective effects against remodeling and could be good candidates for gene therapy.

Limitations of gene therapyGene therapies are hopeful treatments that might in the future help PAH patients. The aim of PAH treat-ment is to reduce pulmonary hypertension without affecting systemic circulation. Gene delivery into the lung via aerosol is one of these strategies. This could avoid many problems associated with systemic delivery by intravenous injection, such as immediate nuclease degradation and difficulty penetrating the endothelial barrier. Successful gene delivery via inhalation strongly

www.future-science.com future science group364

EDITORIAL Paulin, Barrier & Bonnet

Page 30: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

depends on the development of advanced gene vectors. They must be able to protect the plasmid or sequence, provide a specific targeting site and effectively release these plasmids for the desired pharmacological effect. In the past two decades, numerous preclinical and clinical trials have been performed for thoracic malignancies, and lessons must be learned from these experiments.

First trials with adenovirus and adeno-associated viral vectors appeared to be ineffective and associated with detrimental immunologic responses and toxicity  [17]. Recent work show some ingenious constructions, such as antibiotic coupling adenovirus or ‘helper-depen-dent adenovirus’ in which the proinflammatory virus sequences are missing. This permits a decreased inflam-mation associated with gene therapy. Lentiviruses are more efficient than other viral vectors but they need to be frequently readministered and lead to the same inflammatory problems.

Nonviral gene transfers are generally less efficient. In fact, plasmids as well as vectors used, require improve-ments. Some nucleotide sequences such as CpG motifs have been demonstrated to enhance proinflammatory responses [18] and have to be avoided in plasmid con-struct. Promoters, which regulate, in part, duration and tissue-specific localization of gene expression, have to be carefully chosen, particularly in the case of a desired restricted area of gene delivery (e.g., the vascular bed).

With the development of nanotechnologies, the emergence of new kinds of construction can be hoped for. Some nonviral vectors, for example liposomal ones, seem to cause less immunologic lung responses [19], which could worsen PAH. This is also true with nanomicelles. Formulations such as polyethylenimine (25 kDa, Sigma), cationic lipid 67 (GL67A, Genzyme corporation) or DNA nanoparticles (Copernicus), were shown to increase efficiency and duration of transgene expression.

Physical methods have also opened a new window in lung gene transfer improvement. Magnetic particles linked to DNA enhance a DNA response to magnetic fields (magnetofaction) and increase transfection rates in vitro. Unfortunately, application of this method in vivo is unsuccessful. Ultrasound (sonoporation) and electroporation have been shown to improve gene transfer in various tissues, but are not applicable in vivo because they are associated with lung damage such as hemorrhage.

Future perspectiveThe increased knowledge and understanding of stem cells has provided the scientific community hope to find cell therapies for PAH. Some are already used for genetic pathologies, mainly embryonic stem cells. The concept of using the patients’ own stem cells is strongly

emerging, avoiding ethical and immune issues. EPCs appear to be promising candidates, as described previ-ously. Mesenchymal stem cells are also very promis-ing [20]. The understanding of smooth muscle cell differ-entiation mechanisms are increasing and could be very useful for all vascular diseases, including atherosclerosis, heart failure and PAH.

The principle of coupling gene and cell therapies is also coming to the fore: isolate stem cells from a patient, make them express a gene of interest and then re-inject them into the patient. Coupling gene and cell therapy is already in trials using eNOS-transformed EPCs to treat PAH [4], and it could be speculated that targeting genes involved in proliferation and antiapoptotic mechanisms would be even more efficient. Furthermore, using mesen chymal stem cell-coupled gene therapy could be very promising considering their multipotency and their ability to accumulate at the site of tissue/organ damage and inflammation in vivo [21].

Targeting genes involved in the pro-proliferative and antiapoptotic phenotype by RNAi or siRNA could be one other possibility. As RNAi is increasly studied and better understood its use as a therapeutic is slowly emerg-ing. Aerosol coupling siRNA and transfecting agent would theoretically be able to silence the pathologically overexpressed gene(s) observed in PAH PASMCs. This is undoubtedly a promising concept to work on, even though technical issues remain to be resolved. siRNA lifespan, application modes and frequencies have to be characterized. The specificity, structural role and off-target effects also remain to be determined. Regarding the emerging concept of microRNA, which have a natu-ral RNAi action on genes and whose aberrant expression is implicated in human diseases and in PAH, we can also speculate that progress will continue in the next years. Administered mimics or antagomir in order to restore aberrant microRNA expressions could be another therapeutic intervention, but one that also requires improvements in transfectant agents.

ConclusionTreating PAH by gene therapy is very promising and targets and methods are being extensively studied. Treatments based on inhaled siRNA targeting different genes once a week or durable gene/cell therapies restor-ing one or few gene expression are achievable. Some technical issues remain to be solved and time is needed to realize the trials. Nevertheless, these therapeutics are associated with high technical costs. In this way, it will be difficult for these new treatments to be competitive against cheaper agents, such as dehydroepiandrosterone, dichloro acetic acid and trimetazidine [22]. These agents, already in trials, need much less expensive manufacturing costs for the relative same efficiency.

New therapeutics for pulmonary arterial hypertension: do gene therapies have translational values? EDITORIAL

future science group Clin. Invest. (2011) 1(3) 365

Page 31: Today’s and tomorrow’s imaging and circulating biomarkers ... · 2) Pulmonary hypertension with left-sided heart disease Systolic dysfunction Diastolic dysfunction Valvular disease

Financial & competing interests disclosureThe authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manu-script. This includes employment, consultancies, honoraria, stock ownership or options, expert t estimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.

Bibliography1 Archer S, Rich S. Primary pulmonary

hypertension: a vascular biology and translational research ‘Work in progress’. Circulation 102(22), 2781–2791 (2000).

2 Rubin LJ. Therapy of pulmonary hypertension: the evolution from vasodilators to antiproliferative agents. Am. J. Respir. Crit. Care Med. 166(10), 1308–1309 (2002).

3 Geraci MW, Moore M, Gesell T et al. Gene expression patterns in the lungs of patients with primary pulmonary hypertension: a gene microarray ana lysis. Circ. Res. 88(6), 555–562 (2001).

4 Jurasz P, Courtman D, Babaie S, Stewart DJ. Role of apoptosis in pulmonary hypertension: from experimental models to clinical trials. Pharmacol. Ther.126(1), 1–8 (2010).

5 Wang XX, Zhang FR, Shang YP et al. Transplantation of autologous endothelial progenitor cells may be beneficial in patients with idiopathic pulmonary arterial hypertension: a pilot randomized controlled trial. J. Am. Coll. Cardiol. 49(14), 1566–1571 (2007).

6 Deng Z, Morse JH, Slager SL et al. Familial primary pulmonary hypertension (gene PPH1) is caused by mutations in the bone morphogenetic protein receptor-II gene. Am. J. Hum. Genet. 67(3), 737–744 (2000).

7 Reynolds AM, Xia W, Holmes MD et al. Bone morphogenetic protein type 2 receptor gene therapy attenuates hypoxic pulmonary hypertension. Am. J. Physiol. Lung Cell Mol. Physiol. 292(5), L1182–1192 (2007).

8 McMurtry MS, Moudgil R, Hashimoto K, Bonnet S, Michelakis ED, Archer SL. Overexpression of human bone morphogenetic protein receptor 2 does not ameliorate monocrotaline pulmonary arterial hypertension. Am. J. Physiol. Lung Cell Mol. Physiol. 292(4), L872–878 (2007).

9 Michelakis ED, Dyck JR, McMurtry MS et al. Gene transfer and metabolic modulators as new therapies for pulmonary hypertension. Increasing expression and activity of potassium channels in rat and human models. Adv. Exp. Med. Biol. 502, 401–418 (2001).

10 Pozeg ZI, Michelakis ED, McMurtry MS et al. In vivo gene transfer of the O

2-sensitive

potassium channel Kv1.5 reduces pulmonary hypertension and restores hypoxic pulmonary vasoconstriction in chronically hypoxic rats. Circulation 107(15), 2037–2044 (2003).

11 Brevnova EE, Platoshyn O, Zhang S, Yuan JX. Overexpression of human KCNA5 increases IK V and enhances apoptosis. Am. J. Physiol. Cell Physiol. 287(3), C715–C722 (2004).

12 McMurtry MS, Archer SL, Altieri DC et al. Gene therapy targeting survivin selectively induces pulmonary vascular apoptosis and reverses pulmonary arterial hypertension. J. Clin. Invest. 115(6), 1479–1491 (2005).

13 St Hilaire RC, Kadowitz PJ, Jeter JR Jr. Adenoviral transfer of vasoactive intestinal peptide (VIP) gene inhibits rat aortic and pulmonary artery smooth muscle cell proliferation. Peptides 30(12), 2323–2329 (2009).

14 Bivalacqua TJ, Hyman AL, Kadowitz PJ, Paolocci N, Kass DA, Champion HC. Role of calcitonin gene-related peptide (CGRP) in

chronic hypoxia-induced pulmonary hypertension in the mouse. Influence of gene transfer in vivo. Regul. Pept. 108(2–3), 129–133 (2002).

15 Zhao YD, Courtman DW, Ng DS et al. Microvascular regeneration in established pulmonary hypertension by angiogenic gene transfer. Am. J. Respir. Cell Mol. Biol. 35(2), 182–189 (2006).

16 Ono M, Sawa Y, Mizuno S et al. Hepatocyte growth factor suppresses vascular medial hyperplasia and matrix accumulation in advanced pulmonary hypertension of rats. Circulation 110(18), 2896–2902 (2004).

17 Moss RB, Milla C, Colombo J et al. Repeated aerosolized AAV-CFTR for treatment of cystic fibrosis: a randomized placebo-controlled Phase IIB trial. Hum. Gene Ther. 18(8), 726–732 (2007).

18 Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu. Rev. Immunol. 20, 709–760 (2002).

19 Hyde SC, Southern KW, Gileadi U et al. Repeat administration of DNA/liposomes to the nasal epithelium of patients with cystic fibrosis. Gene Ther. 7(13), 1156–1165 (2000).

20 Myers TJ, Granero-Molto F, Longobardi L, Li T, Yan Y, Spagnoli A. Mesenchymal stem cells at the intersection of cell and gene therapy. Expert Opin. Biol. Ther.10(12), 1663–1679 (2010).

21 Ozawa K, Sato K, Oh I et al. Cell and gene therapy using mesenchymal stem cells (MSCs). J. Autoimmun. 30(3), 121–127 (2008).

22 Sutendra G, Bonnet S, Rochefort G et al. Fatty acid oxidation and malonyl-CoA decarboxylase in the vascular remodeling of pulmonary hypertension. Sci. Transl. Med. 2(44), 44ra58 (2010).

www.future-science.com future science group366

EDITORIAL Paulin, Barrier & Bonnet