ArticlePDF AvailableLiterature Review

Update in Pulmonary Vascular Diseases 2012

Authors:
Update in Pulmonary Vascular Diseases 2012
Martin R. Wilkins
1
, John Wharton
1
, and Mark T. Gladwin
2
1
Centre for Pharmacology and Therapeutics, Department of Medicine, Imperial College London, United Kingdom; and
2
Department
of Medicine and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
Pulmonary vascular disease presents a considerable therapeu-
tic challenge. A look at the past year shows that it is a highly
active field of research. About 2,500 publications from many
countries appeared on PubMed in 2012. Here we focus on
some major emerging themes and follow an established
tradition (1), reviewing primarily articles published in
AJRCCM.
Some of the results of these studies have been previously re-
ported in the form of an abstract (2–4).
PULMONARY ARTERIAL HYPERTENSION IN THE
MODERN ERA: PATHOLOGY, DEMOGRAPHICS,
AND ASSOCIATIONS
Textbook descriptions of the vascular pathology of pulmonary
arterial hypertension (PAH) precede the era of modern therapeu-
tics. Stacher and colleagues, using a large collection of explanted
lung tissue (62 patients with PAH), courtesy of the Pulmonary Hy-
pertension Breakthrough Initiative, have applied systematic sam-
pling and analysis to provide an important update (5). Significant
observations include heterogeneity in structural remodeling
within the same lung and between lungs from different patients;
that the primary differences between PAH and healthy lung
reside in intima fractional thickness; that medial fractional
thickness overlapped with measurements made in healthy
tissue; and the presence of marked perivascular inflamma-
tion. The study supports the concept of pathologically dis-
tinct subphenotypes, even in idiopathic PAH (IPAH), and
poses the challenge to identify these at an earlier stage of the
disease (6).
This report should be understood in the context of the changing
demographics of PAH. Ling and colleagues published the largest
cohort of incident PAH, with 482 newly diagnosed patients pre-
senting between 2001 and 2009 (7). The mean age was 50 years,
compared with 36 years in the landmark National Institutes of
Health registry from 1987. Patients older than 50 years were
more obese, had more comorbidities, and worse survival pros-
pects than younger patients. The shift in demographics likely
reflects increased awareness of PAH and a change in referral to
specialist centers. The diagnosis of PAH in older patients, who
are more likely to suffer from coincident systemic hypertension
and ischemic heart disease, demands an obsessional approach
to measuring pulmonary capillary wedge pressure to distin-
guish PAH from pulmonary venous hypertension associated
with left ventricular (LV) dysfunction (8). The high prevalence
of diabetes in older patients with PAH may be simply
contemporaneous, but there is increasing awareness of an asso-
ciation between PAH and insulin resistance and metabolic dis-
turbance may have an intimate role in the development of
PAH (9).
In common with other registries (10), the U.K. and Ire-
land cohort shows improved patient outcomes in the last 10
years. Survival at 1, 2, 3, and 5 years was 92.7, 84, 73.3, and
61.1%, respectively. Although tempting to attribute this im-
provement to new treatments, changing demographics and
a general improvement in patient follow-up and manage-
ment are also factors. Higher mortality and increased risk
of clinical worsening are associated with lower socioeco-
nomic class, independent of clinical features and baseline
hemodynamics (11).
Pulmonary hypertension (PH) is a significant cause of mor-
bidity and mortality in children, and the Tracking Outcomes
and Practice in Pediatric Pulmonary Hypertension Registry
has been established as a global prospective study to explore
the demographics, treatment, and outcomes in pediatric PH
(12). Of the 362 patients recruited with confirmed PH 87%
had PAH and several age-specific characteristics were iden-
tified that emphasize the need for further pediatric-based
studies. These include the type and distribution of associated
disorders (notably congenital heart disease), the high preva-
lence of comorbidities such as trisomy 21, the frequent oc-
currence of syncope, and apparent preservation of right-heart
function despite severe PH (12).
FRESH INSIGHTS INTO MECHANISMS
The ENCODE (Encyclopedia of DNA Elements) Project showed
that less than 2% of the human genome encodes protein whereas
about 60% is transcribed into RNA sequences, many of which
may be concerned with gene regulation (13). Short RNA tran-
scripts, microRNAs (miRs), have emerged as critical posttran-
scriptional regulators of gene expression. More than 1,400 miRs
have been identified and the levels of several are affected by or
predicted to change in experimental PH (14, 15). Pullamsetti and
colleagues have provided proof-of-concept that miR inhibitors,
antagomirs, may offer a novel therapeutic strategy in PAH; inhi-
bition of miR-17 attenuated the PH phenotype in both hypoxia-
and monocrotaline-induced PH (16). Further investigations are
required to establish which miR(s) might be targeted and taken
to clinical trials in patients with PAH. Other RNA molecules are
also being developed as therapeutic agents, including Spiegelmers.
These are stable mirror isomers of RNA that bind specific
chemokines with high (subnanomolar) affinity. Spiegelmer
NOX-A12 blocks stromal cell–derived factor-1 (CXCL12)
and, given to monocrotaline-treated rats with established
PH, inhibits the accumulation of pulmonary inflammatory
cells, reduces cardiovascular remodeling, and improves he-
modynamics (17).
Epigenetic differences (i.e., variation in gene expression and
phenotype that do not result from alteration in DNA sequence)
are now thought to have a role in PAH. In addition to the suppres-
sion of gene expression by miRNAs, the principal mechanisms of
(Received in original form March 11, 2013; accepted in final form May 6, 2013)
Correspondence and requests for reprints should be addressed to Martin Wilkins,
M.D., NIHR–Wellcome Trust Imperial CRF, Imperial Centre for Translational
and Experimental Medicine, Imperial College London W12 0NN, UK. E-mail:
m.wilkins@imperial.ac.uk
Am J Respir Crit Care Med Vol 188, Iss. 1, pp 23–28, Jul 1, 2013
Copyright ª2013 by the American Thoracic Society
DOI: 10.1164/rccm.201303-0470UP
Internet address: www.atsjournals.org
epigenetic regulation include DNA methylation and the modifica-
tion of histone proteins associated with DNA in chromatin. Acet-
ylation is one the most frequent posttranslational modifications
of histones and is a dynamic process regulated by histone acetyl-
transferases and histone deacetylases. Studies in cells and animal
models have demonstrated antiinflammatory and antiremodeling
properties of histone deacetylase inhibitors that could be benefi-
cial in patients (18, 19). But these compounds target multiple cell
types and selectivity is the key to identifying an inhibitor to take
forward for PAH. Concerns about myocardial toxicity also dictate
that we should proceed with caution.
Stacher and colleagues observed a correlation between peri-
vascular infiltration and remodeling of the intima and media
in lungs of patients with PAH (5). Savai and colleagues quan-
tified the cellular infiltration in explanted lungs from patients
with IPAH and found that, except for FoxP3
1
regulatory T
(Treg) cells, the number of mast cells, macrophages, dendritic
cells, and subsets of T cells and B cells in the adventitia was
increased throughout small (diameter, 20–50 mm) to large (di-
ameter, .150 mm) remodeled pulmonary arteries (17). Large
accumulations of lymphocytes have also been localized to ter-
tiary (ectopic) lymphoid follicles adjacent to remodeled vessels,
with chemokines (e.g., CXCL13), lymphotoxin-a/-b, and immu-
noregulatory cytokines (IL-7 and IL-21) implicated in their for-
mation and function (20). Interestingly, lymphoid neogenesis
was not observed in patients with flow-induced PAH (Eisenmenger
syndrome). Together with the accumulation of CD138
1
plasma
cells and deposition of immunoglobulins, this suggests that local
generation of antibodies and autoimmunity are important in
IPAH (20). In-depth studies of the perturbation of specific cell
subtypes (e.g., Treg cells [21] and natural killer cells [22]) in the
circulation as well as lung tissues are now critical to our under-
standing the role of inflammatory mechanisms and altered im-
munity in the progression of PAH (1).
Infection with HIV and intravenous drug use are known to be
independent risk factors for the development of PAH. The con-
comitant use of opioids and amphetamines represents a potential
“second hit” in the pathogenesis of HIV-PAH (23). Spikes and
colleagues treated macaques with morphine before and after
simian immunodeficiency virus inoculation and observed pul-
monary vascular remodeling, including intimal and medial
thickening and plexiform-like lesions, and more severe perivas-
cular inflammation than animals receiving morphine or simian
immunodeficiency virus inoculation alone (24). The combined
insult was also associated with Pneumocystis infection, raised
circulating IL-8 and monocyte chemotactic protein-1 (CCL2)
levels, and evidence of enhanced pulmonary endothelial apo-
ptosis and proliferation. Morphine potentiated the apoptotic
and proliferative effects of HIV viral proteins (Tat, Nef, and
gp-120
CM
), promoted oxidative stress, and affected vascular en-
dothelial growth factor expression/activation in human pulmo-
nary microvascular endothelial cells (24). It remains to be seen
whether other forms of drug abuse have a similar effect, but
these novel findings support a multiple hit hypothesis in the
pathogenesis of HIV-PAH and provide a new model for lon-
gitudinal investigations of pulmonary hemodynamics as well as
vascular remodeling in PAH (25).
Growth factors and the altered expression/activity of receptor
tyrosine kinases (RTKs) are implicated in PAH. Investigation of
RTK transduction pathways identified a common adaptor pro-
tein, p130
Cas
, for major growth factors involved in PAH, includ-
ing platelet-derived growth factor, fibroblast growth factor-2,
and epidermal growth factor (26). Importantly, patients with
IPAH exhibited increased circulating levels and expression/
activity of p130
Cas
in distal pulmonary arteries, modulating
the proliferative and migratory effects of the growth factors
in cultured pulmonary artery smooth muscle cells (PASMCs) and
endothelial cells. Adaptor p130
Cas
protein expression/activity
was also increased in the lungs of rodents with hypoxia- and
monocrotaline-induced PH, and treatment with RTK inhibitors
(gefitinib, dovitinib, and imatinib) reduced both p130
Cas
levels
and inhibited the progression of PH and cardiovascular remod-
eling (26). These observations need to be confirmed and, while
selective inhibitors are not currently available, p130
Cas
may rep-
resent a circulating biomarker reflecting the progression of pul-
monary vascular disease and response to therapy.
The elevation of intracellular Ca
21
levels in PASMCs is con-
sidered to be an important step in pulmonary contraction and
vascular remodeling (27). The entry of Ca
21
into the cytoplasm
occurs via receptor-operated and store-operated Ca
21
channels,
and their initial opening is caused by the activation of a G
protein–coupled receptor known as the extracellular Ca
21
-sensing
receptor (CaSR or GPRC2A). Data have implicated enhanced
CaSR expression and function in the proliferation of PASMCs
from patients with IPAH, as well as the development of PH in
experimental animal models, and suggest that pharmacological
blockade of the receptor or inhibition of expression may be
a potential therapeutic strategy in the future (27).
Increasing evidence indicates that mineralocorticoid recep-
tor signaling has a role in cardiovascular disease, modulating
macrophage and immune cell function and promoting inflamma-
tion and oxidative stress in the heart and vascular tissues (28),
and the renin–angiotensin–aldosterone system is activated in
patients with IPAH (29). de Man and colleagues showed that
circulating levels of renin, angiotensin I, and angiotensin II
(AngII) are raised in IPAH and associated with disease progres-
sion and outcome (30). Local angiotensin-converting enzyme
activity, AngII type I receptor expression, and downstream sig-
naling were increased in pulmonary arteries and endothelial
cells from patients with IPAH. Furthermore, AngII selectively
induced the proliferation of PASMCs from patients with IPAH
and this was blocked by the AngII type I receptor antagonist,
losartan. The potential therapeutic significance of these obser-
vations was underlined by the finding that chronic losartan treat-
ment delayed the progression of pulmonary vascular remodeling
and right ventricular (RV) dysfunction in the monocrotaline rat
model (30). Other agents are also available that target the renin–
angiotensin–aldosterone system. Preclinical studies have shown the
therapeutic potential of aldosterone inhibitors in PAH (31), and
the effect of spironolactone on collagen metabolism in PAH is
now undergoing clinical investigation (ClinicalTrials.gov identi-
fier NCT01468571).
THE ROLE OF ESTROGENS
The role of female sex, and specifically estrogenic hormones, in the
penetrance, severity, and even treatment of PAH is the subject of
active research. However, the findings are not congruent and in
fact paradoxical. For example, there is a female predominance
in IPAH and increased penetrance of PAH in female members
of families with mutations in the bone morphogenetic protein
receptor type 2 (BMPR2) gene (32). But 2-methoxyestradiol ther-
apy is protective in monocrotaline-induced PH and female ro-
dents often develop less severe hypoxia-induced PH and RV
failure (33). These findings have been referred to as the “estrogen
paradox and the potential mechanisms comprehensively re-
viewed (32, 34).
Lahm and colleagues investigated the role of direct estrogen
receptor activation by 17b-estradiol versus the downstream
effects of its metabolites in the setting of hypoxia-induced PH
in mice (35). 17b-Estradiol therapy was protective, inhibiting
cell proliferation and activating autophagy to limit PH and
24 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 188 2013
RV hypertrophy. Using selective estrogen receptor, catechol
O-methyltransferase, and CYP450 inhibitors, 17b-estradiol
appeared to directly activate estrogen receptor-a, indepen-
dent of the estrogen receptor-band downstream metabolites,
reduce extracellular signal–regulated kinase-1/2 (ERK1/2)
activation, increase p27
Kip1
expression, and increase auto-
phagy, leading to improvement in pulmonary pressures and
RV function (35). It is somewhat surprising that the effects
of 17b-estradiol were mediated predominantly by activation of es-
trogen receptor-a, as the estrogen receptor-bknockout mouse
develops hypertension and vascular dysfunction and activation of
this receptor has antiproliferative effects. Further studies are re-
quired to fully reconcile these observations if nonhormonal selec-
tive estrogen receptor ligands are to be developed for PAH.
SIX-MINUTE WALK, HEART RATE, AND RV
FUNCTION IN PAH
Right heart function is the most important determinant of sur-
vival in patients with PAH. A number of studies have refined
specific parameters of right heart function, such as 6-minute walk
distance, heart rate recovery (HRR), and novel measures of RV
glycolytic activity, to assess clinical risk. Although the 6-minute
walk test (6MWT) is the “workhorse” efficacy end point in
clinical trials, the magnitude of improvement in walk distance
with therapy that is clinically meaningful remains unknown. In
a retrospective analysis of data from the Pulmonary Hyperten-
sion Response to Tadalafil (PHIRST) trial, Mathai and col-
leagues used distributional and anchor-based methodologies
to conclude that a distance of greater than 33 m is associated
with improvement in quality-of-life measures (36). These results
are consistent with a more extensive analysis of 10 randomized
clinical trials of PAH-targeted therapy, which found that an
increase greater than 42 m best predicted a reduction in the
time to clinical worsening (37). It has been appreciated for some
time, however, that the threshold level (e.g., achieving .380 m)
is more important than a small improvement over baseline and
is likely to assume a greater role in the assessment of new
therapies in the future (38).
Information obtained after completion of the 6MWT also ap-
pears to have value. Minai and colleagues assessed the associa-
tion between HRR (defined as the difference in heart rate at the
end of 6MWT and 1 min after completion of the 6MWT) and
clinical worsening in 75 consecutive patients with PAH (39).
The best predictors of clinical worsening were an HRR less than
16 (hazard ratio, 5.2), which occurred in 40% of patients eval-
uated, and the mean pulmonary arterial pressure (hazard ratio,
1.04). HRR was also a better predictor of clinical worsening
than 6MWT. The accompanying editorial emphasized the im-
portance of heart rate control in cardiovascular risk assessment
and noted two caveats: the relatively high number of subjects
who were receiving b-blocker therapy (25%) in the group with
an HRR less than 16 and lack of information about whether the
HRR profile can be improved with therapy (40). Future studies
in larger cohorts evaluated before and after therapy will help to
address these questions and further support the proposed use-
fulness of measuring HRR in patients with PAH (39, 41).
Whereas PAH is characterized by right heart failure, progres-
sive diastolic RV pressure overload can adversely affect LV di-
astolic filling and output. Displacement of the interventricular
septum during the diastolic phase with right heart failure can
compromise LV filling and volume (referred to as diastolic ven-
tricular interaction or interdependence). Kasner and colleagues
explored this mechanism in patients with nonsevere PAH
(mean pulmonary artery pressure, 29 mm Hg), using invasive
pressure–volume loop analysis during simultaneous atrial
pacing to a heart rate of 120 beats/minute (42). Remarkably,
even in these mildly affected patients their stroke volumes
dropped by about 25% during pacing. They then occluded
the inferior vena cava to transiently reduce RV filling. With
this maneuver, the LV end-diastolic volume increased 7% and
end-diastolic pressures dropped, resulting in an 11% increase
in cardiac output. This elegant study provides proof that the
heart rate–dependent increase in RV pressure/volume over-
load adversely impacts LV cardiac filling and output, even in
the setting of normal LV systolic function. This has clear impli-
cations for diseases such as sickle cell anemia and portopulmo-
nary hypertension, which are characterized by high baseline
cardiac output states and mild-to-moderate elevations in pulmo-
nary pressures, yet poor prospective survival rates (43, 44).
Cardiac magnetic resonance imaging provides a noninvasive
means of assessing pulmonary hemodynamics and cardiac struc-
ture and function, and studies have explored the use of modern
techniques that may help indentify local abnormalities in RV
function and predict survival in patients with PAH (45, 46).
Another approach that could be useful in evaluating the sever-
ity of PAH and response to therapy is the measurement of
a shift from oxidative phosphorylation to glycolytic metabolism,
using positron emission tomographic imaging of
18
F-labeled
deoxyglucose (FDG) uptake. FDG uptake, via glucose transporter-
1, is increased in the RV of patients with IPAH and in IPAH
pulmonary vascular cells in culture (47). Marsboom and col-
leagues evaluated the relationship between FDG uptake, cellu-
lar glycolytic activity, and PH severity in monocrotaline and
Sugen/hypoxia rat models (48). FDG uptake increased in the
pulmonary vasculature as well as the RV, and the signal in the
vasculature was related to increased cellular glycolytic activity
in proliferating smooth muscle and endothelial cells. This was
characterized by increased glucose transporter-1 expression,
inhibition of pyruvate dehydrogenase activity by pyruvate de-
hydrogenase kinase, and reductions in mitochondrial oxidative
phosphorylation. The increase in glycolytic activity correlated
closely with disease progression and response to two therapies:
inhibition of pyruvate dehydrogenase kinase by dichloroacetate
and inhibition of tyrosine kinase by imatinib. Hypoxia-inducible
factor-1awas also implicated in the development of the glycolytic
phenotype. The clinical usefulness of measurements of FDG up-
take in IPAH remains to be determined.
SICKLE CELL DISEASE–ASSOCIATED PH
Mehari and colleagues summarized the hemodynamic param-
eters of 84 patients with sickle cell disease and clinical suspicion
of PH from a cohort of 529 monitored up to 9 years, the largest
prospectively screened sickle cell cohort to date (44). Fifty-five
patients (10% of the total cohort) had PH and of these 31 (6%
of the cohort) had PAH. PH was the major independent risk
factor for death and risk was determined primarily by the
severity of precapillary pulmonary artery pressures, specifi-
cally pulmonary vascular resistance and transpulmonary pres-
sure gradients, but not pulmonary artery occlusion pressure
values. These findings are consistent with those of a Brazilian
cohort, in which the 10% of patients with PH had the highest
risk of death (49). In both studies, the risk of PH was associated
with the severity of hemolytic anemia, suggesting that patients
with all forms of chronic hemolytic anemia should be evaluated
for possible PH.
GENETICS OF PAH
Familial PAH is inherited as an autosomal dominant condition
and may show anticipation, with younger age of onset and death
Pulmonary, Sleep, and Critical Care Updates 25
in subsequent generations, as well as reduced penetrance (only
z20% of individuals carrying the mutant gene develop the dis-
ease). Having allowed sufficient time to elapse to capture all
affected cases in the analysis of age at diagnosis and death,
Larkin and colleagues reanalyzed 355 individuals with BMPR2
mutations from 53 PAH families in the Vanderbilt University
PH registry (50). Their results suggest that genetic anticipation
is an artifact, reflecting an ascertainment bias where only the
early-onset cases are apparent in recent generations. Nonethe-
less, additional genetic and environmental factors are likely to
be important in determining which genetically susceptible indi-
viduals develop the disease (51) and studies provide evidence
that the phenomenon of reduced penetrance in BMPR2 mutant
carriers is due in part to alterations in alternative splicing (52).
Analysis of lymphocytes from patients and unaffected carriers
with BMPR2 mutations showed that patients produced more of
the alternatively spliced BMPR2 isoform B (missing exon 12)
compared with the full-length isoform A, the ratio between the
two being controlled by an exonic splice enhancer in exon 12
and its associated splicing factor, SRSF2. The study represents
a significant step toward understanding which BMPR2 mutant
carriers may eventually manifest the disease and could facilitate
the development of novel therapies (53).
Flynn and colleagues developed an original approach to
explore the molecular pathways that may influence disease
penetrance in PAH, using gene expression profiling of cultured
lymphocytes to compare patients with heritable disease and
asymptomatic familial control subjects who carry nonsense-
mediated decay-positive (NMD
1
)BMPR2 mutations (54).
The NMD pathway is responsible for degrading mRNA tran-
scripts that contain premature termination codons, thereby pre-
venting translation of unnecessary or harmful transcripts.
Patients with NMD
1
BMPR2 mutations are thought to develop
PAH because of haploinsufficiency and exhibited a “PAH pen-
etrance signature,” comprising 23 up-regulated and 12 down-
regulated genes. This was subjected to in silico and Connectivity
Map (cMap) analysis, revealing differences in the production of
reactive oxygen species between patients and unaffected rela-
tives. More importantly, the combination of gene expression pro-
files and bioinformatic analysis may identify novel pathways in
the pathogenesis of pulmonary vascular disease that are respon-
sive to U.S. Food and Drug Administration (FDA)–approved
drugs, the actions of which are already included in the cMap
database (54).
THERAPY AND CLINICAL TRIALS
The high mortality of PAH documented in registry studies
underlines the need for new treatments. The study by Stacher
and colleagues indicates that current treatments have little di-
rect impact on vascular remodeling (5). For example, plexi-
form lesions were more common in patients who had received
a prostanoid than in patients who had not. Nonetheless, drugs
based on current pathways remain the most likely to enter clinical
practice in 2013. In addition to the oral prostanoid selexipag (55),
a new dual endothelin antagonist (macitentan) (2) and a soluble
guanylate cyclase stimulator (riociguat) (3, 4) were both reported
to have positive clinical results in 2012.
The nitric oxide (NO)–cyclic guanylate cyclase signaling path-
way remains an attractive treatment target for PAH, at least in
adults. Baliga and colleagues have reported that dietary nitrate,
via conversion to nitrite by oral bacteria with nitrate reductase
capacity, and dietary nitrite reduced RV pressure and hypertrophy,
and pulmonary vascular remodeling in experimental PH (56). The
proposed mechanism is dependent on reduction of nitrite to form
NO by both endothelial NO synthase and xanthine oxidoreductase.
The authors suggest that exploitation of this mechanism (i.e., dietary
nitrate/nitrite supplementation) represents a viable, orally ac-
tive therapy for PH. Along these lines, inhaled nitrite is also
being examined as a potential therapy for PAH (ClinicalTrials.
gov identifier NCT01431313).
In contrast, the FDA has issued a warning about the use of
sildenafil in children with PAH, due to an apparent increase
in long-term mortality. The phosphodiesterase type 5 inhibitor
sildenafil is approved for use in the treatment of PAH in children
in Europe but not the United States. The FDA concerns are
based on a single-center, placebo-controlled trial that evalu-
ated the effects of sildenafil at low, medium, or high doses in
treatment-naive children who were diagnosed with IPAH or
PAH associated with congenital heart disease (57). Increased
mortality was reported in patients randomized to high-dose sil-
denafil monotherapy at 3 years when compared with lower dose
groups. There was no untreated control group. The FDA warn-
ing raised objections from the expert community and readers
are referred to their advice with respect to the continued use of
sildenafil in children and the future use of this drug (58).
The strategy of moving from vasorelaxing agents to drugs that
target directly the vascular remodeling in PAH has raised safety
concerns in humans. A striking example of this is the differential
effect of dasatinib in rodent models of PH and humans. Like
other RTK inhibitors, dasatinib is effective in attenuating PH
in monocrotaline and hypoxic rat models (59), but Montani
and colleagues detected an increase in PAH in patients taking
dasatinib for chronic myeloid leukemia (60). RTK inhibitors are
a large drug class and dasatinib is comparatively unselective.
Elucidating the mechanism of the association of PAH with
dasatinib would not only help steer the development of other
RTK inhibitors but also may provide important insight into the
pathophysiology of PAH.
CONCLUSIONS
The studies published over the last year have advanced but also
tested our understanding of the pathology and management of
pulmonary vascular disease. Considering all the data, including
the apparent anomalies (i.e., letting all the data speak), is the
secret to building on this foundation in the year to come.
Author disclosures are available with the text of this article at www.atsjournals.org.
References
1. Hassoun PM, Adnot S. Update in pulmonary vascular diseases 2011. Am
J Respir Crit Care Med 2012;185:1177–1182.
2. Rubin L, Pulido T, Channick R, Delcroix M, Galie N, Ghofrani H, Jansa
P, Le Brun F, Mehta S, Mittelholzer C, et al.Effectofmacitentan
on morbidity and mortality in pulmonary arterial hypertension
(PAH): results from the SERAPHIN trial [abstract]. Chest 2012;
142:1026A.
3. Ghofrani H, Galie N, Grimminger F, Humbert M, Keogh A, Langleben
D, Kilama MO, Neuser D, Rubin L. A randomized, double-blind,
placebo-controlled study (PATENT-1) [abstract]. Chest 2012;142:
1027A.
4. Ghofrani H, Grimminger F, Hoeper M, Kim N, Mayer E, Neuser D, Pena
J, Simonneau G, Wilkins M. Riociguat for the treatment of inoperable
chronic thromboembolic pulmonary hypertension: a randomized,
double-blind, placebo-controlled study (CHEST-1) [abstract]. Chest
2012;142:1023A.
5. Stacher E, Graham BB, Hunt JM, Gandjeva A, Groshong SD, McLaughlin
VV, Jessup M, Grizzle WE, Aldred MA, Cool CD, et al.Modernage
pathology of pulmonary arterial hypertension. Am J Respir Crit Care
Med 2012;186:261–272.
6. Dorfmüller P, Humbert M. Progress in pulmonary arterial hypertension
pathology: relighting a torch inside the tunnel. Am J Respir Crit Care
Med 2012;186:210–212.
26 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 188 2013
7. Ling Y, Johnson MK, Kiely DG, Condliffe R, Elliot CA, Gibbs JS,
Howard LS, Pepke-Zaba J, Sheares KK, Corris PA, et al. Changing
demographics, epidemiology, and survival of incident pulmonary arterial
hypertension: results from the pulmonary hypertension registry of
the United Kingdom and Ireland. AmJRespirCritCareMed2012;
186:790–796.
8. Thenappan T, Ryan JJ, Archer SL. Evolving epidemiology of pulmonary
arterial hypertension. Am J Respir Crit Care Med 2012;186:707–709.
9. West J, Niswender KD, Johnson JA, Pugh ME, Gleaves L, Fessel JP,
Hemnes AR. A potential role for insulin resistance in experimental
pulmonary hypertension. Eur Respir J 2013;41:861–871.
10. Benza RL, Miller DP, Barst RJ, Badesch DB, Frost AE, McGoon MD.
An evaluation of long-term survival from time of diagnosis in pulmo-
nary arterial hypertension from the REVEAL Registry. Chest 2012;
142:448–456.
11. Wu WH, Yang L, Peng FH, Yao J, Zou LL, Liu D, Jiang X, Li J, Gao L,
Qu JM, et al. Lower socioeconomic status is associated with worse
outcomes in pulmonary arterial hypertension. Am J Respir Crit Care
Med 2013;187:303–310.
12. Berger RM, Beghetti M, Humpl T, Raskob GE, Ivy DD, Jing ZC,
Bonnet D, Schulze-Neick I, Barst RJ. Clinical features of paediatric
pulmonary hypertension: a registry study. Lancet 2012;379:537–546.
13. Dunham I, Kundaje A, Aldred SF, Collins PJ, Davis CA, Doyle F,
Epstein CB, Frietze S, Harrow J, Kaul R, et al.; ENCODE Project
Consortium. An integrated encyclopedia of DNA elements in the
human genome. Nature 2012;489:57–74.
14. Parikh VN, Jin RC, Rabello S, Gulbahce N, White K, Hale A, Cottrill
KA, Shaik RS, Waxman AB, Zhang YY, et al. MicroRNA-21 inte-
grates pathogenic signaling to control pulmonary hypertension: re-
sults of a network bioinformatics approach. Circulation 2012;125:
1520–1532.
15. Caruso P, Dempsie Y, Stevens HC, McDonald RA, Long L, Lu R, White
K, Mair KM, McClure JD, Southwood M, et al. A role for miR-145 in
pulmonary arterial hypertension: evidence from mouse models and
patient samples. Circ Res 2012;111:290–300.
16. Pullamsetti SS, Doebele C, Fischer A, Savai R, Kojonazarov B, Dahal
BK, Ghofrani HA, Weissmann N, Grimminger F, Bonauer A, et al.
Inhibition of microRNA-17 improves lung and heart function in ex-
perimental pulmonary hypertension. Am J Respir Crit Care Med 2012;
185:409–419.
17. Savai R, Pullamsetti SS, Kolbe J, Bieniek E, Voswinckel R, Fink L,
Scheed A, Ritter C, Dahal BK, Vater A, et al. Immune and
inflammatory cell involvement in the pathology of idiopathic
pulmonary arterial hypertension. AmJRespirCritCareMed
2012;186:897–908.
18. Cavasin MA, Demos-Davies K, Horn TR, Walker LA, Lemon DD,
Birdsey N, Weiser-Evans MC, Harral J, Irwin DC, Anwar A, et al.
Selective class I histone deacetylase inhibition suppresses hypoxia-
induced cardiopulmonary remodeling through an antiproliferative
mechanism. Circ Res 2012;110:739–748.
19. Zhao L, Chen CN, Hajji N, Oliver E, Cotroneo E, Wharton J, Wang D,
Li M, McKinsey TA, Stenmark KR, et al. Histone deacetylation
inhibition in pulmonary hypertension: therapeutic potential of valproic
acid and suberoylanilide hydroxamic acid. Circulation 2012;126:
455–467.
20. Perros F, Dorfmüller P, Montani D, Hammad H, Waelput W, Girerd B,
Raymond N, Mercier O, Mussot S, Cohen-Kaminsky S, et al. Pulmonary
lymphoid neogenesis in idiopathic pulmonary arterial hypertension.
Am J Respir Crit Care Med 2012;185:311–321.
21. Huertas A, Tu L, Gambaryan N, Girerd B, Perros F, Montani D, Fabre
D, Fadel E, Eddahibi S, Cohen-Kaminsky S, et al. Leptin and
regulatory T-lymphocytes in idiopathic pulmonary arterial hypertension.
EurRespirJ2012;40:895–904.
22. Ormiston ML, Chang C, Long LL, Soon E, Jones D, Machado R, Treacy
C, Toshner MR, Campbell K, Riding A, et al. Impaired natural killer
cell phenotype and function in idiopathic and heritable pulmonary
arterial hypertension. Circulation 2012;126:1099–1109.
23. George MP, Champion HC, Gladwin MT, Norris KA, Morris A. Injection
drug use as a “second hit” in the pathogenesis of HIV-associated
pulmonary hypertension. AmJRespirCritCareMed2012;185:1144–
1146.
24. Spikes L, Dalvi P, Tawfik O, Gu H, Voelkel NF, Cheney P, O’Brien-
Ladner A, Dhillon NK. Enhanced pulmonary arteriopathy in simian
immunodeficiency virus–infected macaques exposed to morphine. Am
J Respir Crit Care Med 2012;185:1235–1243.
25. George MP, Champion HC, Simon M, Guyach S, Tarantelli R, Kling
HM, Brower A, Janssen C, Murphy J, Carney JP, et al. Physiologic
changes in a nonhuman primate model of HIV-associated pulmonary
arterial hypertension. Am J Respir Cell Mol Biol 2013;48:374–381.
26. Tu L, De Man FS, Girerd B, Huertas A, Chaumais MC, Lecerf F,
Franc¸ois C, Perros F, Dorfmüller P, Fadel E, et al. A critical role
for p130
Cas
in the progression of pulmonary hypertension in humans
and rodents. Am J Respir Crit Care Med 2012;186:666–676.
27. Yamamura A, Guo Q, Yamamura H, Zimnicka AM, Pohl NM, Smith
KA, Fernandez RA, Zeifman A, Makino A, Dong H, et al. Enhanced
Ca
21
-sensing receptor function in idiopathic pulmonary arterial
hypertension. Circ Res 2012;111:469–481.
28. Shen JZ, Young MJ. Corticosteroids, heart failure, and hypertension:
a role for immune cells? Endocrinology 2012;153:5692–5700.
29. Ranayhossaini D, Pagano PJ. TrACEing angiotensin II type 1 to right
ventricular hypertrophy: are the “sartans” a viable course to treating
pulmonary arterial hypertension? Am J Respir Crit Care Med 2012;
186:705–707.
30. de Man FS, Tu L, Handoko ML, Rain S, Ruiter G, Franc¸ ois C, Schalij I,
Dorfmüller P, Simonneau G, Fadel E, et al. Dysregulated renin–
angiotensin–aldosterone system contributes to pulmonary arterial
hypertension. Am J Respir Crit Care Med 2012;186:780–789.
31. Maron BA, Zhang YY, White K, Chan SY, Handy DE, Mahoney CE,
Loscalzo J, Leopold JA. Aldosterone inactivates the endothelin-B
receptor via a cysteinyl thiol redox switch to decrease pulmonary
endothelial nitric oxide levels and modulate pulmonary arterial hy-
pertension. Circulation 2012;126:963–974.
32. Austin ED, Cogan JD, West JD, Hedges LK, Hamid R, Dawson EP,
Wheeler LA, Parl FF, Loyd JE, Phillips JA III. Alterations in
oestrogen metabolism: implications for higher penetrance of familial
pulmonary arterial hypertension in females. Eur Respir J 2009;34:
1093–1099.
33. Tofovic SP, Salah EM, Mady HH, Jackson EK, Melhem MF. Estradiol
metabolites attenuate monocrotaline-induced pulmonary hyperten-
sion in rats. J Cardiovasc Pharmacol 2005;46:430–437.
34. Umar S, Rabinovitch M, Eghbali M. Estrogen paradox in pulmonary
hypertension: current controversies and future perspectives. Am J
Respir Crit Care Med 2012;186:125–131.
35. Lahm T, Albrecht M, Fisher AJ, Selej M, Patel NG, Brown JA, Justice
MJ, Brown MB, Van Demark M, Trulock KM, et al.17b-Estradiol
attenuates hypoxic pulmonary hypertension via estrogen receptor–
mediated effects. Am J Respir Crit Care Med 2012;185:965–980.
36. Mathai SC, Puhan MA, Lam D, Wise RA. The minimal important
difference in the 6-Minute Walk Test for patients with pulmonary
arterial hypertension. Am J Respir Crit Care Med 2012;186:428–433.
37. Gabler NB, French B, Strom BL, Palevsky HI, Taichman DB, Kawut
SM, Halpern SD. Validation of 6-minute walk distance as a surrogate
end point in pulmonary arterial hypertension trials. Circulation 2012;
126:349–356.
38. Rubin LJ. The 6-Minute Walk Test in pulmonary arterial hypertension:
how far is enough? Am J Respir Crit Care Med 2012;186:396–397.
39. Minai OA, Gudavalli R, Mummadi S, Liu X, McCarthy K, Dweik RA.
Heart rate recovery predicts clinical worsening in patients with
pulmonary arterial hypertension. Am J Respir Crit Care Med 2012;
185:400–408.
40. Hassoun PM. Drumming up prognostic significance in a heartbeat in
pulmonary arterial hypertension. Am J Respir Crit Care Med 2012;
185:354–355.
41. Ramos RP, Arakaki JS, Barbosa P, Treptow E, Valois FM, Ferreira EV,
Nery LE, Neder JA. Heart rate recovery in pulmonary arterial
hypertension: relationship with exercise capacity and prognosis. Am
Heart J 2012;163:580–588.
42. Kasner M, Westermann D, Steendijk P, Dröse S, Poller W, Schultheiss
HP, Tschöpe C. Left ventricular dysfunction induced by nonsevere
idiopathic pulmonary arterial hypertension: a pressure–volume
relation ship study. Am J Respir Crit Care Med 2012;186:181–189.
Pulmonary, Sleep, and Critical Care Updates 27
43. Mehari A, Gladwin MT, Tian X, Machado RF, Kato GJ. Mortality in
adults with sickle cell disease and pulmonary hypertension. JAMA
2012;307:1254–1256.
44. Mehari A, Alam S, Tian X, Cuttica MJ, Barnett CF, Miles G, Xu D,
Seamon C, Adams-Graves P, Castro OL, et al. Hemodynamic predictors
of mortality in adults with sickle cell disease. Am J Res pir Crit Care
Med 2013;187:840–847.
45. Skrok J, Shehata ML, Mathai S, Girgis RE, Zaiman A, Mudd JO,
Boyce D, Lechtzin N, Lima JA, Bluemke DA, et al. Pulmonary
arterial hypertension: MR imaging–derived first-pass bolus ki-
netic parameters are biomarkers for pulmonary hemodynamics,
cardiac function, and ventricular remodeling. Radiology 2012;263:
678–687.
46. Shehata ML, Harouni AA, Skrok J, Basha TA, Boyce D, Lechtzin N,
Mathai SC, Girgis R, Osman NF, Lima JA, et al. Regional and global
biventricular function in pulmonary arterial hypertension: a cardiac
MR imaging study. Radiology 2013;266:114–122.
47. Xu W, Koeck T, Lara AR, Neumann D, DiFilippo FP, Koo M, Janocha
AJ, Masri FA, Arroliga AC, Jennings C, et al. Alterations of cellular
bioenergetics in pulmonary artery endothelial cells. Proc Natl Acad
Sci USA 2007;104:1342–1347.
48. Marsboom G, Wietholt C, Haney CR, Toth PT, Ryan JJ, Morrow E,
Thenappan T, Bache-Wiig P, Piao L, Paul J, et al. Lung
18
F-
fluorodeoxyglucose positron emission tomography for diagnosis and
monitoring of pulmonary arterial hypertension. Am J Respir Crit Care
Med 2012;185:670–679.
49. Fonseca GH, Souza R, Salemi VM, Jardim CV, Gualandro SF. Pulmonary
hypertension diagnosed by right heart catheterisation in sickle cell
disease. Eur Respir J 2012;39:112–118.
50. Larkin EK, Newman JH, Austin ED, Hemnes AR, Wheeler L, Robbins
IM, West JD, Phillips JA III, Hamid R, Loyd JE. Longitudinal
analysis casts doubt on the presence of genetic anticipation in heritable
pulmonary arterial hypertension. AmJRespirCritCareMed2012;186:
892–896.
51. Aldred MA, Morrell NW. Waiting in anticipation: the genetics of
pulmonary arterial hypertension. Am J Respir Crit Care Med 2012;
186:820–821.
52. Cogan J, Austin E, Hedges L, Womack B, West J, Loyd J, Hamid R.
Role of BMPR2 alternative splicing in heritable pulmonary arterial
hypertension penetrance. Circulation 2012;126:1907–1916.
53. White RJ, Morrell NW. Understanding the low penetrance of bone
morphogenetic protein receptor 2 gene mutations: another needle
in the haystack. Circulation 2012;126:1818–1820.
54. Flynn C, Zheng S, Yan L, Hedges L, Womack B, Fessel J, Cogan J,
Austin E, Loyd J, West J, et al. Connectivity map analysis of
nonsense-mediated decay-positive BMPR2-related hereditary pul-
monary arterial hypertension provides insights into disease pene-
trance. Am J Respir Cell Mol Biol 2012;47:20–27.
55. Simonneau G, Torbicki A, Hoeper MM, Delcroix M, Karlócai K, Galiè
N, Degano B, Bonderman D, Kurzyna M, Efficace M, et al. Selexipag:
an oral, selective prostacyclin receptor agonist for the treatment of
pulmonary arterial hypertension. Eur Respir J 2012;40:874–880.
56. Baliga RS, Milsom AB, Ghosh SM, Trinder SL, Macallister RJ, Ahluwalia
A, Hobbs AJ. Dietary nitrate ameliorates pulmonary hypertension:
cytoprotective role for endothelial nitric oxide synthase and xanthine
oxidoreductase. Circulation 2012;125:2922–2932.
57. Barst RJ, Ivy DD, Gaitan G, Szatmari A, Rudzinski A, Garcia AE,
Sastry BK, Pulido T, Layton GR, Serdarevic-Pehar M, et al.A
randomized, double-blind, placebo-controlled, dose-ranging study of
oral sildenafil citrate in treatment-naive children with pulmonary ar-
terial hypertension. Circulation 2012;125:324–334.
58. Abman SH, Kinsella JP, Rosenzweig EB, Krishnan U, Kulik T, Mullen
M, Wessel DL, Steinhorn R, Adatia I, Hanna B, et al.; Pediatric
Pulmonary Hypertension Network (PPHNet). Implications of the U.S.
Food and Drug Administration warning against the use of sildenafil for
the treatment of pediatric pulmonary hypertension. Am J Respir Crit
Care Med 2013;187:572–575.
59. Pullamsetti SS, Berghausen EM, Dabral S, Tretyn A, Butrous E, Savai
R, Butrous G, Dahal BK, Brandes RP, Ghofrani HA, et al.Roleof
Src tyrosine kinases in experimental pulmonary hypertension. Arterioscler
Thromb Vasc Biol 2012;32:1354–1365.
60. Montani D, Bergot E, Günther S, Savale L, Bergeron A, Bourdin A, Bouvaist
H, Canuet M, Pison C, Macro M, et al. Pulmonary arterial hypertension
in patients treated by dasatinib. Circulation 2012;125:2128–2137.
28 AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE VOL 188 2013
... "Sickle cell acute lung disease, " the acute thoracic syndrome (ATS), is associated with the highest mortality of all disease-related complications and is one of the most common causes of hospitalization (6)(7)(8)(9). The pathophysiology of the chronic form of lung involvement, known as "sickle cell chronic lung disease, " is still not fully understood, and no specific therapeutic options are yet available (10). ...
Article
Full-text available
Background and objectiveSickle cell disease (SCD) is a very common autosomal recessive hemoglobinopathy leading to multiple pulmonary complications that are closely associated with mortality. The pathophysiology of chronic pulmonary involvement is not yet fully understood and no specific therapies are available.Methods The aim of this cross-sectional study was to characterize the lung function of children and young adolescents with SCD in a German single-center cohort and to extend conventional lung function testing by the use of a new imaging method. We performed spirometry and body plethysmography in 35 children and young adults with hemoglobin SS, SC, S/β-thalassemia as well as 50 controls. These data were compared with clinical characteristics and typical laboratory parameters of hemolysis and disease activity in SCD. To identify lung inhomogeneities, for example due to atelectasis, hyperinflation, air trapping or vascular occlusions, we used the promising new method of electrical impedance tomography (EIT) and calculated global inhomogeneity indices.ResultsLung function of patients with SCD was significantly reduced compared to that of healthy controls. When the result was found to be pathological, the most commonly observed type of breathing disorder was classified as restrictive. Laboratory parameters showed typical features of SCD including decreased levels of hemoglobin and hematocrit and elevated levels of leucocytes, platelets, lactate dehydrogenase and total bilirubin. However, there was no correlation between blood values and reduced lung function. Electrical impedance tomography (EIT) revealed no abnormalities in SCD patients compared to healthy controls. In particular, we were unable to demonstrate any regional inhomogeneities in lung ventilation.Conclusion In our study, SCD patients showed impaired lung function, with a relevant percentage of patients suffering from restrictive breathing disorder. Signs of obstruction could not be detected. Electrical impedance tomography (EIT) measurements revealed no unevenness that would suggest air entrapment, blockage of blood vessels, excessive inflation, obstruction, or other forms of lung disease. Additionally, the reduction in lung function observed in SCD patients was not related to the disease severity or laboratory test results.
... [1][2][3] Although many studies in patients or animal models have demonstrated the critical role of multiple growth factors or pathways (such as prostacyclin, nitric oxide, and endothelin) in pulmonary vascular remodeling, knowledge of the precise mechanism underlying this process in COPD remains limited. [4][5][6][7][8] As a subfamily of the nuclear receptor superfamily, NR4A contains three kinds of mammalian nuclear receptors: NR4A1 (Nurr77), NR4A2 (Nurr1), and NR4A3 (neuronderived orphan receptor 1[NOR1]). 9 Acting as a kind of earlyresponse gene, NOR1 could respond rapidly to various extracellular stimuli. ...
Article
Full-text available
Introduction Chronic hypoxia-induced pulmonary vascular remodeling is a feature of chronic obstructive pulmonary disease (COPD). Our previous reports indicate that neuron-derived orphan receptor 1 (NOR1) promoted pulmonary smooth muscle cell proliferation in vitro. But it remains unclear whether NOR1 participated into hypoxia-induced pulmonary vascular remodeling in COPD patients. Patients and methods For this study, we collected peripheral lung tissues of 26 male COPD patients with or without hypoxemia. We detected the pulmonary vascular remodeling in all the peripheral lung tissues. Primary human pulmonary arterial smooth muscle cells were also cultured in vitro and stimulated with hypoxia or normoxia. Cell proliferation and protein levels were detected. Results COPD patients with hypoxemia showed significantly enlarged pulmonary vessels wall thickness and increased protein levels of HIF-1α, smooth muscle actin, cyclin D1, and NOR1 when compared with those in normoxic patients. Moreover, hypoxia induced human pulmonary arterial smooth muscle cell proliferation and NOR1 overexpression in vitro. The plasmid-based NOR1 gene overexpression markedly promoted DNA synthesis and proliferation in hypoxia or normoxic cells. Human NOR1 gene-specific siRNA intensively suppressed DNA synthesis and proliferation. Transfection of NOR1 overexpression plasmid raised cyclin D1 protein levels, which could be significant inhibited by NOR1-specific siRNA or a CDK4/6 inhibitor PD0332991. Conclusion We concluded that NOR1 upregulation is associated with hypoxia-induced pulmonary vascular remodeling in COPD via promoting human pulmonary arterial smooth muscle cell proliferation.
... Untreated pulmonary arterial hypertension (PAH) is a progressive fatal disease (1,2). A common histopathology among patients with PAH suggests a consistent mechanism of disease progression likely involving similar endogenous regulatory substances (3)(4)(5)(6). Endothelin (ET)-1 is central to the pathophysiology of PAH. ET-1 has potent mitogenic effects and is abundantly expressed in the pulmonary vasculature (7)(8)(9)(10)(11)(12)(13). ET-1 expression is increased in the lungs of patients with primary pulmonary hypertension (PH) (14), and plasma levels of ET-1 are elevated in all cases of PAH, regardless of cause (15)(16)(17)(18)(19). Clinically, ET receptor antagonists (ERAs) in patients with PAH improve functional capacity, New York Heart Association (NYHA) functional class, time to clinical worsening, and perhaps mortality (20)(21)(22)(23)(24)(25)(26)(27). ...
Article
Pulmonary arterial hypertension (PAH) is a progressive fatal disease. Variable response and tolerability to PAH therapeutics suggests that genetic differences may influence outcomes. The endothelin pathway is central to pulmonary vascular function, and several polymorphisms and/or mutations in the genes coding for endothelin-1 (ET-1) and its receptors correlate with the clinical manifestations of other diseases. This study examined the interaction of ET-1 pathway polymorphisms and treatment responses of PAH patients treated with endothelin receptor antagonists (ERAs). 1198 PAH patients were prospectively enrolled from 45 US and Canadian PH centers or retrospectively from global sites participating in the Sitaxsentan To Relieve Impaired Exercise (STRIDE) trials. Comprehensive objective measures including a 6-minute walk test, Borg dyspnea score, functional class and laboratories were completed at baseline, prior to the initiation of ERAs and repeated serially. SNPs from ET-1 pathway candidate genes were selected from a completed genome wide association study (GWAS) performed on the study cohort. Patient efficacy outcomes were analyzed for a relationship between ET-1 pathway polymorphisms and clinical efficacy using pre-defined, composite positive and negative outcome measures in 715 European decent samples. A SNP (rs11157866) in the G-protein alpha and gamma subunits gene was significantly associated, accounting for multiple testing, with a combined improvement in functional class and 6 minute walk distance at 12 and 18 months and marginally significant at 24 months. ET-1 pathway associated polymorphisms may influence the clinical efficacy of ERA therapy for PAH. Further prospective studies are needed.
Article
As a transcription factor of the nuclear receptor superfamily, neuron-derived orphan receptor 1 (NOR1) is induced rapidly in response to various extracellular stimuli. But, it is still unclear its role in pulmonary artery smooth muscle cells proliferation. Human PASMCs were cultured in vitro and stimulated by serum. The special antisense oligodeoxynucleotides (AS-ODNs) were used to knockdown human NOR1 gene expression. Real-time PCR and Western-blot were used to evaluate the gene expression and protein levels. Fetal bovine serum (FBS) induced human PASMCs proliferation in a dose dependent manner. Furthermore, FBS promoted NOR1 gene expression in a dose dependent manner and a time dependent manner. 10% FBS induced a maximal NOR1 mRNA levels at 2 h. FBS also induced a significant higher NOR1 protein levels as compared with control. The NOR1 over-expressed plasmid significantly promoted DNA synthesis and cells proliferation. Moreover, the special AS-ODNs against human NOR1 not only prevented NOR1 expression but also inhibited DNA synthesis and cells proliferation significantly. The NOR1 over-expression plasmid could up-regulate cyclin D1 expression markedly, but the AS-ODNs inhibited cyclin D1 expression significantly. So, we concluded that NOR1 could promote human PASMCs proliferation. Cyclin D1 might be involved in this process.
Article
Full-text available
Background: Pulmonary hypertension (PH) in adults with sickle cell disease (SCD) is associated with early mortality, but no prior studies have evaluated quantitative relationships of mortality to physiological measures of pre- and postcapillary PH. Objectives: To identify risk factors associated with mortality and to estimate the expected survival in a cohort of patients with SCD with PH documented by right heart catheterization. Methods: Nine-year follow-up data (median, 4.7 yr) from the National Institutes of Health SCD PH screening study are reported. A total of 529 adults with SCD were screened by echocardiography between 2001 and 2010 with no exclusion criteria. Hemodynamic data were collected from 84 patients. PH was defined as mean pulmonary artery pressure (PAP) ≥ 25 mm Hg. Survival rates were estimated by the Kaplan-Meier method, and mortality risk factors were analyzed by the Cox proportional hazards regression. Measurements and main results: Specific hemodynamic variables were independently related to mortality: mean PAP (hazard ratio [HR], 1.61; 95% confidence interval [CI], 1.05-2.45 per 10 mm Hg increase; P = 0.027), diastolic PAP (HR, 1.83; 95% CI, 1.09-3.08 per 10 mm Hg increase; P = 0.022), diastolic PAP - pulmonary capillary wedge pressure (HR, 2.19; 95% CI, 1.23-3.89 per 10 mm Hg increase; P = 0.008), transpulmonary gradient (HR, 1.78; 95% CI, 1.14-2.79 per 10 mm Hg increase; P = 0.011), and pulmonary vascular resistance (HR, 1.44; 95% CI, 1.09-1.89 per Wood unit increase; P = 0.009) as risk factors for mortality. Conclusions: Mortality in adults with SCD and PH is proportional to the physiological severity of precapillary PH, demonstrating its prognostic and clinical relevance despite anemia-induced high cardiac output and less severely elevated pulmonary vascular resistance.
Article
Full-text available
Rationale: Lower socioeconomic status (SES) confers a heightened risk of common cardiovascular and pulmonary diseases and increased mortality. The association of SES with outcomes in patients with pulmonary arterial hypertension (PAH) is less clear. Objectives: To determine the association between SES and outcomes in patients with PAH. Methods: We performed a prospective cohort study at a national referral center for patients with PAH in China. Two hundred sixty-two consecutive incident patients aged 18 to 65 years with a diagnosis of idiopathic PAH were recruited between January 2007 and June 2011 and followed up until November 2011. The primary endpoint was all-cause mortality. An SES score for each patient was derived from their educational level, annual household income, occupation, and medical reimbursement rate. Measurements and main results: Patients with a lower SES had higher unadjusted mortality rates, with 3-year survival estimates of 50.1, 70.8, and 86.0% in increasing tertiles of SES (P for trend < 0.001). After adjustment for clinical features, hemodynamics, and type of PAH treatment, the hazard ratios for death were 2.98 (95% confidence interval, 1.51-5.89) in the lowest tertile of SES and 1.80 (95% confidence interval, 0.89-3.63) in the middle tertile of SES compared with the upper tertile (P for trend = 0.006). Conclusions: A lower SES is strongly associated with a higher risk of death in idiopathic PAH. This association was independent of clinical characteristics, hemodynamics, and treatment. Addressing the health disparities associated with a lower SES may improve the outcomes of patients with PAH.
Article
Full-text available
Purpose: To determine whether chronic pulmonary arterial pressure (PAP) elevation affects regional biventricular function and whether regional myocardial function may be reduced in pulmonary arterial hypertension (PAH) patients with preserved global right ventricular (RV) function. Materials and methods: After informed consent, 35 PAH patients were evaluated with right heart catheterization and cardiac magnetic resonance (MR) imaging and compared with 13 healthy control subjects. Biventricular segmental, section, and mean ventricular peak systolic longitudinal strain (E(LL)), as well as left ventricular (LV) circumferential and RV tangential strains were compared between PAH patients and control subjects and correlated with global function and catheterization of the right heart indexes. Spearman ρ correlation with Bonferroni correction was used. Multiple linear regression analysis was performed to determine predictors for regional myocardial function. Results: In the RV of PAH patients, longitudinal contractility was reduced at the basal, mid, and apical levels, and tangential contractility was reduced at the midventricular level. Mean RV E(LL) positively correlated with mean PAP (r = 0.62, P < .0014) and pulmonary vascular resistance index (PVRI) (r = 0.77, P < .0014). Mean PAP was a predictor of mean RV E(LL) (β = .19, P = .005) in a multiple linear regression analysis. In the LV, reduced LV longitudinal and circumferential contractility were noted at the base. LV anteroseptal E(LL) positively correlated with increased mean PAP (r = 0.5, P = .03) and septal eccentricity index (r = 0.5, P = .01). In a subgroup of PAH patients with normal global RV function, significantly reduced RV longitudinal contractility was noted at basal and mid anterior septal insertions, as well as the mid anterior RV wall (P < .05 for all). Conclusion: In PAH patients, reduced biventricular regional function is associated with increased RV afterload (mean PAP and PVRI). Cardiac MR imaging helps identify regional RV dysfunction in PAH patients with normal global RV function. Supplemental material: http://radiology.rsna.org/lookup/suppl/doi:10.1148/radiol.12111599/-/DC1.
Article
Pulmonary arterial hypertension (PAH) is a rare condition characterized by an elevated pulmonary arterial resistance that, if untreated, leads to right heart failure. PAH can be sporadic (idiopathic PAH, or primary pulmonary hypertension), familial or related to other conditions including connective tissue diseases, congenital heart disease, human immunodeficiency virus infection, portal hypertension, appetite suppressant exposure .... The incidence of idiopathic PAH has been estimated to be 2 cases per million per year, but this figure may be underestimated due to the lack of specificity of PAH clinical signs (mostly the dyspnea). Echocardiography is the investigation of choice for non invasive screening. Measurement of haemodynamic parameters during right-heart catheterization is mandatory to establish definite diagnosis (mean pulmonary artery pressure > 25 mmHg with a pulmonary artery wedge pressure < 15 mmHg). Recent advances in the management of PAH including prostacyclin derivatives and endothelin receptor antagonists have improved markedly the patients' prognosis. Novel treatments such as type 5 phosphodies terase inhibitors are promising in this setting. Lung transplantation is the last option for patients deteriorating despite optimal medical treatment.
Article
SESSION TYPE: Late-Breaking AbstractsPRESENTED ON: Tuesday, October 23, 2012 at 04:30 PM - 05:45 PMPURPOSE: Riociguat is the first member of a novel class of compounds, the soluble guanylate cyclase (sGC) stimulators. With a dual mode of action, it synergizes with endogenous nitric oxide (NO) and also directly stimulates sGC independent of NO availability. Riociguat restores the NO-sGC-cGMP pathway resulting in a significant improvement in pulmonary vascular hemodynamics and an increase in exercise ability. Here, we present results from the Phase III, multicenter, randomized, double-blind, placebo-controlled CHEST-1 study, which investigated the efficacy and safety of riociguat in patients with inoperable chronic thromboembolic pulmonary hypertension (CTEPH).METHODS: Patients with inoperable CTEPH (as assessed by an independent adjudication committee) or with persisting or recurrent pulmonary hypertension (PH) after pulmonary endarterectomy (PEA), and a mean pulmonary vascular resistance (PVR) >480 dyn·s/cm5 were enrolled in the trial. Treatment with supportive PH therapies was permitted but patients were treatment naïve with regards to pulmonary arterial hypertension-specific therapies. Patients were randomized to either placebo or oral riociguat, which was titrated from a starting dose of 1 mg three times daily (t.i.d.) according to systolic blood pressure (range 0.5-2.5 mg t.i.d.). The primary outcome measure was the change from baseline in 6-minute walking distance after 16 weeks of treatment. Secondary endpoints included change from baseline in PVR, functional class, time to clinical worsening, and safety and tolerability.RESULTS: The study has enrolled a total of 263 patients. Results will be available and presented at the time of the congress.CONCLUSIONS: The CHEST-1 study is the largest placebo-controlled trial to date in CTEPH and will assess the therapeutic benefit of the sGC stimulator riociguat.CLINICAL IMPLICATIONS: At present, no approved pharmacologic therapy exists for CTEPH and as a result there is an urgent unmet need in patients who are not eligible for PEA, or who have persistent or recurrent PH after PEA.DISCLOSURE: Hossein Ghofrani: Grant monies (from sources other than industry): Hossein A. Ghofrani has received sponsored grants over the past 3 years from the German Research Foundation, Excellence Cluster Cardiopulmonary Research and Germany Ministry for Education and Research, Grant monies (from industry related sources): Hossein A. Ghofrani has received industry-sponsored grants over the past 3 years from Bayer HealthCare AG, Aires, Encysive/Pfizer and Novartis, Consultant fee, speaker bureau, advisory committee, etc.: Hossein A. Ghofrani has relationships with the following drug companies: Bayer HealthCare AG, Actelion, Encysive, Pfizer, Ergonex, Novartis and GlaxoSmithKlineFriedrich Grimminger: Consultant fee, speaker bureau, advisory committee, etc.: Friedrich Grimminger has relationships with Bayer AGMarius Hoeper: Consultant fee, speaker bureau, advisory committee, etc.: Marius Hoeper has relationships with the following drug companies: Actelion, Bayer, Gilead, GlaxoSmithKline, Lilly, Novartis and PfizerNick Kim: Grant monies (from industry related sources): Nick H. Kim has received industry related grant monies from the following drug companies: Actelion, Gilead and United Therapeutics, Consultant fee, speaker bureau, advisory committee, etc.: Nick H. Kim has relationships with the following drug companies: Bayer (consultant), Gilead, United Therapeutics (advisory meeting)Eckhard Mayer: Consultant fee, speaker bureau, advisory committee, etc.: Eckhard Mayer has relationships with the following drug companies: Bayer Schering, Pfizer, ActelionDieter Neuser: Employee: Dieter Neuser is an employee of Bayer Pharma AGJanethe Pena: Employee: Janethe Pena is an employee of Bayer HealthCareGerald Simonneau: University grant monies: Gerald Simonneau has received university grant monies from the following drug companies: Actelion, Bayer, GlaxoSmithKline, Lilly, Novartis and Pfizer, Grant monies (from industry related sources): Gerald Simonneau has received industry related grant monies from the following drug companies: Actelion, Bayer, GlaxoSmithKline, Lilly, Novartis and Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: Gerald Simonneau has relationships with the following drug companies: Actelion, Bayer, GlaxoSmithKline, Lilly, Novartis and PfizerMartin Wilkins: Grant monies (from industry related sources): Martin Wilkins has received grant monies from Bayer, Consultant fee, speaker bureau, advisory committee, etc.: Martin Wilkins is a Chair on a research abstract panel for BayerRiociguat is an investigational new drug currently in process for registration submission to health authorities, after successful finalisation of an RCT in patients with PAH.Department of Internal Medicine, University Hospital Giessen and Marburg, Giessen, Germany.
Article
SESSION TYPE: Late-Breaking AbstractsPRESENTED ON: Tuesday, October 23, 2012 at 04:30 PM - 05:45 PMPURPOSE: Riociguat is the first member of a novel class of compounds, the soluble guanylate cyclase (sGC) stimulators. With a dual mode of action, it synergizes with endogenous nitric oxide (NO) and also directly stimulates sGC independent of NO availability. Therefore, riociguat may restore the NO-sGC-cGMP pathway. The Phase III, double-blind, randomized, placebo-controlled PATENT-1 study investigated the efficacy and safety of riociguat in patients with symptomatic pulmonary arterial hypertension (PAH).METHODS: Treatment-naïve patients and patients pre-treated with endothelin receptor antagonists (ERAs) or prostanoids (inhaled or subcutaneous) were eligible. Patients were assigned to placebo or riociguat administered according to a titration regimen (starting dose: 1 mg three times daily [t.i.d.]; range: 0.5-2.5 mg t.i.d.). The primary outcome was the placebo-corrected change from baseline in 6-minute walking distance (6MWD) at week 12. Secondary endpoints included the change from baseline in pulmonary vascular resistance (PVR), NT-proBNP, functional class, clinical worsening, living with pulmonary hypertension questionnaire and Borg dyspnea score.RESULTS: Overall, 445 patients were randomized. Preliminary analysis showed an increase in 6MWD from baseline of 35.8 m with riociguat versus placebo (95% CI 20.1-51.5 m, p<0.0001). Predefined exploratory analyses indicated that riociguat improved 6MWD in pretreated patients (+35.7 m [95% CI 15.0-56.3 m]) as well as treatment-naïve patients (+38.4 m [95% CI 14.5-62.3 m]). Significant improvements were also seen in PVR (p<0.0001), NT-proBNP (p<0.0001), functional class (p=0.003), clinical worsening (p=0.0046), living with pulmonary hypertension questionnaire (p=0.002), and Borg dyspnea score (p=0.002). Riociguat was well tolerated and had a favorable safety profile.CONCLUSIONS: In patients with PAH, riociguat was well tolerated, significantly improved 6MWD, and consistently improved clinically relevant secondary efficacy endpoints.CLINICAL IMPLICATIONS: Riociguat represents a new treatment option for patients with PAH. Efficacy of riociguat was consistently shown in both treatment-naïve patients and patients pre-treated with ERA or inhaled or subcutaneous prostanoid monotherapy. PATENT-2 will evaluate the long-term safety of riociguat in patients with PAH.DISCLOSURE: Hossein Ghofrani: Grant monies (from sources other than industry): Hossein A. Ghofrani has received sponsored grants over the past 3 years from the German Research Foundation, Excellence Cluster Cardiopulmonary Research and Germany Ministry for Education and Research, Grant monies (from industry related sources): Hossein A. Ghofrani has received industry-sponsored grants over the past 3 years from Bayer HealthCare AG, Aires, Encysive/Pfizer and Novartis, Consultant fee, speaker bureau, advisory committee, etc.: Hossein A. Ghofrani has relationships with the following drug companies: Bayer HealthCare AG, Actelion, Encysive, Pfizer, Ergonex, Novartis and GlaxoSmithKlineNazzareno Galie: University grant monies: Nazzareno Galie has relationships with the following drug companies: Actelion, Pfizer, Bayer-Schering, GlaxoSmithKline and Novartis, Consultant fee, speaker bureau, advisory committee, etc.: Nazzareno Galie has relationships with the following drug companies: Actelion, Pfizer, Bayer-Schering, GlaxoSmithKline and NovartisFriedrich Grimminger: Consultant fee, speaker bureau, advisory committee, etc.: Friedrich Grimminger has relationships with Bayer AGMarc Humbert: Consultant fee, speaker bureau, advisory committee, etc.: Marc Humbert has relationships with drug companies including Actelion, Aires, Bayer, GlaxoSmithKline, Novartis, and Pfizer. In addition to being investigator in trials involving these companies, relationships include consultancy service and membership of scientific advisory boardsAnne Keogh: University grant monies: Anne Keogh has received university grant monies from the following drug companies: Actelion, GlaxoSmithKline, Pfizer, United Therapeutics and BayerDavid Langleben: Grant monies (from industry related sources): David Langleben has received industry sponsored grant monies from Bayer, Consultant fee, speaker bureau, advisory committee, etc.: David Langleben has relationships with BayerMichael Ochan Kilama: Employee: Michael Ochan Kilama is an employee of Bayer HealthCare, Global Clinical Development. He is the company medical expert (research physician) for the PATENT study.Dieter Neuser: Employee: Dieter Neuser is an employee of Bayer Pharma AGLewis Rubin: Consultant fee, speaker bureau, advisory committee, etc.: Lewis J. Rubin has relationships with the following drug companies: United therapeutics, Actelion, Pfizer, Lung LLC, Gilead, GlaxoSmithKline and BayerRiociguat is an investigational new drug currently in process for registration submission to health authorities, after successful finalisation of an RCT in patients with PAH.Department of Internal Medicine, University Hospital Giessen and Marburg, Giessen, Germany.
Article
SESSION TYPE: Late-Breaking AbstractsPRESENTED ON: Tuesday, October 23, 2012 at 04:30 PM - 05:45 PMPURPOSE: Current oral PAH therapies have been approved based on short-term trials with exercise capacity as the primary endpoint. In this event-driven trial, the effect of macitentan, a novel dual endothelin receptor antagonist (ERA) with enhanced tissue penetration, was assessed on a primary endpoint of morbidity and mortality in PAH patients.METHODS: In this double-blind, placebo-controlled, Phase III, event-driven study, patients (≥12 years) with symptomatic PAH were randomized (1:1:1) to placebo, macitentan 3mg or macitentan 10mg, once daily. Stable background oral or inhaled PAH therapy, other than an ERA, was allowed. The primary endpoint was time from treatment initiation to first morbidity or mortality event in all randomized patients. This composite endpoint, defined as death, atrial septostomy, lung transplantation, initiation of intravenous/subcutaneous prostanoids or worsening of PAH, was blindly and independently adjudicated. Secondary endpoints included the composite of mortality due to PAH or hospitalization for PAH.RESULTS: 742 patients with PAH were randomized to placebo (n=250), macitentan 3mg (n=250) or macitentan 10mg (n=242); mean treatment duration was 85.3, 99.5 and 103.9 weeks, respectively. Macitentan reduced the risk of occurrence of morbidity and mortality events versus placebo by 30% in the 3mg group (97.5%CI:4-48%;P=0.0108) and 45% in the 10 mg group (97.5%CI:24-61%;P<0.0001). The effect of macitentan on this endpoint was observed irrespective of background PAH therapy (mainly phosphodiesterase type-5 inhibitors); risk reduction for macitentan 3mg and 10mg was 17% (95%CI:-16-41%) and 38% (95%CI:11-57%) in the presence of background PAH therapy and 47% (95%CI:15-66%) and 55% (95%CI:28-72%) in the absence of background PAH therapy. Macitentan 3mg and 10mg also reduced the risk of the secondary endpoint, mortality due to PAH/hospitalization for PAH, by 33% (97.5%CI:3-54%;P=0.0146) and 50% (97.5%CI:25-67%;P<0.0001), respectively. Macitentan was well tolerated, with similar incidences of elevated liver aminotransferases and peripheral edema across all groups. Adverse events more frequently associated with macitentan versus placebo were headache, nasopharyngitis and anemia.CONCLUSIONS: Macitentan demonstrated a significant effect on the combined endpoint of morbidity and mortality, with a favorable safety profile, in the largest and longest randomized controlled trial of therapy in PAH to date.CLINICAL IMPLICATIONS: Macitentan has the potential to improve long-term outcomes in PAH patients.DISCLOSURE: Lewis Rubin: Consultant fee, speaker bureau, advisory committee, etc.: Actelion Pharmaceuticals Ltd, Consultant fee, speaker bureau, advisory committee, etc.: Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: United Therapeutics, Consultant fee, speaker bureau, advisory committee, etc.: Lung LLC, Consultant fee, speaker bureau, advisory committee, etc.: Gilead, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: Bayer, Consultant fee, speaker bureau, advisory committee, etc.: GeNoTomás Pulido: Consultant fee, speaker bureau, advisory committee, etc.: Actelion , Consultant fee, speaker bureau, advisory committee, etc.: Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: Lily, Consultant fee, speaker bureau, advisory committee, etc.: Bayer, Grant monies (from sources other than industry): National Heart Insitute, Grant monies (from industry related sources): Actelion, Grant monies (from industry related sources): Bayer, Grant monies (from industry related sources): United TherapeuticsRichard Channick: Grant monies (from industry related sources): Actelion, Consultant fee, speaker bureau, advisory committee, etc.: ActelionMarion Delcroix: Grant monies (from industry related sources): Actelion, Grant monies (from industry related sources): GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: United Therapeutics, Consultant fee, speaker bureau, advisory committee, etc.: BayerNazzareno Galie: Grant monies (from industry related sources): Actelion, Grant monies (from industry related sources): Pfizer, Grant monies (from industry related sources): Eli Lilly, Grant monies (from industry related sources): Bayer, Grant monies (from industry related sources): GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: Eli Lilly, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: BayerHossein-Ardeschir Ghofrani: Grant monies (from industry related sources): Actelion, Grant monies (from industry related sources): Pfizer, Grant monies (from industry related sources): Bayer, Grant monies (from industry related sources): Ergonex, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: Bayer, Consultant fee, speaker bureau, advisory committee, etc.: Ergonex, Consultant fee, speaker bureau, advisory committee, etc.: Gilead, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: Merck, Consultant fee, speaker bureau, advisory committee, etc.: NovartisPavel Jansa: Grant monies (from industry related sources): Actelion, Consultant fee, speaker bureau, advisory committee, etc.: United Therapeutics, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: AOP Orphan, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: BayerFranck-Olivier Le Brun: Employee: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: ActelionSanjay Mehta: Grant monies (from industry related sources): Actelion, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: Takeda Nycomed, Grant monies (from sources other than industry): Heart & Stroke Foundation of Ontario/Canada (HSFO/C), Grant monies (from industry related sources): Canadian Institute of Health Research / CIHR, Grant monies (from sources other than industry): Ontario Thoracic Society (OTS), Consultant fee, speaker bureau, advisory committee, etc.: Actelion Pharmaceuticals Canada, Consultant fee, speaker bureau, advisory committee, etc.: Takeda - Nycomed, Consultant fee, speaker bureau, advisory committee, etc.: Boehringer-Ingelheim CanadaCamilla Mittelholzer: Employee: Actelion, Shareholder: ActelionLoic Perchenet: Employee: Actelion, Shareholder: ActelionBkS Sastry: Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKlineOlivier Sitbon: Grant monies (from industry related sources): GlaxoSmithKline, Grant monies (from industry related sources): Actelion, Grant monies (from industry related sources): Pfizer, Grant monies (from industry related sources): bayer, Grant monies (from industry related sources): Eli Lilly, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: United Therapuetics, Consultant fee, speaker bureau, advisory committee, etc.: Eli Lilly, Consultant fee, speaker bureau, advisory committee, etc.: United Therapuetics, Consultant fee, speaker bureau, advisory committee, etc.: BayerRogerio Souza: Grant monies (from industry related sources): Actelion, Consultant fee, speaker bureau, advisory committee, etc.: ActelionAdam Torbicki: Grant monies (from industry related sources): Actelion, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: Eli Lilly, Consultant fee, speaker bureau, advisory committee, etc.: Bayer, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: AOP Orphan Pharmaceutics, Grant monies (from industry related sources): Bayer, Grant monies (from industry related sources): United Therapeutics, Grant monies (from industry related sources): AOP Orphan Pharmaceutics, Grant monies (from industry related sources): Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: Bristol Myers SquibbGerald Simonneau: Grant monies (from industry related sources): Actelion, Grant monies (from industry related sources): GlaxoSmithKline, Grant monies (from industry related sources): Pfizer, Grant monies (from industry related sources): Eli Lilly, Consultant fee, speaker bureau, advisory committee, etc.: Actelion, Consultant fee, speaker bureau, advisory committee, etc.: GlaxoSmithKline, Consultant fee, speaker bureau, advisory committee, etc.: Pfizer, Consultant fee, speaker bureau, advisory committee, etc.: Eli Lilly, Consultant fee, speaker bureau, advisory committee, etc.: United Therapeutics, Consultant fee, speaker bureau, advisory committee, etc.: NovartisMacitentan is under investigation for the treatment of pulmonary arterial hypertension. SERAPHIN, the Phase 3 study reported here, is the registration trial for macitentan in this indication. University of California, San Diego, CA.
Article
Rationale: Pulmonary arterial hypertension (PAH) is increased in HIV, but its pathogenesis is not fully understood. Non-human primates infected with simian immunodeficiency virus (SIV) or SIV-HIV chimeric virus (SHIV) exhibit histologic changes characteristic of human PAH, but whether hemodynamic changes accompany this pathology is unknown. Repeated measurements of pulmonary artery pressures would permit longitudinal assessment of disease development and provide insight into pathogenesis. Objective: To test the hypothesis that SIV and SHIV-infected macaques develop physiologic manifestations of PAH. Methods: We performed right heart catheterizations, echocardiography and CT scans in macaques infected with either SIV (delta B670) or SHIV (89.6P) and compared right heart and pulmonary artery pressures, as well as pulmonary vascular changes on CT scans to those in uninfected control animals. Measurements and Main Results: Right atrial, right ventricular systolic, and pulmonary artery pressures (PAP) were significantly elevated in 100% of macaques infected with either SIV or SHIV compared to controls with no difference in pulmonary capillary wedge pressure. PAP were increased as early as 3 months post-SIV infection. Radiographic evidence of pulmonary vascular pruning was also found. Both SIV and SHIV-infected macaques had histologic changes in pulmonary arteries predominantly consisting of intimal and medial hyperplasia. Conclusions: This report is the first to demonstrate SHIV and SIV-infected macaques develop pulmonary hypertension at a high frequency, with physiologic changes occurring as early as 3 months post-infection. These studies establish an important non-human primate model of HIV-associated PAH that will be useful in studies of disease pathogenesis and efficacy of interventions.
Article
Pulmonary arterial hypertension (PAH) contributes to disability and death in children with diverse cardiac, pulmonary or systemic diseases, and therapeutic options are currently limited. Data from adult studies provide the basis for most PAH-specific therapies, however, many of these medications are commonly used in children on an off-label basis due to the life-threatening nature of PAH. Although currently approved for use in adult PAH, sildenafil is used extensively off-label for the treatment of neonates, infants and children with PAH. Past studies have generally suggested favorable effects and outcomes in infants and young children with PAH, but these reports are generally uncontrolled observations, except for one single center trial for persistent pulmonary hypertension of the newborn. Despite extensive clinical experience with sildenafil therapy in children and approval by the EMA for its pediatric use in Europe, the FDA recently issued a warning against the use of sildenafil for pediatric PAH between 1 and 17 years of age due to an apparent increase in mortality during long-term therapy. Although these data are extremely limited, this FDA review challenges the pediatric PAH community to further assess the efficacy and safety of sildenafil, especially with chronic treatment. Although low doses of sildenafil are likely safe in pediatric PAH, further studies should carefully examine its role in the long-term therapy of children, especially with diverse causes of PAH. Pediatric patients with PAH require close surveillance and frequent monitoring and persistent sildenafil monotherapy is likely insufficient with disease progression.