You are on page 1of 16

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Darren B. Taichman, M.D., Ph.D., Editor

Pulmonary Arterial Hypertension


Paul M. Hassoun, M.D.​​

P
ulmonary hypertension is a syndrome characterized by marked From the Division of Pulmonary and Crit-
remodeling of the pulmonary vasculature and a progressive rise in the pul- ical Care Medicine, Department of Medi-
cine, Johns Hopkins University School of
monary vascular load, leading to hypertrophy and remodeling of the right Medicine, Baltimore. Dr. Hassoun can be
ventricle. Death results from right ventricular failure if pulmonary hypertension is contacted at ­phassoun@​­jhmi​.­edu or at
left untreated. Pulmonary hypertension is currently defined hemodynamically by the Division of Pulmonary and Critical
Care Medicine, Johns Hopkins University
a mean pulmonary arterial pressure of higher than 20 mm Hg at rest, as measured School of Medicine, 1830 E. Monument
by right heart catheterization.1 Precapillary pulmonary hypertension due to pulmo- St., Baltimore, MD 21287.
nary vascular disease is further defined by an elevation in pulmonary vascular N Engl J Med 2021;385:2361-76.
resistance of at least 3 Wood units (WU), in contrast to isolated postcapillary DOI: 10.1056/NEJMra2000348
pulmonary hypertension, in which the pulmonary vascular resistance is less than Copyright © 2021 Massachusetts Medical Society.

3 WU and the elevation in the mean pulmonary arterial pressure is due to elevated CME
filling pressures on the left side of the heart. at NEJM.org
The several forms of pulmonary hypertension are categorized into five clinical
groups (Fig. 1). This review focuses on the relatively rare form of pulmonary arte-
rial hypertension (group 1). Immense progress has been achieved in gaining an
understanding of the mechanisms, natural history, and genetic features of pulmo-
nary arterial hypertension and in establishing targeted therapy. A full appreciation
of the pathophysiology of the syndrome is important, since the diagnosis requires
a thorough clinical investigation to rule out other, more common forms of pulmo-
nary hypertension, for which treatment of the underlying disease should be the
primary goal.
The first anatomical description of pulmonary hypertension is credited to von
Romberg.2 However, it is the advent of human right heart catheterization, first
performed by Forssmann on himself in 1929,3 that led to a flurry of physiological
observations on the heart and pulmonary circulation by Cournand and Richards
in the 1940s. The three investigators received the Nobel Prize in Physiology or
Medicine in 1956 for their seminal work. In his illuminating Nobel Lecture, Richards
noted that, as a result of their collective work, “many forms and degrees of failure
were defined, and their responses to treatment measured.”4
In 1951, Dresdale, a disciple of Cournand and Richards, and colleagues pre-
sented the first case series of patients with pulmonary hypertension of unknown
cause, defined as “primary pulmonary hypertension.”5 Greater awareness of the
disease developed in the 1960s, after an epidemic of primary pulmonary hyperten-
sion that was associated with the use of the appetite suppressant aminorex.6 This
prompted the World Health Organization to convene a first meeting of pulmonary
hypertension experts in 1973, to standardize the clinical and pathological nomen-
clature of primary pulmonary hypertension, the first attempt at an organized
classification.7 The first and second meetings about pulmonary hypertension were
separated by 25 years, but thereafter the World Symposium on Pulmonary Hyper-
tension (WSPH) was convened every 5 years. The meetings brought further refine-
ment to the classification of pulmonary hypertension, with five distinct groups

n engl j med 385;25  nejm.org  December 16, 2021 2361


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

1 PAH
Subcategory Clinical characteristics
1.1 Idiopathic PAH • Symptoms and signs of PAH (exertional dyspnea,
2 PH due to left-sided heart disease
1.2 Heritable PAH syncope, elevated jugular-vein pressure, pedal
Subcategory
edema)
1.3 Drug- and toxin-induced PAH
• Signs of connective tissue disease, HIV infection, 2.1 PH due to heart failure with preserved LVEF
1.4 PAH associated with: or liver disease 2.2 PH due to heart failure with reduced LVEF
1.4.1 Connective-tissue disease
• Echocardiographic signs limited to right heart 2.3 Valvular heart disease
1.4.2 HIV infection chambers
1.4.3 Portal hypertension 2.4 Congenital or acquired cardiovascular conditions
• Other features (history of substance abuse [e.g., leading to postcapillary PH
1.4.4 Congenital heart disease methamphetamines], exposure to drugs or toxins
1.4.5 Schistosomiasis [e.g., fenfluramine, dexfenfluramine, dasatinib], Clinical characteristics
1.5 PAH long-term responders to calcium- family history) • Symptoms and signs of left-sided or valvular heart
channel blockers disease
Hemodynamic profile
1.6 PAH with overt features of venous or • Echocardiographic signs of left-sided or valvular
capillary involvement (PVOD or PCH) • Precapillary PH (mPAP >20 mm Hg, PAWP ≤15 heart disease in addition to right-sided heart signs
1.7 Persistent PH of the newborn mm Hg, PVR ≥3 WU)
Hemodynamic profile
• Isolated postcapillary PH (mPAP >20 mm Hg, PAWP
>15 mm Hg, PVR <3 WU)
mPAP
>20 mm Hg • Combined precapillary and postcapillary PH (mPAP
>20 mm Hg, PAWP >15 mm Hg, PVR ≥3 WU)

COPD, ILD, OSA


3 PH due to lung disease, hypoxia, or both
Subcategory
3.1 Obstructive lung disease
3.2 Restrictive lung disease
Pulmonary vascular remodeling, 3.3 Other lung disease with mixed restrictive–
plexiform lesions obstructive pattern
3.4 Hypoxia without lung disease
3.5 Developmental lung disorders
Clot
Clinical characteristics
• Predominant clinical feature: chronic hypoxia at rest
or with exercise
• Abnormal pulmonary-function tests and chest
RV hypertrophy, imaging
dilatation, and • Abnormal polysomnogram
remodeling Left-sided
valvular diseases, Hemodynamic profile
cardiomyopathies • Precapillary PH unless associated with coexisting
condition (e.g., left-sided heart disease)

5 PH with multifactorial or unclear mechanisms


Subcategory 4 PH due to pulmonary-artery obstructions
5.1 Hematologic disorders Subcategory
5.2 Systemic and metabolic disorders (e.g., pulmonary Langerhans-cell 4.1 Chronic thromboembolic PH
histiocytosis, Gaucher’s disease, glycogen storage disease,
neurofibromatosis, and sarcoidosis) 4.2 Other pulmonary-artery obstructions
5.3 Others (e.g., chronic renal failure with or without hemodialysis and fibrosing Clinical characteristics
mediastinitis)
• Symptoms and signs of PH
5.4 Complex congenital heart disease • Abnormal ventilation–perfusion scan, computed
Clinical characteristics tomographic angiography, or both
• Symptoms and signs of underlying disease (e.g., anemia, metabolic disorders) • Cancer

Hemodynamic profile Hemodynamic profile


• Precapillary PH (e.g.,hemoglobinopathies or myeloproliferative disease) • Precapillary PH unless associated with coexisting
• Isolated postcapillary PH or combined precapillary and postcapillary PH condition (e.g., left-sided heart disease)

Figure 1. Clinical Classification of Pulmonary Hypertension (PH).


The PH classification based on the 2018 meeting of the World Symposium on Pulmonary Hypertension is shown, along with the clinical
characteristics and hemodynamic profile of each group.1 COPD denotes chronic obstructive pulmonary disease, HIV human immunode-
ficiency virus, ILD interstitial lung disease, LVEF left ventricular ejection fraction, mPAP mean pulmonary arterial pressure, OSA ob-
structive sleep apnea, PAH pulmonary arterial hypertension, PAWP pulmonary arterial wedge pressure, PCH pulmonary capillary hem-
angiomatosis, PVOD pulmonary veno-occlusive disease, PVR pulmonary vascular resistance, RV right ventricular, and WU Wood units.

2362 n engl j med 385;25 nejm.org December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

based on similar clinical and pathological find- ly in developing countries,13 it is estimated that
ings and treatment responses (for a discussion there are 25 cases of congenital heart disease–
of the historical background, see the Supple- associated pulmonary arterial hypertension per
mentary Appendix, available with the full text of 1 million population worldwide.14 Valvular and
this article at NEJM.org). Idiopathic pulmonary left-sided heart disease are much more com-
arterial hypertension has replaced the term pri- mon,15,16 and more than 100 million persons
mary pulmonary hypertension, in recognition of may have pulmonary hypertension due to left-
the hemodynamic and clinical similarities to sided heart disease (group 2) worldwide.
other conditions that directly affect the pulmo- Similarly, pulmonary hypertension compli-
nary arterial vasculature and for which targeted cates chronic lung diseases, such as chronic
therapy is available. obstructive pulmonary disease (worldwide bur-
The recognition that some persons have a den, >500 million cases) and interstitial lung
genetic predisposition to the disorder (familial disease (estimated incidence, 10 to 70%); the
pulmonary arterial hypertension) led to the land- prevalence is increased among patients with ad-
mark discovery of mutations in the gene encod- vanced disease.14 In addition, more than 140 mil-
ing bone morphogenetic protein (BMP) receptor lion persons live at high altitude (above 2500 m),17
type 2 (BMPR2).8,9 Since 80% of cases of familial but the prevalence of pulmonary hypertension
pulmonary arterial hypertension and up to 20% due to chronic hypoxia among persons living in
of sporadic cases have germline BMPR2 muta- high-altitude areas or persons relocating to such
tions, and since additional mutations in various areas is unclear. Furthermore, pulmonary hyper-
genes have been identified, the term familial tension complicates highly prevalent viral infec-
pulmonary arterial hypertension was subsequent- tions (e.g., human immunodeficiency virus [HIV]
ly changed to heritable pulmonary arterial hyper- infection) and parasitic diseases (e.g., schistoso-
tension. miasis), as well as hemoglobinopathies such as
In the first two decades of this century, a sickle cell disease and thalassemia; therefore,
flurry of novel oral, injectable, and inhaled drugsvery large numbers of patients are affected in
emerged, prompted by a growing interest in and low- and middle-income areas of Africa, Asia,
understanding of pulmonary arterial hyperten- and South and Central America.18,19
sion. The development of these drugs followed Thus, it is estimated that 1% of the world
and was based on various well-conducted, placebo- population and up to 10% of persons older than
controlled studies (Fig. S1 in the Supplementary 65 years of age have pulmonary hypertension.14
Appendix). Moreover, 80% of these persons live in develop-
ing countries and, because of prohibitive cost,20
lack of approved drugs, or limited access to
A Gl ob a l He a lth Probl em
necessary medical and surgical support (e.g., for
The prevalence of pulmonary hypertension var- the treatment of chronic thromboembolic pul-
ies according to the WSPH group classification. monary hypertension [group 4]), are unlikely to
Pulmonary arterial hypertension (group 1) af- receive therapy.12,14
fects 25 persons (most of whom are women) per
1 million population in Western countries, with Pathol o gic a l Fe at ur e s
an annual incidence of 2 to 5 cases per million.10
The disease is most severe in elderly men, al- The histologic features of pulmonary arterial
11

though it is less common in this population, hypertension are complex and variable because
which is more likely to have group 2 disease. For of the multiplicity of underlying diseases. How-
other groups in the pulmonary hypertension ever, there are common pathological features of
classification, the prevalence varies according to the disorder, such as remodeling of the three
the cause and disease state but is likely to be layers of the distal pulmonary vasculature
greatly underestimated worldwide.12 (Fig. 2), which involves uncontrolled growth of
Many widespread diseases of the cardiopul- endothelial and smooth-muscle cells and fibro-
monary systems are complicated by pulmonary blasts,22 and infiltration of inflammatory cells,23
hypertension, which greatly increases morbidity which affects primarily precapillary vessels with
and mortality. Owing to the high prevalence of a caliber of 50 to 500 μm. There is also exten-
congenital heart diseases worldwide, particular- sion of the smooth-muscle cell layer to typically

n engl j med 385;25  nejm.org  December 16, 2021 2363


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A Shared Features of Vascular Remodeling in PAH

Histologic Appearance of Vascular Remodeling


Normal distal pulmonary artery (50-500 µm)
B C

Intima Media Adventitia

Endothelium IEL EEL


SMC
Plexiform lesion
formation
Luminal narrowing
or occlusion
D E
Intimal fibrosis
EC apoptosis and proliferation

Medial hypertrophy
SMC hyperplasia

Inflammatory-cell
recruitment
Fibroblast proliferation
Collagen disruption
Remodeled pulmonary vessel in PAH

F Genes and Proteins Implicated in PAH


Heritable PAH

BMP9, BMP10
(GDF2, BMP10)

K2p3.1 Aquaporin 1 KATP PVOD and PCH


Endoglin (KCNK3) (AQP1) (ABCC8)
(ENG)
P P
P P
K+ K+
Mutation in: Caveolin 1 H2O
80% of inherited cases BMPR-II ACVRL1 (CAV1)
(BMPR2) (ACVRL1)
20% of sporadic cases
P
SMAD8 GCN2
(SMAD9)
H+ ATP13A3 SMAD4 (EIF2AK4)
(ATP13A3) Nucleus
(SMAD4)
Transcription SMAD8 P
factors Gene expression
ATP ADP TBX4 SMAD4
SOX17 TBX4
(TBX4) BRE
Endosome

SOX17 Integrated stress response


(SOX17)

2364 n engl j med 385;25 nejm.org December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

Figure 2 (facing page). Pathobiologic Features of Pulmonary nonmuscularized distal capillaries. Postcapillary
Arterial Hypertension. vessels with similar vein remodeling may be in-
Panel A shows a normal distal pulmonary artery and a PAH ves- volved in specific syndromes, such as pulmonary
sel characterized by remodeling of the three vessel-wall layers veno-occlusive diseases and pulmonary capillary
(intima, media, and adventitia [EEL denotes external elastic lami- hemangiomatosis, scleroderma-associated pulmo-
na, IEL internal elastic lamina, and SMC smooth-muscle cells]).
nary arterial hypertension,24 chronic thrombo-
Endothelial-cell (EC) apoptosis and proliferation cause intimal
thickening, smooth-muscle cell hyperplasia causes medial embolic pulmonary hypertension,25 and group 2
thickening, and infiltration of inflammatory cells such as mono- cardiac diseases in which vascular remodeling
cytes, B and T lymphocytes, and dendritic cells, along with colla- may start in the postcapillary compartment.26 In
gen disruption, contribute to adventitial remodeling. In Panel B, situ thrombosis involving small muscular arteries
an elastic stain of a distal pulmonary vessel shows marked nar-
has long been recognized22 as being due to plate-
rowing of the arterial lumen, with concentric laminar intimal
proliferation and fibrosis, and destruction of the IEL (arrow). let activation and loss of endothelial integrity.27
The elastic stain in Panel C shows total occlusion of a small These changes result in luminal narrowing or
pulmonary artery (arrow). Panel D (hematoxylin and eosin) complete obliteration of small vessels. Plexiform
shows a plexiform lesion with an occlusive lesion proximally lesions, which may arise from anastomoses in-
(arrowhead) and excessive, poorly formed capillaries and large
volving bronchial arteries or vasa vasorum pene-
clusters of endothelial-like cells distally (arrows). Panel E (hema-
toxylin and eosin) shows a characteristic lesion of PVOD, with trating the wall structure of pulmonary vessels,28
intimal fibrosis (arrow) and marked obliteration of the vessel lu- are common features of pulmonary arterial
men. The diagram in Panel F (adapted from Southgate et al.21) hypertension (Fig. 2). Events leading to severe
shows signal transduction by transforming growth factor β remodeling have not been clearly identified,
(TGF-β) superfamily members and related genes and proteins
although endothelial dysfunction induced by
implicated in heritable PAH, including membrane-bound re-
ceptors such as bone morphogenetic protein receptor type II shear stress, hypoxia, autoimmune phenomena,
(BMPR2); activin A receptor type II–like 1 (ACVRL1), also known viral infections, drugs and toxins, or genetic al-
as activin receptor–like kinase 1 (ALK1); endoglin (ENG); caveo- terations may initiate the process of excess vaso-
lin-1 (CAV1); and transcription factors SMAD4 and SMAD8 constriction, inflammation, and uncontrolled
(SMAD4 and SMAD9). Bone morphogenetic protein (BMP)
cellular growth.27 The findings of highly orga-
signaling is mediated, in a cell specific manner, by the binding of
ligands BMP9 and BMP10 to a heterometric complex of BMPR-II, nized lymphoid follicles juxtaposed against pul-
a cognate type 1 receptor (ACVRL1 or ALK1), and endoglin, an monary arterial hypertension lesions, infiltra-
auxiliary receptor thought to sequester ligand, inhibiting recep- tion of T and B lymphocytes,29 and circulating
tor binding and thus modulating the effects of BMPs. Activation inflammatory markers correlating with disease
of the receptor complex results in phosphorylation of the recep-
severity,30 combined with the fact that pulmo-
tor and SMAD8, which then associates with SMAD4 before
translocation to the nucleus, regulating genes that contain nary arterial hypertension often complicates auto-
BMP-responsive elements (BREs). KCNK3, the gene encoding immune or inflammatory diseases, have all lent
potassium-channel subfamily K, member 3 (K2P3.1), contributes substantial credence to a role of inflammation
to pulmonary vascular tone and is a subunit of the ATP-sensitive in the pathogenesis of pulmonary arterial hyper-
potassium (K ATP) channel (ABCC8). SOX17 and TBX4 (transcrip-
tension.23,31
tion factors important for organogenesis) also participate in
BMP signaling. The probable cation-transporting ATPase 13A3
(ATP13A3) is not directly related to BMP signaling. However, R igh t V en t r icl e
loss-of-function mutations in ATP13A3 disrupt polyamine homeo-
stasis and lead to endothelial dysfunction, thus contributing to Right ventricular function is the major determi-
PAH pathogenesis. Aquaporin 1 (AQP1), also not a member of
nant of clinical outcomes and survival among
the TGF-β superfamily, mediates the migration and proliferation
of pulmonary-artery smooth-muscle cells by interacting with patients with pulmonary hypertension.32 In re-
β-catenin to activate cell growth–related target genes. Biallelic sponse to an increase in pulmonary vascular
mutations in GCN2 (general control nonderepressible 2), also resistance by a factor of 5 to 10, the right ven-
known as EIF2AK4 (eukaryotic translation initiation factor 2 alpha tricle undergoes hypertrophy, chamber dilata-
kinase 4), which encodes an important kinase in the integrated
tion, fat deposition, fibrosis, and metabolic shifts
stress response, are associated with the pathogenesis of PVOD
and PCH. as pulmonary hypertension progresses.32
Right ventricular remodeling may be adap-

n engl j med 385;25  nejm.org  December 16, 2021 2365


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

tive, with concentric hypertrophy, preservation of


transforming growth factor β (TGF-β) super-
myocardial microcirculation, and minimal fibro-
family. This breakthrough, combined with ad-
sis, or it may be maladaptive, with eccentric vances in genetic technology such as whole-
hypertrophy, microvascular rarefaction leadinggenome and whole-exome sequencing, has
to an imbalance between oxygen demand and substantially advanced our understanding of the
supply, and myocardial fibrosis.32 The mecha- role certain genes play in the pathogenesis of
nisms leading to such changes, or to the transi-
pulmonary arterial hypertension.
tion between these two states, remain poorly BMPR2 mutations are identified in approxi-
understood but may involve altered angiogene- mately 80% of patients with familial pulmonary
sis, a shift from glucose oxidation to glycolysis
arterial hypertension, with variable penetrance
and fatty acid oxidation, and altered mitochon-
between male and female carriers, and in up to
drial bioenergetics.33 20% of patients with sporadic disease. Identifi-
Pressure–volume loop technology with a high-
cation of mutations in ACVRL1 (encoding activin
fidelity conductance catheter, the standard for
A receptor type II–like 1 [also known as activin
assessing right ventricular intrinsic myocardial
receptor–like kinase 1]) and ENG (encoding en-
function and right ventricular–pulmonary vascu-
doglin)44,45 in families with hereditary hemor-
lar coupling, is invasive and requires special ex-
rhagic telangiectasia, a syndrome occasionally
pertise.32 Surrogate noninvasive techniques (echo-
complicated by pulmonary arterial hypertension,
cardiography or cardiac magnetic resonance followed rapidly. Both ACVRL1 and endoglin
imaging [MRI])34-36 remain to be validated participate in BMPR-II signaling through dimer-
against this standard, although they do predict
ization (Fig. 2).
outcomes.34,37 The mechanisms of right ventricu- Further analysis of large cohorts of patients
lar dysfunction, lack of current right ventricle–
with pulmonary arterial hypertension has identi-
targeted therapeutics, and remaining gaps in fied additional mutations21 in genes coding for
progress have recently been emphasized.32,38 the transcription factors SMAD1, SMAD4, and
Better right ventricular ejection fractions in
SMAD9,46 part of the complex BMPR-II down-
women than in men free of cardiovascular dis- stream signaling, and other genes in families
ease39 have been attributed to sex hormone dif-
that are negative for BMPR2 mutations,47 includ-
ferences40 and sex-related responses to certain
ing the gene encoding caveolin-1 (CAV1)48 (which
drugs (e.g., phosphodiesterase inhibitors and serves to colocalize BMP receptors) and the gene
endothelin receptor antagonists).41 However, fur-
encoding potassium-channel subfamily K, mem-
ther research is warranted. ber 3 (KCNK3),49 which is implicated in mem-
In vitro study of cardiomyocytes has provided
brane potential maintenance and pulmonary vas-
great insight into intrinsic myocardial contrac-
cular tone. Mutations in TBX4 (encoding T-box
tility, revealing a hypercontractile phenotype in
transcription factor 4), a gene associated with
patients with idiopathic or congenital heart the small patella syndrome,50 were detected in a
disease–associated pulmonary arterial hyperten-
number of children with intellectual disabilities
sion,42 in sharp contrast to a hypocontractileand dystrophic features, in some of their par-
phenotype in patients with scleroderma-associ-ents, and in an additional small cohort of adults
ated pulmonary arterial hypertension.43 These with pulmonary arterial hypertension.50 BMPR2
findings, correlating with in vivo measurements
mutations predominate in large cohorts (Fig.
of right ventricular contractility, may explain the
S2). Other new mutations involve ATP13A3 (en-
worse clinical outcomes and shorter survival in
coding ATPase 13A3); SOX17, which encodes
the latter group. The molecular underpinnings SRY-box 17 and is a major risk factor for con-
of these phenotypes remain poorly studied. genital heart disease–associated pulmonary arte-
rial hypertension)51; AQP1 (encoding aquaporin 1);
and GDF2 (encoding growth differentiation fac-
Gene t ic Fe at ur e s
tor 2, also known as BMP9).47
A major breakthrough occurred in 2000, when Biallelic mutations in EIF2AK4, which encodes
two independent groups8,9 described heterozy- the eukaryotic translation initiation factor 2 alpha
gous mutations in BMPR2, a member of the kinase 4, have been reported in heritable pulmo-

2366 n engl j med 385;25  nejm.org  December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

nary capillary hemangiomatosis52 and pulmo- tially delayed diagnosis in many cases. The pres-
nary veno-occlusive disease53 and in up to 25% ence of an underlying disease such as HIV infec-
of sporadic cases of these diseases. More re- tion or liver or connective-tissue disease or a
cently, germline mutations of TET2, encoding history of exposure to drugs or toxins should
ten-eleven translocation (tet) methylcytosine di- heighten suspicion concerning pulmonary arte-
oxygenase 2, a key enzyme in DNA demethyl- rial hypertension (Fig. 1). A major diagnostic
ation, were reported in a large cohort of patients challenge is to rule out other forms of pulmo-
with pulmonary arterial hypertension.54 nary hypertension for which management should
The role of altered BMPR-II signaling in the focus principally on the underlying disease, so
pathogenesis of pulmonary arterial hypertension risk factors for or symptoms of left-sided heart
cannot be overestimated. Most discovered muta- disease or chronic lung disease are important to
tions involve BMPR2 or genes encoding proteins consider.
that form complexes or interact with BMP or Physical findings that are suggestive of pul-
BMPR-II signaling (Fig. 2). BMPR-II functional monary hypertension include an increased sec-
loss leads to endothelial dysfunction and the ond pulmonic sound, a murmur of tricuspid re-
altered balance between proliferation and apo­ gurgitation, and evidence of right ventricular
ptosis that is characteristic of pulmonary arterial fluid overload (e.g., increased jugular venous
hypertension, which explains the growing inter- pressure and pedal edema). Other findings might
est in therapy aimed at increasing BMPR-II ex- suggest an underlying cause of pulmonary hy-
pression55 or ligand levels, as attempted in pre- pertension, including sequelae of chronic liver or
clinical models of BMP9 administration.56 rheumatologic disorders.
Clinicians have an ethical obligation to inform
patients and their families about any existing Diagnostic Testing
genetic condition, particularly in the case of idio- Transthoracic echocardiography (TTE), the sin-
pathic or heritable pulmonary arterial hyperten- gle most important screening test (Fig. 3), pro-
sion, pulmonary veno-occlusive disease, or pulmo- vides a set of measures for gauging the preva-
nary capillary hemangiomatosis, and congenital lence, cause, and severity of the disease. These
heart disease–associated pulmonary arterial hy- measures include dilatation of right-sided cham-
pertension. The implications for family members bers; presence and severity of tricuspid regurgi-
and their offspring who may be mutation carri- tation, which allows for an estimation of right
ers should be considered, along with screening, ventricular systolic pressure; the presence of
genetic and psychological counseling by a mul- pericardial effusion; and abnormal septal devia-
tidisciplinary team of experts, and patient edu- tion due to right ventricular volume and pressure
cation.57 Several affordable technologies and overload. TTE can also identify left ventricular
platforms can be used to probe multiple genes systolic or diastolic dysfunction and valvular ab-
simultaneously. normalities, findings that shift the focus toward
Genetic testing, which promises to further group 2 pulmonary hypertension.
our understanding of the disease and improve In addition to a complete blood count and
therapy through specific targeting, is a task of metabolic panel, measurement of antinuclear
several national and international collaborative antibody titers and HIV serologic testing may
studies58 and the National Institutes of Health– help uncover a specific underlying disorder such
sponsored PVDOMICS (Pulmonary Vascular Dis- as connective-tissue disease or HIV disease, re-
ease Phenomics) initiative.59 spectively. The serum N-terminal natriuretic
peptide level, measured as a nonspecific cardiac
biomarker, can be incorporated into risk strati-
Di agnosis
fication (see the discussion below), since the
History and Physical Examination value tracks with the severity of pulmonary arte-
Symptoms of pulmonary arterial hypertension rial hypertension and can be used to predict
are nonspecific (exertional dyspnea, fatigue, chest survival.61
pain, and fluid retention, as well as syncope in A chest radiograph may suggest cardiac en-
advanced cases), which accounts for a substan- largement and dilated pulmonary arteries, as

n engl j med 385;25  nejm.org  December 16, 2021 2367


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Figure 3. Diagnostic Algorithm for Suspected PH.


Clinical suspicion of PH based on symptoms When PH is suspected on the basis of symptoms and
and clinical findings
signs, transthoracic echocardiography is the single
most important screening test ordered by the general
practitioner. Additional routine blood and imaging tests
Transthoracic echocardiogram are integrated in the clinical assessment to determine
the best approach in diagnosing PH and possible sub-
groups or associated conditions (group 1) or other dis-
eases (e.g., groups 2 through 5). Patients suspected of
Integration of clinical findings and tests to best having group 1 (PAH), group 4 (chronic thromboem-
assign a patient to a PH group
bolic PH), or other forms of severe PH with right ven-
tricular (RV) dysfunction (on echocardiography) are
typically referred to specialized PH centers, where ad-
ditional tests, imaging, and right heart catheterization
Initial routine tests: Initial imaging: are performed to further define the nature and sever­
Blood: CBC, basic metabolic panel, Chest radiography ity of PH. A polysomnogram helps rule out associated
uric acid, TSH, BNP or High-resolution CT of the chest
NT-proBNP, ANA, LFTs sleep disorders, which may contribute to or worsen PH.
Electrocardiogram A vasodilator challenge with inhaled nitric oxide (NO),
Full pulmonary-function tests or alternatively, an inhaled prostacyclin (PGI2) analogue
or intravenous adenosine or PGI2 analogue, is usually
reserved for patients with idiopathic PAH, heritable
PAH, or PAH associated with drugs or toxins.60 Provo-
Referral to PH expert center cation tests with fluid challenge or exercise are used
(particularly for clinical suspicion of group 1 to unmask left-sided heart disease or exercise-induced
or 4 or severe PH with RV dysfunction PH, respectively. Cardiopulmonary exercise testing
for all PH groups)
(CPET) can help identify cardiac versus pulmonary
causes of dyspnea and obtain measures (e.g., the max-
imum oxygen consumption or peak) that can be incor-
Additional tests and imaging: porated into the European Society of Cardiology–Euro-
ANA, hepatitis, and HIV serologic tests
V/Q scan
pean Respiratory Society (ESC-ERS) risk stratification
Six-minute walk test (see Fig. S4 in the Supplementary Appendix for details
CPET of the ESC-ERS risk score and the REVEAL [Registry to
Polysomnogram (if indicated) Evaluate Early and Long-Term PAH Disease Manage-
Chest CTA or pulmonary angiography
(thromboembolic disease suspected)
ment] score). The algorithm is adapted from Galiè et al.60
CMR ANA denotes antinuclear antibody, BNP B-type natriuret-
ic peptide, CBC complete blood count, CMR cardiac
magnetic resonance, LFTs liver-function tests, NT-
Right heart catheterization:
proBNP N-terminal proBNP, and TSH thyrotropin.
Vasodilator challenge (e.g., inhaled NO or PGI2,
or intravenous adenosine or PGI2)
Provocation tests tion–perfusion scan, may reveal signs of chronic
Fluid challenge
Exercise (e.g., ergometer while supine) thromboembolic disease such as filling defects
or wedge-shaped or irregular linear opacities
from previous thrombi. It also helps to deter-
Risk stratification and treatment algorithm mine the surgical accessibility of the lesions and
can rule out other diagnoses (e.g., pulmonary-
artery stenosis or tumor and fibrosing medias-
tinitis).
well as parenchymal lung or chest-wall abnor- Pulmonary-function tests may suggest ob-
malities. Computed tomography (CT) of the structive or restrictive lung disease. The single-
chest is routinely performed to rule out paren- breath diffusing capacity of the lungs for carbon
chymal disease. A ventilation–perfusion scan monoxide, which is typically decreased in pul-
remains essential in the clinical algorithm, since monary arterial hypertension, can be incorpo-
normal perfusion makes chronic thromboem- rated, along with other clinical findings and
bolic pulmonary hypertension (CTEPH) an un- TTE findings (right atrial enlargement and tri-
likely diagnosis. CT angiography of the chest, cuspid regurgitation velocity), in an evidence-
although considered less sensitive than a ventila- based algorithm to detect pulmonary arterial

2368 n engl j med 385;25  nejm.org  December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

hypertension in asymptomatic patients with the ESC-ERS “risk table” and other risk scores (e.g.,
scleroderma spectrum of diseases.62 An electro- the REVEAL [Registry to Evaluate Early and
cardiogram is important to look for evidence of Long-Term PAH Disease Management] risk score
atrial or ventricular hypertrophy, signs of ische­ calculator66) (Fig. S4) have been successfully
mic heart disease, or dysrhythmias. used to assess survival in retrospective analyses
Cardiac MRI (Fig. S3) is the standard for right of data from pulmonary arterial hypertension
ventricular assessment because it provides accu- registries67-69 and a post hoc analysis of data
rate measurements of the cardiac chamber anat- from a large prospective clinical trial.70 The pre-
omy and volume, mass, function, and flow, as dictive ability of stratification methods is im-
well as myocardial perfusion.38 Not widely avail- proved by machine-learning algorithms, which
able, it is increasingly used in centers with ex- reveal a dynamic interdependent influence of
pertise in diagnosing and managing pulmonary multiple risk factors, thus avoiding the assump-
hypertension. Other advanced imaging tech- tion that limited clinical measures have indepen-
niques that remain in the realm of research in- dent relationships to a specific outcome.71
clude three-dimensional echocardiography, four-
dimensional flow MRI, and positron-emission Ther a py
tomography, which can provide unique insights
into right ventricular metabolic activity.63 Basic supportive measures, a constant compo-
nent of treatment long before the availability of
Hemodynamics targeted therapy, include diuretics to achieve
Right heart catheterization is required for the euvolemia and supplemental oxygen when need-
diagnosis of pulmonary arterial hypertension in ed at rest, during sleep, or with exercise, to main-
order to directly assess pulmonary hemodynam- tain adequate hemoglobin oxygen saturation
ics and cardiac output and to calculate pulmo- (Fig. 4). Sleep-disordered breathing, which can
nary vascular resistance. This is a necessary step complicate any form of cardiopulmonary disor-
in the diagnostic algorithm before treatment der, is prevalent among patients with precapil-
(Fig. 3). It is essential for confirmation of the lary pulmonary hypertension72 and should be
presence and type of pulmonary hypertension diagnosed and treated when appropriate. Anti-
(precapillary, postcapillary, or combined) and coagulant therapy, once recommended on the
provides essential measures for risk stratification. basis of retrospective analyses showing a sur-
A structured clinical assessment (Fig. 3) helps vival benefit,73,74 is now recommended only for
assign a patient to a specific pulmonary hyper- idiopathic pulmonary arterial hypertension (not
tension group (Fig. 1), which is essential in de- for other forms of pulmonary arterial hyperten-
termining the appropriate therapy, although it is sion, according to data from a European regis-
increasingly clear that any given patient may try),75 on a case-by-case basis and risk–benefit
belong in more than one group. analysis, and for group 4 pulmonary hyperten-
sion (CTEPH), in which increased clotting is a
primary issue. A cardiopulmonary exercise pro-
R isk S t r at ific at ion
gram is advised on the basis of a meta-analysis
The importance of risk assessment was recog- of controlled trials,76 as tolerated by the patient
nized early in the study of idiopathic pulmonary (Fig. 4). Immunizations should be kept up to date.
arterial hypertension, at which time the focus Although four decades separated the initial
was essentially on baseline hemodynamics.64 The clinical description of pulmonary hypertension
2015 European Society of Cardiology–European and approval of the first effective therapy for
Respiratory Society (ESC-ERS) guidelines for pulmonary arterial hypertension, based on a ran-
pulmonary hypertension65 highlighted as an in- domized, non-placebo-controlled trial of prosta-
creasing priority the stratification of patients, at cyclin in patients with “primary pulmonary hy-
baseline and follow-up, into low-, intermediate-, pertension,”77 the past 20 years have witnessed a
and high-risk groups on the basis of a combina- series of clinical trials targeting essentially three
tion of clinical, functional, and hemodynamic signaling pathways identified in pulmonary ar-
measures, as a tool for choosing therapy. The terial hypertension78 (Fig. 5). These trials have

n engl j med 385;25  nejm.org  December 16, 2021 2369


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Confirmed PAH

General supportive measures No acute vasodilator response Positive acute vasodilator response
Diuresis to achieve euvolemia before (>90% of patients) (<10% of patients)
therapy and after initiation of targeted Drop in mPAP >10 mm Hg to absolute
therapy value ≤40 mm Hg
Supplemental O2 to treat hypoxemia No change or increase in CO
at rest, during sleep, or with exercise
Anticoagulation for idiopathic PAH
or as indicated
Diet and exercise
High-dose calcium-channel blocker
Contraceptive measures for women
of childbearing age Risk assessment Ensure adequate response (FC I or II
Referral for counseling and genetic ESC-ERS risk stratification and improved hemodynamics)
testing as appropriate REVEAL score Add PAH-targeted therapy for lack of
Vaccinations as appropriate adequate response or loss of acute
vasodilator response on repeat
testing at 1 yr

Low or intermediate risk High risk

Oral monotherapy if response Oral dual therapy, either Combination therapy including Evaluation for lung
is adequate or if dual therapy up-front or sequential parenteral PGI2 agent transplantation
is poorly tolerated (for patients with incomplete
response to monotherapy)

Structured follow-up at 3–6 mo Triple therapy including


parenteral PGI2 agent;
Physical examination
escalation within drug
Transthoracic echocardiogram
class if appropriate
Six-minute walk test/Borg dyspnea score
NT-proBNP
Risk stratification
Quality-of-life assessment
Repeat right heart catheterization

Intermediate or
Low risk
high risk

Figure 4. Treatment Algorithm for Confirmed PAH.


The algorithm is adapted from Galiè et al.60 See Fig. S4 for details of the ESC-ERS risk score and the REVEAL score. The Borg dyspnea
scale is used to assess dyspnea as reported by a patient during the 6-minute walk test; the scale ranges from 0 to 10, with higher scores
indicating more severe dyspnea. Patients in whom triple therapy is recommended are typically evaluated for lung transplantation while a
structured follow-up is being performed. FC denotes functional class.

established current targeted therapy for pulmo- ception of inhaled treprostinil, which is now
nary arterial hypertension (group 1) and CTEPH approved by the Food and Drug Administration
(group 4). The findings do not apply to other (FDA) for pulmonary hypertension associated
groups in the WSPH classification, with the ex- with interstitial lung disease (group 3) on the

2370 n engl j med 385;25  nejm.org  December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

Endothelin-1 Pathway Prostacyclin (PGI2) Pathway Nitric Oxide Pathway

Endothelin 1 Dysfunctional endothelium in PAH Nitric oxide

BLOOD-VESSEL
L-citrulline
LUMEN PGI2
ENDOTHELIAL
CELL eNOS
Proendothelin Arachadonic L-arginine
acid Nitric oxide
Cox 1/2

Endothelin 1 Epoprostenol PGI2


Endothelin Treprostinil Riociguat
receptorA Iloprost Nitric oxide
Endothelin
receptorB IP receptor
+ Selexipag +

+
Gs Sildenafil
sGC PDE5 – Tadalafil
Adenylate
Vasoconstriction Vasodilatation cyclase GTP cGMP GMP
Proliferation Proliferation
ATP cAMP
SMOOTH-MUSCLE CELLS

– – Vasodilatation
Bosentan Vasodilatation Proliferation
Macitentan Proliferation
Ambrisentan

Figure 5. Three Classic Pathways of Targeted Therapy for PAH.


Current targeted therapy is aimed at correcting endothelial dysfunction27 by inhibiting the endothelin pathway and enhancing the prosta-
cyclin (PGI2) and NO pathways. Endothelin 1 (ET1), which is increased in PAH, can bind to either the endothelin A (ETA) receptor, caus-
ing vasoconstriction (of smooth-muscle cells) and cell proliferation, or the endothelin B (ETB) receptor, causing vasodilatation and anti-
proliferation. Thus, there are dual ETA –ETB receptor antagonists (e.g., bosentan and macitentan) or selective ETA receptor antagonists
(e.g., ambrisentan), which leave the ETB receptor functional. The expression and function of the PGI2 and NO pathways are decreased
in PAH, resulting, respectively, in diminished cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP),
which are second messengers responsible for vasodilatation and antiproliferation. Agents that increase cAMP include PGI2 analogues
given intravenously (e.g., epoprostenol and treprostinil), subcutaneously (e.g., treprostinil), by inhalation (e.g., iloprost and treprostinil),
orally (treprostinil), or with the use of oral PGI2 receptor (IP) agonists (e.g., selexipag). Increased cGMP release can be achieved with in-
haled NO (used essentially in the cardiac catheterization laboratory or intensive care unit), which stimulates soluble guanylate cyclase
(sGC), or by inhibiting phosphodiesterase type 5 (PDE5, which degrades cGMP into GMP) with the use of oral PDE5 inhibitors (silde-
nafil or tadalafil). Direct sGC stimulators (e.g., oral riociguat) can increase the release of cGMP independently of NO release. These
drugs have been approved by the Food and Drug Administration for patients with PAH who have an mPAP of 25 mm Hg or higher. Al-
though they usually are associated with acceptable adverse-event profiles, these drugs have common side effects that are due essential-
ly to their vasodilatory effects, including headache and lightheadedness (particularly at the initiation of treatment), flushing and upper
respiratory congestion, systemic hypotension (more frequent with systemically administered drugs but also common with certain drugs
that need slower dose escalation, such as riociguat and selexipag), gastrointestinal symptoms (e.g., bloating, nausea or vomiting, and
diarrhea), and rash (PGI2 analogues). Fluid retention (e.g., pedal edema) is a common problem with the initiation of therapy and requires
adjustment of diuretic doses. Additional specific side effects include cough (e.g., with inhaled treprostinil), elevation of aminotrans-
ferase levels (with bosentan), anemia (with macitentan), visual changes (with PDE5 inhibitors but very rare), skin irritation or cellulitis
(with subcutaneous treprostinil), and thrombocytopenia or bone marrow suppression (with systemically administered PGI2 analogues).
AA denotes arachidonic acid, AC adenylate cyclase, Cox 1/2 cyclooxygenase 1/2, eNOS endothelial isoform of nitric oxide synthase, GS
G-protein–coupled receptor, and GTP guanosine triphosphate.

basis of a recent randomized phase 3 clinical therapy with pulmonary endarterectomy should
trial.79 An assessment for CTEPH is best per- be considered first. Medical therapy, pulmonary
formed at specialized centers, where definitive balloon angioplasty, or both are considered for

n engl j med 385;25 nejm.org December 16, 2021 2371


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

patients with inoperable disease or residual pul- therapy on survival in most of the more recent
monary hypertension after endarterectomy. large RCTs,81,82,84 a large meta-analysis of early
There have been important changes in the RCTs evaluating therapy for pulmonary arterial
design of randomized, controlled trials (RCTs) hypertension, averaging 12 to 16 weeks in dura-
over approximately the past decade. The 6-min- tion, showed a significant reduction in mortality
ute walk distance was the primary end point in with therapy as compared with placebo,85 which
most early RCTs, typically with a 12-week study is consistent with data obtained from large reg-
period. However, in response to various calls for istries.86,87
establishing more relevant end points,80 RCTs Treatment algorithms designed to guide ther-
shifted to composite end points, including a apy, with classes of recommendations and the
combination of hospital admission, worsening level of evidence for the various therapies ap-
of pulmonary arterial hypertension, mortality, proved for pulmonary arterial hypertension, are
and escalation of therapy. Another important available in comprehensive guidelines.65 Patients
change in RCT design was an evaluation of the who have pulmonary vasoreactivity (typically to
new therapy added to background treatment inhaled nitric oxide during initial right heart
for pulmonary arterial hypertension or up-front catheterization), based on strict criteria (a re-
combined therapy rather than monotherapy. duction in mean pulmonary arterial pressure of
These changes required the enrollment of many ≥10 mm Hg, to an absolute value of ≤40 mm Hg,
more patients and a longer time to reach the accompanied by an increase or no change in
primary outcomes. An example is SERAPHIN cardiac output), can be treated with high-dose
(Study with an Endothelin Receptor Antagonist calcium-channel blockers alone, provided that
in Pulmonary Arterial Hypertension to Improve this therapy results in New York Heart Associa-
Clinical Outcome), which revealed the efficacy of tion (NYHA) functional class I or II with hemo-
macitentan, a dual endothelin receptor antago- dynamic improvement maintained on repeat
nist, in reducing the first occurrence of a com- testing after at least 1 year of therapy1 (achieved
posite primary end point in a study population in less than 10% of patients with idiopathic
of more than 700 patients with symptomatic pulmonary arterial hypertension88). In case of
pulmonary arterial hypertension who were re- clinical deterioration or loss of vasoreactivity,
ceiving placebo or background therapy (inhaled pulmonary arterial hypertension–specific ther-
or oral drugs, excluding other endothelin re- apy should be added according to accepted algo-
ceptor antagonists).81 A subsequent RCT, the rithms.
AMBITION (Ambrisentan and Tadalafil in Pa- Monotherapy can be used for patients with a
tients with Pulmonary Arterial Hypertension) positive response to acute vasoreactivity and
trial, compared up-front combination therapy those with a good historical response (NYHA
with two FDA-approved drugs (ambrisentan and functional class I or II with sustained hemody-
tadalafil) with each drug alone in patients who namic improvement), elderly patients (>75 years
had received no previous treatment for pulmo- old) with important risk factors for left-sided
nary arterial hypertension.82 The risk of the pri- heart disease (e.g., systemic hypertension, coro-
mary end point (the first event of clinical failure nary artery disease, or atrial fibrillation), those
in a time-to-event analysis) was reduced with suspected of having pulmonary veno-occlusive
combination therapy as compared with mono- disease or pulmonary capillary hemangiomato-
therapy with either drug. In both of these large sis, patients with very mild disease (NYHA func-
trials, however, the primary end point was driv- tional class I and pulmonary vascular resistance
en predominantly by decreased hospitalization of 3 to 4 WU, with normal right ventricular
rates (essentially due to worsening pulmonary function on echocardiography), and patients in
arterial hypertension), a clinically relevant out- whom combination therapy is associated with
come, since admission for right ventricular fail- an unacceptable side-effect profile.60 Otherwise,
ure, the main cause of hospitalization among most patients with pulmonary arterial hyperten-
patients with pulmonary arterial hypertension, sion are currently treated with up-front combi-
portends a very poor prognosis.83 nation therapy consisting of two oral agents,
Despite the lack of an effect of combined oral with dose escalation within a drug class when

2372 n engl j med 385;25  nejm.org  December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

appropriate, or sequential combination therapy. pulmonary arterial hypertension. Integrating a


Referral for evaluation for lung transplantation molecular classification into the current classifi-
is recommended when medical therapy fails to cation is now a realistic goal, as indicated by
reduce the risk to a low or intermediate level. proteomics studies91 and genomics studies.59
The role of up-front triple combination therapy In keeping with the notion that dysregulated
in patients with pulmonary arterial hypertension immunity may trigger or contribute to the patho-
remains unclear. genesis of pulmonary hypertension, clinical
Atrial septostomy is considered occasionally trials targeting specific immune pathways have
in patients with end-stage pulmonary arterial recently been launched. Targeting B cells in
hypertension or those awaiting lung transplan- scleroderma-associated pulmonary arterial hyper-
tation. Atrial septostomy has the advantage of tension appeared to benefit a subgroup of pa-
unloading the right atrium and right ventricle tients identified by machine-learning analysis of
and delaying right ventricular failure while im- biomarkers, suggesting a potential role as ad-
proving left ventricular preload and cardiac out- junctive immunotherapy for this disease.92 Simi-
put at a cost of reduced oxygenation from right larly, targeting altered growth factor signaling
to left shunting. continues to generate interest, despite the cau-
tionary tale of the tyrosine kinase inhibitor
imatinib, which showed encouraging results in a
F u t ur e Dir ec t ions
phase 2 trial but serious side effects (i.e., subdu-
Increasingly sophisticated computational power, ral hemorrhage) in a phase 3 trial, preventing
combined with advanced proteomics platforms89 FDA approval of imatinib for the treatment of
or imaging,90 has led to machine-learning tech- pulmonary arterial hypertension.93
niques that can be used to develop promising Finally, a trial of the calcineurin inhibitor
and powerful diagnostic tools for pulmonary FK506,94 which was shown to up-regulate BMPR-
arterial hypertension. The current trend to push II expression,55 is noteworthy, considering the
large-scale investigation into biomarkers and importance of rescuing BMPR-II signaling to
various other “-omics” (proteomics and genom- counterbalance proproliferative and proinflamma-
ics) should facilitate characterization of specific tory TGF-β pathways. Similarly, a recent phase 2
mechanistic pathways (common or distinct clinical trial showed that sotatercept (a first-in-
among pulmonary hypertension groups) in a class fusion protein designed to bind the TGF-β
totally agnostic fashion; the trend should also ligand activin) reduced pulmonary vascular re-
lead to precision medicine that accounts for ge- sistance and serum N-terminal pro–B-type natri-
netic, environmental, and lifestyle factors, a pro- uretic peptide levels and improved functional
cess similar to the one that led to current cancer capacity in patients with pulmonary arterial hy-
therapy. However, to be successful, this will re- pertension who were receiving background ther-
quire strong collaborative efforts among centers, apy.95 This very promising new treatment is now
at the national and international levels, to create being tested in phase 3 clinical trials.
large registries (for clinical and imaging pheno- Disclosure forms provided by the author are available with the
typing) and biobanks for tissue, biomarkers, full text of this article at NEJM.org.
I thank Carol Farver, University of Michigan, and Jody Hooper,
genetics, and proteomics. This is particularly Johns Hopkins University, for providing the histopathological
important in the case of a rare syndrome such as images.

References
1. Simonneau G, Montani D, Celermajer 4. Richards DW. The contributions of and the pulmonary circulation. Thorax
DS, et al. Haemodynamic definitions and right heart catheterization to physiology 1971;​26:​262-70.
updated clinical classification of pulmo- and medicine, with some observations 7. Hatano S, Strasser T, eds. Primary pul-
nary hypertension. Eur Respir J 2019;​53:​ on the physiopathology of pulmonary monary hypertension:​report on a WHO
1801913. heart disease. Am Heart J 1957;​54:​161- meeting, Geneva, 15–17 October, 1973.
2. von Romberg E. Über sklerose der 71. Geneva:​World Health Organization, 1975.
lungenarterie. Dtsch Arch Klin Med 1891;​ 5. Dresdale DT, Schultz M, Michtom RJ. 8. Lane KB, Machado RD, Pauciulo MW,
48:​197-206. Primary pulmonary hypertension. I. Clin- et al. Heterozygous germline mutations
3. Forssmann W. Die sondierung des ical and hemodynamic study. Am J Med in BMPR2, encoding a TGF-beta receptor,
rechten herzens. Klin Wochenschr 1929;​ 1951;​11:​686-705. cause familial primary pulmonary hyper-
8:​2085-7. 6. Kay JM, Smith P, Heath D. Aminorex tension. Nat Genet 2000;​26:​81-4.

n engl j med 385;25  nejm.org  December 16, 2021 2373


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

9. Deng Z, Morse JH, Slager SL, et al. Fa- 23. Price LC, Wort SJ, Perros F, et al. In- 37. Nie L, Li J, Zhang S, et al. Correlation
milial primary pulmonary hypertension flammation in pulmonary arterial hyper- between right ventricular-pulmonary ar-
(gene PPH1) is caused by mutations in the tension. Chest 2012;​141:​210-21. tery coupling and the prognosis of pa-
bone morphogenetic protein receptor-II 24. Launay D, Sobanski V, Hachulla E, tients with pulmonary arterial hyperten-
gene. Am J Hum Genet 2000;​67:​737-44. Humbert M. Pulmonary hypertension in sion. Medicine (Baltimore) 2019;​98(40):​
10. Humbert M, Sitbon O, Chaouat A, et al. systemic sclerosis: different phenotypes. e17369.
Pulmonary arterial hypertension in France: Eur Respir Rev 2017;​26:​170056. 38. Lahm T, Douglas IS, Archer SL, et al.
results from a national registry. Am J 25. Gerges C, Gerges M, Friewald R, et al. Assessment of right ventricular function
Respir Crit Care Med 2006;​173:​1023-30. Microvascular disease in chronic throm- in the research setting: knowledge gaps
11. Hoeper MM, Huscher D, Ghofrani boembolic pulmonary hypertension: hemo- and pathways forward. An official Ameri-
HA, et al. Elderly patients diagnosed with dynamic phenotyping and histomorpho- can Thoracic Society research statement.
idiopathic pulmonary arterial hyperten- metric assessment. Circulation 2020;​141:​ Am J Respir Crit Care Med 2018;​198(4):​
sion: results from the COMPERA registry. 376-86. e15-e43.
Int J Cardiol 2013;​168:​871-80. 26. Fayyaz AU, Edwards WD, Maleszew­ 39. Kawut SM, Lima JAC, Barr RG, et al.
12. Rich S, Haworth SG, Hassoun PM, Ya- ski JJ, et al. Global pulmonary vascular Sex and race differences in right ventric-
coub MH. Pulmonary hypertension: the remodeling in pulmonary hypertension ular structure and function: the multi-
unaddressed global health burden. Lancet associated with heart failure and pre- ethnic study of atherosclerosis-right
Respir Med 2018;​6:​577-9. served or reduced ejection fraction. Circu- ventricle study. Circulation 2011;​123:​2542-
13. Wu W, He J, Shao X. Incidence and lation 2018;​137:​1796-810. 51.
mortality trend of congenital heart dis- 27. Budhiraja R, Tuder RM, Hassoun PM. 40. Ventetuolo CE, Ouyang P, Bluemke
ease at the global, regional, and national Endothelial dysfunction in pulmonary DA, et al. Sex hormones are associated
level, 1990–2017. Medicine (Baltimore) hypertension. Circulation 2004;​109:​159-65. with right ventricular structure and func-
2020;​99(23):​e20593. 28. Galambos C, Sims-Lucas S, Abman tion: the MESA-right ventricle study. Am J
14. Hoeper MM, Humbert M, Souza R, SH, Cool CD. Intrapulmonary broncho- Respir Crit Care Med 2011;​183:​659-67.
et al. A global view of pulmonary hyper- pulmonary anastomoses and plexiform 41. Jacobs W, van de Veerdonk MC, Trip P,
tension. Lancet Respir Med 2016;​4:​306-22. lesions in idiopathic pulmonary arterial et al. The right ventricle explains sex dif-
15. Gorter TM, Hoendermis ES, van Veld- hypertension. Am J Respir Crit Care Med ferences in survival in idiopathic pulmo-
huisen DJ, et al. Right ventricular dys- 2016;​193:​574-6. nary arterial hypertension. Chest 2014;​
function in heart failure with preserved 29. Perros F, Dorfmüller P, Montani D, 145:​1230-6.
ejection fraction: a systematic review and et al. Pulmonary lymphoid neogenesis in 42. Rain S, da Silva Goncalves Bos D,
meta-analysis. Eur J Heart Fail 2016;​18:​ idiopathic pulmonary arterial hyperten- Handoko ML, et al. Protein changes con-
1472-87. sion. Am J Respir Crit Care Med 2012;​185:​ tributing to right ventricular cardiomyo-
16. Miller WL, Grill DE, Borlaug BA. 311-21. cyte diastolic dysfunction in pulmonary
Clinical features, hemodynamics, and 30. Soon E, Holmes AM, Treacy CM, et al. arterial hypertension. J Am Heart Assoc
outcomes of pulmonary hypertension due Elevated levels of inflammatory cytokines 2014;​3(3):​e000716.
to chronic heart failure with reduced ejec- predict survival in idiopathic and familial 43. Hsu S, Kokkonen-Simon KM, Kirk JA,
tion fraction: pulmonary hypertension pulmonary arterial hypertension. Circula- et al. Right ventricular myofilament func-
and heart failure. JACC Heart Fail 2013;​1:​ tion 2010;​122:​920-7. tional differences in humans with system-
290-9. 31. Hassoun PM, Mouthon L, Barberà JA, ic sclerosis-associated versus idiopathic
17. Moore LG, Niermeyer S, Zamudio S. et al. Inflammation, growth factors, and pulmonary arterial hypertension. Circula-
Human adaptation to high altitude: re- pulmonary vascular remodeling. J Am tion 2018;​137:​2360-70.
gional and life-cycle perspectives. Am J Coll Cardiol 2009;​54:​Suppl 1:​S10-S19. 44. Trembath RC, Thomson JR, Machado
Phys Anthropol 1998;​107:​Suppl 27:​25-64. 32. Vonk Noordegraaf A, Chin KM, Had- RD, et al. Clinical and molecular genetic
18. Deol AK, Fleming FM, Calvo-Urbano dad F, et al. Pathophysiology of the right features of pulmonary hypertension in
B, et al. Schistosomiasis — assessing ventricle and of the pulmonary circula- patients with hereditary hemorrhagic tel-
progress toward the 2020 and 2025 global tion in pulmonary hypertension: an up- angiectasia. N Engl J Med 2001;​345:​325-
goals. N Engl J Med 2019;​381:​2519-28. date. Eur Respir J 2019;​53:​1801900. 34.
19. GBD 2017 HIV collaborators. Global, 33. Ryan JJ, Archer SL. The right ventricle 45. Harrison RE, Berger R, Haworth SG,
regional, and national incidence, preva- in pulmonary arterial hypertension: dis- et al. Transforming growth factor-beta
lence, and mortality of HIV, 1980–2017, orders of metabolism, angiogenesis and receptor mutations and pulmonary arte-
and forecasts to 2030, for 195 countries adrenergic signaling in right ventricular rial hypertension in childhood. Circula-
and territories: a systematic analysis for failure. Circ Res 2014;​115:​176-88. tion 2005;​111:​435-41.
the Global Burden of Diseases, Injuries, 34. Vanderpool RR, Pinsky MR, Naeije R, 46. Nasim MT, Ogo T, Ahmed M, et al.
and Risk Factors Study 2017. Lancet HIV et al. RV-pulmonary arterial coupling pre- Molecular genetic characterization of
2019;​6(12):​e831-e859. dicts outcome in patients referred for pul- SMAD signaling molecules in pulmonary
20. Sikirica M, Iorga SR, Bancroft T, Pot- monary hypertension. Heart 2015;​101:​37- arterial hypertension. Hum Mutat 2011;​
ash J. The economic burden of pulmonary 43. 32:​1385-9.
arterial hypertension (PAH) in the US on 35. Trip P, Kind T, van de Veerdonk MC, 47. Gräf S, Haimel M, Bleda M, et al.
payers and patients. BMC Health Serv Res et al. Accurate assessment of load-inde- Identification of rare sequence variation
2014;​14:​676. pendent right ventricular systolic func- underlying heritable pulmonary arterial
21. Southgate L, Machado RD, Gräf S, tion in patients with pulmonary hyper- hypertension. Nat Commun 2018;​9:​1416.
Morrell NW. Molecular genetic frame- tension. J Heart Lung Transplant 2013;​32:​ 48. Austin ED, Ma L, LeDuc C, et al.
work underlying pulmonary arterial hy- 50-5. Whole exome sequencing to identify a
pertension. Nat Rev Cardiol 2020;​17:​85- 36. Sanz J, Kariisa M, Dellegrottaglie S, novel gene (caveolin-1) associated with
95. et al. Evaluation of pulmonary artery stiff- human pulmonary arterial hypertension.
22. Wagenvoort CA. The pathology of pri- ness in pulmonary hypertension with car- Circ Cardiovasc Genet 2012;​5:​336-43.
mary pulmonary hypertension. J Pathol diac magnetic resonance. JACC Cardio- 49. Ma L, Roman-Campos D, Austin ED,
1970;​101:​Pi. vasc Imaging 2009;​2:​286-95. et al. A novel channelopathy in pulmo-

2374 n engl j med 385;25  nejm.org  December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

nary arterial hypertension. N Engl J Med in pulmonary hypertension: emerging try of Newly Initiated Therapies for Pul-
2013;​369:​351-61. techniques and applications. Int J Cardio- monary Hypertension (COMPERA). Circu-
50. Kerstjens-Frederikse WS, Bongers vasc Imaging 2019;​35:​1407-20. lation 2014;​129:​57-65.
EMHF, Roofthooft MTR, et al. TBX4 muta- 64. D’Alonzo GE, Barst RJ, Ayres SM, et al. 76. Buys R, Avila A, Cornelissen VA. Exer-
tions (small patella syndrome) are associ- Survival in patients with primary pulmo- cise training improves physical fitness in
ated with childhood-onset pulmonary ar- nary hypertension. Results from a national patients with pulmonary arterial hyper-
terial hypertension. J Med Genet 2013;​50:​ prospective registry. Ann Intern Med 1991;​ tension: a systematic review and meta-
500-6. 115:​343-9. analysis of controlled trials. BMC Pulm
51. Zhu N, Welch CL, Wang J, et al. Rare 65. Galiè N, Humbert M, Vachiery JL, et al. Med 2015;​15:​40.
variants in SOX17 are associated with pul- 2015 ESC/ERS guidelines for the diagno- 77. Barst RJ, Rubin LJ, Long WA, et al. A
monary arterial hypertension with con- sis and treatment of pulmonary hyper- comparison of continuous intravenous
genital heart disease. Genome Med 2018;​ tension: the Joint Task Force for the di- epoprostenol (prostacyclin) with conven-
10:​56. agnosis and treatment of pulmonary tional therapy for primary pulmonary
52. Best DH, Sumner KL, Austin ED, et al. hypertension of the European Society of hypertension. N Engl J Med 1996;​334:​296-
EIF2AK4 mutations in pulmonary capil- Cardiology (ESC) and the European Re- 301.
lary hemangiomatosis. Chest 2014;​ 145:​ spiratory Society (ERS). Eur Respir J 2015;​ 78. Humbert M, Lau EM, Montani D, Jaïs
231-6. 46:​903-75. X, Sitbon O, Simonneau G. Advances in
53. Eyries M, Montani D, Girerd B, et al. 66. Benza RL, Farber HW, Frost A, et al. therapeutic interventions for patients with
EIF2AK4 mutations cause pulmonary REVEAL risk scores applied to riociguat- pulmonary arterial hypertension. Circula-
veno-occlusive disease, a recessive form treated patients in PATENT-2: impact of tion 2014;​130:​2189-208.
of pulmonary hypertension. Nat Genet changes in risk score on survival. J Heart 79. Waxman A, Restrepo-Jaramillo R,
2014;​46:​65-9. Lung Transplant 2018;​37:​513-9. Thenappan T, et al. Inhaled treprostinil in
54. Potus F, Pauciulo MW, Cook EK, et al. 67. Hoeper MM, Kramer T, Pan Z, et al. pulmonary hypertension due to intersti-
Novel mutations and decreased expres- Mortality in pulmonary arterial hyperten- tial lung disease. N Engl J Med 2021;​384:​
sion of the epigenetic regulator TET2 in sion: prediction by the 2015 European 325-34.
pulmonary arterial hypertension. Circula- pulmonary hypertension guidelines risk 80. Gaine S, Simonneau G. The need to
tion 2020;​141:​1986-2000. stratification model. Eur Respir J 2017;​50:​ move from 6-minute walk distance to out-
55. Spiekerkoetter E, Tian X, Cai J, et al. 1700740. come trials in pulmonary arterial hyper-
FK506 activates BMPR2, rescues endothe- 68. Kylhammar D, Kjellström B, Hjalmars­ tension. Eur Respir Rev 2013;​22:​487-94.
lial dysfunction, and reverses pulmonary son C, et al. A comprehensive risk stratifi- 81. Pulido T, Adzerikho I, Channick RN,
hypertension. J Clin Invest 2013;​123:​3600- cation at early follow-up determines prog- et al. Macitentan and morbidity and mor-
13. nosis in pulmonary arterial hypertension. tality in pulmonary arterial hypertension.
56. Long L, Ormiston ML, Yang X, et al. Eur Heart J 2018;​39:​4175-81. N Engl J Med 2013;​369:​809-18.
Selective enhancement of endothelial 69. Boucly A, Weatherald J, Savale L, et al. 82. Galiè N, Barberà JA, Frost AE, et al.
BMPR-II with BMP9 reverses pulmonary Risk assessment, prognosis and guideline Initial use of ambrisentan plus tadalafil
arterial hypertension. Nat Med 2015;​21:​ implementation in pulmonary arterial hy- in pulmonary arterial hypertension. N Engl
777-85. pertension. Eur Respir J 2017;​50:​1700889. J Med 2015;​373:​834-44.
57. Morrell NW, Aldred MA, Chung WK, 70. Galiè N, Jansa P, Pulido T, et al. 83. Campo A, Mathai SC, Le Pavec J, et al.
et al. Genetics and genomics of pulmo- SERAPHIN haemodynamic substudy: the Outcomes of hospitalisation for right
nary arterial hypertension. Eur Respir J effect of the dual endothelin receptor heart failure in pulmonary arterial hyper-
2019;​53:​1801899. antagonist macitentan on haemodynamic tension. Eur Respir J 2011;​38:​359-67.
58. Zhu N, Swietlik EM, Welch CL, et al. parameters and NT-proBNP levels and 84. Sitbon O, Channick R, Chin KM, et al.
Rare variant analysis of 4241 pulmonary their association with disease progres- Selexipag for the treatment of pulmonary
arterial hypertension cases from an inter- sion in patients with pulmonary arterial arterial hypertension. N Engl J Med 2015;​
national consortium implicates FBLN2, hypertension. Eur Heart J 2017;​38:​1147- 373:​2522-33.
PDGFD, and rare de novo variants in PAH. 55. 85. Galiè N, Manes A, Negro L, Palazzini
Genome Med 2021;​13:​80. 71. Kanwar MK, Gomberg-Maitland M, M, Bacchi-Reggiani ML, Branzi A. A meta-
59. Hemnes AR, Beck GJ, Newman JH, Hoeper M, et al. Risk stratification in pul- analysis of randomized controlled trials
et al. PVDOMICS: a multi-center study to monary arterial hypertension using Bayes- in pulmonary arterial hypertension. Eur
improve understanding of pulmonary vas- ian analysis. Eur Respir J 2020;​56:​2000008. Heart J 2009;​30:​394-403.
cular disease through phenomics. Circ Res 72. Thurnheer R, Ulrich S, Bloch KE. Pre- 86. Humbert M, Sitbon O, Chaouat A, et al.
2017;​121:​1136-9. capillary pulmonary hypertension and Survival in patients with idiopathic, fa-
60. Galiè N, Channick RN, Frantz RP, sleep-disordered breathing: is there a link? milial, and anorexigen-associated pulmo-
et al. Risk stratification and medical ther- Respiration 2017;​93:​65-77. nary arterial hypertension in the modern
apy of pulmonary arterial hypertension. 73. Rich S, Kaufmann E, Levy PS. The ef- management era. Circulation 2010;​122:​
Eur Respir J 2019;​53:​1801889. fect of high doses of calcium-channel 156-63.
61. Fijalkowska A, Kurzyna M, Torbicki blockers on survival in primary pulmo- 87. Benza RL, Miller DP, Barst RJ,
A, et al. Serum N-terminal brain natri- nary hypertension. N Engl J Med 1992;​ Badesch DB, Frost AE, McGoon MD. An
uretic peptide as a prognostic parameter 327:​76-81. evaluation of long-term survival from
in patients with pulmonary hypertension. 74. Fuster V, Steele PM, Edwards WD, time of diagnosis in pulmonary arterial
Chest 2006;​129:​1313-21. Gersh BJ, McGoon MD, Frye RL. Primary hypertension from the REVEAL Registry.
62. Coghlan JG, Denton CP, Grünig E, et al. pulmonary hypertension: natural history Chest 2012;​142:​448-56.
Evidence-based detection of pulmonary and the importance of thrombosis. Circu- 88. Sitbon O, Humbert M, Jaïs X, et al.
arterial hypertension in systemic sclero- lation 1984;​70:​580-7. Long-term response to calcium channel
sis: the DETECT study. Ann Rheum Dis 75. Olsson KM, Delcroix M, Ghofrani blockers in idiopathic pulmonary arterial
2014;​73:​1340-9. HA, et al. Anticoagulation and survival in hypertension. Circulation 2005;​111:​3105-
63. Rengier F, Melzig C, Derlin T, Marra pulmonary arterial hypertension: results 11.
AM, Vogel-Claussen J. Advanced imaging from the Comparative, Prospective Regis- 89. Sweatt AJ, Hedlin HK, Balasubrama-

n engl j med 385;25  nejm.org  December 16, 2021 2375


The New England Journal of Medicine
Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
Pulmonary Arterial Hypertension

nian V, et al. Discovery of distinct immune sion: an observational cohort study. Lan- of the randomized IMPRES study. Circu-
phenotypes using machine learning in cet Respir Med 2017;​5:​717-26. lation 2013;​127:​1128-38.
pulmonary arterial hypertension. Circ Res 92. Zamanian RT, Badesch D, Chung L, 94. Spiekerkoetter E, Sung YK, Sudheen-
2019;​124:​904-19. et al. Safety and efficacy of B-cell deple- dra D, et al. Randomised placebo-con-
90. Swift AJ, Lu H, Uthoff J, et al. A ma- tion with rituximab for the treatment of trolled safety and tolerability trial of
chine learning cardiac magnetic reso- systemic sclerosis-associated pulmonary FK506 (tacrolimus) for pulmonary arte-
nance approach to extract disease fea- arterial hypertension: a multicenter, dou- rial hypertension. Eur Respir J 2017;​50:​
tures and automate pulmonary arterial ble-blind, randomized, placebo-controlled 1602449.
hypertension diagnosis. Eur Heart J Car- trial. Am J Respir Crit Care Med 2021;​204:​ 95. Humbert M, McLaughlin V, Gibbs
diovasc Imaging 2021;​22:​236-45. 209-21. JSR, et al. Sotatercept for the treatment of
91. Rhodes CJ, Wharton J, Ghataorhe P, 93. Hoeper MM, Barst RJ, Bourge RC, et al. pulmonary arterial hypertension. N Engl J
et al. Plasma proteome analysis in pa- Imatinib mesylate as add-on therapy for Med 2021;​384:​1204-15.
tients with pulmonary arterial hyperten- pulmonary arterial hypertension: results Copyright © 2021 Massachusetts Medical Society.

images in clinical medicine


The Journal welcomes consideration of new submissions for Images in Clinical
Medicine. Instructions for authors and procedures for submissions can be found
on the Journal’s website at NEJM.org. At the discretion of the editor, images that
are accepted for publication may appear in the print version of the Journal,
the electronic version, or both.

2376 n engl j med 385;25  nejm.org  December 16, 2021

The New England Journal of Medicine


Downloaded from nejm.org by Vanina MARTIN on December 15, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.

You might also like