Hypercoagulability in
Pulmonary Hypertension
Isabel S. Bazan, MD
*
, Wassim H. Fares,
MD, MSC
INTRODUCTION
Pulmonary hypertension (PH) is defined as
increased pressure in the pulmonary circulation,
defined by convention and consensus as a mean
pulmonary artery pressure of at least 25 mm Hg
at rest.
1
The World Health Organization (WHO)
has classified PH into 5 major groups: pulmonary
arterial hypertension (PAH), PH caused by left
heart disease, PH caused by lung disease or
chronic hypoxia, PH caused by chronic thrombo-
embolic disease, and a miscellaneous group.
2,3
PAH is a clinical condition that falls under
WHO group 1, and can be idiopathic (IPAH), heri-
table (HPAH), caused by drugs and toxins
(DTPAH), or associated with several other condi-
tions (APAH) including connective tissue disease,
congenital heart disease, HIV infection, or portal
hypertension. PAH is characterized by molecular
and pathologic alterations in the pulmonary circu-
lation that result primarily in progressive vascular
remodeling of the pulmonary arteries, increased
pulmonary vascular resistance, and eventually
right heart failure and death.
4,5
These alterations
are caused by several inflammatory, metabolic,
and cellular changes that ultimately result in occlu-
sive lesions, in situ thromboses, and plexiform
lesions, that are all representative of the patho-
logic findings of PAH.
4,6,7
There is evidence of
pro-thrombotic pathobiology which suggests an
increased hypercoagulable state in PAH patients.
Based on limited evidence, anticoagulation ther-
apy is recommended in certain PH patients; how-
ever, the degree of hypercoagulability and benefit
of anticoagulant therapy are not known.
PATHOPHYSIOLOGY OF PULMONARY
ARTERIAL HYPERTENSION
PAH is characterized by excessive vasoconstric-
tion of the distal pulmonary arteries (although the
vasculopathy is not strictly limited to the pulmo-
nary arterial system
8
). This is related to endothelial
dysfunction and smooth muscle cell hypertrophy
and proliferation (that at least in part is related
to abnormal function or expression of potassium
channels on smooth muscle cells), which
leads to impaired production of vasodilator and
Disclosure Statement: The authors have no financial or commercial conflicts of interest to disclose. No funding
was used for this article.
Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, 300 Cedar Street, PO
Box 208057, New Haven, CT 06520-8057, USA
* Corresponding author. Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, 300
Cedar Street, TAC-441 South, PO Box 208057, New Haven, CT 06520-8057.
E-mail address: [email protected]
KEYWORDS
Pulmonary arterial hypertension
Hypercoagulability
Thromboembolism
Anticoagulation
Pulmonary hypertension
Pulmonary embolism
Right heart failure
Right ventricle
KEY POINTS
Patients with pulmonary arterial hypertension are at increased risk of developing thrombi.
There are known and suspected pathways that contribute to the hypercoagulability in patients with
pulmonary arterial hypertension.
The benefit of anticoagulation and antiplatelet therapy is not known in this patient population.
Hypercoagulability is an etiology and a consequence of pulmonary hypertension.
Clin Chest Med 39 (2018) 595–603
https://doi.org/10.1016/j.ccm.2018.04.005
0272-5231/18/Ó 2018 Elsevier Inc. All rights reserved.
chestmed.theclinics.com
antiproliferative agents such as nitric oxide and
prostacyclin, as well as overexpression of vaso-
constrictor and proliferative substances such as
thromboxane A2 and endothelin-1.
5
Other path-
ways and molecules, including serotonin,
9
have
also been implicated in the pathogenesis of PAH.
Activation of the endothelin pathway has been
demonstrated in both plasma and lung tissue of
PAH patients.
10
Although it is unclear whether
increased endothelin-1 is a cause or consequence
of PAH, it is known that endothelin-1 causes vaso-
constrictive and mitogenic effects by binding to
pulmonary vascular smooth muscle cells.
11
Endo-
thelin receptor antagonists are efficacious in
improving symptoms, exercise capacity, hemody-
namics, and time to clinical worsening in PAH
patients.
12,13
The nitric oxide and cyclic guanosine mono-
phosphate (cGMP) pathway is also important
in the pathogenesis of PAH. Inhibition of
cGMP destruction by phosphodiesterase type 5
(PDE-5) inhibitors results in pulmonary vasodila-
tion. PDE-5 inhibitors also have antiproliferative ef-
fects.
14
PDE-5 inhibitors and guanylate cyclase
stimulators are approved for the treatment of
PAH and have shown to varying degrees benefits
in hemodynamics, exercise capacity, and time
to clinical worsening as with endothelin receptor
antagonists.
2,15
The third pathway that has been a major thera-
peutic target for PAH is the prostacyclin pathway.
Prostacyclin is predominantly produced by endo-
thelial cells, and it induces potent vasodilation. It
also inhibits platelet aggregation, and has cytopro-
tective and antiproliferative effects.
16
PAH patients
have a reduction in prostacyclin synthase expres-
sion in pulmonary arteries and prostacyclin urinary
metabolites.
17
Synthetic analogs of prostacyclins
have been developed that share similar pharma-
codynamic effects of prostacyclin.
18–20
Efficacy
of prostanoids is also seen in APAH and CTEPH
(although currently not approved for CTEPH).
21–23
A common feature in all forms of PAH is the
vascular remodeling of the distal region of pulmo-
nary arteries. This pathologic remodeling results in
the formation of a layer of myofibroblasts and
extracellular matrix between the endothelium and
the internal elastic lamina, termed the neointima.
The cellular processes underlying the musculariza-
tion of the usually nonmuscular distal arteries is
incompletely understood, but the adventitial fibro-
blast is thought to be the first cell activated to
proliferate and synthesize matrix proteins in
response to a pulmonary hypertensive stimulus.
24
Upregulation of matrix metalloproteinases occurs,
and these metalloproteinases are involved in the
migration of the adventitial fibroblasts into the
media layer. PAH is also associated with alter-
ations of proliferation and apoptosis, resulting in
thickened and obstructive pulmonary arteries.
5
Endothelial cells also play a key role in vascular
remodeling. Disorganized endothelial cell prolifer-
ation leads to the formation of plexiform lesions
that are characteristic of PAH. The initiating stim-
ulus that results in abnormal endothelial prolifera-
tion is not known, but may be hypoxia, shear
stress, inflammation, response to drug or toxin,
or a combination of these with a background ge-
netic/genomic susceptibility. Defects in growth-
suppressive genes have been reported in plexi-
form lesions, including growth factors such as
platelet-derived growth factor, fibroblast growth
factor, transforming growth factor beta (TGFb),
and bone morphogenic proteins.
2,25
Inflammation also contributes to the pathogen-
esis of PAH. Pathologic specimens of patients
with PAH show an accumulation of perivascular in-
flammatory cells including macrophages, dendritic
cells, T and B lymphocytes, and mast cells. There
is also an increased level of circulating cytokines
and chemokines.
26–28
The role of inflammation
is particularly noted in certain groups of PAH
including HIV APAH and connective tissue disease
APAH. Interestingly, patients with systemic lupus
erythematosus APAH have improved on immuno-
suppressive therapy, emphasizing the role of
inflammation in this subset of patients.
29–31
The
pathogenesis of PH in patients with sickle cell dis-
ease (WHO group 5 PH) is also linked to inflamma-
tion, as elevated inflammatory markers and levels
of cytokines and chemokines are associated
with worse hemodynamics and poorer clinical out-
comes.
32,33
Mitochondrial dysfunction has also
been shown to be pathologic in PAH.
34
Pathologically, PAH results in medial hypertro-
phy, intimal proliferative and fibrotic changes,
adventitial thickening, plexiform lesions, and
thrombotic lesions in the distal pulmonary arteries.
Thrombi are present in both the small distal pulmo-
nary arteries and in proximal elastic pulmonary
arteries.
2
HYPERCOAGULABILITY IN PULMONARY
ARTERIAL HYPERTENSION
There is a high prevalence of vascular thrombotic
lesions found postmortem in patients with IPAH,
as described in several studies.
35–38
These in situ
thromboses may be caused by abnormalities in
the coagulation cascade, endothelial cells, and/or
platelets. Reduced plasma fibrinolysis was first re-
ported in1973.
39
Since then, studies have shown
that PAH patients have elevated plasma levels of
fibrinopeptide A- and D-dimers,
5
and 1 study found
Bazan & Fares
596
increased levels of fibrinogen and decreased
fibrinolytic response in patients with IPAH, compa-
rable to that of patients with CTEPH.
40
Fibrinopep-
tide A is generated when thrombin cleaves
fibrinogen, suggesting an elevated level of plasma
thrombin activity, and indeed studies have shown
increased thrombin activity in treatment-naı
¨ve
PAH patients.
41
Procoagulant activity and fibrinolytic function of
the pulmonary artery endothelium are also altered.
This dysfunction is reflected by the increased
levels of von Willebrand factor and plasminogen
activator inhibitor type-1 in the plasma of PAH pa-
tients. Plasminogen activator inhibitor was found
in much higher concentrations in arterial blood
than in mixed venous blood, suggesting intrapul-
monary production.
42
Additionally, shear stress
of blood flow toward vessel walls generates a
thrombogenic surface, resulting in thrombotic le-
sions. The effect of shear stress and vessel injury
can be seen in other types of PH as well, suggest-
ing that the prothrombotic state is not unique to
PAH.
5
Tissue factor is a transmembrane glycoprotein
that initiates the coagulation cascade, and it is
thought to play a role in angiogenesis and cancer
metastasis.
43–45
Tissue factor binds to factor VII
to catalyze the activation of factor X, leading to
the generation of thrombin and the formation of a
fibrin clot. Tissue factor expression is sensitive to
changes in blood flow, hypoxia, growth factors
such as platelet-derived growth factor, and che-
mokines. One study found that tissue factor
was upregulated in the diseased vessels of PAH
patients.
46
Another study found tissue factor-
expressing endothelial microparticles in the circu-
lation of PAH patients.
47
Tissue factor expression
may be a key contributor to the formation of in
situ thromboses.
There is growing evidence that the interaction
between platelets, and the arterial wall may
contribute to functional and structural alterations
in the pulmonary vessels. Apart from their known
role in coagulation, platelets release procoagulant,
vasoactive, and mitogenic mediators in response
to vascular abnormalities, such as thromboxane
A2, platelet-activating factor, serotonin, platelet-
derived growth factor, TGFb, and VEGF.
9,35,40
Thromboxane A2, which stimulates the activation
of new platelets and increases platelet aggrega-
tion, is increased in PAH patients, with a corre-
sponding reduction in prostacyclin metabolites.
48
Abnormal platelet aggregation has been described
in in vitro, in vivo, and human studies.
49–51
PAH
patients have higher levels of megakaryocyte-
stimulating hormone thrombopoietin, and 1 study
found that the pulmonary vasculature seemed to
be the site of production of thrombopoietin.
52
Increased platelet production, activation, and ag-
gregation may lead to a vicious cycle that contrib-
utes to thrombosis (Fig. 1). It is unclear whether
thrombosis and platelet dysfunction are causes
or consequences of PAH; however, the over-
arching evidence seems to be pointing toward an
underlying pathology of hypercoagulability as a
contributing etiology to PAH, and it gets worse
as PAH and right heart dysfunction ensue.
In addition to the previously mentioned patho-
physiologic abnormalities, patients with PAH may
also be at increased risk for venous thromboem-
bolism (VTE). PAH can cause significant dyspnea
with exertion, and right heart failure can result in
peripheral edema, both of which can lead a patient
to be immobile.
2
Additionally, heart failure alone is
an independent risk factor for VTE.
53
CHRONIC THROMBOEMBOLIC PULMONARY
HYPERTENSION
CTEPH results from the chronic obstruction of pul-
monary arteries due to thromboembolic disease.
Usually, acute pulmonary emboli are resorbed by
local fibrinolysis, with complete restoration of the
pulmonary arterial bed.
54
CTEPH arises when prior
acute pulmonary emboli for unknown reasons do
not completely resorb. These unresolved clots
then undergo fibrosis into an organized clot, ulti-
mately leading to mechanical obstruction of the
pulmonary arteries.
2,54
This obstruction causes
the release of inflammatory and vasculotropic me-
diators, resulting in vascular remodeling. Micro-
vascular disease is also thought to occur, which
can be related to shear stress in nonobstructed
areas, postcapillary remodeling related to
bronchial-to-pulmonary venous shunting, pres-
sure, and inflammation.
54
Low blood flow states
are created as a result of obstructed arteries and
can result in the in situ thromboses, related to
those of PAH. Thrombophilic factors such as
antiphospholipid antibodies, lupus anticoagulant,
protein S and C deficiency, activated protein C
resistance including factor V Leiden mutation,
prothrombin gene mutation, antithrombin III defi-
ciency, and elevated factor VIII have been statisti-
cally associated with approximately one-a third of
CTEPH patients.
2
Obstruction of pulmonary ar-
teries and secondary remodeling of small, periph-
eral pulmonary vessels most likely contribute to
elevated total pulmonary resistance.
8,55–57
Pathologically, organized thrombi are tightly
attached to the medial layer in the elastic pulmo-
nary arteries, and subsequently replace normal in-
tima. The thrombi occlude the lumen or form
different grades of stenosis, webs, and/or bands.
Hypercoagulability in Pulmonary Hypertension
597
Collateral vessels from the systemic circulation
(bronchial, costal, diaphragmatic, and coronary
arteries) can grow to attempt reperfusion of areas
distal to the obstructed territories. The microvas-
cular changes that occur in occluded and nonoc-
cluded areas are similar to those seen in PAH,
although plexiform lesions are uncommon.
2,58
It is estimated that 1% to 4% of acute pulmo-
nary embolism survivors develop CTEPH
within 2 years from their first embolic event.
55,59,60
Anywhere from 25% to 50% of CTEPH patients do
not have a past medical history of pulmonary em-
bolism or deep vein thrombosis.
55,61
It is sus-
pected that thrombotic and/or inflammatory
lesions exist in the pulmonary vasculature, result-
ing in the formation of thromboses and vascular
remodeling without a clinically evident acute
thromboembolic event. Conditions that cause
chronic inflammatory states such as myeloprolifer-
ative disorders and inflammatory bowel disease
Fig. 1. Summary of hypercoagulable pathways in pulmonary arterial hypertension. This figure depicts a schematic
of the pulmonary artery, with the combined known and suspected pathways that contribute to hypercoagulabil-
ity in PAH. This figure is not comprehensive or inclusive of all known hypercoagulability pathways implicated in
PAH. A condensed version of the coagulation cascade is shown, and in PAH, tissue factor (TF) is increased, result-
ing in increased thrombin, fibrin, and formation of clot. Plasminogen activator inhibitor (PAI) is also increased,
decreasing clot inhibitors. Decreased thrombomodulin reduces inhibition of the clot formation. There are also
more activated platelets in patients with PAH, and the effects of activated platelets are shown, including
increased inflammatory mediators, vasoconstriction, platelet aggregation and prothrombotic factors, fibrinogen,
and other adhesion molecules. Activated platelets stimulate growth factors, resulting in proliferation and angio-
genesis. Finally, the prostacyclin, endothelin-1, and nitric oxide pathways are shown, highlighting their differen-
tial effects on vasoconstriction and proliferation, and are the primary targets of the currently available
treatments for PAH. 5HT, serotonin; APC, activated protein C; aPlt, activated platelet; cGMP, cyclic guanosine
monophosphate; eNOS, endothelial nitric oxide synthase; FV, factor V; FVa, activated factor V; FVIIa, activated
factor VII; FX, factor X; FXa, activated factor X; GTP, guanosine triphosphate; NADPH, nicotinamide adenine dinu-
cleotide phosphate hydrogenase; O2, oxygen; PAI, plasminogen activator inhibitor; PC, protein C; PDGF, platelet-
derived growth factor; PGH2, prostaglandin H2; Plt, platelet; PS, protein S; TF, tissue factor; TGFb, transforming
growth factor b; tPA, tissue plasminogen activator; VEGF, vascular endothelial growth factor; vWF, von
Willebrand factor.
Bazan & Fares
598
and postsplenectomy patients have been associ-
ated with the development of CTEPH.
62
Unlike all
other forms of PH, patients with CTEPH are poten-
tially curable via a pulmonary endarterectomy.
63
The feasibility and success of this surgery depend
on the surgical accessibility of thromboembolic
residues and the underlying comorbidities of
the patient.
63,64
However, even with successful
removal of chronic clots, some patients continue
to have PH postoperatively. It is speculated that
persistent PH is partly due to the remodeled
microvasculature.
58
Balloon pulmonary angio-
plasty has also been gaining momentum in CTEPH
management; however, its exact role is yet to be
determined, as its technique, strategy, and cathe-
ters are rapidly evolving.
65
ANTICOAGULATION AND PULMONARY
HYPERTENSION
Currently, anticoagulation is rec ommended for
patients with IPAH, HPAH, and DTPAH. Accord-
ing to the 2015 European Society of Car diology
and the European Respiratory Society guidelines,
anticoagulation is a class IIb recommendation,
meaning that its usefulness and efficacy are
not we ll established, but it may be considered
(Table 1).
2
There have been some retrospective
and observational single-center studies that
have shown survival benefit in patients who get
anticoagulation with warfari n.
36,66
These early
studies were done prior to the available PAH-
targeted therapies.
36,66–68
More recent random-
ized control trials and registry data have been
inconclusive . For example, in 2014, the Compar-
ative, Prospective Registry of Newly Initiated
Therapies for Pulmonary Hypertension (COM-
PERA) regis try examined the survival rates of
PAH patient s based on use of anticoagulatio n.
It found that in the subgroup of IPAH patients,
there was a significant improvement in 3-year
survival.
69
A major caveat to the COMPERA reg-
istry is tha t the median age of this population
receiving anticoagulation was 70 years, and it
does not ref lect the typical demographics of the
PAH popul ation.
In contrast, the Registry to Evaluate Early and
Long-term PAH Disease Management (REVEAL)
compared the survival of IPAH patients on warfarin
with those who have never been anticoagulated,
and found no difference in survival.
70
These limited
and inconclusive data likely reflect the heteroge-
neity of PAH patients. Clinical use of anticoagula-
tion in IPAH patients is widely variable and
provider dependent. It is generally recommended
that in the absence of contraindications, patients
on long-term intravenous prostanoid analogues
should receive anticoagulation therapy because
of the risk of catheter-associated thrombosis.
2
The potential benefits of anticoagulation for pa-
tients with APAH is even less clear. A subgroup
analysis of from the REVEAL registry found that
patients with systemic sclerosis APAH had an
increased mortality when treated with warfarin.
70
The COMPERA registry also found to have other
forms of APAH including those associated with
connective tissue diseases, congenital heart dis-
ease, and portopulmonary hypertension, had no
Table 1
Summary of evidence for anticoagulation by
World Health Organization groups
WHO Group Class of Recommendation
a
1 PAH IPAH IIb
HPAH IIb
DTPAH IIb
CTD-PAH III
Porto-PH III
HIV-PAH Unknown
CHD-PAH Unknown
2 PH caused by
left heart
disease
Unknown
3 PH caused by
lung disease/
chronic
hypoxia
Unknown
4 CTEPH I
5 Miscellaneous Unknown
Abbreviations: CHD-PAH, congenital heart disease-
pulmonary arterial hypertension; CTD-PAH, connective tis-
sue disease-pulmonary arterial hypertension; CTEPH,
chronic thromboembolic pulmonary hypertension; DTPAH,
drugs/toxins induced pulmonary arterial hypertension;
HIV-PAH, HIV-induced pulmonary arterial hypertension;
HPAH, heritable pulmonary arterial hypertension; IPAH,
idiopathicpulmonaryarterialhypertension;PH, pulmonary
hypertension; porto-PH, portopulmonary hypertension.
a
Class of recommendation. Class I recommendation: is
recommended/indicated (evidence and/or general agree-
ment that a given treatment of procedure is beneficial,
useful, effective). Class IIa recommendation: should be
considered (conflicting evidence with weight of evi-
dence/opinion in favor of usefulness/efficacy). Class IIb:
may be considered (conflicting evidence with usefulness/
efficacy less well established). Class III: is not recommen-
ded (evidence or general agreement that given treatment
is not useful/effective, and in some cases may be harmful).
Data from Galie
`
N, Humbert M, Vachiery JL, et al. 2015
ESC/ERS guidelines for the diagnosis and treatment of pul-
monary hypertension. The joint task force for the diag-
nosis and treatment of pulmonary hypertension of the
European Society of Cardiology (ESC) and the European
Respiratory Society (ERS) endorsed by: Association for Eu-
ropean Paediatric and Congenital Cardiology (AEPC), In-
ternational Society for Heart and Lung Transplantation
(ISHLT). Eur Heart J 2015:37(1);67–119.
Hypercoagulability in Pulmonary Hypertension
599
survival benefit with anticoagulation.
69
Specific to
congenital heart disease APAH, the use of antico-
agulation in patients with Eisenmenger syndrome
is controversial. These patients have a high inci-
dence of pulmonary artery thrombosis and stroke,
but also have an elevated risk of hemorrhage and
hemoptysis.
71
Although there are no data to guide
this clinical dilemma, the authors’ own practice is
to avoid anticoagulation in congenital heart dis-
ease APAH. Hemoptysis is also a known compli-
cation of patients with IPAH and CTEPH, with
prevalence varying from 1% to 6%,
72
and it can
limit the use of anticoagulation. Because many pa-
tients with portopulmonary hypertension have
coagulopathy and thrombocytopenia, they have
an elevated bleeding risk; anticoagulation is not
recommended in these patients, although it has
not been studied.
The preferred treatment for patients with
CTEPH, as mentioned previously, is a pulmonary
endarterectomy. Supplemental medical therapy
for CTEPH includes anticoagulation, as well as di-
uretics and supplemental oxygen if needed for
heart failure or hypoxemia, respectively. Although
there are no studies comparing indefinite anticoa-
gulation to no therapy or a shorter duration of anti-
coagulation, data extrapolated from treatment of
acute and recurrent venous thromboembolism
have led to the recommendation of lifelong antico-
agulation for CTEPH patients, even after success-
ful surgical intervention. The placement of routine
inferior vena cava filter placement in this patient
population is not justified by evidence.
2
The role of direct oral anticoagulants (DOACs)
for any group of PH is unknown. Several studies
have shown that DOACs are at least as effective
as warfarin for the management of venous throm-
boembolism and atrial fibrillation, and some have
shown a reduction in bleeding and mortality.
73,74
DOACs have not yet been studied in people with
PAH, but in a monocrotaline-induced PAH rat
model, rivaroxaban attenuated the increase in
right ventricular systolic pressure and right ventric-
ular hypertrophy caused by monocrotaline.
75
Although the generalizability of this to people is
limited, it does warrant further investigation into
the role of DOACs in PAH.
Since dysregulated platelets have been identi-
fied in the pathophysiology of PAH, antiplatelet
therapy has been studied. However, although
some studies have shown reduction in throm-
boxane A2 levels and reduction in platelet activa-
tion markers with aspirin, none have shown any
improvement in exercise tolerance.
76,77
These
studies are small, and it is possible that larger
studies or the investigation of newer antiplatelet
agents may identify a role for platelet inhibitors in
the treatment of PAH. It is worth noting, however,
that many of the PAH medications, as mentioned
previously, do have antiplatelet effects.
50,78
SUMMARY
Given how heterogeneous the etiologies and
pathophysiology are for each WHO group o f
PH, it is no surprise that the degree of hyperco-
agulability and benefit of anticoagula tion would
be variable between groups and subtypes.
CTEPH is usually caused by an initial ac ute
venous thromboembolism and has been associ-
ated with thrombophilic disorders; anticoagula-
tion has a clear benefit in these patients. There
is pathophysiological evidence that PAH is a pro-
thrombotic state caused by the dysregulation of
coagulation, fibrinolysis, and endothelial cells.
These abnormalities, combined with in situ
thromboses found in pulmonary arteries, argue
that PAH patients are in a hypercoagulable state
and may benefit from anticoa gulation therapy.
Studies investigating the b enefit of anticoagula-
tion in IPAH patients have yielde d mixed results.
Each individual subtype of APAH will need to be
further investigated to assess the benefit of anti-
coagulation. Given that an increased bleeding
risk limits the use of anticoagulation even in pa -
tients who would derive benefit, further study of
DOACs and antiplatelet agents is also overdue.
ACKNOWLEDGMENTS
The authors have grateful to Ms Isabella Siegel
for her artistic and creative skills in drawing the
figure.
REFERENCES
1. Maron BA, Wertheim BM, Gladwin MT. Under pres-
sure to clarify pulmonary hypertension clinical risk.
Am J Respir Crit Care Med 2018;197(4):423–6.
2. Galie
`
N, Humbert M, Vachiery JL, et al. 2015 ESC/
ERS guidelines for the diagnosis and treatment of
pulmonary hypertension. The joint task force for
the diagnosis and treatment of pulmonary hyperten-
sion of the European Society of Cardiology (ESC)
and the European Respiratory Society (ERS)
endorsed by: Association for European Paediatric
and Congenital Cardiology (AEPC), International So-
ciety for Heart and Lung Transplantation (ISHLT).
Eur Heart J 2015;37(1):67–119.
3. Bazan IS, Fares WH. Pulmonary hypertension: diag-
nostic and therapeutic challenges. Ther Clin Risk
Manag 2015;11:1221–33.
4. Robinson JC, Pugliese SC, Fox DL, et al. Anticoagu-
lation in pulmonary arterial hypertension. Curr Hy-
pertens Rep 2016;18(6):47.
Bazan & Fares
600
5. Humbert M, Morrell NW, Archer SL, et al. Cellular
and molecular pathobiology of pulmonary arterial
hypertension. J Am Coll Cardiol 2004;43(12 Suppl
S):13S–24S.
6. Tuder RM, Stacher E, Robinson J, et al. Pathology of
pulmonary hypertension. Clin Chest Med 2013;
34(4):639–50.
7. Tuder RM, Voelkel NF. Plexiform lesion in severe pul-
monary hypertension: association with glomeruloid
lesion. Am J Pathol 2001;159(1):382–3.
8. Fares WH. The other vascular beds in pulmonary
arterial hypertension. Surrogates or associated?
Ann Am Thorac Soc 2014;11(4):596–7.
9. Bazan IS, Fares WH. Review of the ongoing story of
appetite suppressants, serotonin pathway, and
pulmonary vascular disease. Am J Cardiol 2016;
117(10):1691–6.
10. Giaid A, Yanagisawa M, Langleben D, et al. Expres-
sion of endothelin-1 in the lungs of patients with pul-
monary hypertension. New Engl J Med 1993;
328(24):1732–9.
11. Galie N, Manes A, Branzi A. The endothelin system
in pulmonary arterial hypertension. Cardiovasc Res
2004;61(2):227–37.
12. Galie N, Olschewski H, Oudiz RJ, et al.
Ambrisentan for the treatment of pulmonary arte-
rial hypertension: results of the ambrisentan in
pulmonary arterial hypertension, randomized,
double-blind, placebo-controlled, multicenter, effi-
cacy (ARIES) study 1 and 2. Circulation 2008;
117(23):3010–9.
13. Pulido T, Adzerikho I, Channick RN, et al. Macitentan
and morbidity and mortality in pulmonary arterial hy-
pertension. N Engl J Med 2013;369(9):809–18.
14. Wharton J, Strange JW, Møller GM, et al. Antiprolifer-
ative effects of phosphodiesterase type 5 inhibition
in human pulmonary artery cells. Am J Respir Crit
Care Med 2005;172(1):105–13.
15. Galie
`
N, Ghofrani HA, Torbicki A, et al. Sildenafil cit-
rate therapy for pulmonary arterial hypertension.
New Engl J Med 2005;353(20):2148–57.
16. Jones DA, Benjamin CW, Linseman DA. Activation of
thromboxane and prostacyclin receptors elicits
opposing effects on vascular smooth muscle cell
growth and mitogen-activated protein kinase
signaling cascades. Mol Pharmacol 1995;48(5):
890–6.
17. Galie N, Manes A, Branzi A. Prostanoids for pulmo-
nary arterial hypertension. Am J Respir Med 2003;
2(2):123–37.
18. Barst RJ, Rubin LJ, Long WA, et al. A comparison of
continuous intravenous epoprostenol (prostacyclin)
with conventional therapy for primary pulmonary hy-
pertension. N Engl J Med 1996;334(5):296–301.
19. Rubin LJ, Mendoza J, Hood M, et al. Treatment of
primary pulmonary hypertension with continuous
intravenous prostacyclin (epoprostenol). Results of
a randomized trial. Ann Intern Med 1990;112(7):
485–91.
20. Badesch DB, Tapson VF, McGoon MD, et al. Contin-
uous intravenous epoprostenol for pulmonary hyper-
tension due to the scleroderma spectrum of
disease. A randomized, controlled trial. Ann Intern
Med 2000;132(6):425–34.
21. Nunes H, Humbert M, Sitbon O, et al. Prognostic
factors for survival in human immunodeficiency
virus-associated pulmonary arterial hypertension.
Am J Respir Crit Care Med 2003;167(10):1433–9.
22. Rosenzweig EB, Kerstein D, Barst RJ. Long-term
prostacyclin for pulmonary hypertension with asso-
ciated congenital heart defects. Circulation 1999;
99(14):1858–65.
23. Cabrol S, Souza R, Jais X, et al. Intravenous epo-
prostenol in inoperable chronic thromboembolic pul-
monary hypertension. J Heart Lung Transplant 2007;
26(4):357–62.
24. Stenmark KR, Gerasimovskaya E, Nemenoff RA,
et al. Hypoxic activation of adventitial fibroblasts:
role in vascular remodeling. Chest 2002;122(6
Suppl):326s–34s.
25. Yeager ME, Halley GR, Golpon HA, et al. Micro-
satellite instability of endothelial cell growth and
apoptosis genes within plexiform lesions in pri-
mary pulmonary hypertension. Circ Res 2001;
88(1):E2–11.
26. Humbert M, Monti G, Fartoukh M, et al. Platelet-
derived growth factor expression in primary pulmo-
nary hypertension: comparison of HIV seropositive
and HIV seronegative patients. Eur Respir J 1998;
11(3):554–9.
27. Fares WH, Ford HJ, Ghio AJ, et al. Safety and feasi-
bility of obtaining wedged pulmonary artery samples
and differential distribution of biomarkers in pulmo-
nary hypertension. Pulm Circ 2012;2(4):477–82.
28. Marshall JD, Sauler M, Tonelli A, et al. Complexity of
macrophage migration inhibitory factor (MIF) and
other angiogenic biomarkers profiling in pulmonary
arterial hypertension. Pulm Circ 2017;7(3):730–3.
29. Dorfmuller P, Perros F, Balabanian K, et al. Inflamma-
tion in pulmonary arterial hypertension. Eur Respir J
2003;22(2):358–63.
30. Mensah KA, Yadav R, Trow TK, et al. Lupus-associ-
ated pulmonary arterial hypertension: variable
course and importance of prompt recognition.
Case Rep Med 2015;2015:328435.
31. Bazan IS, Mensah KA, Rudkovskaia AA, et al. Pul-
monary arterial hypertension in the setting of sclero-
derma is different than in the setting of lupus: a
review. Respir Med 2018;134:42–6.
32. Parent F, Bachir D, Inamo J, et al. A hemodynamic
study of pulmonary hypertension in sickle cell dis-
ease. N Engl J Med 2011;365(1):44–53.
33. Niu X, Nouraie M, Campbell A, et al. Angiogenic and
inflammatory markers of cardiopulmonary changes
Hypercoagulability in Pulmonary Hypertension
601
in children and adolescents with sickle cell disease.
PLoS One 2009;4(11):e7956.
34. Marshall JD, Bazan I, Zhang Y, et al. Mitochondrial
dysfunction and pulmonary hypertension: cause, ef-
fect, or both. Am J Physiol Lung Cell Mol Physiol
2018;314(5):L782–96.
35. Herve P, Humbert M, Sitbon O, et al. Pathobiology of
pulmonary hypertension. The role of platelets and
thrombosis. Clin Chest Med 2001;22(3):451–8.
36. Fuster V, Steele PM, Edwards WD, et al. Primary
pulmonary hypertension: natural history and the
importance of thrombosis. Circulation 1984;70(4):
580–7.
37. Wagenvoort CA, Mulder PGH. Thrombotic lesions in
primary plexogenic arteriopathy. Chest 1993;103(3):
844–9.
38. Johnson SR, Granton JT, Mehta S. Thrombotic arte-
riopathy and anticoagulation in pulmonary hyperten-
sion. Chest 2006;130(2):545–52.
39. Inglesby TV, Singer JW, Gordon DS. Abnormal fibri-
nolysis in familial pulmonary hypertension. Am J
Med 1973;55(1):5–14.
40. Huber K, Beckmann R, Frank H, et al. Fibrinogen,
t-PA, and PAI-1 plasma levels in patients with pulmo-
nary hypertension. Am J Respir Crit Care Med 1994;
150(4):929–33.
41. Tournier A, Wahl D, Chaouat A, et al. Calibrated
automated thrombography demonstrates hyperco-
agulability in patients with idiopathic pulmonary
arterial hypertension. Thromb Res 2010;126(6):
e418–22.
42. Hoeper MM, Sosada M, Fabel H. Plasma coagula-
tion profiles in patients with severe primary pulmo-
nary hypertension. Eur Respir J 1998;12(6):1446–9.
43. Ruf W, Mueller BM. Tissue factor signaling. Thromb
Haemost 1999;82(2):175–82.
44. Riewald M, Ruf W. Orchestration of coagulation pro-
tease signaling by tissue factor. Trends Cardiovasc
Med 2002;12(4):149–54.
45. Turitto VT, Hall CL. Mechanical factors affecting he-
mostasis and thrombosis. Thromb Res 1998;92(6
Suppl 2):S25–31.
46. White RJ, Meoli DF, Swarthout RF, et al. Plexiform-
like lesions and increased tissue factor expression
in a rat model of severe pulmonary arterial hyperten-
sion. Am J Physiol Lung Cell Mol Physiol 2007;
293(3):L583–90.
47. Bakouboula B, Morel O, Faure A, et al. Procoagulant
membrane microparticles correlate with the severity
of pulmonary arterial hypertension. Am J Respir Crit
Care Med 2008;177(5):536–43.
48. Christman BW, McPherson CD, Newman JH, et al.
An imbalance between the excretion of throm-
boxane and prostacyclin metabolites in pulmonary
hypertension. N Engl J Med 1992;327(2):70–5.
49. Lopes AA, Maeda NY, Almeida A, et al. Circulating
platelet aggregates indicative of in vivo platelet
activation in pulmonary hypertension. Angiology
1993;44(9):701–6.
50. Clapp LH, Gurung R. The mechanistic basis of pros-
tacyclin and its stable analogues in pulmonary arte-
rial hypertension: role of membrane versus nuclear
receptors. Prostaglandins Other Lipid Mediat 2015;
120:56–71.
51. Friedman R, Mears JG, Barst RJ. Continuous infu-
sion of prostacyclin normalizes plasma markers of
endothelial cell injury and platelet aggregation in pri-
mary pulmonary hypertension. Circulation 1997;
96(9):2782–4.
52. Haznedaro
glu IC, Atalar E, Oztu
¨
rk MA, et al. Throm-
bopoietin inside the pulmonary vessels in patients
with and without pulmonary hypertension. Platelets
2002;13(7):395–9.
53. Tang L, Wu YY, Lip GY, et al. Heart failure and risk of
venous thromboembolism: a systematic review and
meta-analysis. Lancet Haematol 2016;3(1):e30–44.
54. Dartevelle P, Fadel E, Mussot S, et al. Chronic throm-
boembolic pulmonary hypertension. Eur Respir J
2004;23(4):637–48.
55. Dorfmuller P, Gu
¨
nther S, Ghigna MR, et al. Microvas-
cular disease in chronic thromboembolic pulmonary
hypertension: a role for pulmonary veins and sys-
temic vasculature. Eur Respir J 2014;44(5):1275–88.
56. Moser KM, Bloor CM. Pulmonary vascular lesions
occurring in patients with chronic major vessel
thromboembolic pulmonary hypertension. Chest
1993;103(3):685–92.
57. Hoeper MM, Mayer E, Simonneau G, et al.
Chronic thromboembolic pulmonary hypertension.
Circulation 2006;113(16):2011–20.
58. Galie N, Kim NH. Pulmonary microvascular disease
in chronic thromboembolic pulmonary hypertension.
Proc Am Thorac Soc 2006;3(7):571–6.
59. Pengo V, Lensing AW, Prins MH, et al. Incidence of
chronic thromboembolic pulmonary hypertension af-
ter pulmonary embolism. N Engl J Med 2004;
350(22):2257–64, 2004 Massachusetts Medical So-
ciety: United States.
60. Becattini C, Agnelli G, Pesavento R, et al. Incidence
of chronic thromboembolic pulmonary hypertension
after a first episode of pulmonary embolism. Chest
2006;130(1):172–5.
61. Pepke-Zaba J, Delcroix M, Lang I, et al. Chronic
thromboembolic pulmonary hypertension (CTEPH):
results from an international prospective registry.
Circulation 2011;124(18):1973–81.
62. Galie
`
N, Hoeper MM, Humbert M, et al. Guidelines
for the diagnosis and treatment of pulmonary hyper-
tension: the task force for the diagnosis and treat-
ment of pulmonary hypertension of the European
Society of Cardiology (ESC) and the European Res-
piratory Society (ERS), endorsed by the International
Society of Heart and Lung Transplantation (ISHLT).
Eur Heart J 2009;30:2493–537.
Bazan & Fares
602
63. Jamieson SW, Kapelanski DP, Sakakibara N, et al.
Pulmonary endarterectomy: experience and lessons
learned in 1,500 cases. Ann Thorac Surg 2003;
76(5):1457–62 [discussion: 1462–4].
64. Jenkins DP, Madani M, Mayer E, et al. Surgical treat-
ment of chronic thromboembolic pulmonary hyper-
tension. Eur Respir J 2013;41(3):735–42.
65. Auger WR, Kim NH. Balloon pulmonary angioplasty
for chronic thromboembolic pulmonary hyperten-
sion: more work to be done. Circ Cardiovasc Qual
Outcomes 2017;10(11) [pii:e004230].
66. Rich S, Kaufmann E, Levy PS. Levy the effect of high
doses of calcium-channel blockers on survival in
primary pulmonary hypertension. New Engl J Med
1992;327(2):76–81.
67. Ogata M, Ohe M, Shirato K, et al. Effects of a com-
bination therapy of anticoagulant and vasodilator on
the long-term prognosis of primary pulmonary hy-
pertension. Jpn Circ J 1993;57(1):63–9.
68. Frank H, Mlczoch J, Huber K, et al. The effect of anti-
coagulant therapy in primary and anorectic drug-
induced pulmonary hypertension. Chest 1997;
112(3):714–21.
69. Olsson KM, Delcroix M, Ghofrani HA, et al. Anticoa-
gulation and survival in pulmonary arterial hyperten-
sion: results from the comparative, prospective
registry of newly initiated therapies for pulmonary
hypertension (COMPERA). Circulation 2014;129(1):
57–65.
70. Preston IR, Roberts KE, Miller DP, et al. Effect
of warfarin treatment on survival of patients with
Pulmonary Arterial Hypertension (PAH) in the Regis-
try to Evaluate Early and Long-Term PAH Disease
Management (REVEAL). Circulation 2015;132(25):
2403–11.
71. Broberg CS, Ujita M, Prasad S, et al. Pulmonary
arterial thrombosis in eisenmenger syndrome is
associated with biventricular dysfunction and
decreased pulmonary flow velocity. J Am Coll Car-
diol 2007;50(7):634–42.
72. Zylkowska J, Kurzyna M, Pietura R, et al. Recurrent
hemoptysis: an emerging life-threatening complica-
tion in idiopathic pulmonary arterial hypertension.
Chest 2011;139(3):690–3.
73. Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban
versus warfarin in nonvalvular atrial fibrillation. New
Engl J Med 2011;365(10):883–91.
74. Schulman S, Kearon C, Kakkar AK, et al. Dabigatran
versus Warfarin in the treatment of acute venous
thromboembolism. New Engl J Med 2009;361(24):
2342–52.
75. Delbeck M, Nickel KF, Perzborn E, et al. A role for
coagulation factor Xa in experimental pulmonary
arterial hypertension. Cardiovasc Res 2011;92(1):
159–68.
76. Kawut SM, Bagiella E, Lederer DJ, et al.
A randomized clinical trial of aspirin and simvastatin
for pulmonary arterial hypertension: ASA-STAT. Cir-
culation 2011;123(25):2985–93.
77. Robbins IM, Kawut SM, Yung D, et al. A study of
aspirin and clopidogrel in idiopathic pulmonary arte-
rial hypertension. Eur Respir J 2006;27(3):578–84.
78. Makowski CT, Rissmiller RW, Bullington WM. Rioci-
guat: a novel new drug for treatment of pulmonary
hypertension. Pharmacotherapy 2015;35(5):502–19.
Hypercoagulability in Pulmonary Hypertension
603