You are on page 1of 10

Am J Cardiovasc Drugs (2015) 15:225–234

DOI 10.1007/s40256-015-0125-4

LEADING ARTICLE

Novel Targets of Drug Treatment for Pulmonary Hypertension


Jian Hu1 • Qinzi Xu1 • Charles McTiernan1 • Yen-Chun Lai1 •

David Osei-Hwedieh1 • Mark Gladwin1,2

Published online: 28 May 2015


Ó Springer International Publishing Switzerland 2015

Abstract Biomedical advances over the last decade have of categorizing patients on the basis of molecular pheno-
identified the central role of proliferative pulmonary arterial type(s) for effective treatment of the disease.
smooth muscle cells (PASMCs) in the development of pul-
monary hypertension (PH). Furthermore, promoters of pro-
liferation and apoptosis resistance in PASMCs and endothelial
Key Points
cells, such as aberrant signal pathways involving growth
factors, G protein-coupled receptors, kinases, and mi-
Several novel strategies have emerged for pulmonary
croRNAs, have also been described. As a result of these dis-
hypertension in recent years.
coveries, PH is currently divided into subgroups based on the
underlying pathology, which allows focused and targeted Pre-clinical and clinical trials have been performed
treatment of the condition. The defining features of PH, which to evaluate the novel strategies.
subsequently lead to vascular wall remodeling, are dys- The following drugs, acting on novel targets, are
regulated proliferation of PASMCs, local inflammation, and currently being evaluated in clinical trials: imatinib
apoptosis-resistant endothelial cells. Efforts to assess the (phase III); nilotinib, AT-877ER, rituximab,
relative contributions of these factors have generated several tacrolimus, paroxetine, sertraline, fluoxetine,
promising targets. This review discusses recent novel targets bardoxolone methyl (phase II); and sorafenib,
of therapies for PH that have been developed as a result of FK506, aviptadil, endothelial progenitor cells (EPCs)
these advances, which are now in pre-clinical and clinical (phase I).
trials (e.g., imatinib [phase III]; nilotinib, AT-877ER, ritux-
imab, tacrolimus, paroxetine, sertraline, fluoxetine, bardox-
olone methyl [phase II]; and sorafenib, FK506, aviptadil,
endothelial progenitor cells (EPCs) [phase I]). While sub-
stantial progress has been made in recent years in targeting key 1 Introduction
molecular pathways, PH still remains without a cure, and these
novel therapies provide an important conceptual framework Pulmonary hypertension (PH) is a progressive vascular dis-
ease caused by vasoconstriction and structural remodeling of
arterioles that result in the reduction of vessel elasticity and
inner diameter. These changes increase the resting mean
& Jian Hu pulmonary arterial pressure (PAP) (C25 mmHg) and can
jih67@pitt.edu lead to dyspnea, fatigue, cough, chest pain, palpitations,
1 peripheral edema, syncope, right heart failure, and death [1,
Pittsburgh Heart, Lung, Blood and Vascular Medicine
Institute, University of Pittsburgh, E1226 BST 200 Lothrop 2]. In the US population, the age-standardized mortality rate
Street, Pittsburgh, PA 15213, USA for PH has significantly increased from 5.5 per 100,000
2
Division of Pulmonary, Allergy and Critical Care Medicine, population in 2001 to 6.5 per 100,000 in 2010. In 2010,
University of Pittsburgh, Pittsburgh, USA women showed significantly higher mortality rates than
226 J. Hu et al.

men, and non-Hispanic blacks showed significantly higher humans, and to exert pro-apoptotic effects in isolated
mortality rates than non-Hispanic whites. Aged population PASMCs [7, 8]. Imatinib has also been reported to prevent
cohorts (75–84 and C85 years of age) show the highest PH right ventricular and pulmonary vascular remodeling and to
mortality (47.9/100,000 and 108.7/100,000, respectively, improve pulmonary hemodynamics in animal models [5,
2010 data) [3]. Mechanisms for the development of PH are 9]. After several reported cases of improved exercise ca-
complex and may be idiopathic, genetic, or secondary to left pacity and hemodynamics of PH patients given imatinib
heart disease, lung diseases, and/or chronic thromboem- treatment [10–14], the compound was evaluated for safety,
bolism. Five subgroups of PH have been defined on the basis tolerability, and efficacy in 59 PH patients in a double-
of the etiology [1], and current research progress on the novel blinded, placebo-controlled multicenter, phase II trial [15].
therapeutic targets is focused on group 1 PH [pulmonary This clinical trial has shown that imatinib decreases PVR
arterial hypertension (PAH)]. and increases cardiac output, but has no significant im-
PAH is characterized by a progressive increase in pul- provement in the 6-min walk distance (6MWD). However,
monary vascular resistance (PVR) caused by pulmonary subgroup analysis revealed that patients with severely im-
artery remodeling and vaso-occlusion. The most important proved PVR showed a significant increase in 6MWD and
mechanism for PAH development is endothelial dysfunction hemodynamics after receiving imatinib. In a phase III trial
and/or injury, which leads to imbalanced production of en- of 202 patients with severe PH, patients receiving imatinib
dogenous vasodilators, such as prostacyclin and nitric oxide, showed a significant improvement in 6MWD, decreased
and vasoconstrictors, such as endothelin and serotonin. mean PAP and PVR, and increased cardiac output [16].
Without treatment, the survival rates of PAH at 1 year, 3, and Nilotinib is another TKI that shows greater potency and
5 years are 68, 48, and 34 %, respectively [4]. Contemporary selectivity in inhibition of the BCR-ABL tyrosine kinase than
drugs used to treat PAH target three pathways: the cyclic does imatinib. Nilotinib has been demonstrated to prevent an-
guanosine monophosphate signaling pathway (e.g., sildenafil gioproliferation in a rodent model of PH with systemic sclerosis
and tadalafil), prostacyclin signaling pathway (e.g., epopros- [17]. However, a phase II clinical trial conducted in 23 patients
tenol and iloprost), and endothelin signaling pathway (e.g., was terminated because of serious adverse effects including
bosentan and ambrisentan). More recently, new drugs tar- cardiopulmonary disorders (e.g., pulseless electrical activity,
geting these major pathways (e.g., riociguat, selexipag, and right ventricular dysfunction, acute respiratory failure, and
macitentan) have been shown to be effective in the treatment PAH worsening) and hepatobiliary disorders [e.g., cholecystitis
of PAH and have been approved by US Food and Drug and cholelithiasis (NCT01179737)] [18]. Sorafenib is a multi-
Administration (FDA). Although current treatments appear to kinase inhibitor targeting PDGF receptor (PDGFR), VEGF
relieve symptoms, improve exercise capacity, and prevent receptor (VEGFR), c-kit, and Raf. In a mouse PH model, it has
hospitalizations, only limited evidence for disease reversal been shown to prevent hemodynamic changes and attenuate
has been observed. In recent years, several novel strategies PH-associated vascular remodeling by abolishing the
have emerged for PH treatment, and this article focuses on upregulation of the mitogen-activated protein kinase (MAPK)
new and promising therapeutic approaches. cascade [19]. In a clinical trial of 22 patients with PH, sorafenib
treatment generated a modest improvement in the exercise
capacity without any alteration in cardiac function [20]. It was
2 Novel Targets of Drug Treatment well tolerated at a 200-mg twice-daily dose, with the most
common adverse effects being moderate skin reaction and
2.1 Receptor Tyrosine Kinase Inhibitors alopecia.
However, there are reports showing that some other agents
Receptor tyrosine kinases (RTKs) are a class of high-affi- in the family of TKIs induce PH. For example, treatment of
nity cell surface receptors for a variety of growth factors, dasatinib in patients with leukemia induced PH [21–23].
hormones, and cytokine ligands. Platelet-derived growth Sugen5416, another protein kinase inhibitor, also induces PH
factor (PDGF), epidermal growth factor (EGF), fibroblast in rodents in combination with hypoxia exposure [24]. In
growth factor (FGF), and c-kit-receptors play pathogenic summary, although some TKIs offer promising new treat-
roles in the development of PH [5, 6], as these RTK ligands ments for PH, more studies are needed to understand how to
increase proliferation and migration of smooth muscle cells use them with minimal adverse effects and maximal efficacy.
(SMCs) and endothelial cells (ECs) in lungs.
Several tyrosine kinase inhibitors (TKIs) are currently 2.2 Rho-Kinase Inhibitor
under development. Imatinib (GleevecÒ) inhibits PDGF,
c-kit, and c-Abl receptors. The use of imatinib has been RhoA is a small monomeric G-protein that belongs to Ras
shown to inhibit proliferation and migration of pulmonary homologous (Rho) family. RhoA and its downstream
arterial smooth muscle cells (PASMCs) in rodents and substrate Rho-kinase (ROCK) play significant roles in the
Developing Drugs for Pulmonary Hypertension 227

pathogenesis of PH, mediating vascular remodeling and in this trial have been reported to have a reduction in PVR
vasoconstriction [25–28]. Long-term inhibition of ROCK of [20 %. Additionally, a combination of VIP and en-
with fasudil, a selective ROCK inhibitor, improved dothelin receptor antagonist, bosentan, has been tested in
pathological parameters of PH and pulmonary artery re- pre-clinical PH models. This combination therapy was more
modeling in monocrotaline- or hypoxia-induced rodent effective than either drug alone, perhaps because of the
models of PH [29–31]. Fasudil was the first ROCK in- synergistic effects of both drugs on suppression of the en-
hibitor approved in 1995 for the treatment of subarachnoid dothelin-endothelin (ET-ET) receptor pathway [51].
hemorrhage–induced brain vessel vasospasms [32, 33].
Clinical trials have shown that intravenous administration 2.4 Selective Serotonin Reuptake Inhibitors
of fasudil has acute beneficial vasodilatory effects on the
pulmonary circulation in PH patients [34, 35]. Inhalation of Upregulation of serotonin released from the pulmonary
fasudil was shown to be as effective as nitric oxide in- endothelium is associated with the development of PH in
halation in PH patients [36]. It was also reported that fa- animal models, likely through its effects on pulmonary
sudil reduced pulmonary artery pressure in patients with vasoconstriction and SMC proliferation [52–61]. Selective
high-altitude PH in a randomized, double-blind study [37]. serotonin reuptake inhibitors (SSRIs) can attenuate and
AT-877ER (fasudil hydrochloride) is a specific ROCK even reverse the development of PH induced by chronic
inhibitor with an extended-release formulation. It has been hypoxia or monocrotaline in animal models [62–67].
used in a double-blind, placebo-controlled clinical trial to Several clinical studies have suggested that SSRIs may be
treat PH in Japan. Although there was no significant dif- associated with a decreased development of PH and mor-
ference observed in 6MWD between the AT-877ER group tality in PH patients [68, 69]. However, one phase II
and placebo group, the mid-term results showed improved clinical trial failed to show the beneficial effects of SSRIs
pulmonary hemodynamics and cardiac index in the AT- (paroxetine, sertraline, or fluoxetine) in the treatment of
877ER–treated group [38, 39]. PH, while a separate trial showed that the use of SSRIs
correlated to even higher mortality and a greater risk of
2.3 Vasoactive Intestinal Peptide clinical worsening [70, 71]. Because these trials may have
failed to appropriately match all clinical variables of the
Vasoactive intestinal peptide (VIP) is a neuropeptide hor- treated and control patient populations, further studies are
mone that induces pulmonary vasodilation, inhibits vascular necessary to conclusively determine whether SSRIs are an
SMC proliferation and platelet aggregation, and shows anti- appropriate therapeutic category for PH.
inflammatory properties [40–46]. Mediated by G-protein
coupled receptors (VPAC1, VPAC2, and PAC1), VIP can 2.5 Endothelial Progenitor Cells
stimulate the adenylate cyclase signaling pathway, activate
phospholipase-D, tyrosine kinases, and RhoA-GTPAses, Since PH is associated with loss of or damage to the pul-
and increase intracellular calcium via calcium channel ac- monary vascular endothelium, it has been theorized that the
tivation. Downregulation of VIP has been proposed as one of delivery of additional endothelial progenitor cells (EPCs)
the defining features of PH on the basis of the observation of may provide a therapeutic benefit. The main sources of
pulmonary vascular remodeling and hypertension in VIP- EPCs are bone marrow and peripheral blood [72–78]. EPC
deficient mice and a reversal of the pathology with VIP re- population is most commonly characterized through ex-
placement therapy [47]. Lungs from VIP knockout mice pression of CD34, CD133, and kinase domain receptor
showed increased expression of gene transcripts associated (KDR) [79–81]. Some studies have suggested that patients
with vasoconstriction/proliferation and inflammation, and with PH had reduced peripheral EPCs [82–86]; however,
decreased expression of gene transcripts associated with the relationship between the number of peripheral EPCs
vasodilation/anti-proliferation. This gene expression profile, and PH remains controversial [87–90]. EPCs derived from
however, was reversed to that seen in wild-type mice by VIP hypoxia-derived PH mice were found to have altered
treatment [48]. Interestingly, serum and pulmonary levels of functions such as decreased migratory response to stromal
VIP are decreased in PH patients, but the expression levels of cell-derived factor-1a (SDF-1a), adhesion to fibronectin,
VIP receptors are increased in PASMCs isolated from PH and incorporation into the vascular network [88]. It has
patients [49]. A clinical trial of 20 PH patients receiving a been demonstrated that treatment with autologous or um-
single inhalation dose of 100 lg aviptadil, an analog of VIP, bilical cord blood-derived EPC can prevent and reverse PH
showed minimal side effects and yielded an acute and mild, in small and large animal models, with improved hemo-
but statistically significant, decrease of pulmonary arterial dynamics and amelioration in the medial thickness of the
pressure with a significant increase in stroke volume and small pulmonary arteries [78, 90, 91]. Studies in animal
mixed venous oxygen saturation [50]. A total of six patients models of PH have shown that EPCs transfected with genes
228 J. Hu et al.

of adrenomedullin [92] or endothelial nitric oxide synthase inflammation and immunity in the pathogenesis of PH.
(eNOS) [93] caused greater improvement in PH than EPCs Perivascular inflammatory infiltration is a complex inter-
alone. The combination of EPCs and sildenafil therapy also play between T- and B-lymphocytes, macrophages, den-
has synergistic effects when compared with a single dritic cells, and mast cells [106]. It was found that
treatment with EPCs [94]. EPCs may benefit the prevention inflammation precedes vascular remodeling in animal PH
of PH by vascular endothelial repair [95–97] and providing models, which suggests that altered immunity is the cause
protective paracrine effects [98–101]. Several clinical rather than a consequence of vascular disease [107]. In
studies have shown that EPC treatment is safe, feasible, addition, the circulating levels of cytokines and chemoki-
and can improve exercise capacity and hemodynamics [42, nes such as interleukin-1b (IL-1b), IL-6, IL-8, monocyte
44, 76, 77, 102]. There are two main types of EPCs; one is chemoattractant protein 1 (MCP1), fractalkine, CCL5/
colony-forming unit-Hill (CFU-Hill) cells, and the other RANTES, and tumor necrosis factors-a (TNF-a) are in-
one is endothelial colony-forming cells (ECFCs). It has creased. These cytokines may lead to pulmonary vascular
been reported that ECFCs can form functional vessels remodeling by mediating or controlling proliferation, mi-
in vivo, while CFU-Hill cells cannot [103]. There is evi- gration and differentiation of pulmonary vascular cells
dence showing that CFU-Hill cells are hematopoietic cells [108]. In pre-clinical studies, targeting of several immune
which may never become long-term intimal ECs in vivo mediators or processes, such as CD20, IL-1, transforming
[103, 104]. Studies showed that the ECFCs could prevent growth factor-b (TGF-b), nuclear factor of activated T cells
PH in rats treated with bleomycin [105], but no reports (NFAT), tumor necrosis factor–related apoptosis-inducing
have suggested CFU-Hill cells in preventing or improving ligand (TRAIL), 5-lipoxygenase/5-lipoxygenase-activating
PH. While more clinical studies are needed to confirm the protein (5-LO/FLAP), Leukotriene B4 (LTB4), and purine
safety, adverse effects, and efficacy of EPC treatment, it is synthesis, has been shown to be effective in preventing or
a novel approach for the future treatment of PH (Table 1). even reversing PH [108]. Immunosuppressive agents such
as dexamethasone, mycophenolate mofetil, cyclosporine,
2.6 Inflammation and Immunity and etanercept have also been shown to improve PH in
animal models [109]. The highly selective elastase in-
As perivascular inflammation is observed in patients and hibitor elafin has been demonstrated to attenuate fully de-
animals with PH, several studies have assessed the role of veloped PAH in an animal model, with a significant

Table 1 Summary of the action of the drugs


Class Agent Mechanism of action

RTKI Imatinib Inhibition of PDGF, c-kit, and c-Abl receptors


Nilotinib Inhibition of BCR-ABL tyrosine kinase
Sorafenib Inhibition of PDGFR, VEGFR, c-kit, and Raf
Rho-kinase inhibitor AT-877ER (fasudil Inhibition of Rho-kinase
hydrochloride)
VIP Aviptadil Inducing pulmonary vasodilation, inhibiting vascular SMCs proliferation, platelet
aggregation
SSRI Paroxetine, sertraline, Inhibition of serotonin reuptake
fluoxetine
EPCs Repairing pulmonary vascular endothelium
Inflammation and Elafin Inhibition of elastase
immunity Bardoxolone methyl Inducing Nrf2 and suppressing NF-jB activation
Tacrolimus Inhibition of the NFAT family
Rituximab Antibody of CD20 on B cells
CCR5 Antagonist Maraviroc Inhibition of CCR5, protective effect on vascular lesions
Wnt Mesd (blocker of Wnt ligands) Competitively blocking the binding of Wnt ligands to LRP5/6
miRNAs MRX34 Mimic of miRNA tumor suppressor miR-34
Miravirsen Inhibitor of miRNA-122
CCR5 C-C motif chemokine receptor 5, EPC endothelial progenitor cell, LRP5/6 low-density lipoprotein receptor-related protein 5/6, miRNA
microRNA, NFAT nuclear factor of activated T cells, NF-kB nuclear factor kappa-light-chain-enhancer of activated B cells, Nrf2 nuclear factor
erythroid 2–related factor 2, PDGF platelet-derived growth factor, PDGFR PDGF receptor, RTKI receptor tyrosine kinase inhibitor, SMC smooth
muscle cell, SSRI selective serotonin reuptake inhibitor, VEGFR VEGF receptor, VIP vasoactive intestinal peptide
Developing Drugs for Pulmonary Hypertension 229

improvement of the vascular pathology, parameters of hypoxia exposure, and maraviroc treatment markedly at-
pulmonary hemodynamics, and right ventricular function tenuated hypoxia-induced PH development (distal pul-
[110]. In fact, elafin is an FDA-approved orphan drug for monary artery muscularization) in CCR5 knock-in mice
the treatment of PH. Bardoxolone methyl (RTA402) is an [122]. In addition to the preventive effect, maraviroc can
orally available semi-synthetic triterpenoid that induces also partially reverse PH in chronically hypoxic mice
nuclear factor erythroid 2–related factor 2 (Nrf2) and [122]. Although no clinical trials evaluating the effects of
suppresses nuclear factor kappa-light-chain-enhancer of CCR5 inhibitors on PH have been reported, it may have
activated B cells (NF-jB) activation. Bardoxolone methyl therapeutic potential for the treatment of PH.
can suppress activation of proinflammatory mediators,
enhance endothelial nitric oxide (NO) bioavailability, im- 2.8 Wnt Signaling
prove metabolic dysfunction, inhibit vascular proliferation,
and prevent maladaptive remodeling. Bardoxolone methyl Wnt signaling has been linked to more than 20 clinical
also targets multiple cell types relevant to PH, including diseases. Its effects in regulating angiogenesis and cell
ECs, SMCs, and macrophages. A phase II clinical trial is growth have prompted researchers to hypothesize that it
currently underway to evaluate its efficacy as a therapeutic can play a role in the treatment of PH in which none of the
for PH (NCT02036970) [111]. Recently, an inhibitor of the current therapies can promote angiogenesis or reverse
NFAT family, FK506 (Tacrolimus), was reported to reverse medial thickening. Both the Wnt/b-catenin (Wnt/bC)
severe PH in the SUGEN/hypoxia model by restoring bone pathway and Wnt/planar cell polarity (Wnt/PCP) pathway
morphogenetic protein type 2 receptor (BMPR2) signaling have been shown to be involved in the development of PH
[112], and its safety and efficacy are being evaluated in a [123–127]. Wnt7a and Wnt5 downregulate b-catenin and
clinical trial (NCT01647945) [113]. Rituximab is a chimeric have antiproliferative effects on PASMC cultured under
monoclonal antibody recognizing CD20 on B cells and is hypoxic conditions [127, 128]. In the lung tissue of PH
currently in use in a National Institute of Allergy and In- patients, it was found that the expression of PCP mediators
fectious Diseases (NIAID)-sponsored trial (NCT01086540) are upregulated, suggesting a pathophysiological role of the
for the treatment of systemic sclerosis-associated PH [114]. Wnt/PCP pathway [129]. Low-density lipoprotein receptor-
However, no results have been reported yet for the clinical related protein 5/6 (LRP5/6) are Wnt co-receptors that bind
trials utilizing bardoxolone methyl, FK506, or rituximab for to Wnt ligands and mediate canonical Wnt/bC signaling,
the treatment of PH. The effectiveness and safety of nu- while Mesd is a cellular-encoded chaperone protein for
merous immune targets still need to be evaluated, and the LRP5/6 that competitively blocks the binding of Wnt li-
immunotherapies may have to be adjusted on a patient-by- gands to LRP5/6 [130]. Systemic delivery of Mesd protein
patient basis according to different subtypes of PH charac- attenuated PH and pulmonary vascular remodeling in a
terized by distinct inflammatory profiles. rodent model of hyperoxia-induced PH [131]. ICG001, a
novel b-catenin inhibitor, has been shown to increase
2.7 G-Protein-Coupled Receptor CCR5 alveolarization and reduce pulmonary vascular remodeling
and PH in the rodent model of hyperoxia-induced PH.
CCR5 is strongly expressed in principal cell types impli- ICG001 could also decrease PASMC proliferation and the
cated in PH progression, including ECs, SMCs, T cells, and expression of extracellular matrix remodeling molecules
macrophages [115–122]. It is considered a therapeutic under hyperoxic conditions [132]. Gene expression array
target in human immunodeficiency virus (HIV) infection analyses of different cell types from PH patients revealed
because it is a co-receptor for HIV cell entry [120, 122]. that molecular lesions associated with PH were present in
The CCR5 pathway also plays an important role in all cell types, and the Wnt signaling pathway was a com-
atherogenesis, and studies have suggested that inhibition of mon molecular defect in both heritable and idiopathic PH
the CCR5 pathway has a protective effect on vascular le- [133]. However, due to a wide range of cellular functions
sions [117, 122]. Pulmonary expression of CCR5 is in- and molecular targets of Wnt signaling, extensive screen-
creased in mice subjected to hypoxia-induced PH, as well ing of Wnt modulators will be required to find therapeutic
as in the explanted lungs from patients with idiopathic PH. candidates for PH that have minimal adverse effects and
Notably, CCR5 expression is more marked in areas char- toxicity with maximal efficacy.
acterized by greater severity of vascular medial hypertro-
phy. The activation of CCR5 leads to the proliferation of 2.9 MicroRNAs
cultured human PASMCs, while a human CCR5 antagonist
developed as an HIV therapeutic, maraviroc, inhibits this MicroRNAs (miRNAs) are a category of small noncoding
proliferation [115]. It is reported that CCR5-deficient mice RNAs, which regulate an array of proteins that affect many
developed less severe PH than wild-type mice after biological processes including cell differentiation, survival,
230 J. Hu et al.

Fig. 1 Drugs currently in discovery, development, and/or approved for the treatment of pulmonary hypertension. miRNA microRNA

and proliferation [134]. As many miRNAs are associated 3 Summary


with cancer development and have a potential as anti-cancer
agents, and because there are many overlapping pathways PH is a disease with complex and diverse underlying
(e.g., BMPR2, Src/STAT3/Pim1, and hypoxia) in both mechanisms and a poor prognosis. In recent years, sig-
cancer and PH [135, 136], the evaluation of miRNAs in PH nificant progress has been made toward understanding the
treatment remains an attractive idea. Some miRNAs are pathophysiology and biochemistry of PH development,
thought to be modulators of signaling in pathogenic protein which has led to considerable advances in the search for
expression, which leads to pulmonary vascular remodeling, new treatments. Alongside the refinement of existing
PH development, right ventricular (RV) hypertrophy, and treatment strategies, many new molecular and cellular
RV failure [137, 138]. There are two strategies for miRNA- pathways and targets involved in PH have been explored in
based therapies: miRNAs mimetics and miRNA antagonists. the development of new drugs and therapies (Fig. 1).
Mimetics of miR-204, miR-424, miR-503, and let-7f are Although there are new therapies in pre-clinical or early-
reported to ameliorate or reverse PH in animal models [139– stage clinical trials, most of the clinical trials conducted are
141]. While miRNA antagonism may have more side effects short term. More studies need to be conducted to assess the
than miRNA mimetics, several studies have demonstrated safety and efficacy of such novel therapeutics before such
that specific miRNA antagonists, such as anti-miR-17, can promising approaches can be added to our armamentarium
provide a beneficial therapy for PH [142]. Two drugs in this of treatments for PH.
category are being evaluated in clinical trials for other dis-
eases. MRX34, a mimic of miRNA tumor suppressor miR- Acknowledgments We thank Dr. Sergei Snovida for helpful com-
ments on the manuscript and Elfy Chiang for the graphic assistance.
34, is being evaluated for treatment of liver cancer by in-
travenous (IV) delivery [143]. Miravirsen, an inhibitor of Compliance with ethical standards There are no conflicts of in-
miRNA-122, is in clinical trials for the treatment of hepatitis terests to declare for Jian Hu, Qinzi Xu, Charles McTiernan, Yen-
C virus infection, via subcutaneous administration [144]. Chun Lai, and David Osei-Hwedieh. Mark Gladwin receives National
Institutes of Health (NIH) grant funding, and is a consultant for Bayer
Currently, there are no clinical trials for miRNA-based
for sickle cell disease. He is also the co-inventor of an NIH patent for
therapy for PH. However, further investigation may lead to the use of nitrite for cardiovascular indications. He also receives
the development of novel and effective therapeutic royalties as co-author of a textbook of medical students.
strategies. No financial assistance was received for preparing this manuscript.
Developing Drugs for Pulmonary Hypertension 231

References 20. Gomberg-Maitland M, Maitland ML, Barst RJ, et al. A dosing/


cross-development study of the multikinase inhibitor sorafenib
1. Galiè N, Hoeper MM, Humbert M, et al. Guidelines for the in patients with pulmonary arterial hypertension. Clin Pharma-
diagnosis and treatment of pulmonary hypertension. Eur Respir col Ther. 2010;87(3):303–10.
J. 2009;34(6):1219–63. 21. Taçoy G, Çengel A, Özkurt ZN, et al. Dasatinib-induced pul-
2. Pullamsetti SS, Schermuly R, Ghofrani A, et al. Novel and monary hypertension in acute lymphoblastic leukemia: case
emerging therapies for pulmonary hypertension. Am J Respir report. Turk Kardiyol Dern Ars. 2015;43(1):78–81.
Crit Care Med. 2014;189(4):394–400. 22. Hong JH, Lee SE, Choi SY, et al. Reversible pulmonary arterial
3. George MG, Schieb LJ, Ayala C, et al. Pulmonary hypertension hypertension associated with dasatinib for chronic myeloid
surveillance. Chest. 2014;146(2):476–95. leukemia. Cancer Res Treat. 2014;17:1–6.
4. D’Alonzo GE, Barst RJ, Ayres SM, et al. Survival in patients 23. Buchelli Ramirez HL, Álvarez Álvarez CM, Rodrı́guez Reguero
with primary pulmonary hypertension. Results from a national JJ, et al. Reversible pre-capillary pulmonary hypertension due to
prospective registry. Ann Intern Med. 1991;115(5):343–9. dasatinib. Respir Care. 2014;59:e77–80.
5. Schermuly RT, Dony E, Ghofrani HA, et al. Reversal of ex- 24. Ciuclan L, Bonneau O, Hussey M, et al. A novel murine model
perimental pulmonary hypertension by PDGF inhibition. J Clin of severe pulmonary arterial hypertension. Am J Respir Crit
Invest. 2005;115:2811–21. Care Med. 2011;184(10):1171–82.
6. Perros F, Montani D, Dorfmüller P, et al. Platelet-derived growth 25. Robertson TP, Dipp M, Ward JP, et al. Inhibition of sustained
factor expression and function in idiopathic pulmonary arterial hypoxic vasoconstriction by Y-27632 in isolated intrapulmonary
hypertension. Am J Respir Crit Care Med. 2008;178:81–8. arteries and perfused lung of the rat. Br J Pharmacol. 2000;
7. Nakamura K, Akagi S, Ogawa A, et al. Pro-apoptotic effects of 131(1):5–9.
imatinib on PDGF stimulated pulmonary artery smooth muscle 26. Hu J, Tarantelli R, Simon M, et al. Simian immunodeficiency
cells from patients with idiopathic pulmonary arterial hyper- virus induces pulmonary arterial hypertension in rhesus maca-
tension. Int J Cardiol. 2011;159(2):100–6. ques: a hemodynamic study. Circulation. 2013;128(22):A18705.
8. Vantler M, Karikkineth BC, Naito H, et al. PDGF-BB protects 27. Robertson TP, Aaronson PI, Ward JP. Ca2? sensitization during
cardiomyocytes from apoptosis and improves contractile func- sustained hypoxic pulmonary vasoconstriction is endothelium
tion of engineered heart tissue. J Mol Cell Cardiol. 2010;48: dependent. Am J Physiol Lung Cell Mol Physiol. 2003;
1316–23. 284(6):L1121–6.
9. Panky EA, Thammasiboon S, Lasker GF, et al. Imatinib at- 28. Wang Z, Lanner MC, Jin N, et al. Hypoxia inhibits myosin
tenuates monocrotaline pulmonary hypertension and has potent phosphatase in pulmonary arterial smooth muscle cells: role of
vasodilator activity in pulmonary and systemic vascular beds in Rho-kinase. Am J Respir Cell Mol Biol. 2003;29(4):465–71.
the rat. Am J Physiol Heart Circ Physiol. 2013;305:H1288–96. 29. Abe K, Shimokawa H, Morikawa K, et al. Long-term treatment
10. Ghofrani HA, Seeger W, Grimminger F. Imatinib for the treat- with a Rho-kinase inhibitor improves monocrotaline-induced fa-
ment of pulmonary arterial hypertension. N Engl J Med. tal pulmonary hypertension in rats. Circ Res. 2004;94(3):385–93.
2005;353:1412–3. 30. Abe K, Tawara S, Oi K, et al. Long-term inhibition of Rho-
11. Patterson KC, Weissmann A, Ahmadi T, et al. Imatinib mesylate kinase ameliorates hypoxia-induced pulmonary hypertension in
in the treatment of refractory idiopathic pulmonary arterial hy- mice. J Cardiovasc Pharmacol. 2006;48(6):280–5.
pertension. Ann Intern Med. 2006;145:152–3. 31. Li F, Xia W, Li A, et al. Long-term inhibition of Rho kinase
12. Souza R, Sitbon O, Parent G, et al. Long term imatinib treatment with fasudil attenuates high flow induced pulmonary artery re-
in pulmonary arterial hypertension. Thorax. 2006;61:736. modeling in rats. Pharmacol Res. 2007;55(1):64–71.
13. Ten Freyhaus H, Dumitrescu D, Bovenschulte H, et al. Sig- 32. Hisaoka T, Yano M, Ohkusa T, et al. Enhancement of Rho/Rho-
nificant improvement of right ventricular function by imatinib kinase system in regulation of vascular smooth muscle con-
mesylate in scleroderma-associated pulmonary arterial hyper- traction in tachycardia-induced heart failure. Cardiovasc Res.
tension. Clin Res Cardiol. 2009;98:265–7. 2001;49(2):319–29.
14. Tapper EB, Knowles D, Heffron T, et al. Portopulmonary hy- 33. Yamashita K, Kotani Y, Nakajima Y, et al. Fasudil, a Rho ki-
pertension: imatinib is a novel treatment and the Emory expe- nase (ROCK) inhibitor, protects against ischemic neuronal
rience with this condition. Transplant Proc. 2009;41:1969–71. damage in vitro and in vivo by acting directly on neurons. Brain
15. Ghofrani HA, Morrell NW, Hoeper MM, et al. Imatinib in Res. 2007;1154:215–24.
pulmonary arterial hypertension patients with inadequate re- 34. Fukumoto Y, Matoba T, Ito A, et al. Acute vasodilator effects of
sponse to established therapy. Am J Respir Crit Care Med. a Rho-kinase inhibitor, fasudil, in patients with severe pul-
2010;182:1171–7. monary hypertension. Heart. 2005;91(3):391–2.
16. Hoeper MM, Barst RJ, Bourge RC, et al. Imatinib mesylate as 35. Ishikura K, Yamada N, Ito M, et al. Beneficial acute effects of
add-on therapy for pulmonary arterial hypertension: results of Rho-kinase inhibitor in patients with pulmonary arterial hyper-
the randomized IMPRES study. Circulation. 2013;127(10): tension. Circ J. 2006;70(2):174–8.
1128–38. 36. Fujita H, Fukumoto Y, Saji K, et al. Acute vasodilator effects of
17. Maurer B, Reich N, Juengel A, et al. Fra-2 transgenic mice as a inhaled fasudil, a specific Rho-kinase inhibitor, in patients with
novel model of pulmonary hypertension associated with sys- pulmonary arterial hypertension. Heart Vessels. 2010;25(2):144–9.
temic sclerosis. Ann Rheum Dis. 2012;71(8):1382–7. 37. Kojonazarov B, Myrzaakhmatova A, Sooronbaev T, et al. Ef-
18. Efficacy, safety, tolerability and pharmocokinetics (PK) of fects of Fasudil in patients with high-altitude pulmonary hy-
nilotinib (AMN107) in pulmonary arterial hypertension (PAH). pertension. Eur Respir J. 2012;39(2):496–8.
Novartis Pharmaceuticals. https://clinicaltrials.gov/show/ 38. Uehata M, Ishizaki T, Satoh H, et al. Calcium sensitization of
NCT01179737. Accessed April 2014. smooth muscle mediated by a Rho-associated protein kinase in
19. Moreno-Vinasco L, Gomberg-Maitland M, Maitland ML, et al. hypertension. Nature. 1997;389(6654):990–4.
Genomic assessment of a multikinase inhibitor, sorafenib, in a 39. Fukumoto Y, Yamada N, Matsubara H, et al. Double-blind,
rodent model of pulmonary hypertension. Physiol Genomics. placebo-controlled clinical trial with a Rho-kinase inhibitor in
2008;33(2):278–91. pulmonary arterial hypertension. Circ J. 2013;77(10):2619–25.
232 J. Hu et al.

40. Said SI, Mutt V. Polypeptide with broad biological activity: 61. Vane J, Corin RE. Prostacyclin: a vascular mediator. Eur J Vasc
isolation from small intestine. Science. 1970;169:1217–8. Endovasc Surg. 2003;26(6):571–8.
41. Maruno K, Absood A, Said SI. VIP inhibits basal and histamine- 62. Morecroft I, Loughlin L, Nilsen M, et al. Functional interactions
stimulated proliferation of human airway smooth muscle cells. between 5-hydroxytryptamine receptors and the serotonin
Am J Physiol. 1995;268:L1047–51. transporter in pulmonary arteries. J Pharmacol Exp Ther.
42. Saga T, Said SI. Vasoactive intestinal peptide relaxes isolated 2005;313(2):539–48.
strips of human bronchus, pulmonary artery, and lung 63. Guignabert C, Raffestin B, Benferhat R, et al. Serotonin trans-
parenchyma. Trans Assoc Am Physicians. 1984;97:304–10. porter inhibition prevents and reverses monocrotaline-induced
43. Hu J, Sharifi-Sanjani M, Shiva S, et al. Nitrite prevents right pulmonary hypertension in rats. Circulation. 2005;111:2812–9.
ventricular failure and remodeling induced by pulmonary artery 64. Lushaj EB, Hu J, Haworth R, et al. Intravital microscopy to
banding. Circulation. 2013;128(22):A16923. study myocardial engraftment. Interact CardioVasc Thorac
44. Nandiwada PA, Kadowitz PJ, Said SI, et al. Pulmonary va- Surg. 2012;15(1):5–9.
sodilator responses to vasoactive intestinal peptide in the cat. 65. Morecroft I, Pang L, Baranowska M, et al. In vivo effects of a
J Appl Physiol. 1985;58(5):1723–8. combined 5-HT1B receptor/SERT antagonist in experimental
45. Hashimoto H, Negishi M, Ichikawa A. Identification of a pulmonary hypertension. Cardiovasc Res. 2010;85(3):593–603.
prostacyclin receptor coupled to the adenylate cyclase system 66. Marcos E, Adnot S, Pham MH, et al. Serotonin transporter in-
via a stimulatory GTP-binding protein in mouse mastocytoma hibitors protect against hypoxic pulmonary hypertension. Am J
P-815 cells. Prostaglandins. 1990;40(5):491–505. Respir Crit Care Med. 2003;168:487–93.
46. Said SI, Hamidi SA, Dickman KG, et al. Moderate pulmonary 67. Zhu SP, Mao ZF, Huang J, et al. Continuous fluoxetine ad-
arterial hypertension in male mice lacking the vasoactive in- ministration prevents recurrence of pulmonary arterial hyper-
testinal peptide gene. Circulation. 2007;115:1260–8. tension and prolongs survival in rats. Clin Exp Pharmacol
47. Haydar S, Sarti JF, Grisoni ER. Intravenous vasoactive intestinal Physiol. 2009;36:e1–5.
polypeptide lowers pulmonary-to-systemic vascular resistance 68. Shah SJ, Gomberg-Maitland M, Thenappan T, et al. Selective
ratio in a neonatal piglet model of pulmonary arterial hyper- serotonin reuptake inhibitors and the incidence and outcome of
tension. J Pediatr Surg. 2007;42:758–64. pulmonary hypertension. Chest. 2009;136(3):694–700.
48. Said SI, Hamidi SA. Pharmacogenomics in pulmonary arterial 69. Kawut SM, Horn EM, Berekashvili KK, et al. Selective sero-
hypertension: toward a mechanistic, target-based approach to tonin reuptake inhibitor use and outcomes in pulmonary arterial
therapy. Pulm Circ. 2011;1(3):383–8. hypertension. Pulm Pharmacol Ther. 2006;19(5):370–4.
49. Petkov V, Mosgoeller W, Ziesche R, et al. Vasoactive intestinal 70. Dhalla IA, Juurlink DN, Gomes T, et al. Selective serotonin
peptide as a new drug for treatment of primary pulmonary hy- reuptake inhibitors and pulmonary arterial hypertension: a case–
pertension. J Clin Invest. 2003;111:1339–46. control study. Chest. 2012;141(2):348–53.
50. Leuchte HH, Baezner C, Baumgartner RA, et al. Inhalation of 71. Sadoughi A, Roberts KE, Preston IR, et al. Use of selective
vasoactive intestinal peptide in pulmonary hypertension. Eur serotonin reuptake inhibitors and outcomes in pulmonary arterial
Respir J. 2008;32(5):1289–94. hypertension. Chest. 2013;144(2):531–41.
51. Hamidi SA, Lin RZ, Szema AM, et al. VIP and endothelin receptor 72. Lin RZ, Dreyzin A, Aamodt K, et al. Functional endothelial
antagonist: an effective combination against experimental pul- progenitor cells from cryopreserved umbilical cord blood. Cell
monary arterial hypertension. Respir Res. 2011;12:141. Transplant. 2011;20(4):515–22.
52. Dempsie Y, MacLean MR. Pulmonary hypertension: therapeutic 73. Ormiston ML, Deng Y, Stewart DJ, et al. Innate immunity in the
targets within the serotonin system. Br J Pharmacol. 2008; therapeutic actions of endothelial progenitor cells in pulmonary
155:455–62. hypertension. Am J Respir Cell Mol Biol. 2010;43(5):546–54.
53. Eddahibi S, Adnot S. The serotonin pathway in pulmonary hy- 74. Vanneaux V, El-Ayoubi F, Delmau C, et al. In vitro and in vivo
pertension. Arch Mal Coeur Vaiss. 2006;99:621–5. analysis of endothelial progenitor cells from cryopreserved
54. Eddahibi S, Raffestin B, Hamon M, et al. Is the serotonin umbilical cord blood: are we ready for clinical application? Cell
transporter involved in the pathogenesis of pulmonary hyper- Transplant. 2010;19(9):1143–55.
tension? J Lab Clin Med. 2002;139:194–201. 75. Lozonschi L, Bombien R, Osaki S, et al. Transapical mitral
55. Eddahibi S, Raffestin B, Pham I, et al. Treatment with 5-HT po- valved stent implantation: a survival series in swine. J Thorac
tentiates development of pulmonary hypertension in chronically Cardiovasc Surg. 2010;140(2):422–6.
hypoxic rats. Am J Physiol. 1997;272(3 Pt 2):H1173–81. 76. Wang XX, Zhang FR, Shang YP, et al. Transplantation of au-
56. Guibert C, Marthan R, Savineau JP. 5-HT induces an arachidonic tologous endothelial progenitor cells may be beneficial in pa-
acid-sensitive calcium influx in rat small intrapulmonary artery. tients with idiopathic pulmonary arterial hypertension: a pilot
Am J Physiol Lung Cell Mol Physiol. 2004;286(6):L1228–36. randomized controlled trial. J Am Coll Cardiol. 2007;49(14):
57. Liu JQ, Folz RJ. Extracellular superoxide enhances 5-HT-in- 1566–71.
duced murine pulmonary artery vasoconstriction. Am J Physiol 77. Yip HK, Chang LT, Sun CK, et al. Autologous transplantation
Lung Cell Mol Physiol. 2004;287:L111–8. of bone marrow-derived endothelial progenitor cells attenuates
58. Deuchar GA, Hicks MN, MacLean MR. The role of 5-hydrox- monocrotaline-induced pulmonary arterial hypertension in rats.
ytryptamine in the control of pulmonary vascular tone in a rabbit Crit Care Med. 2008;36(3):873–80.
model of pulmonary hypertension secondary to left ventricular 78. Zhao YD, Courtman DW, Deng Y, et al. Rescue of monocro-
dysfunction. Pulm Pharmacol Ther. 2005;18(1):23–31. taline-induced pulmonary arterial hypertension using bone
59. Liu Y, Tian H, Yan X, et al. Serotonin inhibits apoptosis of marrow derived endothelial-like progenitor cells: efficacy of
pulmonary artery smooth muscle cells through 5-HT2A receptors combined cell and eNOS gene therapy in established disease.
involved in the pulmonary artery remodeling of pulmonary Circ Res. 2005;96(4):442–50.
artery hypertension. Exp Lung Res. 2013;39(2):70–9. 79. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative
60. Liu DB, Hu J, Zhang MK, et al. Low-dose ketamine combined progenitor endothelial cells for angiogenesis. Science. 1997;
with pentobarbital in a miniature porcine model for a car- 275(5302):964–7.
diopulmonary bypass procedure: a randomized controlled study. 80. Peichev M, Naiyer AJ, Pereira D, et al. Expression of VEGFR-2
Eur J Anaesthesiol. 2009;26(5):389–95. and AC133 by circulating human CD34(?) cells identifies a
Developing Drugs for Pulmonary Hypertension 233

population of functional endothelial precursors. Blood. cells and cardiac resident progenitor cells. J Mol Cell Cardiol.
2000;95(3):952–8. 2005;39(5):733–42.
81. Urbich C, Dimmeler S. Endothelial progenitor cells: charac- 102. Zhu JH, Wang XX, Zhang FR, et al. Safety and efficacy of
terization and role in vascular biology. Circ Res. 2004; autologous endothelial progenitor cells transplantation in chil-
95(4):343–53. dren with idiopathic pulmonary arterial hypertension: open-label
82. Ding DC, Shyu WC, Lin SZ, et al. The role of endothelial pilot study. Pediatr Transplant. 2008;12(6):650–5.
progenitor cells in ischemic cerebral and heart diseases. Cell 103. Yoder MC, Mead LE, Prater D, et al. Redefining endothelial
Transplant. 2007;16(3):273–84. progenitor cells via clonal analysis and hematopoietic stem/
83. Marsboom G, Janssens S. Endothelial progenitor cells: new progenitor cell principals. Blood. 2007;109:1801–9.
perspectives and applications in cardiovascular therapies. Expert 104. Hirschi KK, Ingram DA, Yoder MC. Assessing identity, phe-
Rev Cardiovasc Ther. 2008;6(5):687–701. notype, and fate of endothelial progenitor cells. Arterioscler
84. Diller GP, van Eijl S, Okonko DO, et al. Circulating endothelial Thromb Vasc Biol. 2008;28:1584–95.
progenitor cells in patients with Eisenmenger syndrome and 105. Baker CD, Seedorf GJ, Wisniewski BL, et al. Endothelial col-
idiopathic pulmonary arterial hypertension. Circulation. ony-forming cell conditioned media promote angiogenesis
2008;117(23):3020–30. in vitro and prevent pulmonary hypertension in experimental
85. Fadini GP, Schiavon M, Rea F, et al. Depletion of endothelial bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol
progenitor cells may link pulmonary fibrosis and pulmonary Physiol. 2013;305(1):L73–81.
hypertension. Am J Respir Crit Care Med. 2007;176(7):724–5. 106. Stacher E, Graham BB, Hunt JM, et al. Modern age pathology of
86. Junhui Z, Xingxiang W, Guosheng F, et al. Reduced number and pulmonary arterial hypertension. Am J Respir Crit Care Med.
activity of circulating endothelial progenitor cells in patients 2012;186:261–72.
with idiopathic pulmonary arterial hypertension. Respir Med. 107. Tamosiuniene R, Tian W, Dhillon G, et al. Regulatory T cells
2008;102(7):1073–9. limit vascular endothelial injury and prevent pulmonary hyper-
87. Asosingh K, Aldred MA, Vasanji A, et al. Circulating angio- tension. Circ Res. 2011;109:867–79.
genic precursors in idiopathic pulmonary arterial hypertension. 108. Rabinovitch M, Guignabert C, Humbert M, et al. Inflammation
Am J Pathol. 2008;172(3):615–27. and immunity in the pathogenesis of pulmonary arterial hyper-
88. Marsboom G, Pokreisz P, Gheysens O, et al. Sustained endothelial tension. Circ Res. 2014;115:165–75.
progenitor cell dysfunction after chronic hypoxia-induced pul- 109. Meloche J, Renard S, Provencher S, et al. Anti-inflammatory
monary hypertension. Stem Cells. 2008;26(4):1017–26. and immunosuppressive agents in PAH. Handb Exp Pharmacol.
89. Toshner M, Voswinckel R, Southwood M, et al. Evidence of 2013;218:437–76.
dysfunction of endothelial progenitors in pulmonary arterial 110. Kim YM, Haghighat L, Spiekerkoetter E, et al. Neutrophil elastase
hypertension. Am J Respir Crit Care Med. 2009;180(8):780–7. is produced by pulmonary artery smooth muscle cells and is linked
90. Takahashi M, Nakamura T, Toba T. Transplantation of en- to neointimal lesions. Am J Pathol. 2011;179(3):1560–72.
dothelial progenitor cells into the lung to alleviate pulmonary 111. Bardoxolone methyl evaluation in patients with pulmonary ar-
hypertension in dogs. Tissue Eng. 2004;10(5–6):771–9. terial hypertension (PAH)—LARIAT. Reata Pharmaceuticals.
91. Xia L, Fu GS, Yang JX, et al. Endothelial progenitor cells may http://clinicaltrials.gov/ct2/show/NCT02036970. Accessed May
inhibit apoptosis of pulmonary microvascular endothelial cells: 2015.
new insights into cell therapy for pulmonary arterial hyperten- 112. Spiekerkoetter E, Tian X, Cai J, et al. FK506 activates BMPR2,
sion. Cytotherapy. 2009;11(4):492–502. rescues endothelial dysfunction, and reverses pulmonary hy-
92. Nagaya N, Kangawa K, Kanda M, et al. Hybrid cell-gene therapy pertension. J Clin Invest. 2013;123:3600–13.
for pulmonary hypertension based on phagocytosing action of 113. Spiekerkoetter E, Zamanian R. FK506 (Tacrolimus) in pul-
endothelial progenitor cells. Circulation. 2003;108:889–95. monary arterial hypertension (TransformPAH). http://
93. Wei L, Zhu W, Xia L, et al. Therapeutic effect of eNOS- clinicaltrials.gov/ct2/show/NCT01647945. Accessed May 2015.
transfected endothelial progenitor cells on hemodynamic pul- 114. Nicolls M, Badesch DB, Medsger TA. Rituximab for treatment
monary arterial hypertension. Hypertens Res. 2013;36:414–21. of systemic sclerosis-associated pulmonary arterial hypertension
94. Diller GP, van Eijl S, Okonko DO, et al. Circulating endothelial (SSc-PAH). http://clinicaltrials.gov/ct2/show/NCT01086540.
progenitor cells in patients with Eisenmenger syndrome and Accessed May 2015.
idiopathic pulmonary arterial hypertension. Circulation. 115. Amsellem V, Lipskaia L, Abid S, et al. CCR5 as a treatment
2008;117:3020–30. target in pulmonary arterial hypertension. Circulation.
95. Hill JM, Zalos G, Halcox JP, et al. Circulating endothelial 2014;130(11):880–91.
progenitor cells, vascular function, and cardiovascular risk. 116. Lutter G, Quaden R, Osaki S, et al. Off-pump transapical mi-
N Engl J Med. 2003;348(7):593–600. tral valve replacement. Eur J Cardiothorac Surg. 2009;36(1):
96. Asahara T, Takahashi T, Masuda H, et al. VEGF contributes to 124–8.
postnatal neovascularization by mobilizing bone marrow-derived 117. Charo IF, Ransohoff RM. The many roles of chemokines and
endothelial progenitor cells. EMBO J. 1999;18(14):3964–72. chemokine receptors in inflammation. N Engl J Med.
97. Werner N, Junk S, Laufs U, et al. Intravenous transfusion of 2006;354:610–21.
endothelial progenitor cells reduces neointima formation after 118. Schecter AD, Calderon TM, Berman AB, et al. Human vascular
vascular injury. Circ Res. 2003;93(2):e17–24. smooth muscle cells possess functional CCR5. J Biol Chem.
98. Rehman J. Feeling the elephant of cardiovascular cell therapy. 2000;275:5466–71.
Circulation. 2010;121(2):197–9. 119. Ishida Y, Kimura A, Kuninaka Y, et al. Pivotal role of the CCL5/
99. Gnecchi M, Zhang Z, Ni A, et al. Paracrine mechanisms in adult CCR5 interaction for recruitment of endothelial progenitor cells in
stem cell signaling and therapy. Circ Res. 2008;103(11):1204–19. mouse wound healing. J Clin Invest. 2012;122:711–21.
100. Santhanam AV, Smith LA, He T, et al. Endothelial progenitor cells 120. Dragic T, Litwin V, Allaway GP, et al. HIV-1 entry into CD4?
stimulate cerebrovascular production of prostacyclin by paracrine cells is mediated by the chemokine receptor CC-CKR-5. Nature.
activation of cyclooxygenase-2. Circ Res. 2007;100(9):1379–88. 1996;381:667–73.
101. Urbich C, Aicher A, Heeschen C, et al. Soluble factors released 121. Deng H, Liu R, Ellmeier W, et al. Identification of a major co-
by endothelial progenitor cells promote migration of endothelial receptor for primary isolates of HIV-1. Nature. 1996;381:661–6.
234 J. Hu et al.

122. Combadiere C, Potteaux S, Rodero M, et al. Combined inhibi- 133. West JD, Austin ED, Gaskill C, et al. Identification of a common
tion of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Wnt-associated genetic signature across multiple cell types in
Ly6C(lo) monocytosis and almost abolishes atherosclerosis in pulmonary arterial hypertension. Am J Physiol Cell Physiol.
hypercholesterolemic mice. Circulation. 2008;117:1649–57. 2014;307:C415–30.
123. Papkoff J, Rubinfeld B, Schryver B, et al. Wnt-1 regulates free 134. Farh KK, Grimson A, Jan C, et al. The widespread impact of
pools of catenins and stabilizes APC-catenin complexes. Mol mammalian microRNAs on mRNA repression and evolution.
Cell Biol. 1996;16:2128–34. Science. 2005;310:1817–21.
124. Shulman JM, Perrimon N, Axelrod JD, et al. Frizzled signaling 135. Voelkel NF, Cool C, Lee SD, et al. Primary pulmonary hyper-
and the developmental control of cell polarity. Trends Genet. tension between inflammation and cancer. Chest. 1998;114(3
1998;14:452–8. Suppl.):225S–30S.
125. Veeman MT, Axelrod JD, Moon RT. A second canon. Functions 136. Paulin R, Courboulin A, Barrier M, et al. From oncopro-
and mechanisms of beta-catenin-independent Wnt signaling. teins/tumor suppressors to microRNAs, the newest therapeutic
Dev Cell. 2003;5:367–77. targets for pulmonary arterial hypertension. J Mol Med.
126. Laumanns IP, Fink L, Wilhelm J, et al. The noncanonical WNT 2011;89:1089–101.
pathway is operative in idiopathic pulmonary arterial hyper- 137. Drake JI, Bogaard HJ, Mizuno S, et al. Molecular signature of a
tension. Am J Respir Cell Mol Biol. 2009;40:683–91. right heart failure program in chronic severe pulmonary hyper-
127. de Jesus Perez VA, Alastalo TP, Wu JC, et al. Bone morpho- tension. Am J Respir Cell Mol Biol. 2011;45:1239–47.
genetic protein 2 induces pulmonary angiogenesis via Wnt-beta- 138. McDonald RA, Hata A, MacLean MR, et al. MicroRNA and
catenin and Wnt-RhoA-Rac1 pathways. J Cell Biol. vascular remodelling in acute vascular injury and pulmonary
2009;184:83–99. vascular remodeling. Cardiovasc Res. 2012;93:594–604.
128. Yu XM, Wang L, JF Li, et al. Wnt5a inhibits hypoxia-induced 139. Courboulin A, Paulin R, Giguère NJ, et al. Role for miR-204 in
pulmonary arterial smooth muscle cell proliferation by down- human pulmonary arterial hypertension. J Exp Med.
regulation of b-catenin. Am J Physiol Lung Cell Mol Physiol. 2011;208:535–48.
2013;304:L103–11. 140. Kim J, Kang Y, Kojima Y, et al. An endothelial apelin-FGF link
129. Laumanns IP, Fink L, Wilhelm J, et al. The noncanonical WNT mediated by miR-424 and miR-503 is disrupted in pulmonary
pathway is operative in idiopathic pulmonary arterial hyper- arterial hypertension. Nat Med. 2013;19:74–82.
tension. Am J Respir Cell Mol Biol. 2009;40(6):683–91. 141. Caruso P, MacLean MR, Khanin R, et al. Dynamic changes in
130. Lin C, Lu W, Zhai L, et al. Mesd is a general inhibitor of lung microRNA profiles during the development of pulmonary
different Wnt ligands in Wnt/LRP signaling and inhibits PC-3 hypertension due to chronic hypoxia and monocrotaline. Arte-
tumor growth in vivo. FEBS Lett. 2011;585(19):3120–5. rioscler Thromb Vasc Biol. 2010;30:716–23.
131. Alapati D, Rong M, Chen S, et al. Inhibition of LRP5/6-medi- 142. Pullamsetti SS, Doebele C, Fischer A, et al. Inhibition of microRNA-
ated Wnt/b-catenin signaling by Mesd attenuates hyperoxia-in- 17 improves lung and heart function in experimental pulmonary
duced pulmonary hypertension in neonatal rats. Pediatr Res. hypertension. Am J Respir Crit Care Med. 2012;185:409–19.
2013;73(6):719–25. 143. Bouchie A. First microRNA mimic enters clinic. Nat Biotech-
132. Alapati D, Rong M, Chen S, et al. Inhibition of b-catenin sig- nol. 2013;31:577.
naling improves alveolarization and reduces pulmonary hyper- 144. Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV
tension in experimental bronchopulmonary dysplasia. Am J infection by targeting microRNA. N Engl J Med.
Respir Cell Mol Biol. 2014;51:104–13. 2013;368:1685–94.

You might also like