Am J Cardiovasc Drugs 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



Ó Springer International Publishing Switzerland 2015

Abstract Biomedical advances over the last decade have identified the central role of proliferative pulmonary arterial smooth muscle cells (PASMCs) in the development of pulmonary hypertension (PH). Furthermore, promoters of proliferation and apoptosis resistance in PASMCs and endothelial cells, such as aberrant signal pathways involving growth factors, G protein-coupled receptors, kinases, and microRNAs, have also been described. As a result of these discoveries, PH is currently divided into subgroups based on the underlying pathology, which allows focused and targeted treatment of the condition. The defining features of PH, which subsequently lead to vascular wall remodeling, are dysregulated proliferation of PASMCs, local inflammation, and apoptosis-resistant endothelial cells. Efforts to assess the relative contributions of these factors have generated several promising targets. This review discusses recent novel targets of therapies for PH that have been developed as a result of these advances, which are now in pre-clinical and clinical trials (e.g., imatinib [phase III]; nilotinib, AT-877ER, rituximab, tacrolimus, paroxetine, sertraline, fluoxetine, bardoxolone methyl [phase II]; and sorafenib, FK506, aviptadil, endothelial progenitor cells (EPCs) [phase I]). While substantial progress has been made in recent years in targeting key molecular pathways, PH still remains without a cure, and these novel therapies provide an important conceptual framework

& Jian Hu [email protected] 1

Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, E1226 BST 200 Lothrop Street, Pittsburgh, PA 15213, USA

2

Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA

of categorizing patients on the basis of molecular phenotype(s) for effective treatment of the disease.

Key Points Several novel strategies have emerged for pulmonary hypertension in recent years. Pre-clinical and clinical trials have been performed to evaluate the novel strategies. The following drugs, acting on novel targets, are currently being evaluated in clinical trials: imatinib (phase III); nilotinib, AT-877ER, rituximab, tacrolimus, paroxetine, sertraline, fluoxetine, bardoxolone methyl (phase II); and sorafenib, FK506, aviptadil, endothelial progenitor cells (EPCs) (phase I).

1 Introduction Pulmonary hypertension (PH) is a progressive vascular disease 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 pulmonary arterial pressure (PAP) (C25 mmHg) and can lead to dyspnea, fatigue, cough, chest pain, palpitations, peripheral edema, syncope, right heart failure, and death [1, 2]. In the US population, the age-standardized mortality rate for PH has significantly increased from 5.5 per 100,000 population in 2001 to 6.5 per 100,000 in 2010. In 2010, women showed significantly higher mortality rates than

J. Hu et al.

men, and non-Hispanic blacks showed significantly higher mortality rates than non-Hispanic whites. Aged population cohorts (75–84 and C85 years of age) show the highest PH mortality (47.9/100,000 and 108.7/100,000, respectively, 2010 data) [3]. Mechanisms for the development of PH are complex and may be idiopathic, genetic, or secondary to left heart disease, lung diseases, and/or chronic thromboembolism. Five subgroups of PH have been defined on the basis of the etiology [1], and current research progress on the novel therapeutic targets is focused on group 1 PH [pulmonary arterial hypertension (PAH)]. PAH is characterized by a progressive increase in pulmonary vascular resistance (PVR) caused by pulmonary artery remodeling and vaso-occlusion. The most important mechanism for PAH development is endothelial dysfunction and/or injury, which leads to imbalanced production of endogenous vasodilators, such as prostacyclin and nitric oxide, and vasoconstrictors, such as endothelin and serotonin. Without treatment, the survival rates of PAH at 1 year, 3, and 5 years are 68, 48, and 34 %, respectively [4]. Contemporary drugs used to treat PAH target three pathways: the cyclic guanosine monophosphate signaling pathway (e.g., sildenafil and tadalafil), prostacyclin signaling pathway (e.g., epoprostenol and iloprost), and endothelin signaling pathway (e.g., bosentan and ambrisentan). More recently, new drugs targeting these major pathways (e.g., riociguat, selexipag, and macitentan) have been shown to be effective in the treatment of PAH and have been approved by US Food and Drug Administration (FDA). Although current treatments appear to relieve symptoms, improve exercise capacity, and prevent hospitalizations, only limited evidence for disease reversal has been observed. In recent years, several novel strategies have emerged for PH treatment, and this article focuses on new and promising therapeutic approaches.

2 Novel Targets of Drug Treatment 2.1 Receptor Tyrosine Kinase Inhibitors Receptor tyrosine kinases (RTKs) are a class of high-affinity cell surface receptors for a variety of growth factors, hormones, and cytokine ligands. Platelet-derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), and c-kit-receptors play pathogenic roles in the development of PH [5, 6], as these RTK ligands increase proliferation and migration of smooth muscle cells (SMCs) and endothelial cells (ECs) in lungs. Several tyrosine kinase inhibitors (TKIs) are currently under development. Imatinib (GleevecÒ) inhibits PDGF, c-kit, and c-Abl receptors. The use of imatinib has been shown to inhibit proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) in rodents and

humans, and to exert pro-apoptotic effects in isolated PASMCs [7, 8]. Imatinib has also been reported to prevent right ventricular and pulmonary vascular remodeling and to improve pulmonary hemodynamics in animal models [5, 9]. After several reported cases of improved exercise capacity and hemodynamics of PH patients given imatinib treatment [10–14], the compound was evaluated for safety, tolerability, and efficacy in 59 PH patients in a doubleblinded, placebo-controlled multicenter, phase II trial [15]. This clinical trial has shown that imatinib decreases PVR and increases cardiac output, but has no significant improvement in the 6-min walk distance (6MWD). However, subgroup analysis revealed that patients with severely improved PVR showed a significant increase in 6MWD and hemodynamics after receiving imatinib. In a phase III trial of 202 patients with severe PH, patients receiving imatinib showed a significant improvement in 6MWD, decreased mean PAP and PVR, and increased cardiac output [16]. Nilotinib is another TKI that shows greater potency and selectivity in inhibition of the BCR-ABL tyrosine kinase than does imatinib. Nilotinib has been demonstrated to prevent angioproliferation in a rodent model of PH with systemic sclerosis [17]. However, a phase II clinical trial conducted in 23 patients was terminated because of serious adverse effects including cardiopulmonary disorders (e.g., pulseless electrical activity, right ventricular dysfunction, acute respiratory failure, and PAH worsening) and hepatobiliary disorders [e.g., cholecystitis and cholelithiasis (NCT01179737)] [18]. Sorafenib is a multikinase inhibitor targeting PDGF receptor (PDGFR), VEGF receptor (VEGFR), c-kit, and Raf. In a mouse PH model, it has been shown to prevent hemodynamic changes and attenuate PH-associated vascular remodeling by abolishing the upregulation of the mitogen-activated protein kinase (MAPK) 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 well tolerated at a 200-mg twice-daily dose, with the most common adverse effects being moderate skin reaction and alopecia. However, there are reports showing that some other agents in the family of TKIs induce PH. For example, treatment of dasatinib in patients with leukemia induced PH [21–23]. Sugen5416, another protein kinase inhibitor, also induces PH in rodents in combination with hypoxia exposure [24]. In summary, although some TKIs offer promising new treatments for PH, more studies are needed to understand how to use them with minimal adverse effects and maximal efficacy. 2.2 Rho-Kinase Inhibitor RhoA is a small monomeric G-protein that belongs to Ras homologous (Rho) family. RhoA and its downstream substrate Rho-kinase (ROCK) play significant roles in the

Developing Drugs for Pulmonary Hypertension

pathogenesis of PH, mediating vascular remodeling and vasoconstriction [25–28]. Long-term inhibition of ROCK with fasudil, a selective ROCK inhibitor, improved pathological parameters of PH and pulmonary artery remodeling in monocrotaline- or hypoxia-induced rodent models of PH [29–31]. Fasudil was the first ROCK inhibitor approved in 1995 for the treatment of subarachnoid hemorrhage–induced brain vessel vasospasms [32, 33]. Clinical trials have shown that intravenous administration of fasudil has acute beneficial vasodilatory effects on the pulmonary circulation in PH patients [34, 35]. Inhalation of fasudil was shown to be as effective as nitric oxide inhalation in PH patients [36]. It was also reported that fasudil reduced pulmonary artery pressure in patients with high-altitude PH in a randomized, double-blind study [37]. AT-877ER (fasudil hydrochloride) is a specific ROCK inhibitor with an extended-release formulation. It has been used in a double-blind, placebo-controlled clinical trial to treat PH in Japan. Although there was no significant difference observed in 6MWD between the AT-877ER group and placebo group, the mid-term results showed improved pulmonary hemodynamics and cardiac index in the AT877ER–treated group [38, 39]. 2.3 Vasoactive Intestinal Peptide Vasoactive intestinal peptide (VIP) is a neuropeptide hormone that induces pulmonary vasodilation, inhibits vascular SMC proliferation and platelet aggregation, and shows antiinflammatory properties [40–46]. Mediated by G-protein coupled receptors (VPAC1, VPAC2, and PAC1), VIP can stimulate the adenylate cyclase signaling pathway, activate phospholipase-D, tyrosine kinases, and RhoA-GTPAses, and increase intracellular calcium via calcium channel activation. Downregulation of VIP has been proposed as one of the defining features of PH on the basis of the observation of pulmonary vascular remodeling and hypertension in VIPdeficient mice and a reversal of the pathology with VIP replacement therapy [47]. Lungs from VIP knockout mice showed increased expression of gene transcripts associated with vasoconstriction/proliferation and inflammation, and decreased expression of gene transcripts associated with vasodilation/anti-proliferation. This gene expression profile, however, was reversed to that seen in wild-type mice by VIP treatment [48]. Interestingly, serum and pulmonary levels of VIP are decreased in PH patients, but the expression levels of VIP receptors are increased in PASMCs isolated from PH patients [49]. A clinical trial of 20 PH patients receiving a single inhalation dose of 100 lg aviptadil, an analog of VIP, showed minimal side effects and yielded an acute and mild, but statistically significant, decrease of pulmonary arterial pressure with a significant increase in stroke volume and mixed venous oxygen saturation [50]. A total of six patients

in this trial have been reported to have a reduction in PVR of [20 %. Additionally, a combination of VIP and endothelin receptor antagonist, bosentan, has been tested in pre-clinical PH models. This combination therapy was more effective than either drug alone, perhaps because of the synergistic effects of both drugs on suppression of the endothelin-endothelin (ET-ET) receptor pathway [51]. 2.4 Selective Serotonin Reuptake Inhibitors Upregulation of serotonin released from the pulmonary endothelium is associated with the development of PH in animal models, likely through its effects on pulmonary vasoconstriction and SMC proliferation [52–61]. Selective serotonin reuptake inhibitors (SSRIs) can attenuate and even reverse the development of PH induced by chronic hypoxia or monocrotaline in animal models [62–67]. Several clinical studies have suggested that SSRIs may be associated with a decreased development of PH and mortality in PH patients [68, 69]. However, one phase II clinical trial failed to show the beneficial effects of SSRIs (paroxetine, sertraline, or fluoxetine) in the treatment of PH, while a separate trial showed that the use of SSRIs correlated to even higher mortality and a greater risk of clinical worsening [70, 71]. Because these trials may have failed to appropriately match all clinical variables of the treated and control patient populations, further studies are necessary to conclusively determine whether SSRIs are an appropriate therapeutic category for PH. 2.5 Endothelial Progenitor Cells Since PH is associated with loss of or damage to the pulmonary vascular endothelium, it has been theorized that the delivery of additional endothelial progenitor cells (EPCs) may provide a therapeutic benefit. The main sources of EPCs are bone marrow and peripheral blood [72–78]. EPC population is most commonly characterized through expression of CD34, CD133, and kinase domain receptor (KDR) [79–81]. Some studies have suggested that patients with PH had reduced peripheral EPCs [82–86]; however, the relationship between the number of peripheral EPCs and PH remains controversial [87–90]. EPCs derived from hypoxia-derived PH mice were found to have altered functions such as decreased migratory response to stromal cell-derived factor-1a (SDF-1a), adhesion to fibronectin, and incorporation into the vascular network [88]. It has been demonstrated that treatment with autologous or umbilical cord blood-derived EPC can prevent and reverse PH in small and large animal models, with improved hemodynamics and amelioration in the medial thickness of the small pulmonary arteries [78, 90, 91]. Studies in animal models of PH have shown that EPCs transfected with genes

J. Hu et al.

of adrenomedullin [92] or endothelial nitric oxide synthase (eNOS) [93] caused greater improvement in PH than EPCs alone. The combination of EPCs and sildenafil therapy also has synergistic effects when compared with a single treatment with EPCs [94]. EPCs may benefit the prevention of PH by vascular endothelial repair [95–97] and providing protective paracrine effects [98–101]. Several clinical studies have shown that EPC treatment is safe, feasible, and can improve exercise capacity and hemodynamics [42, 44, 76, 77, 102]. There are two main types of EPCs; one is colony-forming unit-Hill (CFU-Hill) cells, and the other one is endothelial colony-forming cells (ECFCs). It has been reported that ECFCs can form functional vessels in vivo, while CFU-Hill cells cannot [103]. There is evidence showing that CFU-Hill cells are hematopoietic cells which may never become long-term intimal ECs in vivo [103, 104]. Studies showed that the ECFCs could prevent PH in rats treated with bleomycin [105], but no reports have suggested CFU-Hill cells in preventing or improving PH. While more clinical studies are needed to confirm the safety, adverse effects, and efficacy of EPC treatment, it is a novel approach for the future treatment of PH (Table 1). 2.6 Inflammation and Immunity As perivascular inflammation is observed in patients and animals with PH, several studies have assessed the role of

inflammation and immunity in the pathogenesis of PH. Perivascular inflammatory infiltration is a complex interplay between T- and B-lymphocytes, macrophages, dendritic cells, and mast cells [106]. It was found that inflammation precedes vascular remodeling in animal PH models, which suggests that altered immunity is the cause rather than a consequence of vascular disease [107]. In addition, the circulating levels of cytokines and chemokines such as interleukin-1b (IL-1b), IL-6, IL-8, monocyte chemoattractant protein 1 (MCP1), fractalkine, CCL5/ RANTES, and tumor necrosis factors-a (TNF-a) are increased. These cytokines may lead to pulmonary vascular remodeling by mediating or controlling proliferation, migration and differentiation of pulmonary vascular cells [108]. In pre-clinical studies, targeting of several immune mediators or processes, such as CD20, IL-1, transforming growth factor-b (TGF-b), nuclear factor of activated T cells (NFAT), tumor necrosis factor–related apoptosis-inducing ligand (TRAIL), 5-lipoxygenase/5-lipoxygenase-activating protein (5-LO/FLAP), Leukotriene B4 (LTB4), and purine synthesis, has been shown to be effective in preventing or even reversing PH [108]. Immunosuppressive agents such as dexamethasone, mycophenolate mofetil, cyclosporine, and etanercept have also been shown to improve PH in animal models [109]. The highly selective elastase inhibitor elafin has been demonstrated to attenuate fully developed PAH in an animal model, with a significant

Table 1 Summary of the action of the drugs Class RTKI

Agent

Mechanism of action

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 hydrochloride)

Inhibition of Rho-kinase

VIP

Aviptadil

Inducing pulmonary vasodilation, inhibiting vascular SMCs proliferation, platelet aggregation

SSRI

Paroxetine, sertraline, fluoxetine

Inhibition of serotonin reuptake

EPCs Inflammation and immunity

Repairing pulmonary vascular endothelium Elafin

Inhibition of elastase

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 Miravirsen

Mimic of miRNA tumor suppressor miR-34 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

improvement of the vascular pathology, parameters of pulmonary hemodynamics, and right ventricular function [110]. In fact, elafin is an FDA-approved orphan drug for the treatment of PH. Bardoxolone methyl (RTA402) is an orally available semi-synthetic triterpenoid that induces nuclear factor erythroid 2–related factor 2 (Nrf2) and suppresses nuclear factor kappa-light-chain-enhancer of activated B cells (NF-jB) activation. Bardoxolone methyl can suppress activation of proinflammatory mediators, enhance endothelial nitric oxide (NO) bioavailability, improve metabolic dysfunction, inhibit vascular proliferation, and prevent maladaptive remodeling. Bardoxolone methyl also targets multiple cell types relevant to PH, including ECs, SMCs, and macrophages. A phase II clinical trial is currently underway to evaluate its efficacy as a therapeutic for PH (NCT02036970) [111]. Recently, an inhibitor of the NFAT family, FK506 (Tacrolimus), was reported to reverse severe PH in the SUGEN/hypoxia model by restoring bone morphogenetic protein type 2 receptor (BMPR2) signaling [112], and its safety and efficacy are being evaluated in a clinical trial (NCT01647945) [113]. Rituximab is a chimeric monoclonal antibody recognizing CD20 on B cells and is currently in use in a National Institute of Allergy and Infectious Diseases (NIAID)-sponsored trial (NCT01086540) for the treatment of systemic sclerosis-associated PH [114]. However, no results have been reported yet for the clinical trials utilizing bardoxolone methyl, FK506, or rituximab for the treatment of PH. The effectiveness and safety of numerous immune targets still need to be evaluated, and the immunotherapies may have to be adjusted on a patient-bypatient basis according to different subtypes of PH characterized by distinct inflammatory profiles. 2.7 G-Protein-Coupled Receptor CCR5 CCR5 is strongly expressed in principal cell types implicated in PH progression, including ECs, SMCs, T cells, and macrophages [115–122]. It is considered a therapeutic target in human immunodeficiency virus (HIV) infection because it is a co-receptor for HIV cell entry [120, 122]. The CCR5 pathway also plays an important role in atherogenesis, and studies have suggested that inhibition of the CCR5 pathway has a protective effect on vascular lesions [117, 122]. Pulmonary expression of CCR5 is increased in mice subjected to hypoxia-induced PH, as well as in the explanted lungs from patients with idiopathic PH. Notably, CCR5 expression is more marked in areas characterized by greater severity of vascular medial hypertrophy. The activation of CCR5 leads to the proliferation of cultured human PASMCs, while a human CCR5 antagonist developed as an HIV therapeutic, maraviroc, inhibits this proliferation [115]. It is reported that CCR5-deficient mice developed less severe PH than wild-type mice after

hypoxia exposure, and maraviroc treatment markedly attenuated hypoxia-induced PH development (distal pulmonary artery muscularization) in CCR5 knock-in mice [122]. In addition to the preventive effect, maraviroc can also partially reverse PH in chronically hypoxic mice [122]. Although no clinical trials evaluating the effects of CCR5 inhibitors on PH have been reported, it may have therapeutic potential for the treatment of PH. 2.8 Wnt Signaling Wnt signaling has been linked to more than 20 clinical diseases. Its effects in regulating angiogenesis and cell growth have prompted researchers to hypothesize that it can play a role in the treatment of PH in which none of the current therapies can promote angiogenesis or reverse medial thickening. Both the Wnt/b-catenin (Wnt/bC) pathway and Wnt/planar cell polarity (Wnt/PCP) pathway have been shown to be involved in the development of PH [123–127]. Wnt7a and Wnt5 downregulate b-catenin and have antiproliferative effects on PASMC cultured under hypoxic conditions [127, 128]. In the lung tissue of PH patients, it was found that the expression of PCP mediators are upregulated, suggesting a pathophysiological role of the Wnt/PCP pathway [129]. Low-density lipoprotein receptorrelated protein 5/6 (LRP5/6) are Wnt co-receptors that bind to Wnt ligands and mediate canonical Wnt/bC signaling, while Mesd is a cellular-encoded chaperone protein for LRP5/6 that competitively blocks the binding of Wnt ligands to LRP5/6 [130]. Systemic delivery of Mesd protein attenuated PH and pulmonary vascular remodeling in a rodent model of hyperoxia-induced PH [131]. ICG001, a novel b-catenin inhibitor, has been shown to increase alveolarization and reduce pulmonary vascular remodeling and PH in the rodent model of hyperoxia-induced PH. ICG001 could also decrease PASMC proliferation and the expression of extracellular matrix remodeling molecules under hyperoxic conditions [132]. Gene expression array analyses of different cell types from PH patients revealed that molecular lesions associated with PH were present in all cell types, and the Wnt signaling pathway was a common molecular defect in both heritable and idiopathic PH [133]. However, due to a wide range of cellular functions and molecular targets of Wnt signaling, extensive screening of Wnt modulators will be required to find therapeutic candidates for PH that have minimal adverse effects and toxicity with maximal efficacy. 2.9 MicroRNAs MicroRNAs (miRNAs) are a category of small noncoding RNAs, which regulate an array of proteins that affect many biological processes including cell differentiation, survival,

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 with cancer development and have a potential as anti-cancer agents, and because there are many overlapping pathways (e.g., BMPR2, Src/STAT3/Pim1, and hypoxia) in both cancer and PH [135, 136], the evaluation of miRNAs in PH treatment remains an attractive idea. Some miRNAs are thought to be modulators of signaling in pathogenic protein expression, which leads to pulmonary vascular remodeling, PH development, right ventricular (RV) hypertrophy, and RV failure [137, 138]. There are two strategies for miRNAbased therapies: miRNAs mimetics and miRNA antagonists. Mimetics of miR-204, miR-424, miR-503, and let-7f are reported to ameliorate or reverse PH in animal models [139– 141]. While miRNA antagonism may have more side effects than miRNA mimetics, several studies have demonstrated that specific miRNA antagonists, such as anti-miR-17, can provide a beneficial therapy for PH [142]. Two drugs in this category are being evaluated in clinical trials for other diseases. MRX34, a mimic of miRNA tumor suppressor miR34, is being evaluated for treatment of liver cancer by intravenous (IV) delivery [143]. Miravirsen, an inhibitor of miRNA-122, is in clinical trials for the treatment of hepatitis C virus infection, via subcutaneous administration [144]. Currently, there are no clinical trials for miRNA-based therapy for PH. However, further investigation may lead to the development of novel and effective therapeutic strategies.

3 Summary PH is a disease with complex and diverse underlying mechanisms and a poor prognosis. In recent years, significant progress has been made toward understanding the pathophysiology and biochemistry of PH development, which has led to considerable advances in the search for new treatments. Alongside the refinement of existing treatment strategies, many new molecular and cellular pathways and targets involved in PH have been explored in the development of new drugs and therapies (Fig. 1). Although there are new therapies in pre-clinical or earlystage clinical trials, most of the clinical trials conducted are short term. More studies need to be conducted to assess the safety and efficacy of such novel therapeutics before such promising approaches can be added to our armamentarium of treatments for PH. Acknowledgments We thank Dr. Sergei Snovida for helpful comments on the manuscript and Elfy Chiang for the graphic assistance. Compliance with ethical standards There are no conflicts of interests to declare for Jian Hu, Qinzi Xu, Charles McTiernan, YenChun Lai, and David Osei-Hwedieh. Mark Gladwin receives National Institutes of Health (NIH) grant funding, and is a consultant for Bayer for sickle cell disease. He is also the co-inventor of an NIH patent for the use of nitrite for cardiovascular indications. He also receives royalties as co-author of a textbook of medical students. No financial assistance was received for preparing this manuscript.

Developing Drugs for Pulmonary Hypertension

References 1. Galie` N, Hoeper MM, Humbert M, et al. Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Respir J. 2009;34(6):1219–63. 2. Pullamsetti SS, Schermuly R, Ghofrani A, et al. Novel and emerging therapies for pulmonary hypertension. Am J Respir Crit Care Med. 2014;189(4):394–400. 3. George MG, Schieb LJ, Ayala C, et al. Pulmonary hypertension surveillance. Chest. 2014;146(2):476–95. 4. D’Alonzo GE, Barst RJ, Ayres SM, et al. Survival in patients with primary pulmonary hypertension. Results from a national prospective registry. Ann Intern Med. 1991;115(5):343–9. 5. Schermuly RT, Dony E, Ghofrani HA, et al. Reversal of experimental pulmonary hypertension by PDGF inhibition. J Clin Invest. 2005;115:2811–21. 6. Perros F, Montani D, Dorfmu¨ller P, et al. Platelet-derived growth factor expression and function in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2008;178:81–8. 7. Nakamura K, Akagi S, Ogawa A, et al. Pro-apoptotic effects of imatinib on PDGF stimulated pulmonary artery smooth muscle cells from patients with idiopathic pulmonary arterial hypertension. Int J Cardiol. 2011;159(2):100–6. 8. Vantler M, Karikkineth BC, Naito H, et al. PDGF-BB protects cardiomyocytes from apoptosis and improves contractile function of engineered heart tissue. J Mol Cell Cardiol. 2010;48: 1316–23. 9. Panky EA, Thammasiboon S, Lasker GF, et al. Imatinib attenuates monocrotaline pulmonary hypertension and has potent vasodilator activity in pulmonary and systemic vascular beds in the rat. Am J Physiol Heart Circ Physiol. 2013;305:H1288–96. 10. Ghofrani HA, Seeger W, Grimminger F. Imatinib for the treatment of pulmonary arterial hypertension. N Engl J Med. 2005;353:1412–3. 11. Patterson KC, Weissmann A, Ahmadi T, et al. Imatinib mesylate in the treatment of refractory idiopathic pulmonary arterial hypertension. Ann Intern Med. 2006;145:152–3. 12. Souza R, Sitbon O, Parent G, et al. Long term imatinib treatment in pulmonary arterial hypertension. Thorax. 2006;61:736. 13. Ten Freyhaus H, Dumitrescu D, Bovenschulte H, et al. Significant improvement of right ventricular function by imatinib mesylate in scleroderma-associated pulmonary arterial hypertension. Clin Res Cardiol. 2009;98:265–7. 14. Tapper EB, Knowles D, Heffron T, et al. Portopulmonary hypertension: imatinib is a novel treatment and the Emory experience with this condition. Transplant Proc. 2009;41:1969–71. 15. Ghofrani HA, Morrell NW, Hoeper MM, et al. Imatinib in pulmonary arterial hypertension patients with inadequate response to established therapy. Am J Respir Crit Care Med. 2010;182:1171–7. 16. Hoeper MM, Barst RJ, Bourge RC, et al. Imatinib mesylate as add-on therapy for pulmonary arterial hypertension: results of the randomized IMPRES study. Circulation. 2013;127(10): 1128–38. 17. Maurer B, Reich N, Juengel A, et al. Fra-2 transgenic mice as a novel model of pulmonary hypertension associated with systemic sclerosis. Ann Rheum Dis. 2012;71(8):1382–7. 18. Efficacy, safety, tolerability and pharmocokinetics (PK) of nilotinib (AMN107) in pulmonary arterial hypertension (PAH). Novartis Pharmaceuticals. https://clinicaltrials.gov/show/ NCT01179737. Accessed April 2014. 19. Moreno-Vinasco L, Gomberg-Maitland M, Maitland ML, et al. Genomic assessment of a multikinase inhibitor, sorafenib, in a rodent model of pulmonary hypertension. Physiol Genomics. 2008;33(2):278–91.

20. Gomberg-Maitland M, Maitland ML, Barst RJ, et al. A dosing/ cross-development study of the multikinase inhibitor sorafenib in patients with pulmonary arterial hypertension. Clin Pharmacol Ther. 2010;87(3):303–10. ¨ zkurt ZN, et al. Dasatinib-induced pul21. Tac¸oy G, C¸engel A, O monary hypertension in acute lymphoblastic leukemia: case report. Turk Kardiyol Dern Ars. 2015;43(1):78–81. 22. Hong JH, Lee SE, Choi SY, et al. Reversible pulmonary arterial hypertension associated with dasatinib for chronic myeloid leukemia. Cancer Res Treat. 2014;17:1–6. ´ lvarez A ´ lvarez CM, Rodrı´guez Reguero 23. Buchelli Ramirez HL, A JJ, et al. Reversible pre-capillary pulmonary hypertension due to dasatinib. Respir Care. 2014;59:e77–80. 24. Ciuclan L, Bonneau O, Hussey M, et al. A novel murine model of severe pulmonary arterial hypertension. Am J Respir Crit Care Med. 2011;184(10):1171–82. 25. Robertson TP, Dipp M, Ward JP, et al. Inhibition of sustained hypoxic vasoconstriction by Y-27632 in isolated intrapulmonary arteries and perfused lung of the rat. Br J Pharmacol. 2000; 131(1):5–9. 26. Hu J, Tarantelli R, Simon M, et al. Simian immunodeficiency virus induces pulmonary arterial hypertension in rhesus macaques: a hemodynamic study. Circulation. 2013;128(22):A18705. 27. Robertson TP, Aaronson PI, Ward JP. Ca2? sensitization during sustained hypoxic pulmonary vasoconstriction is endothelium dependent. Am J Physiol Lung Cell Mol Physiol. 2003; 284(6):L1121–6. 28. Wang Z, Lanner MC, Jin N, et al. Hypoxia inhibits myosin phosphatase in pulmonary arterial smooth muscle cells: role of Rho-kinase. Am J Respir Cell Mol Biol. 2003;29(4):465–71. 29. Abe K, Shimokawa H, Morikawa K, et al. Long-term treatment with a Rho-kinase inhibitor improves monocrotaline-induced fatal pulmonary hypertension in rats. Circ Res. 2004;94(3):385–93. 30. Abe K, Tawara S, Oi K, et al. Long-term inhibition of Rhokinase ameliorates hypoxia-induced pulmonary hypertension in mice. J Cardiovasc Pharmacol. 2006;48(6):280–5. 31. Li F, Xia W, Li A, et al. Long-term inhibition of Rho kinase with fasudil attenuates high flow induced pulmonary artery remodeling in rats. Pharmacol Res. 2007;55(1):64–71. 32. Hisaoka T, Yano M, Ohkusa T, et al. Enhancement of Rho/Rhokinase system in regulation of vascular smooth muscle contraction in tachycardia-induced heart failure. Cardiovasc Res. 2001;49(2):319–29. 33. Yamashita K, Kotani Y, Nakajima Y, et al. Fasudil, a Rho kinase (ROCK) inhibitor, protects against ischemic neuronal damage in vitro and in vivo by acting directly on neurons. Brain Res. 2007;1154:215–24. 34. Fukumoto Y, Matoba T, Ito A, et al. Acute vasodilator effects of a Rho-kinase inhibitor, fasudil, in patients with severe pulmonary hypertension. Heart. 2005;91(3):391–2. 35. Ishikura K, Yamada N, Ito M, et al. Beneficial acute effects of Rho-kinase inhibitor in patients with pulmonary arterial hypertension. Circ J. 2006;70(2):174–8. 36. Fujita H, Fukumoto Y, Saji K, et al. Acute vasodilator effects of inhaled fasudil, a specific Rho-kinase inhibitor, in patients with pulmonary arterial hypertension. Heart Vessels. 2010;25(2):144–9. 37. Kojonazarov B, Myrzaakhmatova A, Sooronbaev T, et al. Effects of Fasudil in patients with high-altitude pulmonary hypertension. Eur Respir J. 2012;39(2):496–8. 38. Uehata M, Ishizaki T, Satoh H, et al. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature. 1997;389(6654):990–4. 39. Fukumoto Y, Yamada N, Matsubara H, et al. Double-blind, placebo-controlled clinical trial with a Rho-kinase inhibitor in pulmonary arterial hypertension. Circ J. 2013;77(10):2619–25.

J. Hu et al. 40. Said SI, Mutt V. Polypeptide with broad biological activity: isolation from small intestine. Science. 1970;169:1217–8. 41. Maruno K, Absood A, Said SI. VIP inhibits basal and histaminestimulated proliferation of human airway smooth muscle cells. Am J Physiol. 1995;268:L1047–51. 42. Saga T, Said SI. Vasoactive intestinal peptide relaxes isolated strips of human bronchus, pulmonary artery, and lung parenchyma. Trans Assoc Am Physicians. 1984;97:304–10. 43. Hu J, Sharifi-Sanjani M, Shiva S, et al. Nitrite prevents right ventricular failure and remodeling induced by pulmonary artery banding. Circulation. 2013;128(22):A16923. 44. Nandiwada PA, Kadowitz PJ, Said SI, et al. Pulmonary vasodilator responses to vasoactive intestinal peptide in the cat. J Appl Physiol. 1985;58(5):1723–8. 45. Hashimoto H, Negishi M, Ichikawa A. Identification of a prostacyclin receptor coupled to the adenylate cyclase system via a stimulatory GTP-binding protein in mouse mastocytoma P-815 cells. Prostaglandins. 1990;40(5):491–505. 46. Said SI, Hamidi SA, Dickman KG, et al. Moderate pulmonary arterial hypertension in male mice lacking the vasoactive intestinal peptide gene. Circulation. 2007;115:1260–8. 47. Haydar S, Sarti JF, Grisoni ER. Intravenous vasoactive intestinal polypeptide lowers pulmonary-to-systemic vascular resistance ratio in a neonatal piglet model of pulmonary arterial hypertension. J Pediatr Surg. 2007;42:758–64. 48. Said SI, Hamidi SA. Pharmacogenomics in pulmonary arterial hypertension: toward a mechanistic, target-based approach to therapy. Pulm Circ. 2011;1(3):383–8. 49. Petkov V, Mosgoeller W, Ziesche R, et al. Vasoactive intestinal peptide as a new drug for treatment of primary pulmonary hypertension. J Clin Invest. 2003;111:1339–46. 50. Leuchte HH, Baezner C, Baumgartner RA, et al. Inhalation of vasoactive intestinal peptide in pulmonary hypertension. Eur Respir J. 2008;32(5):1289–94. 51. Hamidi SA, Lin RZ, Szema AM, et al. VIP and endothelin receptor antagonist: an effective combination against experimental pulmonary arterial hypertension. Respir Res. 2011;12:141. 52. Dempsie Y, MacLean MR. Pulmonary hypertension: therapeutic targets within the serotonin system. Br J Pharmacol. 2008; 155:455–62. 53. Eddahibi S, Adnot S. The serotonin pathway in pulmonary hypertension. Arch Mal Coeur Vaiss. 2006;99:621–5. 54. Eddahibi S, Raffestin B, Hamon M, et al. Is the serotonin transporter involved in the pathogenesis of pulmonary hypertension? J Lab Clin Med. 2002;139:194–201. 55. Eddahibi S, Raffestin B, Pham I, et al. Treatment with 5-HT potentiates development of pulmonary hypertension in chronically hypoxic rats. Am J Physiol. 1997;272(3 Pt 2):H1173–81. 56. Guibert C, Marthan R, Savineau JP. 5-HT induces an arachidonic acid-sensitive calcium influx in rat small intrapulmonary artery. Am J Physiol Lung Cell Mol Physiol. 2004;286(6):L1228–36. 57. Liu JQ, Folz RJ. Extracellular superoxide enhances 5-HT-induced murine pulmonary artery vasoconstriction. Am J Physiol Lung Cell Mol Physiol. 2004;287:L111–8. 58. Deuchar GA, Hicks MN, MacLean MR. The role of 5-hydroxytryptamine in the control of pulmonary vascular tone in a rabbit model of pulmonary hypertension secondary to left ventricular dysfunction. Pulm Pharmacol Ther. 2005;18(1):23–31. 59. Liu Y, Tian H, Yan X, et al. Serotonin inhibits apoptosis of pulmonary artery smooth muscle cells through 5-HT2A receptors involved in the pulmonary artery remodeling of pulmonary artery hypertension. Exp Lung Res. 2013;39(2):70–9. 60. Liu DB, Hu J, Zhang MK, et al. Low-dose ketamine combined with pentobarbital in a miniature porcine model for a cardiopulmonary bypass procedure: a randomized controlled study. Eur J Anaesthesiol. 2009;26(5):389–95.

61. Vane J, Corin RE. Prostacyclin: a vascular mediator. Eur J Vasc Endovasc Surg. 2003;26(6):571–8. 62. Morecroft I, Loughlin L, Nilsen M, et al. Functional interactions between 5-hydroxytryptamine receptors and the serotonin transporter in pulmonary arteries. J Pharmacol Exp Ther. 2005;313(2):539–48. 63. Guignabert C, Raffestin B, Benferhat R, et al. Serotonin transporter inhibition prevents and reverses monocrotaline-induced pulmonary hypertension in rats. Circulation. 2005;111:2812–9. 64. Lushaj EB, Hu J, Haworth R, et al. Intravital microscopy to study myocardial engraftment. Interact CardioVasc Thorac Surg. 2012;15(1):5–9. 65. Morecroft I, Pang L, Baranowska M, et al. In vivo effects of a combined 5-HT1B receptor/SERT antagonist in experimental pulmonary hypertension. Cardiovasc Res. 2010;85(3):593–603. 66. Marcos E, Adnot S, Pham MH, et al. Serotonin transporter inhibitors protect against hypoxic pulmonary hypertension. Am J Respir Crit Care Med. 2003;168:487–93. 67. Zhu SP, Mao ZF, Huang J, et al. Continuous fluoxetine administration prevents recurrence of pulmonary arterial hypertension and prolongs survival in rats. Clin Exp Pharmacol Physiol. 2009;36:e1–5. 68. Shah SJ, Gomberg-Maitland M, Thenappan T, et al. Selective serotonin reuptake inhibitors and the incidence and outcome of pulmonary hypertension. Chest. 2009;136(3):694–700. 69. Kawut SM, Horn EM, Berekashvili KK, et al. Selective serotonin reuptake inhibitor use and outcomes in pulmonary arterial hypertension. Pulm Pharmacol Ther. 2006;19(5):370–4. 70. Dhalla IA, Juurlink DN, Gomes T, et al. Selective serotonin reuptake inhibitors and pulmonary arterial hypertension: a case– control study. Chest. 2012;141(2):348–53. 71. Sadoughi A, Roberts KE, Preston IR, et al. Use of selective serotonin reuptake inhibitors and outcomes in pulmonary arterial hypertension. Chest. 2013;144(2):531–41. 72. Lin RZ, Dreyzin A, Aamodt K, et al. Functional endothelial progenitor cells from cryopreserved umbilical cord blood. Cell Transplant. 2011;20(4):515–22. 73. Ormiston ML, Deng Y, Stewart DJ, et al. Innate immunity in the therapeutic actions of endothelial progenitor cells in pulmonary hypertension. Am J Respir Cell Mol Biol. 2010;43(5):546–54. 74. Vanneaux V, El-Ayoubi F, Delmau C, et al. In vitro and in vivo analysis of endothelial progenitor cells from cryopreserved umbilical cord blood: are we ready for clinical application? Cell Transplant. 2010;19(9):1143–55. 75. Lozonschi L, Bombien R, Osaki S, et al. Transapical mitral valved stent implantation: a survival series in swine. J Thorac Cardiovasc Surg. 2010;140(2):422–6. 76. Wang XX, Zhang FR, Shang YP, et al. Transplantation of autologous endothelial progenitor cells may be beneficial in patients with idiopathic pulmonary arterial hypertension: a pilot randomized controlled trial. J Am Coll Cardiol. 2007;49(14): 1566–71. 77. Yip HK, Chang LT, Sun CK, et al. Autologous transplantation of bone marrow-derived endothelial progenitor cells attenuates monocrotaline-induced pulmonary arterial hypertension in rats. Crit Care Med. 2008;36(3):873–80. 78. Zhao YD, Courtman DW, Deng Y, et al. Rescue of monocrotaline-induced pulmonary arterial hypertension using bone marrow derived endothelial-like progenitor cells: efficacy of combined cell and eNOS gene therapy in established disease. Circ Res. 2005;96(4):442–50. 79. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997; 275(5302):964–7. 80. Peichev M, Naiyer AJ, Pereira D, et al. Expression of VEGFR-2 and AC133 by circulating human CD34(?) cells identifies a

Developing Drugs for Pulmonary Hypertension

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

93.

94.

95.

96.

97.

98. 99. 100.

101.

population of functional endothelial precursors. Blood. 2000;95(3):952–8. Urbich C, Dimmeler S. Endothelial progenitor cells: characterization and role in vascular biology. Circ Res. 2004; 95(4):343–53. Ding DC, Shyu WC, Lin SZ, et al. The role of endothelial progenitor cells in ischemic cerebral and heart diseases. Cell Transplant. 2007;16(3):273–84. Marsboom G, Janssens S. Endothelial progenitor cells: new perspectives and applications in cardiovascular therapies. Expert Rev Cardiovasc Ther. 2008;6(5):687–701. Diller GP, van Eijl S, Okonko DO, et al. Circulating endothelial progenitor cells in patients with Eisenmenger syndrome and idiopathic pulmonary arterial hypertension. Circulation. 2008;117(23):3020–30. Fadini GP, Schiavon M, Rea F, et al. Depletion of endothelial progenitor cells may link pulmonary fibrosis and pulmonary hypertension. Am J Respir Crit Care Med. 2007;176(7):724–5. Junhui Z, Xingxiang W, Guosheng F, et al. Reduced number and activity of circulating endothelial progenitor cells in patients with idiopathic pulmonary arterial hypertension. Respir Med. 2008;102(7):1073–9. Asosingh K, Aldred MA, Vasanji A, et al. Circulating angiogenic precursors in idiopathic pulmonary arterial hypertension. Am J Pathol. 2008;172(3):615–27. Marsboom G, Pokreisz P, Gheysens O, et al. Sustained endothelial progenitor cell dysfunction after chronic hypoxia-induced pulmonary hypertension. Stem Cells. 2008;26(4):1017–26. Toshner M, Voswinckel R, Southwood M, et al. Evidence of dysfunction of endothelial progenitors in pulmonary arterial hypertension. Am J Respir Crit Care Med. 2009;180(8):780–7. Takahashi M, Nakamura T, Toba T. Transplantation of endothelial progenitor cells into the lung to alleviate pulmonary hypertension in dogs. Tissue Eng. 2004;10(5–6):771–9. Xia L, Fu GS, Yang JX, et al. Endothelial progenitor cells may inhibit apoptosis of pulmonary microvascular endothelial cells: new insights into cell therapy for pulmonary arterial hypertension. Cytotherapy. 2009;11(4):492–502. Nagaya N, Kangawa K, Kanda M, et al. Hybrid cell-gene therapy for pulmonary hypertension based on phagocytosing action of endothelial progenitor cells. Circulation. 2003;108:889–95. Wei L, Zhu W, Xia L, et al. Therapeutic effect of eNOStransfected endothelial progenitor cells on hemodynamic pulmonary arterial hypertension. Hypertens Res. 2013;36:414–21. Diller GP, van Eijl S, Okonko DO, et al. Circulating endothelial progenitor cells in patients with Eisenmenger syndrome and idiopathic pulmonary arterial hypertension. Circulation. 2008;117:3020–30. Hill JM, Zalos G, Halcox JP, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003;348(7):593–600. Asahara T, Takahashi T, Masuda H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J. 1999;18(14):3964–72. Werner N, Junk S, Laufs U, et al. Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ Res. 2003;93(2):e17–24. Rehman J. Feeling the elephant of cardiovascular cell therapy. Circulation. 2010;121(2):197–9. Gnecchi M, Zhang Z, Ni A, et al. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103(11):1204–19. Santhanam AV, Smith LA, He T, et al. Endothelial progenitor cells stimulate cerebrovascular production of prostacyclin by paracrine activation of cyclooxygenase-2. Circ Res. 2007;100(9):1379–88. Urbich C, Aicher A, Heeschen C, et al. Soluble factors released by endothelial progenitor cells promote migration of endothelial

102.

103.

104.

105.

106.

107.

108.

109.

110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

cells and cardiac resident progenitor cells. J Mol Cell Cardiol. 2005;39(5):733–42. Zhu JH, Wang XX, Zhang FR, et al. Safety and efficacy of autologous endothelial progenitor cells transplantation in children with idiopathic pulmonary arterial hypertension: open-label pilot study. Pediatr Transplant. 2008;12(6):650–5. Yoder MC, Mead LE, Prater D, et al. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/ progenitor cell principals. Blood. 2007;109:1801–9. Hirschi KK, Ingram DA, Yoder MC. Assessing identity, phenotype, and fate of endothelial progenitor cells. Arterioscler Thromb Vasc Biol. 2008;28:1584–95. Baker CD, Seedorf GJ, Wisniewski BL, et al. Endothelial colony-forming cell conditioned media promote angiogenesis in vitro and prevent pulmonary hypertension in experimental bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2013;305(1):L73–81. Stacher E, Graham BB, Hunt JM, et al. Modern age pathology of pulmonary arterial hypertension. Am J Respir Crit Care Med. 2012;186:261–72. Tamosiuniene R, Tian W, Dhillon G, et al. Regulatory T cells limit vascular endothelial injury and prevent pulmonary hypertension. Circ Res. 2011;109:867–79. Rabinovitch M, Guignabert C, Humbert M, et al. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res. 2014;115:165–75. Meloche J, Renard S, Provencher S, et al. Anti-inflammatory and immunosuppressive agents in PAH. Handb Exp Pharmacol. 2013;218:437–76. Kim YM, Haghighat L, Spiekerkoetter E, et al. Neutrophil elastase is produced by pulmonary artery smooth muscle cells and is linked to neointimal lesions. Am J Pathol. 2011;179(3):1560–72. Bardoxolone methyl evaluation in patients with pulmonary arterial hypertension (PAH)—LARIAT. Reata Pharmaceuticals. http://clinicaltrials.gov/ct2/show/NCT02036970. Accessed May 2015. Spiekerkoetter E, Tian X, Cai J, et al. FK506 activates BMPR2, rescues endothelial dysfunction, and reverses pulmonary hypertension. J Clin Invest. 2013;123:3600–13. Spiekerkoetter E, Zamanian R. FK506 (Tacrolimus) in pulmonary arterial hypertension (TransformPAH). http:// clinicaltrials.gov/ct2/show/NCT01647945. Accessed May 2015. Nicolls M, Badesch DB, Medsger TA. Rituximab for treatment of systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH). http://clinicaltrials.gov/ct2/show/NCT01086540. Accessed May 2015. Amsellem V, Lipskaia L, Abid S, et al. CCR5 as a treatment target in pulmonary arterial hypertension. Circulation. 2014;130(11):880–91. Lutter G, Quaden R, Osaki S, et al. Off-pump transapical mitral valve replacement. Eur J Cardiothorac Surg. 2009;36(1): 124–8. Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med. 2006;354:610–21. Schecter AD, Calderon TM, Berman AB, et al. Human vascular smooth muscle cells possess functional CCR5. J Biol Chem. 2000;275:5466–71. Ishida Y, Kimura A, Kuninaka Y, et al. Pivotal role of the CCL5/ CCR5 interaction for recruitment of endothelial progenitor cells in mouse wound healing. J Clin Invest. 2012;122:711–21. Dragic T, Litwin V, Allaway GP, et al. HIV-1 entry into CD4? cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381:667–73. Deng H, Liu R, Ellmeier W, et al. Identification of a major coreceptor for primary isolates of HIV-1. Nature. 1996;381:661–6.

J. Hu et al. 122. Combadiere C, Potteaux S, Rodero M, et al. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008;117:1649–57. 123. Papkoff J, Rubinfeld B, Schryver B, et al. Wnt-1 regulates free pools of catenins and stabilizes APC-catenin complexes. Mol Cell Biol. 1996;16:2128–34. 124. Shulman JM, Perrimon N, Axelrod JD, et al. Frizzled signaling and the developmental control of cell polarity. Trends Genet. 1998;14:452–8. 125. Veeman MT, Axelrod JD, Moon RT. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev Cell. 2003;5:367–77. 126. Laumanns IP, Fink L, Wilhelm J, et al. The noncanonical WNT pathway is operative in idiopathic pulmonary arterial hypertension. Am J Respir Cell Mol Biol. 2009;40:683–91. 127. de Jesus Perez VA, Alastalo TP, Wu JC, et al. Bone morphogenetic protein 2 induces pulmonary angiogenesis via Wnt-betacatenin and Wnt-RhoA-Rac1 pathways. J Cell Biol. 2009;184:83–99. 128. Yu XM, Wang L, JF Li, et al. Wnt5a inhibits hypoxia-induced pulmonary arterial smooth muscle cell proliferation by downregulation of b-catenin. Am J Physiol Lung Cell Mol Physiol. 2013;304:L103–11. 129. Laumanns IP, Fink L, Wilhelm J, et al. The noncanonical WNT pathway is operative in idiopathic pulmonary arterial hypertension. Am J Respir Cell Mol Biol. 2009;40(6):683–91. 130. Lin C, Lu W, Zhai L, et al. Mesd is a general inhibitor of different Wnt ligands in Wnt/LRP signaling and inhibits PC-3 tumor growth in vivo. FEBS Lett. 2011;585(19):3120–5. 131. Alapati D, Rong M, Chen S, et al. Inhibition of LRP5/6-mediated Wnt/b-catenin signaling by Mesd attenuates hyperoxia-induced pulmonary hypertension in neonatal rats. Pediatr Res. 2013;73(6):719–25. 132. Alapati D, Rong M, Chen S, et al. Inhibition of b-catenin signaling improves alveolarization and reduces pulmonary hypertension in experimental bronchopulmonary dysplasia. Am J Respir Cell Mol Biol. 2014;51:104–13.

133. West JD, Austin ED, Gaskill C, et al. Identification of a common Wnt-associated genetic signature across multiple cell types in pulmonary arterial hypertension. Am J Physiol Cell Physiol. 2014;307:C415–30. 134. Farh KK, Grimson A, Jan C, et al. The widespread impact of mammalian microRNAs on mRNA repression and evolution. Science. 2005;310:1817–21. 135. Voelkel NF, Cool C, Lee SD, et al. Primary pulmonary hypertension between inflammation and cancer. Chest. 1998;114(3 Suppl.):225S–30S. 136. Paulin R, Courboulin A, Barrier M, et al. From oncoproteins/tumor suppressors to microRNAs, the newest therapeutic targets for pulmonary arterial hypertension. J Mol Med. 2011;89:1089–101. 137. Drake JI, Bogaard HJ, Mizuno S, et al. Molecular signature of a right heart failure program in chronic severe pulmonary hypertension. Am J Respir Cell Mol Biol. 2011;45:1239–47. 138. McDonald RA, Hata A, MacLean MR, et al. MicroRNA and vascular remodelling in acute vascular injury and pulmonary vascular remodeling. Cardiovasc Res. 2012;93:594–604. 139. Courboulin A, Paulin R, Gigue`re NJ, et al. Role for miR-204 in human pulmonary arterial hypertension. J Exp Med. 2011;208:535–48. 140. Kim J, Kang Y, Kojima Y, et al. An endothelial apelin-FGF link mediated by miR-424 and miR-503 is disrupted in pulmonary arterial hypertension. Nat Med. 2013;19:74–82. 141. Caruso P, MacLean MR, Khanin R, et al. Dynamic changes in lung microRNA profiles during the development of pulmonary hypertension due to chronic hypoxia and monocrotaline. Arterioscler Thromb Vasc Biol. 2010;30:716–23. 142. Pullamsetti SS, Doebele C, Fischer A, et al. Inhibition of microRNA17 improves lung and heart function in experimental pulmonary hypertension. Am J Respir Crit Care Med. 2012;185:409–19. 143. Bouchie A. First microRNA mimic enters clinic. Nat Biotechnol. 2013;31:577. 144. Janssen HL, Reesink HW, Lawitz EJ, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368:1685–94.

Novel Targets of Drug Treatment for Pulmonary Hypertension.

Biomedical advances over the last decade have identified the central role of proliferative pulmonary arterial smooth muscle cells (PASMCs) in the deve...
742KB Sizes 0 Downloads 7 Views