Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Introduction

Pulmonary hypertension (PH) is a complex condition that encompasses an array of underlying disease processes and affects a diverse population of infants, including those with congenital heart disease, congenital diaphragmatic hernia, persistent PH of the newborn, and those with lung disease such as bronchopulmonary dysplasia. While there are treatments available to adults with PH, limited data exists for infants, especially for the newer medications. Therapies that target the three main pathophysiologic pathways of pulmonary hypertension appear to benefit infants, but which are best for each individual disease process is unclear.

Areas covered

A review of the therapies to treat pulmonary hypertension is covered in this article including the prostacyclin pathway, endothelin pathway, and the nitric oxide pathway. Other adjunctive treatments are also discussed. Findings are based on a PubMed literature search of research papers spanning 1990-2023 and a search of ongoing trials registered with clinicaltrials.gov.

Expert opinion

Overall therapies seem to improve outcomes with most infants with PH. However, given the diverse population of infants with PH, it is imperative to understand the basis for the PH in individual patients and understand which therapies can be applicable. Further research into tailored therapy for the specific populations is warranted.

Free full text 


Logo of nihpaLink to Publisher's site
Expert Opin Pharmacother. Author manuscript; available in PMC 2024 Sep 14.
Published in final edited form as:
PMCID: PMC10843401
NIHMSID: NIHMS1930865
PMID: 37707346

Current and Emerging Pharmacotherapies for the Treatment of Pulmonary Arterial Hypertension in Infants

Abstract

Introduction:

Pulmonary hypertension (PH) is a complex condition that encompasses an array of underlying disease processes and affects a diverse population of infants, including those with congenital heart disease, congenital diaphragmatic hernia, persistent PH of the newborn, and those with lung disease such as bronchopulmonary dysplasia. While there are treatments available to adults with PH, limited data exists for infants, especially for the newer medications. Therapies that target the three main pathophysiologic pathways of pulmonary hypertension appear to benefit infants, but which are best for each individual disease process is unclear.

Areas covered:

A review of the therapies to treat pulmonary hypertension is covered in this article including the prostacyclin pathway, endothelin pathway, and the nitric oxide pathway. Other adjunctive treatments are also discussed. Findings are based on a PubMed literature search of research papers spanning 1990–2023 and a search of ongoing trials registered with clinicaltrials.gov.

Expert opinion:

Overall therapies seem to improve outcomes with most infants with PH. However, given the diverse population of infants with PH, it is imperative to understand the basis for the PH in individual patients and understand which therapies can be applicable. Further research into tailored therapy for the specific populations is warranted.

Keywords: pulmonary hypertension, infant, treatment, therapy, pulmonary arterial hypertension, pulmonary vascular resistance

1. Introduction

Pulmonary hypertension (PH) in infants is a complex condition with a multitude of underlying causes. Common causes of PH in infants include (but are not limited to) persistent pulmonary hypertension of the newborn (PPHN), PH related to lung disease – especially bronchopulmonary dysplasia (PH-BPD), PH related to congenital heart disease (PH-CHD), and PH related to congenital diaphragmatic hernia (PH-CDH).

The previous definition of pulmonary hypertension in children and infants had been the same as adults: mean pulmonary artery pressure (mPAP) >= 25 mmHg. This definition was first described in 1973 at the first World Symposium on Pulmonary Hypertension (WSPH). The 6th World Symposium on Pulmonary Hypertension (WSPH) in Nice, France in 2018 proposed a new definition for both pediatrics and adults: mPAP >20 mmHg and to include indexed pulmonary vascular resistance (PVR) >= 3 indexed Woods units (WUxm2) to identify pre-capillary PH.[1] This definition is helpful in that it captures patients that are borderline for PH and includes PVR in the definition. Estimated incidence of PH is 4–10 cases per million children (under 18 years old) per year and prevalence of 20–40 cases per million children.[2, 3] Many infants (82% in one series of over 3000 pediatric patients with PH diagnosis) will have transient PH such as PPHN and repairable cardiac defects (e.g., patent ductus arteriosus, ventricular septal defect).[2] Others will unfortunately have long term chronic pulmonary hypertension or develop PH later in life.

To standardize this diverse clinical entity, the World Health Organization (WHO) developed a classification schema that separates PH into 5 groups (Table 1). Group 1 is pulmonary arteriolar hypertension (PAH) and includes idiopathic and hereditary PAH, PAH-CHD, PPHN, drug-induced PAH, and PAH associated with connective tissue disease (PAH-CTD). Group 2 is PH secondary to left-sided heart disease (post-capillary PH) and includes left ventricular diastolic dysfunction, pulmonary vein stenosis, mitral stenosis/regurgitation, and assorted cardiomyopathies. Group 3 is PH due to lung diseases and hypoxia, including PH-BPD, PH-CDH, lung hypoplasia disorders, and surfactant abnormalities. Group 4 is PH due to pulmonary artery obstructions such as chronic thromboembolic pulmonary hypertension (CTEPH) which is relatively uncommon in infants and children. And finally, Group 5 is PH with unclear or multifactorial mechanisms such as single ventricle patients, including those with Glenn or Fontan physiology (passive pulmonary blood flow system).[4]

Table 1.

World Health Organization Pulmonary Hypertension Groups

GROUPCauseMechanismExamples
Group 1Pulmonary Arterial HypertensionPH due to vascular remodeling of the pulmonary arteriesIdiopathic PAH
PAH caused by congenital heart disease
Persistent PH of the newborn (PPHN)
Drug-induced PH
PAH associated with connective tissue diseases
Group 2Left Heart DiseasePH due to left heart disease causing congestion and back up of blood flowLV systolic dysfunction
LV diastolic dysfunction
Pulmonary vein stenosis
Cardiomyopathy
Group 3Lung DiseasePH due to chronic lung disease and hypoxemiaPH related to bronchopulmonary dysplasia
PH related to congenital diaphragmatic hernia
Group 4Chronic Thromboembolic Pulmonary HypertensionPH due to chronic thromboembolic pulmonary hypertensionUncommon in infants and children
Group 5Multifactorial or Unclear CauseMiscellaneous or unknown causesSingle ventricle patients including those with Glenn and Fontan physiology

Many of the diagnoses linked to the different WHO groups serve as risk factors for the development of pulmonary hypertension in infants such as BPD, congenital heart disease, CDH, and lung hypoplasia. Other risk factors include meconium aspiration syndrome, trisomy 21 and other genetic/metabolic syndromes, sepsis, lung disease, diazoxide use, maternal smoking and/or drug use, and placental dysfunction.[1]

Once diagnosed with PH, patients can be risk stratified by several variables. Higher risk patients include those with right ventricular failure, progressive symptoms, poor growth, syncope, WHO functional class 3 or 4, rising B-type natriuretic peptide (BNP), or pericardial effusion.[5, 6]

Clinical evaluation of pulmonary hypertension includes electrocardiogram, chest x-ray, echocardiogram, laboratory testing including inflammatory markers and BNP, and genetic studies as well as cardiac catheterization. Ventilation-to-Perfusion (V/Q) scans and cross-sectional imaging can also be useful in select cases. In older children and adults, WHO functional classification, the 6-minute walk test, and cardiopulmonary exercise testing (CPET) can be helpful in assessing treatment goals, but these are not applicable to infants.[6, 7] Often, a multidisciplinary approach involving a diverse mix of care teams is beneficial for the evaluation and management of pulmonary hypertension including but not limited to: cardiologists, pulmonologists, intensivists, pharmacists, nutrition experts, rheumatologists, social work, surgeons, and palliative care specialists.

2. Pharmacotherapy treatment (Table 2)

Table 2.

Common Pharmacotherapies for Infants with Pulmonary Arterial Hypertension

MedicationRoutes of AdministrationClassTherapeutic TargetFDA indicationSummary / Trials
BosentanOralEndothelin-1 Receptor AntagonistOff-label in infants; in pediatric patients aged 3 years and older with idiopathic or congenital PAH to improve pulmonary vascular resistance (PVR), which is expected to result in an improvement in exercise ability.More 2016 (37) - Cochrane review: Two RCTs were included in the Cochrane review. There was inadequate evidence to support the use of oral bosentan.
Ambrisentan and MacitentanOralEndothelin-1 Receptor AntagonistInfants - Off Label; Adults - Indication for WHO Group 1 pulmonary artery hypertensionMay have less hepatotoxicity and drug-drug interactions
EpoprostenolIV or inhaledProstacyclinActivation of adenylate cyclase and production of cyclic AMP in vascular smooth musclesInfants - Off Label; Adults - Indication for WHO Group 1 pulmonary artery hypertensionIn patients with severe PPHN despite inhaled nitric oxide, about 40% will respond to IV epoprostenol with an average improvement in OI by 11. Ahmad 2018 (45); Short half life of 6 minutes.
IloprostIV or inhaledProstacyclinActivation of adenylate cyclase and production of cyclic AMP in vascular smooth musclesInfants - Off Label; Adults - Indication for WHO Group 1 pulmonary artery hypertensionShown to improve PaO2 and OI in subset of responders (n=12) Verma 2022 (47).
TreprostinilIV, oral, subcutaneousProstacyclinActivation of adenylate cyclase and production of cyclic AMP in vascular smooth musclesInfants - Off Label; Adults - Indication for WHO Group 1 pulmonary artery hypertensionMore stable than epoprostenol with half-life of 3 hours in adults.
Prostaglandin E1IVProstacyclinIncreases cyclic AMP in vascular smooth muscles; promotes ductal patencyPGE1 is indicated for palliative, not definitive, therapy to temporarily maintain the patency of the ductus arteriosus until corrective or palliative surgery can be performed in neonates who have congenital heart defects and who depend upon the patent ductus for survivalIn CDH or PPHN can be considered for neonates with severe PPHN but definitive studies of benefit are lacking.
Inhaled nitric oxideInhalationMiscellaneous respiratory agentActivation of guanylate cyclase leading to accumulation of cyclic GMPIndicated for term and near term (>34 weeks gestation) with hypoxic respiratory failure to improve oxygenation and lower risk of ECMO. Off-label use in premature infants with pulmonary hypertension.Cochrane meta-analysis found that inhaled nitric oxide at 20 parts per million effective in reducing use of ECMO for acute hypoxic respiratory failure. Risks and benefits unknown in preterm (<34 weeks gestation) populationpie (8, 12).
SildenafilIV, oralPhosphodiesterase-5 inhibitorsInhibits the breakdown of vasodilatory cGMPIndicated to treat pulmonary arterial hypertension WHO Group 1 in children 1–17 years old. Used off-label to treat lung disease associated pulmonary hypertension (WHO Group 3).Limited data on treatment for PPHN in resource-limited settings where iNO not available (Kamran, Imam) and as adjunctive therapy to iNO (21, 22). Safety and efficacy for treatment of WHO Group 1 PH in pediatric population older than 1 year described in STARTS-1 trial. Limited safety and efficacy data in infants born premature with BPD-related PH.
TadalafilOralPhosphodiesterase-5 inhibitorsInhibits the breakdown of vasodilatory cGMPAdults with pulmonary artery hypertension WHO Group 1Small trials have shown tadalafil can be used in children (28,29).
MilrinoneIVInotropic agent; phosphodiesterase inhibitorImproves cardiac contractility (inotropy), cardiac relaxation (lusitropy), and promotes pulmonary vasodilation through inhibition of phosphodiesterasesLimited data on treatment for PPHN in resource-limited settings where iNO not available and PPHN refractory to iNO (20, 30, 32). Case series in CDH found improved right ventricular function on echo with milrinone (88).
RiociguatOralSoluble guanylate cyclase stimulatorIt promotes formation of cGMP and promotes relaxation of the vascular smooth muscle cellsAdults with pulmonary artery hypertension WHO Group 1 and Group 4Clinical trials are currently ongoing in children to assess riociguat (NCT 02562235).

2.1. Nitric Oxide Pathway

Nitric oxide (NO) released by vascular endothelial cells is the primary mediator of pulmonary vascular tone. The production of NO via nitric oxide synthase (NOS) results in activation of soluble guanylate cyclase in smooth muscle cells leading to an increase in cyclic guanosine monophosphate (cGMP) and smooth muscle relaxation. Type 5 phosphodiesterase, the predominant isoform in the lung, regulates cGMP-specific signaling pathways by breaking down cGMP. The NO-cGMP pathway is targeted in two common treatments of PH: inhaled NO (iNO) and sildenafil, a potent inhibitor of type 5 phosphodiesterase.

Inhaled NO is the most studied therapeutic for the treatment of PH. It results in selective pulmonary vasodilation without reducing systemic vascular tone. When used in conjunction with ventilatory support and other appropriate agents, iNO is approved by the Food and Drug Administration to improve oxygenation and reduce the need for extracorporeal membrane oxygenation in infants >34 weeks’ gestation with PPHN and hypoxic respiratory failure. A Cochrane Review of 17 randomized controlled trials in term and near-term infants with hypoxia found that an initial concentration of 20 parts per million (ppm) was effective in reducing the use of extracorporeal membrane oxygenation (ECMO).[8] Because it combines with hemoglobin in the circulation to form methemoglobin, infants treated with iNO need to have methemoglobin levels checked periodically.

The evidence supporting the use of iNO in preterm infants (<=34 weeks) is less consistent. Furthermore, there are safety concerns related to interventricular hemorrhage in this high-risk population due to the known effects of iNO on inhibition of platelet aggregation.[9, 10] Clinical guidelines released in 2014 by the American Academy of Pediatrics recommended against routine and rescue use of iNO in preterm infants with respiratory failure based on available evidence.[11] A Cochrane Review meta-analysis of 17 randomized controlled trials reported a similar conclusion that early or late iNO use in preterm infants did not reduce mortality or bronchopulmonary dysplasia.[12] Despite these findings, authors from the Pediatric Pulmonary Hypertension Network point out that the majority of clinical trials of iNO in preterm infants were focused on the prevention of BPD and note that iNO therapy may be beneficial for a subset of preterm infants with severe hypoxemia that is primarily attributed to PPHN physiology, rather than parenchymal lung disease.[13] Several case series of preterm infants with a history of preterm, premature rupture of membranes, prolonged oligohydramnios, or pulmonary hypoplasia (and therefore at higher risk for PPHN) demonstrated improved oxygenation with iNO, consistent with the expected response in term infants with PPHN.[14, 15, 16, 17] Due to a lack of clinical equipoise for performing randomized, placebo-controlled trials of iNO in preterm infants with PPHN, prospective observational studies have been proposed to evaluate clinical evidence for the safety and efficacy of iNO in preterm infants with PPHN. A multicenter, prospective registry study of infants with PPHN receiving iNO in the first week of life found that iNO was at least as effective in improving oxygenation index in infants born at 27–34 weeks compared to those born at term or near-term.[18]

Sildenafil inhibits type 5 phosphodiesterase (PDE-5) leading to pulmonary vasodilation. Sildenafil is approved by the FDA for the treatment of pulmonary arterial hypertension in adults and pediatric patients 1 to 17 years old. In term and near-term infants with PPHN, sildenafil has been studied in resource-limited settings where iNO is not available[19, 20] and as adjunctive therapy to iNO.[21, 22] These studies found an improvement in oxygenation index and pulmonary arterial pressure with sildenafil use in the setting of PPHN, however more robust trials are needed to demonstrate safety and efficacy in this population. Sildenafil is frequently used off-label to treat PH associated with BPD.[23] Case series of enteral sildenafil at doses 1.5–8 mg/kg/day have demonstrated improvement in BPD-associated PH based on echocardiography.[24, 25, 26, 27] Given the widespread use of sildenafil to treat BPD-associated PH, there likely is a lack of equipoise to conduct placebo-controlled trials evaluating the safety and efficacy of sildenafil in this population. Tadalafil is a long-acting type 5 phosphodiesterase inhibitor this is once daily dosing. Given it is a relatively newer agent, it has less data available in infants compared to sildenafil, but it appears to be well tolerated and similarly efficacious. [28, 29]

Milrinone is a type 3 phosphodiesterase inhibitor which promotes pulmonary vasodilation by preventing cyclic AMP breakdown. In addition to relaxation of vascular smooth muscle, milrinone increases myocardial contractility and improves myocardial relaxation. Preliminary studies have investigated the use of milrinone for PPHN in settings where iNO is not available[20, 30] and in cases where PPHN is refractory to iNO alone.[31, 32] Milrinone may be particularly effective in the context of left ventricular dysfunction and pulmonary venous hypertension by increasing cardiac output.[32] In the clinical arena, it is also noteworthy to consider the effects of vasoactive infusion therapy on both systemic vascular resistance (SVR) and pulmonary vascular resistance (PVR). For example, there is evidence that vasopressin will increase SVR and lower PVR, while norepinephrine raises both SVR and PVR. Therefore, care must be taken when using vasoactive agents in patients with or at-risk for PH.[33]

2.2. Endothelin Pathway

Endothelin-1 (ET-1) is a potent vasoconstrictor peptide that acts on two receptors: ETA and ETB. ET-1 is made by vascular endothelial cells. ETA receptor promotes vasoconstriction while ETB promotes vasodilation. This is the pathway targeted by endothelin receptor antagonists (ERA); in infants the most studied drug in this class is bosentan.

Bosentan is an ET-1 receptor antagonist and works at both ETA and ETB receptors. Bosentan is beneficial in adults with PH and there are limited studies in infants with some signal of benefit. In infants with PPHN a study by Kumar et al showed elevated levels of ET-1 in infants with PPHN compared to controls but this finding was not found in another study by McDonald et al who looked at ET-1 levels in infants with PPHN requiring ECMO.[34, 35] A study by de Laguisie in infants with CDH showed that ET receptors, and preferentially ETA receptors, demonstrated over-expression and could contribute to PPHN in CDH.[36]

A 2016 Cochrane review examined 2 ERA studies for PPHN in infants. [37, 38, 39] In the first study by Mohamed et al., they compared bosentan alone vs placebo in 47 infants >= 34 weeks gestation. That study showed better oxygenation index and shorter duration of mechanical ventilation in the bosentan group. The second study by Steinhorn et al. compared inhaled nitric oxide plus bosentan vs inhaled nitric oxide plus placebo in 21 infants with PPHN who were >=34 weeks gestation. This study showed that bosentan was well tolerated by infants but there was no significant clinical improvement with treatment. The study was confounded by imbalance in markers of disease illness between groups (higher OI and use of vasoactives in the bosentan group). The pharmacokinetics from this study demonstrated that steady state was achieved by day 5 of treatment. Overall, the Cochrane review concluded that: “There is inadequate evidence to support the use of ERAs either as stand-alone therapy or as adjuvant to inhaled nitric oxide in PPHN. Adequately powered RCTs are needed.”[37]

Bosentan is metabolized by the liver and excreted via the biliary system. It induces the CYP3A4 enzyme which is clinically relevant especially because it will decrease the plasma concentration of sildenafil (which is cleared by hepatic metabolism). [40] It is only available in oral form. The infant dose of bosentan is typically 2–4 mg/kg/day divided twice a day.[41] It requires monitoring via the REMS program due to adverse reactions related to transaminitis and liver injury as well as teratogenicity. Per the company guidelines, bosentan should not be initiated or continued if the alanine transaminases and aspartate aminotransferases are three times the upper limit of normal or greater OR the total bilirubin is two times the upper limit of normal or greater. Patients require continued surveillance of their liver enzymes (and methods of contraception if childbearing age) while on the medication.[42]

Ambrisentan and macitentan are other endothelin receptor antagonists approved for adults with pulmonary hypertension, however there is limited data in children and specifically, infants. Ambrisentan is an oral, once-daily, ETA-selective ERA. In adults it was associated with better exercise tolerance as measured by 6-minute walk test. It also has shown less risk of hepatotoxicity obviating the need for laboratory monitoring as well as less drug-drug interactions compared to bosentan (most notably bosentan interacts with sildenafil and warfarin, which was not seen in studies of ambrisentan). [43] Macitentan is an oral, once-daily, ET-1 receptor antagonist and works at both ETA and ETB receptors (similar to bosentan). Like ambrisentan, macitentan has less concern for hepatotoxicity and drug-drug interactions.[44] There is currently a multi-center study investigating macitentan in infants and children with pulmonary arterial hypertension (NCT02932410).

2.3. Prostacyclin Pathway

Prostacyclin medications are another class of pulmonary vasodilator therapy that work via the arachidonic acid-prostacyclin pathway and lead to activation of adenylate cyclase and production of cyclic AMP in vascular smooth muscle cells, specifically the pulmonary artery smooth muscle in PH. Prostaglandin I2 (PGI2) analogs are the main prostacyclins used clinically in PH therapy. Three main analogs of PGI2 are available and each has specific routes of administration: epoprostenol, iloprost, and treprostinil.

Epoprostenol is given commonly as an IV continuous infusion. It was the first prostacyclin analog approved for PH therapy in the 1990s. It has a short half-life of 6 minutes which can make it a challenging medication to manage as an outpatient. It also can reduce systemic blood pressure prohibitively, which can limit achieving ideal dosage and effect. A study of 36 patients demonstrated response in 15 patients when added to inhaled nitric oxide. In the responder group, the average improvement in oxygenation index was 11 within 4 hours.[45] Systemic hypotension can be avoided by using the inhaled version of epoprostenol. Typical dosage of inhaled epoprostenol is 50 ng/kg/min.[46]

Iloprost is another prostacyclin analog that can be used to treat PH via intravenous route or inhaled. In a study of infants with PPHN it has been shown to improve PaO2 and oxygenation index in a subset of responders (n=12).[47] Another study by Kahveci et al also showed clinical improvement (lower systolic pulmonary artery pressure and lower oxygenation index) in infants with PPHN treated with inhaled iloprost in comparison to oral sildenafil.[48] The dose in this study for infants was 1–2.5 mcg/kg/dose with an interval of 2–4 hr between doses for all patients. For reference, the recommended dose for older pediatric patients is 2.5 or 5 μg of inhaled iloprost. It is recommended to start with the lower dose and then increase the dose for the second inhalation, if tolerated. Iloprost should not be taken less than 6 times per day and up to 9 times are allowed. Max dosing is 45 mcg per day.[6, 7, 49]

Treprostinil is a prostacyclin analog that was developed to be more stable than epoprostenol, with a longer half-life of 3 hours in adults. It can be delivered by oral, subcutaneous, or intravenous route. There is limited literature in infants and children but that limited data seems to support that treprostinil may be safe in infants and children.[50, 51] In a study of 5 pediatric patients started on subcutaneous treprostinil for PH, the initial dose was 1.25 ng/kg/min with an up-titration of 1.25 ng/kg/min every 12–48 hours to a target dose of 20 ng/kg/min and then titrated up weekly to a range of 20–50 ng/kg/min. This medication should be used cautiously in patients with hepatic dysfunction and may require dose reduction or discontinuation. Hypotension has also been reported.[51]

Prostaglandin E1 (PGE1) also known as alprostadil is commonly used medication as a continuous IV infusion to keep the patent ductus arteriosus open in neonates with critical congenital heart disease as a palliation to promote either systemic or pulmonary blood flow. Similar to other prostacyclins, it acts by increasing cyclic AMP. It also has been theorized to be of benefit for neonates with severe PH consisting of supra-systemic pulmonary artery pressures by keeping the PDA open to allow for a right to left pop-off shunt to reduce the pressure load of the right ventricle. This concept is supported by a 2 small studies in CDH infants [52, 53] and a brief report of 51 infants with PPHN without congenital heart disease. In the PPHN study, PGE1 therapy was associated with shorter LOS but no difference in hospital mortality.[54] In summary, PGE1 can be considered for neonates with severe PH, particularly patients with CDH or PPHN.

In general, this group of medications has a common side effect profile that should be considered. Hypotension can be seen especially in IV formulations. Rebound pulmonary hypertension can occur with abrupt discontinuation of the medication. Flushing, syncope, and hypotension can also occur and may require dose reduction or discontinuation of the medication. These medications may also inhibit platelet aggregation and lead to bleeding complications. Significant jaw pain can develop in patients and limit tolerance to treatment. With inhalation forms of prostacyclin analogs, bronchospasm may occur.[7, 41]

2.4. Other Pharmacotherapies for Pulmonary Hypertension

Other common medications have been considered for treatment of PH including steroids, oxygen, calcium channel blockers, and diuretics. Clinical data regarding steroid use in infants with pulmonary hypertension is limited to case reports and case series.[55, 56] There is limited animal data that supports a possible role for glucocorticoids in PPHN via inhibition of phosphodiesterase-5, but clinical data is lacking.[57, 58] Oxygen therapy and the treatment of hypoxia promotes pulmonary vasodilation in preterm and term infants. However, this needs to be balanced with the risks of hyperoxia in preterm infants such as retinopathy of prematurity.[59] Optimal oxygen saturations are unclear but targeting 90–97% oxygen saturations appear to be a reasonable goal.[60] Although a common treatment in adults for pulmonary hypertension, calcium channel blockers have limited data in infants. One study of 9 infants with PH-BPD showed no improvement in PAP or PVR/SVR with treatment of a CCB.[61] Diuretics (including loops, thiazides, and K-sparing diuretics) can be used for symptomatic relief and decongestion experienced from RV or LV dysfunction in patients with PH. Interestingly, one study of PH in BPD demonstrated that 25% of infants with PH-BPD had evidence of LV diastolic dysfunction at cardiac catheterization.[7, 61] Use of diuretics requires careful management is required because over-diuresis can reduce the preload to the right ventricle and lead to impaired cardiac output.[7]

Pharmacotherapy for WHO Group 2 PH which is PH secondary to left-sided heart disease (post-capillary PH) includes a variety of medications to treat the underlying disease process. This group includes diseases such as left ventricular diastolic dysfunction, pulmonary vein stenosis, mitral stenosis/regurgitation, and assorted cardiomyopathies. Often these patients are placed on heart failure medications beyond the scope of this review but typically can consist of IV vasoactive support, beta-blockade, angiotensin converting enzyme inhibition, and diuretics including spironolactone. [62]

Newer PH drugs for adults have limited exposure to infants including selexipag, sotatercept, and riociguat. Selexipag is an oral prostacyclin receptor agonist. In adults it has demonstrated benefit but studies in infants and children are lacking.[63] Koo et al described a case of successful transition from IV Treprostinil to enteral selexipag in a 11-month-old infant but no actual studies have occurred.[64] Sotatercept is a protein that works on transforming growth factor beta and has been shown to improve 6 minute walk test in adults with PAH. Currently there is a trial in children (NCT 05587712) but no studies in infants have been performed. Riociguat was approved for use in adults with pulmonary hypertension in 2013 [65] specifically those patients with Group 1 PAH or Group 4 CTEPH. Riociguat is a soluble guanylate cyclase stimulator. This simulation of soluble guanylate cyclase acts to promote formation of cyclic guanosine monophosphate (cGMP) from guanosine triphosphate (GTP) and promotes relaxation of the vascular smooth muscle cells. Riociguat also promotes nitric oxide binding to soluble guanylate cyclase to further simulate cGMP production and vasodilation. Thus, it has a nitric oxide independent and dependent mechanism of action. Limited studies have been performed in infants but recently a Phase 3 study of 24 children (age 6–17) was completed demonstrate a suitable dosing strategy for riociguat in children and adolescents with PH but did not assess efficacy. [66] A 2023 article reviewed a series of 10 infants treated with riociguat and demonstrated improvement of some clinical markers and echocardiographic markers of pulmonary hypertension, however 2 patients experienced a marked clinical change when transitioning from sildenafil to riociguat as these two medications cannot be administered together due to risk of hypotension.[67] Clinical trials are currently ongoing in children to assess riociguat (NCT 02562235).

2.5. Therapies Other Than Medications

Medications are the main treatment for pulmonary hypertension. However, treating any underlying lung disease can also be an important therapy. This is paramount in the intubated patient wherein optimizing lung recruitment to the functional residual capacity, avoiding acidosis and hypoxemia, and considering tracheostomy are key to minimizing the risks of PH crises. For intubated patients, administration of pain and sedation medications as well as consideration for neuromuscular blockade can also be helpful in avoidance of PH crises per the American Heart Association (AHA) and American Thoracic Society (ATS) 2015 pulmonary hypertension guidelines.[7]

3. Specific infant populations

3.1. Infants with Congenital Heart Disease

Infants with certain congenital heart disease (CHD) conditions are at particular risk for PH. Generally, any lesion leading to a large left to right shunt post-tricuspid valve (e.g., ventricular septal defect (VSD), patent ductus arteriosus (PDA)), any lesion related to pulmonary venous obstructive disease (e.g., obstructed total anomalous pulmonary venous return (TAPVR), functional univentricular heart disease such as tricuspid atresia or hypoplastic left heart syndrome, and left heart obstructive disease (e.g. mitral stenosis, pulmonary vein stenosis) are associated with higher risks of developing pulmonary hypertension. Simple lesions including atrial septal defects (ASDs), VSDs, and PDAs can lead to significant pulmonary arterial hypertension, but this generally can be easily avoided if they are corrected at the appropriate age and time. More complex congenital heart disease needs a thoughtful approach of how to manage the disease and timing of any repair or palliation as well as how best to mitigate risk of pulmonary hypertension.[68] Two common scenarios leading to the development of PH are seen in CHD patients. One is the scenario where a patient had delayed repair for their condition and develops PH prior to repair. And the second scenario is in the early post operative period after repair. The effects of the PH in CHD were highlighted in an adult study that demonstrated a more two-fold increase in mortality in patients with congenital heart disease and a diagnosis of pulmonary hypertension.[69] Two recent articles described the significance of post-operative PH in congenital heart surgery patients. In one report of 818 patients, 100 (12%) had some evidence of post-operative pulmonary hypertension requiring medication administration (in this study it was sildenafil). Over 80% of the patients in the study were < 12 months old and common operations necessitating intervention for pulmonary hypertension included the arterial switch operation, VSD closure, AVSD repair, truncus arteriosus repair, and TAPVR repair.[70] Another study of the Pediatric Health Information Systems (PHIS) database found 4% of patients who underwent congenital heart surgery had a concomitant diagnosis of pulmonary hypertension and 2% required inhaled nitric oxide during their hospitalization.[71] Generally, early repair (prior to age 1–2 years old) is recommended to avoid progression of PH and development of Eisenmenger’s syndrome. Eisenmenger’s syndrome occurs when a right to left shunt with hypoxemia develops secondary to the development of elevated PVR and pulmonary hypertension in a patient with congenital heart disease born with a large systemic to pulmonary shunt such as a VSD or truncus arteriosus.[72] If there is a concern of PH in a patient with CHD prior to repair, often cardiac catheterization is recommended to measure pulmonary vascular resistance and assess candidacy of repair.[7, 73]

3.2. Infants with Persistent Pulmonary Hypertension of the Newborn

At birth the normal transition of the pulmonary system is an immediate decrease in PVR leading to an increase in pulmonary blood flow to support the infants new need for respiration.[74] However, in infants with PPHN there is a failure of this normal transition leading to persistently elevated PVR and limited pulmonary blood flow causing hypoxemia and respiratory failure. Risk factors for PPHN include delivery via Caesarean, meconium aspiration syndrome, maternal smoking, and maternal use of medications including SSRI and NSAIDS.[75]

Inhaled nitric oxide is the most studied pulmonary vasodilators in this population. A recent Cochrane review showed that it is an effective therapy for term infants with hypoxemic respiratory failure including those with PPHN.[8] Sildenafil is also utilized as a treatment for PPHN and has some small studies that support its use as described in a recent 2017 Cochrane review article.[76] The authors found 5 trials comprising of 166 participants. The data suggests reduced mortality and improved oxygenation with sildenafil but the quality of evidence was rated as low and in need of a large scale study. Another recent randomized controlled trial (RCT) of IV sildenafil added to iNO compared to placebo in 59 infants demonstrated no improvement but that IV sildenafil was relatively safe with minimal significant adverse events.[21]

Multiple prostacyclin analogues have been studied in PPHN. A retrospective study of PGE1 in infants with PPHN demonstrated a benefit of shorter hospital length of stay but no benefit in mortality.[54] Iloprost was shown to be beneficial in patients with PPHN who were not responding to iNO in a small study of 22 patients.[47] IV treprostinil has limited data in PPHN: 2 case reports showed clinical improvement and tolerance of the medication. [77]

Studies of bosentan in PPHN have mostly been limited by small sample size. Generally, the studies have shown that bosentan is safe and tolerated in this population. Benefit is limited but there is some evidence that it can reduce PA pressures and improve oxygenation. Mortality benefit has not been established. [38, 78, 79, 80]

3.3. Infants with Congenital Diaphragmatic Hernia

Congenital diaphragmatic hernia (CDH) is a condition due to improper formation of the diaphragm during fetal development. It can present with varying severity often related to the size of the defect. The defect in the diaphragm leads to abdominal contents to enter the thoracic cavity leading to a mass effect on the developing lungs. This causes abnormal lung development leading to pulmonary hypoplasia and pulmonary hypertension, which can be fatal. Studies show that an abnormal pulmonary vasculature develops with arterial remodeling, altered vascular response, and decreased vascular bed density [81, 82, 83]. Various PH therapies have been studied in the CDH population. Nitric oxide was studied in a multicenter study in 1997 in n=53 infants with CDH[84]. Patients were randomized to receive inhaled nitric oxide (treatment arm) or 100% oxygen (control arm). Treatment patients exhibited short-term improvements of oxygenation but no reduction in need for ECMO or death. Another study concluded that there may be a subset or CDH patients that respond to inhaled nitric oxide and that it can be beneficial in the extubated patient via nasal cannula [85]. Case reports of use of sildenafil in CDH patients show improvement in pulmonary artery pressures and oxygenation as well as echocardiographic indices of PH.[86, 87]

A case series in 6 infants with CDH and PH reviewed effects of milrinone and saw an improvement of echocardiographic indices of RV function, however rigorous data is limited.[88] There is an ongoing clinical trial evaluating milrinone in CDH patients for PH.[89] Bosentan has theoretical benefit in CDH patients with PH but no clinical trials have been done in CDH patients to show benefit.[90] Prostacyclins and prostaglandin E1 also have limited data but have been used to treat PH in CDH infants.[53, 91]

A related disease process to PH-CDH is PH secondary to lung hypoplasia. This can be seen in infants with premature rupture of membranes, kidney disorders, and omphaloceles. Research is limited on this topic. One large multicenter study showed no evidence of improved survival with inhaled nitric oxide in extremely premature infants and pulmonary hypoplasia; however, another case report demonstrated significant improvement with iNO.[92, 93] The mixed conclusions are likely a result of the heterogeneity of underlying causes of PH, leading to different patients having different responses to the limited array of PH therapies available.

3.4. Infants with Acute Right Ventricular Failure / Pulmonary Hypertensive Crisis

In patients with pulmonary hypertension, a sudden increase in pulmonary arterial pressures because of an abrupt increase in pulmonary vascular resistance can lead to acute right ventricular failure, also known as a pulmonary hypertensive crisis (PHC). PHC can be lethal and lead to profound hypoxemia and hypotension. Often, a PHC occurs after cardiac surgery or heart or lung transplantation, however, any patient with significant pulmonary hypertension is at risk for a PHC. It can be triggered by acute illness such as a viral or bacterial pneumonia but also triggered by any stressor such as surgery, anesthesia, withdrawal of PH therapies, or airway instrumentation (like intubation or suctioning). Pharmacotherapy for a PHC is directed at fast acting therapies typically administered in the critical care setting. Common agents include inhaled nitric oxide, IV sildenafil, and inhaled/IV prostacyclins. Other management includes intubation and mechanical ventilation as well as correcting acidosis, hypoxemia, and hypercarbia. Certain patients may need vasoactive support to avoid right ventricular ischemia caused by significant hypotension and treat the right ventricular dysfunction. For intubated patients, sedation and possibly neuromuscular blockade can be helpful. Refractory cases may require mechanical circulatory support such as ECMO. Select cases can benefit from balloon atrial septostomy as well.[5, 7]

4. Conclusion

Pulmonary hypertension is a broad syndrome that encompasses a multitude of underlying disease processes and significantly affects a diverse population of infants including those with congenital heart disease, congenital diaphragmatic hernia, persistent PH of the newborn, and those with lung disease including bronchopulmonary dysplasia. While there is a wide array of treatments available to adults with PH, limited data exists for infants, especially for the newer generation of medications. Therapies that target the three main pathophysiologic pathways of pulmonary hypertension appear to benefit infants, but which are best for each individual disease process is still yet to be elucidated.

5. Expert opinion

There has been significant improvement in the care in infants with PH over the last few decades. The advent of inhaled nitric oxide has led to significant improvements for infants with PPHN especially for the risk of death or need of ECMO support.[8] The use of sildenafil has also been of benefit as an oral therapy for PH in infants.[20, 21] Also, the benefits (and risks) of supplemental oxygen in infants has become better understood.[59, 60] These three agents are the common therapies for PH currently employed in infants. As described throughout the paper, many of the other adult PH therapies have been tried in infants but often research is limited to small studies or case reports. Most therapies have been shown to be tolerated well by infants, but which therapy is best suited for each group of PH is not clear at this point.

Ideally, PH therapy in infants would have a tailored treatment plan for the different disease processes that lead to PH. There is potential for achieving this goal given the advent of newer medications that are available for the adult population but are only recently beginning to be studied in the pediatric population. These medications first need to be shown to be safe for the general infant population. And then they need to be studied in the different groups of infants with PH. For example, the same medication or combination of medications may be effective for PH related to congenital diaphragmatic hernia but not to PH related to congenital heart disease. To this end, multiple investigational studies are underway including studies of sildenafil, inhaled iloprost, nebulized magnesium sulfate, inhaled nitric oxide, IV remodulin (Treprostinil), milrinone, sotatercept, and riociguat (clinicaltrials.gov).

Various treatment algorithms have been proposed that give a general guideline for practitioners and can be viewed elsewhere including a guideline from the American Heart Association (AHA) and The European Pediatric Pulmonary Vascular Disease Network (EPPVDN) that are quite helpful.[7, 68] Briefly, once pulmonary arterial hypertension is diagnosed and post-capillary PH is excluded, the initial management depends on how sick the patient appears. Often monotherapy with either inhaled nitric oxide or a PDE-5 inhibitor is first line. This is then followed by the addition of an ERA or a prostacyclin agent for second line and then third line therapy would be triple therapy with treatment directed at all three pathways (nitic oxide, endothelin, and prostacyclin).

Another weakness in PH treatment in infants is the lack of reliable intermediate outcomes or surrogate markers of disease. While studies of adult patients can quantify the functional class of a study participant or the six-minute walk test measure, in infants there are only a few surrogate markers of clinical outcomes, mainly limited to echocardiographic measurements and growth. Possible biologic measures could include B-type natriuretic peptide (BNP) and inflammatory markers, both of which are currently under investigation (clinicaltrials.gov). Cardiac MRI could be another measure that has yet to be explored as an endpoint for efficacy.

Given the heterogeneity of the infant population with PH and the small numbers of patients, it is important to also consider the use of multi-center studies to achieve meaningful results. The growth of collaborative network and databases such as the Pediatric Pulmonary Hypertension Network (PPHNet), pediatric cardiac critical care consortium (PC4), and Congenital Diaphragmatic Hernia Registry (CDHR) should hopefully aid in this crucial avenue of research. Additionally, more studies evaluating the genetic basis for the different types of PH would be beneficial to all patients with PH including infants. And finally, long term follow-up of these patients including neurodevelopmental outcomes would clarify this disease process and how to optimally treat this delicate patient population to best support these patients and their families.

Article highlights:

  • Pulmonary hypertension is a complex condition that affects a diverse population of infants.

  • Management strategies should be tailored to address specific characteristics of pulmonary hypertension in infants based on co-morbidities such as congenital heart disease, congenital diaphragmatic hernia, and bronchopulmonary dysplasia.

  • We describe the nitric oxide, endothelin, and prostacyclin pathways and the mechanisms of action of drugs targeting these pathways to treat pulmonary hypertension.

  • We present a review of current and future therapies to improve outcomes in infants with pulmonary hypertension.

  • We highlight treatment algorithms based on consensus guidelines from the American Heart Association and the European Pediatric Pulmonary Vascular Disease Network.

Funding

This paper is supported by a mentoring award from the National Heart, Lung, and Blood Institute for M M Laughon (1K24HL143283).

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Footnotes

Reviewer disclosures

A reviewer on this manuscript has disclosed being a Consultant for Johnson & Johnson, UT, and Bayer. Peer reviewers on this manuscript have no other relevant financial relationships or otherwise to disclose.

References

Papers of special note have been highlighted as either of interest (*) or of considerable interest (**) to readers.

1. Rosenzweig EB, Abman SH, Adatia I, et al. Paediatric pulmonary arterial hypertension: updates on definition, classification, diagnostics and management. Eur Respir J. 2019. Jan;53(1). 10.1183/13993003.01916-2018. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
2. van Loon RL, Roofthooft MT, Hillege HL, et al. Pediatric pulmonary hypertension in the Netherlands: epidemiology and characterization during the period 1991 to 2005. Circulation. 2011. Oct 18;124(16):1755–64. 10.1161/CIRCULATIONAHA.110.969584. [Abstract] [CrossRef] [Google Scholar]
3. Li L, Jick S, Breitenstein S, et al. Pulmonary arterial hypertension in the USA: an epidemiological study in a large insured pediatric population. Pulm Circ. 2017. Mar;7(1):126–136. 10.1086/690007. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. Simonneau G, Montani D, Celermajer DS, et al. Haemodynamic definitions and updated clinical classification of pulmonary hypertension. Eur Respir J. 2019. Jan;53(1). 10.1183/13993003.01913-2018. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
5. Galie N, Humbert M, Vachiery JL, et al. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur Heart J. 2016. Jan 1;37(1):67–119. 10.1093/eurheartj/ehv317. [Abstract] [CrossRef] [Google Scholar]
6. Hansmann G, Apitz C. Treatment of children with pulmonary hypertension. Expert consensus statement on the diagnosis and treatment of paediatric pulmonary hypertension. The European Paediatric Pulmonary Vascular Disease Network, endorsed by ISHLT and DGPK. Heart. 2016. May;102 Suppl 2:ii67–85. 10.1136/heartjnl-2015-309103. [Abstract] [CrossRef] [Google Scholar]
7. Abman SH, Hansmann G, Archer SL, et al. Pediatric Pulmonary Hypertension: Guidelines From the American Heart Association and American Thoracic Society. Circulation. 2015. Nov 24;132(21):2037–99. 10.1161/CIR.0000000000000329. [Abstract] [CrossRef] [Google Scholar]**Of considerable interest due to inclusion of consensus guidelines with recommendations for treatment of pediatric pulmonary hypertension.
8. Barrington KJ, Finer N, Pennaforte T, et al. Nitric oxide for respiratory failure in infants born at or near term. Cochrane Database Syst Rev. 2017. Jan 5;1(1):CD000399. 10.1002/14651858.CD000399.pub3. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
9. Samama CM, Diaby M, Fellahi JL, et al. Inhibition of platelet aggregation by inhaled nitric oxide in patients with acute respiratory distress syndrome. Anesthesiology. 1995. Jul;83(1):56–65. 10.1097/00000542-199507000-00007. eng. [Abstract] [CrossRef] [Google Scholar]
10. Högman M, Frostell C, Arnberg H, et al. Bleeding time prolongation and NO inhalation. Lancet. 1993. Jun 26;341(8861):1664–5. 10.1016/0140-6736(93)90802-n. eng. [Abstract] [CrossRef] [Google Scholar]
11. Kumar P Use of inhaled nitric oxide in preterm infants. Pediatrics. 2014. Jan;133(1):164–70. 10.1542/peds.2013-3444. eng. [Abstract] [CrossRef] [Google Scholar]
12. Barrington KJ, Finer N, Pennaforte T. Inhaled nitric oxide for respiratory failure in preterm infants. Cochrane Database Syst Rev. 2017. Jan 3;1(1):Cd000509. 10.1002/14651858.CD000509.pub5. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Kinsella JP, Steinhorn RH, Krishnan US, et al. Recommendations for the Use of Inhaled Nitric Oxide Therapy in Premature Newborns with Severe Pulmonary Hypertension. J Pediatr. 2016. Mar;170:312–4. 10.1016/j.jpeds.2015.11.050. eng. [Abstract] [CrossRef] [Google Scholar]
14. Peliowski A, Finer NN, Etches PC, et al. Inhaled nitric oxide for premature infants after prolonged rupture of the membranes. J Pediatr. 1995. Mar;126(3):450–3. 10.1016/s0022-3476(95)70467-1. eng. [Abstract] [CrossRef] [Google Scholar]
15. Chock VY, Van Meurs KP, Hintz SR, et al. Inhaled nitric oxide for preterm premature rupture of membranes, oligohydramnios, and pulmonary hypoplasia. Am J Perinatol. 2009. Apr;26(4):317–22. 10.1055/s-0028-1104743. eng. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
16. Shah DM, Kluckow M. Early functional echocardiogram and inhaled nitric oxide: usefulness in managing neonates born following extreme preterm premature rupture of membranes (PPROM). J Paediatr Child Health. 2011. Jun;47(6):340–5. 10.1111/j.1440-1754.2010.01982.x. eng. [Abstract] [CrossRef] [Google Scholar]
17. Semberova J, O’Donnell SM, Franta J, et al. Inhaled nitric oxide in preterm infants with prolonged preterm rupture of the membranes: a case series. J Perinatol. 2015. Apr;35(4):304–6. 10.1038/jp.2015.2. eng. [Abstract] [CrossRef] [Google Scholar]
18. Nelin L, Kinsella JP, Courtney SE, et al. Use of inhaled nitric oxide in preterm vs term/near-term neonates with pulmonary hypertension: results of the PaTTerN registry study. J Perinatol. 2022. 2022/01/01;42(1):14–18. 10.1038/s41372-021-01252-x. [Abstract] [CrossRef] [Google Scholar]
19. Kamran A, Rafiq N, Khalid A, et al. Effectiveness of oral sildenafil for neonates with persistent pulmonary hypertension of newborn (PPHN): a prospective study in a tertiary care hospital. J Matern Fetal Neonatal Med. 2022. Dec;35(25):6787–6793. 10.1080/14767058.2021.1923003. eng. [Abstract] [CrossRef] [Google Scholar]
20. Imam SS, El-Farrash RA, Taha AS, et al. Milrinone Versus Sildenafil in Treatment of Neonatal Persistent Pulmonary Hypertension: A Randomized Control Trial. J Cardiovasc Pharmacol. 2022. Nov 1;80(5):746–752. 10.1097/fjc.0000000000001332. eng. [Abstract] [CrossRef] [Google Scholar]
21. Pierce CM, Zhang MH, Jonsson B, et al. Efficacy and Safety of IV Sildenafil in the Treatment of Newborn Infants with, or at Risk of, Persistent Pulmonary Hypertension of the Newborn (PPHN): A Multicenter, Randomized, Placebo-Controlled Trial. J Pediatr. 2021. Oct;237:154–161 e3. 10.1016/j.jpeds.2021.05.051. [Abstract] [CrossRef] [Google Scholar]
22. Steinhorn RH, Kinsella JP, Pierce C, et al. Intravenous sildenafil in the treatment of neonates with persistent pulmonary hypertension. J Pediatr. 2009. Dec;155(6):841–847.e1. 10.1016/j.jpeds.2009.06.012. eng. [Abstract] [CrossRef] [Google Scholar]
23. Thompson EJ, Perez K, Hornik CP, et al. Sildenafil Exposure in the Neonatal Intensive Care Unit. Am J Perinatol. 2019. Feb;36(3):262–267. 10.1055/s-0038-1667378. eng. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
24. Caputo S, Furcolo G, Rabuano R, et al. Severe pulmonary arterial hypertension in a very premature baby with bronchopulmonary dysplasia: normalization with long-term sildenafil. J Cardiovasc Med (Hagerstown). 2010. Sep;11(9):704–6. 10.2459/JCM.0b013e328332e745. eng. [Abstract] [CrossRef] [Google Scholar]
25. Hon KL, Cheung KL, Siu KL, et al. Oral sildenafil for treatment of severe pulmonary hypertension in an infant. Biol Neonate. 2005;88(2):109–12. 10.1159/000085646. eng. [Abstract] [CrossRef] [Google Scholar]
26. Mourani PM, Sontag MK, Ivy DD, et al. Effects of long-term sildenafil treatment for pulmonary hypertension in infants with chronic lung disease. J Pediatr. 2009. Mar;154(3):379–84, 384.e1–2. 10.1016/j.jpeds.2008.09.021. eng. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
27. Trottier-Boucher MN, Lapointe A, Malo J, et al. Sildenafil for the Treatment of Pulmonary Arterial Hypertension in Infants with Bronchopulmonary Dysplasia. Pediatr Cardiol. 2015. Aug;36(6):1255–60. 10.1007/s00246-015-1154-0. eng. [Abstract] [CrossRef] [Google Scholar]
28. Youssef DE, Handler SS, Richards SM, et al. Multicenter review of a tadalafil suspension formulation for infants and children with pulmonary hypertension: A North American experience. Front Pediatr. 2023;11:1055131. 10.3389/fped.2023.1055131. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Takatsuki S, Calderbank M, Ivy DD. Initial experience with tadalafil in pediatric pulmonary arterial hypertension. Pediatr Cardiol. 2012. Jun;33(5):683–8. 10.1007/s00246-012-0180-4. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
30. El-Ghandour M, Hammad B, Ghanem M, et al. Efficacy of Milrinone Plus Sildenafil in the Treatment of Neonates with Persistent Pulmonary Hypertension in Resource-Limited Settings: Results of a Randomized, Double-Blind Trial. Paediatr Drugs. 2020. Dec;22(6):685–693. 10.1007/s40272-020-00412-4. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
31. McNamara PJ, Laique F, Muang-In S, et al. Milrinone improves oxygenation in neonates with severe persistent pulmonary hypertension of the newborn. J Crit Care. 2006. Jun;21(2):217–22. 10.1016/j.jcrc.2006.01.001. [Abstract] [CrossRef] [Google Scholar]
32. McNamara PJ, Shivananda SP, Sahni M, et al. Pharmacology of milrinone in neonates with persistent pulmonary hypertension of the newborn and suboptimal response to inhaled nitric oxide. Pediatr Crit Care Med. 2013. Jan;14(1):74–84. 10.1097/PCC.0b013e31824ea2cd. [Abstract] [CrossRef] [Google Scholar]
33. Jeon Y, Ryu JH, Lim YJ, et al. Comparative hemodynamic effects of vasopressin and norepinephrine after milrinone-induced hypotension in off-pump coronary artery bypass surgical patients. Eur J Cardiothorac Surg. 2006. Jun;29(6):952–6. 10.1016/j.ejcts.2006.02.032. [Abstract] [CrossRef] [Google Scholar]
34. Kumar P, Kazzi NJ, Shankaran S. Plasma immunoreactive endothelin-1 concentrations in infants with persistent pulmonary hypertension of the newborn. Am J Perinatol. 1996. Aug;13(6):335–41. 10.1055/s-2007-994352. [Abstract] [CrossRef] [Google Scholar]
35. Macdonald PD, Paton RD, Logan RW, et al. Endothelin-1 levels in infants with pulmonary hypertension receiving extracorporeal membrane oxygenation. J Perinat Med. 1999;27(3):216–20. 10.1515/JPM.1999.030. [Abstract] [CrossRef] [Google Scholar]
36. de Lagausie P, de Buys-Roessingh A, Ferkdadji L, et al. Endothelin receptor expression in human lungs of newborns with congenital diaphragmatic hernia. J Pathol. 2005. Jan;205(1):112–8. 10.1002/path.1677. [Abstract] [CrossRef] [Google Scholar]
37. More K, Athalye-Jape GK, Rao SC, et al. Endothelin receptor antagonists for persistent pulmonary hypertension in term and late preterm infants. Cochrane Database Syst Rev. 2016. Aug 18;2016(8):CD010531. 10.1002/14651858.CD010531.pub2. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]*Of interest due to providing high level of evidence for the use of endothelin receptor antagonists for PPHN.
38. Steinhorn RH, Fineman J, Kusic-Pajic A, et al. Bosentan as Adjunctive Therapy for Persistent Pulmonary Hypertension of the Newborn: Results of the Randomized Multicenter Placebo-Controlled Exploratory Trial. J Pediatr. 2016. Oct;177:90–96 e3. 10.1016/j.jpeds.2016.06.078. [Abstract] [CrossRef] [Google Scholar]
39. Mohamed WA, Ismail M. A randomized, double-blind, placebo-controlled, prospective study of bosentan for the treatment of persistent pulmonary hypertension of the newborn. J Perinatol. 2012. Aug;32(8):608–13. 10.1038/jp.2011.157. [Abstract] [CrossRef] [Google Scholar]
40. Paul GA, Gibbs JS, Boobis AR, et al. Bosentan decreases the plasma concentration of sildenafil when coprescribed in pulmonary hypertension. Br J Clin Pharmacol. 2005. Jul;60(1):107–12. 10.1111/j.1365-2125.2005.02383.x. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
41. Lakshminrusimha S, Mathew B, Leach CL. Pharmacologic strategies in neonatal pulmonary hypertension other than nitric oxide. Semin Perinatol. 2016. Apr;40(3):160–73. 10.1053/j.semperi.2015.12.004. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
42. Bosentan REMS Program Prescriber Guide [cited 2022 December 6th, 2022]. Available from: https://bosentanremsprogram.com
43. Barst RJ. A review of pulmonary arterial hypertension: role of ambrisentan. Vasc Health Risk Manag. 2007;3(1):11–22. [Europe PMC free article] [Abstract] [Google Scholar]
44. Khadka A, Singh Brashier DB, Tejus A, et al. Macitentan: An important addition to the treatment of pulmonary arterial hypertension. J Pharmacol Pharmacother. 2015. Jan-Mar;6(1):53–7. 10.4103/0976-500X.149151. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
45. Ahmad KA, Banales J, Henderson CL, et al. Intravenous epoprostenol improves oxygenation index in patients with persistent pulmonary hypertension of the newborn refractory to nitric oxide. J Perinatol. 2018. Sep;38(9):1212–1219. 10.1038/s41372-018-0179-7. [Abstract] [CrossRef] [Google Scholar]
46. Kelly LK, Porta NF, Goodman DM, et al. Inhaled prostacyclin for term infants with persistent pulmonary hypertension refractory to inhaled nitric oxide. J Pediatr. 2002. Dec;141(6):830–2. 10.1067/mpd.2002.129849. [Abstract] [CrossRef] [Google Scholar]
47. Verma S, Lumba R, Kazmi SH, et al. Effects of Inhaled Iloprost for the Management of Persistent Pulmonary Hypertension of the Newborn. Am J Perinatol. 2022. Oct;39(13):1441–1448. 10.1055/s-0040-1722653. [Abstract] [CrossRef] [Google Scholar]
48. Kahveci H, Yilmaz O, Avsar UZ, et al. Oral sildenafil and inhaled iloprost in the treatment of pulmonary hypertension of the newborn. Pediatr Pulmonol. 2014. Dec;49(12):1205–13. 10.1002/ppul.22985. [Abstract] [CrossRef] [Google Scholar]
49. Tissot C, Beghetti M. Review of inhaled iloprost for the control of pulmonary artery hypertension in children. Vasc Health Risk Manag. 2009;5(1):325–31. 10.2147/vhrm.s3222. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
50. McIntyre CM, Hanna BD, Rintoul N, et al. Safety of epoprostenol and treprostinil in children less than 12 months of age. Pulm Circ. 2013. Dec;3(4):862–9. 10.1086/674762. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
51. Ferdman DJ, Rosenzweig EB, Zuckerman WA, et al. Subcutaneous treprostinil for pulmonary hypertension in chronic lung disease of infancy. Pediatrics. 2014. Jul;134(1):e274–8. 10.1542/peds.2013-2330. [Abstract] [CrossRef] [Google Scholar]
52. Le Duc K, Mur S, Sharma D, et al. Prostaglandin E1 in infants with congenital diaphragmatic hernia (CDH) and life-threatening pulmonary hypertension. J Pediatr Surg. 2020. Sep;55(9):1872–1878. 10.1016/j.jpedsurg.2020.01.008. [Abstract] [CrossRef] [Google Scholar]
53. Lawrence KM, Berger K, Herkert L, et al. Use of prostaglandin E1 to treat pulmonary hypertension in congenital diaphragmatic hernia. J Pediatr Surg. 2019. Jan;54(1):55–59. 10.1016/j.jpedsurg.2018.10.039. [Abstract] [CrossRef] [Google Scholar]
54. Gupta N, Kamlin CO, Cheung M, et al. Prostaglandin E1 use during neonatal transfer: potential beneficial role in persistent pulmonary hypertension of the newborn. Arch Dis Child Fetal Neonatal Ed. 2013. Mar;98(2):F186–8. 10.1136/archdischild-2012-303294. [Abstract] [CrossRef] [Google Scholar]
55. Aggarwal M, Grady RM. Glucocorticoids for treating paediatric pulmonary hypertension: a novel use for a common medication. Cardiol Young. 2017. Sep;27(7):1410–1412. 10.1017/S1047951117000464. [Abstract] [CrossRef] [Google Scholar]
56. da Costa DE, Nair AK, Pai MG, et al. Steroids in full term infants with respiratory failure and pulmonary hypertension due to meconium aspiration syndrome. Eur J Pediatr. 2001. Mar;160(3):150–3. 10.1007/s004310000678. [Abstract] [CrossRef] [Google Scholar]
57. Perez M, Lakshminrusimha S, Wedgwood S, et al. Hydrocortisone normalizes oxygenation and cGMP regulation in lambs with persistent pulmonary hypertension of the newborn. Am J Physiol Lung Cell Mol Physiol. 2012. Mar 15;302(6):L595–603. 10.1152/ajplung.00145.2011. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
58. Perez M, Wedgwood S, Lakshminrusimha S, et al. Hydrocortisone normalizes phosphodiesterase-5 activity in pulmonary artery smooth muscle cells from lambs with persistent pulmonary hypertension of the newborn. Pulm Circ. 2014. Mar;4(1):71–81. 10.1086/674903. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
59. Hellstrom A, Smith LE, Dammann O. Retinopathy of prematurity. Lancet. 2013. Oct 26;382(9902):1445–57. 10.1016/S0140-6736(13)60178-6. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
60. Chandrasekharan P, Lakshminrusimha S. Oxygen therapy in preterm infants with pulmonary hypertension. Semin Fetal Neonatal Med. 2020. Apr;25(2):101070. 10.1016/j.siny.2019.101070. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
61. Mourani PM, Ivy DD, Gao D, et al. Pulmonary vascular effects of inhaled nitric oxide and oxygen tension in bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2004. Nov 1;170(9):1006–13. 10.1164/rccm.200310-1483OC. [Abstract] [CrossRef] [Google Scholar]
62. Castaldi B, Cuppini E, Fumanelli J, et al. Chronic Heart Failure in Children: State of the Art and New Perspectives. J Clin Med. 2023. Mar 30;12(7). 10.3390/jcm12072611. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
63. Coghlan JG, Channick R, Chin K, et al. Targeting the Prostacyclin Pathway with Selexipag in Patients with Pulmonary Arterial Hypertension Receiving Double Combination Therapy: Insights from the Randomized Controlled GRIPHON Study. Am J Cardiovasc Drugs. 2018. Feb;18(1):37–47. 10.1007/s40256-017-0262-z. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
64. Koo R, Lo J, Bock MJ. Transition from intravenous treprostinil to enteral selexipag in an infant with pulmonary arterial hypertension. Cardiol Young. 2019. Jun;29(6):849–851. 10.1017/S1047951119001082. [Abstract] [CrossRef] [Google Scholar]
65. Khaybullina D, Patel A, Zerilli T. Riociguat (adempas): a novel agent for the treatment of pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. P T. 2014. Nov;39(11):749–58. [Europe PMC free article] [Abstract] [Google Scholar]
66. Garcia Aguilar H, Gorenflo M, Ivy DD, et al. Riociguat in children with pulmonary arterial hypertension: The PATENT-CHILD study. Pulm Circ. 2022. Jul;12(3):e12133. 10.1002/pul2.12133. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
67. Giesinger RE, Stanford AH, Thomas B, et al. Safety and Feasibility of Riociguat Therapy for the Treatment of Chronic Pulmonary Arterial Hypertension in Infancy. J Pediatr. 2023. Apr;255:224–229 e1. 10.1016/j.jpeds.2022.11.026. [Abstract] [CrossRef] [Google Scholar]
68. Hansmann G, Koestenberger M, Alastalo TP, et al. 2019 updated consensus statement on the diagnosis and treatment of pediatric pulmonary hypertension: The European Pediatric Pulmonary Vascular Disease Network (EPPVDN), endorsed by AEPC, ESPR and ISHLT. J Heart Lung Transplant. 2019. Sep;38(9):879–901. 10.1016/j.healun.2019.06.022. [Abstract] [CrossRef] [Google Scholar]**Of considerable interest due to inclusion of consensus guidelines with recommendations for treatment of pediatric pulmonary hypertension.
69. Lowe BS, Therrien J, Ionescu-Ittu R, et al. Diagnosis of pulmonary hypertension in the congenital heart disease adult population impact on outcomes. J Am Coll Cardiol. 2011. Jul 26;58(5):538–46. 10.1016/j.jacc.2011.03.033. [Abstract] [CrossRef] [Google Scholar]
70. Nemoto S, Sasaki T, Ozawa H, et al. Oral sildenafil for persistent pulmonary hypertension early after congenital cardiac surgery in children. Eur J Cardiothorac Surg. 2010. Jul;38(1):71–7. 10.1016/j.ejcts.2010.01.045. [Abstract] [CrossRef] [Google Scholar]
71. Wong J, Loomba RS, Evey L, et al. Postoperative Inhaled Nitric Oxide Does Not Decrease Length of Stay in Pediatric Cardiac Surgery Admissions. Pediatr Cardiol. 2019. Dec;40(8):1559–1568. 10.1007/s00246-019-02187-z. [Abstract] [CrossRef] [Google Scholar]
72. Landzberg MJ. Congenital heart disease associated pulmonary arterial hypertension. Clin Chest Med. 2007. Mar;28(1):243–53, x. 10.1016/j.ccm.2006.12.004. [Abstract] [CrossRef] [Google Scholar]
73. Suesaowalak M, Cleary JP, Chang AC. Advances in diagnosis and treatment of pulmonary arterial hypertension in neonates and children with congenital heart disease. World J Pediatr. 2010. Feb;6(1):13–31. 10.1007/s12519-010-0002-9. [Abstract] [CrossRef] [Google Scholar]
74. Rudolph AM. The changes in the circulation after birth. Their importance in congenital heart disease. Circulation. 1970. Feb;41(2):343–59. 10.1161/01.cir.41.2.343. [Abstract] [CrossRef] [Google Scholar]
75. Pedersen J, Hedegaard ER, Simonsen U, et al. Current and Future Treatments for Persistent Pulmonary Hypertension in the Newborn. Basic Clin Pharmacol Toxicol. 2018. Oct;123(4):392–406. 10.1111/bcpt.13051. [Abstract] [CrossRef] [Google Scholar]
76. Kelly LE, Ohlsson A, Shah PS. Sildenafil for pulmonary hypertension in neonates. Cochrane Database Syst Rev. 2017. Aug 4;8(8):CD005494. 10.1002/14651858.CD005494.pub4. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]*Of interest due to providing high level evidence for the efficacy of sildenafil for PH in infants.
77. Park BY, Chung SH. Treprostinil for persistent pulmonary hypertension of the newborn, with early onset sepsis in preterm infant: 2 Case reports. Medicine (Baltimore). 2017. Jun;96(26):e7303. 10.1097/MD.0000000000007303. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
78. Vijay Kumar JR, Natraj Setty HS, Jayaranganath M, et al. Efficacy, safety and tolerability of bosentan as an adjuvant to sildenafil and sildenafil alone in persistant pulmonary hypertension of newborn (PPHN). Interv Med Appl Sci. 2021. Aug;11(4):216–220. 10.1556/1646.2020.00004. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
79. Maneenil G, Thatrimontrichai A, Janjindamai W, et al. Effect of bosentan therapy in persistent pulmonary hypertension of the newborn. Pediatr Neonatol. 2018. Feb;59(1):58–64. 10.1016/j.pedneo.2017.02.003. [Abstract] [CrossRef] [Google Scholar]
80. Adnan M, Arshad MS, Anwar-Ul-Haq HM, et al. Persistent Pulmonary Hypertension of the Newborn: The Efficacy Comparison of Vasodilators Sildenafil Plus Bosentan Versus Sildenafil Plus Beraprost at a Tertiary Childcare Health Facility. Cureus. 2021. Nov;13(11):e20020. 10.7759/cureus.20020. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
81. O’Toole SJ, Irish MS, Holm BA, et al. Pulmonary Vascular Abnormalities in Congenital Diaphragmatic Hernia. Clinics in Perinatology. 1996;23(4):781–794. 10.1016/s0095-5108(18)30209-4. [Abstract] [CrossRef] [Google Scholar]
82. Yamataka T, Puri P. Pulmonary artery structural changes in pulmonary hypertension complicating congenital diaphragmatic hernia. J Pediatr Surg. 1997. Mar;32(3):387–90. 10.1016/s0022-3468(97)90587-x. [Abstract] [CrossRef] [Google Scholar]
83. Yang MJ, Russell KW, Yoder BA, et al. Congenital diaphragmatic hernia: a narrative review of controversies in neonatal management. Transl Pediatr. 2021. May;10(5):1432–1447. 10.21037/tp-20-142. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
84. Inhaled nitric oxide and hypoxic respiratory failure in infants with congenital diaphragmatic hernia. The Neonatal Inhaled Nitric Oxide Study Group (NINOS). Pediatrics. 1997. Jun;99(6):838–45. 10.1542/peds.99.6.838. [Abstract] [CrossRef] [Google Scholar]
85. Kinsella JP, Parker TA, Ivy DD, et al. Noninvasive delivery of inhaled nitric oxide therapy for late pulmonary hypertension in newborn infants with congenital diaphragmatic hernia. J Pediatr. 2003. Apr;142(4):397–401. 10.1067/mpd.2003.140. [Abstract] [CrossRef] [Google Scholar]
86. Bialkowski A, Moenkemeyer F, Patel N. Intravenous sildenafil in the management of pulmonary hypertension associated with congenital diaphragmatic hernia. Eur J Pediatr Surg. 2015. Apr;25(2):171–6. 10.1055/s-0033-1357757. [Abstract] [CrossRef] [Google Scholar]
87. Noori S, Friedlich P, Wong P, et al. Cardiovascular effects of sildenafil in neonates and infants with congenital diaphragmatic hernia and pulmonary hypertension. Neonatology. 2007;91(2):92–100. 10.1159/000097125. [Abstract] [CrossRef] [Google Scholar]
88. Patel N Use of milrinone to treat cardiac dysfunction in infants with pulmonary hypertension secondary to congenital diaphragmatic hernia: a review of six patients. Neonatology. 2012;102(2):130–6. 10.1159/000339108. [Abstract] [CrossRef] [Google Scholar]
89. Lakshminrusimha S, Keszler M, Kirpalani H, et al. Milrinone in congenital diaphragmatic hernia - a randomized pilot trial: study protocol, review of literature and survey of current practices. Matern Health Neonatol Perinatol. 2017;3:27. 10.1186/s40748-017-0066-9. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
90. Mous DS, Buscop-van Kempen MJ, Wijnen RMH, et al. Changes in vasoactive pathways in congenital diaphragmatic hernia associated pulmonary hypertension explain unresponsiveness to pharmacotherapy. Respir Res. 2017. Nov 7;18(1):187. 10.1186/s12931-017-0670-2. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
91. Shiyanagi S, Okazaki T, Shoji H, et al. Management of pulmonary hypertension in congenital diaphragmatic hernia: nitric oxide with prostaglandin-E1 versus nitric oxide alone. Pediatr Surg Int. 2008. Oct;24(10):1101–4. 10.1007/s00383-008-2225-6. [Abstract] [CrossRef] [Google Scholar]
92. Ellsworth KR, Ellsworth MA, Weaver AL, et al. Association of Early Inhaled Nitric Oxide With the Survival of Preterm Neonates With Pulmonary Hypoplasia. JAMA Pediatr. 2018. Jul 2;172(7):e180761. 10.1001/jamapediatrics.2018.0761. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
93. Geary C, Whitsett J. Inhaled nitric oxide for oligohydramnios-induced pulmonary hypoplasia: a report of two cases and review of the literature. J Perinatol. 2002. Jan;22(1):82–5. 10.1038/sj.jp.7210580. [Abstract] [CrossRef] [Google Scholar]

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.

Funding 


Funders who supported this work.

NHLBI NIH HHS (1)

National Heart, Lung, and Blood Institute (1)