Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Aug 16, 2025; 13(23): 107279
Published online Aug 16, 2025. doi: 10.12998/wjcc.v13.i23.107279
Off-label use of treprostinil in adult patients in clinical cases
Christina Mouratidou, Serafeim-Chrysovalantis Kotoulas, Eleni Mouloudi, Intensive Care Unit, Hippokration General Hospital, Thessaloniki 54642, Greece
Efstathios T Pavlidis, Ioannis N Galanis, Theodoros E Pavlidis, The 2nd Department of Propaedeutic Surgery, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
Georgios Katsanos, Georgios Tsoulfas, Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University of Thessaloniki, School of Medicine, Thessaloniki 54642, Greece
Ioannis A Taitzoglou, Laboratory of Physiology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
ORCID number: Christina Mouratidou (0009-0007-8657-2032); Efstathios T Pavlidis (0000-0002-7282-8101); Georgios Katsanos (0000-0002-5845-8175); Georgios Tsoulfas (0000-0001-5043-7962); Eleni Mouloudi (0000-0003-0079-2012); Ioannis A Taitzoglou (0000-0003-4897-7366); Ioannis N Galanis (0009-0001-4283-0788); Theodoros E Pavlidis (0000-0002-8141-1412).
Co-corresponding authors: Efstathios T Pavlidis and Theodoros E Pavlidis.
Author contributions: Pavlidis ET and Pavlidis TE contribute equally to this study as co-corresponding authors; Mouratidou C, Pavlidis ET, Mouloudi E, Taitzoglou IA, designed research and analysed data; Kotoulas SC, Tsoulfas G performed research contributed new analytic tools, evaluated data and review the paper; Katsanos G, Galanis IN contributed to data collection and analysed data; Pavlidis TE analysed data review and approved the paper.
Conflict-of-interest statement: There is no conflict of interest associated with any of the senior author or other coauthors contributed their efforts in this manuscript.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Theodoros E Pavlidis, MD, PhD, Professor, The 2nd Department of Propaedeutic Surgery, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, Konstantinoupoleos 49, Thessaloniki 54642, Greece. pavlidth@auth.gr
Received: March 20, 2025
Revised: April 3, 2025
Accepted: May 7, 2025
Published online: August 16, 2025
Processing time: 77 Days and 2 Hours

Abstract

Treprostinil is a relatively new tricyclic prostacyclin analog with a stable structure, extended half-life and improved potency. Currently, treprostinil is indicated by the Food and Drug Administration in the United States for the treatment of pulmonary arterial hypertension (group 1 in the pulmonary hypertension classification of the World Health Organization). It has a potent vasodilating effect along with the inhibition of platelet aggregation and the attenuation of the inflammatory response in pulmonary and systemic circulation. It is available in the following formulations: Subcutaneous, intravenous, inhaled and oral. Although unknown to many clinicians, several encouraging reports of off-label treprostinil use in the adult population suggest its potential effectiveness in other clinical conditions. Currently under investigation are digital ischemia secondary to systemic sclerosis, chronic limb ischemia, hepatic ischemia-reperfusion injury and group 3 and 4 pulmonary hypertension. Based on review and analysis of the available literature, this article provides a thorough update on the off-label use of treprostinil in adult patients.

Key Words: Treprostinil; Off-label use; Pulmonary hypertension; Digital ischemia; Chronic limb ischemia; Hepatic ischemia–reperfusion injury

Core Tip: Treprostinil is a synthetic prostacyclin analog with a stable structure that is currently used for the treatment of pulmonary arterial hypertension. By way of vasodilation and the inhibition of platelet aggregation, treprostinil increases the blood supply in pulmonary circulation and reduces the cardiac workload. In addition to its main indication, several positive reports of off-label treprostinil use in the adult population indicate its benefit in other clinical conditions. Hepatic ischemia–reperfusion injury, chronic limb ischemia, digital ischemia and group 3 and 4 pulmonary hypertension are currently under study as possible indications for treprostinil use outside its approved labeling.



INTRODUCTION

Treprostinil is a prostacyclin (PGI2) analog primarily approved by the Food and Drug Administration (FDA) in the United States for the treatment of pulmonary arterial hypertension (PAH). PAH is a subtype of pulmonary hypertension in which pulmonary artery remodeling results in right heart failure and death[1]. In addition to several supportive methods to offer symptomatic relief, several medical therapeutic options that target mainly endothelin receptors, nitric oxide–guanylyl cyclase and PGI2 signaling pathways exist. Treprostinil is a synthetic, chemically stable PGI2 analog with potent vasodilating and antithrombotic effects. In the pulmonary circulatory system, it causes relaxation of the pulmonary vasculature, reduces platelet aggregation, reverses pulmonary artery remodeling and decreases the proliferation rate of muscle cells through the activation of peroxisome proliferator-activated receptor γ[2]. Treprostinil is also known for its anti-inflammatory effect while reducing cytokine release and lipid peroxidation[3]. The main biochemical interactions of treprostinil are summarized in Table 1.

Table 1 Treprostinil mechanisms of action.
Number
Effect
1Vasodilation
2Inhibition of platelets aggregation
3Inhibition of platelets adherence
4Reversion of pulmonary remodelling
5Decrease of muscle cell proliferation rate
6Regulation of vascular homeostasis
7Attenuation of inflammatory response
8Upregulation of apoptosis rate
9Elevation of ATP production
10Preservation of mitochondrial biogenesis

Treprostinil is available via four possible routes of administration: Intravenous, subcutaneous, oral and inhaled. In vitro studies have shown high affinity for the prostaglandin DP1, EP2, and IP receptors, all of which activate the intracellular cyclic adenosine monophosphate signaling pathway[4,5]. Treprostinil has shown clinical effectiveness in the treatment of PAH by delaying disease progression. The simplified mechanisms of action of treprostinil are shown in Figure 1. However, several other clinical conditions appear to benefit from treprostinil administration under certain circumstances. In this mini-review, we discuss the off-label uses of treprostinil based on its mechanisms of action and the results of clinical studies.

Figure 1
Figure 1 Simplified mechanisms of actions of treprostinil. IP: Prostacyclin.
SYSTEMIC SCLEROSIS AND DIGITAL ISCHEMIA

Digital ischemia is a serious complication of systemic sclerosis, resulting in significant morbidity and gangrene, necessitating amputation in some cases. The activation of innate immune responses, progressive fibrosis and the development of vasculopathy constitute some of the causative mechanisms, but the pathogenesis of systemic sclerosis is not completely understood[6]. Peripheral altered vascular reactivity presents as a common evolution of the underlying disease, resulting in Raynaud’s phenomenon. At the early stages of the disease, Raynaud’s phenomenon is characterized by pallor and cyanosis followed by hyperemia of the digits due to symmetrical transient vascular events triggered by exposure to low temperature or emotional stress. Recurrent episodes of digital ischemia can result in digital ulcers with the loss of continuous epithelial coverage. However, a prolonged reduction in digital perfusion can cause critical damage to tissues with impaired endurance[7,8]. The etiology of the ischemic events includes a combination of intimal hyperplasia, episodes of vasospasm and in situ thrombosis[9]. Among other therapeutic options to enhance peripheral microcirculation, PGI2 analogs have improved microvascular arterial flow and velocity, remission of digital ulcers and attenuation of ischemic pain. Treprostinil has been used in a small population of patients with systemic sclerosis to improve peripheral blood flow and prevent or heal digital ulcers. Continuous subcutaneous treprostinil infusion resulted in complete resolution of baseline digital ulcers in most of these patients. Additionally, no new lesions were observed during continuous therapy, suggesting the important role of treprostinil in preventing the development of new ulcers. However, injection site reactions and diarrhea were observed in some patients, resulting in drug discontinuation in 5 patients. The high rate of adverse events suggests that the use of treprostinil in particular formulations is challenging and indicates an urgent need for larger studies to determine the route, length and dose of therapy[10,11]. The administration of treprostinil as an oral sustained-release osmotic tablet (treprostinil diethanolamine) was associated with improved digital perfusion and mild or moderate adverse events possibly related to the study drug following dose escalation. In detail, the plasma drug concentrations of oral doses were similar to those reported during continuous infusion. Moreover, significant improvement in perfusion was observed after treprostinil administration, as assessed by laser Doppler imaging. Finally, digital skin temperature was positively associated with the drug plasma concentration[12].

Treprostinil administration via cathodal iontophoresis enables local noninvasive transdermal delivery of the drug, avoiding systemic side effects (Figure 2). Recent studies have demonstrated improvements in skin vascular blood flux, with an extended duration of the effect even after the procedure and a very low rate of digital ulcer recurrence. Noninvasive laser speckle contrast imaging was used to determine the effectiveness of local therapy, but no systemic or local side effects were described[13-15]. Overall, recent reports have shown favorable effects of locally delivered treprostinil in patients with systemic sclerosis and digital ischemia, but larger clinical studies are needed.

Figure 2
Figure 2 Schematic representation of iontophoretic treprostinil delivery.
CHRONIC LIMB ISCHEMIA

Chronic limb-threatening ischemia is a severe peripheral vascular disease caused by progressive occlusion of the arterial branches of the lower extremities. The development of atherosclerotic plaques leads to advanced arterial stenosis with a reduced blood supply, hypoxia, reactive oxygen species formation, oxidative stress, arteriolar vasospasm, platelet aggregation, leucocyte activation and inflammation, microthrombosis, microcirculatory disfunction and muscle cell necrosis[16]. In the later stages of the disease, the degeneration of blood vessels leads to the formation of nonhealing ulcers, necrotic lesions and/or gangrene[17,18]. Bypass surgical techniques and endovascular procedures are the gold standard strategies that aim to restore the blood supply, but not all patients are suitable for these therapies due to their comorbidities or advanced age. Therefore, a number of pharmacological agents have been used in patients with critical limb ischemia and microcirculatory dysfunction who do not meet the criteria for revascularization[19].

During the past thirty years, PGI2 analogs have been studied for the management of patients in the early stages of peripheral arterial disease and critical limb ischemia[20]. Based on acknowledged mechanisms of action and previous reports, Berman et al[21] conducted an open-label, single-center pilot study to assess the safety and effectiveness of treprostinil administration in patients with critical limb ischemia and no planned revascularization. Each patient with Fontaine stage III to IV (Rutherford class 4-6) peripheral arterial disease received an escalated dose of treprostinil via continuous subcutaneous infusion for 12 weeks. All patients reported improvement in pain, while a positive effect on ischemic lesion healing was observed. Moreover, treprostinil is considered a safe and well-tolerated pharmacological agent in these particular populations.

HEPATIC ISCHEMIA–REPERFUSION INJURY

Hepatic ischemia–reperfusion injury is a common pathophysiological event that usually follows major surgical procedures for liver and liver transplantation. Severe inflammatory reactions and oxidative stress during reperfusion of ischemic hepatic tissue lead to extreme tissue damage and cell death with widespread local and systemic consequences[22-24]. The molecular mechanisms responsible for cell damage are exceedingly complicated; therefore, various types of pharmacological interventions and antioxidant agents have been used to intervene in the different phases of hepatic ischemia–reperfusion injury to suppress this phenomenon[25].

A shortage of organs suitable for transplantation has resulted in a major organ shortage crisis and necessitated the use of extended-criteria donor livers, which are more susceptible to ischemia–reperfusion injury[26]. For nearly forty years, numerous studies have investigated the advantages of PGI2 analog administration in hepatic ischemia–reperfusion injury, with rather encouraging results.

In an orthotopic rat liver transplant model, treprostinil administration protected liver grafts from ischemia–reperfusion injury by restoring energy levels, reducing platelet aggregation and preserving the blood supply[27]. Treprostinil has also been used in adult liver transplant patients in a prospective, single-center, nonrandomized, interventional study to assess the safety and effectiveness of the study drug in the aforementioned population. Patients who underwent liver transplantation received treprostinil at a dose of 5 ng/kg/minute for 2 days or 2.5, 5, or 7.5 ng/ minute/kg for 5 days as a continuous infusion. With no reported cases of primary graft nonfunction, treprostinil infusion up to 5 ng/ minute/kg for 120 hours was safe and well tolerated. Furthermore, the authors reported a minimized need for ventilation support, reduced hospitalization time, enhanced hepatobiliary excretory function and 100% graft and patient survival[28,29].

OTHER SUBTYPES OF PULMONARY HYPERTENSION
Pulmonary hypertension in interstitial lung disease

Interstitial lung disease is a heterogeneous group of diffuse parenchymal lung disorders that are characterized primarily by fibrosis of the lung parenchyma. Up to 86% of patients with interstitial lung disease have been shown to develop pulmonary hypertension with a higher demand for supplementary oxygen, decreased quality of life and worse outcomes[30,31]. In the pulmonary hypertension classification of the World Health Organization (WHO), pulmonary hypertension due to lung disease is classified into group 3 (Table 2). Regardless of the widespread occurrence worldwide and the progressive nature of the disease, there are currently no approved therapies for these patients.

Table 2 World Health Classification of pulmonary hypertension and treprostinil indications.
Group
Indication
Treprostinil use status
Group 1Pulmonary arterial hypertensionApproved
Group 2Pulmonary hypertension due to left heart diseaseHas not been tested
Group 3Pulmonary hypertension associated with lung disordersOff-label use
Group 4Pulmonary hypertension due to pulmonary artery obstructionsOff-label use
Group 5Miscellaneous (unclear or multifactorial mechanisms)Has not been tested

Based on data that PGI2 analogs improve exercise capacity in patients with PAH, vasodilator therapies have been used to treat group 3 patients[32-34]. Although not originally approved for this purpose, treprostinil has shown efficacy in treating pulmonary hypertension associated with interstitial lung disease. Patients treated with inhaled treprostinil had significant improvements in exercise capacity and increased forced vital capacity at 16 weeks[35-37]. The exacerbation of the underlying lung disease rate was significantly lower in the treprostinil group than in the placebo group, whereas no statistically significant differences in serious adverse events were detected between the drug and placebo groups.

Chronic thromboembolic pulmonary hypertension

The pulmonary hypertension classification of the WHO categorizes chronic thromboembolic pulmonary hypertension (CTEPH) into group 4. Chronic obstruction of the pulmonary vascular bed by nonresolving organized thromboemboli causes increased resistance in pulmonary circulation, right ventricular overload and progressive right heart failure[38]. The thromboembolic material remains resistant for at least three months of uninterrupted anticoagulation therapy. To this degree, pulmonary endarterectomy is the treatment of choice for these patients; however, not all individuals are ideal candidates for surgical intervention, mainly due to significant distal small vessel disease[39]. Although riociguat is the only pharmacological agent approved for treating CTEPH, treprostinil has been explored as an alternative therapeutic option to improve symptoms in inoperable patients or those with residual postoperative disease[40,41].

Published data on the effects of long-term treprostinil therapy (> 2 years) demonstrated that long-term treatment ameliorated hemodynamic measurements and improved survival[42]. Patients with inoperable CTEPH (distal small vessel disease, resistant or recurrent pulmonary hypertension after endarterectomy, severe comorbidities) and typical abnormal pulmonary angiography findings received continuous subcutaneous treprostinil at an adjusted dose with the use of a positive pressure microinfusion pump. Exercise capacity was significantly improved in patients treated with treprostinil. Moreover, B-type natriuretic peptide plasma levels, pulmonary resistance, cardiac output and right ventricular end-diastolic pressure were also improved. Although the mean pulmonary arterial pressure was reduced, the difference was not statistically significant. The study also confirmed the successful management of local pain at the infusion site and maintained efficacy over an extended period of time.

Idiopathic pulmonary fibrosis with pulmonary hypertension

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by progressive lung fibrosis, ongoing respiratory failure and premature mortality[43]. Pulmonary hypertension is common in patients with IPF and is classified as Group 3 according to the WHO classification[44]. As mentioned above, there are limited pharmacological therapeutic options available for these patients, but lung transplantation constitutes the optimal treatment[45].

An inhaled treprostinil formulation has been considered to improve symptoms and exercise capacity in patients with pulmonary hypertension secondary to IPF. The TETON program, an ongoing phase III clinical trial, assesses the long-term safety and tolerability of inhaled treprostinil in patients with a diagnosis of IPF[46]. Based on the combination of preclinical evidence of treprostinil antifibrotic activity and results from previous studies, the TETON program aims to study changes in absolute forced vital capacity and overall survival at week 52 in IPF patients who received inhaled treprostinil.

STRATEGIES FOR FUTURE STUDIES ON TREPROSTINIL OFF-LABEL USE

A number of reports have investigated the use of treprostinil beyond its approved labeling, but clinical evidence remains limited. Treprostinil has a known side effect profile consisting of coughing, headache, nausea, diarrhea, hypotension, flushing, jaw pain and extremity pain, which may be hard to manage in an off-label context[47]. Furthermore, prescribing high-cost treatment involves negotiating a complex regulatory framework, as its unapproved administration is not supported by the latest clinical guidelines[48]. The lack of established protocols may also be challenging, as interruptions in treprostinil therapy or difficulty in reaching the optimal dose can lead to negative outcomes[49]. The proposed treprostinil dosing schemes for selected diseases according to current data, which demonstrate the efficacy of treprostinil, are shown in Table 3. The use of treprostinil for other clinical conditions requires extensive research, potentially prolonged and expensive processes, without any guarantee of positive results.

Table 3 Proposed treprostinil dosing schemes in selected diseases according to current data, which demonstrated the efficacy of treprostinil.
Ref.
Type of study
Disease
Number of patients
Dosage
Follow-up
Results
Engel et al[10], 2005Case reportDigital necrosis in systemic sclerosis115 ng/kg/min
subcutaneous infusion for 16 weeks
1 yearHealing of lesions
Chung et al[11], 2006Open-label, single center trialDigital ulcers in systemic sclerosis1215 ng/kg/min
subcutaneous infusion for 12 weeks
8 weeksResolution of baseline digital ulcers
Shah et al[12], 2013Open-label, dual center, phase I pharmacokinetic studyDigital ulcers secondary to scleroderma204 mg BID per os for 8 weeks8 weeksIncrease in digital perfusion
Hellmann et al[13], 2015Double-blind randomized pharmacology studyDiabetic ulcers240,1 mg/mL solution for iontophoresis-Increase in microvascular blood flux in the malleolus area
Roustit et al[14], 2014Randomized controlled trialDigital ulcers in systemic sclerosis12Iontophoresis at 240 mC/cm2-Increase in digital skin perfusion
Gaillard-Bigot et al[15], 2016Prospective studyDigital ulcers in systemic sclerosis11Simultaneous double-blinded iontophoresis (0.1 mg/mL)-Increase in skin blood flow
Berman et al[21], 2006Open-label, single-center pilot studyCritical limb ischemia1015 ng/kg/min
subcutaneous infusion
12 weeksReduction in ischemic rest pain
Almazroo O et al[28], 2021Prospective, single-center, non-randomized, interventional studyIschemia – reperfusion injury in adult liver transplantation352.5, 5, 7.5 ng/kg/min in continuous IV infusion for 120 hours180 daysNo primary graft non-function, minimized need for ventilation support, reduced hospitalization time, 100% graft and patient survival
Nathan et al[36], 2021Multicenter, randomized, double-blind, placebo-controlled trialPulmonary hypertension due to interstitial lung disease32672 μg inhaled treprostinil 16 weeksImprovement of exercise capacity
Sadushi-Kolici and Lang[41], 2019Multicenter, randomised, double-blind controlled trialChronic thromboembolic pulmonary hypertension1053 ng/kg/min or 30 ng/kg/min subcoutaneous treprostinil for 12 weeks24 weeksImprovement of exercise capacity
Skoro-Sajer et al, 2007[42]Open-label uncontrolled study252 ng/kg/minute continuous subcutaneous infusion for 12 weeks24 ± 18 monthsImprovement of exercise capacity, haemodynamics and survival
Nathan et al, 2022[46]2 randomised, double-blind placebo-controlled, phase 3 studiesIdiopathic pulmonary fibrosis79272 μg inhaled treprostinil52 weeksOngoing study

Although treprostinil has high potential for off-label use, further research is necessary to ensure patient safety and therapeutic efficacy. Future studies should follow methodized strategies to provide scientific accuracy and clinical implementation. Preclinical studies using cellular and animal models should provide detailed information on the vasodilating, anti-inflammatory, antithrombotic and antifibrotic effects of treprostinil, along with dosing and toxicity levels. Small-scale and large-scale randomized controlled trials could determine the effectiveness of off-label treprostinil compared with standard treatment in certain groups of patients. To achieve that, approved criteria for primary and secondary outcomes should be established, such as signs of clinical improvement, reduction in inflammatory or fibrotic markers, formulation of quality-of-life scores and development of long-term disease progression markers. Overcoming the lack of scientific data, funding limitations and regulatory considerations, along with securing FDA guidance for drug repurposing, will become key to expanding treprostinil clinical applications[50].

Additionally, there are limitations regarding the off-label use of treprostinil, including its cost, the risk of complications, and the lack of standardized protocols[47-49]. Therefore, the decision to use this drug should be made based on the best possible treatment for each patient. However, this article opens new horizons for both clinical and experimental preclinical studies, which are necessary for drawing safe conclusions. The above limitations of the research should be indicated in future research studies.

CONCLUSION

Off-label treprostinil administration includes the use of the medication for medical conditions beyond its primary indication in a certain unapproved group of patients. Decision-making is often based on known pharmacological mechanisms and evidence from clinical studies. Current evidence-based indications for off-label treprostinil use are digital ischemia secondary to systemic sclerosis, chronic limb ischemia, hepatic ischemia–reperfusion injury and group 3 and 4 pulmonary hypertension. Treprostinil unapproved use provides alternative options to patients who have exhausted the standard choices of treatment. The present knowledge demonstrates the safety, efficacy and tolerability of treprostinil use. However, clinicians should consider the possibility of a favorable benefit–risk ratio for each patient separately. More clinical trials need to be conducted on larger populations to support the use of treprostinil in the discussed clinical conditions to gain approval.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country of origin: Greece

Peer-review report’s classification

Scientific Quality: Grade B, Grade B

Novelty: Grade B, Grade B

Creativity or Innovation: Grade B, Grade B

Scientific Significance: Grade B, Grade B

P-Reviewer: Mogulkoc R S-Editor: Lin C L-Editor: A P-Editor: Xu ZH

References
1.  Feldman J, Habib N, Fann J, Radosevich JJ. Treprostinil in the treatment of pulmonary arterial hypertension. Future Cardiol. 2020;16:547-558.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
2.  Maron BA, Abman SH, Elliott CG, Frantz RP, Hopper RK, Horn EM, Nicolls MR, Shlobin OA, Shah SJ, Kovacs G, Olschewski H, Rosenzweig EB. Pulmonary Arterial Hypertension: Diagnosis, Treatment, and Novel Advances. Am J Respir Crit Care Med. 2021;203:1472-1487.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 101]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
3.  Lindegaard Pedersen M, Krüger M, Grimm D, Infanger M, Wehland M. The prostacyclin analogue treprostinil in the treatment of pulmonary arterial hypertension. Basic Clin Pharmacol Toxicol. 2020;126:32-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 28]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
4.  Kumar P, Thudium E, Laliberte K, Zaccardelli D, Nelsen A. A Comprehensive Review of Treprostinil Pharmacokinetics via Four Routes of Administration. Clin Pharmacokinet. 2016;55:1495-1505.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 32]  [Cited by in RCA: 55]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
5.  Whittle BJ, Silverstein AM, Mottola DM, Clapp LH. Binding and activity of the prostacyclin receptor (IP) agonists, treprostinil and iloprost, at human prostanoid receptors: treprostinil is a potent DP1 and EP2 agonist. Biochem Pharmacol. 2012;84:68-75.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 95]  [Cited by in RCA: 127]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
6.  Galluccio F, Matucci-Cerinic M. Registry evaluation of digital ulcers in systemic sclerosis. Int J Rheumatol. 2010;2010:363679.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
7.  Nihtyanova SI, Brough GM, Black CM, Denton CP. Clinical burden of digital vasculopathy in limited and diffuse cutaneous systemic sclerosis. Ann Rheum Dis. 2008;67:120-123.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 121]  [Cited by in RCA: 98]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
8.  Herrick AL, Wigley FM. Raynaud's phenomenon. Best Pract Res Clin Rheumatol. 2020;34:101474.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 49]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
9.  Romero-Karam LA, Honan KA, Arain SA, Mayes MD. Percutaneous revascularization for the treatment of refractory digital ischemia in systemic sclerosis. J Scleroderma Relat Disord. 2023;8:36-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
10.  Engel G, Rockson SG. Treprostinil for the treatment of severe digital necrosis in systemic sclerosis. Vasc Med. 2005;10:29-32.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
11.  Chung L, Fiorentino D. A pilot trial of treprostinil for the treatment and prevention of digital ulcers in patients with systemic sclerosis. J Am Acad Dermatol. 2006;54:880-882.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 45]  [Cited by in RCA: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
12.  Shah AA, Schiopu E, Hummers LK, Wade M, Phillips K, Anderson C, Wise R, Boin F, Seibold JR, Wigley F, Rollins KD. Open label study of escalating doses of oral treprostinil diethanolamine in patients with systemic sclerosis and digital ischemia: pharmacokinetics and correlation with digital perfusion. Arthritis Res Ther. 2013;15:R54.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
13.  Hellmann M, Roustit M, Gaillard-Bigot F, Cracowski JL. Cutaneous iontophoresis of treprostinil, a prostacyclin analog, increases microvascular blood flux in diabetic malleolus area. Eur J Pharmacol. 2015;758:123-128.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 14]  [Cited by in RCA: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
14.  Roustit M, Gaillard-Bigot F, Blaise S, Stanke-Labesque F, Cracowski C, Seinturier C, Jourdil JF, Imbert B, Carpentier PH, Cracowski JL. Cutaneous iontophoresis of treprostinil in systemic sclerosis: a proof-of-concept study. Clin Pharmacol Ther. 2014;95:439-445.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
15.  Gaillard-Bigot F, Roustit M, Blaise S, Cracowski C, Seinturier C, Imbert B, Carpentier P, Cracowski JL. Treprostinil Iontophoresis Improves Digital Blood Flow during Local Cooling in Systemic Sclerosis. Microcirculation. 2016;23:266-270.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
16.  Marsico G, Martin-Saldaρa S, Pandit A. Therapeutic Biomaterial Approaches to Alleviate Chronic Limb Threatening Ischemia. Adv Sci (Weinh). 2021;8:2003119.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
17.  Rajagopalan S, Grossman PM. Management of chronic critical limb ischemia. Cardiol Clin. 2002;20:535-545.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 11]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
18.  Management of peripheral arterial disease (PAD). TransAtlantic Inter-Society Consensus (TASC). Int Angiol. 2000;19:I-XXIV, 1.  [PubMed]  [DOI]
19.  Mangiafico RA, Mangiafico M. Medical treatment of critical limb ischemia: current state and future directions. Curr Vasc Pharmacol. 2011;9:658-676.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 26]  [Cited by in RCA: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
20.  Dormandy JA. Prostanoid drug therapy for peripheral arterial occlusive disease--the European experience. Vasc Med. 1996;1:155-158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 21]  [Cited by in RCA: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
21.  Berman S, Quick R, Yoder P, Voigt S, Strootman D, Wade M. Treprostinil sodium (Remodulin), a prostacyclin analog, in the treatment of critical limb ischemia: open-label study. Vascular. 2006;14:142-148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
22.  Yu B, Zhang Y, Wang T, Guo J, Kong C, Chen Z, Ma X, Qiu T. MAPK Signaling Pathways in Hepatic Ischemia/Reperfusion Injury. J Inflamm Res. 2023;16:1405-1418.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
23.  Luo L, Mo G, Huang D. Ferroptosis in hepatic ischemiareperfusion injury: Regulatory mechanisms and new methods for therapy (Review). Mol Med Rep. 2021;23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
24.  Nastos C, Kalimeris K, Papoutsidakis N, Tasoulis MK, Lykoudis PM, Theodoraki K, Nastou D, Smyrniotis V, Arkadopoulos N. Global consequences of liver ischemia/reperfusion injury. Oxid Med Cell Longev. 2014;2014:906965.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 155]  [Cited by in RCA: 223]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
25.  George J, Lu Y, Tsuchishima M, Tsutsumi M. Cellular and molecular mechanisms of hepatic ischemia-reperfusion injury: The role of oxidative stress and therapeutic approaches. Redox Biol. 2024;75:103258.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 19]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
26.  Saidi RF, Hejazii Kenari SK. Challenges of organ shortage for transplantation: solutions and opportunities. Int J Organ Transplant Med. 2014;5:87-96.  [PubMed]  [DOI]
27.  Ghonem N, Yoshida J, Stolz DB, Humar A, Starzl TE, Murase N, Venkataramanan R. Treprostinil, a prostacyclin analog, ameliorates ischemia-reperfusion injury in rat orthotopic liver transplantation. Am J Transplant. 2011;11:2508-2516.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 21]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
28.  Almazroo OA, Miah MK, Pillai VC, Shaik IH, Xu R, Dharmayan S, Johnson HJ, Ganesh S, Planinsic RM, Demetris AJ, Al-Khafaji A, Lopez R, Molinari M, Tevar AD, Hughes C, Humar A, Venkataramanan R. An evaluation of the safety and preliminary efficacy of peri- and post-operative treprostinil in preventing ischemia and reperfusion injury in adult orthotopic liver transplant recipients. Clin Transplant. 2021;35:e14298.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
29.  Almazroo OA, Shaik IH, Hughes CB, Humar A, Venkataramanan R. Treprostinil Supplementation Ameliorates Hepatic Ischemia Reperfusion Injury and Regulates Expression of Hepatic Drug Transporters: An Isolated Perfused Rat Liver (IPRL) Study. Pharm Res. 2022;39:2979-2990.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
30.  Cerro Chiang G, Parimon T. Understanding Interstitial Lung Diseases Associated with Connective Tissue Disease (CTD-ILD): Genetics, Cellular Pathophysiology, and Biologic Drivers. Int J Mol Sci. 2023;24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 26]  [Reference Citation Analysis (0)]
31.  King CS, Shlobin OA. The Trouble With Group 3 Pulmonary Hypertension in Interstitial Lung Disease: Dilemmas in Diagnosis and the Conundrum of Treatment. Chest. 2020;158:1651-1664.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 59]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
32.  Wang L, Jin YZ, Zhao QH, Jiang R, Wu WH, Gong SG, He J, Liu JM, Jing ZC. Hemodynamic and gas exchange effects of inhaled iloprost in patients with COPD and pulmonary hypertension. Int J Chron Obstruct Pulmon Dis. 2017;12:3353-3360.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
33.  Faria-Urbina M, Oliveira RKF, Agarwal M, Waxman AB. Inhaled Treprostinil in Pulmonary Hypertension Associated with Lung Disease. Lung. 2018;196:139-146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 20]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
34.  McLaughlin VV, Benza RL, Rubin LJ, Channick RN, Voswinckel R, Tapson VF, Robbins IM, Olschewski H, Rubenfire M, Seeger W. Addition of inhaled treprostinil to oral therapy for pulmonary arterial hypertension: a randomized controlled clinical trial. J Am Coll Cardiol. 2010;55:1915-1922.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 359]  [Cited by in RCA: 391]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
35.  Behr J. Inhaled Treprostinil in Pulmonary Hypertension in the Context of Interstitial Lung Disease: A Success, Finally. Am J Respir Crit Care Med. 2022;205:144-145.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
36.  Nathan SD, Waxman A, Rajagopal S, Case A, Johri S, DuBrock H, De La Zerda DJ, Sahay S, King C, Melendres-Groves L, Smith P, Shen E, Edwards LD, Nelsen A, Tapson VF. Inhaled treprostinil and forced vital capacity in patients with interstitial lung disease and associated pulmonary hypertension: a post-hoc analysis of the INCREASE study. Lancet Respir Med. 2021;9:1266-1274.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 80]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
37.  Waxman A, Restrepo-Jaramillo R, Thenappan T, Ravichandran A, Engel P, Bajwa A, Allen R, Feldman J, Argula R, Smith P, Rollins K, Deng C, Peterson L, Bell H, Tapson V, Nathan SD. Inhaled Treprostinil in Pulmonary Hypertension Due to Interstitial Lung Disease. N Engl J Med. 2021;384:325-334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 162]  [Cited by in RCA: 355]  [Article Influence: 88.8]  [Reference Citation Analysis (0)]
38.  Teerapuncharoen K, Bag R. Chronic Thromboembolic Pulmonary Hypertension. Lung. 2022;200:283-299.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
39.  Coulden R. State-of-the-art imaging techniques in chronic thromboembolic pulmonary hypertension. Proc Am Thorac Soc. 2006;3:577-583.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 75]  [Cited by in RCA: 69]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
40.  Ghofrani HA, Gomez Sanchez MA, Humbert M, Pittrow D, Simonneau G, Gall H, Grόnig E, Klose H, Halank M, Langleben D, Snijder RJ, Escribano Subias P, Mielniczuk LM, Lange TJ, Vachiιry JL, Wirtz H, Helmersen DS, Tsangaris I, Barberα JA, Pepke-Zaba J, Boonstra A, Rosenkranz S, Ulrich S, Steringer-Mascherbauer R, Delcroix M, Jansa P, Šimková I, Giannakoulas G, Klotsche J, Williams E, Meier C, Hoeper MM; NEW COLLABORATORS LIST. Riociguat treatment in patients with chronic thromboembolic pulmonary hypertension: Final safety data from the EXPERT registry. Respir Med. 2021;178:106220.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
41.  Sadushi-Kolici R, Lang IM. Treprostinil for the treatment of chronic thromboembolic pulmonary hypertension. Expert Rev Respir Med. 2019;1-7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
42.  Skoro-Sajer N, Bonderman D, Wiesbauer F, Harja E, Jakowitsch J, Klepetko W, Kneussl MP, Lang IM. Treprostinil for severe inoperable chronic thromboembolic pulmonary hypertension. J Thromb Haemost. 2007;5:483-489.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 102]  [Cited by in RCA: 97]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
43.  Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, Behr J, Cottin V, Danoff SK, Morell F, Flaherty KR, Wells A, Martinez FJ, Azuma A, Bice TJ, Bouros D, Brown KK, Collard HR, Duggal A, Galvin L, Inoue Y, Jenkins RG, Johkoh T, Kazerooni EA, Kitaichi M, Knight SL, Mansour G, Nicholson AG, Pipavath SNJ, Buendνa-Roldαn I, Selman M, Travis WD, Walsh S, Wilson KC; American Thoracic Society, European Respiratory Society, Japanese Respiratory Society, and Latin American Thoracic Society. Diagnosis of Idiopathic Pulmonary Fibrosis. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am J Respir Crit Care Med. 2018;198:e44-e68.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1705]  [Cited by in RCA: 2617]  [Article Influence: 436.2]  [Reference Citation Analysis (0)]
44.  Collum SD, Amione-Guerra J, Cruz-Solbes AS, DiFrancesco A, Hernandez AM, Hanmandlu A, Youker K, Guha A, Karmouty-Quintana H. Pulmonary Hypertension Associated with Idiopathic Pulmonary Fibrosis: Current and Future Perspectives. Can Respir J. 2017;2017:1430350.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 41]  [Cited by in RCA: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
45.  Copeland CR, Lancaster LH. Management of Progressive Fibrosing Interstitial Lung Diseases (PF-ILD). Front Med (Lausanne). 2021;8:743977.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
46.  Nathan SD, Behr J, Cottin V, Lancaster L, Smith P, Deng CQ, Pearce N, Bell H, Peterson L, Flaherty KR. Study design and rationale for the TETON phase 3, randomised, controlled clinical trials of inhaled treprostinil in the treatment of idiopathic pulmonary fibrosis. BMJ Open Respir Res. 2022;9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 31]  [Reference Citation Analysis (0)]
47.  Brewer J, Wilson M, Coons JC, Schmit A, Whittenhall ME, Kimber A, Broderick M, Lee D, Patzlaff N, Miller C, Ataya A, LaRoy V, King CS, Ravichandran AK, Kingrey JF, Sahay S. Practical management of oral treprostinil in patients with pulmonary arterial hypertension: Lessons from ADAPT, EXPEDITE, and expert consensus. Respir Med. 2024;231:107734.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
48.  Epstein RS, Huang SM. The many sides of off-label prescribing. Clin Pharmacol Ther. 2012;91:755-758.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
49.  Narechania S, Torbic H, Tonelli AR. Treatment Discontinuation or Interruption in Pulmonary Arterial Hypertension. J Cardiovasc Pharmacol Ther. 2020;25:131-141.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
50.  Corsello SM, Bittker JA, Liu Z, Gould J, McCarren P, Hirschman JE, Johnston SE, Vrcic A, Wong B, Khan M, Asiedu J, Narayan R, Mader CC, Subramanian A, Golub TR. The Drug Repurposing Hub: a next-generation drug library and information resource. Nat Med. 2017;23:405-408.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 437]  [Cited by in RCA: 620]  [Article Influence: 103.3]  [Reference Citation Analysis (0)]