Abstract
2022 ESC/ERS pulmonary hypertension guidelines incorporate changes and adaptations focusing on clinical management https://bit.ly/3QtUvb4
Table of contents
1. Preamble 7
2. Introduction 8
2.1. What is new 8
2.2. Methods 18
3. Definitions and classifications 19
3.1. Definitions 19
3.2. Classifications 20
4. Epidemiology and risk factors 21
4.1. Group 1, pulmonary arterial hypertension 21
4.2. Group 2, pulmonary hypertension associated with left heart disease 23
4.3. Group 3, pulmonary hypertension associated with lung diseases and/or hypoxia 23
4.4. Group 4, pulmonary hypertension associated with chronic pulmonary artery obstruction 23
4.5. Group 5, pulmonary hypertension with unclear and/or multifactorial mechanisms 23
5. Pulmonary hypertension diagnosis 23
5.1. Diagnosis 23
5.1.1. Clinical presentation 23
5.1.2. Electrocardiogram 23
5.1.3. Chest radiography 24
5.1.4. Pulmonary function tests and arterial blood gases 24
5.1.5. Echocardiography 25
5.1.6. Ventilation/perfusion lung scan 26
5.1.7. Non-contrast and contrast-enhanced chest computed tomography examinations, and digital subtraction angiography 27
5.1.8. Cardiac magnetic resonance imaging 28
5.1.9. Blood tests and immunology 29
5.1.10. Abdominal ultrasound 29
5.1.11. Cardiopulmonary exercise testing 29
5.1.12. Right heart catheterization, vasoreactivity, exercise, and fluid challenge 29
5.1.12.1. Right heart catheterization 29
5.1.12.2. Vasoreactivity testing 30
5.1.12.3. Exercise right heart catheterization 30
5.1.12.4. Fluid challenge 30
5.1.13. Genetic counselling and testing 31
5.2. Diagnostic algorithm 31
5.2.1 Step 1 (suspicion) 31
5.2.2. Step 2 (detection) 31
5.2.3. Step 3 (confirmation) 33
5.3. Screening and early detection 35
5.3.1. Systemic sclerosis 36
5.3.2. BMPR2 mutation carriers 36
5.3.3. Portal hypertension 36
5.3.4. Pulmonary embolism 36
6. Pulmonary arterial hypertension (group 1) 38
6.1. Clinical characteristics 38
6.2. Severity and risk assessment 38
6.2.1. Clinical parameters 38
6.2.2. Imaging 39
6.2.2.1. Echocardiography 39
6.2.2.2. Cardiac magnetic resonance imaging 39
6.2.3. Haemodynamics 39
6.2.4. Exercise capacity 40
6.2.5. Biochemical markers 41
6.2.6. Patient-reported outcome measures 41
6.2.7. Comprehensive prognostic evaluation, risk assessment, and treatment goals 42
6.3. Therapy 43
6.3.1. General measures 43
6.3.1.1. Physical activity and supervised rehabilitation 43
6.3.1.2. Anticoagulation 43
6.3.1.3. Diuretics 44
6.3.1.4. Oxygen 44
6.3.1.5. Cardiovascular drugs 44
6.3.1.6. Anaemia and iron status 44
6.3.1.7. Vaccination 44
6.3.1.8. Psychosocial support 44
6.3.1.9. Adherence to treatments 44
6.3.2. Special circumstances 45
6.3.2.1. Pregnancy and birth control 45
6.3.2.1.1. Pregnancy 45
6.3.2.1.2. Contraception 45
6.3.2.2. Surgical procedures 45
6.3.2.3. Travel and altitude 45
6.3.3. Pulmonary arterial hypertension therapies 46
6.3.3.1. Calcium channel blockers 46
6.3.3.2. Endothelin receptor antagonists 47
6.3.3.2.1. Ambrisentan 48
6.3.3.2.2. Bosentan 48
6.3.3.2.3. Macitentan 49
6.3.3.3. Phosphodiesterase 5 inhibitors and guanylate cyclase stimulators 49
6.3.3.3.1. Sildenafil 50
6.3.3.3.2. Tadalafil 50
6.3.3.3.3. Riociguat 50
6.3.3.4. Prostacyclin analogues and prostacyclin receptoragonists 50
6.3.3.4.1. Epoprostenol 50
6.3.3.4.2. Iloprost 50
6.3.3.4.3. Treprostinil 50
6.3.3.4.4. Beraprost 50
6.3.3.4.5. Selexipag 50
6.3.4. Treatment strategies for patients with idiopathic, heritable, drug-associated, or connective tissue disease-associated pulmonary arterial hypertension 50
6.3.4.1. Initial treatment decision in patients without cardiopulmonary comorbidities 51
6.3.4.2. Treatment decisions during follow-up in patients without cardiopulmonary comorbidities 52
6.3.4.3. Pulmonary arterial hypertension with cardiopulmonary comorbidities 53
6.3.5. Drug interactions 54
6.3.6. Interventional therapy 54
6.3.6.1. Balloon atrial septostomy and Potts shunt 54
6.3.6.2. Pulmonary artery denervation 54
6.3.7. Advanced right ventricular failure 55
6.3.7.1. Intensive care unit management 55
6.3.7.2. Mechanical circulatory support 55
6.3.8. Lung and heart-lung transplantation 55
6.3.9. Evidence-based treatment algorithm 56
6.3.10. Diagnosis and treatment of pulmonary arterial hypertension complications 56
6.3.10.1. Arrhythmias 56
6.3.10.2. Haemoptysis 56
6.3.10.3. Mechanical complications 56
6.3.11. End-of-life care and ethical issues 57
6.3.12. New drugs in advanced clinical development (phase 3 studies) 57
7. Specific pulmonary arterial hypertension subsets 57
7.1. Pulmonary arterial hypertension associated with drugs and toxins 57
7.2. Pulmonary arterial hypertension associated with connective tissue disease 58
7.2.1. Epidemiology and diagnosis 58
7.2.2. Therapy 58
7.3. Pulmonary arterial hypertension associated with human immunodeficiency virus infection 59
7.3.1. Diagnosis 59
7.3.2. Therapy 59
7.4. Pulmonary arterial hypertension associated with portal hypertension 60
7.4.1. Diagnosis 60
7.4.2. Therapy 60
7.4.2.1. Liver transplantation 60
7.5. Pulmonary arterial hypertension associated with adult congenital heart disease 61
7.5.1. Diagnosis and risk assessment 61
7.5.2. Therapy 62
7.6. Pulmonary arterial hypertension associated with schistosomiasis 63
7.7. Pulmonary arterial hypertension with signs of venous/capillary involvement 63
7.7.1. Diagnosis 64
7.7.2. Therapy 64
7.8. Paediatric pulmonary hypertension 64
7.8.1. Epidemiology and classification 64
7.8.2. Diagnosis and risk assessment 66
7.8.3. Therapy 66
8. Pulmonary hypertension associated with left heart disease (group 2) 68
8.1. Definition, prognosis, and pathophysiology 68
8.2. Diagnosis 70
8.2.1. Diagnosis and control of the underlying left heart disease 70
8.2.2. Evaluation of pulmonary hypertension and patient phenotyping 70
8.2.3. Invasive assessment of haemodynamics 70
8.3. Therapy 71
8.3.1. Pulmonary hypertension associated with left-sided heart failure 71
8.3.1.1. Heart failure with reduced ejection fraction 71
8.3.1.2. Heart failure with preserved ejection fraction 71
8.3.1.3. Interatrial shunt devices 72
8.3.1.4. Remote pulmonary arterial pressure monitoring in heart failure 72
8.3.2. Pulmonary hypertension associated with valvular heart disease 72
8.3.2.1. Mitral valve disease 72
8.3.2.2. Aortic stenosis 72
8.3.2.3. Tricuspid regurgitation 72
8.3.3. Recommendations on the use of drugs approved for PAH in PH-LHD 72
9. Pulmonary hypertension associated with lung diseases and/or hypoxia (group 3) 73
9.1. Diagnosis 75
9.2. Therapy 75
9.2.1. Pulmonary hypertension associated with chronic obstructive pulmonary disease or emphysema 75
9.2.2. Pulmonary hypertension associated with interstitial lung disease 75
9.2.3. Recommendations on the use of drugs approved for PAH in PH associated with lung disease 76
10. Chronic thrombo-embolic pulmonary hypertension (group 4) 76
10.1. Diagnosis 77
10.2. Therapy 78
10.2.1. Surgical treatment 78
10.2.2. Medical therapy 78
10.2.3. Interventional treatment 79
10.2.4. Multimodal treatment 80
10.2.5. Follow-up 81
10.3. Chronic thrombo-embolic pulmonary hypertension team and experience criteria 81
11. Pulmonary hypertension with unclear and/or multifactorial mechanisms (group 5) 82
11.1. Haematological disorders 82
11.2. Systemic disorders 83
11.3. Metabolic disorders 83
11.4. Chronic kidney failure 83
11.5. Pulmonary tumour thrombotic microangiopathy 83
11.6. Fibrosing mediastinitis 83
12. Definition of a pulmonary hypertension centre 84
12.1. Facilities and skills required for a pulmonary hypertension centre 85
12.2. European Reference Network 85
12.3. Patient associations and patient empowerment 85
13. Key messages 86
14. Gaps in evidence 86
14.1. Pulmonary arterial hypertension (group 1) 86
14.2. Pulmonary hypertension associated with left heart disease (group 2) 86
14.3. Pulmonary hypertension associated with lung diseases and/ or hypoxia (group 3) 87
14.4. Chronic thrombo-embolic pulmonary hypertension (group 4) 87
14.5. Pulmonary hypertension with unclear and/or multifactorial mechanisms (group 5) 87
15. ‘What to do’ and ‘What not to do’ messages from the Guidelines 87
16. Quality indicators 93
17. Supplementary data 94
18. Data availability statement 94
19. Author information 94
20. Appendix 94
21. References 95
Tables of recommendations
Recommendation Table 1 — Recommendations for right heart catheterization and vasoreactivity testing 31
Recommendation Table 2 — Recommendations for diagnostic strategy 35
Recommendation Table 3 — Recommendations for screening and improved detection of pulmonary arterial hypertension and
chronic thrombo-embolic pulmonary hypertension 37
Recommendation Table 4 — Recommendations for evaluating the disease severity and risk of death in patients with pulmonary
arterial hypertension 43
Recommendation Table 5 — Recommendations for general
measures and special circumstances 45
Recommendation Table 6 — Recommendations for women of childbearing potential 46
Recommendation Table 7 — Recommendations for the treatment of vasoreactive patients with idiopathic, heritable, or
drug-associated pulmonary arterial hypertension 47
Recommendation Table 8 — Recommendations for the treatment of non-vasoreactive patients with idiopathic, heritable, or drug-associated pulmonary arterial hypertension who present without cardiopulmonary comorbiditiesa 51
Recommendation Table 9 — Recommendations for initial oral drug combination therapy for patients with idiopathic, heritable, or drug-associated pulmonary arterial hypertension without cardiopulmonary comorbidities 52
Recommendation Table 10 — Recommendations for sequential drug combination therapy for patients with idiopathic, heritable, or drug-associated pulmonary arterial hypertension 53
Recommendation Table 11 — Recommendations for the treatment of non-vasoreactive patients with idiopathic, heritable, or drug-associated pulmonary arterial hypertension who present with cardiopulmonary comorbiditiesa 54
Recommendation Table 12 — Recommendations for intensive care management for pulmonary arterial hypertension 55
Recommendation Table 13 — Recommendations for lung transplantation 56
Recommendation Table 14 — Recommendations for pulmonary arterial hypertension associated with drugs or toxins 57
Recommendation Table 15 — Recommendations for pulmonary arterial hypertension associated with connective tissue disease 58
Recommendation Table 16 — Recommendations for pulmonary arterial hypertension associated with human immunodeficiency virus infection 59
Recommendation Table 17 — Recommendations for pulmonary arterial hypertension associated with portal hypertension 60
Recommendation Table 18 — Recommendations for shunt closure in patients with pulmonary-systemic flow ratio .1.5:1 based on calculated pulmonary vascular resistance 63
Recommendation Table 19 — Recommendations for pulmonary arterial hypertension associated with adult congenital heart disease 63
Recommendation Table 20 — Recommendations for pulmonary arterial hypertension with signs of venous/capillary involvement 64
Recommendation Table 21 — Recommendations for paediatric pulmonary hypertension 68
Recommendation Table 22 — Recommendations for pulmonary hypertension associated with left heart disease 73
Recommendation Table 23 — Recommendations for pulmonary hypertension associated with lung disease and/or hypoxia 76
Recommendation Table 24 — Recommendations for chronic thrombo-embolic pulmonary hypertension and chronic thrombo-embolic pulmonary disease without pulmonary hypertension 81
Recommendation Table 25 — Recommendations for pulmonary hypertension centres 85
List of tables
Table 1 Strength of the recommendations according to GRADE 18
Table 2 Quality of evidence grades and their definitions 18
Table 3 Classes of recommendations 19
Table 4 Levels of evidence 19
Table 5 Haemodynamic definitions of pulmonary hypertension 20
Table 6 Clinical classification of pulmonary hypertension 21
Table 7 Drugs and toxins associated with pulmonary arterial hypertension 23
Table 8 Electrocardiogram abnormalities in patients with pulmonary hypertension 26
Table 9 Radiographic signs of pulmonary hypertension and concomitant abnormalities 26
Table 10 Additional echocardiographic signs suggestive of pulmonary hypertension 28
Table 11 Haemodynamic measures obtained during right heart catheterization 29
Table 12 Route of administration, half-life, dosages, and duration of administration of the recommended test compounds for vasoreactivity testing in pulmonary arterial hypertension 30
Table 13 Phenotypic features associated with pulmonary arterial hypertension mutations 32
Table 14 Characteristic diagnostic features of patients with different forms of pulmonary hypertension 34
Table 15 World Health Organization classification of functional status of patients with pulmonary hypertension 39
Table 16 Comprehensive risk assessment in pulmonary arterial hypertension (three-strata model) 40
Table 17 Suggested assessment and timing for the follow-up of patients with pulmonary arterial hypertension 41
Table 18 Variables used to calculate the simplified four-strata risk-assessment tool 42
Table 19 Dosing of pulmonary arterial hypertension medication in adults 47
Table 20 Criteria for lung transplantation and listing in patients with pulmonary arterial hypertension 55
Table 21 Clinical classification of pulmonary arterial hypertension associated with congenital heart disease 61
Table 22 Use of pulmonary arterial hypertension therapies in children 67
Table 23 Patient phenotyping and likelihood for left heart disease as cause of pulmonary hypertension 71
Table 24 Pulmonary hypertension with unclear and/or multifactorial mechanisms 82
List of figures
Figure 1 Central illustration 22
Figure 2 Symptoms in patients with pulmonary hypertension 24
Figure 3 Clinical signs in patients with pulmonary hypertension 25
Figure 4 Transthoracic echocardiographic parameters in the assessment of pulmonary hypertension 27
Figure 5 Echocardiographic probability of pulmonary hypertension and recommendations for further assessment 28
Figure 6 Diagnostic algorithm of patients with unexplained dyspnoea and/or suspected pulmonary hypertension 33
Figure 7 Pathophysiology and current therapeutic targets of pulmonary arterial hypertension (group 1) 38
Figure 8 Vasoreactivity testing algorithm of patients with presumed diagnosis of idiopathic, heritable, or drug-associated pulmonary arterial hypertension 48
Figure 9 Evidence-based pulmonary arterial hypertension treatment algorithm for patients with idiopathic, heritable, drug-associated, and connective tissue disease-associated pulmonary arterial hypertension 49
Figure 10 Neonatal and paediatric versus adult pulmonary hypertension 65
Figure 11 Pathophysiology of pulmonary hypertension associated with left heart disease (group 2) 69
Figure 12 Pathophysiology of pulmonary hypertension associated with lung disease (group 3) 74
Figure 13 Diagnostic strategy in chronic thrombo-embolic pulmonary hypertension 77
Figure 14 Management strategy in chronic thrombo-embolic pulmonary hypertension 79
Figure 15 Overlap in treatments/multimodality approaches in chronic thrombo-embolic pulmonary hypertension 80
Figure 16 Pulmonary hypertension centre schematic 84
1. Preamble
Guidelines summarize and evaluate available evidence, with the aim of assisting health professionals in proposing the best management strategies for an individual patient with a given condition. Guidelines and their recommendations should facilitate decision-making of health professionals in their daily practice. However, guidelines are not a substitute for the patient's relationship with their practitioner. The final decisions concerning an individual patient must be made by the responsible health professional(s), based on what they consider to be the most appropriate in the circumstances. These decisions are made in consultation with the patient and caregiver as appropriate.
Guidelines are intended for use by health professionals. To ensure that all physicians have access to the most recent recommendations, both the European Society of Cardiology (ESC) and European Respiratory Society (ERS) make their guidelines freely available in their own journals. The ESC and ERS warn non-medical readers that the technical language may be misinterpreted and decline any responsibility in this respect.
Many Guidelines have been issued in recent years by the ESC and ERS. Because of their impact on clinical practice, quality criteria for the development of guidelines have been established in order to make all decisions transparent to the user. The ERS and ESC guidance and procedure to formulate and issue clinical practice recommendations can be found on the societies’ relevant website or journal (https://www.escardio.org/Guidelines and https://openres.ersjournals.com/content/8/1/00655-2021). The ESC and ERS Guidelines represent the official position of the ESC and ERS on a given topic and are regularly updated.
The panel of experts of these specific guidelines comprised an equal number of ERS and ESC members, including representatives from relevant subspecialty groups involved in the medical care of patients with this pathology.
The experts of the writing and reviewing panels provided declaration of interest forms for all relationships that might be perceived as real or potential sources of conflicts of interest. Their declarations of interest were reviewed according to the ESC declaration of interest rules and can be found on the ESC website (http://www.escardio.org/Guidelines). They have been compiled in a report and co-published in a supplementary document of the guidelines. This process ensures transparency and prevents potential biases in the development and review processes. Any changes in declarations of interest that arose during the writing period were notified to the ESC and updated. The Task Force received its entire financial support from the ESC and ERS without any involvement from the health care industry.
The ESC Clinical Practice Guidelines (CPG) Committee and the ERS Guidelines Director reporting to the ERS Science Council supervise and co-ordinate the preparation of new guidelines. These Guidelines underwent extensive review by the ESC CPG Committee, the ERS Guidelines Working Group, and external experts. The guidelines were developed after careful consideration of the scientific and medical knowledge and the evidence available at the time of drafting. After appropriate revisions, the guidelines were signed off by all the experts in the Task Force. The finalized document was signed off by the ESC CPG Committee and endorsed by the ERS Executive Committee before being simultaneously published in the European Heart Journal (EHJ) and the European Respiratory Journal (ERJ). The decision to publish the guidelines in both journals was made to ensure adequate dissemination of the recommendations in both the cardiology and respiratory fields.
The task of developing the ESC/ERS Guidelines also included creating educational tools and implementation programmes for the recommendations, including condensed pocket guidelines versions, summary slides, a lay summary, and an electronic version for digital applications (smartphones, etc.). These versions are abridged and thus, for more detailed information, the user should always access the full-text version of the guidelines, which is freely available via the ESC and ERS websites, and hosted on the EHJ and ERJ websites. The National Cardiac Societies of the ESC are encouraged to endorse, adopt, translate, and implement all ESC Guidelines. Pulmonary national societies are also encouraged to share these guidelines with their members and develop a summary or editorials in their own language, if appropriate. Implementation programmes are needed because it has been shown that the outcome of disease may be favourably influenced by the thorough application of clinical recommendations.
Health professionals are encouraged to take the ESC/ERS Guidelines fully into account when exercising their clinical judgement, as well as in determining and implementing preventive, diagnostic, or therapeutic medical strategies. However, the ESC/ERS Guidelines do not override, in any way, the individual responsibility of health professionals to make appropriate and accurate decisions in considering each patient's health condition and in consulting with that patient or the patient's caregiver where appropriate and/or necessary. It is also the health professional's responsibility to verify the rules and regulations applicable to drugs and devices at the time of prescription and, where appropriate, to respect the ethical rules of their profession in each country.
Off-label use of medication may be presented in these guidelines if a sufficient level of evidence shows that it can be considered medically appropriate to a given condition and if patients could benefit from the recommended therapy. However, the final decisions concerning an individual patient must be made by the responsible health professional, giving special consideration to:
The specific situation of the patient. In this respect, it is specified that, unless otherwise provided for by national regulations, off-label use of medication should be limited to situations where it is in the patient's interest to do so, with regards to the quality, safety, and efficacy of care, and only after the patient has been fully informed and provided consent.
Country-specific health regulations, indications by governmental drug regulatory agencies, and the ethical rules to which health professionals are subject, where applicable.
2. Introduction
Pulmonary hypertension (PH) is a pathophysiological disorder that may involve multiple clinical conditions and may be associated with a variety of cardiovascular and respiratory diseases. The complexity of managing PH requires a multifaceted, holistic, and multidisciplinary approach, with active involvement of patients with PH in partnership with clinicians. Streamlining the care of patients with PH in daily clinical practice is a challenging but essential requirement for effectively managing PH. In recent years, substantial progress has been made in detecting and managing PH, and new evidence has been timeously integrated in this fourth edition of the ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Reflecting the multidisciplinary input into managing patients with PH and interpreting new evidence, the Task Force included cardiologists and pneumologists, a thoracic surgeon, methodologists, and patients. These comprehensive clinical practice guidelines cover the whole spectrum of PH, with an emphasis on diagnosing and treating pulmonary arterial hypertension (PAH) and chronic thrombo-embolic pulmonary hypertension (CTEPH).
2.1. What is new
One of the most important proposals from the 6th World Symposium on Pulmonary Hypertension (WSPH) was to reconsider the haemodynamic definition of PH [1]. After careful evaluation, the new definitions of PH have been endorsed and expanded in these guidelines, including a revised cut-off level for pulmonary vascular resistance (PVR) and a definition of exercise PH.
The classification of PH has been updated, including repositioning of vasoreactive patients with idiopathic pulmonary arterial hypertension (IPAH) and a revision of group 5 PH, including repositioning of PH in lymphangioleiomyomatosis in group 3.
Concerning the diagnosis of PH, a new algorithm has been developed aiming at earlier detection of PH in the community. In addition, expedited referral is recommended for high-risk or complex patients. Screening strategies are also proposed.
The risk-stratification table has been expanded to include additional echocardiographic and cardiac magnetic resonance imaging (cMRI) prognostic indicators. The recommendations for initial drug therapies have been simplified, building on this revised, three-strata, multiparametric risk model to replace functional classification. At follow-up, a four-strata risk-assessment tool is now proposed based on refined cut-off levels for World Health Organization functional class (WHO-FC), 6-minute walking distance (6MWD), and N-terminal pro-brain natriuretic peptide (NT-proBNP), categorizing patients as low, intermediate–low, intermediate–high, or high risk.
The PAH treatment algorithm has been modified, highlighting the importance of cardiopulmonary comorbidities, risk assessment both at diagnosis and follow-up, and the importance of combination therapies. Treatment strategies during follow-up have been based on the four-strata model intended to facilitate more granular decision-making.
The recommendations for managing PH associated with left heart disease (PH-LHD) and lung disease have been updated, including a new haemodynamic definition of severe PH in patients with lung disease.
In group 4 PH, the term chronic thrombo-embolic pulmonary disease (CTEPD) with or without PH has been introduced, acknowledging the presence of similar symptoms, perfusion defects, and organized fibrotic obstructions in patients with or without PH at rest. Interventional treatment by balloon pulmonary angioplasty (BPA) in combination with medical therapy has been upgraded in the therapeutic algorithm of CTEPH.
New standards for PH centres have been presented and, for the first time, patient representatives were actively involved in developing these guidelines.
Questions with direct consequences for clinical practitioners regarding each PH classification subgroup were selected and addressed, namely guidance on: initial treatment strategy for group 1 PH (Population, Intervention, Control, Outcome [PICO] I); use of oral phosphodiesterase 5 inhibitors (PDE5is) for the treatment of group 2 PH (PICO II); use of oral PDE5is for the treatment of group 3 PH (PICO III); and use of PH drugs prior to BPA for the treatment of group 4 PH (PICO IV). These questions were considered to be important because: most contemporary PH registries describe variable use of initial oral monotherapy and combination therapy; large case series show widespread use of PDE5is in group 2 PH, despite a class III recommendation in the 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension; large case series show widespread use of PDE5is in group 3 PH, despite a class III recommendation in the 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension; and there is no clear guidance for therapy with PH drugs in patients with inoperable CTEPH prior to BPA.
2.2. Methods
Three main methodological approaches were used in these guidelines, depending on the type of questions addressed:
i) Four questions that were considered highly important were formulated in the PICO format, and assessed with full systematic reviews and application of the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) approach [2] and the Evidence to Decision (EtD) framework [3] (see Supplementary Data, Section 2.1 for full methodology description and supportive material). The resulting recommendations were rated as strong or conditional, based on four potential levels of evidence (high, moderate, low, or very low; Tables 1 and 2). All Task Force members approved the recommendations. In addition, these recommendations were also presented and voted following the usual ESC approach.
ii) Eight questions that were considered of key importance (key narrative questions) were assessed with systematic literature searches and application of the EtD framework [6]. The evidence grading was performed following the usual ESC approach.
iii) The remaining topics of interest were assessed using the process commonly followed in ESC Guidelines. Structured literature searches were undertaken and grading tables, as outlined in Tables 3 and 4, were created to describe level of confidence in the recommendation provided and the quality of evidence supporting the recommendation. The Task Force discussed each draft recommendation during web-based conference calls dedicated to specific sections, followed by consensus modifications and an online vote on each recommendation. Only recommendations that were supported by at least 75% of the Task Force members were included in the guidelines. The recommendation tables were colour-coded for ease of interpretation.
3. Definitions and classifications
3.1. Definitions
The definitions for PH are based on haemodynamic assessment by right heart catheterization (RHC). Although haemodynamics represent the central element of characterizing PH, the final diagnosis and classification should reflect the whole clinical context and consider the results of all investigations.
Pulmonary hypertension is defined by a mean pulmonary arterial pressure (mPAP) >20 mmHg at rest (Table 5). This is supported by studies assessing the upper limit of normal pulmonary arterial pressure (PAP) in healthy subjects [7–9], and by studies investigating the prognostic relevance of increased PAP (key narrative question 1, Supplementary Data, Section 3.1) [10–12].
It is essential to include PVR and pulmonary arterial wedge pressure (PAWP) in the definition of pre-capillary PH, in order to discriminate elevated PAP due to pulmonary vascular disease (PVD) from that due to left heart disease (LHD), elevated pulmonary blood flow, or increased intrathoracic pressure (Table 5). Based on the available data, the upper limit of normal PVR and the lowest prognostically relevant threshold of PVR is ∼2 Wood units (WU) [7, 8, 13, 14]. Pulmonary vascular resistance depends on body surface area and age, with elderly healthy subjects having higher values. The available data on the best threshold for PAWP discriminating pre- and post-capillary PH are contradictory. Although the upper limit of normal PAWP is considered to be 12 mmHg [15], previous ESC/ERS Guidelines for the diagnosis and treatment of PH, as well as the recent consensus recommendation of the ESC Heart Failure Association [16], suggest a higher threshold for the invasive diagnosis of heart failure (HF) with preserved ejection fraction (HFpEF) (PAWP ≥15 mmHg). In addition, almost all therapeutic studies of PAH have used the PAWP ≤15 mmHg threshold. Therefore, it is recommended keeping PAWP ≤15 mmHg as the threshold for pre-capillary PH, while acknowledging that any PAWP threshold is arbitrary and that the patient phenotype, risk factors, and echocardiographic findings, including left atrial (LA) volume, need to be considered when distinguishing pre- from post-capillary PH.
Patients with PAH are haemodynamically characterized by pre-capillary PH in the absence of other causes of pre-capillary PH, such as CTEPH and PH associated with lung diseases. All PH groups may comprise both pre- and post-capillary components contributing to PAP elevation. In particular, older patients may present with several conditions predisposing them to PH. The primary classification should be based on the presumed predominant cause of the pulmonary pressure increase.
Post-capillary PH is haemodynamically defined as mPAP >20 mmHg and PAWP >15 mmHg. Pulmonary vascular resistance is used to differentiate between patients with post-capillary PH who have a significant pre-capillary component (PVR >2 WU—combined post- and pre-capillary PH [CpcPH]) and those who do not (PVR ≤2 WU—isolated post-capillary PH [IpcPH]).
There are patients with elevated mPAP (>20 mmHg) but low PVR (≤2 WU) and low PAWP (≤15 mmHg). These patients are frequently characterized by elevated pulmonary blood flow and, although they have PH, they do not fulfil the criteria of pre- or post-capillary PH. This haemodynamic condition may be described by the term ‘unclassified PH’. Patients with unclassified PH may present with congenital heart disease (CHD), liver disease, airway disease, lung disease, or hyperthyroidism explaining their mPAP elevation. Clinical follow-up of these patients is generally recommended. In the case of elevated pulmonary blood flow, its aetiology should be explored.
As the groups of PH according to clinical classification represent different clinical conditions, there may be additional clinically relevant haemodynamic thresholds (e.g. for PVR) for the individual PH groups besides the general thresholds of the haemodynamic definition of PH, which are discussed in the corresponding sections.
Exercise PH, defined by an mPAP/cardiac output (CO) slope >3 mmHg/L/min between rest and exercise [17], has been re-introduced. The mPAP/CO slope is strongly age dependent and its upper limit of normal ranges from 1.6–3.3 mmHg/L/min in the supine position [17]. An mPAP/CO slope >3 mmHg/L/min is not physiological in subjects aged <60 years and may rarely be present in healthy subjects aged >60 years [17]. A pathological increase in pulmonary pressure during exercise is associated with impaired prognosis in patients with exercise dyspnoea [18] and in several cardiovascular conditions [19–22]. Although an increased mPAP/CO slope defines an abnormal haemodynamic response to exercise, it does not allow for differentiation between pre- and post-capillary causes. The PAWP/CO slope with a threshold >2 mmHg/L/min may best differentiate between pre- and post-capillary causes of exercise PH [23, 24].
3.2. Classifications
The basic structure of the classification from the 2015 ESC/ERS Guidelines for the diagnosis and treatment of PH [25, 26] and the Proceedings of the 6th WSPH [1] has been kept (Table 6). The general purpose of the clinical classification of PH remains to categorize clinical conditions associated with PH, based on similar pathophysiological mechanisms, clinical presentation, haemodynamic characteristics, and therapeutic management (Figure 1). The main changes are as follows:
i) The subgroups ‘non-responders at vasoreactivity testing’ and ‘acute responders at vasoreactivity testing’ have been added to IPAH as compared with the 2015 ESC/ERS Guidelines for the diagnosis and treatment of PH [25, 26]. In addition to patients with IPAH, some patients with heritable PAH (HPAH) or drug- or toxin-associated PAH (DPAH) might be acute responders.
ii) The groups ‘PAH with features of venous/capillary (pulmonary veno-occlusive disease/pulmonary capillary haemangiomatosis [PVOD/PCH]) involvement’ and ‘persistent PH of the newborn (PPHN)’ have been included in group 1 (PAH) as compared with the 2015 ESC/ERS Guidelines for the diagnosis and treatment of PH and in line with the Proceedings of the 6th WSPH [1].
iii) Instead of the general term ‘sleep-disordered breathing’, the term ‘hypoventilation syndromes’ should be used within group 3 to describe conditions with increased risk of PH. Sole nocturnal obstructive sleep apnoea is generally not a cause of PH, but PH is frequent in patients with hypoventilation syndromes causing daytime hypercapnia.
4. Epidemiology and risk factors
Pulmonary hypertension is a major global health issue. All age groups are affected. Present estimates suggest a PH prevalence of ∼1% of the global population. Due to the presence of cardiac and pulmonary causes of PH, prevalence is higher in individuals aged >65 years [29]. Globally, LHD is the leading cause of PH [29]. Lung disease, especially chronic obstructive pulmonary disease (COPD), is the second most common cause [29]. In the UK, the observed PH prevalence has doubled in the last 10 years and is currently 125 cases/million inhabitants [30]. Irrespective of the underlying condition, developing PH is associated with worsening symptoms and increased mortality [29]. In developing countries, CHD, some infectious diseases (schistosomiasis, human immunodeficiency virus [HIV]), and high altitude represent important but under-studied causes of PH [29].
4.1. Group 1, pulmonary arterial hypertension
Recent registry data from economically developed countries indicate a PAH incidence and prevalence of ∼6 and 48–55 cases/million adults, respectively [31]. It has been thought to predominantly affect younger individuals, mostly females [32, 33]; this is currently true for HPAH, which affects twice as many females as males. However, recent data from the USA and Europe suggest that PAH is now frequently diagnosed in older patients (i.e. those aged ≥65 years, who often present with cardiovascular comorbidities, resulting in a more equal distribution between sexes) [32]. In most PAH registries, IPAH was the most common subtype (50–60% of all cases), followed by PAH associated with connective tissue disease (CTD), CHD, and portal hypertension (porto-pulmonary hypertension [PoPH]) [32].
A number of drugs and toxins are associated with the development of PAH [1, 34–45]. The association between exposure to drugs and toxins and PAH is classified as definite or possible, as proposed at the 6th WSPH (Table 7) [1]. There is a definite association with drugs, with available data based on outbreaks, epidemiological case-control studies, or large multicentre series. A possible association is suggested by multiple case series or cases with drugs with similar mechanisms of action [1].
4.2. Group 2, pulmonary hypertension associated with left heart disease
In 2013, the Global Burden of Disease Study reported 61.7 million cases of HF worldwide, which represented almost a doubling since 1990 [46]. In Europe and the USA, >80% of patients with HF are aged ≥65 years. Post-capillary PH, either isolated or combined with a pre-capillary component, is a frequent complication mainly in HFpEF, affecting at least 50% of these patients [47, 48]. The prevalence of PH increases with severity of left-sided valvular diseases, and PH can be found in 60–70% of patients with severe and symptomatic mitral valve disease [49] and in up to 50% of those with symptomatic aortic stenosis [50].
4.3. Group 3, pulmonary hypertension associated with lung diseases and/or hypoxia
Mild PH is common in advanced parenchymal and interstitial lung disease. Studies have reported that ∼1–5% of patients with advanced COPD with chronic respiratory failure or candidates for lung volume reduction surgery or lung transplantation (LTx) have an mPAP >35–40 mmHg [51, 52]. In idiopathic pulmonary fibrosis, an mPAP ≥25 mmHg has been reported in 8–15% of patients upon initial work-up, with greater prevalence in advanced (30–50%) and end-stage (>60%) disease [52]. Hypoxia is a public health problem for the estimated 120 million people living at altitudes >2500 m. Altitude dwellers are at risk of developing PH and chronic mountain sickness. However, it remains unclear to what extent PH and right HF are public health problems in high-altitude communities; this should be addressed with updated methodology and large-scale population studies [53].
4.4. Group 4, pulmonary hypertension associated with chronic pulmonary artery obstruction
The number of patients diagnosed with CTEPH is increasing, probably due to a deeper understanding of the disease and more active screening for this condition in patients who remain dyspnoeic after pulmonary embolism (PE) or who have risk factors for developing CTEPH. Registry data indicate a CTEPH incidence and prevalence of 2–6 and 26–38 cases/million adults, respectively [31, 54, 55]. Patients with chronic thrombo-embolic pulmonary disease (CTEPD) without PH still represent a small proportion of the patients referred to CTEPH centres [56].
4.5. Group 5, pulmonary hypertension with unclear and/or multifactorial mechanisms
Group 5 PH consists of a complex group of disorders that are associated with PH [57]. The cause is often multifactorial and can be secondary to increased pre- and post-capillary pressure, as well as direct effects on pulmonary vasculature. The incidence and prevalence of PH in most of these disorders are unknown. However, high-quality registries have recently enabled estimation of PH prevalence in adult patients with sarcoidosis [58, 59]. Studies suggest that PH can be common and its presence is often associated with increased morbidity and mortality [58, 59].
5. Pulmonary hypertension diagnosis
5.1. Diagnosis
The diagnostic approach to PH is mainly focused on two tasks. The primary goal is to raise early suspicion of PH and ensure fast-track referral to PH centres in patients with a high likelihood of PAH, CTEPH, or other forms of severe PH. The second objective is to identify underlying diseases, especially LHD (group 2 PH) and lung disease (group 3 PH), as well as comorbidities, to ensure proper classification, risk assessment, and treatment.
5.1.1. Clinical presentation
Symptoms of PH are mainly linked to right ventricle (RV) dysfunction, and typically associated with exercise in the earlier course of the disease [25, 26]. The cardinal symptom is dyspnoea on progressively minor exertion. Other common symptoms are related to the stages and severity of the disease, and are listed in Figure 2 [60–62]. Potential clinical signs and physical findings are summarized in Figure 3 [60, 61]. Importantly, the physical examination may also be the key to identifying the underlying cause of PH (see Figure 3).
5.1.2. Electrocardiogram
Electrocardiogram (ECG) abnormalities (Table 8) may raise suspicion of PH, deliver prognostic information, and detect arrhythmias and signs of LHD. In adults with clinical suspicion of PH (e.g. unexplained dyspnoea on exertion), right axis deviation has a high predictive value for PH [63]. A normal ECG does not exclude the presence of PH, but a normal ECG in combination with normal biomarkers (BNP/NT-proBNP) is associated with a low likelihood of PH in patients referred for suspected PH or at risk of PH (i.e. after acute PE) [64, 65].
5.1.3. Chest radiography
Chest radiography presents abnormal findings in most patients with PH; however, a normal chest X-ray does not exclude PH [68]. Radiographic signs of PH include a characteristic configuration of the cardiac silhouette due to right heart (right atrium [RA]/RV) and PA enlargement, sometimes with pruning of the peripheral vessels. In addition, signs of the underlying cause of PH, such as LHD or lung disease, may be found (Table 9) [25, 26, 60, 69, 70].
5.1.4. Pulmonary function tests and arterial blood gases
Pulmonary function tests (PFTs) and analysis of arterial blood gas (ABG) or arterialized capillary blood are necessary to distinguish between PH groups, assess comorbidities and the need for supplementary oxygen, and determine disease severity. The initial work-up of patients with suspected PH should comprise forced spirometry, body plethysmography, lung diffusion capacity for carbon monoxide (DLCO), and ABG.
In patients with PAH, PFTs are usually normal or may show mild restrictive, obstructive, or combined abnormalities [71, 72]. More severe PFT abnormalities are occasionally found in patients with PAH associated with CHD [73], and those with group 3 PH. The DLCO may be normal in patients with PAH, although it is usually mildly reduced [71]. A severely reduced DLCO (<45% of the predicted value) in the presence of otherwise normal PFTs can be found in PAH associated with systemic sclerosis (SSc), PVOD, in PH group 3—associated with emphysema, interstitial lung disease (ILD), or combined pulmonary fibrosis and emphysema—and in some PAH phenotypes [74]. A low DLCO is associated with a poor prognosis in several forms of PH [75–78].
Patients with PAH usually have normal or slightly reduced partial pressure of arterial oxygen (PaO2). Severe reduction of PaO2 might raise suspicion for patent foramen ovale, hepatic disease, other abnormalities with right-to-left shunt (e.g. septal defect), or low-DLCO-associated conditions.
Partial pressure of arterial carbon dioxide (PaCO2) is typically lower than normal due to alveolar hyperventilation [79]. Low PaCO2 at diagnosis and follow-up is common in PAH and associated with unfavourable outcomes [80]. Elevated PaCO2 is very unusual in PAH and reflects alveolar hypoventilation, which in itself may be a cause of PH. Overnight oximetry or polysomnography should be performed if there is suspicion of sleep-disordered breathing or hypoventilation [81].
5.1.5. Echocardiography
Independent of the underlying aetiology, PH leads to RV pressure overload and dysfunction, which can be detected by echocardiography [82–84]. When performed accurately, echocardiography provides comprehensive information on right and left heart morphology, RV and LV function, and valvular abnormalities, and gives estimates of haemodynamic parameters. Echocardiography is also a valuable tool with which to detect the cause of suspected or confirmed PH, particularly with respect to PH associated with LHD or CHD. Yet, echocardiography alone is insufficient to confirm a diagnosis of PH, which requires RHC.
Given the heterogeneous nature of PH and the peculiar geometry of the RV, there is no single echocardiographic parameter that reliably informs about PH status and underlying aetiology. Therefore, a comprehensive echocardiographic evaluation for suspected PH includes estimating the systolic pulmonary arterial pressure (sPAP) and detecting additional signs suggestive of PH, aiming at assigning an echocardiographic level of probability of PH. Echocardiographic findings of PH, including estimating pressure and signs of RV overload and/or dysfunction, are summarized in Figure 4.
Estimates of sPAP are based on the peak tricuspid regurgitation velocity (TRV) and the TRV-derived tricuspid regurgitation pressure gradient (TRPG)—after excluding pulmonary stenosis—taking into account non-invasive estimates of RA pressure (RAP). Considering the inaccuracies in estimating RAP and the amplification of measurement errors by using derived variables [85–87], these guidelines recommend using the peak TRV (and not the estimated sPAP) as the key variable for assigning the echocardiographic probability of PH. A peak TRV >2.8 m/s may suggest PH; however, the presence or absence of PH cannot be reliably determined by TRV alone [88]. Lowering the TRV threshold in view of the revised haemodynamic definition of PH is not supported by available data (key narrative question 2, Supplementary Data, Section 5.1) [89–92]. Tricuspid regurgitation (TR) velocity may underestimate (e.g. in patients with severe TR) [28] or overestimate (e.g. in patients with high CO in liver disease or sickle cell disease [SCD] [93, 94], misinterpretation of tricuspid valve closure artefact for the TR jet, or incorrect assignment of a peak TRV in the case of maximum velocity boundary artefacts) pressure gradients. Hence, additional variables related to RV morphology and function are used to define the echocardiographic probability of PH (Table 10) [82–84, 95], which may then be determined as low, intermediate, or high. When interpreted in a clinical context, this probability can be used to decide the need for further investigation, including cardiac catheterization in individual patients (Figure 5).
Echocardiographic measures of RV function include the tricuspid annular plane systolic excursion (TAPSE), RV fractional area change (RV-FAC), RV free-wall strain, and tricuspid annulus velocity (S′ wave) derived from tissue Doppler imaging, and potentially RV ejection fraction (RVEF) derived from 3D echocardiography. Furthermore, the TAPSE/sPAP ratio—representing a non-invasive measure of RV–PA coupling [96]—may aid in diagnosing PH [90, 97, 98]. The pattern of RV outflow tract (RVOT) blood flow (mid-systolic ‘notching’) may suggest pre-capillary PH [99, 100].
To separate between group 2 PH and other forms of PH, and to assess the likelihood of left ventricle (LV) diastolic dysfunction, LA size and signs of LV hypertrophy should always be measured, and Doppler echocardiographic signs (e.g. E/A ratio, E/E′) should be assessed even if the reliability of the latter is considered low [16]. To identify CHD, 2D Doppler and contrast examinations are helpful, but transoesophageal contrast echocardiography or other imaging techniques (e.g. computer tomography [CT] angiography, cMRI) are needed in some cases to detect or exclude sinus venosus atrial septal defects, patent ductus arteriosus, and/or anomalous pulmonary venous return [101]. The clinical value of exercise Doppler echocardiography in identifying exercise PH remains uncertain because of the lack of validated criteria and prospective confirmatory data. In most cases, increases in sPAP during exercise are caused by diastolic LV dysfunction [16].
5.1.6. Ventilation/perfusion lung scan
A ventilation/perfusion (V/Q) lung scan (planar or single-photon emission computed tomography [SPECT]) is recommended in the diagnostic work-up of patients with suspected or newly diagnosed PH, to rule out or detect signs of CTEPH [102, 103]. The V/Q SPECT is superior to planar imaging and is the methodology of choice; however, SPECT has been widely evaluated in assessing PE, but not to the same degree in CTEPH [68]. In the absence of parenchymal lung disease, a normal perfusion scan excludes CTEPH with a negative predicted value of 98% [104, 105]. In most patients with PAH, V/Q scintigraphy is normal or shows a speckled pattern but no typical perfusion defects characteristic of PE or CTEPH, whereas matched V/Q defects may be found in patients with lung disease (i.e. group 3 PH). Non-matched perfusion defects similar to those seen in CTEPH may be present in 7–10% of patients with PVOD/PCH or PAH [106, 107]. Deposition of the perfusion agent in extrapulmonary organs may hint to cardiac or pulmonary right-to-left shunting and has been reported in CHD, hepato-pulmonary syndrome, and pulmonary arteriovenous malformations (PAVMs) [68].
5.1.7. Non-contrast and contrast-enhanced chest computed tomography examinations, and digital subtraction angiography
Computed tomography (CT) imaging may provide important information for patients with unexplained dyspnoea or suspected/confirmed PH. The CT signs suggesting the presence of PH include an enlarged PA diameter, a PA-to-aorta ratio >0.9, and enlarged right heart chambers [68]. A combination of three parameters (PA diameter ≥30 mm, RVOT wall thickness ≥6 mm, and septal deviation ≥140° [or RV:LV ratio ≥1]) is highly predictive of PH [108]. Non-contrast chest CT can help determine the cause of PH when there are features of parenchymal lung disease, and may also point towards the presence of PVOD/PCH by showing centrilobular ground-glass opacities (which may also be found in PAH), septal lines, and lymphadenopathy [68].
Computed tomography pulmonary angiography (CTPA) is mainly used to detect direct or indirect signs of CTEPH, such as filling defects (including thrombus adhering to the vascular wall), webs or bands in the PAs, PA retraction/dilatation, mosaic perfusion, and enlarged bronchial arteries. Importantly, the diagnostic accuracy of CTPA for CTEPH is limited (at the patient level, sensitivity and specificity are 76% and 96%, respectively) [109], but was reported to be higher when modern, high-quality multi-detector CT scanners were used and when interpreted by experienced readers [109, 110]. Computed tomography pulmonary angiography may also be used to detect other cardiovascular abnormalities, including intracardiac shunts, abnormal pulmonary venous return, patent ductus arteriosus, and PAVMs.
In patients presenting with a clinical picture of acute PE, chest CT may be helpful in detecting signs of hitherto undetected CTEPH, which may include the presence of the above CTEPH signs, and RV hypertrophy as a sign for chronicity [111, 112]. Detecting ‘acute on chronic’ PE is important, as it may impact the management of patients with presumed acute PE.
Dual-energy CT (DECT) angiography and iodine subtraction mapping may provide additional diagnostic information by creating iodine maps [113], which reflect lung perfusion, thereby possibly increasing the diagnostic accuracy for CTEPH [114]. Although increasingly used, the diagnostic value of DECT in the work-up of patients with PH has not been established.
Digital subtraction angiography (DSA) is mainly used to confirm the diagnosis of CTEPH and to assess treatment options (i.e. operability or accessibility for BPA). Most centres use conventional two- or three-planar DSA. However, C-arm CT imaging may provide a higher spatial resolution, potentially identifying more target vessels for BPA and providing procedural guidance [115, 116].
5.1.8. Cardiac magnetic resonance imaging
Cardiac magnetic resonance imaging accurately and reproducibly assesses atrial and ventricular size, morphology, and function. Additional information on RV/LV myocardial strain can be obtained by applying tagging or by post-processing feature tracking. In addition, cMRI can be used to measure blood flow in the PA, aorta, and vena cava, allowing for quantifying stroke volume (SV), intracardiac shunt, and retrograde flow. By combining contrast magnetic resonance (MR) angiography and pulmonary perfusion imaging with late gadolinium-enhancement imaging of the myocardium, a complete picture of the heart and pulmonary vasculature can be obtained (see Supplementary Data, Table S2 for cMRI indices and normal values). A limitation is that there is no established method with which to estimate PAP. Even though the cost and availability of the technique precludes its use in the early diagnosis of PAH, it is sensitive in detecting early signs of PH and diagnosing CHD [117].
5.1.9. Blood tests and immunology
The initial diagnostic assessment of patients with newly diagnosed PH/PAH aims to identify comorbidities and possible causes or complications of PH. Laboratory tests that should be obtained at the time of PH diagnosis include: blood counts (including haemoglobin [Hb]); serum electrolytes (sodium, potassium); kidney function (creatinine, calculation of estimated glomerular filtration rate, and urea); uric acid; liver parameters (alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase, γ-glutamyl transpeptidase, bilirubin); iron status (serum iron, transferrin saturation, and ferritin); and BNP or NT-proBNP. In addition, serological studies should include testing for hepatitis viruses and HIV. Basic immunology laboratory work-up is recommended, including screening tests for anti-nuclear antibodies, anti-centromere antibodies, and anti-Ro. Screening for biological markers of antiphospholipid syndrome is recommended in patients with CTEPH. Additional thrombophilia screening is not generally recommended, unless therapeutic consequences are to be expected [118]. Pulmonary arterial hypertension and other forms of severe PH can be associated with thyroid function disorders; hence, laboratory screening should include at least thyroid-stimulating hormone.
5.1.10. Abdominal ultrasound
An abdominal ultrasound examination should be part of the comprehensive diagnostic work-up of patients with newly diagnosed PH, particularly if liver disease is suspected. A major objective is to search for liver disease and/or portal hypertension, or portocaval shunt (Abernethy malformation). During the course of the disease, patients with PH may develop secondary organ dysfunction mainly affecting the liver and kidneys [119]. In these patients, abdominal ultrasound is needed for differential diagnostic reasons and to assess the extent of organ damage.
5.1.11. Cardiopulmonary exercise testing
Cardiopulmonary exercise testing (CPET) is a useful tool to assess the underlying pathophysiologic mechanisms leading to exercise intolerance. Patients with PAH show a typical pattern, with a low end-tidal partial pressure of carbon dioxide (PETCO2), high ventilatory equivalent for carbon dioxide (VE/VCO2), low oxygen pulse (VO2/HR), and low peak oxygen uptake (VO2) [120]. These findings should prompt consideration of PVD. In patients with LHD or COPD, such a pattern may indicate an additional pulmonary vascular limitation [121, 122]. In populations at risk of PAH, such as those with SSc, a normal peak VO2 seems to exclude the diagnosis of PAH [123].
5.1.12. Right heart catheterization, vasoreactivity, exercise, and fluid challenge
5.1.12.1. Right heart catheterization
Right heart catheterization is the gold standard for diagnosing and classifying PH. Performing RHC requires expertise and meticulous methodology following standardized protocols. In addition to diagnosing and classifying PH, clinical indications include haemodynamic assessment of heart or LTx candidates [124] and evaluating congenital cardiac shunts. Interpreting invasive haemodynamics should be done in the context of the clinical picture and other diagnostic investigations. When performed in PH centres, the frequencies of serious adverse events (1.1%) and procedure-related mortality (0.055%) are low [125]. A known thrombus or tumour in the RV or RA, recently implanted (<1 month) pacemaker, mechanical right heart valve, TriClip, and an acute infection are contraindications to RHC; the risk:benefit ratio should be individually assessed before each examination and discussed with the patient. The most feared complication of RHC is perforation of a PA.
The adequate preparation of patients for RHC is of major relevance. Pre-existing medical conditions should be optimally controlled at the time of the examination (particularly blood pressure and volume control). In the supine position, the mid-thoracic level is recommended as the zero reference level, which is at the level of the LA in most patients [126].
For a complete assessment of cardiopulmonary haemodynamics, all measures listed in Table 11 must be measured or calculated. Incomplete assessments must be avoided, as this may lead to misdiagnosis. As a minimum, mixed venous oxygen saturation (SvO2) and arterial oxygen saturation (SaO2) should be determined. A stepwise assessment of oxygen saturation should be performed in patients with SvO2 >75% and whenever a left-to-right shunt is suspected. Cardiac output (CO) should be assessed by the direct Fick method or thermodilution (mean values of at least three measurements). The indirect Fick method is considered to be less reliable than thermodilution [127]; however, thermodilution should not be used in the presence of shunts. Pulmonary vascular resistance ([mPAP−PAWP]/CO) should be calculated for each patient. All pressure measurements, including PAWP, should be performed at end expiration (without breath-holding manoeuvre). In patients with large intrathoracic pressure changes during the respiratory cycle (i.e. COPD, obesity, during exercise), it is appropriate to average over at least three to four respiratory cycles. If no reliable PAWP curve can be obtained, or if the PAWP values are implausible, additional measurement of LV end-diastolic pressure should be considered to avoid misclassification. Saturations taken with the catheter in the wedged position can confirm an accurate PAWP [128].
5.1.12.2. Vasoreactivity testing
The purpose of vasoreactivity testing in PAH is to identify acute vasoresponders who may be candidates for treatment with high-dose calcium channel blockers (CCBs). Pulmonary vasoreactivity testing is only recommended in patients with IPAH, HPAH, or DPAH. Inhaled nitric oxide [129] or inhaled iloprost [130, 131] are the recommended test compounds for vasoreactivity testing (Table 12). There is similar evidence for intravenous (i.v.) epoprostenol, but due to incremental dose increases and repetitive measurements, testing takes much longer and is therefore less feasible [129]. Adenosine i.v. is no longer recommended due to frequent side effects [132]. A positive acute response is defined as a reduction in mPAP by ≥10 mmHg to reach an absolute value ≤40 mmHg, with increased or unchanged CO [129]. In patients with PH-LHD, vasoreactivity testing is restricted to evaluating heart transplantation candidacy (see Section 8.1), and in patients with PH in the context of CHD with initial systemic-to-pulmonary shunting, vasoreactivity testing can be performed to evaluate the possibility of defect closure (see Section 7.5) [101].
5.1.12.3. Exercise right heart catheterization
Right heart catheterization is the gold standard method to assess cardiopulmonary haemodynamics during exercise and to define exercise PH [133]. The main reason to perform exercise RHC is to investigate patients with unexplained dyspnoea and normal resting haemodynamics in order to detect early PVD or left heart dysfunction. In addition, exercise haemodynamics may reveal important prognostic and functional information in patients at risk of PAH and CTEPH [22, 134, 135]. To maximize the amount of information, exercise RHC may be combined with CPET. According to the available data and experience, exercise RHC is not associated with an additional risk of complications compared with resting RHC and CPET [133].
Incremental exercise tests (step or ramp protocol) with repeated haemodynamic measurements provide the most clinical information on pulmonary circulation. The minimally required haemodynamic variables measured at each exercise level include mPAP, sPAP, diastolic PAP (dPAP), PAWP, CO, heart rate, and systemic blood pressure. In addition, RAP, SvO2, and SaO2 should at least be measured at rest and peak exercise. Total pulmonary resistance (TPR), PVR, and cardiac index (CI) should be calculated at each exercise level, as well as arteriovenous difference in oxygen at peak exercise. The mPAP/CO and PAWP/CO slopes should also be calculated [136, 137]. In patients with early PVD, PVR may be normal or mildly elevated at rest, but may change during exercise with a steep increase in mPAP, reflected by an mPAP/CO slope >3 mmHg/L/min, while the PAWP/CO slope usually remains <2 mmHg/L/min. Patients with left heart dysfunction, such as those with HFpEF [23] and/or dynamic mitral regurgitation [138], and a normal PAWP at rest, usually show a steep increase in mPAP and PAWP (and mPAP/CO, PAWP/CO slope) during exercise.
According to recent studies, a PAWP/CO slope >2 mmHg/L/min may be helpful in recognizing an abnormal PAWP increase and, therefore, a cardiac exercise limitation, especially in patients with PAWP 12–15 mmHg at rest [23, 24, 139]. A PAWP cut-off of >25 mmHg during supine exercise has been recommended for diagnosing HFpEF [16]. In patients with lung disease, increased intrathoracic pressure may contribute to mPAP elevation; this is exaggerated during exercise and can be recognized by a concomitant increase in RAP [140]. Some exercise haemodynamics are age dependent, with healthy elderly subjects presenting with steeper mPAP/CO and PAWP/CO slopes than healthy young individuals [9, 141].
5.1.12.4. Fluid challenge
Fluid challenge may reveal LV diastolic dysfunction in patients with PAWP ≤15 mmHg, but a clinical phenotype suggestive of LHD. Most available data are derived from studies aiming to uncover HFpEF (increase in PAWP) rather than identify group 2 PH (increase in PAP; see Section 8.1). It is generally accepted that rapid infusion (over 5–10 min) of ∼500 mL (7–10 mL/kg) of saline would be sufficient to detect an abnormal increase in PAWP to ≥18 mmHg (suggestive of HFpEF) [142], although validation and long-term evaluation of these data are needed [143]. There are insufficient data on the haemodynamic response to fluid challenge in patients with PAH. Recent data suggest that passive leg raise during RHC may also help to uncover occult HFpEF [144].
Recommendations | Classa | Levelb |
Right heart catheterisation (RHC) | ||
RHC is recommended to confirm the diagnosis of PH (especially PAH or CTEPH), and to support treatment decisions [25, 26] | I | B |
In patients with suspected or known PH, it is recommended to perform RHC in experienced centres [125] | I | C |
It is recommended that RHC comprises a complete set of haemodynamics and is performed following standardized protocols [25, 26, 145] | I | C |
Vasoreactivity testing | ||
Vasoreactivity testing is recommended in patients with I/H/DPAH to detect patients who can be treated with high doses of a CCB [129, 146] | I | B |
It is recommended that vasoreactivity testing is performed at PH centres | I | C |
It is recommended to consider a positive response to vasoreactivity testing by a reduction in mPAP ≥10 mmHg to reach an absolute value of mPAP ≤40 mmHg with an increased or unchanged COc [129] | I | C |
Inhaled nitric oxide, inhaled iloprost, or i.v. epoprostenol are recommended for performing vasoreactivity testing [129–132] | I | C |
Vasoreactivity testing, for identifying candidates for CCB therapy, is not recommended in patients with PAH other than I/H/DPAH, and in PH groups 2, 3, 4, and 5 [124, 129] | III | C |
CCB, calcium channel blocker; CO, cardiac output; CTEPH, chronic thrombo-embolic pulmonary hypertension; I/H/DPAH, idiopathic, heritable, drug-associated pulmonary arterial hypertension; i.v., intravenous; mPAP, mean pulmonary arterial pressure; PAH, pulmonary arterial hypertension; PH, pulmonary hypertension; RHC, right heart catheterisation. aClass of recommendation. bLevel of evidence. cTesting should also be performed in patients with a baseline mPAP ≤40 mmHg, in whom the same responder criteria apply.
5.1.13. Genetic counselling and testing
Mutations in PAH genes have been identified in familial PAH, IPAH, PVOD/PCH, and anorexigen-associated PAH (Table 13) [148]. The screening recommendations herein specifically relate to patients with an a priori diagnosis of PAH and not ‘at-risk’ populations being screened for PAH (see Section 5.3). All patients with these conditions should be informed about the possibility of a genetic condition and that family members could carry a mutation that increases the risk of PAH, allowing for screening and early diagnosis [33, 148]. Even if genetic testing is not performed, family members should be made aware of early signs and symptoms, to ensure that a timely and appropriate diagnosis is made [148].
Genetic counselling by appropriately trained PAH providers or geneticists should be performed prior to genetic testing, to address the complex questions related to penetrance, genetically at-risk family members, reproduction, genetic discrimination, and psychosocial issues. Careful genetic counselling with genetic counsellors or medical geneticists is critical prior to genetic testing for asymptomatic family members [148].
If the familial mutation is known and an unaffected family member tests negative for that mutation, the risk of PAH for that person is the same as for the general population [148].
Many of the less common mutations outlined have a potential additional set of syndromic features. These are summarized in Table 13 where specific clinical history, examination, and investigations are suggested. In particular, clinicians should undertake a thorough history and examination, as syndromic PAH diagnoses may be missed if not interrogated. For example, in one of the largest studies to date, TBX4, ALK1, and ENG mutations were represented in the top six most common genetic findings in adults with previously diagnosed IPAH [149]. These findings have been confirmed and extended in international genetics consortia in 4241 patients with PAH [150]. It is therefore apparent that there is either phenotypic heterogeneity of these syndromes or missed diagnostic features. As more genes associated with PAH are discovered, it will become increasingly difficult to individually test for each. Next-generation sequencing has enabled the development of gene panels to simultaneously interrogate several genes [151]. It is, however, important to check the genes included in the panel at the time of testing, since the composition changes as genetic discoveries advance.
5.2. Diagnostic algorithm
A multistep, pragmatic approach to diagnosis should be considered in patients with unexplained dyspnoea or symptoms/signs raising suspicion of PH. This strategy is depicted in detail in Figure 6 and Table 14. The diagnostic algorithm does not address screening for specific groups at risk of PH.
5.2.1 Step 1 (suspicion)
Patients with PH are likely to be seen by first-line physicians, mainly general practitioners, for non-specific symptoms. Initial evaluation should include a comprehensive medical (including familial) history, thorough physical examination (including measurement of blood pressure, heart rate, and pulse oximetry), blood test to determine BNP/NT-proBNP, and resting ECG. This first step may raise a suspicion of a cardiac or respiratory disorder causing the symptoms.
5.2.2. Step 2 (detection)
The second step includes classical, non-invasive lung and cardiac testing. Among those tests, echocardiography is an important step in the diagnostic algorithm (Figure 6), as it assigns a level of probability of PH, irrespective of the cause. In addition, it is an important step in identifying other cardiac disorders. Based on this initial assessment, if causes other than PH are identified and/or in case of low probability of PH, patients should be managed accordingly.
5.2.3. Step 3 (confirmation)
Patients should be referred to a PH centre for further evaluation in the following situations: (1) when an intermediate/high probability of PH is established; (2) in the presence of risk factors for PAH, or a history of PE. A comprehensive work-up should be performed, with the goal of establishing the differential diagnoses and distinguishing between the various causes of PH according to the current clinical classification. The PH centre is responsible for performing an invasive assessment according to the clinical scenario.
At any time, warning signs must be recognized, as they are associated with worse outcomes and warrant immediate intervention. Such warning signs include: rapidly evolving or severe symptoms (WHO-FC III/IV), clinical signs of RV failure, syncope, signs of low CO state, poorly tolerated arrhythmias, and compromised or deteriorated haemodynamic status (hypotension, tachycardia). Such cases must be immediately managed as inpatients for initial work-up at a nearby hospital or PH centre. The presence of RV dysfunction by echocardiography, elevated levels of cardiac biomarkers, and/or haemodynamic instability must prompt referral to a PH centre for immediate assessment.
This diagnostic process emphasizes the importance of sufficient awareness and collaboration between first-line, specialized medicine and PH centres. Effective and rapid collaboration between each partner permits earlier diagnosis and management, and improves outcomes.
Recommendation | Classa | Levelb |
Echocardiography | ||
Echocardiography is recommended as the first-line, non-invasive, diagnostic investigation in suspected PH [82, 84, 91] | I | B |
It is recommended to assign an echocardiographic probability of PH, based on an abnormal TRV and the presence of other echocardiographic signs suggestive of PH (see Table 10) [91, 92, 162] | I | B |
It is recommended to maintain the current threshold for TRV (>2.8 m/s) for echocardiographic probability of PH according to the updated haemodynamic definition [88] | I | C |
Based on the probability of PH by echocardiography, further testing should be considered in the clinical context (i.e. symptoms and risk factors or associated conditions for PAH/CTEPH) [92] | IIa | B |
In symptomatic patients with intermediate echocardiographic probability of PH, CPET may be considered to further determine the likelihood of PH [123, 163] | IIb | C |
Imaging | ||
Ventilation/perfusion or perfusion lung scan is recommended in patients with unexplained PH to assess for CTEPH [105] | I | C |
CT pulmonary angiography is recommended in the work-up of patients with suspected CTEPH [104] | I | C |
Routine biochemistry, haematology, immunology, HIV testing, and thyroid function tests are recommended in all patients with PAH, to identify associated conditions | I | C |
Abdominal ultrasound is recommended for the screening of portal hypertension [164] | I | C |
Chest CT should be considered in all patients with PH | IIa | C |
Digital subtraction angiography should be considered in the work-up of patients with CTEPH | IIa | C |
Other diagnostic tests | ||
Pulmonary function tests with DLCO are recommended in the initial evaluation of patients with PH [78] | I | C |
Open or thoracoscopic lung biopsy is not recommended in patients with PAH | III | C |
CPET, cardiopulmonary exercise testing; CT, computed tomography; CTEPH, chronic thrombo-embolic pulmonary hypertension; DLCO, Lung diffusion capacity for carbon monoxide; HIV, human immunodeficiency virus; PAH, pulmonary arterial hypertension; PH, pulmonary hypertension; TRV, tricuspid regurgitation velocity. aClass of recommendation. bLevel of evidence.
5.3. Screening and early detection
Despite the advent of PAH therapies that prevent clinical worsening [166–168] and effective interventions for CTEPH [102], the time from symptom onset to PH diagnosis remains at >2 years [169, 170], with most patients presenting with advanced disease. Decreasing the time to diagnosis may reduce emotional uncertainty in patients [171], reduce the use of health care resources, and enable treatment at an earlier stage when therapies may be more effective [172].
A proposed multifaceted approach [172] to facilitate an earlier diagnosis includes: (1) screening asymptomatic, high-risk groups (with high prevalence or where the diagnosis significantly impacts the proposed intervention), including individuals with SSc (prevalence: 5–19%) [173, 174], BMPR2 mutation carriers (14–42%) [33], first-degree relatives of patients with HPAH [148], and patients undergoing assessment for liver transplantation (2–9%) [175]; (2) early detection of symptomatic patients in at-risk groups with conditions such as portal hypertension [176], HIV infection (0.5%) [177], and non-SSc CTD, where the lower prevalence rates do not support asymptomatic screening; and (3) applying population-based strategies by deploying early detection approaches in PE follow-up clinics [178, 179], breathlessness clinics [172], or in at-risk patients identified from their health care behaviour and/or previous investigations [180].
Screening can be defined as the systematic application of a test or tests to identify at-risk, asymptomatic individuals. Screening approaches can also be extended to individuals who would not otherwise have sought medical attention on account of their symptoms, to facilitate early detection. Tools used to screen for PH have primarily been assessed, but not exclusively, in SSc [172, 174], and include blood biomarkers (NT-proBNP), ECG, echocardiography (primarily using estimates of sPAP at rest, but also exercise studies) [182], PFTs (DLCO and forced vital capacity [FVC]/DLCO ratio), and exercise testing including CPET (which has been used in combination with screening algorithms to reduce the need for RHC) [123, 163].
5.3.1. Systemic sclerosis
In SSc, the prevalence of PAH is 5–19% [174], with an annual incidence of developing PAH of 0.7–1.5% [183–185]. Evidence for the clinical value of detecting PAH early in SSc was provided by a screening programme [186], which showed less severe haemodynamic impairment and better survival in screened patients compared with a contemporaneous, non-screened cohort [187], providing a strong rationale for screening for PAH in patients with SSc.
The diagnostic accuracy of echocardiography or other tests alone in detecting PAH is suboptimal [173]. Several screening algorithms have been studied using a combination of clinical features, echocardiography, PFTs, and NT-proBNP to select patients with SSc for RHC (DETECT [173]; Australian Scleroderma Interest Group [ASIG] [188]). Such combined approaches have improved diagnostic accuracy compared with the use of echocardiography, NT-proBNP, or PFTs alone, and are able to prevent unnecessary RHC and identify patients with mPAP 21–24 mmHg [189]. Therefore, a multimodal approach is warranted when screening patients with SSc for PAH; the echocardiographic assessment should follow the strategy described in Section 5.1.5.
Beyond initial screening, the frequency with which screening should be undertaken in asymptomatic subjects with SSc is unclear. A study from the Australian Scleroderma Study Cohort, where annual screening was recommended (some patients were screened up to 10 times), noted that most patients were diagnosed with PAH at their first screening; however, those diagnosed on subsequent screening had a lower mPAP, PVR, and WHO-FC, and better survival than those diagnosed at first screening [190]. Based on current evidence, annual screening for PAH in patients with SSc is sufficient. Given the financial and emotional cost associated with regular screening, stratifying subjects with SSc into those at highest and lowest risk of PAH would be desirable. Risk factors for PAH include: (1) clinical and demographic factors (i.e. breathlessness, longer disease duration, sicca symptoms, digital ulceration, older age, and male sex); and (2) the results of investigations (e.g. positive anti-centromere antibody profile, mild ILD, low DLCO, elevated FVC/DLCO ratio, or elevated NT-proBNP) [174, 191]. A recent meta-analysis showed that reduced digital capillary density, as assessed by video-capillaroscopy, or progression to a severe active/late pattern of vascular involvement is also a risk factor for PAH [192]. In addition to identifying patients at increased risk of PAH, a simple prediction model integrating symptoms, DLCO, and NT-proBNP identified subjects at very low probability of PAH who could potentially avoid further specific testing for PH [183]. Furthermore, CPET may help to identify patients with SSc with a low risk of having PAH and thus to avoid unnecessary RHC [123].
The recommendations on screening for PAH in SSc have been established based on key narrative question 3 (Supplementary Data, Section 5.2).
5.3.2. BMPR2 mutation carriers
In the evolving list of genes known to be associated with PAH, experience is largely restricted to BMPR2 mutation carriers who carry a lifetime risk of developing PAH of ∼20%, with penetrance higher in female carriers (42%) compared with male carriers (14%) [33, 148, 193]. There is currently no accepted screening strategy for evaluating PAH in BMPR2 mutation carriers. At present, based on expert consensus, asymptomatic relatives who screen positive for PAH-causing mutations are often offered yearly screening echocardiography [25, 26] The DELPHI-2 study, which prospectively screened carriers and relatives, recently demonstrated a 9.1% pick up over 47±27 months of PAH, with 2/55 diagnosed at baseline and 3/55 at follow-up; this equates to an incidence of 2.3%/year [33]. The screening schedule included ECG, NT-proBNP, DLCO, echocardiography, CPET, and optional RHC; however, none of the cases would have been picked up by echocardiography alone. Screening programmes should adopt a multimodal approach, although the optimal strategy and screening period remains undefined and will require multinational, multicentre study.
5.3.3. Portal hypertension
An estimated 1–2% of patients with liver disease and portal hypertension develop PoPH [176, 194], which is of particular relevance in patients considered for transjugular portosystemic shunting or liver transplantation. In such patients, echocardiography is recommended to screen for PAH, even in the absence of symptoms. By using echocardiography, sPAP can be measured in ∼80% of patients with portal hypertension, which aids decisions to perform RHC. In patients assessed for liver transplantation, one study showed that an sPAP of >50 mmHg had 97% sensitivity and 77% specificity for detecting moderate-to-severe PAH [195]. Other investigators have recommended RHC when sPAP is >38 mmHg [196]. When screening for PoPH, it is advised to assess the echocardiographic probability of PH (see Section 5.1.5). In agreement with the International Liver Transplant Society, for patients awaiting liver transplantation, it is recommended to reassess for PAH annually, although the optimal interval remains unclear [175].
5.3.4. Pulmonary embolism
Chronic thrombo-embolic pulmonary hypertension is an uncommon and under-diagnosed complication of acute PE [112]. The reported cumulative incidence of CTEPH after acute, symptomatic PE ranges 0.1–11.8%, depending on the collective investigated [112, 178, 197–199]. A systematic review and meta-analysis reported a CTEPH incidence of 0.6% in all patients with acute PE, 3.2% in survivors, and 2.8% in survivors without major comorbidities [178]. A multicentre, observational, screening study reported a CTEPH incidence of 3.7/1000 patient-years and a 2 year cumulative incidence of 0.79% following acute PE [200]. A recent prospective observational study (FOCUS, Follow-up After Acute Pulmonary Embolism) showed a cumulative 2 year incidence of 2.3% and 16.0% for CTEPH and post-PE impairment, respectively, which were both associated with a higher risk of rehospitalization and death [201]. Due to insufficient awareness, some patients may have a delayed diagnosis of CTEPH because they may initially be misclassified as acute PE [112]. In this context, the current guidelines do not recommend routine follow-up of patients with PE by imaging methods of the pulmonary vascular tree, but suggest evaluating the index imaging test used to diagnose acute PE for signs of CTEPH. Echocardiography is the preferred first-line diagnostic test in patients with suspected CTEPH [103].
Up to 50% of patients have persistent perfusion defects after an acute PE; however, the clinical relevance is unclear [202–204]. All patients in whom symptoms can be attributed to post-thrombotic deposits within PAs are considered to have CTEPD, with or without PH [54]. While persistent dyspnoea is common after acute PE [205], the prevalence of CTEPD without PH is unknown and requires further study (see Section 10.1). A study exploring screening for CTEPH following acute PE identified, using echocardiography, a low yield of additional CTEPH diagnoses in asymptomatic patients [206]. Current PE guidelines recommend that further diagnostic evaluation may be considered in asymptomatic patients with risk factors for CTEPH at 3–6 months’ follow-up [103, 207]. Approaches to early detection of CTEPH following acute PE are based on identifying patients at increased risk [208]. In patients with persistent or new-onset dyspnoea after PE, non-invasive approaches use echocardiography to assess for PH and cross-sectional imaging to assess for persistent perfusion defects. Limited data exist on strategies using DECT, CT lung subtraction iodine mapping, or 3D MR perfusion imaging. Scoring systems, including the Leiden CTEPH rule-out criteria [206, 209] can be used to inform diagnostic strategies. Cardiopulmonary exercise testing may identify characteristic features of exercise limitation due to PVD, or suggest an alternative diagnosis. The optimal timing for assessing symptoms to aid early detection of CTEPH may be 3–6 months after acute PE, coinciding with the routine evaluation of anticoagulant treatment, but earlier assessment may be necessary in highly symptomatic or deteriorating patients [54, 103].
Recommendations | Classc | Levelb |
Systemic sclerosis | ||
In patients with SSc, an annual evaluation of the risk of having PAH is recommended [183, 186] | I | B |
In adult patients with SSc with >3 years' disease duration, an FVC ≥40%, and a DLCO <60%, the DETECT algorithm is recommended to identify asymptomatic patients with PAH [173, 186] | I | B |
In patients with SSc, where breathlessness remains unexplained following non-invasive assessment, RHC is recommended to exclude PAH [185–187] | I | C |
Assessing the risk of having PAH, based on an evaluation of breathlessness, in combination with echocardiogram or PFTs and BNP/NT-proBNP, should be considered in patients with SSc [172, 173, 186, 188, 190] | IIa | B |
Policies to evaluate the risk of having PAH should be considered in hospitals managing patients with SSc | IIa | C |
In symptomatic patients with SSc, exercise echocardiography or CPET, or CMR may be considered to aid decisions to perform RHC | IIb | C |
In patients with CTD with overlap features of SSc, an annual evaluation of the risk of PAH may be considered | IIb | C |
CTEPH/CTEPD | ||
In patients with persistent or new-onset dyspnoea or exercise limitation following PE, further diagnostic evaluation to assess for CTEPH/CTEPD is recommended [103] | I | C |
For symptomatic patients with mismatched perfusion lung defects beyond 3 months of anticoagulation for acute PE, referral to a PH/CTEPH centre is recommended after considering the results of echocardiography, BNP/NT-proBNP, and/or CPET [203, 206] | I | C |
Other | ||
Counselling regarding the risk of PAH, and annual screening is recommended in individuals who test positive for PAH-causing mutations and in first-degree relatives of patients with HPAH [33] | I | B |
In patients referred for liver transplantation, echocardiography is recommended as a screening test for PH | I | C |
Further tests (echocardiography, BNP/NT-proBNP, PFTs, and/or CPET) should be considered in symptomatic patients with CTD, portal hypertension, or HIV to screen for PAH [172] | IIa | B |
BNP, brain natriuretic peptide; CMR, cardiac magnetic resonance; CPET, cardiopulmonary exercise testing; CTD, connective tissue disease; CTEPD, chronic thrombo-embolic pulmonary disease; CTEPH, chronic thrombo-embolic pulmonary hypertension; DLCO, Lung diffusion capacity for carbon monoxide; FVC, forced vital capacity; HIV, human immunodeficiency virus; HPAH, heritable pulmonary arterial hypertension; NT-proBNP, N-terminal pro-brain natriuretic peptide; PAH, pulmonary arterial hypertension; PE, pulmonary embolism; PFT, pulmonary function test; PH, pulmonary hypertension; RHC, right heart catheterisation; SSc, systemic sclerosis. aClass of recommendation. bLevel of evidence.
6. Pulmonary arterial hypertension (group 1)
6.1. Clinical characteristics
The symptoms of PAH are non-specific and mainly related to progressive RV dysfunction (see Section 5.1.1) as a consequence of progressive pulmonary vasculopathy (Figure 7). The presentation of PAH may be modified by diseases that are associated with PAH, as well as comorbidities. More detailed descriptions of the individual PAH subsets are reported in Section 7.
6.2. Severity and risk assessment
6.2.1. Clinical parameters
Clinical assessment is a key part of evaluating patients with PAH, as it provides valuable information for determining disease severity, improvement, deterioration, or stability. At follow-up, changes in WHO-FC (Table 15), episodes of chest pain, arrhythmias, haemoptysis, syncope, and signs of right HF provide important information. Physical examination should assess heart rate, rhythm, blood pressure, cyanosis, enlarged jugular veins, oedema, ascites, and pleural effusions. The WHO-FC is one of the strongest predictors of survival, both at diagnosis and follow-up [210–212], and worsening WHO-FC is one of the most alarming indicators of disease progression, which should trigger further investigations to identify the cause(s) of clinical deterioration [210, 213, 214].
6.2.2. Imaging
Imaging of the heart plays an essential role in the follow-up of patients with PAH. Several echocardiographic and cMRI parameters have been proposed to monitor RV function during the course of PAH. Table S2 provides a list of imaging parameters and relative cut-off values associated with increased and decreased risk of adverse events.
6.2.2.1. Echocardiography
Echocardiography is a widely available imaging modality and is readily performed at the patient's bedside. It is crucial that a high-quality echocardiographic assessment by PH specialists is undertaken to reduce intraobserver and interobserver variability. Of note, estimated sPAP at rest is not prognostic and irrelevant to therapeutic decision-making [212, 215, 216]. An increase in sPAP does not necessarily reflect disease progression and a decrease in sPAP does not necessarily reflect improvement.
Despite the complex geometry of the right heart, echocardiographic surrogates of the true right heart dimensions, which include a description of RV and RA areas, and the LV eccentricity index, provide useful clinical information in PAH [217, 218]. Right ventricular dysfunction can be evaluated measuring fractional area change, TAPSE, tissue Doppler, and 2D speckle tracking myocardial strain recording of RV free-wall motion, all of which represent isovolumetric and ejection-phase indices of load-induced RV pump failure [219–224]. The rationale for the reported measurements is strong, as RV systolic function metrics assess the adaptation of RV contractility to increased afterload, and increased right heart dimension and inferior vena cava dilation reflect failure of this mechanism, hence maladaptation [225]. Pericardial effusion and tricuspid regurgitation (TR) grading further explore RV overload and are of prognostic relevance in these patients [218, 226–228]. All of these variables are physiologically interdependent and their combination provides additional prognostic information over single measurements [223].
Echocardiography also enables combined parameters to be measured, such as the TAPSE/sPAP ratio, which is tightly linked to RV–PA coupling and predicts outcome [96, 97]. Echocardiographic measurements of RV and RA sizes combined with LV eccentricity index are crucial for assessing RV reverse remodelling as an emerging marker of treatment efficacy [220, 229]. Three-dimensional echocardiography may achieve better estimation than standard 2D assessment, but underestimations of volumes and ejection fraction have been reported, and technical issues are, as yet, unresolved [230].
6.2.2.2. Cardiac magnetic resonance imaging
The role of cMRI in evaluating patients with PAH has been addressed in several studies, and RV volumes, RVEF, and SV are essential prognostic determinants in PAH [225, 231–236]. In patients with PAH, initial cMRI measurements added prognostic value to current risk scores [231, 232]. In addition, risk assessment at 1 year of follow-up based on cMRI was at least equal to risk assessment based on RHC [237]. The cMRI risk-assessment variables based on the current literature are included in Table 16 [117, 225, 231–235, 237]. The stroke volume index (SVI) cut-off levels are based on the consensus of the literature [238]; a change of 10 mL in SV (LV end-diastolic volume−LV end-systolic volume) during follow-up is considered clinically significant [239]. The value of cMRI in the follow-up of patients has been shown in several studies, and cMRI enables treatment effects to be monitored and treatment strategies adapted in time to prevent clinical failure [240–242].
6.2.3. Haemodynamics
Cardiopulmonary haemodynamics assessed by RHC provide important prognostic information, both at the time of diagnosis and at follow-up [129, 212, 213, 216, 238, 243–245, 247, 248]. Currently available risk-stratification tools include haemodynamic variables for prognostication: RAP and PVR in REVEAL risk scores [213, 249, 250], and RAP, CI, and SvO2 in the ESC/ERS risk-stratification table [25, 26]. The mPAP provides little prognostic information, except in acute vasodilator responders [129]. A recent study from France, which combined clinical and haemodynamic parameters, found that WHO-FC, 6MWD, RAP, and SVI (but not SV and SvO2) were independent predictors of outcome [238].
To refine the risk-stratification table (Table 16), SVI criteria are now added with the cut-off values of >38 mL/m2 and <31 mL/m2 to determine low-risk and high-risk status, respectively [238].
The optimal timing of follow-up RHC has not been determined. While some centres regularly perform invasive follow-up assessments, others perform them as clinically indicated, and there is no evidence that any of these strategies is associated with better outcomes (Table 17).
6.2.4. Exercise capacity
The 6-minute walking test (6MWT) is the most widely used measure of exercise capacity in PH centres. The 6MWT is easy to perform, inexpensive, and widely accepted by patients, health professionals, and medicines agencies as an important and validated variable in PH. As with all PH assessments, 6MWT results must always be interpreted in the clinical context. The 6MWD is influenced by factors such as sex, age, height, weight, comorbidities, need for oxygen, learning curve, and motivation. Test results are usually given in absolute distance (metres) rather than the percentage of predicted values. Change in 6MWD is one of the most commonly used parameters in PAH clinical trials as a primary endpoint, key secondary endpoint, or component of clinical worsening [251]. A recent investigation showed that the best absolute-threshold values for 1 year mortality and 1 year survival, respectively, were 165 m and 440 m, respectively [252]. Improvements in 6MWD have had less predictive value than deterioration on key clinical outcomes (mortality and survival) [250, 252, 253]. These results are consistent with observations from clinical trials and registries [254, 255]; however, there is no single threshold that would apply to all patients [256]. Some studies have also suggested that adding SaO2 measured by pulse oximetry and heart rate responses may improve prognostic relevance [246, 257]. Hypoxaemia observed during the 6MWT is associated with worse survival, but these findings still await confirmation in large multicentre studies.
The incremental shuttle walking test (ISWT) is an alternative maximal test for assessing patients with PAH. The ISWT has a potential advantage over the 6MWT in that it does not have a ceiling effect; furthermore, it keeps the simplicity of a simple-to-perform field test, in contrast to CPET. However, the ISWT experience in PAH is currently limited [258].
Cardiopulmonary exercise testing is a non-invasive method for assessing functional capacity and exercise limitation. It is usually performed as a maximal exercise test, and is safe even in patients with severe exercise limitation [259, 260]. Most PH centres use an incremental ramp protocol, although the test has not yet been standardized for this patient population. Robust prognostic evidence for peak VO2 and VE/VCO2 has been found in three studies, all powered for multivariable analysis [261–263]. When associated with SVI, peak VO2 provided useful information to further stratify patients with PAH at intermediate risk [264]. However, the added value of CPET on top of common clinical and haemodynamic variables remains largely unexplored.
6.2.5. Biochemical markers
Considerable efforts have been made to identify additional biomarkers of PVD, addressing prognosis [265–272], diagnosis, and differentiation of PH subtypes [270, 273–276], as well as PAH treatment response [266]. Emerging proteins related to PAH and vascular remodelling include bone morphogenetic proteins 9 and 10 and translationally controlled tumour protein [270, 277, 278]. Proteome-wide screening in IPAH and HPAH identified a multimarker panel with prognostic information in addition to the REVEAL risk score [271]. Another study found that early development of SSc-associated PAH (PAH-SSc) was predicted by high circulating levels of C-X-C motif chemokine 4 in patients with SSc [276]. However, none of these biomarkers have been introduced in clinical practice.
Thus, BNP and NT-proBNP remain the only biomarkers routinely used in clinical practice at PH centres, correlating with myocardial stress and providing prognostic information [279]. Brain natriuretic peptide and NT-proBNP are not specific for PH, as they can be elevated in other forms of heart disease, exhibiting great variability. The previously proposed cut-off levels of BNP (<50, 50–300, and >300 ng/L) and NT-proBNP (<300, 300–1400, and >1400 ng/L) for low, intermediate, and high risk, respectively, in the ESC/ERS risk-assessment model at baseline and during follow-up are prognostic for long-term outcomes and can be used to predict response to treatment [266]. Refined cut-off values for BNP (<50, 50–199, 200–800, and >800 ng/L) and NT-pro-BNP (<300, 300–649, 650–1100, and >1100 ng/L) for low, intermediate–low, intermediate–high, and high risk, respectively, have recently been introduced as part of a four-strata risk-assessment strategy (see Section 6.2.7) [280].
6.2.6. Patient-reported outcome measures
A patient-reported outcome measure (PROM) is a term for health outcomes that are ‘self-reported’ by the patient. It is the patient's experience of living with PH and its impact on them and their caregivers, including symptomatic, intellectual, psychosocial, spiritual, and goal-orientated dimensions of the disease and its treatment. Despite treatment advances improving survival, patients with PAH present with a range of non-specific yet debilitating symptoms, which affect health-related quality of life (HR-QoL) [281, 282].
Patient-reported outcome measures remain an underused outcome measure. Tools validated in patients with PAH should be used to assess HR-QoL [282, 283] in individual patients. There has been a reliance on generic PROMs, which have been studied in patients with PAH but may lack sensitivity to detect changes in PAH [284, 285]. To address this, a number of PH-specific HR-QoL instruments have been developed and validated (e.g. Cambridge Pulmonary Hypertension Outcome Review [CAMPHOR] [286], emPHasis-10 [282, 287], Living with Pulmonary Hypertension [288], and Pulmonary Arterial Hypertension-Symptoms and Impact [PAH-SYMPACT]) [289]. These disease-specific PROMs track functional status, clinical deterioration, and prognosis in PAH, and are more sensitive to the differences in the risk status than generic PROMs [290, 291]. In addition, HR-QoL scores provide independent prognostic information [287].
6.2.7. Comprehensive prognostic evaluation, risk assessment, and treatment goals
In the 2015 ESC/ERS Guidelines for the diagnosis and treatment of PH, risk assessment was based on a multiparametric approach using a three-strata model to classify patients at low, intermediate, or high risk of death. Originally, these strata were based on estimated 1 year mortality rates of <5%, 5–10%, and >10%, respectively [25, 26]. Since then, registry data have shown that observed 1 year mortality rates in the intermediate- and high-risk groups were sometimes higher than predicted (i.e. up to 20% in the intermediate-risk group and >20% in the high-risk group). These numbers have been updated accordingly in the revised three-strata risk model (Table 16) [292–294].
Several abbreviated approaches of the 2015 ESC/ERS risk-stratification tool have been introduced and independently validated using the Swedish Pulmonary Arterial Hypertension Registry (SPAHR) [292], the Comparative, Prospective Registry of Newly Initiated Therapies for PH (COMPERA) [293], and the French PH Registry (FPHR) [295]. Other risk-stratification tools have been developed from the US REVEAL, including the REVEAL 2.0 risk score calculator, and an abridged version (REVEAL Lite 2) [249, 296]. In all these studies, WHO-FC, 6MWD, and BNP/NT-proBNP emerged as the variables with the highest predictive value.
The main limitation of the 2015 ESC/ERS three-strata, risk-assessment tool is that 60–70% of the patients are classified as intermediate risk [292–295, 297–303]. An initial attempt to substratify the intermediate-risk group has been proposed, using a modified mean score in the SPAHR equation (with low–intermediate, 1.5–1.99 and high–intermediate, 2.0–2.49 as cut-offs), where the high–intermediate group was associated with worse survival [302]. There have also been attempts to further improve risk stratification by exploring the additional value of new biomarkers [304], or by measuring RV structure and function by echocardiography and cMRI [231, 305, 306]. Other strategies have included incorporating renal function [307] or combining 6MWD with TAPSE/sPAP ratio [96, 97]; however, all of these strategies have to be further validated.
Two recent registry studies have evaluated a four-strata, risk-assessment tool based on refined cut-off levels for WHO-FC, 6MWD, and NT-proBNP (Table 18) [280, 308]. Patients were categorized as low, intermediate–low, intermediate–high, or high risk. Together, these studies included >4000 patients with PAH and showed that the four-strata model performed at least as well as the three-strata model in predicting mortality. The four-strata model predicted survival in patients with IPAH, HPAH, DPAH, and PAH associated with CTD (including the SSc subgroup), and in patients with PoPH. The observed 1-year mortality rates in the four risk strata were 0–3%, 2–7%, 9–19%, and >20%, respectively. Compared with the three-strata model, the four-strata model was more sensitive to changes in risk from baseline to follow-up, and these changes were associated with changes in the long-term mortality risk. The main advantage of the four-strata model over the three-strata model is better discrimination within the intermediate-risk group, which helps guide therapeutic decision-making (see Section 6.3.4). For these reasons, the four-strata model is included in the updated treatment algorithm (see Figure 9). However, the three-strata model is maintained for initial assessment, which should be comprehensive and include echocardiographic and haemodynamic variables, for which cut-off values for the four-strata model have yet to be established.
Several studies have identified WHO-FC, 6MWD, and BNP/NT-proBNP as the strongest prognostic predictors [293, 295, 296]. With the abbreviated risk-assessment tools, missing values become an important limitation. REVEAL Lite 2 provides accurate prediction when one key variable (WHO-FC, 6MWD, or BNP/NT-proBNP) is unavailable, but is no longer accurate when two of these variables are missing [293, 296]. The original three-strata SPAHR/COMPERA risk tool was developed with at least two variables available, while the four-strata model was developed and validated in patients for whom all three variables were available. It is therefore recommended to use at least these three variables for risk stratification. However, two components may be used when variables are missing, especially when a functional criterion (WHO-FC or 6MWD) is combined with BNP or NT-proBNP [296].
Collectively, the available studies support a risk-based, goal-orientated treatment approach in patients with PAH, where achieving and/or maintaining a low-risk status is favourable and recommended (key narrative question 4, Supplementary Data, Section 6.1) [298, 300, 303, 309, 310]. For risk stratification at diagnosis, use of the three-strata model is recommended taking into account as many factors as possible (Table 16), with a strong emphasis on disease type, WHO-FC, 6MWD, BNP/NT-proBNP, and haemodynamics. At follow-up, the four-strata model (Table 18) is recommended as a basic risk-stratification tool, but additional variables should be considered as needed, especially right heart imaging and haemodynamics. At any stage, individual factors such as age, sex, disease type, comorbidities, and kidney function should also be considered.
Recommendations | Classa | Levelb |
It is recommended to evaluate disease severity in patients with PAH with a panel of data derived from clinical assessment, exercise tests, biochemical markers, echocardiography, and haemodynamic evaluations [212, 213, 216, 249, 292, 293, 295, 296, 302, 307] | I | B |
Achieving and maintaining a low-risk profile on optimised medical therapy is recommended as a treatment goal in patients with PAH [210, 212, 213, 216, 298, 300, 303, 309, 310] | I | B |
For risk stratification at the time of diagnosis, the use of a three-strata model (low, intermediate, and high risk) is recommended, taking into account all available data, including haemodynamics [292, 293, 295] | I | B |
For risk stratification during follow-up, the use of a four-strata model (low, intermediate-low, intermediate-high, and high risk) based on WHO-FC, 6MWD, and BNP/NT-proBNP is recommended, with additional variables taken into account as necessary [280, 308] | I | B |
In some PAH aetiologies and in patients with comorbidities, optimisation of therapy should be considered on an individual basis while acknowledging that a low-risk profile is not always achievable [293, 294, 299, 311] | IIa | B |
6MWD, 6-minute walking distance; BNP, brain natriuretic peptide; NT-proBNP, N-terminal pro-brain natriuretic peptide; PAH, pulmonary arterial hypertension; WHO-FC, World Health Organization functional class. aClass of recommendation. bLevel of evidence.
6.3. Therapy
According to the revised haemodynamic definition, PAH may be diagnosed in patients with mPAP >20 mmHg and PVR >2 WU. Yet, the efficacy of drugs approved for PAH has only been demonstrated in patients with mPAP ≥25 mmHg and PVR >3 WU (see Supplementary Data, Table S1). No data are available for the efficacy of drugs approved for PAH in patients whose mPAP is <25 mmHg and whose PVR is <3 WU. Hence, for such patients, the efficacy of drugs approved for PAH has not been established. The same is true for patients with exercise PH, who, by definition, do not fulfil the diagnostic criteria for PAH. Patients at high risk of developing PAH, for instance patients with SSc or family members of patients with HPAH, should be referred to a PH centre for individual decision-making.
6.3.1. General measures
Managing patients with PAH requires a comprehensive treatment strategy and multidisciplinary care. In addition to applying PAH drugs, general measures and care in special situations represent integral components of optimized patient care. In this context, the systemic consequences of PH and right-sided HF, often contributing to disease burden, should be appropriately managed [119].
6.3.1.1. Physical activity and supervised rehabilitation
The 2015 ESC/ERS Guidelines for the diagnosis and treatment of PH suggested that patients with PAH should be encouraged to be active within symptom limits [25, 26]. Since then, additional studies have shown the beneficial impact of exercise training on exercise capacity (6MWD) and quality of life [312–316]. A large, randomized controlled trial (RCT) in 11 centres across 10 European countries, including 116 patients with PAH/CTEPH on PAH drugs, showed a significant improvement in 6MWD of 34.1±8.3 m, quality of life, WHO-FC, and peak VO2 compared with standard of care [315]. Since most of the studies included patients who were stable on medical treatment, patients with PAH should be treated with the best standard of pharmacological treatment and be in a stable clinical condition before embarking on a supervised rehabilitation programme. Establishing specialized rehabilitation programmes for patients with PH would further enhance patient access to this intervention [317].
6.3.1.2. Anticoagulation
There are several reasons to consider anticoagulation in patients with PAH. Histopathological specimens from PAH patients’ lungs have shown in situ thrombosis of pulmonary vessels. Patients with CHD or PA aneurysms may develop thrombosis of the central PAs. Abnormalities in the coagulation and fibrinolytic system indicating a pro-coagulant state have been reported in patients with PAH [318].
Data from RCTs on anticoagulation in PAH are lacking, and registry data have yielded conflicting results. The largest registry analysis so far suggested a potential survival benefit associated with anticoagulation in patients with IPAH [319], but this finding was not confirmed by others [320]. Two recent meta-analyses also concluded that using anticoagulants may improve survival in patients with IPAH [321, 322]; however, none of the included studies were methodologically robust. Despite the lack of evidence, registry data obtained between 2007 and 2016 showed that anticoagulation was used in 43% of patients with IPAH [293]. In PAH associated with SSc, registry data and meta-analyses uniformly indicated that anticoagulation may be harmful [320–322]. In CHD, there are also no RCTs on anticoagulation. There is also no consensus about the use of anticoagulants in patients who have permanent i.v. lines for therapy with prostacyclin analogues; this is left to local centre practice.
As anticoagulation is associated with an increased bleeding risk, and in the absence of robust data, no general recommendation has been made for or against the use of anticoagulants in patients with PAH; therefore, individual decision-making is required.
6.3.1.3. Diuretics
Right HF is associated with systemic fluid retention, reduced renal blood flow, and activation of the renin–angiotensin–aldosterone system. Increased right-sided filling pressures are transmitted to the renal veins, increasing interstitial and tubular hydrostatic pressure within the encapsulated kidney, which decreases net glomerular filtration rate and oxygen delivery [119].
Avoiding fluid retention is one of the key objectives in managing patients with PH. Once these patients develop signs of right-sided HF and oedema, restricting fluid intake and using diuretics is recommended. The three main classes of diuretics—loop diuretics, thiazides, and mineralocorticoid receptor antagonists—are used as monotherapy or in combination, as determined by the patient's clinical need and kidney function. Patients requiring diuretic therapy should be advised to regularly monitor their body weight and to seek medical advice in case of weight gain. Close collaboration between patients, PH centres, especially PH nurses, and primary care physicians plays a vital role. Kidney function and serum electrolytes should be regularly monitored, and intravascular volume depletion must be avoided as it may cause a further decline in CO and systemic blood pressure. Physicians should bear in mind that fluid retention and oedema may not necessarily signal right-sided HF, but may also be a side effect of PAH therapy [323].
6.3.1.4. Oxygen
Although oxygen administration reduces PVR and improves exercise tolerance in patients with PAH, there are no data to suggest that long-term oxygen therapy has sustained benefits on the course of the disease. Most patients with PAH, except those with CHD and pulmonary-to-systemic shunts, have minor degrees of arterial hypoxaemia at rest, unless they have a patent foramen ovale. Data show that nocturnal oxygen therapy does not modify the natural history of advanced Eisenmenger syndrome [324]. In the absence of robust data on the use of oxygen in patients with PAH, guidance is based on evidence in patients with COPD [325]; when PaO2 is <8 kPa (60 mmHg; alternatively, SaO2 <92%) on at least two occasions, patients are advised to take oxygen to achieve a PaO2 >8 kPa. Ambulatory oxygen may be considered when there is evidence of symptomatic benefit and correctable desaturation on exercise [326, 327]. Nocturnal oxygen therapy should be considered in case of sleep-related desaturation [328].
6.3.1.5. Cardiovascular drugs
No data from rigorous clinical trials are available on the usefulness and safety of drugs that are effective in systemic hypertension or left-sided HF, such as angiotensin-converting enzyme inhibitors, angiotensin receptor blockers (ARBs), angiotensin receptor–neprilysin inhibitors (ARNIs), sodium–glucose cotransporter-2 inhibitors (SGLT-2is), beta-blockers, or ivabradine in patients with PAH. In this group of patients, these drugs may lead to potentially dangerous drops in blood pressure, heart rate, or both. Likewise, the efficacy of digoxin/digitoxin has not been documented in PAH, although these drugs may be administered to slow ventricular rate in patients with PAH who develop atrial tachyarrhythmias.
6.3.1.6. Anaemia and iron status
Iron deficiency is common in patients with PAH and is defined by serum ferritin <100 µg/L, or serum ferritin 100–299 µg/L and transferrin saturation <20% [329]. The underlying pathological mechanisms are complex [330–333]. In patients with PAH, iron deficiency is associated with impaired myocardial function, aggravated symptoms, and increased mortality risk [333, 334]. Based on these data, regular monitoring of iron status (serum iron, ferritin, transferrin saturation, soluble transferrin receptors) is recommended in patients with PAH.
In patients with severe iron deficiency anaemia (Hb <7–8 g/dL), i.v. supplementation is recommended [335–337]. Oral iron formulations containing ferrous (Fe2+) sulfate, ferrous gluconate, and ferrous fumarate are often poorly tolerated, and drug efficacy may be impaired in patients with PAH [330, 331]. Ferric maltol is a new, orally available formulation of ferric (Fe3+) iron and maltol. One small, open-label study suggested good tolerability and efficacy in patients with severe PH with mild-to-moderate iron deficiency and anaemia [338]. In contrast, two small, 12 week, randomized, cross-over trials studying iron supplementation in PAH patients without anaemia provided no significant clinical benefit [339]. Randomized controlled trials comparing oral and i.v. iron supplementation in patients with PAH are lacking.
6.3.1.7. Vaccination
As a general health care measure, it is recommended that patients with PAH be vaccinated at least against influenza, Streptococcus pneumoniae, and SARS-CoV-2.
6.3.1.8. Psychosocial support
Receiving a diagnosis of PH—often after a substantial delay—and experiencing the physical limitations have a substantial impact on psychological, emotional, and social aspects of patients and their families. Symptoms of depression and anxiety, as well as adjustment disorders, have a high prevalence in patients with PAH. Pulmonary arterial hypertension also has grave repercussions on ability to work and income [281, 340–344].
Empathic and hopeful communication is essential for physicians caring for patients with PAH. Awareness and knowledge about the disease and its treatment options empower patients to engage in shared decision-making. Adequate diagnostic screening tools are the key to identifying patients in need of referral for psychological/psychiatric support, including psychopharmacological medication [345], or social assistance. Patient support groups may play an important role, and patients should be advised to join such groups. Given the life-limiting character of PAH, advanced care planning with referral to specialist palliative care services should be supported at the right time [346].
6.3.1.9. Adherence to treatments
Adhering to medical therapy is key to successfully managing PAH. In general, factors that affect adherence are patient related (e.g. demographics, cognitive impairment, polypharmacy, adverse reactions/side effects, psychological health, health literacy, patient understanding of the treatment rationale, and comorbidities), physician related (expertise, awareness of guidelines, and multidisciplinary team approach), and health care system related (work setting, access to treatments, and cost) [347].
Recent studies have indicated that adherence to drug therapy in patients with PAH may be suboptimal [348, 349]. Given the complexity of PAH treatment, potential side effects, and risks associated with treatment interruptions, adherence should be periodically monitored by a member of the multidisciplinary team, to identify non-adherence and any changes to the treatment regimen spontaneously triggered by patients or non-expert physicians. To promote adherence, it is important to ensure that patients are involved in care decisions and appropriately informed about treatment options and rationale, expectations, side effects, and potential consequences of non-adherence. Patients should be advised that any changes in treatment should be made in cooperation with the PH centre.
6.3.2. Special circumstances
6.3.2.1. Pregnancy and birth control
6.3.2.1.1. Pregnancy
Historically, pregnancy in women with PAH and other forms of severe PH has been associated with maternal mortality rates of up to 56% and neonatal mortality rates of up to 13% [350]. With improved treatment of PAH and new approaches to managing women during pregnancy and the peri-partum period, maternal mortality has declined but remains high, ranging 11–25% [351–355]. For these reasons, previous ESC/ERS Guidelines for the diagnosis and treatment of PH have recommended that patients with PAH should avoid pregnancy [25, 26]. However, there are reports of favourable pregnancy outcomes in women with PH, including, but not limited to, women with IPAH who respond to CCB therapy [353, 354, 356, 357]. Nonetheless, pregnancy remains associated with unforeseeable risks, and may accelerate PH progression [358]. Women with PH can deteriorate at any time during or after pregnancy. Therefore, physicians have a responsibility to inform patients about the risks of pregnancy, so that women and their families can make informed decisions.
Women with poorly controlled disease, indicated by an intermediate- or high-risk profile and signs of RV dysfunction, are at high risk of adverse outcomes; in the event of pregnancy, they should be carefully counselled and early termination should be advised. For patients with well-controlled disease, a low-risk profile, and normal or near-normal resting haemodynamics who consider becoming pregnant, individual counselling and shared decision-making are recommended. In such cases, alternatives such as adoption and surrogacy may also be explored. Pre-conception genetic counselling should also be considered in HPAH.
Women with PH who become pregnant or present during pregnancy with newly diagnosed PAH should be treated, whenever possible, in centres with a multidisciplinary team experienced in managing PH in pregnancy. If pregnancy is continued, PAH therapy may have to be adjusted. It is recommended to stop endothelin receptor antagonists (ERAs), riociguat, and selexipag because of potential or unknown teratogenicity [359]. Despite limited evidence, CCBs, PDE5is, and inhaled/i.v./subcutaneous (s.c.) prostacyclin analogues are considered safe during pregnancy [356, 360].
Pregnancy in PH is a very sensitive topic and requires empathic communication. Psychological support should be offered whenever needed.
6.3.2.1.2. Contraception
Women with PH of childbearing potential should be provided with clear contraceptive advice, considering the individual needs of the woman but recognizing that the implications of contraceptive failure are significant in PH. With appropriate use, many forms of contraception, including oral contraceptives, are highly effective. In patients treated with bosentan, reduced efficacy of hormonal contraceptives should be carefully considered [361]. Using hormonal implants or an intrauterine device are alternative options with low failure rates. Surgical sterilization may be considered but is associated with peri-operative risks. Emergency post-coital hormonal contraception is safe in PH.
6.3.2.2. Surgical procedures
Surgical procedures in patients with PH are associated with an elevated risk of right HF and death. In a prospective, multinational registry including 114 patients with PAH who underwent non-cardiac and non-obstetric surgery, the peri-operative mortality rate was 2% in elective procedures and 15% in emergency procedures [362]. The mortality risk was associated with the severity of PH. The decision to perform surgery should be made by a multidisciplinary team involving a PH physician, and must be based on an individual risk:benefit assessment considering various factors, including indication, urgency, PH severity, and patient preferences. Risk scores to predict the peri-operative mortality risk have been developed but require further validation [363]. General recommendations cannot be made. The same is true regarding the preferred mode of anaesthesia. Pre-operative optimization of PAH therapy should be attempted whenever possible (see also the 2022 ESC Guidelines on cardiovascular assessment and management of patients undergoing non-cardiac surgery) [364].
6.3.2.3. Travel and altitude
Hypobaric hypoxia may induce arterial hypoxaemia, additional hypoxic pulmonary vasoconstriction, and increased RV load in PAH [365, 366]. Cabin aircraft pressures are equivalent to altitudes up to 2438 m [367], at which the PaO2 decreases to that of an inspired O2 fraction of 15.1% at sea level [365]. However, evidence suggests that short-term (less than 1 day) normobaric hypoxia is generally well tolerated in clinically stable patients with PAH [365, 368–372]. In-flight oxygen administration is advised for patients using oxygen at sea level and for those with PaO2 <8 kPa (60 mmHg) or SaO2 <92% [25, 26, 325, 369, 372]. An oxygen flow rate of 2 L/min will raise inspired oxygen pressure to values as at sea level, and patients already using oxygen at sea level should increase their oxygen flow rate [25, 26, 373].
As the effects of moderate to long-term (hours–days) hypoxia exposure in PAH remain largely unexplored [374, 375], patients should avoid altitudes >1500 m without supplemental oxygen [25, 26, 369]. However, patients with PAH who are not hypoxaemic at sea level have tolerated day trips to 2500 m reasonably well [376]. Patients should travel with written information about their disease, including a medication list, bring extra doses of their medication, and be informed about local PH centres near their travel destination [25, 26].
Recommendations | Classa | Levelb |
General measures | ||
Supervised exercise training is recommended in patients with PAH under medical therapy [314, 315, 317] | I | A |
Psychosocial support is recommended in patients with PAH | I | C |
Immunization of patients with PAH against SARS-CoV-2, influenza, and Streptococcus pneumoniae is recommended | I | C |
Diuretic treatment is recommended in patients with PAH with signs of RV failure and fluid retention | I | C |
Long-term oxygen therapy is recommended in patients with PAH whose arterial blood oxygen pressure is <8 kPa (60 mmHg)c | I | C |
In the presence of iron-deficiency anaemia, correction of iron status is recommended in patients with PAH | I | C |
In the absence of anaemia, iron repletion may be considered in patients with PAH with iron deficiency | IIb | C |
Anticoagulation is not generally recommended in patients with PAH but may be considered on an individual basis | IIb | C |
The use of ACEis, ARBs, ARNIs, SGLT-2is, beta-blockers, or ivabradine is not recommended in patients with PAH unless required by comorbidities (i.e. high blood pressure, coronary artery disease, left HF, or arrhythmias) | III | C |
Special circumstances | ||
In-flight oxygen administration is recommended for patients using oxygen or whose arterial blood oxygen pressure is <8 kPa (60 mmHg) at sea level | I | C |
For interventions requiring anaesthesia, multidisciplinary consultation at a PH centre to assess risk and benefit should be considered | IIa | C |
ACEi, angiotensin-converting enzyme inhibitor; ARB, angiotensin receptor blocker; ARNI, angiotensin receptor–neprilysin inhibitor; HF, heart failure; PAH, pulmonary arterial hypertension; PH, pulmonary hypertension; RV, right ventricle; SARS-CoV-2, severe acute respiratory syndrome coronavirus-2; SGLT-2i, sodium–glucose cotransporter-2 inhibitor. aClass of recommendation. bLevel of evidence. cMeasured on at least two occasions.
Recommendations | Classa | Levelb |
It is recommended that women of childbearing potential with PAH are counselled at the time of diagnosis about the risks and uncertainties associated with becoming pregnant; this should include advice against becoming pregnant, and referral for psychological support where needed | I | C |
It is recommended to provide women of childbearing potential with PAH with clear contraceptive advice, considering the individual needs of the woman but recognizing that the implications of contraceptive failure are significant in PAH | I | C |
It is recommended that women with PAH who consider pregnancy or who become pregnant receive prompt counselling in an experienced PH centre, to facilitate genetic counselling and shared decision-making, and to provide psychological support to the patients and their families where needed | I | C |
For women with PAH having termination of pregnancy, it is recommended that this be performed in PH centres, with psychological support provided to the patient and their family | I | C |
For women with PAH who desire to have children, where available, adoption and surrogacy with pre-conception genetic counselling may be considered | IIb | C |
As teratogenic potential has been reported in preclinical models for endothelin receptor antagonists and riociguat, these drugs are not recommended during pregnancy [359, 377] | III | B |
PAH, pulmonary arterial hypertension; PH, pulmonary hypertension. aClass of recommendation. bLevel of evidence.
6.3.3. Pulmonary arterial hypertension therapies
6.3.3.1. Calcium channel blockers
Patients with PAH who respond favourably to acute vasoreactivity testing (Figure 8) may respond favourably to treatment with CCBs [129, 146]. Less than 10% of patients with IPAH, HPAH, or DPAH are responders, while an acute vasodilator response does not predict a favourable long-term response to CCBs in patients with other forms of PAH [129, 146, 378]. The CCBs that have predominantly been used in PAH are nifedipine, diltiazem, and amlodipine [129, 146]. Amlodipine and felodipine are increasingly being used in clinical practice due to their long half-life and good tolerability. The daily doses that have shown efficacy in PAH are relatively high and they must be reached progressively (Table 19). The most common adverse events are systemic hypotension and peripheral oedema.
Patients who meet the criteria for a positive acute vasodilator response and treated with CCBs should be closely followed for safety and efficacy, with a complete reassessment after 3–6 months of therapy, including RHC. Additional acute vasoreactivity testing should be performed at re-evaluation to detect persistent vasodilator response, supporting possible increases in CCB dosage. Patients with a satisfactory chronic response present with WHO-FC I/II and marked haemodynamic improvement (ideally, mPAP <30 mmHg and PVR <4 WU) while on CCB therapy. In the absence of a satisfactory response, additional PAH therapy should be instituted. In some cases, a combination of CCBs with approved PAH drugs is required because of clinical deterioration with CCB withdrawal attempts. Patients who have not undergone a vasoreactivity study or those with a negative test should not be started on CCBs because of potentially severe side effects (e.g. severe hypotension, syncope, and RV failure), unless prescribed at standard doses for other indications [379].
Recommendations | Classa | Levelb |
High doses of CCBs are recommended in patients with IPAH, HPAH, or DPAH who are responders to acute vasoreactivity testing | I | C |
Close follow-up with complete reassessment after 3–4 months of therapy (including RHC) is recommended in patients with IPAH, HPAH, or DPAH treated with high doses of CCBs | I | C |
Continuing high doses of CCBs is recommended in patients with IPAH, HPAH, or DPAH in WHO-FC I or II with marked haemodynamic improvement (mPAP <30 mmHg and PVR <4 WU) | I | C |
Initiating PAH therapy is recommended in patients who remain in WHO-FC III or IV or those without marked haemodynamic improvement after high doses of CCBs | I | C |
In patients with a positive vasoreactivity test but insufficient long-term response to CCBs who require additional PAH therapy, continuation of CCB therapy should be considered | IIa | C |
CCBs are not recommended in patients without a vasoreactivity study or non-responders, unless prescribed for other indications (e.g. Raynaud's phenomenon) | III | C |
CCB, calcium channel blocker; DPAH, drug-associated pulmonary arterial hypertension; HPAH, heritable pulmonary arterial hypertension; IPAH, idiopathic pulmonary arterial hypertension; mPAP, mean pulmonary arterial pressure; PAH, pulmonary arterial hypertension; PVR, pulmonary vascular resistance; RHC, right heart catheterization; WHO-FC, World Health Organization functional class; WU, Wood units. aClass of recommendation. bLevel of evidence.
6.3.3.2. Endothelin receptor antagonists
Binding of endothelin-1 to endothelin receptors A and B on PA smooth-muscle cells promotes vasoconstriction and proliferation (Figure 7) [380]. Endothelin B receptors are mostly expressed on pulmonary endothelial cells, promoting vasodilation through accelerated production of prostacyclin and nitric oxide, and clearance of endothelin-1 [380]. Nevertheless, selective blocking of endothelin A receptors alone or non-selective blocking of both A and B receptors has shown similar effectiveness in PAH [380]. Endothelial receptor antagonists have teratogenic effects and should not be used during pregnancy [381].
6.3.3.2.1. Ambrisentan
Ambrisentan is an oral ERA that preferentially blocks the endothelin A receptors. The approved dosages in adults are 5 mg and 10 mg o.d. In patients with PAH, it has demonstrated efficacy for symptoms, exercise capacity, haemodynamics, and time to clinical worsening [382]. An increased incidence of peripheral oedema was reported with ambrisentan use, while there was no increased incidence of abnormal liver function.
6.3.3.2.2. Bosentan
Bosentan is an oral, dual ERA that improves exercise capacity, WHO-FC, haemodynamics, and time to clinical worsening in patients with PAH [383]. The approved target dose in adults is 125 mg b.i.d. Dose-dependent increases in liver transaminases can occur in ∼10% of treated patients (reversible after dose reduction or discontinuation) [384]. Thus, liver function testing should be performed monthly in patients receiving bosentan [384]. Due to pharmacokinetic interactions, bosentan may render hormonal contraceptives unreliable and lower serum levels of warfarin, sildenafil, and tadalafil [361, 385–387].
6.3.3.2.3. Macitentan
Macitentan is an oral, dual ERA that has been found to increase exercise capacity and reduce a composite endpoint of clinical worsening in patients with PAH [167]. While no liver toxicity has been shown, a reduction in Hb to ≤8 g/dL was observed in 4.3% of patients receiving 10 mg of macitentan [167].
6.3.3.3. Phosphodiesterase 5 inhibitors and guanylate cyclase stimulators
Stimulating soluble guanylate cyclase (sGC) by nitric oxide results in production of the intracellular second messenger cyclic guanosine monophosphate (cGMP) (Figure 7). This pathway is controlled by a negative feedback loop through degradation of cGMP via different phosphodiesterases, among which subtype 5 (PDE5) is abundantly expressed in the pulmonary vasculature [388]. Phosphodiesterase 5 inhibitors and sGC stimulators must not be combined with each other and with nitrates, as this can result in systemic hypotension [389].
6.3.3.3.1. Sildenafil
Sildenafil is an orally active, potent, and selective inhibitor of PDE5. Several RCTs of patients with PAH treated with sildenafil (with or without background therapy) have confirmed favourable results on exercise capacity, symptoms, and/or haemodynamics [390–392]. The approved dose of sildenafil is 20 mg t.i.d. Most side effects of sildenafil are mild to moderate and mainly related to vasodilation (headache, flushing, and epistaxis).
6.3.3.3.2. Tadalafil
Tadalafil is a once-daily administered PDE5i. An RCT of 406 patients with PAH (53% on background bosentan therapy) treated with tadalafil at doses up to 40 mg o.d. showed favourable results on exercise capacity, symptoms, haemodynamics, and time to clinical worsening [393]. The side effect profile was similar to that of sildenafil.
6.3.3.3.3. Riociguat
While PDE5is augment the nitric oxide–cGMP pathway by slowing cGMP degradation, sGC stimulators enhance cGMP production by directly stimulating the enzyme, both in the presence and absence of endogenous nitric oxide [394]. An RCT of 443 patients with PAH (44% and 6% on background therapy with ERAs or prostacyclin analogues, respectively) treated with riociguat up to 2.5 mg t.i.d. showed favourable results on exercise capacity, haemodynamics, WHO-FC, and time to clinical worsening [395]. The side effect profile was similar to that of PDE5is.
6.3.3.4. Prostacyclin analogues and prostacyclin receptor agonists
The prostacyclin metabolic pathway (Figure 7) is dysregulated in patients with PAH, with less prostacyclin synthase expressed in PAs and reduced prostacyclin urinary metabolites [396]. Prostacyclin analogues and prostacyclin receptor agonists induce potent vasodilation, inhibit platelet aggregation, and also have both cytoprotective and anti-proliferative activities [397]. The most common adverse events observed with these compounds are related to systemic vasodilation and include headache, flushing, jaw pain, and diarrhoea.
6.3.3.4.1. Epoprostenol
Epoprostenol has a short half-life (3–5 min) and needs continuous i.v. administration via an infusion pump and a permanent tunnelled catheter. A thermo-stable formulation is available to maintain stability up to 48 h [398]. Its efficacy has been demonstrated in three unblinded RCTs in patients with IPAH (WHO-FC III and IV) [399, 400] and SSc-associated PAH [401]. Epoprostenol improved symptoms, exercise capacity, haemodynamics, and mortality [399]. Long-term, persistent efficacy has also been shown in IPAH [212, 245], as well as in other associated PAH conditions [402–404]. Serious adverse events related to the delivery system include pump malfunction, local site infection, catheter obstruction, and sepsis. Recommendations for preventing central venous catheter bloodstream infections have been proposed [405, 406].
6.3.3.4.2. Iloprost
Iloprost is a prostacyclin analogue approved for inhaled administration. Inhaled iloprost has been evaluated in one RCT, in which six to nine repetitive iloprost inhalations were compared with placebo in treatment-naïve patients with PAH or CTEPH [407]. The study showed an increase in exercise capacity and improvement in symptoms, PVR, and clinical events in the iloprost group compared with the placebo group.
6.3.3.4.3. Treprostinil
Treprostinil is available for s.c., i.v., inhaled, and oral administration. Treprostinil s.c. improved exercise capacity, haemodynamics, and symptoms in PAH [408]. Infusion-site pain was the most common adverse effect, which led to treatment discontinuation in 8% of cases [408]. Based on its chemical stability, i.v. treprostinil may also be administered via implantable pumps, improving convenience and likely decreasing the occurrence of line infections [409, 410].
Inhaled treprostinil improved the 6MWD, NT-proBNP, and quality of life measures in patients with PAH on background therapy with either bosentan or sildenafil [411]. Inhaled treprostinil is not approved in Europe.
Oral treprostinil has been evaluated in two RCTs of patients with PAH on background therapy with bosentan and/or sildenafil. In both trials, the primary endpoint—6MWD—did not reach statistical significance [412, 413]. An additional RCT in treatment-naïve patients with PAH showed improved 6MWD [414]. An event-driven RCT that enrolled 690 patients with PAH demonstrated that oral treprostinil reduced the risk of clinical worsening events in patients who were receiving oral monotherapy with ERAs or PDE5is [415]. Oral treprostinil is not approved in Europe.
6.3.3.4.4. Beraprost
Beraprost is a chemically stable and orally active prostacyclin analogue. Two RCTs have shown a modest, short-term improvement in exercise capacity in patients with PAH [416, 417]; however, there were no haemodynamic improvements or long-term outcome benefits. Beraprost is not approved in Europe.
6.3.3.4.5. Selexipag
Selexipag is an orally available, selective, prostacyclin receptor agonist that is chemically distinct from prostacyclin, with different pharmacology. In a pilot RCT in patients with PAH (receiving stable ERA and/or PDE5i therapy), selexipag reduced PVR after 17 weeks [418]. An event-driven, phase 3 RCT that enrolled 1156 patients [419] showed that selexipag alone or on top of mono or double therapy with an ERA and/or a PDE5i reduced the relative risk of composite morbidity/mortality events by 40%. The most common side effects were headache, diarrhoea, nausea, and jaw pain.
6.3.4. Treatment strategies for patients with idiopathic, heritable, drug-associated, or connective tissue disease-associated pulmonary arterial hypertension
Pulmonary arterial hypertension is a rare and life-threatening disease and should be managed, where possible, at PH centres in close collaboration with the patient's local physicians.
This section describes drug treatment and is focused on non-vasoreactive patients with IPAH/HPAH/DPAH and on patients with PAH associated with connective tissue disease (PAH-CTD). Information on the dosing of PAH medication is summarized in Table 19. For other forms of PAH, treatment strategies have to be modified (see Section 7). The approach to vasoreactive patients with IPAH/HPAH/DPAH is described in Section 6.3.3.1.
In addition to targeted drug treatment, the comprehensive management of patients with PAH includes general measures that may include supplementary oxygen, diuretics to optimize volume status, psychosocial support, and standardized exercise training (Section 6.3.1) [315]. Prior to the treatment decisions, patients and their next of kin should be provided with appropriate and timely information about the risks and benefits of the treatment options so they can make the final, informed, and joint decision about the treatment with the medical team. Treatment decisions in patients with IPAH/HPAH/DPAH or PAH-CTD should be stratified according to the presence or absence of cardiopulmonary comorbidities (Section 6.3.4.3) and according to disease severity assessed by risk stratification (Section 6.2.7).
6.3.4.1. Initial treatment decision in patients without cardiopulmonary comorbidities
The initial treatment of patients with PAH should be based on a comprehensive, multiparameter risk assessment, considering disease type and severity, comorbidities, access to therapies, economic aspects, and patient preference.
The following considerations predominantly apply to patients with IPAH/HPAH/DPAH or PAH-CTD without cardiopulmonary comorbidities, as patients with comorbidities were under-represented in the clinical studies addressing treatment strategies and combination therapy in patients with PAH. Treatment considerations for patients with PAH and cardiopulmonary comorbidities are summarized in Section 6.3.4.3.
For patients presenting at low or intermediate risk, initial combination therapy with an ERA and a PDE5i is recommended. This approach was assessed in the AMBITION study, which compared initial combination therapy using ambrisentan at a target dose of 10 mg o.d. and tadalafil at a target dose of 40 mg o.d. with initial monotherapy with either drug [166]. AMBITION predominantly included patients with IPAH/HPAH/DPAH or PAH-CTD. The primary endpoint was the time to first clinical failure event (a composite of death, hospitalization for worsening PAH, disease progression, or unsatisfactory long-term clinical response). The hazard ratio (HR) for the primary endpoint in the combination-therapy group versus the pooled monotherapy group was 0.50 (95% confidence interval [CI], 0.35–0.72; P<0.001) and there were significant improvements in 6MWD and NT-proBNP with initial combination therapy. At the end of the study, 10% of the patients assigned to initial combination therapy had died compared with 14% of the patients assigned to initial monotherapy (HR 0.67; 95% CI, 0.42–1.08) [420].
In the TRITON study, treatment-naïve patients with PAH were assigned to initial dual-combination therapy with macitentan and tadalafil, or initial triple-combination therapy with macitentan 10 mg o.d., tadalafil at a target dose of 40 mg o.d., and selexipag up to 1600 µg o.d [421]. TRITON predominantly included patients with IPAH/HPAH/DPAH or PAH-CTD. At week 26, PVR was reduced by 52% and 54%, with double- or triple-combination therapy, respectively, and 6MWD had increased by 55 m and 56 m, respectively. The geometric means of the NT-proBNP ratio from baseline to week 26 were 0.25 and 0.26, respectively. Hence, TRITON did not show a benefit of oral triple- versus oral double-combination therapy but confirmed that substantial improvements in haemodynamics and exercise capacity can be obtained with initial ERA/PDE5i combination therapy. Further studies are needed to determine whether oral triple-combination therapy impacts long-term outcomes.
Based on the evidence generated by these and other studies [303, 422–424], initial dual-combination therapy with an ERA and a PDE5i is recommended for newly diagnosed patients who present at low or intermediate risk. Initial oral triple-combination therapy is not recommended, given the current lack of evidence supporting this strategy. In patients presenting at high risk, initial triple-combination therapy including an i.v./s.c. prostacyclin analogue should be considered [426, 427]. While it is acknowledged that the evidence for this approach is limited to case series, there is consensus that this strategy has the highest likelihood of success, especially in view of registry data from France showing that initial triple-combination therapy including an i.v./s.c. prostacyclin analogue was associated with better long-term survival than monotherapy or dual-combination therapy [428]. Initial triple-combination therapy including an i.v./s.c. prostacyclin analogue should also be considered in patients at intermediate risk presenting with severe haemodynamic impairment (e.g. RAP ≥20 mmHg, CI <2.0 L/min/m2, SVI <31 mL/m2, and/or PVR ≥12 WU) [238, 426].
The recommendations for initial oral double-combination therapy are based on PICO question I (Supplementary Data, Section 6.2). Although the quality of evidence is low, initial oral combination therapy with an ERA and a PDE5i achieves important targets in symptom improvement (functional class), exercise capacity, cardiac biomarkers, and reduction of hospitalizations.
Recommendations | Classb | Levelc |
Recommendations for initial therapy | ||
In patients with IPAH/HPAH/DPAH who present at high risk of death, initial combination therapy with a PDE5i, an ERA, and i.v./s.c. prostacyclin analogues should be consideredd | IIa | C |
Recommendations for treatment decisions during follow-up | ||
In patients with IPAH/HPAH/DPAH who present at intermediate–low risk of death while receiving ERA/PDE5i therapy, addition of selexipag should be considered [419] | IIa | B |
In patients with IPAH/HPAH/DPAH who present at intermediate–high or high risk of death while receiving ERA/PDE5i therapy, addition of i.v./s.c. prostacyclin analogues and referral for LTx evaluation should be considered | IIa | C |
In patients with IPAH/HPAH/DPAH who present at intermediate–low risk of death while receiving ERA/PDE5i therapy, switching from PDE5i to riociguat may be considered [429] | IIb | B |
See Recommendation Table 8B for footnotes.
Recommendations | GRADE | Classb | Levelc | |
Quality of evidence | Strength of recommendation | |||
Recommendations for initial therapy | ||||
In patients with IPAH/HPAH/DPAH who present at low or intermediate risk of death, initial combination therapy with a PDE5i and an ERA is recommended [166] | Low | Conditional | I | B |
CI, cardiac index; DLCO, Lung diffusion capacity for carbon monoxide; DPAH, drug-associated pulmonary arterial hypertension; ERA, endothelin receptor antagonist; HFpEF, heart failure with preserved ejection fraction; HPAH, heritable pulmonary arterial hypertension; IPAH, idiopathic pulmonary arterial hypertension; i.v., intravenous; LTx, lung transplantation; PAH, pulmonary arterial hypertension; PDE5i, phosphodiesterase 5 inhibitor; PVR, pulmonary vascular resistance; RAP, right atrial pressure; s.c., subcutaneous; SVI, stroke volume index; WU, Wood units. aCardiopulmonary comorbidities are predominantly encountered in elderly patients and include risk factors for HFpEF such as obesity, diabetes, coronary heart disease, a history of hypertension, and/or a low DLCO. bClass of recommendation. cLevel of evidence. dInitial triple-combination therapy including i.v./s.c. prostacyclin analogues may also be considered in patients presenting at intermediate risk but severe haemodynamic impairment (e.g. RAP ≥20 mmHg, CI <2.0 L/min/m2, SVI <31 mL/m2, and/or PVR ≥12 WU).
Recommendations | Classa | Levelb |
Initial combination therapy with ambrisentan and tadalafil is recommended [166, 420, 423] | I | B |
Initial combination therapy with macitentan and tadalafil is recommended [421, 430] | I | B |
Initial combination therapy with other ERAs and PDE5is should be considered [430] | IIa | B |
Initial combination therapy with macitentan, tadalafil, and selexipag is not recommended [421] | III | B |
ERA, endothelin receptor antagonist; PDE5i, phosphodiesterase 5 inhibitor. aClass of recommendation. bLevel of evidence.
6.3.4.2. Treatment decisions during follow-up in patients without cardiopulmonary comorbidities
Patients with PAH require regular follow-up, including risk stratification and an assessment of patient concordance with therapy. Patients who achieve a low-risk status have a much superior long-term survival compared with patients with intermediate- or high-risk status [292, 295, 296]. Achieving and maintaining a low-risk profile is therefore a key objective in managing patients with PAH.
Several clinical trials have assessed the safety and efficacy of sequential combination therapy in patients with PAH. SERAPHIN enrolled 742 patients with PAH, mostly with IPAH/HPAH/DPAH and PAH-CTD, of whom 63.7% were receiving other PAH medication at the time of enrolment, mostly sildenafil [167]. In the subgroup of patients with background PAH therapy, macitentan at a daily dose of 10 mg reduced the risk of clinical worsening events compared with placebo (HR 0.62; 95% CI, 0.43–0.89) [167].
GRIPHON assessed the safety and efficacy of selexipag [419]. This study enrolled 1156 patients with PAH, also mostly with IPAH/HPAH/DPAH or PAH-CTD, who were treatment naïve or receiving background therapy with an ERA, PDE5i, or a combination of both. Selexipag at a dose of up to 1600 µg b.i.d. was associated with a reduced risk of clinical worsening events independent of the background medication. In patients receiving ERA/PDE5i combination therapy (n=376), the risk of clinical worsening events was lower with selexipag than with placebo (HR 0.63; 95% CI, 0.44–0.90) [431].
The effects of combination therapy on long-term survival in patients with PAH remain unclear. A 2016 meta-analysis demonstrated that combination therapy (initial and sequential) was associated with a significant risk reduction for clinical worsening (relative risk [RR] 0.65; 95% CI, 0.58–0.72; P<0.0001) [432]; however, all-cause mortality was not improved (RR 0.86; 95% CI, 0.72–1.03; P=0.09) and a substantial proportion of patients had clinical worsening events or died despite receiving combination therapy. In addition, registry data showed that the use of combination therapy increased since 2015 but there was no clear improvement in overall survival rates [428, 433, 434]. These data were corroborated by a study showing that less than half of patients receiving initial combination therapy with an ERA and a PDE5i achieved and maintained a low-risk profile [422].
Switching from PDE5is to riociguat has also been investigated as a treatment-escalation strategy [429, 435]. REPLACE was a randomized, controlled, open-label study that enrolled patients on a PDE5i-based therapy who were in WHO-FC III and had a 6MWD of 165–440 m [429]. The study predominantly included patients with IPAH/HPAH/DPAH or PAH-CTD who were randomized to continue their PDE5i or to switch from a PDE5i to riociguat up to 2.5 mg t.i.d. The study met its primary endpoint, termed ‘clinical improvement’, which was a composite of pre-specified improvements in 6MWD, WHO-FC, and NT-proBNP at week 24. Clinical improvement at week 24 was demonstrated in 41% of the patients who switched to riociguat and in 20% of the patients who maintained their PDE5i (odds ratio [OR] 2.78; 95% CI, 1.53–5.06; P=0.0007). In addition, fewer patients in the riociguat group experienced a clinical worsening event (OR 0.10; 95% CI, 0.01–0.73; P=0.0047).
Based on the evidence summarized above, the following recommendations for treatment decisions during follow-up are:
i) In patients who achieve a low-risk status with their initial PAH therapy, continuation of treatment is recommended.
ii) In patients who are at intermediate–low risk despite receiving ERA/PDE5i therapy, adding selexipag should be considered to reduce the risk of clinical worsening. In these patients, switching from PDE5i to riociguat may also be considered.
iii) In patients who are at intermediate–high or high risk while receiving oral therapies, the addition of i.v. epoprostenol or i.v./s.c. treprostinil and referral for LTx evaluation should be considered [309, 436]. If adding i.v./s.c. prostacyclin analogues is unfeasible, adding selexipag or switching from PDE5i to riociguat may be considered.
Recommendations | Classa | Levelb |
General recommendation for sequential combination therapy | ||
It is recommended to base treatment escalations on risk assessment and general treatment strategies (see Figure 9) | I | C |
Evidence from studies with a composite morbidity/mortality endpoint as primary outcome measure | ||
Addition of macitentan to PDE5is or oral/inhaled prostacyclin analogues is recommended to reduce the risk of morbidity/mortality events [167, 168, 437] | I | B |
Addition of selexipag to ERAsc and/or PDE5is is recommended to reduce the risk of morbidity/mortality events [418, 419] | I | B |
Addition of oral treprostinil to ERA or PDE5i/riociguat monotherapy is recommended to reduce the risk of morbidity/mortality events [412, 413, 415] | I | B |
Addition of bosentan to sildenafil is not recommended to reduce the risk of morbidity/mortality events [419] | III | B |
Evidence from studies with change in 6MWD as primary outcome measure | ||
Addition of sildenafil to epoprostenol is recommended to improve exercise capacity [392, 438] | I | B |
Addition of inhaled treprostinil to sildenafil or bosentan monotherapy should be considered to improve exercise capacity [411, 439] | IIa | B |
Addition of riociguat to bosentan should be considered to improve exercise capacity [395, 440] | IIa | B |
Addition of tadalafil to bosentan may be considered to improve exercise capacity [393] | IIb | C |
Addition of inhaled iloprost to bosentan may be considered to improve exercise capacity [441, 442] | IIb | B |
Addition of ambrisentan to sildenafil may be considered to improve exercise capacity [443] | IIb | C |
Addition of bosentan to sildenafil may be considered to improve exercise capacity [419, 444] | IIb | C |
Addition of sildenafil to bosentan may be considered to improve exercise capacity [444–446] | IIb | C |
Other sequential double- or triple-combination therapies may be considered to improve exercise capacity and/or alleviate PH symptoms | IIb | C |
Evidence from studies with safety of combination therapy as primary outcome measure | ||
Combining riociguat and PDE5is is not recommendedd [389] | III | B |
6MWD, 6-minute walking distance; ERA, endothelin receptor antagonist; PDE5i, phosphodiesterase 5 inhibitor; PH, pulmonary hypertension. aClass of recommendation. bLevel of evidence. cERAs used in the GRIPHON study were bosentan and ambrisentan. dThe PATENT plus study investigated the combination of sildenafil and riociguat; however, combining riociguat with any PDE5i is contraindicated.
6.3.4.3. Pulmonary arterial hypertension with cardiopulmonary comorbidities
Over the past decade, the demographics and characteristics of patients with IPAH have changed, especially in industrialized countries [447]. In several contemporary registries, the average age of patients diagnosed with IPAH is ∼60 years or older [161, 295, 299, 447, 448]. Many elderly patients have cardiopulmonary comorbidities, making the distinction from group 2 and group 3 PH challenging. Among elderly patients diagnosed with IPAH, two main disease phenotypes have emerged. One phenotype (herein called the left heart phenotype) consists of elderly, mostly female patients with risk factors for HFpEF (e.g. hypertension, obesity, diabetes, or coronary heart disease) but pre-capillary PH rather than post-capillary PH [449, 450]; ∼30% of these patients have a history of atrial fibrillation [161]. The other phenotype (called the cardiopulmonary phenotype) consists of elderly, predominantly male patients who have a low DLCO (<45% of the predicted value), are often hypoxaemic, have a significant smoking history, and have risk factors for LHD [77, 78, 161, 451]. In a cluster analysis of 841 newly diagnosed patients with IPAH from the COMPERA registry, 12.6% had a classic phenotype of young, mostly female patients without cardiopulmonary comorbidities, while 35.8% presented with a left heart phenotype and 51.6% with a cardiopulmonary phenotype [161].
There are no evidence-based rules for determining a patient's phenotype. The AMBITION study used the presence of more than three risk factors for LHD together with certain haemodynamic criteria to exclude patients from the primary analysis [166]. However, the COMPERA cluster analysis mentioned above found that the presence of a single risk factor may change the phenotype [161]. Pending further data, it is the overall profile that should be used to determine a patient's phenotype.
Compared with patients without cardiopulmonary comorbidities, patients with cardiopulmonary comorbidities respond less well to PAH medication, are more likely to discontinue this medication due to efficacy failure or lack of tolerability, are less likely to reach a low-risk status, and have a higher mortality risk. While the age-adjusted mortality of patients with the left heart phenotype seems to be similar to that of patients with classical PAH, patients with a cardiopulmonary phenotype and a low DLCO have a particularly high mortality risk [77, 78, 161, 450, 451].
As patients with cardiopulmonary comorbidities were under-represented in or excluded from PAH trials, no evidence-based treatment recommendations can be made for this patient population. Registry data suggest that most physicians use PDE5is as primary treatment for these patients. Endothelin receptor antagonists or PDE5i/ERA combinations are occasionally used, but the drug discontinuation rate is higher than in patients with classical PAH [447, 450]. A subgroup analysis from AMBITION, which assessed the response to PAH therapy in 105 patients who were excluded from the primary analysis set because of a left heart phenotype, found that these patients—compared with patients in the primary analysis set—had less clinical improvement and a higher likelihood of drug discontinuations due to safety and tolerability with both monotherapy and initial combination therapy [449]. Data from the ASPIRE registry demonstrated that patients with IPAH and a cardiopulmonary phenotype had less improvement in exercise capacity and PROMs compared with patients with classical IPAH [451].
In patients with a left heart phenotype, ERA therapy is associated with an elevated risk of fluid retention [449]. Moreover, in patients with a cardiopulmonary phenotype, PAH medication may cause a decline in the peripheral oxygen saturation [452]. There is little published experience on the use of prostacyclin analogues or prostacyclin receptor agonists in this patient population [453].
The lack of solid evidence for treating elderly patients with PAH and cardiopulmonary comorbidities makes treatment recommendations challenging, and patients should be counselled accordingly. In the absence of evidence on treatment strategies in these patients, risk stratification is of limited usefulness in guiding therapeutic decision-making. Initial monotherapy (see Supplementary Data, Table S3) is recommended for most of these patients, with PDE5is being the most widely used compounds according to registry data [161]. Further treatment decisions should be made on an individual basis in collaboration with the PH centre and local physicians.
The treatment algorithm for patients with PAH is shown in Figure 9 and the accompanying section describing the treatment algorithm.
Recommendations | Classb | Levelc |
Recommendations for initial therapy | ||
In patients with IPAH/HPAH/DPAH and cardiopulmonary comorbidities, initial monotherapy with a PDE5i or an ERA should be considered | IIa | C |
Recommendations for treatment decisions during follow-up | ||
In patients with IPAH/HPAH/DPAH with cardiopulmonary comorbidities who present at intermediate or high risk of death while receiving PDE5i or ERA monotherapy, additional PAH medications may be considered on an individual basis | IIb | C |
DPAH, drug-associated pulmonary arterial hypertension; ERA, endothelin receptor antagonist; HPAH, heritable pulmonary arterial hypertension; IPAH, idiopathic pulmonary arterial hypertension; PAH, pulmonary arterial hypertension; PDE5i, phosphodiesterase 5 inhibitor. aCardiopulmonary comorbidities are predominantly encountered in elderly patients and include risk factors for HFpEF such as obesity, diabetes, coronary heart disease, a history of hypertension, and/or a low DLCO. bClass of recommendation. cLevel of evidence.
6.3.5. Drug interactions
Among PAH drugs, clinically relevant pharmacokinetic interactions are observed between bosentan and sildenafil (reduced sildenafil plasma concentration [385]), bosentan and hormonal contraceptives (reduced contraception efficacy [361]), and bosentan and vitamin K antagonists (VKAs) (potential need for VKA dose adjustment [386]). Additional pharmacokinetic interactions of potential clinical relevance are listed in Supplementary Data, Table S4.
6.3.6. Interventional therapy
6.3.6.1. Balloon atrial septostomy and Potts shunt
Balloon atrial septostomy [454, 455], by creating an interatrial shunt, and Potts shunt [456–459], by connecting the left PA and descending aorta, aim to decompress the right heart and increase systemic blood flow, thereby improving systemic oxygen transport despite arterial oxygen desaturation. As these procedures are complex and associated with high risk, including substantial procedure-related mortality, they are rarely performed in patients with PAH and may only be considered in centres with experience in the techniques.
6.3.6.2. Pulmonary artery denervation
The rationale for performing a PA denervation (PADN) is based on the increased sympathetic overdrive characterizing PAH, which is associated with poor outcome [460, 461]. Although the contribution of this mechanism to developing PAH is not completely understood, it is associated with vasoconstriction and vascular remodelling through a baroreflex mediated by stretch receptors located at the bifurcation of the Pas [462, 463]. Applying radiofrequency at the latter acutely and chronically improves haemodynamic variables [464]. However, there is little evidence yet from multicentre RCTs demonstrating a benefit of PADN in patients already receiving recommended medical therapy. A small multicentre study tested the feasibility of PADN using an intravascular ultrasound catheter in patients receiving dual or triple therapy for PAH [465]; the procedure was safe and associated with a reduction in PVR, and increases in 6MWD and daily activity. Although potentially promising, PADN should be considered experimental.
6.3.7. Advanced right ventricular failure
6.3.7.1. Intensive care unit management
Patients with PH may require intensive care treatment for right HF, comorbidities (including major surgery), or both. The mortality risk is high in such patients [466, 467], and specialized centres should be involved whenever possible. In addition to basic intensive care unit (ICU) standards, RV function in these patients should be carefully monitored. Non-specific clinical signs of right HF with low CO include pale skin with peripheral cyanosis, hypotension, tachycardia, declining urine output, and increasing lactate levels. Non-invasive monitoring should include biomarkers (NT-proBNP and troponin) and echocardiography. Minimum invasive monitoring consists of an upper body central venous catheter to measure central venous pressure and central venous oxygen saturation, the latter reflecting CO. Right heart catheterization or other forms of advanced haemodynamic assessment should be considered in patients with advanced right HF or in complex situations [468].
Treating right HF should focus on treatable triggers such as infection, arrhythmia, anaemia, and other comorbidities. Fluid management is of utmost importance in these patients, most of whom require a negative fluid balance to reduce RV pre-load, thereby improving RV geometry and function [468]. Patients with a low CO may benefit from treatment with inotropes; dobutamine and milrinone are the most frequently used substances in this setting. Maintaining the mean systemic blood pressure >60 mmHg is a key objective when treating right HF, and patients with persistent hypotension may require vasopressors such as norepinephrine or vasopressin. Intubation and invasive mechanical ventilation should be avoided whenever possible in patients with advanced RV failure because of a high risk of further haemodynamic deterioration and death. Pulmonary arterial hypertension medication should be considered on an individual basis, taking into account underlying disease, comorbidities, and existing medication. In patients with newly diagnosed PAH presenting with low CO, combination therapy including i.v./s.c. prostacyclin analogues should be considered [426].
6.3.7.2. Mechanical circulatory support
In specialist centres, various forms of mechanical circulatory support are available for managing RV failure, with veno-arterial extracorporeal membrane oxygenation (ECMO) being the most widely used approach. Mechanical circulatory support has become an established bridging tool to transplantation in patients with irreversible right HF, but is occasionally used as a bridge to recovery in patients with treatable causes and potentially reversible RV failure [468]. No general recommendations can be made regarding the indication for mechanical circulatory support, which needs to be individualized, considering patient factors and local resources [469, 470]. Long-term mechanical support analogous to left ventricular assist devices (LVADs) is not yet available for patients with PH and end-stage right HF.
Recommendations | Classa | Levelb |
When managing patients with right HF in the ICU, it is recommended to involve physicians with expertise, treat causative factors, and use supportive measures, including inotropes and vasopressors, fluid management, and PAH drugs, as appropriate | I | C |
Mechanical circulatory support may be an option for selected patients as bridge to transplantation or to recovery, and interhospital transfer should be considered if such resources are not available on site | IIa | C |
HF, heart failure; ICU, intensive care unit; PAH, pulmonary arterial hypertension. aClass of recommendation. bLevel of evidence.
6.3.8. Lung and heart–lung transplantation
Lung transplantation remains an important treatment option for patients with PAH refractory to optimized medical therapy. In patients with PAH, referral to an LTx centre should be considered early (Table 20): 1) when they present with an inadequate response to treatment despite optimized combination therapy; 2) when they present with an intermediate–high or high risk of death (i.e. 1-year mortality >10% when estimated with established risk-stratification tools) [471] (see Section 6.2.7), which exceeds the current mortality rate after LTx [472]; 3) when patients have a disease variant that poorly responds to medical therapy, such as PVOD or PCH.
Both heart–lung and bilateral LTx have been performed for PAH. Currently, most patients receive bilateral LTx, while combined heart–lung transplantation is reserved for patients who have additional non-correctable cardiac conditions [473]. With the introduction of the lung allocation score (LAS), waiting list mortality has decreased and the odds of receiving a donor organ have increased [474]. In some countries, an ‘exceptional LAS’ can be obtained for patients with severe PH. Some other countries not using the LAS have successfully implemented high-priority programmes for these patients [475]. The patient and their next of kin should be fully engaged in the transplant assessment process and informed of the risks and benefits, and the final decision should be jointly made between the patient and medical team (see Section 6.3.1.8). For patients with PAH who survive the early post-transplant period, long-term outcomes are good. A study found that for primary transplant patients with IPAH who survived to 1 year, conditional median survival was 10.0 years [476].
Recommendations | Classa | Levelb |
It is recommended that potentially eligible candidates are referred for LTx evaluation when they have an inadequate response to oral combination therapy, indicated by an intermediate–high or high risk or by a REVEAL risk score >7 | I | C |
It is recommended to list patients for LTx who present with a high risk of death or with a REVEAL risk score ≥10 despite receiving optimized medical therapy including s.c. or i.v. prostacyclin analogues | I | C |
i.v., intravenous; LTx, lung transplantation; s.c., subcutaneous. aClass of recommendation. bLevel of evidence.
6.3.9. Evidence-based treatment algorithm
A treatment algorithm for patients with IPAH/HPAH/DPAH or PAH-CTD is shown in Figure 9. The evidence supporting this algorithm has mainly been generated in patients with IPAH/HPAH/DPAH or PAH-CTD who present without cardiopulmonary comorbidities. Patients with HIV-associated PAH, PoPH, and PAH associated with congenital heart disease were not enrolled or under-represented in most PAH therapy trials. Treatment recommendations for these patients are provided in Section 7.
6.3.10. Diagnosis and treatment of pulmonary arterial hypertension complications
6.3.10.1. Arrhythmias
The most common types of arrhythmias observed in PAH are supraventricular, mainly atrial fibrillation and atrial flutter, while the frequency of ventricular arrhythmias and bradyarrhythmias appears to be considerably lower [477–479]. Of note, age is an independent risk factor for atrial arrhythmias. In prospective studies, the incidence of atrial arrhythmias was 3–25% over an observation time of 5 years in cohorts primarily containing patients with IPAH [479–481].
In the absence of specific evidence for PAH, managing anticoagulation in patients with PAH and atrial arrhythmia should follow the recommendations for patients with other cardiac conditions [477].
Patients with PAH are especially sensitive to haemodynamic stress during atrial arrhythmias due to tachycardia and loss of atrioventricular synchrony. Maintaining sinus rhythm is an important treatment objective in these patients. New-onset arrhythmias frequently lead to clinical deterioration and are associated with increased mortality [481]. Observational studies have shown that a variety of rhythm control strategies are feasible, including pharmacological cardioversion with anti-arrhythmic drugs, electrical cardioversion, and invasive catheter ablation procedures. To achieve or maintain a stable sinus rhythm, prophylaxis with anti-arrhythmic drugs without negative inotropic effects, such as oral amiodarone, should be considered, even if specific data regarding their efficacy are lacking. Low-dose beta-blockers and/or digoxin may be used on an individual patient basis.
Catheter ablation is the preferred approach in managing atrial flutter and some other atrial tachycardias, although catheter ablation in patients with PAH is often more technically challenging than in patients with a structurally normal right heart chamber [482]. The safety and efficacy of ablation techniques for atrial fibrillation specifically in the PAH population are uncertain, and it is possible that, due to remodelling of the RA, non-pulmonary vein triggers may play a more important role than in patients without PAH [483].
6.3.10.2. Haemoptysis
Haemoptysis, ranging from mild to life-threatening, may occur in all forms of PH but is particularly common in HPAH and PAH associated with CHD. Pulmonary bleeding frequently originates from enlarged bronchial arteries [484–486]; hence, the diagnostic evaluation of patients with PAH and haemoptysis should include a contrast-enhanced CT scan with an arterial phase. Even if the source of bleeding cannot be determined, embolization of enlarged bronchial arteries is recommended in patients who present with moderate-to-severe haemoptysis or recurrent episodes of mild haemoptysis. Lung transplant should be considered in patients with recurrent and severe haemoptysis despite optimized treatment.
6.3.10.3. Mechanical complications
Mechanical complications in patients with PAH usually arise from progressive dilatation of the PA and include PA aneurysms, rupture, and dissection, and compression of adjacent structures such as the left main coronary artery, pulmonary veins, main bronchi, and recurrent laryngeal nerves [487–492].
Pulmonary artery aneurysm was independently related to an increased risk of sudden cardiac death in one study [492]. Symptoms and signs are non-specific; in most cases, patients are asymptomatic and these complications are incidentally diagnosed. Pulmonary artery aneurysms are usually detected during echocardiography and best visualized by contrast-enhanced CT or MRI. Treatment options for asymptomatic PA aneurysm or PA dissection are not well defined. LTx has to be considered on an individual basis [490, 493].
For patients with left main coronary artery compression syndrome, percutaneous coronary stenting is an effective and safe treatment [62]. For patients with asymptomatic left main coronary artery compression or non-severe compromise of its anatomy, evaluation with intravascular ultrasound or coronary pressure wire may help to avoid unnecessary interventions [494].
6.3.11. End-of-life care and ethical issues
The clinical course of PAH may be characterized by progressive deterioration and occasional episodes of acute decompensation. Life expectancy is difficult to predict, as patients may either die slowly because of progressive right HF or experience sudden death.
Patient-orientated care is essential in managing PAH. Information about disease severity and possible prognosis should be provided at initial diagnosis but empathic and hopeful communication, as well as yielding hope, is essential, in line with Section 6.3.1.8. At the right time, open and sensitive communication will enable advanced planning and discussion of a patient's fears, concerns, and wishes, and will ultimately contribute to making the final, well-informed, and joint decision about treatment with the medical team.
Patients approaching end of life require frequent assessment of their full needs by a multidisciplinary team. In advanced stages, recognizing that cardiopulmonary resuscitation in severe PAH has a poor outcome may enable a do not resuscitate order; this may facilitate patients being in their preferred place of care at end of life. Attention should be given to controlling distressing symptoms and prescribing appropriate drugs while withdrawing medication that is no longer needed, which may include PAH medication. Well-informed psychological, social, and spiritual support is also vital. Specialist palliative care should be consulted for patients whose needs are beyond the expertise of the PH team [346].
6.3.12. New drugs in advanced clinical development (phase 3 studies)
Pulmonary arterial hypertension remains an incurable condition with a high mortality rate, despite use of PAH drugs mainly targeting imbalance of vasoactive factors. Novel agents, which are currently in phase 3 development, are ralinepag and sotatercept. Ralinepag is an orally available prostacyclin receptor agonist, which, in a phase 2 RCT that included 61 patients with PAH, improved PVR compared with placebo after 22 weeks of therapy [495]. Sotatercept—a fusion protein comprising the extracellular domain of the human activin receptor type IIA linked to the Fc domain of human immunoglobulin G1—acts as a ligand trap for members of the transforming growth factor (TGF)-β superfamily, thus restoring balance between growth-promoting and growth-inhibiting pathways [496]. In a phase 2 RCT that included 106 patients with PAH treated over 24 weeks, s.c. sotatercept reduced PVR in patients receiving background PAH therapy [496]; improvements were also observed in 6MWD and NT-proBNP [496].
7. Specific pulmonary arterial hypertension subsets
7.1. Pulmonary arterial hypertension associated with drugs and toxins
Several drugs and toxins are associated with developing PAH or PVOD/PCH. Historically, certain appetite suppressants and toxic rapeseed oil were the most prominent examples, whereas methamphetamines, interferons, and some tyrosine kinase inhibitors are more common causes nowadays (Table 7). Pulmonary arterial hypertension is a rare complication in patients exposed to these drugs, and many of these drugs have also been linked to other pulmonary complications such as parenchymal lung disease or pleural effusions. These pulmonary complications may occur concurrently.
Methamphetamine-associated PAH has mainly been reported from the USA, where some centres have found that 20–29% of their otherwise idiopathic cases of PAH were associated with methamphetamine use [497, 498]. Compared with patients with IPAH, those with methamphetamine-associated PAH had more severe haemodynamic impairment and a higher mortality risk [498]. Alpha and beta interferons have also been associated with developing PAH [499]. The same is true of some tyrosine kinase inhibitors, especially dasatinib, but also bosutinib and ponatinib [40, 500].
Drug- or toxin-induced PAH should always be considered in patients presenting with unexplained exertional dyspnoea or other warning signs. The diagnostic approach should be the same as in other forms of PH, and the diagnosis is usually made by excluding other forms of PH in patients who have been exposed to drugs associated with developing PAH.
Treatment of DPAH follows the same basic principles as treating other forms of PAH. Importantly, partial or full reversal of PAH has been reported after discontinuing the causative agent, at least for interferons and dasatinib [499, 500]. Hence, multidisciplinary management of the patient should include discontinuing the presumed causative agents once PAH is diagnosed (also see the 2022 ESC Guidelines on Cardio-Oncology) [501]. In patients with mild PH and a low-risk profile, discontinuing the trigger alone may be sufficient, and it is recommended that these patients be observed over 3–4 months before considering PAH therapy. Pulmonary arterial hypertension therapy should be initiated in patients who do not normalize their haemodynamics after withdrawing or in patients presenting with more advanced PAH at diagnosis. Unlike in other forms of PAH, de-escalation of PAH therapy is often possible during the course of the disease [500]. Physicians should bear in mind that DPAH may have features of PVOD/PCH, especially in patients treated with alkylating agents such as mitomycin C or cyclophosphamide. Health professional awareness is essential in identifying cases of DPAH and reporting adverse effects of pharmaceutical products.
Recommendations | Classa | Levelb |
It is recommended to make a diagnosis of drug- or toxin-associated PAH in patients who had relevant exposure and in whom other causes of PH have been excluded | I | C |
In patients with suspected drug- or toxin-associated PAH, it is recommended to discontinue the causative agent whenever possible | I | C |
Immediate PAH therapy should be considered in patients who present with intermediate/high-risk PAH at diagnosis | IIa | C |
Patients with low-risk PAH should be re-evaluated 3–4 months after discontinuing the suspected drug or toxin, and PAH therapy may be considered when the haemodynamics have not normalized | IIb | C |
PAH, pulmonary arterial hypertension; PH, pulmonary hypertension. aClass of recommendation. bLevel of evidence.
7.2. Pulmonary arterial hypertension associated with connective tissue disease
Pulmonary arterial hypertension is a well-known pulmonary vascular complication of SSc [173, 502–504], systemic lupus erythematosus (SLE) [505–507], mixed CTD [506], and, rarely, dermatomyositis [508] and Sjögren's syndrome [509]. Conversely, the relationship between rheumatoid arthritis and PAH is not established [510]. After IPAH, PAH-CTD is the second most prevalent type of PAH in western countries [511].
Systemic sclerosis, particularly in its limited variant, represents the main cause of PAH-CTD in Europe and the USA (SLE being more common in Asia) [173, 502, 506]. The prevalence of pre-capillary PH in large cohorts of patients with SSc is 5–19% [173, 502]. In these patients, PH may occur in association with ILD [504, 512] or as a result of PAH [173, 502–504, 506], sometimes with features of venous/capillary involvement [504, 513]. Moreover, group 2 PH-LHD is also common due to myocardial SSc involvement [504, 514]. Of note, patients with SLE may also present with PAH, LHD, ILD, and CTEPH (mostly in the setting of antiphospholipid syndrome). It is therefore essential to carefully determine which mechanism is operative in a given patient, since this will dictate treatment in the context of a multifaceted disease.
Cluster analysis performed in patients with SSc has shown that pre-capillary PH can be characterized into distinct clusters that differ in prognosis [503]. One cluster, characterized by the presence of extensive ILD, and another by severely impaired haemodynamics carried a dismal prognosis, while the two others showed either the absence of ILD or the presence of limited ILD, with mild-to-moderate risk PAH and a relatively favourable overall prognosis [503].
7.2.1. Epidemiology and diagnosis
There is a strong female predominance in PAH-CTD (female/male ratio 4:1), and mean age at diagnosis is commonly >50 years, especially in SSc [173, 502–511, 513, 515, 516]. In the setting of a CTD, patients may present with concomitant disorders such as ILD, and have shorter survival compared with patients with IPAH [503]. The unadjusted risk of death for PAH-SSc compared with IPAH is 2.9, and the predictors of outcome are broadly similar to those for IPAH [516, 517]. Symptoms and clinical presentation are also similar to IPAH, and some patients thought to have IPAH can be identified as having an associated CTD by careful clinical examination and immunological screening tests. Chest CT is recommended for evaluating the presence of associated ILD or PVOD/PCH [504, 513, 515]. An isolated reduction of DLCO is common in PAH-CTD [173, 502–504].
Resting echocardiography combined with other tests is recommended as a screening test in asymptomatic patients with SSc, followed by annual assessments. Screening/early detection is discussed in Section 5.3.1. In other CTDs, PH screening in the absence of suggestive symptoms is not recommended, while echocardiography should be performed in the presence of symptoms. As in other forms of PAH, RHC is recommended in all cases of suspected PAH-CTD to confirm diagnosis, determine severity, and rule out LHD [504].
7.2.2. Therapy
Drugs for PAH should be prescribed in PAH-CTD according to the same treatment algorithm as in IPAH (Figure 9). Patients with PAH-CTD have been included in most of the major RCTs for regulatory approval of PAH therapy [518]. Some aspects of PAH-CTD treatment differ according to the associated CTD [506]. Immunosuppressive therapy combining glucocorticosteroids and cyclophosphamide may result in clinical improvement in patients with SLE- or mixed CTD-associated PAH [506], while it is not recommended in PAH-SSc [519]. Patients with SSc and other CTDs may have ILD and/or HFpEF, which needs to be considered when initiating PAH therapy [504, 515]. In SSc, the long-term risk/benefit ratio of oral anticoagulation is unfavourable because of an increased risk of bleeding, while VKAs are recommended in PAH-CTD with a thrombophilic predisposition (e.g. antiphospholipid syndrome) [319].
Subgroup analyses of patients with PAH-SSc enrolled in RCTs performed with monotherapy or combination therapy of ERAs, PDE5is, sGC stimulators, prostacyclin receptor agonists, epoprostenol, and prostacyclin analogues have shown positive effects versus placebo [301, 401, 519, 520]. In some of these trials, the magnitude of the response in the PAH-CTD subgroup was lower than in the IPAH subgroup [519, 520]. Continuous i.v. epoprostenol therapy improved exercise capacity, symptoms, and haemodynamics in a 3-month RCT in PAH-SSc [401]. However, a retrospective analysis showed a better effect of i.v. epoprostenol on survival in IPAH compared with PAH-SSc [521]. The choice of PAH therapy in the context of SSc and its systemic manifestations may consider other vascular damage such as digital ulcers [522].
Connective tissue disease should not be considered as an a priori contraindication for LTx [523]. This has been extensively studied in SSc, where a multidisciplinary approach optimizing SSc management before, during, and after surgery is recommended [523]. Indications and contraindications for transplantation have to be adapted to the specificities of CTD, with a special focus on digestive (gastro-oesophageal reflux disease and intestinal disease), cardiac, renal, and cutaneous involvement [523].
Recommendations | Classa | Levelb |
In patients with PAH associated with CTD, treatment of the underlying condition according to current guidelines is recommended [166, 167, 419, 524] | I | A |
In patients with PAH associated with CTD, the same treatment algorithm as for patients with IPAH is recommended | I | C |
PAH-CTD, pulmonary arterial hypertension associated with connective tissue disease; IPAH, idiopathic pulmonary arterial hypertension. aClass of recommendation. bLevel of evidence.
7.3. Pulmonary arterial hypertension associated with human immunodeficiency virus infection
The use of highly active antiretroviral therapy (HAART), and advances in managing opportunistic infections have contributed to increased life expectancy in patients with HIV [525, 526]. Consequently, the spectrum of complications has shifted towards other long-term conditions, including PAH. Clinical and histopathological findings in PAH associated with HIV infection (PAH-HIV) share many similarities with IPAH [1, 527]. With the availability of HAART given in combination with PAH therapies, the prognosis of PAH-HIV has markedly improved in recent years [526, 528]. In addition, the incidence of PAH-HIV has declined in parallel with the increasing availability of HAART [528]. Taken together, these effects on survival and incidence have resulted in a stable PAH prevalence in patients with HIV over recent decades. A French population study indicated that the prevalence of PAH in individuals with HIV infection was 0.46%, which is very similar to the prevalence before the HAART era [177].
The pathogenesis of PAH-HIV remains unclear. There is no evidence of a direct role of HIV in the pathogenesis of PAH and, although present in inflammatory cells in the lungs, the virus itself has never been found in pulmonary vascular lesions of patients with PAH-HIV [529]. This suggests that an indirect action of viral infection on inflammation and growth factors may act as a trigger in a predisposed patient.
7.3.1. Diagnosis
Pulmonary arterial hypertension associated with HIV shares a clinical presentation with IPAH. Before the availability of HAART most patients were in WHO-FC III or IV at diagnosis. Nowadays, patients are diagnosed with much less severe symptoms and haemodynamics. Patients may present with other risk factors for PAH such as liver disease (chronic viral hepatitis B or C) or exposure to drugs or toxins. Patients with PAH-HIV are more likely to be male and i.v. drug abusers [403, 526]. There is no correlation between the severity of PAH and the stage of HIV infection or the degree of immunodeficiency [403, 530]. Because of its low prevalence, asymptomatic patients with HIV should not be screened for PAH. However, echocardiography should be performed in patients with unexplained dyspnoea to detect HIV-related cardiovascular complications such as myocarditis, cardiomyopathy, or PAH. Right heart catheterization is mandatory to confirm the diagnosis of PAH-HIV and to rule out LHD [527].
Pulmonary arterial hypertension is an independent risk factor for death in patients with HIV. In the 1990s, before the availability of HAART, patients with PAH-HIV had poor outcomes, with a 3 year survival of <50% [403]. The overall survival has now improved and patients with PAH-HIV have a better prognosis than most patients with other forms of PAH [526].
7.3.2. Therapy
Current recommendations for the treatment of PAH-HIV are largely based on data from IPAH [25, 26].
Treatment of PAH-HIV with HAART has improved functional status and survival in some retrospective studies [525, 526, 531]. The use of HAART in PAH-HIV is therefore recommended, irrespective of viral load and CD4+ cell count.
Anticoagulation is not recommended because of an increased risk of bleeding and drug interactions [319, 527]. Patients with PAH-HIV are usually non-responders to acute vasoreactivity testing and therefore should not receive CCBs [378].
The prospective, open-label, BREATHE-4 study showed that bosentan markedly improved WHO-FC, exercise capacity, quality of life, and haemodynamics after 16 weeks in patients with PAH-HIV [532]. In a long-term, retrospective series, bosentan therapy was associated with haemodynamic normalization in 10/59 patients [533]. Bosentan potentially interacts with antiretroviral drugs, and close monitoring is required when combined with HAART. Very few patients with PAH-HIV have been included in RCTs with ambrisentan and macitentan, and no definite conclusion can be drawn from those studies.
Positive effects of sildenafil and tadalafil in PAH-HIV have been established in case studies [534, 535]. Interactions have been reported between PDE5is and protease inhibitors, resulting in major increases in PDE5i concentrations; these drugs should be introduced at low dosages with careful monitoring of potential side effects, including hypotension [536, 537]. There are no data on the use of the sGC stimulator riociguat in PAH-HIV.
Treatment with i.v. epoprostenol resulted in significant improvement in WHO-FC, exercise capacity, haemodynamics, and survival in selected patients with PAH-HIV [403, 538]. There are very few data on the use of i.v. or s.c. treprostinil or inhaled iloprost in PAH-HIV [539, 540].
There are no clinical trial data on the use of combination therapy for PAH-HIV. Given the lack of supporting evidence and potential safety concerns when PAH drugs are co-administered with antiretroviral drugs, initial monotherapy with PAH medication is recommended, followed by an individualized use of combination therapy in patients who do not reach a low-risk profile.
Recommendations | Classa | Levelb |
In patients with PAH associated with HIV infection, antiretroviral treatment according to current guidelines is recommended [541, 542] | I | A |
In patients with PAH associated with HIV infection, initial monotherapy should be considered, followed by sequential combination if necessary, taking into consideration comorbidities and drug–drug interactions | IIa | C |
HIV, Human immunodeficiency virus; PAH, pulmonary arterial hypertension. aClass of recommendation. bLevel of evidence.
7.4. Pulmonary arterial hypertension associated with portal hypertension
Pulmonary arterial hypertension associated with portal hypertension, commonly referred to as PoPH, develops in 2–6% of patients with portal hypertension, with or without liver disease. In PAH registries, PoPH represents 5–15% of the patients [543–545]. Rarely, some patients with PoPH have portosystemic shunts in the absence of portal hypertension (congenital extrahepatic cavoportal shunts) [546]. However, PoPH is distinct from hepatopulmonary syndrome (HPS), which is characterized by intrapulmonary vascular dilatations and hypoxaemia. Of note, HPS and PoPH can occur sequentially or concurrently in patients with portal hypertension [547].
7.4.1. Diagnosis
The diagnosis of PoPH is based on the presence of otherwise unexplained pre-capillary PH in patients with portal hypertension or a portosystemic shunt. The diagnostic approach is the same as in other patients with suspected or newly detected PH. Transthoracic echocardiography is usually the first non-invasive assessment in patients with suspected PH, and echocardiography is also recommended as a screening tool in patients evaluated for liver transplantation. As patients with liver disease often have an elevated CO, TRV tends to overestimate PAP in these patients. Hence, RHC with comprehensive haemodynamic assessment is essential to confirm the diagnosis of PH and to distinguish PAH (with elevated PVR) from unclassified PH (with a normal PVR).
7.4.2. Therapy
Patients with unclassified PH (i.e. mPAP >20 mmHg, elevated CO, and PVR ≤2.0 WU) should be regularly followed-up but should not be treated with drugs approved for PAH.
In patients with an established diagnosis of PoPH, treatment should follow the same general principles as in other patients with PAH, taking into account the severity of underlying liver disease, the indication for liver transplantation, and the potential effects of PAH medication on gas exchange, which may deteriorate with vasodilators in patients with PoPH [548, 549]. All drugs approved for PAH can principally be used to treat patients with PoPH, bearing in mind that these patients are usually excluded from registration studies. Nevertheless, various case series support the use of approved PAH medication in patients with PoPH. The largest series published so far reported on 574 patients with PoPH treated with various PAH drugs, mostly PDE5is or ERAs, alone and in combination [545]. Most patients (56.8%) were in Child–Pugh class A at the time of PAH diagnosis. At the first follow-up, which took place 4.5 months after starting treatment, improvements were seen in haemodynamics, WHO-FC, 6MWD, and BNP/NT-proBNP; survival at 5 years was 51%. In patients presenting with mild liver disease, the main causes of death were PAH progression and malignancy, whereas complications of liver disease were the most common causes of death in patients with advanced liver disease. The 5 year survival of patients who underwent liver transplantation (n=63) was 81%.
The only RCT dedicated to the treatment of PoPH was PORTICO, a 12 week study that randomized 85 patients to macitentan (n=43) or placebo (n=42) [168]. PORTICO met its primary endpoint, demonstrating a significant reduction in PVR from baseline (ratio of geometric mean 0.65; 95% CI, 0.59–0.72; P<0.0001). There were, however, no differences between the two treatment groups in secondary outcome measures, including WHO-FC, 6MWD, and NT-proBNP.
7.4.2.1. Liver transplantation
Porto-pulmonary hypertension is not per se an indication for liver transplantation. Pulmonary arterial hypertension poses a major threat to patients who undergo liver transplantation when indicated for the severity of liver disease. In a historical series from the Mayo Clinic, severe PAH with mPAP ≥50 mmHg was associated with a 100% peri-operative mortality rate. In patients with mPAP 35–50 mmHg and PVR >3.0 WU, mortality was still 50% [550]. In liver transplantation candidates with PAH, targeted medical therapy successfully improves haemodynamics and establishes eligibility for transplantation [545, 551–554]. However, haemodynamic criteria for successful liver transplantation have not been firmly established. The International Liver Transplant Society proposed haemodynamic targets of mPAP <35 mmHg and PVR <5 WU, or mPAP ≥35 mmHg and PVR <3 WU in patients receiving PAH therapy, while acknowledging that these criteria need to be further validated [175]. An mPAP ≥45 mmHg is regarded as an absolute contraindication to liver transplantation [175].
In patients with PoPH who successfully underwent liver transplantation, de-escalation or discontinuation of PAH medication is often feasible, but this has to be performed on an individual basis [551, 554].
Recommendations | Classa | Levelb |
Echocardiography is recommended in patients with liver disease or portal hypertension with signs or symptoms suggestive of PH, and as a screening tool in patients evaluated for liver transplantation or transjugular portosystemic shunt | I | C |
It is recommended that patients with PAH associated with portal hypertension are referred to centres with expertise in managing both conditions | I | C |
In patients with PAH associated with portal hypertension, initial monotherapy should be considered, followed by sequential combination if necessary, taking into consideration the underlying liver disease and the indication for liver transplantation | IIa | C |
Liver transplantation should be considered on an individual basis in patients with PAH associated with portal hypertension, as long as PVR is normal or near normal with PAH therapy | IIa | C |
Drugs approved for PAH are not recommended for patients with portal hypertension and unclassified PH, i.e. elevated mPAP, high CO, and a normal PVR | III | C |
mPAP, mean pulmonary arterial pressure; CO, cardiac output; PAH, pulmonary arterial hypertension; PH, pulmonary hypertension; PVR, pulmonary vascular resistance. aClass of recommendation. bLevel of evidence.
7.5. Pulmonary arterial hypertension associated with adult congenital heart disease
The presence of PH in adults with CHD has a negative impact on the natural course of CHD, and worsens clinical status and overall outcome [555]. Pulmonary arterial hypertension associated with adult CHD is included in group 1 of the PH clinical classification (Table 6) and represents a heterogeneous patient population. Post-capillary PH in adult CHD (e.g. systolic or diastolic, systemic, ventricular dysfunction in combination with shunt lesions or complex adult CHD, and systemic atrioventricular valve dysfunction) should be excluded to determine further management. A specific clinical classification (Table 21) is provided to better characterize PAH associated with adult CHD. Some complex CHDs are associated with congenital abnormalities of the pulmonary vascular tree leading to segmental PH. In segmental PH, one or more, but not all, segments of the lung(s) are hypertensive and each hypertensive area may present with PH of different severity, while other parts of the lung vasculature may be hypoplastic. Pulmonary atresia with ventricular septal defect and systemic-to-pulmonary collaterals is the most frequent condition, but other complex CHDs may also lead to segmental PH.
Approximately 3–7% of patients with adult CHD will eventually develop PAH; it is more frequently encountered in females, and the incidence depends on the underlying lesion and increases with age and age at defect closure [556]. The estimated prevalence of PAH in patients after correcting a simple cardiac defect is 3% [557]. The epidemiology of PAH associated with adult CHD is expected to change due to advances in diagnostic and therapeutic paediatric cardiology, resulting in fewer patients with simple adult CHD and more patients with complex lesions and/or closed defects who develop PAH in adulthood [558].
The clinical presentation of Eisenmenger syndrome, an advanced form of adult CHD-associated PAH, is characterized by the multiorgan effects of chronic hypoxaemia, including cyanosis, and haematological changes, including secondary erythrocytosis and thrombocytopenia; the main symptoms are dyspnoea, fatigue, and syncope. Eisenmenger syndrome may also present with haemoptysis, chest pain, cerebrovascular accidents, brain abscesses, coagulation abnormalities, and sudden death. Patients with adult CHD and Down syndrome are at an increased risk of developing Eisenmenger syndrome.
7.5.1. Diagnosis and risk assessment
The diagnostic work-up of PAH associated with adult CHD should be based on the presence of symptoms and includes medical history, physical examination, PFTs, ABG, imaging (especially echocardiography), and exercise and laboratory testing. Of note, standard echocardiographic criteria for detecting PH may not be applicable in complex adult CHD [559]. Right heart catheterization with compartmental oximetry for calculating pulmonary blood flow/systemic blood flow (Qp/Qs) is required to confirm PAH diagnosis and guide therapeutic interventions. Thermodilution should be avoided in the presence of intracardiac shunts, and direct Fick is the most accurate method. Pulmonary vascular resistance may be overestimated due to erythrocytosis [560]. Interpreting invasive haemodynamics (see Section 5.1.12) should be made in the context of multiparametric assessment of exercise capacity, laboratory testing, and imaging.
Predictors of worse outcomes in adult CHD-associated PAH are WHO-FC III–IV, exercise intolerance assessed by 6MWD or peak VO2, history of hospitalization for right HF, biomarkers (NT-proBNP >500 pg/mL, C-reactive protein >10 mg/mL, high serum creatinine, and low albumin levels), iron deficiency, and echocardiographic indices of RV dysfunction [559, 561]. When compared with patients with IPAH, patients with Eisenmenger syndrome may have a relatively stable long-term clinical course. The right ventricle is unloaded by the right-to-left shunt, sustaining CO at the expense of hypoxaemia and cyanosis. However, due to immortal time bias, prognosis of Eisenmenger syndrome is not as favourable as previously thought [562].
As in other forms of PAH, risk assessment is important to guide therapy, and specific risk factors have been described in Eisenmenger syndrome. A large multicentre study showed that mortality in adults with Eisenmenger syndrome was predicted by the presence of pre-tricuspid shunt, advancing age, low rest oxygen saturation, absence of sinus rhythm, and presence of pericardial effusion [563].
7.5.2. Therapy
Outcomes in adult CHD-associated PAH have improved with the availability of new PAH therapies, advances in surgical and peri-operative management, and a team-based, multidisciplinary approach in PH centres. These patients should be managed by specialized health professionals. Patient education, behavioural modifications, and social and psychological support are all important aspects of management.
Shunt closure (surgical or interventional) may only be considered in patients with prevalent systemic-to-pulmonary shunting without significantly increased PVR. Criteria for defect closure based on Qp/Qs ratio and (baseline and/or after targeted PAH treatment) PVR have been proposed by the 2020 ESC Guidelines for the management of adult congenital heart disease [101]. Decisions on shunt closure should not be made on haemodynamic numbers alone, and a multiparametric strategy should be followed. For instance, shunt closure is not indicated in the case of desaturation during exercise in the 6MWT or CPET, or when there is secondary erythrocytosis suggesting dynamic reversal of shunt. There is no evidence for a long-term benefit of a treat-and-repair approach in patients with adult CHD-associated PAH with prevalent systemic-to-pulmonary shunts; therefore, there is a need for future prospective studies [564]. Defect closure is contraindicated in all patients with Eisenmenger syndrome, and may also adversely affect patients with small/coincidental defects that behave similarly to IPAH [565]. There are no prospective data available on the usefulness of vasoreactivity testing, balloon closure testing, or lung biopsy for assessing operability and normalization of PVR after closure [566].
Patients with adult CHD-associated PAH may present with clinical deterioration in different circumstances, such as arrhythmia, during non-cardiac surgery requiring general anaesthesia, dehydration or bleeding, thrombo-embolism, and lung infections. Surgeries should be limited to those deemed essential, and performed in specialized centres with anaesthetists experienced in adult CHD and PAH. Endocarditis should be suspected in patients with sepsis, whereas a cerebral abscess should be excluded in those with neurological symptoms or new headache, especially in those with low oxygen saturations and complex anatomies. It is recommended to avoid strenuous exercise, but mild and moderate activities seem to be beneficial [567]. Patients should receive all recommended vaccinations and endocarditis prophylaxis in the presence of cyanosis. Although pregnant patients with left-to-right shunts and stable, well-controlled PAH have tolerated pregnancy well under specialized care, pregnancy is still associated with both high maternal mortality and foetal complications in Eisenmenger syndrome and should be discouraged in this setting [568, 569]; hence, effective contraception is highly recommended. Levonorgestrel-based, long-acting, reversible contraception implants or intrauterine devices have been recommended for these patients [570].
Secondary erythrocytosis is beneficial for adequate oxygen transport and delivery, and routine phlebotomy should be avoided whenever possible. Symptoms of hyperviscosity in the presence of haematocrit >65% should be approached with appropriate hydration. Iron deficiency should be corrected. When i.v. iron supplementation is administered, special care should be taken to avoid air emboli during administration [571]. Supplemental oxygen therapy has not been shown to impact survival.
Oral anticoagulant treatment with VKAs may be considered in patients with large PA aneurysms with thrombus, atrial arrhythmias, and previous thrombo-embolic events, but with low bleeding risk. In patients with very high Hb levels (>20 mg/dL), standard international normalized ratio measures are less accurate, and citrate-adjusted blood bottles must be used. Regarding using novel oral anticoagulants (NOACs), a large, nationwide, German, adult CHD database (including 106 NOAC-treated patients with Eisenmenger syndrome) showed that NOAC users had higher long-term risk of bleeding, major adverse cardiovascular events, and mortality compared with those on VKAs, suggesting that initiating NOACs should be reserved for experienced adult CHD centres, carefully weighing potential benefits and risks [572, 573].
Compared with other group 1 subgroups, limited data exist on the use of drugs approved for PAH in patients with adult CHD-associated PAH. Bosentan improved 6MWD and decreased PVR in patients with Eisenmenger syndrome in WHO-FC III [574]. Patients with more complex lesions were less likely to respond to PAH therapies compared with patients with simple lesions. An RCT investigating the efficacy of macitentan found no effect on 6MWD in a mixed cohort of patients with Eisenmenger syndrome (6MWD improved in both treatment and placebo arms), although decreases in NT-proBNP and PVR were noted in the macitentan arm [575].
Experiences with other ERAs and PDE5is have shown favourable functional and haemodynamic results in Eisenmenger syndrome [576]. In a small, single-centre, pilot study, adding nebulized iloprost to a background of oral PAH therapy failed to improve 6MWD in Eisenmenger syndrome [577]. In case symptoms persist or in clinical deterioration, a sequential and symptom-orientated treatment strategy is recommended in Eisenmenger syndrome, starting with an oral ERA (or PDE5i) and escalating therapy. Should symptoms not adequately improve with oral therapies, i.v./s.c. options should be proactively considered [578]. There is a theoretical risk of paradoxical embolism in right-to-left shunt lesions with the presence of a central venous catheter for i.v. therapy; therefore, s.c. prostacyclin analogue infusion may be considered.
The effect of PAH therapies in patients with prevalent systemic-to-pulmonary shunts is less well established. Patients with small/coincidental defects should be treated with PAH medication [557]. This is also the case for patients with PAH after defect correction who have increased mortality compared with those with Eisenmenger syndrome [579]. These patients were included in major RCTs with PAH therapies and should be evaluated based on comprehensive risk assessment (Table 16) [580]. The effect of PAH therapies in patients with segmental PH remains a matter of debate [101, 581]. While some series have reported promising results, there have been cases where therapies were not tolerated [581]. Similarly, using PAH therapies in Fontan circulation has yielded conflicting results, and results of further studies are awaited [582–584].
Heart–lung transplantation or LTx with heart surgery is an option in highly selected cases not responsive to medical treatment; however, it is limited by organ availability and lesion complexity. Mortality is high during the first year after surgery, especially after heart–lung transplantation, but remains relatively low thereafter [585].
Recommendations | Classa | Levelb |
In patients with ASD, VSD, or PDA and a PVR <3 WU, shunt closure is recommended | I | C |
In patients with ASD, VSD, or PDA and a PVR of 3–5 WU, shunt closure should be considered | IIa | C |
In patients with ASD and a PVR >5 WU that declines to <5 WU with PAH treatment, shunt closure may be considered | IIb | C |
In patients with VSD or PDA and a PVR >5 WU, shunt closure may be considered after careful evaluation in specialized centres | IIb | C |
In patients with ASD and a PVR >5 WU despite PAH treatment, shunt closure is not recommended | III | C |
ASD, atrial septal defect; PAH, pulmonary arterial hypertension; PDA, patent ductus arteriosus; PVR, pulmonary vascular resistance; VSD, ventricular septal defect; WU, Wood units. Decisions on shunt closure should not be made on haemodynamic numbers alone; a multiparametric strategy should be followed (see Section 7.5.2). aClass of recommendation. bLevel of evidence.
Recommendations | Classa | Levelb |
Risk assessment | ||
Risk assessment is recommended for patients with persistent PAH after defect closure | I | C |
Risk assessment should be considered in patients with Eisenmenger syndrome | IIa | C |
Treatment | ||
Bosentan is recommended in symptomatic patients with Eisenmenger's syndrome to improve exercise capacity [574] | I | B |
In patients with Eisenmenger syndrome, the use of supplemental oxygen therapy should be considered in cases where it consistently increases arterial oxygen saturation and reduces symptoms | IIa | C |
Supplemental iron treatment should be considered in patients with iron deficiency | IIa | C |
In patients with adult CHD, including Eisenmenger syndrome, other ERAs, PDE5is, riociguat, prostacyclin analogues, and prostacyclin receptor agonists should be considered | IIa | C |
In patients with PAH after corrected adult CHD, initial oral combination therapy with drugs approved for PAH should be considered for patients at low and intermediate risk, while initial combination therapy including i.v./s.c. prostacyclin analogues should be considered for patients at high risk | IIa | Cc |
In patients with adult CHD, including Eisenmenger syndrome, sequential combination therapy should be considered if patients do not meet treatment goals | IIa | C |
In the absence of significant haemoptysis, oral anticoagulant treatment may be considered in patients with Eisenmenger syndrome with pulmonary artery thrombosis | IIb | C |
In women with Eisenmenger syndrome, pregnancy is not recommended | III | C |
In patients with Eisenmenger syndrome, routine phlebotomy to lower elevated haematocrit is not recommended | III | C |
CHD, congenital heart disease; ERA, endothelin receptor antagonist; i.v., intravenous; PAH, pulmonary arterial hypertension; PDE5i, phosphodiesterase 5 inhibitor; s.c., subcutaneous. aClass of recommendation. bLevel of evidence. cLevel of evidence differs from the 2020 ESC Guidelines for the management of adult congenital heart disease because the number of patients with adult CHD included in the AMBITION study was very low.
7.6. Pulmonary arterial hypertension associated with schistosomiasis
Schistosomiasis is one of the most common chronic infectious diseases worldwide, affecting around 200 million people [586, 587]. Schistosomiasis-associated PAH is present in 5% of patients with the hepatosplenic form of the disease [586]. It is thus a leading cause of PAH, especially in some regions of South America, Africa, and Asia. Compared with patients with IPAH, patients with schistosomiasis-associated PAH present with higher CO and lower PVR, and have a better survival [587]. Registry data suggest that survival in schistosomiasis-associated PAH has improved in recent years with the use of PAH drugs [588].
7.7. Pulmonary arterial hypertension with signs of venous/capillary involvement
The common risk factors, identical genetic substrate, and indistinguishable clinical presentations of PCH and PVOD necessitate their consideration as a single disease belonging to the group 1 PH spectrum of diseases (PAH with signs of venous/capillary involvement) [1, 425, 589]. In PVOD/PCH, post-capillary lesions affecting septal veins and pre-septal venules consist of loose, fibrous remodelling of the intima that may totally occlude the lumen [1, 425, 589, 590]. These changes are frequently associated with PCH consisting of capillary ectasia and proliferation, with doubling and tripling of the alveolar septal capillary layers that may be focally distributed within the alveolar interstitium [425, 590].
The proportion of patients with IPAH that fulfil the criteria for PVOD/PCH is ∼10%, resulting in a lowest estimate of PVOD/PCH incidence and prevalence of <1 case/million [425]. In contrast to IPAH, there is a male predominance in PVOD/PCH and its prognosis is worse [425, 589, 591]. Familial PVOD/PCH typically occurs in the young siblings of one generation, with unaffected and sometimes consanguineous parents, indicating that the disease segregates as a recessive trait [158, 425, 591]. Biallelic mutations in the EIF2AK4 gene cause heritable PVOD/PCH [158]. In addition, PVOD/PCH can complicate the course of associated conditions, such as SSc [425], or be associated with exposure to environmental triggers, such as alkylating agents (cyclophosphamide, mitomycin C) [34] and solvents (trichloroethylene) [38].
7.7.1. Diagnosis
Most patients complain of non-specific dyspnoea on exertion and fatigue [590]. Physical examination may reveal digital clubbing and bibasal crackles on lung auscultation [590]. Pulmonary arterial hypertension and PVOD/PCH share the same haemodynamic profile as pre-capillary PH [590, 591]. The PAWP is not elevated because the pulmonary vascular changes occur in small venulae and capillaries, while the LA filling pressure remains normal [590, 591]. A diagnosis of PVOD/PCH is based on the results of tests suggesting venous post-capillary involvement, chronic interstitial pulmonary oedema, and capillary proliferation [1, 590, 591]. These tests include PFTs (decreased DLCO, frequently <50% theoretical values), ABG (hypoxaemia), and non-contrast chest CT (subpleural thickened septal lines, centrilobular ground-glass opacities, and mediastinal lymphadenopathy) [1, 425, 589, 591, 592]. Importantly, these patients are at risk of drug-induced pulmonary oedema with PAH therapy, a finding suggestive of PVOD/PCH [425, 591]. Detecting biallelic EIF2AK4 mutations is sufficient to confirm a diagnosis of heritable PVOD/PCH [158, 591, 592]. Lung biopsy is hazardous in PH and is not recommended for diagnosing PVOD/PCH [1, 425].
7.7.2. Therapy
There is no established medical therapy for PVOD/PCH [425]. Compared with IPAH, PVOD/PCH has a poor prognosis and limited response to PAH therapy, with a risk of pulmonary oedema due to pulmonary venous obstruction [425, 591]. However, there are reports of incomplete and transient clinical improvement in individual patients with PVOD/PCH treated with PAH therapy, which should be used with great caution in this setting [425, 591]. Diuretics, oxygen therapy, and slow titration of PAH therapy can be used on an individual basis [425]. Therefore, therapy for PVOD/PCH should be undertaken at centres with extensive experience in managing PH, and patients should be fully informed about the risks [425]. Anecdotal reports suggest a potential benefit of immunomodulatory treatments, but this approach requires further study [593]. The only curative therapy for PVOD/PCH is LTx, and eligible patients should be referred to a transplant centre for evaluation upon diagnosis [425, 591]. Pathological examination of the explanted lungs will confirm the diagnosis [590].
Recommendations | Classa | Levelb |
A combination of clinical and radiological findings, ABG, PFTs, and genetic testing is recommended to diagnose PAH with signs of venous and/or capillary involvement (PVOD/PCH) [591] | I | A |
Identification of biallelic EIF2AK4 mutations is recommended to confirm a diagnosis of heritable PVOD/PCH [158, 591] | I | A |
Referral of eligible patients with PVOD/PCH to a transplant centre for evaluation is recommended as soon as the diagnosis is established | I | C |
In patients with PVOD/PCH, the use of drugs approved for PAH may be considered with careful monitoring of clinical symptoms and gas exchange | IIb | C |
Lung biopsy is not recommended to confirm a diagnosis of PVOD/PCH | III | C |
ABG, arterial blood gas analysis; PAH, pulmonary arterial hypertension; PCH, pulmonary capillary haemangiomatosis; PFT, pulmonary function test; PVOD, pulmonary veno-occlusive disease. aClass of recommendation. bLevel of evidence.
7.8. Paediatric pulmonary hypertension
Pulmonary hypertension may present at all ages, including in infants and children. Pulmonary hypertension in childhood shares many common features with PH in adulthood; however, there are also important differences, which concern epidemiology, aetiology, genetic background, age-dependent diagnostic and treatment approaches, and disease monitoring. An important and conceptually distinctive feature of paediatric PH is injury to developing foetal, neonatal, or paediatric lung circulation.
7.8.1. Epidemiology and classification
The reported annual incident rate for paediatric PH is 64/million children [594]. The distribution of the various aetiologies of PH in childhood differs from PH in adulthood [594–596]. Pulmonary arterial hypertension is the most frequent type of PH in children, with the vast majority (82%) of cases being infants with transient PAH (i.e. PPHN or repairable cardiac shunt defects). Of the remaining children with PAH, most have either IPAH, HPAH, or irreversible CHD-associated PAH. The reported incidences of IPAH/HPAH and (non-transient) CHD-associated PAH are 0.7 and 2.2/million children, respectively, with a prevalence of 4.4 and 15.6/million children, respectively [594]. Other conditions associated with PAH (Table 6) do occur in children but are rare.
Another significant proportion (34–49%) of children with non-transient PH are neonates and infants with PH associated with respiratory disease, especially developmental lung diseases, including bronchopulmonary dysplasia (BPD), congenital diaphragmatic hernia (CDH), and congenital pulmonary vascular abnormalities [594–598]. These children form a prominent and distinctive group in paediatric PH and are currently classified as PH group 3 associated with developmental lung disease (Table 6; Table S7). A significant and growing proportion of children with PH associated with respiratory disease is made up of pre-term infants with BPD. Also, newly recognized genetic developmental lung disorders—including alveolar capillary dysplasia, TBX4-mutation-related lung disorders, and surfactant abnormalities—are currently classified in this category (Figure 10) [599].
Another distinctive feature of PH in children is the high burden of genetic disorders. Childhood PH is often associated with chromosomal, genetic, and syndromic anomalies (11–52%). Like in adults, gene mutations implicated in the pathogenesis of HPAH are found in 20–30% of sporadic cases, where paediatric HPAH seems to be characterized by an enrichment in TBX4 and ACVRL1 variations [600, 601]. Additionally, 17% of children with PAH have other disorders known to be associated with PAH, including trisomy 21. Finally, 23% of children with PAH have copy number variations not previously associated with PH [600, 602, 603].
Given the frequent association of paediatric PAH with chromosomal, genetic, and syndromic anomalies (for which the mechanistic basis for PAH is generally uncertain), genetic testing may be considered for defining aetiology and comorbidities, stratifying risk, and identifying family members at risk; however, this should be after appropriate expert genetic counselling for the child and family (see Section 5.1.13).
The clinical PH classification (Table 6) is also followed for paediatric PH. To improve applicability of this classification in infants and children with PH, it has been adapted to give room to PH associated with various congenital cardiovascular and pulmonary diseases or specific paediatric conditions (Tables S5–S8) [599].
7.8.2. Diagnosis and risk assessment
Historically, the definition of PH in children aged >3 months has been the same as in adults. The definition for PH has now been redefined to mPAP >20 mmHg in adults as well as in children. The impact of an mPAP 21–24 mmHg on outcomes in children is unknown. However, in the interest of consistency and to facilitate transition from paediatric to adult PH care, it is recommended that the updated definition for PH also be followed in children. No treatment recommendations currently exist for this group of children (mPAP 21–24 mmHg).
Regarding the newly introduced criterion to include PVR >2 WU to identify pre-capillary PH in adults, PVR had previously been included in the definition for PAH in children. In children, blood flows are traditionally indexed assuming that systemic and pulmonary blood flows change proportionally with body size, while the transpulmonary pressure gradient does not. Since blood flow is the denominator in the equation for calculating PVR, the need for indexing of PVR in children is emphasized, and the criterion of pulmonary vascular resistance index (PVRI) ≥3 WU·m2 in the definition for PAH in children remains unchanged [599].
Since the aetiology of paediatric PH is very diverse, a methodical and comprehensive diagnostic approach is crucial to reach an accurate diagnosis and treatment plan. As in adults, IPAH is a diagnosis ‘per exclusion’. A diagnostic work-up, similar to that in adults but customized for paediatric PH, is recommended [599]. Pre-term infants with BPD should be screened for PH, since PH is prevalent in this population and seriously affects outcome [604].
Also in children, RHC is the gold standard for definitively diagnosing and establishing the nature of PH, and provides important data for stratifying risk [604a, 605]. To identify those suitable for high-dose CCB treatment, acute vasoreactivity testing is recommended in children with IPAH/HPAH. The criteria used in adults for a positive acute response have identified children who will show sustained benefit from CCB therapy; however, these criteria do not define reversibility of PAH or operability in children with CHD. Since RHC in children with PH may be associated with major complications (in 1–3% of cases, especially in young infants and those in worse clinical condition), risks and benefits have to be balanced in the individual child [605]. Heart catheterization in children with PAH should be exclusively performed in experienced paediatric PH centres. Indications for repeated RHC in children with PH are currently not well defined.
Treatment of children with PAH is based on risk stratification [599]. Predictors of worse outcome in paediatric PAH are similar to those in adults, and include clinical evidence of RV failure, progression of symptoms, WHO-FC III–IV, certain echocardiographic parameters (e.g. TAPSE), and elevated serum NT-proBNP. A 6MWD <350 m has also been suggested as a predictor of worse outcome in paediatric PH, but its value in young children is less established. Further prognosticators identified in paediatric PAH are failure to thrive and haemodynamic variables, such as RAP >10 mmHg, the ratio of mean pulmonary-to-systemic blood pressure >0.75, and PVRI >20 WU·m2 [602, 606, 607]. Paediatric risk-assessment tools based on these parameters have been retrospectively validated in observational paediatric registries [599, 604a].
7.8.3. Therapy
The ultimate goal of treatment should be to improve survival and facilitate normal childhood activities without limitations. In the absence of RCTs in paediatric PAH, recommended treatment algorithms are extrapolated from those in adults and enhanced with data from observational studies in children with PAH [599].
Observational cohort studies support treatment algorithms designed for adults to be used for children (including the superiority of combination therapy over monotherapy) [608]. Drugs investigated in children, with or without formal approval by the European Medicines Agency (EMA) for treating children with PAH, are shown in Table 22.
A paediatric treatment algorithm, derived from that for adults, is based on risk stratification, recommending general measures, high-dose CCB therapy for responders to acute vasoreactivity testing (where close follow-up is mandatory, as some patients may fail long-term therapy), oral or inhaled combination therapy for children at low risk, and combination therapy with i.v./s.c. prostacyclin analogues for those at high risk [599].
In the case of insufficient response to recommended drug therapy, or when drugs are unavailable, a Potts shunt (a surgical or interventional connection between the left PA and the descending aorta), BAS, or LTx may be considered in children with severe PH (see Sections 6.3.6.1 and 6.3.8) [599]. Reported clinical experience with Potts shunts is limited to just over 100 patients, predominantly children, with a mortality of 12–25% and long-term clinical benefit in a subset of children with long-term follow-up [456–459].
Monitoring of treatment effect and disease course is pivotal in managing all patients with PAH (adults and in children). In children with PAH, clinical risk scores including WHO-FC, TAPSE, and serum NT-proBNP are potential treatment targets for goal-orientated treatment [604a, 609].
Contemporary treatment algorithms for infants with PPHN have been proposed but are outside the scope of these guidelines [610].
The recommendations discussed above apply to children with PAH, whereas the specific group of infants with neonatal PVD, mostly classified as PH associated with developmental lung disease and with heterogeneous aetiology, require a distinct and customized approach (Figure 10).
In pre-term infants with BPD and PH, the underlying lung disease should primarily be treated. Frequently, these infants are additionally treated with therapies for PAH, including sildenafil and bosentan; however, these are not approved by the EMA for use in infants with group 3 PH and developmental lung diseases (BPD, CDH). Their effects on outcomes in this population are unclear, and data enabling robust treatment recommendations are lacking. These children should be treated by multidisciplinary teams involving cardiologists, neonatologists, pulmonologists, and nutritionists. Pulmonary hypertension in these infants may disappear with lung healing, although long-term cardiovascular sequelae have been reported [611, 612].
Recommendations | Classa | Levelb |
Children | ||
It is recommended to perform the diagnostic work-up, including RHC and acute vasodilator testing, and treat children with PH at centres with specific expertise in paediatric PH | I | C |
In children with PH, a comprehensive work-up for confirming diagnosis and specific aetiology is recommended (similar to that in adults, but adapted for age) | I | C |
For confirming PH diagnosis, RHC is recommended, preferably before initiating any PAH therapy | I | C |
In children with IPAH/HPAH, acute vasoreactivity testing is recommended to detect those who may benefit from CCB therapy | I | C |
It is recommended to similarly define a positive response to acute vasoreactivity testing in children and adults by a reduction in mPAP ≥10 mmHg to reach an absolute value of mPAP ≤40 mmHg, with an increased or unchanged CO | I | C |
In children with PAH, a therapeutic strategy based on risk stratification and treatment response is recommended, extrapolated from that in adults but adapted for age | I | C |
It is recommended to monitor the treatment response in children with PAH by serially assessing a panel of data derived from clinical assessment, echocardiographic evaluation, biochemical markers, and exercise tolerance tests | I | C |
Achieving and maintaining a low-risk profile should be considered as an adequate treatment response for children with PAH | IIa | C |
Infants | ||
It is recommended to screen infants with bronchopulmonary dysplasia for PH [628, 629] | I | B |
In infants with (or at risk of) bronchopulmonary dysplasia and PH, treating lung disease—including hypoxia, aspiration, and structural airway disease—and optimizing respiratory support is recommended before initiating PAH therapy [630] | I | B |
In neonates and infants, a diagnostic and therapeutic approach to PH distinct from that in older children and adults should be considered, given the frequent association with developmental vascular and parenchymal lung disease | IIa | C |
CCB, calcium channel blocker; CO, cardiac output; HPAH, heritable pulmonary arterial hypertension; IPAH, idiopathic pulmonary arterial hypertension; mPAP, mean pulmonary arterial pressure; PAH, pulmonary arterial hypertension; PH, pulmonary hypertension; RHC, right heart catheterization. aClass of recommendation. bLevel of evidence.
8. Pulmonary hypertension associated with left heart disease (group 2)
8.1. Definition, prognosis, and pathophysiology
Among patients with LHD, PH and RV dysfunction are frequently present and associated with high mortality [47]. This includes patients with HF with reduced, mildly reduced, or preserved ejection fraction (HFrEF, HFmrEF, or HFpEF), left-sided valvular heart disease, and congenital/acquired cardiovascular conditions leading to post-capillary PH [13, 631–635]. Arguably, PH-LHD represents the most prevalent form of PH, accounting for 65–80% of cases [47].
Consistent with the general definitions of PH, PH-LHD (group 2 PH) is defined by an mPAP >20 mmHg and a PAWP >15 mmHg. Within this haemodynamic condition of post-capillary PH, IpcPH is defined by PVR ≤2 WU and CpcPH by PVR >2 WU (Table 5). The diastolic pressure gradient (DPG) (calculated as the difference between dPAP and PAWP) is no longer used to distinguish between IpcPH and CpcPH because of conflicting data on prognostication in LHD [142].
Across the spectrum of LHD, increases in PAP and PVR are associated with an increased disease burden and a worse outcome [13, 631, 633, 635]. In a large patient cohort—predominantly with post-capillary PH—a PVR ≥2.2 WU was associated with adverse outcomes and considered abnormal [13]. However, even within this subgroup of patients with LHD and CpcPH, the risk of mortality increases with progressive elevation in PVR. In patients with advanced HFrEF and those with HFpEF or valvular heart disease, a PVR >5 WU carries additional prognostic information and is considered clinically meaningful by physicians [142, 450, 631–639]. Elevated PVR also appears to be associated with decreased survival in special situations, such as in patients undergoing interventions for correcting valvular heart disease [634], heart transplantation [142, 633], or LVAD implantation [142, 637]. Based on available data, a PVR >5 WU may indicate a severe pre-capillary component, the presence of which may prompt physicians to refer patients to PH centres for specialized care.
The prevalence of PH in patients with LHD is difficult to assess and depends on the methodology of diagnostic testing (echocardiography or invasive haemodynamics), cut-off values used to define PH, and populations studied. Observational studies suggest an estimated prevalence of PH of 40–72% in patients with HFrEF and 36–83% in those with HFpEF [48, 639–643]. When PVR is used to define a pre-capillary component in patients with HF and post-capillary PH, ∼20–30% of patients are categorized as having CpcPH [47, 644, 645]. In patients with valvular heart disease, echocardiographic studies have shown that PH is present in up to 65% of patients with symptomatic aortic stenosis [646–651], while virtually all patients with severe mitral valve stenosis develop PH [652], which can also be found in most patients with significant degenerative or functional mitral regurgitation.
The pathophysiology of PH-LHD combines several mechanisms (Figure 11): 1) an initial passive increase in LV filling pressures and backward transmission into the pulmonary circulation; 2) PA endothelial dysfunction (including vasoconstriction); 3) vascular remodelling (which may occur in both venules and/or arterioles); 4) RV dilatation/dysfunction and functional TR [653–656]; and 5) altered RV–PA coupling [655–657]. The haemodynamic profile of CpcPH versus IpcPH and elevated PVR reflects pulmonary vascular abnormalities, which contribute to an increased RV afterload. Resulting dysfunction of the RV is frequent and associated with a worse prognosis in patients with PH-LHD. In HFpEF, where RV dysfunction may occur via distinct mechanisms (Figure S1), deterioration of RV, but not LV systolic function, has been observed over time, and both prevalent and incident RV dysfunction are predictors of mortality [658].
The occurrence of PH in patients with LHD may also be due to other causes, including undetected CTEPH or PAH. Further, respiratory comorbidities such as COPD and sleep apnoea are also common in patients with LHD and may contribute to PH and impact prognosis. Patients with HFpEF and PH associated with HFpEF [75, 76] may also present with a low DLCO, which is an independent predictor of outcome [75].
8.2. Diagnosis
In patients with LHD, symptoms (e.g. exertional dyspnoea) and physical signs of PH (e.g. peripheral oedema) frequently overlap with those of the underlying left heart condition and are mostly non-specific. However, while pulmonary congestion or pleural effusion indicate LHD as the underlying cause of PH, other features may suggest the presence of relevant PH (see Section 5.1.1).
Routine diagnostic tests including BNP/NT-proBNP, ECG, and echocardiography may show signs of underlying LHD, but may also indicate PH. While BNP/NT-proBNP cannot discriminate between left- or right-sided HF, ECG findings such as right axis deviation or RV strain may suggest the presence of PH in patients with LHD. Echocardiography can diagnose HFrEF and HFpEF; identify specific cardiac conditions, including those with restrictive filling pattern; and diagnose additional valvular heart disease; it may also detect elevated sPAP and other features of PH (RA area, PA enlargement, RV/LV ratio, LV eccentricity index, RV forming the apex), leading to an echocardiographic probability of PH (see Section 5.1.5). A stepwise, composite echocardiographic score may discriminate pre- versus post-capillary PH and predict PVD in patients with LHD [659, 660]. Additional information may be gathered from further testing, including biomarkers, imaging-derived markers of RV dysfunction, and CPET-derived variables [142].
Given the complexity and variability of cardiopulmonary haemodynamics in patients with LHD, the distinction between post- and pre-capillary PH and the diagnosis of PH-LHD versus other forms of PH can be challenging. Diagnostic clues in the evaluation of suspected PH in LHD include: 1) diagnosis and control of the underlying LHD; 2) evaluation for PH and patient phenotyping; and 3) invasive haemodynamic evaluation, when indicated.
8.2.1. Diagnosis and control of the underlying left heart disease
Patients with suspected PH-LHD will have an established diagnosis of LHD, such as HFrEF/HFmrEF, HFpEF, valvular heart disease, and/or CHD. The distinction between PH associated with HFpEF and other forms of PH (e.g. PAH, CTEPH) may be challenging, particularly given the increased burden of cardiovascular comorbidities in real-world PAH populations [142, 450, 661]. In this context, validated scores for diagnosing HFpEF (HFA-PEFF, H2FPEF) [16, 662, 663] may be helpful for detecting it as an underlying condition in PH, and the presence or absence of risk factors for PAH or CTEPH should be determined. Patients with signs of predominant RV strain and/or PH should be further evaluated. Patients should be assessed or reassessed when they are fully recompensated and in a clinically stable condition.
8.2.2. Evaluation of pulmonary hypertension and patient phenotyping
Patients with LHD and suspected PH should be evaluated following the diagnostic strategy for PH (see Section 5). This requires identifying clinical features and a multimodal approach using non-invasive diagnostic tests such as echocardiography, ECG, and BNP/NT-proBNP levels. In the presence of mild PH and predominant LHD, no further testing may be necessary. Otherwise, CTEPH and significant lung disease should be ruled out by V/Q scan and PFTs, and additional cardiac imaging including cMRI may be considered in selected cases. For phenotyping, a combination of variables may help to determine the likelihood of LHD, and HFpEF in particular, versus other causes of PH (Table 23). Pulmonary hypertension associated with left heart disease is likely in the presence of known cardiac disease, multiple cardiovascular comorbidities/risk factors, atrial fibrillation at diagnosis, and specific imaging findings (LV hypertrophy, increased LA size, and reduced LA strain). Although exercise echocardiography has been proposed to uncover HFpEF, it is unable to diagnose or classify PH in this context. A combination of clinical findings and phenotyping is required to decide about the need for further invasive assessment.
8.2.3. Invasive assessment of haemodynamics
The decision to perform cardiac catheterization and to invasively assess cardiopulmonary haemodynamics should depend on the presence of an intermediate to high echocardiographic probability of PH, and should be determined by the need to obtain relevant information for prognostication or management. In patients with a high likelihood of LHD as the main cause of PH, or with established underlying LHD and mild PH (Table 23), invasive assessment for PH is usually not indicated. Indications for RHC in LHD include: (1) suspected PAH or CTEPH; (2) suspected CpcPH with a severe pre-capillary component, where further information will aid phenotyping and treatment decisions (Figure S2); and (3) advanced HF and evaluation for heart transplantation. While several haemodynamic measures (mPAP, PVR, pulmonary arterial compliance [PAC], transpulmonary pressure gradient, and DPG) are associated with outcomes in PH-LHD [142, 632, 635], the most robust and consistent data are available for PVR. Invasive assessment should be conducted in experienced centres, when management of the underlying LHD has been optimized and patients are in a clinically stable condition. With respect to respiratory variations of intrathoracic pressures, all pressure readings should be taken at end-expiration.
Additional testing during RHC may be useful for distinguishing between PAH and HFpEF [18, 23, 664–669], and to uncover LHD in patients with a high likelihood of PH-LHD and normal resting PAWP [670–673]; both exercise testing and fluid challenge may be considered in special situations (see Section 5.1.12). Conditions associated with reduced LV diastolic compliance or valvular heart disease may be associated with a rapid increase in PAWP when challenged with increased systemic venous return [674]. While the upper limit of normal remains controversial [142, 143, 665, 667], a PAWP cut-off of >18 mmHg has been suggested to identify HFpEF as the underlying cause of PH, despite normal PAWP at baseline [143]. While this may help to classify PH, therapeutic consequences of such testing remain to be determined.
As differentiating between severe PH associated with HFpEF and IPAH with cardiac comorbidities is challenging, patients with an unclear diagnosis, particularly those with a predominant pre-capillary component (e.g. PVR >5 WU), should be referred to a PH centre for individualized management.
8.3. Therapy
The primary strategy in managing PH-LHD is optimizing treatment of the underlying cardiac disease. Nevertheless, a pathophysiological sequence ranging from left-sided heart disease via pulmonary circulation to chronic right heart strain (at rest or exercise) is present in many patients [47]. Since deterioration of RV function over time is associated with poor outcomes in HFpEF [658], preserving RV function should be considered an important treatment goal. Diuretics remain the cornerstone of medical therapy in the presence of fluid retention due to PH-LHD.
There is limited and conflicting evidence for the use of drugs approved for PAH in patients with group 2 PH. Some medications may have variable and potentially detrimental effects in such patients and are therefore not indicated in PH-LHD. Management strategies for PH in various left heart aetiologies are described below.
8.3.1. Pulmonary hypertension associated with left-sided heart failure
8.3.1.1. Heart failure with reduced ejection fraction
Patients with HFrEF or HFmrEF require guideline-directed treatment including established medical and interventional therapies [27]. In patients with advanced HFrEF, implanting an LVAD may significantly reduce or even normalize mPAP [675], although this is not achieved in all patients [676], and an increased DPG emerged as a negative prognostic factor after LVAD implantation [677]. With regards to PAH drugs, bosentan was assessed in an RCT of patients with PH associated with HFrEF [678], showing no efficacy but an increase in adverse events compared with placebo, predominantly related to fluid retention. Small studies have suggested that sildenafil may improve haemodynamics and exercise capacity in PH and HFrEF [679–681], but RCTs are lacking.
8.3.1.2. Heart failure with preserved ejection fraction
In patients with HFpEF, blood pressure, volume load, and risk factors should be controlled, which may lower filling pressures and PAP [27]. Recently, the SGLT-2i empagliflozin improved outcomes in patients with an LV ejection fraction of 40–60% [682]. Endothelin receptor antagonists have not proved successful in this population, as both bosentan [683] and macitentan [684] failed to show efficacy but rather led to more adverse events (fluid retention) versus placebo in patients with HFpEF-associated PH and HF with ejection fraction >35%-associated CpcPH, respectively. Phosphodiesterase 5 inhibitors were assessed in two small RCTs in patients with HFpEF and PH with distinct haemodynamic characteristics. In patients with a predominantly IpcPH profile, sildenafil had no effect on mPAP (primary endpoint) or other haemodynamic and clinical measures versus placebo [685]. In patients with a predominantly CpcPH profile, sildenafil improved haemodynamics, RV function, and quality of life at 6 and 12 months versus placebo [686]. Furthermore, retrospective analyses and registry data suggested improvements in exercise capacity with PDE5i therapy in patients with HFpEF-associated CpcPH and with a severe pre-capillary component (PVR mostly >5 WU) [450, 687].
8.3.1.3. Interatrial shunt devices
Recent data suggest that specific interventions may be considered in selected cases of HFpEF, such as interatrial shunt devices to unload the left heart. While this was associated with short-term improvements in pulmonary vascular function [688], the long-term effect on the pulmonary circulation remains unknown. The recent REDUCE LAP-HF II trial failed to show a reduction in HF events after placement of an atrial shunt device in a population of HF patients with LVEF ≥40% [689], with worse outcomes in the presence of PVD [690]. In addition, a sustained increase in PA blood flow may be a matter of concern, as this may trigger vascular remodelling in patients with pre-existing PH.
8.3.1.4. Remote pulmonary arterial pressure monitoring in heart failure
The importance of decongestion in patients with HF is underscored by the use of implantable pressure sensors, remotely monitoring PAP as a surrogate of left-sided filling pressure. Pulmonary arterial pressure-based adjustment of HF therapy substantially reduced HF hospitalizations and improved outcomes in both patients with HFpEF and HFrEF [691–694], with adjustment of diuretic therapy being the most prominent therapeutic consequence. Further strategies to optimize management depending on the haemodynamic phenotype in PH-LHD remain to be established. In HFrEF, novel medical therapies such as ARNIs and SGLT-2is reduced remotely monitored PAP and diuretic use [695–698], potentially providing opportunities to further optimize PAP-guided HF therapy.
8.3.2. Pulmonary hypertension associated with valvular heart disease
Pulmonary hypertension frequently occurs as a consequence of valvular heart disease. While surgical or interventional approaches for valvular repair improve cardiopulmonary haemodynamics by reducing PAWP and PAP and improving forward SV [699], persistent PH after correcting valvular heart disease is frequent and associated with adverse outcomes [634, 700].
8.3.2.1. Mitral valve disease
Both mitral stenosis and regurgitation regularly lead to post-capillary PH. Functional (secondary) mitral regurgitation occurs in both HFrEF and HFpEF, and is an important contributor to PH in LHD. Reducing mitral regurgitation according to the recommendations of the 2021 ESC/EACTS Guidelines for the management of valvular heart disease [28] has a crucial role in improving haemodynamics in patients with HFrEF, as this reduces mPAP and PAWP and improves the CI [699]. Nevertheless, registry data have demonstrated that even moderately elevated sPAP negatively impacts post-procedural outcomes after catheter-based therapy [700].
8.3.2.2. Aortic stenosis
In patients with aortic stenosis undergoing surgical or catheter-based aortic valve repair, pre-interventional PH is associated with a higher risk of in-hospital adverse events and adverse long-term outcomes [646–651]. Although post-procedural improvement in PH correlates with symptom relief and favourable outcomes, persistence of PH is common, and even moderate PH is associated with a higher all-cause mortality [646–651].
Of note, medical therapy of PH post-valvular repair may be harmful. A randomized study of 231 patients with surgically corrected valvular heart disease and persistent PH showed that sildenafil therapy versus placebo was associated with worse outcome when compared with placebo [701]; however, this study did not distinguish between different types of PH (pre-capillary, IpcPH, and CpcPH).
8.3.2.3. Tricuspid regurgitation
Severe TR is associated with volume overload, increased RV workload, and maladaptive remodelling, leading to symptomatic right HF and impaired survival [702, 703]. While primary TR is relatively rare, functional TR may arise from annular dilation in the presence of both PH and LHD. Transcatheter tricuspid valve interventions have recently emerged, aiming at reducing TR and RV volume overload. Of note, correcting TR in patients with PAH or PH in (non-valvular) LHD with significantly elevated PVR and/or RV dysfunction must be considered with great caution, as this may be hazardous [704]. Right ventricle–PA coupling is an independent predictor of all-cause mortality in such patients [705]. Patient selection appears crucial, and a comprehensive diagnostic approach integrating imaging modalities and invasive haemodynamic assessment is necessary in the evaluation process prior to tricuspid valve repair, particularly since echocardiography underestimates sPAP in the presence of severe TR.
8.3.3. Recommendations on the use of drugs approved for PAH in PH-LHD
The recommendations on the use of drugs approved for PAH in patients with PH-LHD have been established based on key narrative question 5 (Supplementary Data, Section 8.3).
The recommendations on the use of PDE5is in patients with CpcPH associated with HFpEF are based on PICO question II (Supplementary Data, Section 8.4). Two RCTs that enrolled patients with HFpEF and PH were identified, but no study that specifically enrolled patients with HFpEF and CpcPH. Harmful effects cannot be excluded, even if the available data from clinical studies, case series, and registries suggest that PDE5is may be safely administered to patients with HFpEF-associated CpcPH. As a result, a general recommendation for or against the use of PDE5is in patients with HFpEF and CpcPH cannot be made. However, it is clinically relevant to make a recommendation against their use for patients with HFpEF and IpcPH.
Recommendation | Classa | Levelb |
In patients with LHD, optimizing treatment of the underlying condition is recommended before considering assessment of suspected PH [27, 28] | I | A |
RHC is recommended for suspected PH in patients with LHD, if it aids management decisions | I | C |
RHC is recommended in patients with severe tricuspid regurgitation with or without LHD prior to surgical or interventional valve repair | I | C |
For patients with LHD and suspected PH with features of a severe pre-capillary component and/or markers of RV dysfunction, referral to a PH centre for a complete diagnostic work-up is recommended [29, 47, 142] | I | C |
In patients with LHD and CpcPH with a severe pre-capillary component (e.g. PVR >5 WU), an individualized approach to treatment is recommended | I | C |
When patients with PH and multiple risk factors for LHD, who have a normal PAWP at rest but an abnormal response to exercise or fluid challenge, are treated with PAH drugs, close monitoring is recommended | I | C |
In patients with PH at RHC, a borderline PAWP (13–15 mmHg) and features of HFpEF, additional testing with exercise or fluid challenge may be considered to uncover post-capillary PH [133, 143] | IIb | C |
Drugs approved for PAH are not recommended in PH-LHDc [631, 678, 683, 684, 701, 706] | III | A |
See Recommendation Table 22B for footnotes.
Recommendations | GRADE | Classa | Levelb | |
Quality of evidence | Strength of recommendation | |||
No recommendation can be given for or against the use of PDE5is in patients with HFpEF and combined post- and pre-capillary PH | Low | None | ||
The use of PDE5is in patients with HFpEF and isolated post-capillary PH is not recommended | Low | Conditional | III | C |
CpcPH, combined post- and pre-capillary PH; ERA, endothelin receptor antagonist; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; LHD, left heart disease; PAH, pulmonary arterial hypertension; PAWP, pulmonary arterial wedge pressure; PDE5is, phosphodiesterase 5 inhibitors; PH, pulmonary hypertension; PH-LHD, pulmonary hypertension associated with left heart disease; PVR, pulmonary vascular resistance; RHC, right heart catheterization; RV, right ventricular; WU, Wood units. aClass of recommendation. bLevel of evidence. cSafety concerns have been identified when ERAs are used in patients with HF (HFpEF and HFrEF, with or without PH) and when sildenafil is used in patients with persistent PH after correction of valvular heart disease.
9. Pulmonary hypertension associated with lung diseases and/or hypoxia (group 3)
Pulmonary hypertension is frequently observed in patients with COPD and/or emphysema, ILD, combined pulmonary fibrosis and emphysema (CPFE), and hypoventilation syndromes [52, 165, 707, 708]. Pulmonary hypertension is uncommon in obstructive sleep apnoea unless other conditions coexist, such as COPD or daytime hypoventilation [709]. At high altitude (>2500 m) hypoxia-induced PH is thought to affect >5% of the population, the development of PH being related to geography and genetic factors [710].
A PH screening study performed on a large cohort of >100 patients with lymphangioleiomyomatosis confirmed that PH is usually mild in that setting: from six patients (5.7%) presenting with pre-capillary PH, none had mPAP >30 mmHg and PH was associated with PFT alteration, suggesting that the rise in mPAP is associated with parenchymal involvement [711]. Thus, PH in lymphangioleiomyomatosis is now classified in group 3 PH [1].
In patients with lung disease, PH is categorized as non-severe or severe, depending on haemodynamic findings (Figure 12). In the 2015 ESC/ERS Guidelines for the diagnosis and treatment of PH, severe PH was defined by mPAP >35 mmHg or mPAP ≥25 mmHg with CI <2.5 L/min/m2 [25, 26]. However, two recent studies have demonstrated that a PVR >5 WU is a better threshold for predicting worse prognosis in patients with PH associated with both COPD and ILD [712, 713]. Based on these data, the current guidelines used PVR to distinguish between non-severe PH (PVR ≤5 WU) and severe PH (PVR >5 WU). Whereas non-severe PH is common in advanced COPD and ILD defined by spirometric criteria, severe PH is uncommon, occurring in 1–5% of cases of COPD and <10% of patients with advanced ILD, with limited data in obesity hypoventilation syndrome [714, 715]. Even non-severe PH in lung disease negatively impacts symptoms and survival, and is associated with increased hospitalization [715–717]. Patients with lung disease and severe PH have a worse outcome than those with non-severe PH, providing evidence that this distinction has clinical significance [51, 712, 713, 718, 719]. It is noteworthy that developing severe PH is largely independent of spirometry but usually accompanied by hypoxaemia, low PaCO2, and a significant reduction in DLCO [51, 714, 718, 719].
Pulmonary hypertension presenting in patients with lung disease may be due to a number of causes, including undiagnosed CTEPH or PAH [714, 720]. Cardiac comorbidities are also common in patients with lung disease and may contribute to PH. A number of distinct phenotypes of PH in patients with lung disease, including a pulmonary vascular phenotype, have been proposed [51, 720]. The pulmonary vascular phenotype is characterized by better preserved spirometry, low DLCO, hypoxaemia, a range of parenchymal involvement on lung imaging, and a circulatory limitation to exercise [51, 714, 718–722]. Recent studies have shown that the clinical characteristics, disease trajectory, response to treatment [451, 718, 719], and histological correlates [723, 724] of patients with severe PH and minor lung disease are different to those in patients with IPAH, including a poorer prognosis.
9.1. Diagnosis
In patients with lung disease, symptoms of PH, especially exertional dyspnoea, overlap with those of the underlying condition. Physical findings may also be non-specific, for example: ankle swelling is common during episodes of ventilatory failure in COPD, where activation of the renin–angiotensin–aldosterone system may cause fluid retention, usually in the setting of preserved RV function.
Non-invasive tests—such as ECG showing right axis deviation or RV strain, elevated levels of BNP/NT-proBNP, CPET, or features on cross-sectional imaging—may suggest the diagnosis of PH in patients with lung disease [725, 726]. Echocardiography remains the most widely used non-invasive diagnostic tool for assessing PH; however, the accuracy of echocardiography in patients with advanced respiratory diseases is low, with a TRV unmeasurable in >50% of patients in some studies, and there is a tendency to overestimate PAP and misclassify patients with PH [86, 87, 727]. More recent data suggest that a stepwise, composite, echocardiographic score can identify patients with severe PH, with and without an estimate of TRV, using other echocardiographic features including RA area, RV:LV ratio, and LV eccentricity index [728]. Where PH is suspected, combining echocardiography with a contrast-enhanced CT may aid diagnostic assessment and disease classification [108, 729–731]. Pulmonary artery enlargement, RV outflow hypertrophy, and increased RV:LV ratio may suggest a diagnosis of PH [108]. Ideally, assessments should be made or repeated when the patient is clinically stable, as exacerbations can significantly raise PAP.
Key parts of evaluating suspected PH in lung disease include integrating: 1) the presence or absence of risk factors for PAH, CTEPH, or LHD; 2) clinical features, including disease trajectory (e.g. rapid recent deterioration versus gradual change over years, and oxygen requirements); 3) PFTs, including DLCO and blood gas analysis; 4) NT-proBNP measurements, ECG, and echocardiography; and 5) cross-sectional imaging with contrast-enhanced CT, SPECT, or V/Q lung scan and, in selected cases, cMRI [732] to assess the need for RHC. Cardiopulmonary exercise testing may be helpful in assessing ventilatory or cardiac limitation in patients with lung disease [121, 733], although data are limited regarding its clinical use in identifying patients with PH in lung disease.
Indications for RHC in lung disease include assessment for surgical treatments (selected patients considered for LTx and lung volume reduction surgery), suspected PAH or CTEPH, and where further information will aid phenotyping of disease and consideration of therapeutic interventions (Figure S3) [712, 718, 734]. Such testing should ideally be conducted in PH centres when patients are clinically stable and treatment of underlying lung disease has been optimized. Consideration should be given to how pressure measurements are made, due to the impact of changing intrathoracic pressures on pulmonary haemodynamics during the respiratory cycle (see Section 5.1.12) [735].
9.2. Therapy
The therapeutic approach to group 3 PH starts with optimizing the treatment of the underlying lung disease, including supplementary oxygen and non-invasive ventilation, where indicated, as well as enrolment into pulmonary rehabilitation programmes [736]. There is limited and conflicting evidence for the use of medication approved for PAH in patients with group 3 PH, and these drugs may have variable and sometimes detrimental effects on haemodynamics, exercise capacity, gas exchange, and outcomes in this patient population [181, 737–740].
9.2.1. Pulmonary hypertension associated with chronic obstructive pulmonary disease or emphysema
Studies using drugs approved for PAH in patients with PH associated with COPD or emphysema have yielded conflicting results and are mostly limited by small sample size, short duration, and insufficient haemodynamic characterization of PH [739, 741, 742]. In a 16 week RCT of 28 patients with COPD and severe PH confirmed by RHC, sildenafil therapy resulted in statistically significant improvements in PVR and quality of life [743]. Registry data identified that ∼30% of patients with COPD and severe PH, predominantly treated with PDE5is, had improved WHO-FC, 6MWD, and PVR versus baseline, and those with a treatment response had improved transplant-free survival [51, 718]. However, in the absence of large randomized trials, the evidence is insufficient to support the general use of medication approved for PAH in patients with COPD and PH. Patients with COPD and suspected or confirmed severe PH should be referred to PH centres for individual decision-making.
9.2.2. Pulmonary hypertension associated with interstitial lung disease
Numerous phase 2 and phase 3 studies have investigated the use of ERAs to treat ILD, all with negative results [740, 744, 745]. In addition, the PDE5i sildenafil has been investigated in phase 3 trials of patients with ILD, also with negative results [746, 747]. Few data from RCTs are available for patients with PH associated with ILD, and many of the studies performed for this indication [748, 749] suffered from the same limitations as the aforementioned studies in PH associated with COPD. In addition, there were several adverse safety signals: ambrisentan was associated with an increased risk of clinical worsening in patients with ILD with and without PH [740, 750], while riociguat was associated with an increased risk of clinical worsening events, including potential excess mortality, in patients with PH associated with idiopathic interstitial pneumonia [181].
In contrast, promising results have been obtained with the use of inhaled treprostinil. A phase 3 RCT (INCREASE) examined inhaled treprostinil at a target dose of 72 µg given four times daily in 326 patients with PH associated with ILD [734, 751]. The PH diagnosis was confirmed by RHC within 1 year prior to enrolment. At week 16, the placebo-corrected 6MWD improved by 31 m with inhaled treprostinil. There were also improvements in NT-proBNP and clinical worsening events, the latter driven by a lower proportion of patients whose 6MWD declined by >15% from baseline.
Given the significant impact of even non-severe PH in patients with lung disease, eligible patients should be referred for LTx evaluation. In patients with ILD and PH, inhaled treprostinil may be considered based on the findings from the INCREASE study, but further data are needed, especially on long-term outcomes. The routine use of other medication approved for PAH is not recommended in patients with ILD and non-severe PH. For patients with severe PH and/or severe RV dysfunction, or where there is uncertainty regarding the treatment of PH, referral to a PH centre is recommended for careful evaluation, to facilitate entry into RCTs, and consider PAH therapies on an individual basis (Figure S3). Registry data show that some patients with group 3 PH are being treated with PAH medication, predominantly PDE5is [718, 752, 753], but it is unclear if and to what extent these patients benefit from this treatment.
9.2.3. Recommendations on the use of drugs approved for PAH in PH associated with lung disease
The recommendations on the use of drugs approved for PAH in patients with PH associated with COPD and ILD have been established based on key narrative questions 6 and 7 (Supplementary Data, Sections 9.1 and 9.2, respectively).
The recommendations on the use of PDE5is in patients with severe PH associated with ILD are based on PICO question III (Supplementary Data, Section 9.3). There are no direct data from RCTs on the safety, tolerability, and efficacy of PDE5is in patients with PH associated with ILD. The indirect data included in the guidelines do not enable firm conclusions to be drawn. Given the lack of robust evidence, the Task Force members felt unable to provide a recommendation for or against the use of PDE5is in patients with ILD and severe PH, and recommend that these patients are referred to a PH centre for individualized decision-making.
Recommendations | Classa | Levelb |
If PH is suspected in patients with lung disease, it is recommended that echocardiographyc be performed and results interpreted in conjunction with ABG, PFTs including DLCO, and CT imaging | I | C |
In patients with lung disease and suspected PH, it is recommended to optimize treatment of the underlying lung disease and, where indicated, hypoxaemia, sleep-disordered breathing, and/or alveolar hypoventilation | I | C |
In patients with lung disease and suspected severe PH, or where there is uncertainty regarding the treatment of PH, referral to a PH centre is recommendedd | I | C |
In patients with lung disease and severe PH, an individualized approach to treatment is recommended | I | C |
It is recommended to refer eligible patients with lung disease and PH for LTx evaluation | I | C |
In patients with lung disease and suspected PH, RHC is recommended if the results are expected to aid management decisions | I | C |
Inhaled treprostinil may be considered in patients with PH associated with ILD [734] | IIb | B |
The use of ambrisentan is not recommended in patients with PH associated with IPF [740] | III | B |
The use of riociguat is not recommended in patients with PH associated with IIP [181] | III | B |
The use of PAH medication is not recommended in patients with lung disease and non-severe PHe | III | C |
See Recommendation Table 23B for footnotes.
Recommendations |