Next Article in Journal
Combined Treatment with Ultrasound and Immune Checkpoint Inhibitors for Prostate Cancer
Next Article in Special Issue
Risk Stratification of Patients with Pulmonary Arterial Hypertension: The Role of Echocardiography
Previous Article in Journal
Effect of an Electronic Alert System on Hepatitis B Virus Reactivation in Patients Receiving Immunosuppressive Drug Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exercise-Induced Pulmonary Hypertension Is Associated with High Cardiovascular Risk in Patients with HIV

1
Department of Pathology, Cardiology Division, Azienda Ospedaliera Universitaria Pisana, University of Pisa, 56124 Pisa, Italy
2
Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, 56124 Pisa, Italy
3
Section of Statistics, University Hospital of Pisa, 56124 Pisa, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(9), 2447; https://doi.org/10.3390/jcm11092447
Submission received: 25 March 2022 / Revised: 21 April 2022 / Accepted: 25 April 2022 / Published: 27 April 2022

Abstract

:
Background and Aim: Pulmonary hypertension (PH) at rest can be preceded by the onset of exercise-induced PH (ExPH). We investigated its association with the cardiovascular (CV) risk score in patients with human immunodeficiency virus (HIV). Methods: In 46 consecutive patients with HIV with low (n = 43) or intermediate (n = 3) probability of resting PH, we evaluated the CV risk score based on prognostic determinants of CV risk. Diagnosis of ExPH was made by cardiopulmonary exercise test (CPET) and exercise stress echocardiogram (ESE). Results: Twenty-eight % (n = 13) of the enrolled patients had ExPH at both CPET and ESE, with good agreement between the two methods (Cohen’s kappa = 0.678). ExPH correlated directly with a higher CV score (p < 0.001). Patients with a higher CV score also had lower CD4+ T-cell counts (p = 0.001), a faster progression to acquired immunodeficiency syndrome (p < 0.001), a poor immunological response to antiretroviral therapy (p = 0.035), higher pulmonary vascular resistance (p = 0.003) and a higher right atrial area (p = 0.006). Conclusions: Isolated ExPH is associated with a high CV risk score in patients with HIV. Assessment of ExPH may better stratify CV risk in patients with HIV.

1. Introduction

HIV infection represents a potential risk factor for pulmonary arterial hypertension (PAH) [1]. PAH is the main cause of mortality in patients with HIV [2]. Reduced pulmonary vascular reserve may be silent at rest, while it occurs with PH during exercise. The onset of exercise-induced pulmonary hypertension (ExPH), therefore, represents the first clinical sign of pulmonary vascular disease that can progress to PAH. Similar to other clinical PAH groups, a significant number of patients with HIV may have ExPH [3,4,5,6,7]. However, the prognostic value of ExPH in patients with HIV is poorly studied and Guidelines from the American College of Cardiology/American Heart Association recommend further investigation [8].
Isolated ExPH is also associated with cardiovascular (CV) events in patients with valvular or ischemic heart disease [9,10], while it is associated with clinical worsening in patients with scleroderma [11].
We have recently shown that the onset of ExPH, diagnosed by exercise stress echocardiogram (ESE), is associated with poor control of HIV infection and ExPH is associated with impaired functional capacity, as measured by the World Health Organization functional class (WHO-FC), suggesting that disease progression in the lung leads to a reduction in the vasodilatory reserve of pulmonary circulation and, consequently, PH triggered by exercise [12]. How ExPH correlates with CV risk in patients with HIV has never been previously studied. Here, we hypothesized that an increased CV risk in patients with HIV can be determined early through the work-out for ExPH by ESE and cardiopulmonary stress testing (CPET).

2. Methods

Study Design and Data Collection. We conducted a prospective, observational, cohort study of patients with HIV recruited from the Infectious Disease Clinic at Pisa University Hospital. The study complies with the Helsinki Declaration, and informed consent was obtained from all patients prior to any diagnostic test. Local investigators had full access to patient data and medical records.
All of the enrolled patients underwent evaluation of PH probability at rest by transthoracic echocardiography (TTE) [9,12], followed by ExPH assessment by transthoracic exercise stress echocardiogram (ESE) and cardiopulmonary exercise test (CPET), as detailed in the Supplementary Materials. Patients included had either a “low” PH probability at rest (n = 43) or an “intermediate” PH probability at rest (n = 3). We excluded patients with a “high” PH probability (n = 8). Patients were then classified as either with or without ExPH at ESE or CPET. We evaluated the CV risk score by assessing the presence/absence of the prognostic determinants of cardiovascular (CV) risk, according to the 2015 European Society of Cardiology (ESC)/European Respiratory Society (ERS) Guidelines [1] and, as detailed in the Supplementary Materials. We assigned a severity score of 1–3 to each prognostic determinant and derived an overall CV risk score [11]. We then evaluated the association of a higher CV score with the presence/absence of ExPH, and with several echocardiographic parameters [12] and immuno-virological parameters.
Mono and 2D Transthoracic Echocardiography was performed using a Philips iE33 echocardiograph (Philips iE33 xMATRIX echocardiography system, Andover, MA) [13]. The images were recorded in at least three cardiac cycles. Right atrial pressure (RAP) was assessed by evaluating the inferior vena cava (IVC) diameter and collapsibility during inspirium [13]. Systolic pulmonary arterial pressure (PAPs) was calculated by adding RAP to the maximum systolic pressure gradient from tricuspid regurgitation velocity (TRV). Left atrial volume index (LAVi) was calculated by Simpson’s algorithm in apical four-chamber and two-chamber view [13]. Mitral, aortic and tricuspidal regurgitations were assessed by measuring the vena contracta at apical four-chamber view.
Stress echocardiography. All patients underwent a semi-supine ESE performed with a 2.5-MHz duplex transducer and a Philips iE33 echocardiograph (Philips iE33 xMATRIX echocardiography system, Andover, MA, USA) on a semi-recumbent cycle ergometer (Ergoline, model 900 EL, Germany), according to the European Association of Echocardiography (EAE) Guidelines [13,14,15,16,17,18]. A detailed protocol of the echocardiographic procedures is reported in the Supplementary Materials.
Cardiopulmonary Exercise Test. We performed the CPET on an electronically-braked cycle ergometer, using Vmax 6200 Spectra Series software (SensorMedics, Hochberg, Germany), according to a graded, cycling workload increase protocol. The test was interrupted when one of the following symptoms or signs occurred: angina; electrocardiographic signs of myocardial ischemia or injury; an excessive blood pressure increase (systolic blood pressure ≥ 240 mmHg, diastolic blood pressure ≥ 120 mmHg); dyspnea or maximal predicted heart rate. A detailed protocol is reported in the Supplementary Materials [19,20,21].

Statistical Analyses

Categorical data were expressed by absolute and relative frequency, and continuous data by mean and standard deviation (SD). The Chi square test and the Student’s t-test for independent (two-tailed) samples were used to compare categorial and continuous variables with ExPH, respectively. Pearson’s correlation analysis was used to compare the CV score with continuous factors, while the Student’s t-test for independent samples (two-tailed) was used to compare the CV score with categorical factors. All analyses were performed with the SPSS v.26 statistical software, with statistical significance set at 0.05.

3. Results

We enrolled 54 patients from January 2020 to July 2021 in the outpatient clinic dedicated to the diagnosis and treatment of pulmonary hypertension, University Cardiology Division, University Hospital of Pisa.
All patients included in the study had no abnormalities on chest x-ray, lung function tests or electrocardiogram. We excluded eight patients with a high probability of PH on the resting echocardiogram. The remaining 46 were admitted to the study. Table A1 and Table A2 report the baseline characteristics, medical and drug histories of the entire study cohort with respect to the presence and absence of isolated ExPH at CPET and ESE, respectively. In our cohort, 72% (n = 33) of the enrolled population did not develop ExPH at ESE and 91% (n = 30) of these patients also did not develop ExPH at CPET, with a Cohen’s kappa = 0.678, indicating moderate agreement in the diagnosis of isolated ExPH between the two different methods (Figure 1). The mean age of the 46 participants included in the study was 53 ± 11. The age of those with ExPH diagnosed by CPET (Table A1) and ESE (Table A2) was 52 ± 14 years and 51 ± 13 years, respectively, while the age of those without ExPH at CPET and ESE was 54 ± 10 and 55 ± 10, respectively. We observed no statistically significant differences in patients with and without ExPH on CPET and ESE in terms of mean body surface index (BSA) age, sex, body mass index (BMI), heart rate, systolic and diastolic blood pressure, history of hypertension, comorbidities, CV risk factors and concomitant drug use, bio-humoral data and proinflammatory markers such as IL-6, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP) and fibrinogen (Table A1 and Table A2). Patients diagnosed with ExPH, both on CPET (Table A1) and ESE (Table A2), had reduced functional capacity compared to patients without ExPH (p < 0.001), while they did not show significant differences in terms of morphology and function of the right or left ventricle (Table A3 and Table A4). Patients with ExPH had higher values of sPAP (p < 0.001), TRV (p = 0.048), right atrial area (p = 0.008) and pulmonary vascular resistance (velocity ratio Peak TR and VTIRVOT > 0.20, p <0.003), compared to patients without ExPH (Table A4). We did not observe any correlation between the diagnosis of ExPH at CPET and the echocardiographic parameters associated with the pulmonary circulation and the right chambers, with the exception of sPAP (Table A3).
We evaluated the CV risk score in all 46 patients. The population’s overall CV risk score ranged from 8 (low CV risk) to 14 (high CV risk), with an average of 10.2 ± 2.4. The isolated ExPH diagnosed by both CPET and ESE was directly correlated to a higher CV score (p < 0.001) (Table 1). Patients with a higher CV score also had lower CD4+ T-cell counts (p = 0.001), a higher proportion of clinical progression to AIDS (p < 0.001) and a poor immunological response to anti-retroviral therapy (ART) (p = 0.035) (Table 2) and a higher right atrial area (p = 0.006) at rest TTE compared to patients with a lower CV risk score (Table 3). Furthermore, the proportion of patients with PVRI (ratio of Peak TR velocity to VTIRVOT) > 0.20 and higher CV score was increased compared to patients with a lower CV score (p = 0.003), indicating higher total pulmonary vascular resistance in patients with higher CV risk (Table 3). No associations with time to HIV diagnosis, time to beginning ART, ART discontinuation, virological response to ART, current use of protease inhibitors and proinflammatory markers such as IL-6, ESR, CRP and fibrinogen were found (Table 2).

4. Discussion

We investigated whether isolated ExPH is associated with a higher CV risk score in patients with HIV without a high probability of PH at resting echocardiogram. We have found that patients who develop ExPH have a higher CV risk score, as assessed by the ESC/ERS Guidelines [1]. We have found that isolated ExPH is associated with worse WHO-FC, shorter walk distance at the 6MWT and a lower VO2 peak, indicating worse functional capacity. The worst CV risk score was actually clustered into the isolated ExPH group, suggesting the usefulness of ExPH in assessing CV risk of patients with HIV. This is supported by the fact that patients with a worse CV risk score who also had isolated ExPH showed worse echo parameters related to PH and its consequences on the right cardiac chambers, as these patients had a higher total pulmonary vascular resistance and a higher right atrial area at rest TTE. Our results suggest that the work-out of ExPH using the combined ESE and CPET approach allows for the identification of those patients with HIV who develop ExPH due to reduced pulmonary vasodilatory reserve and who have a worse CV risk profile.
Unlike the previous study, where we only used the echocardiographic approach in diagnosing ExPH [12], here we used a combined CPET-ESE approach to non-invasively assess cardiovascular and pulmonary responses to exercise. Several studies have shown that in different clinical settings, CPET is more sensitive and specific than ESE in identifying pulmonary vascular abnormalities precipitated by exercise [22]. In our cohort we could not evaluate the differential performance of CPET and ESE in detecting ExPH as we did not perform right stress cardiac catheterization, which is the reference gold standard [1]. However, we have shown that there is good agreement in diagnosis between the two different methods, as demonstrated by the high Cohen’s kappa index. However, the benefit of performing a combined CPET-ESE is the identification of concomitant left heart disease as the cause of exertional dyspnea and ExPH. Increased left ventricle filling pressure is known to lead to pulmonary venous congestion and post-capillary pulmonary hypertension, regardless of LVEF [23,24]. In our cohort, TEE at rest and ESE ruled out the existence of left heart disease as a possible cause of ExPH, regardless of the method used for diagnosis.
We have recently shown that the onset of ExPH, as diagnosed by ESE, is associated with poor immunological control of HIV infection [11]. Here we have found that those patients who have worse immunological control of the disease also have a higher CV risk score. Finally, patients with a higher CV risk score also had greater clinical progression to AIDS and poorer immunological response to ART. This confirms previous studies demonstrating the role of a weakened or dysfunctional immune system in determining CV risk and prognosis of HIV-related PAH [2,25,26]. Thus, isolated ExPH can identify patients with HIV at higher CV risk as a consequence of poorer immune control of the disease. PAH often complicates HIV infection and this leads to the increased mortality of these patients. We are now demonstrating that the development of isolated ExPH in such patients without a high PH probability at rest can be considered an early marker of a worsening outcome.
HIV proteins can trigger an inflammatory response, leading to PAH [27]. Patients with HIV have elevated levels of interleukin 6 (IL-6), tumor necrosis factor α (TNFα) and nitric oxide synthase (NO) inhibitors [28,29]. The inflammation theory of PAH is biologically and clinically plausible, as PAH continues to manifest significantly in patients with HIV as an expression of the persistence of this inflammatory component, despite a good response to ART [30]. However, so far there are no clinical studies that correlate high levels of these proinflammatory mediators with the development of PAH in patients with HIV. An exploratory Phase 1 clinical study, the first of its kind to our knowledge, is still ongoing to test the effects of a combination of the HIV protease inhibitors saquinavir and ritonavir on proinflammatory mediators and pulmonary hemodynamics in patients with idiopathic PAH (ClinicalTrials.gov identifier: NCT02023450). Therefore, the blood levels of these mediators may currently be useful biomarkers in the stratification of patients with HIV with a higher atherosclerotic CV risk, but not those who have a propensity to develop HIV-related PAH. On the other hand, these biomarkers are not included in any of the validated risk scores in patients with PAH group 1, including patients with HIV. In our cohort, we did not observe any significant association between levels of IL-6 or other proinflammatory markers and ExPH or a worse CV risk score. However, only a few patients had such biomarkers evaluated. Further investigation with more patients is needed to establish the role of proinflammatory biomarkers in the development of PAH and a worse CV risk score.

Study Limitations

This study has several limitations: (1) the small sample size, for which our report should be intended as a pilot study on the role of ExPH in risk stratification of patients with HIV; (2) patients with ExPH at ESE and CPET did not undergo cardiac catheterization, which is, however, not indicated, and therefore unethical in such patients with low and intermediate probability of PH [1]. Finally, adequate follow-up could allow us to verify if patients with ExPH develop PH at rest, and if this is associated with a worsening of CV risk over time. Our research group is carrying out a long-term follow-up, which will help to clarify the prognostic significance of ExPH in the HIV population.
In conclusion: Isolated ExPH associates with a higher CV risk score in patients with HIV. Assessment of ExPH by CPET or ESE can contribute to the risk stratification of patients with HIV.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm11092447/s1, online text: methods.

Author Contributions

Conceptualization, R.M. (Rosalinda Madonna); methodology, R.M. (Rosalinda Madonna); software, R.M. (Riccardo Morganti); validation, RDC, F.M. (Francesco Menichetti) and R.I.; formal analysis, R.M. (Riccardo Morganti); investigation, R.M. (Rosalinda Madonna), F.B., S.F., L.R. and A.F.; resources, R.D.C.; data curation, R.M. (Rosalinda Madonna) and S.F.; writing—original draft preparation, R.M. (Rosalinda Madonna); writing—review and editing, R.D.C. and S.F.; visualization, R.M. (Rosalinda Madonna); project administration, R.D.C. and F.M.; funding acquisition, R.M. (Rosalinda Madonna). All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by from Ministero dell’Istruzione, Università e Ricerca Scientifica, grant number 549901_2020_Madonna:Ateneo. The APC was funded by 549901_2020_Madonna:Ateneo.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by Comitato Etico di Area Vasta Nord Ovest (CEAVNO) (protocol code 21315_ DE_CATERINA, date of approval 15 March 2022).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study. Written informed consent has been obtained from the patient(s) to publish this paper.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Demographics, medical history and medication use according to presence and absence of ExPH at CPET.
Table A1. Demographics, medical history and medication use according to presence and absence of ExPH at CPET.
Clinical ParameterExPH at CPETMeanSDp-Value
Ageno54.219.760.615
yes52.3813.86
BSAno1.910.200.475
yes1.860.24
SAPno126.3612.200.652
yes124.6210.50
DAPno74.247.080.745
yes75.007.07
ExPH at CPET
noyes
SexF1150.742
M228
Smokeno1630.264
yes169
Functional class FC-WHOI323<0.001
II10
III010
Chronic liver diseaseno1680.425
yes175
Kidney diseaseno29120.664
yes41
Thyroid diseaseno29110.767
yes42
Arterial hypertensionno29130.189
yes40
Dyslipidemiano1060.309
yes237
Diabetes mellitusno29110.767
yes42
Familiarity for CADno26100.891
yes73
Calcium channel blockersno29120.206
yes40
Hypoglycemic drugsno1060.309
yes237
Beta blockersno32130.526
yes10
ACE inhibitors or sartanicsno32130.526
yes10
Thyroid hormonesno29110.767
yes42
Lipid lowering drugsno1060.309
yes237
Abbreviations: ExPH, isolated exercise pulmonary hypertension; CPET, cardiopulmonary exercise test; BSA, body surface area; SAP, systolic arterial pressure; DAP, diastolic arterial pressure; CAD, coronary artery disease; F, female; M, male; ACE, angiotensin converting enzyme.
Table A2. Demographics, medical history and medication use according to presence and absence of ExPH at ESE.
Table A2. Demographics, medical history and medication use according to presence and absence of ExPH at ESE.
FactorExPH at ESEMeanSDp-Value
Ageno54.589.940.391
yes51.4613.33
BSAno1.900.200.932
yes1.890.25
Heart rateno69.036.300.839
yes68.625.98
yes126.929.47
DAPno73.797.180.308
yes76.156.50
ExPH at ESE
Factor noyesp-value
SexF1240.720
M219
Smokeno1540.570
ex11
yes178
Functional class FC-WHOI314<0.001
II10
III19
Chronic liver diseaseno1590.146
yes184
Kidney diseaseno29120.664
yes41
Thyroid diseaseno28120.499
yes51
Arterial hypertensionno30120.880
yes31
Dyslipidemiano970.088
yes246
Diabetes mellitusno28120.499
yes51
Familiarity for CADno2790.351
yes64
Calcium channel blockersno30110.937
yes31
Hypoglycemic drugsno970.088
yes246
Beta blockersno33120.107
yes01
ACE inhibitors or sartanicsno32130.526
yes10
Thyroid hormonesno28120.499
yes51
Lipid lowering drugsno970.088
yes246
Abbreviations: ExPH, isolated exercise pulmonary hypertension; ESE, exercise stress echocardiography; BSA, body surface area; F, female; M, male; CAD, coronary artery disease; ACE, angiotensin converting enzyme.
Table A3. ExPH at CPET vs. continuous or categorical ultrasound parameters.
Table A3. ExPH at CPET vs. continuous or categorical ultrasound parameters.
Continuous Ultrasound ParametersExPH at CPETMeanSDp-Value
LVEDDno43.6565.5220.959
yes43.5585.978
LVESDno26.6566.4390.442
yes24.9177.090
LVEDVno103.87531.2700.299
yes93.00028.451
iLVEDVno36.67610.5970.163
yes31.55910.808
LVESVno38.21914.0410.314
yes33.16716.219
iLVESVno13.4164.9440.384
yes11.9235.184
LVno152.88448.2810.443
yes140.81738.913
iLVmassno79.13229.5200.255
yes68.11024.111
LADno48.3598.5710.919
yes48.0835.744
LAVno38.77820.3680.294
yes32.3177.786
iLAVno13.6837.6570.468
yes12.0003.149
LVEFno65.1476.4060.584
yes66.5009.200
FwSVno72.36622.1970.268
yes63.75024.000
iFwSVno25.3677.8100.195
yes21.7458.944
MRno0.6880.7800.027
yes0.2500.452
AOno0.2500.6721.000
yes0.2500.622
E waveno69.76917.9890.993
yes69.81714.658
A waveno71.39730.3840.783
yes74.16726.889
Septal e waveno9.9593.0030.837
yes9.7671.837
E/Ano1.0670.3940.909
yes1.0850.629
E/eno6.4583.4080.299
yes7.5752.193
iRVESAno4.0551.8790.396
yes4.5781.542
iRVEDAno6.9552.1430.991
yes6.9472.343
iRVESVno7.1614.9070.908
yes7.3382.902
iRVEDVno16.0486.5500.220
yes13.5074.273
RD1no36.1287.2700.822
yes36.6676.372
RD2no33.4725.4830.116
yes30.3336.513
RD3no20.5036.4850.041
yes16.0835.351
RVOT prox.no31.7166.0570.627
yes30.7923.905
RVOT dist.no32.3537.4710.166
yes29.0425.163
eccentricity indexno0.9410.1100.475
yes0.9140.103
RV/LV basal diameter rationo0.8640.1620.317
yes0.8080.162
TAPSEno21.9137.1040.707
yes22.8427.665
FACno45.0539.6070.210
yes49.0838.554
RV E/Ano4.33813.3180.389
yes0.9630.408
RV E/eno3.6731.8290.683
yes3.9381.891
RV S velno10.9934.4850.630
yes11.7675.301
RV S VTIno3.6084.0500.024
yes1.8100.926
TRno1.2190.4910.862
yes1.2500.622
sPAPno22.3755.7520.749
yes21.7505.675
sPAP at exercise peakno32.84710.6020.028
yes40.7679.452
mPAPno16.3333.6230.969
yes16.3854.636
TRVno2.0090.3730.622
yes2.0750.449
TRV at exercise peakno2.5440.5550.557
yes2.6580.616
RV outflow ATno102.71442.7170.014
yes138.16734.821
VCI diameterno15.0163.7770.571
yes14.1585.896
RA areano14.7093.2270.295
yes15.8753.294
iRAVno5.3562.1910.410
yes5.9241.420
VTI at rvotno15.8433.811<0.001
yes11.1672.916
VRT/VTI at rvotno0.1560.1420.414
yes0.1910.051
TAPSE/PAPsno1.0330.3470.609
yes1.1000.471
Categorical ultrasound parametersExPH at ESEnoyesp-value
Concentric remodelingno1950.293
yes137
Normal geometryno1870.901
yes145
Concentric hypertrophyno29120.272
yes30
Eccentric hypertrophyno31120.536
yes10
Abbreviations: ExPH, isolated exercise pulmonary hypertension; CPET, cardiopulmonary exercise test; LVEDD, left ventricle end-diastolic diameter; LVESD, left ventricle end-systolic diameter; LVEDV, left ventricle end-diastolic volume; LVESV, left ventricle end-systolic volume; iLVEDV, indexed LVEDV; iLVESV, indexed LVESV; LAD, left atrial diameter; LAV, left atrial volume; iLAV, indexed LAV; LVEF, left ventricle ejection fraction; FwSV, forward stroke volume; iFwSV, indexed FwSV; iRVESA, indexed right ventricle end-systolic area; iRVEDA, indexed right ventricle end-diastolic area; iRVESV, indexed right ventricle end-systolic volume; iRVEDV, indexed right ventricle end-diastolic volume; RD, right diameter; RVOT, right ventricle outflow; TAPSE, tricuspid annular plane excursion; FAC, fractional area change; VTI, velocity time integral; sPAP, systolic pulmonary arterial pressure; mPAP, mean PAP; TR, tricuspid regurgitation; IVC, inferior vena cava, iRAV, indexed right atrial volume; TRV, tricuspid regurgitation velocity; ESE, exercise stress echocardiogram.
Table A4. ExPH at ESE vs. continuous or categorical ultrasound parameters.
Table A4. ExPH at ESE vs. continuous or categorical ultrasound parameters.
Continuous Ultrasound ParametersExPH at ESEMeanSDp-Value
LVEDDno43.4595.3220.745
yes44.0836.445
LVESDno26.6256.4600.472
yes25.0007.058
LVEDVno100.93831.4600.992
yes100.83329.489
iLVEDVno35.85710.9550.568
yes33.74310.596
LVESVno36.40614.5550.752
yes38.00015.486
iLVESVno12.9925.2050.971
yes13.0544.604
LVno149.45344.3580.974
yes149.96751.515
iLV massno77.54428.0610.593
yes72.34329.882
LADno48.5318.4810.737
yes47.6256.057
LAVno38.84419.9550.276
yes32.14210.252
iLAVno13.8957.5070.290
yes11.4523.692
LVEFno66.4597.4370.157
yes63.0006.030
FwSVno73.02224.8490.155
yes62.00013.671
iFwSVno25.6638.9520.090
yes20.9574.347
MRno0.6560.7870.193
yes0.3330.492
AOno0.3130.7380.304
yes0.0830.289
E waveno68.08418.6060.284
yes74.30810.979
A waveno71.40030.3670.784
yes74.15826.943
Septal e waveno10.1282.9580.384
yes9.3171.908
E/Ano1.0370.3970.415
yes1.1660.613
E/eno6.2513.4000.077
yes8.1291.778
iRVESAno4.0831.8620.492
yes4.5051.622
iRVEDAno7.0292.1130.708
yes6.7492.405
iRVESVno7.1834.8860.950
yes7.2783.001
iRVEDVno16.2176.4610.126
yes13.0584.326
RD1no35.9876.9200.660
yes37.0427.344
RD2no33.1885.5380.297
yes31.0926.712
RD3no20.0316.6990.222
yes17.3425.520
RVOT prox.no31.1195.7820.505
yes32.3834.882
RVOT dist.no31.6567.3050.754
yes30.9006.468
Eccentricity indexno0.9430.1120.335
yes0.9080.096
RV/LV basal diameter rationo0.8520.1570.815
yes0.8390.179
TAPSEno21.7076.9490.495
yes23.3927.958
FACno45.3979.3760.391
yes48.1679.609
RV E/Ano4.33313.3190.391
yes0.9760.431
RV E/eno3.7291.8820.946
yes3.7741.739
RV S velno10.9504.4970.561
yes11.8835.251
RV S VTIno3.1743.3280.867
yes2.9684.289
TRno1.3130.4710.077
yes1.0000.603
sPAPno22.3446.0030.794
yes21.8334.896
sPAP at exercise peakno30.9098.846<0.001
yes45.9337.504
mPAPno16.3513.8020.991
yes16.3374.212
TRVno2.0090.3890.622
yes2.0750.409
TRV at exercise peakno2.4720.5020.048
yes2.8500.659
RV outflow ATno105.02743.9900.065
yes132.00036.352
VCI diameterno14.7224.5340.885
yes14.9424.191
RA areano14.2533.1270.008
yes17.0922.710
iRAVno5.2662.1860.189
yes6.1661.300
VTI at rvotno16.0653.552<0.001
yes10.5752.703
VRT/VTI at rvotno0.1530.1430.289
yes0.1980.041
TAPSE/PAPsno1.0240.3210.448
yes1.1230.516
Categorical ultrasound parameterExPH at ESEnoyesp-value
Concentric remodelingno1950.293
yes137
Normal geometryno1870.901
yes145
Concentric hypertrophyno29120.272
yes30
Eccentric hypertrophyno31120.536
yes10
Abbreviations: ExPH, isolated exercise pulmonary hypertension; ESE, exercise stress echocardiogram; LVEDD, left ventricle end-diastolic diameter; LVESD, left ventricle end-systolic diameter; LVEDV, left ventricle end-diastolic volume; LVESV, left ventricle end-systolic volume; iLVEDV, indexed LVEDV; iLVESV, indexed LVESV; LAD, left atrial diameter; LAV, left atrial volume; iLAV, indexed LAV; LVEF, left ventricle ejection fraction; FwSV, forward stroke volume; iFwSV, indexed FwSV; iRVESA, indexed right ventricle end-systolic area; iRVEDA, indexed right ventricle end-diastolic area; iRVESV, indexed right ventricle end-systolic volume; iRVEDV, indexed right ventricle end-diastolic volume; RD, right diameter; RVOT, right ventricle outflow; TAPSE, tricuspid annular plane excursion; FAC, fractional area change; VTI, velocity time integral; sPAP, systolic pulmonary arterial pressure; mPAP, mean PAP; TR, tricuspid regurgitation; IVC, inferior vena cava, iRAV, indexed right atrial volume; TRV, tricuspid regurgitation velocity.

References

  1. Galie, N.; Humbert, M.; Vachiery, J.L.; Gibbs, S.; Lang, I.; Torbicki, A.; Simonneau, G.; Peacock, A.; Vonk Noordegraaf, A.; Beghetti, M.; et al. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur. Heart J. 2016, 37, 67–119. [Google Scholar] [PubMed]
  2. Nunes, H.; Humbert, M.; Sitbon, O.; Morse, J.H.; Deng, Z.; Knowles, J.A.; Le Gall, C.; Parent, F.; Garcia, G.; Hervé, P.; et al. Prognostic factors for survival in human immunodeficiency virus-associated pulmonary arterial hypertension. Am. J. Respir. Crit. Care Med. 2003, 167, 1433–1439. [Google Scholar] [CrossRef] [PubMed]
  3. Tolle, J.J.; Waxman, A.B.; Van Horn, T.L.; Pappagianopoulos, P.P.; Systrom, D.M. Exercise-induced pulmonary arterial hypertension. Circulation 2008, 118, 2183–2189. [Google Scholar] [CrossRef] [PubMed]
  4. Doukky, R.; Lee, W.Y.; Ravilla, M.; Lateef, O.B.; Pelaez, V.; French, A.; Tandon, R. A novel expression of exercise induced pulmonary hypertension in human immunodeficiency virus patients: A pilot study. Open Cardiovasc. Med. J. 2012, 6, 44–49. [Google Scholar] [CrossRef]
  5. Alkotob, M.L.; Soltani, P.; Sheatt, M.A.; Katsetos, M.C.; Rothfield, N.; Hager, W.D.; Foley, R.J.; Silverman, D.I. Reduced exercise capacity and stress-induced pulmonary hypertension in patients with scleroderma. Chest 2006, 130, 176–181. [Google Scholar] [CrossRef]
  6. Saggar, R.; Sitbon, O. Hemodynamics in pulmonary arterial hypertension: Current and future perspectives. Am. J. Cardio. 2012, 110, S9–S15. [Google Scholar] [CrossRef]
  7. Lewis, G.D.; Bossone, E.; Naeije, R.; Grünig, E.; Saggar, R.; Lancellotti, P.; Ghio, S.; Varga, J.; Rajagopalan, S.; Oudiz, R.; et al. Pulmonary vascular hemodynamic response to exercise in cardiopulmonary diseases. Circulation 2013, 128, 1470–1479. [Google Scholar] [CrossRef]
  8. McLaughlin, V.V.; Archer, S.L.; Badesch, D.B.; Barst, R.J.; Farber, H.W.; Lindner, J.R.; Mathier, M.A.; McGoon, M.D.; Park, M.H.; Rosenson, R.S.; et al. ACCF/AHA 2009 expert consensus document on pulmonary hypertension: A report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents and the American Heart Association: Developed in collaboration with the American College of Chest Physicians, American Thoracic Society, Inc., and the Pulmonary Hypertension Association. Circulation 2009, 119, 2250–2294. [Google Scholar]
  9. Madonna, R.; Bonitatibus, G.; Vitulli, P.; Pierdomenico, S.D.; Galie, N.; De Caterina, R. Association of the European Society of Cardiology echocardiographic probability grading for pulmonary hypertension with short and mid-term clinical outcomes after heart valve surgery. Vasc. Pharm. 2020, 125–126, 106648. [Google Scholar] [CrossRef]
  10. Borlaug, B.A.; Nishimura, R.A.; Sorajja, P.; Lam, C.S.; Redfield, M.M. Exercise hemodynamics enhance diagnosis of early heart failure with preserved ejection fraction. Circ. Heart Fail. 2010, 3, 588–595. [Google Scholar] [CrossRef]
  11. Madonna, R.; Morganti, R.; Radico, F.; Vitulli, P.; Mascellanti, M.; Amerio, P.; De Caterina, R. Isolated Exercise-Induced Pulmonary Hypertension Associates with Higher Cardiovascular Risk in Scleroderma Patients. J. Clin. Med. 2020, 9, 1910. [Google Scholar] [CrossRef] [PubMed]
  12. Madonna, R.; Fabiani, S.; Morganti, R.; Forniti, A.; Mazzola, M.; Menichetti, F.; De Caterina, R. Exercise-induced pulmonary hypertension in HIV patients: Association with poor clinical and immunological status. Vasc. Pharmacol. 2021, 139, 106888. [Google Scholar] [CrossRef] [PubMed]
  13. Sicari, R.; Nihoyannopoulos, P.; Evangelista, A.; Kasprzak, J.; Lancellotti, P.; Poldermans, D.; Voigt, J.U.; Zamorano, J.L. Stress echocardiography expert consensus statement: European Association of Echocardiography (EAE) (a registered branch of the ESC). Eur. J. Echocardiogr. 2008, 9, 415–437. [Google Scholar] [CrossRef] [PubMed]
  14. Nagel, C.; Henn, P.; Ehlken, N.; D’Andrea, A.; Blank, N.; Bossone, E.; Böttger, A.; Fiehn, C.; Fischer, C.; Lorenz, H.M.; et al. Stress Doppler echocardiography for early detection of systemic sclerosis-associated pulmonary arterial hypertension. Arthritis Res. Ther. 2015, 17, 165. [Google Scholar] [CrossRef]
  15. Argiento, P.; Chesler, N.; Mule, M.; D’Alto, M.; Bossone, E.; Unger, P.; Naeije, R. Exercise stress echocardiography for the study of the pulmonary circulation. Eur. Respir. J. 2010, 35, 1273–1278. [Google Scholar] [CrossRef]
  16. Rudski, L.G.; Lai, W.W.; Afilalo, J.; Hua, L.; Handschumacher, M.D.; Chandrasekaran, K.; Solomon, S.D.; Louie, E.K.; Schiller, N.B. Guidelines for the echocardiographic assessment of the right heart in adults: A report from the American Society of Echocardiography endorsed by the European Association of Echocardiography, a registered branch of the European Society of Cardiology, and the Canadian Society of Echocardiography. J. Am. Soc. Echocardiogr. 2010, 23, 685–713, quiz 786–688. [Google Scholar]
  17. Baumgartner, H.; Hung, J.; Bermejo, J.; Chambers, J.B.; Evangelista, A.; Griffin, B.P.; Iung, B.; Otto, C.M.; Pellikka, P.A.; Quiñones, M. Echocardiographic assessment of valve stenosis: EAE/ASE recommendations for clinical practice. J. Am. Soc. Echocardiogr. 2009, 22, 1–23, quiz 101–102. [Google Scholar] [CrossRef]
  18. Cagnina, A.; Michaux, S.; Ancion, A.; Cardos, B.; Damas, F.; Lancellotti, P. European guidelines for the diagnosis and management of acute pulmonary embolism. Rev. Med. Liege 2021, 76, 208–215. [Google Scholar]
  19. Belardine, R., III. Il Test da Sforzo Cardiopolmonare; Midia: Monza, Italy, 2006. [Google Scholar]
  20. Wassermann, K. Cardiopulmonary Exercise Testing and Cardiovascular Health; Wiley-Blackwell: Hoboken, NJ, USA, 2002. [Google Scholar]
  21. Badesch, D.B.; Champion, H.C.; Gomez Sanchez, M.A.; Hoeper, M.M.; Loyd, J.E.; Manes, A.; McGoon, M.; Naeije, R.; Olschewski, H.; Oudiz, R.J.; et al. Diagnosis and assessment of pulmonary arterial hypertension. J. Am. Coll. Cardiol. 2009, 54, S55–S66. [Google Scholar] [CrossRef]
  22. Sciume, M.; Mattiello, V.; Cattaneo, D.; Bucelli, C.; Orofino, N.; Gandolfi, L.; Pettine, L.; Lonati, S.; Gianelli, U.; Pierini, A.; et al. Early detection of pulmonary hypertension in primary myelofibrosis: The role of echocardiography, cardiopulmonary exercise testing, and biomarkers. Am. J. Hematol. 2017, 92, E47–E48. [Google Scholar] [CrossRef]
  23. Guazzi, M. Pulmonary hypertension in heart failure preserved ejection fraction: Prevalence, pathophysiology, and clinical perspectives. Circ. Heart Fail. 2014, 7, 367–377. [Google Scholar] [CrossRef] [PubMed]
  24. Kessler, K.M.; Willens, H.J.; Mallon, S.M. Diastolic left ventricular dysfunction leading to severe reversible pulmonary hypertension. Am. Heart J. 1993, 126, 234–235. [Google Scholar] [CrossRef]
  25. Quezada, M.; Martin-Carbonero, L.; Soriano, V.; Vispo, E.; Valencia, E.; Moreno, V.; de Isla, L.P.; Lennie, V.; Almería, C.; Zamorano, J.L. Prevalence and risk factors associated with pulmonary hypertension in HIV-infected patients on regular follow-up. Aids 2012, 26, 1387–1392. [Google Scholar] [CrossRef]
  26. Degano, B.; Guillaume, M.; Savale, L.; Montani, D.; Jaïs, X.; Yaici, A.; Le Pavec, J.; Humbert, M.; Simonneau, G.; Sitbon, O. HIV-associated pulmonary arterial hypertension: Survival and prognostic factors in the modern therapeutic era. Aids 2010, 24, 67–75. [Google Scholar] [CrossRef] [PubMed]
  27. Feinstein, M.J.; Hsue, P.Y.; Benjamin, L.A.; Bloomfield, G.S.; Currier, J.S.; Freiberg, M.S.; Grinspoon, S.K.; Levin, J.; Longenecker, C.T.; Post, W.S.; et al. Characteristics, Prevention, and Management of Cardiovascular Disease in People Living With HIV: A Scientific Statement From the American Heart Association. Circulation 2019, 140, e98–e124. [Google Scholar] [CrossRef]
  28. Pellicelli, A.M.; Palmieri, F.; D’Ambrosio, C.; Rianda, A.; Boumis, E.; Girardi, E.; Antonucci, G.; D’Amato, C.; Borgia, M.C. Role of human immunodeficiency virus in primary pulmonary hypertension—Case reports. Angiology 1998, 49, 1005–1011. [Google Scholar] [CrossRef]
  29. Parikh, R.V.; Scherzer, R.; Nitta, E.M.; Leone, A.; Hur, S.; Mistry, V.; Macgregor, J.S.; Martin, J.N.; Deeks, S.G.; Ganz, P.; et al. Increased levels of asymmetric dimethylarginine are associated with pulmonary arterial hypertension in HIV infection. Aids 2014, 28, 511–519. [Google Scholar] [CrossRef]
  30. Tcherakian, C.; Couderc, L.J.; Humbert, M.; Godot, V.; Sitbon, O.; Devillier, P. Inflammatory mechanisms in HIV-associated pulmonary arterial hypertension. Semin. Respir. Crit. Care Med. 2013, 34, 645–653. [Google Scholar]
Figure 1. Concordance of exercise stress echocardiography and the cardiopulmonary exercise test in the diagnosis of isolated exercise pulmonary hypertension. Abbreviations: ESE, exercise stress echocardiogram; ExPH, exercise-induced pulmonary hypertension; CPET, cardiopulmonary exercise test.
Figure 1. Concordance of exercise stress echocardiography and the cardiopulmonary exercise test in the diagnosis of isolated exercise pulmonary hypertension. Abbreviations: ESE, exercise stress echocardiogram; ExPH, exercise-induced pulmonary hypertension; CPET, cardiopulmonary exercise test.
Jcm 11 02447 g001
Table 1. Comparison between score and ExPH.
Table 1. Comparison between score and ExPH.
CV Scorep-Value
ExPH at CPET <0.001
no8.8 (0.7)
yes12.9 (2.1)
ExPH at ESE <0.001
no8.9 (1.1)
yes12.5 (2.4)
CV score values are shown as mean (SD). Abbreviations: ExPH, isolated exercise pulmonary hypertension; CPET, cardiopulmonary exercise test; ESE, exercise stress echocardiogram.
Table 2. Comparison between score and virological and immunological factors.
Table 2. Comparison between score and virological and immunological factors.
Statisticsp-Value
Time to HIV diagnosis (y)−0.1550.309
CD4+ T-cell count at diagnosis (cell/mmc)−0.4020.012
CD4+ T-cell count at diagnosis (%)−0.3800.020
CD4+ T-cell count last determination (cell/mmc)−0.3860.009
CD4+ T-cell count last determination (%)−0.4800.001
Clinical progression to AIDS <0.001
no8.8 (1.1)
yes12.9 (2)
Development of resistance to ART 0.451
no9.8 (2.1)
yes10.4 (2.7)
HIV-RNA levels at diagnosis (cp/mL)−0.1160.502
HIV-RNA levels last determination (cp/mL)0.0100.949
HIV-RNA levels last determination (cp/mL) 0.949
<20 cp/mL9.9 (2.3)
>20 cp/mL10 (2.3)
Time to beginning of ART−0.1820.226
Current use of protease inhibitors 0.101
no9.8 (2.2)
yes11.5 (2.7)
Combination of ART with booster (ritonavir or cobicistat) 0.827
no10.1 (2.4)
yes9.9 (2.4)
Virologic response to ART 0.690
<20 copies/mL9.7 (2.2)
20–50 copies/mL10.1 (2.3)
50–200 copies/mL11.5 (3.5)
>200 copies/mL10.7 (2.9)
Immunologic response to ART 0.035 *
optimal9.3 (1.8)
acceptable10.6 (2.6)
not acceptable12.3 (2.9)
ART discontinuation 0.536
no10.2 (2.4)
yes9.7 (2.3)
IL-60.2890.152
PCR−0.2470.224
VES0.1940.354
FBG−0.0530.789
Statistics: Pearson’s r or mean (SD). Abbreviations: HIV, Human immunodeficiency virus; ART, antiretroviral therapy; IL-6, interleukin-6; PCR, reactive C protein; VES, Erythrocyte sedimentation rate; FBG, fibrinogen. * only the comparison between “optimal” and “not acceptable” was statistically significant with a p = 0.040, by Dunnett correction using “not acceptable” as the reference category.
Table 3. Comparison between score and ultrasound parameters.
Table 3. Comparison between score and ultrasound parameters.
ParameterStatisticsp-Value
Concentric remodeling 0.557
no9.9 (2.2)
yes10.3 (2.6)
Normal geometry 0.755
no10.1 (2.4)
yes9.9 (2.3)
Concentric hypertrophy 0.014
no10.2 (2.4)
yes8.7 (0.6)
Eccentric hypertrophy 0.383
no10 (2.3)
yes11.5 (3.5)
LAD−0.0550.718
LAV−0.0760.620
iLAV−0.0670.667
LVEF−0.1640.281
FwSV−0.0970.527
iFwSV−0.1120.465
MR−0.1020.503
AO−0.1460.339
MS−0.0690.651
AS−0.0690.651
E wave0.1160.448
A wave0.1530.315
Septal e wave0.0250.871
E/A−0.1070.485
E/e0.1780.242
iRVESA0.1330.385
iRVEDA0.0690.652
iRVESV0.1240.419
iRVEDV−0.1330.385
RD10.0750.626
RD2−0.2150.157
RD3−0.2750.068
RVOT prox.−0.0810.596
RVOT dist.0.0710.644
Eccentricity index−0.2070.171
RV/LV basal diameter ratio−0.1920.205
TAPSE0.0880.564
FAC0.1790.240
RV E/A−0.1400.358
RV E/e−0.0260.869
RV S vel0.1510.322
RV S VTI−0.2450.104
TR−0.1270.407
sPAP−0.0180.906
aPAP at exercise peak0.2690.074
mPAP0.0360.814
TRV0.1150.454
TRV at exercise peak0.0320.834
RV outflow AT0.2070.173
VCI diameter−0.0480.753
RA area0.4010.006
iRAV0.2730.070
VTI at rvot−0.5830.000
VRT/VTI at rvot0.1970.196
TAPSE/PAPs0.1010.508
PVRI 0.003
<0.208.88 (1.23)
>0.2012.10 (2.55)
Statistics: Pearson’s r or mean (SD). Abbreviations: LVEDD, left ventricle end-diastolic diameter; LVESD, left ventricle end-systolic diameter; LVEDV, left ventricle end-diastolic volume; LVESV, left ventricle end-systolic volume; iLVEDV, indexed LVEDV; iLVESV, indexed LVESV; LAD, left atrial diameter; LAV, left atrial volume; iLAV, indexed LAV; LVEF, left ventricle ejection fraction; FwSV, forward stroke volume; iFwSV, indexed FwSV; iRVESA, indexed right ventricle end-systolic area; iRVEDA, indexed right ventricle end-diastolic area; iRVESV, indexed right ventricle end-systolic volume; iRVEDV, indexed right ventricle end-diastolic volume; RD, right diameter; RVOT, right ventricle outflow; TAPSE, tricuspid annular plane excursion; FAC, fractional area change; VTI, velocity time integral; sPAP, systolic pulmonary arterial pressure; mPAP, mean PAP; TR, tricuspid regurgitation; IVC, inferior vena cava; iRAV, indexed right atrial volume; TRV, tricuspid regurgitation velocity; PVRI, Pulmonary Vascular Resistance Index.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Madonna, R.; Fabiani, S.; Morganti, R.; Forniti, A.; Biondi, F.; Ridolfi, L.; Iapoce, R.; Menichetti, F.; De Caterina, R. Exercise-Induced Pulmonary Hypertension Is Associated with High Cardiovascular Risk in Patients with HIV. J. Clin. Med. 2022, 11, 2447. https://doi.org/10.3390/jcm11092447

AMA Style

Madonna R, Fabiani S, Morganti R, Forniti A, Biondi F, Ridolfi L, Iapoce R, Menichetti F, De Caterina R. Exercise-Induced Pulmonary Hypertension Is Associated with High Cardiovascular Risk in Patients with HIV. Journal of Clinical Medicine. 2022; 11(9):2447. https://doi.org/10.3390/jcm11092447

Chicago/Turabian Style

Madonna, Rosalinda, Silvia Fabiani, Riccardo Morganti, Arianna Forniti, Filippo Biondi, Lorenzo Ridolfi, Riccardo Iapoce, Francesco Menichetti, and Raffaele De Caterina. 2022. "Exercise-Induced Pulmonary Hypertension Is Associated with High Cardiovascular Risk in Patients with HIV" Journal of Clinical Medicine 11, no. 9: 2447. https://doi.org/10.3390/jcm11092447

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop