Skip to main content
Erschienen in: Critical Care 1/2020

Open Access 01.12.2020 | COVID-19 | Research

Right ventricular-arterial uncoupling independently predicts survival in COVID-19 ARDS

verfasst von: Michele D’Alto, Alberto M. Marra, Sergio Severino, Andrea Salzano, Emanuele Romeo, Rosanna De Rosa, Francesca Maria Stagnaro, Gianpiero Pagnano, Raffaele Verde, Patrizia Murino, Andrea Farro, Giovanni Ciccarelli, Maria Vargas, Giuseppe Fiorentino, Giuseppe Servillo, Ivan Gentile, Antonio Corcione, Antonio Cittadini, Robert Naeije, Paolo Golino

Erschienen in: Critical Care | Ausgabe 1/2020

Abstract

Aim

To investigate the prevalence and prognostic impact of right heart failure and right ventricular-arterial uncoupling in Corona Virus Infectious Disease 2019 (COVID-19) complicated by an Acute Respiratory Distress Syndrome (ARDS).

Methods

Ninety-four consecutive patients (mean age 64 years) admitted for acute respiratory failure on COVID-19 were enrolled. Coupling of right ventricular function to the pulmonary circulation was evaluated by a comprehensive trans-thoracic echocardiography with focus on the tricuspid annular plane systolic excursion (TAPSE) to systolic pulmonary artery pressure (PASP) ratio

Results

The majority of patients needed ventilatory support, which was noninvasive in 22 and invasive in 37. There were 25 deaths, all in the invasively ventilated patients. Survivors were younger (62 ± 13 vs. 68 ± 12 years, p = 0.033), less often overweight or usual smokers, had lower NT-proBNP and interleukin-6, and higher arterial partial pressure of oxygen (PaO2)/fraction of inspired O2 (FIO2) ratio (270 ± 104 vs. 117 ± 57 mmHg, p < 0.001). In the non-survivors, PASP was increased (42 ± 12 vs. 30 ± 7 mmHg, p < 0.001), while TAPSE was decreased (19 ± 4 vs. 25 ± 4 mm, p < 0.001). Accordingly, the TAPSE/PASP ratio was lower than in the survivors (0.51 ± 0.22 vs. 0.89 ± 0.29 mm/mmHg, p < 0.001). At univariate/multivariable analysis, the TAPSE/PASP (HR: 0.026; 95%CI 0.01–0.579; p: 0.019) and PaO2/FIO2 (HR: 0.988; 95%CI 0.988–0.998; p: 0.018) ratios were the only independent predictors of mortality, with ROC-determined cutoff values of 159 mmHg and 0.635 mm/mmHg, respectively.

Conclusions

COVID-19 ARDS is associated with clinically relevant uncoupling of right ventricular function from the pulmonary circulation; bedside echocardiography of TAPSE/PASP adds to the prognostic relevance of PaO2/FIO2 in ARDS on COVID-19.
Hinweise
Michele D’Alto and Alberto M. Marra take responsibility for all aspects of the reliability and freedom from bias of the data presented and their discussed interpretation
Michele D’Alto and Alberto M. Marra have contributed equally to this manuscript.
Robert Naeije and Paolo Golino have contributed equally to this manuscript.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ARF
Acute respiratory failure
ARDS
Acute respiratory distress syndrome
AUC
Area under the curve
COVID-19
Corona Virus Infectious Disease 2019
CT
Computed tomography
Ea
Arterial elastance
Ees
End-systolic elastance
FIO2
Fraction of inspired O2
IQR
Interquartile range
LV
Left ventricle
PaO2
Arterial partial pressure of oxygen
PASP
Systolic pulmonary artery pressure
ROC
Receiver operator characteristics
RV
Right ventricle
SARS-CoV-2
Severe acute respiratory syndrome-coronavirus-2
SD
Standard deviation
TAPSE
Tricuspid annular plane systolic excursion

Background

Severe Acute Respiratory Syndrome-CoronaVirus-2 (SARS-CoV-2) infection, or Corona Virus Infectious Disease 2019 (COVID-19), may be complicated by the acute respiratory distress syndrome (ARDS) with reported high mortality rates between 26 and 61% [1, 2]. There are data suggesting that COVID-19 respiratory failure differs from “typical” ARDS by several aspects, including preserved respiratory system compliance [3], good tolerance to hypoxemia ("happy hypoxemia") [4], and prominent micro- and macrovascular thrombotic changes in relation with extensive endothelial injury [5, 6]. However, whether the respiratory physiology of COVID-19-induced ARDS really differs from other types of ARDS remains discussed [7]. On the cardiac side, COVID-19 has also been associated with myocardial injury [8] and altered right ventricle (RV) strain as an independent predictor of poor prognosis [9]. There are data suggesting that COVID-19 may predominantly affect the RV and that is clinically relevant [10].
Right heart failure (“acute cor pulmonale”) is a long-recognized complication of ARDS, in relation to severity of the disease and ventilatory strategies associated with hyper-inflated lungs and permissive hypercapnia [11]. We hypothesized that myocardial injury and inflammatory changes in COVID-19 could be an additional cause of ARDS-related acute right heart failure. We therefore assessed the coupling of RV function to the pulmonary circulation in COVID-19 ARDS patients. To this purpose, we used bedside transthoracic echocardiography with focus on the tricuspid annular plane systolic excursion (TAPSE)/pulmonary artery systolic pressure (PASP) ratio, previously shown to be a valid surrogate of the gold standard ratio of end-systolic to arterial elastance (Ees/Ea) for the assessment of RV-arterial coupling [12] and an independent predictor of outcome in heart failure and pulmonary arterial hypertension [13].

Methods

Study design

This was a prospective study from two Italian centres, Ospedale dei Colli (Monaldi-Cotugno) and Federico II Hospital, Naples, Italy, which are teaching hospitals authorized for COVID-19 patients. All patients were enrolled from 8 March to 8 May 2020. The diagnosis of COVID-19 was confirmed according to the interim guidance of World Health Organization [14]. The study was approved by local Ethics Committees (#AOC/0015171/2020).

Data collection

Patients’ demographics, clinical status, disease duration from the symptoms onset, medical history, comorbidities, laboratory examinations, concomitant treatment, type of ventilation, eventual complications, treatment, and outcomes were recorded. The diagnosis of ARDS rested on the Berlin consensus criteria and PaO2/FIO2 ratios discriminating mild, moderate, and severe forms of the disease [15]. Treatment was in keeping with current expert recommendations, with high-flow nasal O2 as needed to restore arterial oxygenation, and ventilation with positive end-expiratory pressure by facial mask or tracheal intubation following current expert recommendations [16]. Thus, tidal volume was kept as low as possible, on average to 6 ml/kg; positive end-expiratory pressure titrated by 2–3 cmH2O increments to a maximum of 10–15 cmH2O and a plateau pressure < 30 cmH2O. Noninvasive ventilation was applied when endotracheal intubation was not considered necessary.

Transthoracic echocardiography

Bedside transthoracic echocardiographic examinations were performed with the Vivid E9 ultrasound system (General Electrics Medical Systems, Andover, MA, USA), according to the American Society of Echocardiography guidelines [17]. Images were stored and analyzed offline by three independent trained observers (MD, SS and AMM).

Statistical analysis

Kolmogorov–Smirnov test was applied to test the variable distribution. Normally distributed continuous variables were expressed as mean ± standard deviation (SD); skewed distributed continuous data were expressed as median and interquartile range [IQR]; categorical variables were expressed as counts and percentages. Two-tailed t test for paired and unpaired data was used to assess changes between groups. Linear regression analyses and partial correlation test by Pearson’s method were used to assess univariate relations. The association between analyzed variables and outcome (i.e., mortality) was established by using Cox proportional hazard regression analyses. Univariate and multivariable linear models were used to assess potential predictors of outcome. The following variables, selected according to their potential clinical relevance, were included in the analysis: age, sex, disease duration, previous lung disease, previous coronary artery disease, cardiovascular risk factors (hypertension, diabetes, obesity, smoke), therapy for COVID, type of ventilation, PaO2/FiO2 ratio, creatinine, cardiac troponin I, C-reactive protein, activated partial thromboplastin time, N-terminal pro-brain natriuretic peptide, interleukine-6, left ventricle (LV) end-diastolic diameter, LV end-systolic diameter, left atrium diameter, LV ejection fraction, mitral and aortic valve diseases, tricuspid regurgitation, TAPSE, PASP, TAPSE/PASP ratio, inferior vena cava dimension and ratio of RV to LV surface areas on an apical 4-chamber view. Results were expressed as hazard ratios with 95% confidence intervals. Outcome prediction accuracies were tested by calculating the area under the curve (AUC) for the receiver operator characteristics (ROC) curve analysis for TAPSE/PASP and PaO2/FiO2 across the endpoint. Kaplan–Meier curves for cumulative survival were constructed for the endpoint to assess the impact of TAPSE/PASP and PaO2/FiO2 on survival, categorizing patients using optimal cutoff points for TAPSE/PASP and PaO2/FiO2 derived from Youden’s Index from the ROC curve. Further, ROC curve analyses using the same multivariable model, with and without TAPSE/PASP and PaO2/FiO2, were used to investigate the gain in C-statistic for associations with outcome when compared to the same model without these parameters. Statistical analyses were performed using SPSS version 25.0 (SPSS Inc, Chicago, Illinois, USA). A value of p < 0.05 was considered statistically significant.

Results

Ninety-four patients were included in the study; they presented with fever (94/94, 100%), dyspnea (87/94, 93%), fatigue (94/94, 100%) and cough (58/94, 62%). All patients had a computed tomography (CT) scan diagnostic for diffuse or localized pneumonia. The echocardiographic assessment was performed on average 3 days after hospital admission (range 1–7 days) after the patients had been stabilized with either high flow supplemental O2 or invasive/noninvasive ventilation.
The clinical data of the survivor and non-survivor patients are shown in Table 1.
Table 1
Comparison between alive and dead patients affected by COVID-19
 
Alive (n = 69)
Dead (n = 25)
p
Age (year)
62 ± 13
68 ± 12
0.033
Sex M (%)
53 (77)
17 (68)
0.549
Disease duration (day)
7.7 ± 3.3
7.7 ± 3.1
0.942
Lung disease (%)
17 (25)
11 (44)
0.079
Coronary artery disease (%)
14 (20)
3 (12)
0.545
Cardiovascular comorbidities
 Hypertension (%)
44 (64)
19 (76)
0.362
 Diabetes (%)
11 (16)
5 (20)
0.99
 Smoke (%)
7 (10)
8 (32)
0.021
 Obesity (%)
18 (26)
13 (52)
0.025
Treatment
 Anticoagulants (%)
69 (100)
24 (96)
0.097
 Hydroxycloroquine (%)
51 (74)
19 (76)
0.840
 Antivirals (%)
43 (62)
23 (92)
0.005
 Monoclonal antibodies (%)
8 (12)
10 (40)
0.005
 Corticosteroids (%)
14 (20)
6 (24)
0.98
Type of ventilation
 Nasal oxygen (%)
35 (51)
0 (0)
 < 0.001
 Noninvasive ventilation (%)
22 (32)
0 (0)
 < 0.001
 Intubation (%)
12 (17)
25 (100)
 < 0.032
Biochemistry
 Creatinine (mg/dl)
1.3 ± 1.3
2.8 ± 1.4
 < 0.001
 Cardiac Troponin I (pg/l)
365 ± 644
1245 ± 2049
 < 0.002
 D-dimer (ng/ml)
317 ± 557
919 ± 974
 < 0.001
 C-reactive protein (mg/dl)
10.6 ± 19.9
22.8 ± 27.3
 < 0.023
 Procalcitonin (ng/ml)
0.6 ± 1.5
1.8 ± 2.0
 < 0.005
 APTT (sec)
36.8 ± 6.7
40.6 ± 4.3
0.037
 NT-proBNP (pg/ml)
686 ± 1224
3375 ± 3891
 < 0.001
 Interleukine-6 (ng/ml)
33.6 ± 33.4
246.4 ± 87.4
 < 0.001
 PaO2/FiO2 ratio (mmHg)
270 ± 104
117 ± 56
 < 0.001
Value are represented as mean ± standard deviation or absolute value and (%)
APTT partial thromboplastin time, NT-proBNP N-terminal prohormone of brain natriuretic peptide, PaO2 arterial partial pressure of oxygen, FiO2 fraction of inspired O2
The patient population was globally relatively old, predominantly male and presented with pulmonary comorbidities and cardiovascular risk factors. Non-survivors were older by an average of 6 years and were more frequently smokers and overweight. The majority of the patients were anticoagulated and treated with hydroxychloroquine. A proportion of the patients also received antiviral drugs, monoclonal antibodies, and corticosteroids. Non-survivors received more frequently antiviral drugs and invasive mechanical ventilation. Serum creatinine, cardiac troponin I, C-reactive protein, interleukine-6, N-terminal pro-brain natriuretic peptide, activated partial thromboplastin time, and pro-calcitonin were higher and PaO2/FIO2 lower in non-survivors. An angio CT performed when dyspnea was deemed out of proportion of standard CT imaging revealed a pulmonary embolism in nine of the patients.
Echocardiographic findings shown in Table 2 disclosed a significant increase in PASP, inferior vena cava dimensions and a decrease in TAPSE/PASP in non-survivors, as compared to survivors.
Table 2
Echocardiographic features
 
Alive (n = 69)
Dead (n = 25)
p
LVEDD (mm)
48 ± 5
49 ± 4
0.388
LVESD (mm)
29 ± 7
31 ± 5
0.059
LAD (mm)
38 ± 6
40 ± 5
0.082
LVEF (%)
60 ± 7
58 ± 8
0.209
MVD
5 (7)
1 (4)
0.574
AVD
1 (1)
0 (0)
0.550
TR
2 (3)
3 (12)
0.084
TAPSE (mm)
25 ± 4
19 ± 4
 < 0.001
PASP (mmHg)
30 ± 7
42 ± 12
 < 0.001
TAPSE/PASP
0.89 ± 0.29
0.51 ± 0.22
 < 0.001
IVC (mm)
15 ± 4
20 ± 3
 < 0.001
Pericardial effusion
6 (9)
4 (16)
0.375
Echocardiographic phenotypes
 Normal
50 (73)
10 (40)
0.007
 Hyperkinetic
9 (13)
3 (12)
0.99
 Right
3 (4)
12 (48)
 < 0.001
 LV depression
3 (4)
0 (0)
0.57
 Severe pericardial effusion
4 (6)
0 (0)
0.57
Value are represented as mean ± standard deviation or absolute value and (%). p < 0.05 statistically significant
LVEDD left ventricle end-diastolic diameter, LVESD left ventricle end-systolic diameter, LAD left atrium diameter, LVEF left ventricle ejection fraction, MVD mitral valve disease moderate-to-severe, AoVD aortic valve disease moderate-to-severe, TR tricuspid regurgitation, TAPSE tricuspid annulus plane systolic excursion, PASP pulmonary artery systolic pressure, IVC inferior vena cava
A typical right heart phenotype echocardiographic examination is shown in Fig. 1 (Panel A and B).
The results of univariate and multivariable analyses are shown, respectively, in Table 3 and Table 4.
Table 3
Single predictor models of Cox proportional hazard analysis
Variables
HR
95% (CI)
p
Age (year)
1.04
1.003–1.078
0.035
Sex (female)
0.006
0.000–0.216
0.006
Fever (days)
1.001
0.912–1.098
0.98
Pulmonary disease
1.998
0.906–4.408
0.086
Coronary artery disease
0.556
0,166–1.858
0.340
Hypertension
1,767
0.706–4.429
0.224
Diabetes
1.525
0.571–4.072
0.40
Smokers
3.050
1.313–7.086
0.10
Obesity
2.252
1.027–4.936
0.044
Risk factors
0.025
0
Ref
1
6.608
0.853–51
0.07
2
5.126
0.617–42.6
0.13
3
15.518
1.861–129
0.011
4
46.105
2.772–766
0.008
Nasal oxygen
0.019
0.001–0.512
0.018
Noninvasive ventilation
0.031
0.001–1.386
0.073
Intubation
223,89
4.81–10,415
0.006
PaO2/FiO2 ratio (mmHg)
0.986
0.981–0.992
0.001
Heparin
0.274
0.037–2.039
0.206
Antivirals
5.935
1.398–25.186
0.016
Hydroxycloroquine
1.195
0.477–2.995
0.704
Monoclonal antibody
3.301
1.481–7.356
0.003
Corticosteroids
1.308
0.521–3.279
0.568
Creatinine (mg/mL)
1.236
1.067–1.432
0.005
Troponine (pg/l)
1.000
1.000–1.000
0.003
D-dimer (ng/ml)
1.000
1.000–1.001
 < 0.001
C-reactive protein (mg/dl)
1.014
1.004–1.024
0.006
Procalcitonin (ng/ml)
1.108
0.975–1.259
0.115
NT-proBNP (pg/ml)
1.000
1.000–1.000
 < 0.001
APTT (sec)
0.997
0.982–1.012
0.687
Interleukine-6 (ng/ml)
1.010
1.007–1.013
 < 0.001
Heart rate (bpm)
1.031
1.006–1.058
0.016
Systolic blood pressure (mmHg)
0.976
0.949–1.003
0.076
Diastolic blood pressure (mmHg)
0.912
0.868–0.958
 < 0.001
LVEDd (mm)
1.031
0.948–1.121
0.473
LVESd (mm)
1.039
0.99–1.091
0.123
LAD (mm)
1.066
0.995–1.143
0.068
LVEF (%)
0.972
0.934–1.011
0.151
Severe MR
0.526
0.071–3.887
0.529
Severe AR
0.049
0–201,330
0.697
Severe TR
2.671
0.798–8.95
0.111
TAPSE (mm)
0.796
0.727–0.871
 < 0.001
PASP (mmHg)
1.085
1.054–1.118
 < 0.001
TAPSE/PASP (mm/mmHg)
0.013
0.002–0.069
 < 0.001
IVC (mm)
1.335
1.201–1.483
 < 0.001
IVC respiratory changes
1.591
0.702–3.606
0.226
Pericardial effusion
1.693
0.580–4.940
0.335
Pleural effusion
0.868
0.204–3.689
0.848
Right phenotype
4.232
1.505–11.902
0.006
APTT partial thromboplastin time, NT-proBNP N-terminal prohormone of brain natriuretic peptide, LVEDD left ventricle end-diastolic diameter, LVESD left ventricle end-systolic diameter, LAD left atrium diameter, LVEF left ventricle ejection fraction, MVD mitral valve disease moderate-to-severe, AoVD aortic valve disease moderate-to-severe, TR tricuspid regurgitation, TAPSE tricuspid annulus plane systolic excursion, PASP pulmonary artery systolic pressure, IVC inferior vena cava
Table 4
Multivariable models of Cox proportional hazard analysis
Variables
HR
95% (CI)
p
Age (year)
1.002
0.944–1.063
0.953
Obesity
0.626
0.171–2.295
0.480
Creatinine (mg/mL)
1.033
0.746–1.429
0.847
Troponine (pg/L)
1.00
0.999–1.001
0.774
D-dimer (ng/mL)
1.00
0.999–1.001
0.442
C-reactive protein (mg/mL)
1.01
0.996–1.024
0.171
Heart rate (bpm)
0.996
0.961–1.032
0.817
Systolic blood pressure (mmHg)
1.038
0.988–1.09
0.137
Diastolic blood pressure (mmHg)
0.915
0.837–1.002
0.054
LVEDd (mm)
1.064
0.991–1.550
0.508
LVESd (mm)
0.899
0.707–1.143
0.385
LVEF (%)
1.022
0.900–1.161
0.739
LAD (mm)
0.947
0.858–1.046
0.947
TAPSE/PASP (mm/mmHg)
0.026
0.01–0.579
0.019
PaO2/FiO2 ratio (mmHg)
0.988
0.977–0.998
0.018
LVEDD left ventricle end-diastolic diameter, LVESD left ventricle end-systolic diameter, LVEF left ventricle ejection fraction, LAD left atrium diameter, TAPSE tricuspid annulus plane systolic excursion, PASP pulmonary artery systolic pressure, PaO2 arterial partial pressure of oxygen, FiO2 fraction of inspired O2
While at univariate analysis most of the biological and echocardiographic differences between survivors and non-survivors were significantly associated with survival (Table 3), only PaO2/FIO2 and TAPSE/PASP emerged as independent predictors after adjustment at multivariable analysis [hazard ratio (95% confidence interval); p value: 0.988 (0.977–0.998); p = 0.018 and 0.026 (0.01–0.579); p = 0.019, respectively] (Table 4).
Individual values for TAPSE/PASP and PaO2/FIO2 in survivors and non-survivors are presented in Fig. 2. ROC curves to predict outcome of these two variables are shown in Fig. 3. When patients were dichotomised according to the Youden’s Index for optimal cutoff point from the ROC curve (159 mmHg and 0.635 mm/mmHg, PaO2/FIO2 and TAPSE/PASP, respectively), Kaplan–Meyer curves of % survival as a function of time in patients showed that patients with TAPSE/PASP or PaO2/FIO2 below ROC-derived cutoff values have reduced survival (chi square; log rank test p: 26.43; < 0.001 and 42.83; < 0.001, respectively) (Fig. 4). Furthermore, when patients were categorized according to value of both parameters, patients with reduction of both parameters showed the lowest survival (chi square: 45.87; log rank test p: < 0.001), significantly different to those with normal levels (chi square: 50.32, p < 0.001) or only one parameter impaired (chi square: 9.56, p = 0.001). A combination of high TAPSE/PASP or PaO2/FIO2 allowed for a very high likelihood of survival. Exclusion of the 9 pulmonary embolism patients from multivariate analysis did not affect the results.

Discussion

The present results show that COVID 19-induced ARDS is associated with early and pronounced uncoupling of right ventricular function from the pulmonary circulation and that its noninvasive echocardiographic assessment by the TAPSE/PASP ratio adds significantly and independently to the prognostic relevance of the PaO2/FIO2 ratio in these patients.
The reported COVID-19 patients were diagnosed with pneumonia complicated by ARDS based on clinical presentation of dyspnea, cough, and fatigue; compatible chest computed tomography findings; and the PaO2/FIO2 ratio. At the moment of echocardiographic evaluation, the PaO2/FIO2 ratio had been corrected in a proportion of the survivors (Fig. 2). The patients were treated empirically with drugs expected to be of benefit, such as hydroxychloroquine and were anticoagulated. Their ventilatory management included proning, application of positive end-expiratory pressure, and so-called “protective ventilation” with a low as possible tidal volumes [16]. This resulted in a 26% mortality at the lower range of currently reported [18].
Pulmonary hypertension in the present study was mild to moderate as based on echocardiographic estimates of PASP. A PASP of 40 mmHg in the non-survivors would indeed be at the upper limit of normal taken into account age, sex, and body weight [19]. On the other hand, the TAPSE was decreased but still above the lower limit of normal in the non-survivors [20]. Accordingly, the TAPSE/PASP at 0.89 ± 0.29 in survivors was mildly decreased compared to the value of 1.11 ± 0.03 previously reported in 209 subjects older than 60 years [20]. However, it was markedly decreased to 0.51 ± 0.22 mm/mmHg in non-survivors, approaching values below 0.50 mm/mmHg previously shown to be of poor prognosis in heart failure and severe pulmonary hypertension [13].
In a recent report of 200 hospitalized with COVID-19 in non-ICU departments, PASP was > 35 mmHg in 12% and the TAPSE < 17 mm in 14.5%, but increased PASP and not decreased TAPSE was found to predict a poor outcome [21]. Mild pulmonary hypertensions along with moderate decrease in TAPSE in that study are in keeping with the present findings in more severely ill patients with respiratory insufficiency.
The TAPSE/PASP ratio was initially proposed as an estimate of RV myocardial length-tension relationship and as such showed to be of prognostic relevance in heart failure [21]. Subsequent studies confirmed its prognostic capability, not only in heart failure [22] but also in pulmonary arterial hypertension [23] and in patients with chronic lung diseases [24]. In these studies, the TAPSE/PASP was assumed to inform about RV-PA coupling, with TAPSE considered as a load-dependent surrogate of Ees and PASP as an indirect estimate of Ea [2224]. The TAPSE/PASP has been shown to be superior to other composite echocardiographic indices in the assessment of RV-PA and correlated to gold standard invasive [12] or indirectly assessed Ees/Ea ratios [22].
As in the present study the TAPSE/PASP ratio was mostly decreased in invasively ventilated patients, one could wonder if the application of positive end-expiratory pressure could have contributed to increased PAP and RV-PA uncoupling [25]. COVID-19 ARDS patients could have presented with increased transmission of alveolar pressures to pulmonary resistive vessels because of preserved lung compliance [3]. Mechanics of the respiratory system were not assessed in the present study. However, the notion of preserved compliance in COVID-19 ARDS may not be confirmed in most of these patients [7, 26], and the “protective ventilation” approach in the present study would be expected to avoid to high volumes and alveolar pressures as a cause of excessive RV afterload [11]. This was confirmed by only mild increases in PASP disclosed by the echocardiographic examinations.
The reason for RV-PA uncoupling in the presence of only mildly increased PAP is not immediately apparent. The basic response of RV function to increased afterload is homeometric, with increased Ees (contractility) to match Ea (afterload), and uncoupling expected but only in severe or rapidly evolving pulmonary hypertension [27]. However, early RV-PA uncoupling may be observed in severe inflammatory conditions such as sepsis [28] or also in left heart failure because of negative ventricular interactions [29]. Both may occur in COVID-19 patients [8]. Therefore, the right heart in COVID-19 patients may fail even in the presence of only modest increase in afterload.
The present results are in keeping with a recent echocardiographic study in patients with COVID-19 ARDS, in which non-survivors had a PASP at the upper limit of normal, decreased indices of RV systolic function, and longitudinal strain identified as an independent predictor of outcome [9]. Pulmonary hypertension in COVID-19 may belong either to pulmonary hypertension due to lung parenchymal disease or at most probably to chronic thromboembolic pulmonary hypertension. The TAPSE/PASP is easier to assess, can be part of standard bedside echocardiographic assessments as it does not require off-line analysis of images and specific software, and may be a more sensitive assessment of RV-PA coupling. The high prevalence of RV dilatation and dysfunction in the range of 40–50% recently reported in patients with COVID-19 [10, 30] underscore the exquisite sensitivity of the RV to this newly appeared viral infection.
The most potent predictor of outcome in ARDS is the PaO2/FIO2 ratio, which as such is part of the definition of the syndrome [14]. In the present study, the TAPSE/PASP emerged with equally potent prognostic capability, suggesting a major component of acute cor pulmonale in COVID-19 ARDS pathophysiology. Whether this is entirely particular to COVID-19 ARDS is uncertain as there have been no systematic evaluations of RV-PA coupling in more “typical” ARDS or other viral pneumonia ARDS controls.
The present study is limited by relatively small sample size and by the small number of events. This might limit the results of the multivariate analyses and lead to a certain over-fitting. An angio-CT to diagnose acute pulmonary embolism was performed on physician in care's clinical suspicion, so that the frequency of this complication might have been under-estimated. However, PAP in the present study in the present study remained at the upper limit of normal, excluding pulmonary embolism as a cause of afterload-induced RV-PA uncoupling. Furthermore, excluding 9 of the patients with a diagnosis of pulmonary embolism did not affect the predictive capability of the TAPSE/PASP and PaO2/FIO2 ratios. Other limitations might be absence of respiratory system compliance measurements, absence of non-COVID-19 viral pneumonia controls, and exclusively noninvasive evaluations of the right heart and the pulmonary circulation. However, the results call attention to cor pulmonale as an important component of COVID-19 ARDS and plea for systematic bedside echocardiographic assessments added to blood gases and lung mechanics in the management of these patients.
Approximately 4 decades ago, Zapol and Snider called attention to the pulmonary circulation and the right heart in severe ARDS [31]. Pulmonary hypertension in these patients is nowadays uncommon along with progress in management, but “acute cor pulmonale” continues to be reported, albeit generally in the context of ventilatory settings associated with excessive increase in alveolar pressure and permissive hypercapnia [11]. The present investigation shows that acute uncoupling of the right heart from the quasi-normotensive pulmonary circulation may also occur in the context of severe systemic inflammation and vasculitis.

Conclusions

In conclusion, COVID 19-induced ARDS is associated with early and pronounced right ventricular-arterial uncoupling, and its noninvasive echocardiographic assessment by the TAPSE/PASP ratio adds significantly and independently to the prognostic relevance of the PaO2/FIO2 ratio in these patients. These data call for the indispensable integration of bedside echocardiography in the assessment of COVID-19 patients in the intensive care setting.

Acknowledgements

We thank all study participants and staff from Monaldi Hospital, Cotugno Hospital and Federico II University Hospital.
This study was approved by the Institutional Ethics Committees of Monaldi Hospital (#AOC/0015171/2020). All data were anonymized to comply with the provisions of personal data protection legislation. Due to the retrospective nature of this study and due the fact that only historical medical data were collected, written informed consent was not required.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, Cereda D, Coluccello A, Foti G, Fumagalli R, Iotti G, Latronico N, Lorini L, Merler S, Natalini G, Piatti A, Ranieri MV, Scandroglio AM, Storti E, Cecconi M, Pesenti M. COVID-19 lombardy ICU network. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy region, Italy. JAMA. 2020;323:1574–81.CrossRefPubMedPubMedCentral Grasselli G, Zangrillo A, Zanella A, Antonelli M, Cabrini L, Castelli A, Cereda D, Coluccello A, Foti G, Fumagalli R, Iotti G, Latronico N, Lorini L, Merler S, Natalini G, Piatti A, Ranieri MV, Scandroglio AM, Storti E, Cecconi M, Pesenti M. COVID-19 lombardy ICU network. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy region, Italy. JAMA. 2020;323:1574–81.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Bhatraju PK, Ghassemieh BJ, Nichols M, Kim R, Jerome KR, Nalla AK, Greninger AL, Pipavath S, Wurfel MM, Evans L, Kritek PA, West TE, Luks A, Gerbino A, Dale CR, Goldman JD, O’Mahony S, Mikacenic C. Covid-19 in critically Ill patients in the Seattle region—case series. N Engl J Med. 2020;382:2012–22.CrossRefPubMed Bhatraju PK, Ghassemieh BJ, Nichols M, Kim R, Jerome KR, Nalla AK, Greninger AL, Pipavath S, Wurfel MM, Evans L, Kritek PA, West TE, Luks A, Gerbino A, Dale CR, Goldman JD, O’Mahony S, Mikacenic C. Covid-19 in critically Ill patients in the Seattle region—case series. N Engl J Med. 2020;382:2012–22.CrossRefPubMed
3.
Zurück zum Zitat Gattinoni L, Coppola S, Cressoni M, Busana M, Chiumello D, Rossi S. COVID-19 does not lead to a “typical” acute respiratory distress syndrome. Am J Respir Crit Care Med. 2020;201:1299–300.CrossRefPubMedPubMedCentral Gattinoni L, Coppola S, Cressoni M, Busana M, Chiumello D, Rossi S. COVID-19 does not lead to a “typical” acute respiratory distress syndrome. Am J Respir Crit Care Med. 2020;201:1299–300.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Ottestad W, Søvik S. COVID-19 patients with respiratory failure: what can we learn from aviation medicine? Brit J Anaesth 2020;S0007–0912:(20)30226–9. Ottestad W, Søvik S. COVID-19 patients with respiratory failure: what can we learn from aviation medicine? Brit J Anaesth 2020;S0007–0912:(20)30226–9.
5.
Zurück zum Zitat Klok FA, Kruip MJHA, Meer NJM, van der, Arbous MS, Gommers D, Kant KM, Kaptein FHJ, van Paassen J, Stals MAM, Huisman MV, Endeman H. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res 2020;S0049–3848:(20)30120–1. Klok FA, Kruip MJHA, Meer NJM, van der, Arbous MS, Gommers D, Kant KM, Kaptein FHJ, van Paassen J, Stals MAM, Huisman MV, Endeman H. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res 2020;S0049–3848:(20)30120–1.
7.
Zurück zum Zitat Grieco DL, Bongiovanni F, Chen L, Menga LS, Cutuli SL, Pintaudi G, Carelli S, Michi T, Torrini F, Lombardi G, Anzellotti GM, De Pascale G, Urbani A, Bocci MG, Tanzarella ES, Bello G, Dell’Anna AM, Maggiore SM, Brochard L, Antonelli M. Respiratory physiology of COVID-19-induced respiratory failure compared to ARDS of other etiologies. Crit Care. 2020;24(1):529. https://doi.org/10.1186/s13054-020-03253-2.CrossRefPubMedPubMedCentral Grieco DL, Bongiovanni F, Chen L, Menga LS, Cutuli SL, Pintaudi G, Carelli S, Michi T, Torrini F, Lombardi G, Anzellotti GM, De Pascale G, Urbani A, Bocci MG, Tanzarella ES, Bello G, Dell’Anna AM, Maggiore SM, Brochard L, Antonelli M. Respiratory physiology of COVID-19-induced respiratory failure compared to ARDS of other etiologies. Crit Care. 2020;24(1):529. https://​doi.​org/​10.​1186/​s13054-020-03253-2.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Santoso A, Pranata R, Wibowo A, Wibowo A, Al-Farabi MJ, Huang I, Antariksa B. Cardiac injury is associated with mortality and critically ill pneumonia in COVID-19: A meta-analysis. Am J Emerg Med. 2020;S0735–6757:(20)30280–1. Santoso A, Pranata R, Wibowo A, Wibowo A, Al-Farabi MJ, Huang I, Antariksa B. Cardiac injury is associated with mortality and critically ill pneumonia in COVID-19: A meta-analysis. Am J Emerg Med. 2020;S0735–6757:(20)30280–1.
10.
Zurück zum Zitat Szekely Y, Lichter Y, Taieb P, Banai A, Hochstadt A, Merdler I, Gal Oz A, Rothschild E, Baruch G, Peri Y, Arbel Y, Topilsky Y. Spectrum of cardiac manifestations in COVID-19: a systematic echocardiographic study. Circulation. 2020;142:342–53.CrossRefPubMedPubMedCentral Szekely Y, Lichter Y, Taieb P, Banai A, Hochstadt A, Merdler I, Gal Oz A, Rothschild E, Baruch G, Peri Y, Arbel Y, Topilsky Y. Spectrum of cardiac manifestations in COVID-19: a systematic echocardiographic study. Circulation. 2020;142:342–53.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Vieillard-Baron A, Price LC, Matthay MA. Acute cor pulmonale in ARDS. Intens Care Med. 2013;39:1836–8.CrossRef Vieillard-Baron A, Price LC, Matthay MA. Acute cor pulmonale in ARDS. Intens Care Med. 2013;39:1836–8.CrossRef
12.
Zurück zum Zitat Tello K, Wan J, Dalmer A, Vanderpool R, Ghofrani HA, Naeije R, Roller F, Mohajerani E, Seeger W, Herberg U, Sommer N, Gall H, Richter MJ. Validation of the tricuspid annular plane systolic excursion/systolic pulmonary artery pressure ratio for the assessment of right ventricular-arterial coupling in severe pulmonary hypertension. Circ Cardiovasc Imaging. 2019;12(9):e009047.CrossRefPubMedPubMedCentral Tello K, Wan J, Dalmer A, Vanderpool R, Ghofrani HA, Naeije R, Roller F, Mohajerani E, Seeger W, Herberg U, Sommer N, Gall H, Richter MJ. Validation of the tricuspid annular plane systolic excursion/systolic pulmonary artery pressure ratio for the assessment of right ventricular-arterial coupling in severe pulmonary hypertension. Circ Cardiovasc Imaging. 2019;12(9):e009047.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Guazzi M. Use of TAPSE/PASP ratio in pulmonary arterial hypertension: an easy shortcut in a congested road. Int J Cardiol. 2018;266:242–4.CrossRefPubMed Guazzi M. Use of TAPSE/PASP ratio in pulmonary arterial hypertension: an easy shortcut in a congested road. Int J Cardiol. 2018;266:242–4.CrossRefPubMed
14.
Zurück zum Zitat World Health Organization. Clinical management of severe acute respiratory infection when novel coronavirus (nCoV) infection is suspected. Published March 13, 2020. World Health Organization. Clinical management of severe acute respiratory infection when novel coronavirus (nCoV) infection is suspected. Published March 13, 2020.
15.
Zurück zum Zitat Ranieri VM, Rubenfeld GD, Thompson B, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012;307:2526–33.PubMed Ranieri VM, Rubenfeld GD, Thompson B, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012;307:2526–33.PubMed
16.
17.
Zurück zum Zitat Lang RM, Badano LP, Mor-Avi V, Afilalo J, Armstrong A, Ernande L, Flachskampf FA, Foster E, Goldstein SA, Kuznetsova T, Lancellotti P, Muraru D, Picard MH, Rietzschel ER, Rudski L, Spencer KT, Tsang W, Voigt JU. Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J Am Soc Echocardiogr. 2015;28:1–39.CrossRefPubMed Lang RM, Badano LP, Mor-Avi V, Afilalo J, Armstrong A, Ernande L, Flachskampf FA, Foster E, Goldstein SA, Kuznetsova T, Lancellotti P, Muraru D, Picard MH, Rietzschel ER, Rudski L, Spencer KT, Tsang W, Voigt JU. Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J Am Soc Echocardiogr. 2015;28:1–39.CrossRefPubMed
18.
Zurück zum Zitat Wunsch H. Mechanical ventilation in COVID-19. Interpreting the current epidemiology. Am J Respir Crit Care Med 2020; 202:1–4. Wunsch H. Mechanical ventilation in COVID-19. Interpreting the current epidemiology. Am J Respir Crit Care Med 2020; 202:1–4.
19.
Zurück zum Zitat McQuillan BM, Picard MH, Levitt M, Weyman AE. Clinical correlates and reference intervals for pulmonary artery systolic pressure among echocardiographically normal subject. Circulation. 2001;104:2797–802.CrossRefPubMed McQuillan BM, Picard MH, Levitt M, Weyman AE. Clinical correlates and reference intervals for pulmonary artery systolic pressure among echocardiographically normal subject. Circulation. 2001;104:2797–802.CrossRefPubMed
20.
Zurück zum Zitat Ferrara F, Rudski LG, Vriz O, Gargani L, Afilalo J, D’Andrea A, D’Alto M, Marra AM, Acri E, Stanziola AA, Ghio S, Cittadini A, Naeije R, Bossone E. Physiologic correlates of tricuspid annular plane systolic excursion in 1168 healthy subjects. Int J Cardiol. 2016;223:736–43.CrossRefPubMed Ferrara F, Rudski LG, Vriz O, Gargani L, Afilalo J, D’Andrea A, D’Alto M, Marra AM, Acri E, Stanziola AA, Ghio S, Cittadini A, Naeije R, Bossone E. Physiologic correlates of tricuspid annular plane systolic excursion in 1168 healthy subjects. Int J Cardiol. 2016;223:736–43.CrossRefPubMed
21.
Zurück zum Zitat Guazzi M, Bandera F, Pelissero G, Castelvecchio S, Menicanti L, Ghio S, Temporelli PL, Arena R. Tricuspid annular plane systolic excursion and pulmonary arterial systolic pressure relationship in heart failure: an index of right ventricular contractile function and prognosis. Am J Physiol Heart Circ Physiol. 2013;305:H1373-1381.CrossRefPubMed Guazzi M, Bandera F, Pelissero G, Castelvecchio S, Menicanti L, Ghio S, Temporelli PL, Arena R. Tricuspid annular plane systolic excursion and pulmonary arterial systolic pressure relationship in heart failure: an index of right ventricular contractile function and prognosis. Am J Physiol Heart Circ Physiol. 2013;305:H1373-1381.CrossRefPubMed
22.
Zurück zum Zitat Guazzi M, Dixon D, Labate V, Beussink-Nelson L, Bandera F, Cuttica MJ, Shah SJ. RV contractile function and its coupling to pulmonary circulation in heart failure with preserved ejection fraction: stratification of clinical Phenotypes and outcomes. JACC Cardiovasc Imaging. 2017;10:1211–21.CrossRefPubMed Guazzi M, Dixon D, Labate V, Beussink-Nelson L, Bandera F, Cuttica MJ, Shah SJ. RV contractile function and its coupling to pulmonary circulation in heart failure with preserved ejection fraction: stratification of clinical Phenotypes and outcomes. JACC Cardiovasc Imaging. 2017;10:1211–21.CrossRefPubMed
23.
Zurück zum Zitat Tello K, Axmann J, Ghofrani HA, Naeije R, Narcin N, Rieth A, Seeger W, Gall H, Richter MJ. Relevance of the TAPSE/PASP ratio in pulmonary arterial hypertension. Int J Cardiol. 2018;266:229–35.CrossRefPubMed Tello K, Axmann J, Ghofrani HA, Naeije R, Narcin N, Rieth A, Seeger W, Gall H, Richter MJ. Relevance of the TAPSE/PASP ratio in pulmonary arterial hypertension. Int J Cardiol. 2018;266:229–35.CrossRefPubMed
24.
Zurück zum Zitat Tello K, Ghofrani HA, Heinze C, Krueger K, Naeije R, Raubach C, Seeger W, Sommer N, Gall H, Richter MJ. A simple echocardiographic estimate of right ventricular-arterial coupling to assess severity and outcome in pulmonary hypertension on chronic lung disease. Eur Respir J. 2019;54(3):1802435.CrossRefPubMed Tello K, Ghofrani HA, Heinze C, Krueger K, Naeije R, Raubach C, Seeger W, Sommer N, Gall H, Richter MJ. A simple echocardiographic estimate of right ventricular-arterial coupling to assess severity and outcome in pulmonary hypertension on chronic lung disease. Eur Respir J. 2019;54(3):1802435.CrossRefPubMed
25.
Zurück zum Zitat Vieillard-Baron A, Prin S, Chergui K, Dubourg O, Jardin F. Echo-Doppler demonstration of acute cor pulmonale at the bedside in the medical intensive care unit. Am J Respir Crit Care Med. 2002;166:1310–9.CrossRefPubMed Vieillard-Baron A, Prin S, Chergui K, Dubourg O, Jardin F. Echo-Doppler demonstration of acute cor pulmonale at the bedside in the medical intensive care unit. Am J Respir Crit Care Med. 2002;166:1310–9.CrossRefPubMed
26.
Zurück zum Zitat Ziehr DR, Alladina J, Petri CR, Maley JH, Moskowitz A, Medoff BD, Hibbert KA, Thompson BT, Hardin CC. Respiratory pathophysiology of mechanically ventilated patients with COVID-19: a cohort study. Am J Respir Crit Care Med. 2020;201:1560–4.CrossRefPubMedPubMedCentral Ziehr DR, Alladina J, Petri CR, Maley JH, Moskowitz A, Medoff BD, Hibbert KA, Thompson BT, Hardin CC. Respiratory pathophysiology of mechanically ventilated patients with COVID-19: a cohort study. Am J Respir Crit Care Med. 2020;201:1560–4.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Sanz J, Sánchez-Quintana D, Bossone E, Bogaard HJ, Naeije R. Anatomy, function, and dysfunction of the right ventricle: JACC state-of-the-art review. J Am Coll Cardiol. 2019;73:1463–82.CrossRefPubMed Sanz J, Sánchez-Quintana D, Bossone E, Bogaard HJ, Naeije R. Anatomy, function, and dysfunction of the right ventricle: JACC state-of-the-art review. J Am Coll Cardiol. 2019;73:1463–82.CrossRefPubMed
28.
Zurück zum Zitat Lambermont B, Ghuysen A, Kolh P, Tchana-Sato V, Segers P, Gérard P, Morimont P, Magis D, Dogné JM, Masereel B, D’Orio V. Effects of endotoxic shock on right ventricular systolic function and mechanical efficiency. Cardiovasc Res. 2003;59:412–8.CrossRefPubMed Lambermont B, Ghuysen A, Kolh P, Tchana-Sato V, Segers P, Gérard P, Morimont P, Magis D, Dogné JM, Masereel B, D’Orio V. Effects of endotoxic shock on right ventricular systolic function and mechanical efficiency. Cardiovasc Res. 2003;59:412–8.CrossRefPubMed
29.
Zurück zum Zitat Pagnamenta A, Dewachter C, McEntee K, Fesler P, Brimioulle S, Naeije R. Early right ventriculo-arterial uncoupling in borderline pulmonary hypertension on experimental heart failure. J Appl Physiol. 2010;109:1080–5.CrossRefPubMed Pagnamenta A, Dewachter C, McEntee K, Fesler P, Brimioulle S, Naeije R. Early right ventriculo-arterial uncoupling in borderline pulmonary hypertension on experimental heart failure. J Appl Physiol. 2010;109:1080–5.CrossRefPubMed
30.
Zurück zum Zitat Mahmoud-Elsayed HM, Moody WE, Bradlow WM, Khan-Kheil AM, Senior J, Hudsmith LE, Steeds RP. Echocardiographic findings in patients with COVID-19 pneumonia. Can J Cardiol. 2020;36:1203–7.CrossRefPubMed Mahmoud-Elsayed HM, Moody WE, Bradlow WM, Khan-Kheil AM, Senior J, Hudsmith LE, Steeds RP. Echocardiographic findings in patients with COVID-19 pneumonia. Can J Cardiol. 2020;36:1203–7.CrossRefPubMed
31.
Zurück zum Zitat Zapol WM, Snider MT. Pulmonary hypertension in acute severe respiratory failure. N Engl J Med. 1977;296:476–80.CrossRefPubMed Zapol WM, Snider MT. Pulmonary hypertension in acute severe respiratory failure. N Engl J Med. 1977;296:476–80.CrossRefPubMed
Metadaten
Titel
Right ventricular-arterial uncoupling independently predicts survival in COVID-19 ARDS
verfasst von
Michele D’Alto
Alberto M. Marra
Sergio Severino
Andrea Salzano
Emanuele Romeo
Rosanna De Rosa
Francesca Maria Stagnaro
Gianpiero Pagnano
Raffaele Verde
Patrizia Murino
Andrea Farro
Giovanni Ciccarelli
Maria Vargas
Giuseppe Fiorentino
Giuseppe Servillo
Ivan Gentile
Antonio Corcione
Antonio Cittadini
Robert Naeije
Paolo Golino
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Schlagwort
COVID-19
Erschienen in
Critical Care / Ausgabe 1/2020
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-020-03385-5

Weitere Artikel der Ausgabe 1/2020

Critical Care 1/2020 Zur Ausgabe

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Hinter dieser Appendizitis steckte ein Erreger

23.04.2024 Appendizitis Nachrichten

Schmerzen im Unterbauch, aber sonst nicht viel, was auf eine Appendizitis hindeutete: Ein junger Mann hatte Glück, dass trotzdem eine Laparoskopie mit Appendektomie durchgeführt und der Wurmfortsatz histologisch untersucht wurde.

Ärztliche Empathie hilft gegen Rückenschmerzen

23.04.2024 Leitsymptom Rückenschmerzen Nachrichten

Personen mit chronischen Rückenschmerzen, die von einfühlsamen Ärzten und Ärztinnen betreut werden, berichten über weniger Beschwerden und eine bessere Lebensqualität.

Mehr Schaden als Nutzen durch präoperatives Aussetzen von GLP-1-Agonisten?

23.04.2024 Operationsvorbereitung Nachrichten

Derzeit wird empfohlen, eine Therapie mit GLP-1-Rezeptoragonisten präoperativ zu unterbrechen. Eine neue Studie nährt jedoch Zweifel an der Notwendigkeit der Maßnahme.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.