Skip to main content
Erschienen in: BMC Anesthesiology 1/2020

Open Access 01.12.2020 | Research article

Intraoperative mechanical ventilation practice in thoracic surgery patients and its association with postoperative pulmonary complications: results of a multicenter prospective observational study

verfasst von: Christopher Uhlig, Ary Serpa Neto, Meta van der Woude, Thomas Kiss, Jakob Wittenstein, Benjamin Shelley, Helen Scholes, Michael Hiesmayr, Marcos Francisco Vidal Melo, Daniele Sances, Nesil Coskunfirat, Paolo Pelosi, Marcus Schultz, Marcelo Gama de Abreu, LAS VEGAS# investigators, Protective Ventilation Network (PROVEnet), Clinical Trial Network of the European Society of Anaesthesiology

Erschienen in: BMC Anesthesiology | Ausgabe 1/2020

Abstract

Background

Intraoperative mechanical ventilation may influence postoperative pulmonary complications (PPCs). Current practice during thoracic surgery is not well described.

Methods

This is a post-hoc analysis of the prospective multicenter cross-sectional LAS VEGAS study focusing on patients who underwent thoracic surgery. Consecutive adult patients receiving invasive ventilation during general anesthesia were included in a one-week period in 2013. Baseline characteristics, intraoperative and postoperative data were registered. PPCs were collected as composite endpoint until the 5th postoperative day. Patients were stratified into groups based on the use of one lung ventilation (OLV) or two lung ventilation (TLV), endoscopic vs. non-endoscopic approach and ARISCAT score risk for PPCs. Differences between subgroups were compared using χ2 or Fisher exact tests or Student’s t-test. Kaplan–Meier estimates of the cumulative probability of development of PPC and hospital discharge were performed. Cox-proportional hazard models without adjustment for covariates were used to assess the effect of the subgroups on outcome.

Results

From 10,520 patients enrolled in the LAS VEGAS study, 302 patients underwent thoracic procedures and were analyzed. There were no differences in patient characteristics between OLV vs. TLV, or endoscopic vs. open surgery. Patients received VT of 7.4 ± 1.6 mL/kg, a PEEP of 3.5 ± 2.4 cmH2O, and driving pressure of 14.4 ± 4.6 cmH2O. Compared with TLV, patients receiving OLV had lower VT and higher peak, plateau and driving pressures, higher PEEP and respiratory rate, and received more recruitment maneuvers. There was no difference in the incidence of PPCs in OLV vs. TLV or in endoscopic vs. open procedures. Patients at high risk had a higher incidence of PPCs compared with patients at low risk (48.1% vs. 28.9%; hazard ratio, 1.95; 95% CI 1.05–3.61; p = 0.033). There was no difference in the incidence of severe PPCs. The in-hospital length of stay (LOS) was longer in patients who developed PPCs. Patients undergoing OLV, endoscopic procedures and at low risk for PPC had shorter LOS.

Conclusion

PPCs occurred frequently and prolonged hospital LOS following thoracic surgery. Proportionally large tidal volumes and high driving pressure were commonly used in this sub-population. However, large RCTs are needed to confirm these findings.

Trial registration

This trial was prospectively registered at the Clinical Trial Register (www.clinicaltrials.gov; NCT01601223; registered May 17, 2012.)
Hinweise
Christopher Uhlig, Ary Serpa Neto and Meta van der Woude contributed equally to this work.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12871-020-01098-4.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ARISCAT Score
Assess Respiratory Risk in Surgical Patients in Catalonia Score
ESA
European Society of Anaesthesiology
FIO2
Fraction of inspired oxygen
LAS VEGAS
Local assessment of ventilatory management during general anesthesia for surgery and effects on postoperative pulmonary complications
LOS
Length of stay
NMBAs
Neuromuscular blocking agents
OLV
Open lung ventilation
PBW
Predicted body weight
PEEP
Positive end-expiratory pressure
Pplat
Plateau pressure
PPCs
Postoperative pulmonary complications
PROVEnet
Protective Ventilation Network
TLV
Total lung ventilation
TOF
Train–of–four stimulation
SpO2
Peripheral oxygen saturation
STROBE
Strengthening the reporting of observational studies in epidemiology
VT
Tidal volume

Background

Approximately 234 million major surgical procedures are undertaken worldwide every year [1]. Among these, approximately 7 million patients develop major complications resulting in one million deaths during surgery or in-hospital stay, contributing to an estimated mortality rate after anesthesia of 34 per million [1, 2]. According to the ‘Local assessment of ventilatory management during general anesthesia for surgery and effects on postoperative pulmonary complications’ (LAS VEGAS) trial, postoperative pulmonary complications (PPC) occur in a significant proportion of surgical patients [3]. However, since thoracic surgery requires a differentiated ventilatory approach, those patients were excluded from the primary analysis of the LAS VEGAS study. In thoracic surgery, conventional methods to prevent and treat hypoxemia during one lung ventilation (OLV) can be harmful to the lung tissue: high fraction of inspired oxygen (FIO2) and low (or no) positive end–expiratory pressure (PEEP) both can promote atelectasis, whereas high tidal volume (VT) can cause baro- and volutrauma [4]. The type of thoracic surgery (open or endoscopic) as well as the intraoperative mechanical ventilation settings may also influence PPCs.
Intraoperative mechanical ventilation with low VT, low driving pressure, and low to moderate PEEP improved postoperative lung function and even outcome in patients undergoing open abdominal surgery [5, 6]. When low VT was used in abdominal surgery, high PEEP combined with recruitment maneuvers, as compared to low PEEP without recruitment maneuvers, did not add to the protection against PPCs [7].
The present study aimed to characterize the current mechanical ventilation practice during general anesthesia for thoracic surgery, describe the incidence of PPCs, and investigate possible associations between type of surgery (open vs. endoscopic), type of ventilation (OLV or two lung ventilation) and risk for PPCs (low risk vs high) with the incidence of PPCs. We hypothesized that intraoperative mechanical ventilation, as recommended in the literature, namely with low VT, low driving pressure, and low to moderate PEEP [8], is not commonly used during thoracic surgery, and that the incidence of PPCs is higher in this surgical population than in non-thoracic surgery.

Methods

Study design and sites

The present work is a post hoc analysis of the ‘Local assessment of ventilatory management during general anesthesia for surgery and effects on postoperative pulmonary complications’ (LAS VEGAS trial) [3]. The LAS VEGAS trial protocol was first approved by the institutional review board of the Academic Medical Center, Amsterdam, The Netherlands (W12_190#12.17.0227) and registered at clinicaltrials.gov (NCT01601223). The protocol of this trial was published elsewhere [9].

Study population and data collection

Consecutive adult patients receiving invasive ventilation during general anesthesia for elective or non–elective surgery were eligible for participation in the study, which ran for seven predefined days in each country, selected by the national coordinator, in the period between January 14th and March 4th, 2013. Patients were excluded from participation if they were aged < 18 years, or scheduled for pregnancy related surgery, surgical procedures outside the operating room, or procedures involving cardio-pulmonary bypass.
The patient database of the LAS VEGAS trial was searched for eligible patients who received either open thoracic surgery, thoracoscopic or thoracoscopy assisted surgery (both summarized as endoscopic surgery), with or without OLV. These data have not been considered in previous analyses.
Reasonable parameters of baseline characteristics, intraoperative data and preoperative risk factors for PPCs were identified from previous studies [1013]. During the intraoperative period, data describing ventilation settings and vital parameters, as well as episodes of hypoxia (SpO2 < 92%), use of recruitment maneuvers, airway pressure reduction, presence of expiratory flow limitation, hypotension (mean arterial pressure < 60 mmHg), use of vasoactive drugs, and new arrhythmias, was collected. Postoperative residual curarisation with neuromuscular blocking agents (NMBAs), defined as train–of–four stimulation (TOF) ratio <  0.9, was documented.
The definition of protective mechanical ventilation is still under debate. For this analysis it was based on recent recommendations [8, 1416]. Patients were considered to be have been protectively ventilated “as recommended” if PEEP ≥5 cmH2O and VT ≤ 8 ml/kg PBW during TLV [8, 14, 17], and PEEP ≥5 cmH2O and VT ≤ 5 ml/kg PBW during OLV [1820].
The occurrence of PPCs is presented as a collapsed composite of PPCs in the first five postoperative days. The following PPCs were scored daily from the day of surgery until hospital discharge or postoperative day 5: 1) need for supplementary oxygen (due to PaO2 < 60 mmHg or SpO2 < 90% in room air, excluding oxygen supplementation given as standard care or as continuation of preoperative therapy), 2) respiratory failure (PaO2 < 60 mmHg or SpO2 < 90% despite oxygen therapy, or need for non-invasive mechanical ventilation), 3) unplanned new or prolonged invasive or non–invasive mechanical ventilation, 4) acute respiratory distress syndrome, 5) pneumonia. Severe PPCs were defined as the occurrence of one or more of the complications 2–5. Patient data were anonymized before entry onto a password secured, web–based electronic case record form (OpenClinica, Boston, MA, USA).

Statistical analysis

Patients were stratified into groups based on: 1) use or not of OLV (OLV vs. only TLV); 2) use or not of an endoscopic approach (endoscopic vs. open); and 3) risk for PPC according to ARISCAT (low risk [ARISCAT < 26] vs. moderate-to-high risk [ARISCAT ≥26] Supplemental Table 2, Additional file 1). The ventilatory data, which were collected hourly, were first averaged for each patient according to the number of observations (median of the value). In a longitudinal analysis, this data is presented for the first, second, third, fourth and last hour of surgery. All data are presented for the whole population and for the subgroups. In-hospital length of stay (LOS) and in-hospital mortality was censored at postoperative day 28. Proportions are compared using χ2 or Fisher exact tests and continuous variables are compared using the Mann-Whitney U Test, as appropriate.
The distributions of combinations of tidal volume size and PEEP level are presented in scatter plots. Cut-offs of 6 ml/kg PBW for tidal volume, and 5 cmH2O for PEEP were chosen to form the matrices. These cut-offs were based on widely accepted values of each variable, or according to normal daily practice. The driving pressure was defined as plateau pressure (Pplat) minus the PEEP level.
Kaplan–Meier estimates of the cumulative probability of development of PPC and hospital discharge were performed. Cox proportional hazard models without adjustment for covariates were used to assess the effect of the subgroups on outcome. The proportionality assumption was tested with scaled Schoenfeld residuals. Adjustments for multiple comparisons were not performed and no assumption for missing data was done. Statistical significance was considered to be at two-sided p <  0.05. All analyses were performed with R version 3.4.1 (http://​www.​R-project.​org/​).

Results

From 10,520 patients enrolled in the LAS VEGAS study, 302 patients underwent thoracic procedures (Supplemental Figure 1, Additional file 1). Characteristics of patient and surgery are shown in Table 1. In this sub-population of 302 thoracic surgical patients, 55% (168/302) received OLV, 15.2% (46/302) were operated with an endoscopic approach and 87.4% (264/302) had moderate-to-high risk for PPCs.
Table 1
Pre-Operative Characteristics of the Patients According to Subgroups
 
All Patients
(n = 302)
OLV
(n = 168)
TLV
(n = 134)
p value
Endoscopic
(n = 46)
Open
(n = 256)
p value
Low Risk
(n = 38)
High Risk
(n = 264)
p value
Age, years
62.0 (50.0–70.8)
62.0 (50.0–70.0)
62.0 (47.2–71.0)
0.799
62.0 (49.8–70.8)
62.0 (50.0–70.2)
0.839
37.0 (28.2–46.0)
64.0 (54.8–71.2)
<  0.001
Male gender
181 (59.9)
101 (60.1)
80 (59.7)
0.941
23 (50.0)
158 (61.7)
0.135
17 (44.7)
164 (62.1)
0.040
BMI, kg/m2
25.6 (22.9–28.9)
25.7 (23.3–29.3)
25.4 (22.5–28.7)
0.398
25.1 (21.8–27.5)
25.7 (23.0–29.3)
0.129
23.1 (20.9–26.3)
25.8 (23.4–29.2)
0.010
ASA physical status
   
0.573
  
0.880
  
<  0.001
 1
40 (13.3)
22 (13.1)
18 (13.5)
 
5 (11.1)
35 (13.7)
 
20 (54.1)
20 (7.6)
 
 2
109 (36.2)
60 (35.7)
49 (36.8)
 
18 (40.0)
91 (35.5)
 
17 (45.9)
92 (34.8)
 
 3
141 (46.8)
82 (48.8)
59 (44.4)
 
20 (44.4)
121 (47.3)
 
0 (0.0)
141 (53.4)
 
 4
11 (3.7)
4 (2.4)
7 (5.3)
 
2 (4.4)
9 (3.5)
 
0 (0.0)
11 (4.2)
 
ARISCAT score
40.0 (27.0–49.5)
40.0 (27.0–47.0)
39.0 (27.0–50.0)
0.755
35.0 (27.0–43.0)
40.0 (27.0–50.0)
0.018
24.0 (24.0–24.0)
43.0 (27.0–50.0)
< 0.001
Functional status
 Independent
270 (89.4)
153 (91.1)
117 (88.0)
0.536
43 (93.5)
227 (89.0)
0.438
36 (94.7)
234 (89.0)
 
 Partially dependent
26 (8.6)
12 (7.1)
14 (10.5)
 
2 (4.3)
24 (9.4)
 
2 (5.3)
24 (9.1)
0.772
 Totally dependent
5 (1.7)
3 (1.8)
2 (1.5)
 
1 (2.2)
4 (1.6)
 
0 (0.0)
5 (1.9)
 
Smoking
83 (27.5)
43 (25.6)
40 (29.9)
0.410
12 (26.1)
71 (27.7)
0.817
9 (23.7)
74 (28.0)
0.574
Transfusion (<  24 h)
6 (2.0)
2 (1.2)
4 (3.0)
0.411
0 (0.0)
6 (2.3)
0.595
0 (0.0)
6 (2.3)
1.000
Respiratory infection (<  30 d)
24 (7.9)
17 (10.1)
7 (5.2)
0.118
2 (4.3)
22 (8.6)
0.551
0 (0.0)
24 (9.1)
0.054
Recent MV (<  30 d)
8 (2.6)
5 (3.0)
3 (2.2)
1.000
0 (0.0)
8 (3.1)
0.612
0 (0.0)
8 (3.0)
0.602
Co-morbidities
 COPD
54 (17.9)
35 (20.8)
19 (14.2)
0.133
6 (13.0)
48 (18.8)
0.352
1 (2.6)
53 (20.1)
0.005
 Apnea
5 (1.7)
1 (0.6)
4 (3.0)
0.174
1 (2.2)
4 (1.6)
0.564
0 (0.0)
5 (1.9)
1.000
 Liver cirrhosis
2 (0.7)
2 (1.2)
0 (0.0)
0.504
0 (0.0)
2 (0.8)
1.000
0 (0.0)
2 (0.8)
1.000
 Chronic kidney failure
7 (2.3)
5 (3.0)
2 (2.0)
0.468
1 (2.2)
6 (2.3)
1.000
0 (0.0)
7 (2.7)
0.601
 Heart failure
23 (7.6)
9 (5.4)
14 (10.4)
0.097
2 (4.3)
21 (8.2)
0.548
0 (0.0)
23 (8.7)
0.092
 Neuro disease
8 (2.6)
5 (5.0)
3 (2.2)
1.000
2 (4.3)
6 (2.3)
0.350
2 (5.3)
6 (2.3)
0.265
Laboratorial tests and vital signs
SpO2, %
97.0 (95.0–98.0)
97.0 (95.0–98.0)
97.0 (95.0–98.0)
0.347
98.0 (95.0–99.0)
97.0 (95.0–98.0)
0.232
98.0 (97.0–99.0)
97.0 (95.0–98.0)
< 0.001
Hemoglobin, g/dL
13.5 (12.4–14.8)
13.5 (12.4–14.5)
13.4 (12.5–15.0)
0.700
13.3 (12.9–14.6)
13.6 (12.3–14.8)
0.773
14.3 (13.3–15.2)
13.4 (12.3–14.6)
0.002
WBC, cell/mm3
7150.0 (6000.0–9000.0)
7000.0 (6000.0–9000.0)
7565.0 (6000.0–9150.0)
0.755
8000.0 (6000.0–9912.5)
7000.0 (6000.0–9000.0)
0.344
7000.0 (6000.0–8125.0)
7300.0 (6000.0–9000.0)
0.506
Surgical characteristics
Procedurea
 Vascular
2 (0.7)
0 (0.0)
2 (1.5)
0.196
0 (0.0)
2 (0.8)
1.000
0 (0.0)
2 (0.8)
1.000
 Cardiac
12 (4.0)
0 (0.0)
12 (9.0)
<  0.001
1 (2.2)
11 (4.3)
0.700
0 (0.0)
12 (4.5)
0.374
 Lung / Pleural
231 (76.5)
152 (90.5)
79 (59.0)
<  0.001
34 (73.9)
197 (77.0)
0.654
27 (71.1)
204 (77.3)
0.397
 Other
64 (21.2)
21 (12.5)
43 (32.1)
<  0.001
11 (23.9)
53 (20.7)
0.623
12 (31.6)
52 (19.7)
0.093
Condition
 Elective
283 (93.7)
160 (95.2)
123 (91.8)
0.454
41 (91.3)
241 (94.1)
0.148
36 (94.7)
247 (93.6)
 
 Urgency
13 (4.3)
5 (3.0)
8 (6.0)
 
4 (8.7)
9 (3.5)
 
2 (5.3)
11 (4.2)
0.856
 Emergency
6 (2.0)
3 (1.8)
3 (2.2)
 
0 (0.0)
6 (2.3)
 
0 (0.0)
6 (2.3)
 
Planned duration
  ≤ 2 h
136 (45.0)
73 (43.5)
63 (47.0)
0.427
28 (60.9)
108 (42.2)
0.061
30 (78.9)
106 (40.2)
 
 2–3 h
95 (31.5)
58 (34.5)
37 (27.6)
 
11 (23.9)
84 (32.8)
 
6 (15.8)
89 (33.7)
<  0.001
  > 3 h
71 (23.5)
37 (22.0)
34 (25.4)
 
7 (15.2)
64 (25.0)
 
2 (5.3)
69 (26.1)
 
Antibiotic prophylaxis
266 (88.1)
146 (86.9)
120 (89.6)
0.480
38 (82.6)
228 (89.1)
0.213
27 (71.1)
239 (90.5)
<  0.001
Epidural anesthesia
72 (23.8)
48 (28.6)
24 (17.9)
0.307
5 (10.9)
67 (26.2)
0.024
4 (10.5)
68 (25.8)
0.039
Type of tube
 Endotracheal
84 (27.8)
11 (6.5)
73 (54.5)
<  0.001
24 (52.2)
60 (23.4)
0.001
15 (39.5)
69 (26.1)
 
 Bronchus blocker
19 (6.3)
12 (7.1)
7 (5.2)
 
1 (2.2)
18 (7.0)
 
2 (5.3)
17 (6.4)
0.295
 SGA
6 (2.0)
2 (1.2)
4 (3.0)
 
0 (0.0)
6 (2.3)
 
1 (2.6)
5 (1.9)
 
 DLT
193 (63.9)
143 (85.1)
50 (37.3)
 
21 (45.7)
172 (67.2)
 
20 (52.6)
173 (65.5)
 
Duration of surgery, min
105.0 (55.0–174.2)
104.0 (57.5–164.8)
105.0 (55.0–180.0)
0.836
62.5 (45.0–131.2)
110.0 (62.2–180.0)
0.003
55.0 (41.2–80.0)
115.0 (62.2–180.8)
< 0.001
Duration of anesthesia, min
145.0 (90.0–225.0)
147.5 (97.8–225.0)
145.0 (90.0–235.0)
0.651
105.0 (80.0–153.8)
152.0 (100.0–236.0)
0.001
88.0 (64.5–120.0)
160.0 (100.0–240.0)
< 0.001
Values are presented as median (interquartile range) or number (percentage). p values from a Proportions χ2 or Fisher exact tests for proportions and Mann-Whitney U Test for continuous variables
ARISCAT: ASA American Society of Anesthesiology recommended physical status, BMI Body mass index, COPD Chronic obstructive pulmonary disease, DLT Double-lumen tube, MV Mechanical ventilation, OLV One-lung ventilation, SGA Supraglottic airway, SpO2 Pulse oximetry, TLV Total lung ventilation, WBC White blood count;
amore than one option allowed
Characteristics of patients undergoing procedures with OLV vs. TLV, and endoscopic vs. open were comparable. Patients with moderate-to-high risk for PPCs were different from those at low risk with respect to age, gender, BMI, ASA status, COPD prevalence and planned duration of surgery (Table 1).

Intra-operative characteristics

Patients operated under OLV received more often double-lumen tubes and had more frequently lung or pleural surgery than those operated under TLV (Table 1). Use of epidural anesthesia was less and duration of surgery shorter in endoscopic compared to non-endoscopic surgery (Table 1).
Patients at moderate-to-high risk for PPC received more frequently antibiotic prophylaxis and epidural anesthesia, and had longer duration of surgery as well as anesthesia, compared with patients at low risk (Table 1).
The amounts of crystalloids, colloids, albumin and packed red blood cells was higher in open vs. endoscopic surgery, and in patients at moderate-to-high vs. low risk for PPC (Table 2).
Table 2
Intra-Operative Characteristics of the Patients According to Subgroups
 
All Patients
(n = 302)
OLV
(n = 168)
TLV
(n = 134)
p value
Endoscopic
(n = 46)
Open
(n = 256)
p value
Low Risk
(n = 38)
High Risk
(n = 264)
p value
Ventilation and vital signs
Ventilatory mode
 Volume controlled
209 (70.8)
121 (73.3)
88 (67.7)
0.055
28 (65.1)
181 (71.8)
0.285
32 (91.4)
177 (68.1)
 
 Pressure controlled
51 (17.3)
28 (17.0)
23 (17.7)
 
7 (16.3)
44 (17.5)
 
2 (5.7)
49 (18.8)
 
 Pressure support
3 (1.2)
3 (1.8)
0 (0.0)
 
0 (0.0)
3 (1.2)
 
0 (0.0)
3 (1.2)
0.109
 Spontaneous
8 (2.7)
1 (0.6)
7 (5.4)
 
3 (7.0)
5 (2.0)
 
0 (0.0)
8 (3.1)
 
 Other
24 (8.1)
12 (7.3)
12 (9.2)
 
5 (11.6)
19 (7.5)
 
1 (2.9)
23 (8.8)
 
VT, ml
472.2 (400.5–525.0)
453.5 (398.4–510.0)
475.0 (430.6–549.0)
0.015
468.8 (400.0–544.5)
472.2 (405.6–525.0)
0.895
483.2 (443.1–543.9)
468.5 (400.0–525.0)
0.102
VT, ml/kg PBWa
7.6 (6.3–8.4)
7.4 (6.0–8.3)
7.6 (6.6–8.5)
0.050
7.6 (6.2–8.8)
7.5 (6.3–8.3)
0.225
7.6 (7.0–8.3)
7.5 (6.2–8.4)
0.587
Peak pressure, cmH2Oa
20.0 (17.5–24.0)
21.0 (18.0–25.0)
19.0 (16.0–23.0)
0.001
20.0 (16.2–23.0)
20.0 (18.0–24.0)
0.187
18.5 (15.6–22.0)
20.0 (18.0–24.1)
0.068
Plato pressure, cmH2Oa
17.8 (15.0–21.0)
18.2 (16.0–21.4)
16.5 (13.1–20.0)
0.004
16.0 (12.5–20.2)
18.0 (15.0–21.0)
0.118
15.8 (13.0–18.9)
18.0 (15.5–21.0)
0.010
PEEP, cmH2Oa
4.0 (1.5–5.0)
4.5 (2.4–5.0)
3.0 (1.5–5.0)
0.006
3.0 (1.5–5.0)
4.0 (2.0–5.0)
0.176
2.0 (0.0–5.0)
4.0 (2.0–5.0)
0.008
Driving pressure, cmH2Oa
14.0 (11.0–17.0)
14.5 (12.0–17.5)
13.0 (10.5–16.0)
0.016
13.0 (10.8–16.0)
14.0 (11.5–17.0)
0.389
14.0 (10.5–16.0)
14.0 (11.5–17.0)
0.216
Respiratory rate, bpm
12.0 (12.0–14.0)
12.0 (12.0–15.0)
12.0 (12.0–14.0)
0.128
12.0 (11.1–14.4)
12.0 (12.0–14.0)
0.715
12.0 (12.0–13.5)
12.0 (12.0–14.5)
0.644
FiO2, %
65.0 (50.0–80.0)
65.8 (50.0–85.0)
63.5 (50.0–80.0)
0.294
70.0 (58.5–83.6)
64.5 (50.0–80.0)
0.151
68.2 (50.0–75.0)
65.0 (50.0–84.0)
0.543
SpO2, %
99.0 (97.5–100.0)
98.5 (97.0–100.0)
99.0 (98.0–100.0)
0.039
99.0 (98.0–99.9)
99.0 (97.5–100.0)
0.724
99.0 (98.0–100.0)
99.0 (97.5–100.0)
0.352
MAP, mmHg
78.0 (71.0–86.6)
78.0 (71.0–86.0)
78.0 (71.0–88.0)
0.940
80.8 (74.0–95.0)
77.5 (70.0–85.0)
0.009
77.5 (71.5–91.6)
78.0 (71.0–86.0)
0.474
Heart rate, bpm
73.5 (64.5–82.0)
73.5 (64.5–82.5)
73.5 (65.0–81.1)
0.893
73.2 (65.2–79.0)
74.0 (64.5–82.0)
0.663
75.0 (70.0–81.5)
73.5 (63.6–82.0)
0.223
RM
105 (34.8)
71 (42.3)
34 (25.4)
0.002
13 (28.3)
92 (35.9)
0.314
7 (18.4)
98 (37.1)
0.023
 In the last hour
68 (22.5)
52 (31.0)
16 (11.9)
<  0.001
6 (13.0)
62 (24.2)
0.094
2 (5.3)
66 (25.0)
0.006
Number of RM
0.0 (0.0–1.0)
0.0 (0.0–1.0)
0.0 (0.0–0.8)
0.003
0.0 (0.0–1.0)
0.0 (0.0–1.0)
0.397
0.0 (0.0–0.0)
0.0 (0.0–1.0)
0.020
Protective ventilation
41 (14.8)
13 (8.3)
28 (23.5)
<  0.001
10 (24.4)
31 (13.2)
0.091
4 (11.4)
37 (15.4)
0.798
Anesthesia characteristics
Type of anesthesia
 TIVA
56 (18.5)
25 (14.9)
31 (23.1)
0.012
10 (21.7)
46 (18.0)
0.287
6 (15.8)
50 (18.9)
 
 Volatile
188 (62.3)
117 (69.6)
71 (53.0)
 
31 (67.4)
157 (61.3)
 
22 (57.9)
166 (62.9)
0.483
 Mixed
58 (19.2)
26 (15.5)
32 (23.9)
 
5 (10.9)
53 (20.7)
 
10 (26.3)
48 (18.2)
 
Opioids
 Short acting
66 (21.9)
39 (23.2)
27 (20.3)
0.151
14 (30.4)
52 (20.4)
0.306
11 (28.9)
55 (20.9)
 
 Long acting
202 (66.9)
106 (63.1)
96 (72.2)
 
28 (60.9)
174 (68.2)
 
24 (63.2)
178 (67.7)
0.475
 Both
33 (10.9)
23 (13.7)
10 (7.5)
 
4 (8.7)
29 (11.4)
 
3 (7.9)
30 (11.4)
 
Total Fluids
 Crystalloids, ml
1000.0 (875.0–2000.0)
1000.0 (1000.0–1750.0)
1000.0 (800.0–2000.0)
0.949
900.0 (500.0–1100.0)
1130.0 (1000.0–2000.0)
< 0.001
1000.0 (670.0–1000.0)
1100.0 (1000.0–2000.0)
< 0.001
 Colloids, ml
500.0 (67.5–700.0)
500.0 (0.0–500.0)
500.0 (500.0–1000.0)
0.076
0.0 (0.0–500.0)
500.0 (450.0–850.0)
0.027
0.0 (0.0–500.0)
500.0 (500.0–1000.0)
0.007
 Albumin, ml
0.0 (0.0–0.0)
0.0 (0.0–0.0)
0.0 (0.0–12.2)
0.755
0.0 (0.0–0.0)
0.0 (0.0–0.0)
0.212
0.0 (0.0–0.0)
0.0 (0.0–0.0)
0.134
 PRBC, units
0.0 (0.0–2.0)
0.0 (0.0–1.0)
1.0 (0.0–2.0)
0.162
0.0 (0.0–0.0)
0.0 (0.0–2.0)
0.045
0.0 (0.0–0.0)
0.0 (0.0–2.0)
0.017
Reversal of NMBA
115 (38.1)
72 (43.1)
43 (32.1)
0.050
16 (34.8)
99 (38.8)
0.603
14 (36.8)
101 (38.4)
0.853
Values are presented as median (interquartile range) or number (percentage). p values from a Proportions χ2 or Fisher exact tests for proportions and Mann-Whitney U Test for continuous variables bpm beats per minute, etCO2 End-tidal carbon dioxide, FiO2 Inspired fraction of oxygen, MAP Mean arterial pressure, mpm Movements per minute, NMBA Neuromuscular blocking agents, OLV One lung ventilation, PBW Predicted body weight, PEEP Positive end-expiratory pressure, PRBC Packed red blood cells, RM Recruitment maneuver, SpO2 Pulse oximetry, TIVA Total intravenous anesthesia, TLV Total lung ventilation, VT Tidal volume
adata presented as the median used through surgery

Mechanical ventilation

Patients were ventilated with VT of 7.4 ± 1.6 ml/kg PBW, PEEP of 3.5 ± 2.4 cmH2O, and driving pressure of 14.4 ± 4.6 cmH2O (Table 2). Compared to patients operated solely under TLV, patients receiving OLV had lower VT, higher peak, plateau and driving pressures, as well as PEEP and respiratory rate, and received higher number of recruitment maneuvers (Table 2). Protective ventilation was used in 14.8% (41/302) of all patients, mainly during TLV. The ventilatory management of patients undergoing endoscopic and non-endoscopic procedures did not differ significantly. Patients at moderate-to-high risk for PPC had higher levels of PEEP, and received more recruitment maneuvers than patients at low risk (Table 2).
Values of ventilator settings along time are shown in Supplemental Figures 2 through 4 (Additional file 1). Patients operated under OLV had higher FiO2 compared with patients operated under TLV (Supplemental Figure 2, Additional file 1). The combinations of VT and PEEP according to subgroups are shown in Supplemental Figures 5 through 7 (Additional file 1).

Primary outcome

The overall incidence of PPCs in this population was 45.7% (138/302), and did not differ significantly between OLV vs. TLV (82/168 vs. 56/134, 48.8% vs. 41.8%, p = 0.223, total number and percentage respectively), and endoscopic vs. open procedures (16/46 vs. 122/256, 34.8% vs. 47.7%, p = 0.106, total number and percentage respectively, Table 3, Fig. 1). Patients at moderate-to-high risk showed an increased incidence of PPC compared to patients at lower risk (48.1% vs. 28.9%; hazard ratio, 1.95; 95% CI 1.05–3.61; p = 0.033), mainly due to unplanned need for supplemental oxygen (Table 3, Fig. 1).
Table 3
Clinical Outcomes of the Patients According to Subgroups
 
All Patients
(n = 302)
OLV
(n = 168)
TLV
(n = 134)
p value
Endoscopic
(n = 46)
Open
(n = 256)
p value
Low Risk
(n = 38)
High Risk
(n = 264)
p value
Primary outcome
PPC
138 (45.7)
82 (48.8)
56 (41.8)
0.223
16 (34.8)
122 (47.7)
0.106
11 (28.9)
127 (48.1)
0.026
 Need of oxygen
109 (36.1)
65 (38.9)
44 (32.8)
0.274
12 (26.1)
97 (38.0)
0.120
8 (21.1)
101 (38.4)
0.037
 Respiratory failure
26 (8.6)
15 (8.9)
11 (8.2)
0.824
1 (2.2)
25 (9.8)
0.148
2 (5.3)
24 (9.1)
0.755
 Invasive MV
26 (8.6)
14 (8.3)
12 (9.0)
0.848
3 (6.5)
23 (9.0)
0.778
1 (2.6)
25 (9.5)
0.222
 NIV
15 (5.0)
10 (6.0)
5 (3.7)
0.377
2 (4.3)
13 (5.1)
1.000
1 (2.6)
14 (5.3)
0.702
 ARDS
4 (1.3)
4 (2.4)
0 (0.0)
0.132
0 (0.0)
4 (1.6)
1.000
0 (0.0)
4 (1.5)
1.000
 Pneumonia
8 (2.6)
6 (3.6)
2 (1.5)
0.307
2 (4.3)
6 (2.3)
0.350
1 (2.6)
7 (2.7)
1.000
Secondary outcomes
Severe PPCa
53 (17.5)
30 (17.9)
23 (17.2)
0.875
7 (15.2)
46 (18.0)
0.651
4 (10.5)
49 (18.6)
0.223
Intra-OP complications
 Desaturation
61 (20.2)
38 (22.6)
23 (17.2)
0.240
7 (15.2)
54 (21.1)
0.360
5 (13.2)
56 (21.2)
0.247
 Unplanned RM
48 (15.9)
31 (18.5)
17 (12.7)
0.173
7 (15.2)
41 (16.0)
0.891
3 (7.9)
45 (17.0)
0.149
 Pressure reduction
36 (11.9)
27 (16.1)
9 (6.7)
0.012
5 (10.9)
31 (12.1)
0.811
2 (5.3)
34 (12.9)
0.281
 Flow limitation
3 (1.0)
2 (1.2)
1 (0.8)
1.000
0 (0.0)
3 (1.2)
1.000
0 (0.0)
3 (1.1)
1.000
 Hypotension
102 (33.8)
60 (35.7)
42 (31.3)
0.424
8 (17.4)
94 (36.7)
0.010
3 (7.9)
99 (37.5)
<  0.001
 Vasopressors
113 (37.4)
65 (38.7)
48 (35.8)
0.608
13 (28.3)
100 (39.1)
0.163
3 (7.9)
110 (41.7)
<  0.001
 New arrhythmias
6 (2.0)
3 (1.8)
3 (2.2)
1.000
0 (0.0)
6 (2.3)
0.595
0 (0.0)
6 (2.3)
1.000
ICU admissionb
6 (2.0)
2 (1.2)
4 (3.0)
0.411
0 (0.0)
6 (2.3)
0.595
0 (0.0)
6 (2.3)
1.000
Hospital LOS, days
6.0 (3.0–10.0)
6.0 (4.0–11.0)
5.0 (3.0–9.0)
0.010c
3.0 (1.0–7.5)
6.0 (4.0–10.0)
< 0.001c
4.0 (1.0–6.0)
6.0 (4.0–10.0)
< 0.001c
Hospital mortality
1 (0.3)
1 (0.6)
0 (0.0)
1.000
0 (0.0)
1 (0.4)
1.000
0 (0.0)
1 (0.4)
1.000
Values are presented as median (interquartile range) or number (percentage). p values from a Proportions χ2 or Fisher exact tests for proportions and Mann-Whitney U Test for continuous variables ARDS Acute respiratory distress syndrome, ICU Intensive care unit, Intra-OP Intraoperative, LOS Length of stay, MV Mechanical ventilation, NIV Non-invasive ventilation, OLV One lung ventilation, PPC Postoperative pulmonary complication, RM Recruitment maneuvers, TLV Total lung ventilation
aexcluding need of oxygen
bunplanned admission
cp value from the Cox proportional hazard model

Secondary outcomes

The incidence of severe PPCs, unplanned ICU admission and hospital mortality did not differ among groups (Table 3). The incidence of hypotension was decreased in endoscopic compared to open procedures, and in patients at lower compared to moderate-to-high risk of PPCs (Table 3).
The LOS was increased in patients who developed PPCs (Supplemental Figure 8, Additional file 1), and shorter in patients operated under OLV vs. TLV, endoscopic vs. open, and those with low vs. moderate-to-high risk for PPC (Table 3, Fig. 2).

Discussion

In this population of patients undergoing thoracic surgery: 1) mechanical ventilation differed from those recommended for lung protection in 85.2% of all patients; 2) patients under OLV received lower VT, higher peak, plateau and driving pressures, higher PEEP levels and respiratory rate, and received more recruitment maneuvers compared with TLV; 3) the overall incidence of PPCs was as high as 45.7%; 4) PPCs were more common among patients with higher ARISCAT score or co-morbidities, but not increased following open vs. endoscopic procedures, or OLV vs. TLV; 6) PPCs were associated with increased LOS.
To our knowledge, this is the first prospective observational investigation addressing the practice of mechanical ventilation and incidence of PPCs in thoracic anesthesia. The main strengths of our study are that data was stored, analyzed and reported according to international standards [21].
High VT strategies, usually accompanied by low or zero PEEP, have been used to prevent intraoperative atelectasis [22, 23]. However, this may cause overdistension (volutrauma), and repetitive collapse-reopening of lung units (atelectrauma), which can injure the lungs and lead to PPCs [24]. A protective ventilation approach consisting mainly of low VT reduces the incidence of PPCs [7, 25]. This seems to apply also to thoracic anesthesia but this claim is not undisputed [2628]. The present study shows that protective mechanical ventilation, as recommended, was used in less than 15% of patients undergoing thoracic surgery. Different possible reasons might explain this finding: 1) the concept of protective ventilation during surgery is still not widespread among anesthesiologists; 2) the role of single components of mechanical ventilation in lung protection, especially of PEEP, is still poorly defined, leading anesthesiologists to set values according to their own preferences; 3) sound evidence from large RCTs demonstrating the benefit of protective mechanical ventilation in thoracic surgical patients is still missing; 4) thoracic surgical procedures usually last less than 1 hour, which might be deemed as too short to benefit from protective mechanical ventilation; 5) mechanical ventilation settings guided by driving pressure may result in VT and PEEP outside the range that has been recommended for protective mechanical ventilation.
The incidence of PPCs after surgery is influenced by patient-related factors, and type of surgery. In a mixed surgical population without surgery involving cardiopulmonary bypass, 10.4% of patients developed PPCs within the first postoperative 5 days; values ranged from 6.7% in plastic/cutaneous procedures to 38.2% in transplant surgery [3]. In open abdominal surgery, PPCs were reported in 10.5 to 39.0% of patients, despite the use of a protective ventilation strategy [3, 7, 25]. In average, 10.7% of patients at increased risk, for example obese patients, developed PPCs [29]. In patients undergoing thoracic surgery, an incidence of PPCs between 10.7 and 50% has been reported [26, 3032]. This relatively wide range is possibly explained by differences in definition of pulmonary complications among trials. The rate of severe PPCs was 17.5% in our thoracic surgery population, which is comparable to the rate of 18.1% reported by Blank and colleagues [26].
The observation that patients who developed PPCs had more comorbidities and longer LOS is in line with previous studies addressing intraoperative TLV [3, 33]. The difference in LOS in the subgroups is likely explained by the type of procedure per se, where open approaches require a prolonged treatment due to more complex procedures, independent from the type of mechanical ventilation.
Although the incidence of PPCs was relatively high, neither open thoracic surgery procedures, nor OLV itself were associated with them, especially when taking the infrequent use of protective mechanical ventilation in this population into account. The precise role of PEEP for protective intraoperative mechanical ventilation has been challenged in recent trials [7, 34]. In fact, it has been suggested that a strategy aimed at permissive atelectasis might be as protective as a strategy to open lungs during surgery [14, 35]. Our finding that higher VT was not associated with PPCs is intriguingly, but in agreement with data from an observational study reporting that the use of VT as high as 8 mL/kg as even associated with better pulmonary outcome [26]. Together, these findings suggest that protective OLV settings are more complex than previously thought. Cutoff values, although valuable, must not only consider the interaction among variables, but also a possible role of airway pressures.

Limitations

This study has several limitations. First, a one-week inclusion period was relatively short in order to include a high number of patients per center. However, this fact was counterbalanced by the multicenter design. Second, a short inclusion period might have resulted in selection bias, since fluctuation of the severity of cases cannot be ruled out. Nevertheless, the benefits of avoiding changes in therapy during the observation period as a potential confounder should not be underestimated. Third, the definition of protective mechanical ventilation was based on recommendations that are still under debate. Fourth, most study sites included less than 10 patients. This number, however, does not imply lack of experience with the procedure, since thoracic anesthesia per se already requires a substantial degree of expertise. Fifth, the duration of OLV was not investigated and, therefore, the exact contribution of OLV to PPCs cannot be separated from the period under TLV in this sub-population. Sixth, the design of this study precludes the possibility of determining cause-effect relationships, and results must be seen from a hypothesis-generating perspective. Seventh, the fact that data was collected prospectively might have interfered with clinical practice itself, and biased towards the use of protective ventilation. Still, non-protective ventilation was used in a vast majority of patients. Eighth, the total number of patients enrolled allowed analyses of three subgroups only. Potential confounders could be the type of anesthesia (total intravenous anesthesia vs. volatile anesthetics), the type of postoperative analgesia (epidural anesthesia vs. opioids) or the ASA status, which should be subject of future trials.

Conclusions

The present study provides relevant insight into the practice of mechanical ventilation during thoracic surgery. The data might prove useful for the development of scores for risk prediction in this particular population, allocation of human and financial resources, including need for postoperative monitoring in dedicated units, and also estimation of sample size in interventional trials [18]. Mechanical ventilation practice did not follow current recommendations for lung protection in the vast majority of patients undergoing thoracic surgery. Although PPCs were common in this population, and associated with increased LOS, their incidence was not higher following open vs. endoscopic or OLV vs. TLV, and not associated with mechanical ventilation settings. It must be emphasized that the lack of association between mechanical ventilation settings and PPCs does not support use of non-protective VT and PEEP in this population.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12871-020-01098-4.

Acknowledgements

The authors thank the European Society of Anaesthesiology (ESA) for co–sponsoring and endorsement as well as assistance in developing and hosting of the electronic case record forms, database and recruiting study sites.
The LAS VEGAS Study Collaborators
Collaborators are listed in the supplemental material (Additional file 1, pp. 2–5).
The trial protocol was first approved by the institutional review board of the Academic Medical Center, Amsterdam, The Netherlands (W12_190#12.17.0227). Written informed consent was obtained from all participants prior to trial enrollment.
Not applicable.

Competing interests

All 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 Weiser TG, Regenbogen SE, Thompson KD, Haynes AB, Lipsitz SR, Berry WR, Gawande AA. An estimation of the global volume of surgery: a modelling strategy based on available data. Lancet. 2008;372(9633):139–44.CrossRef Weiser TG, Regenbogen SE, Thompson KD, Haynes AB, Lipsitz SR, Berry WR, Gawande AA. An estimation of the global volume of surgery: a modelling strategy based on available data. Lancet. 2008;372(9633):139–44.CrossRef
2.
Zurück zum Zitat Bainbridge D, Martin J, Arango M, Cheng D, Evidence-based Peri-operative clinical outcomes research G. Perioperative and anaesthetic-related mortality in developed and developing countries: a systematic review and meta-analysis. Lancet. 2012;380(9847):1075–81.CrossRef Bainbridge D, Martin J, Arango M, Cheng D, Evidence-based Peri-operative clinical outcomes research G. Perioperative and anaesthetic-related mortality in developed and developing countries: a systematic review and meta-analysis. Lancet. 2012;380(9847):1075–81.CrossRef
3.
Zurück zum Zitat Investigators LV. Epidemiology, practice of ventilation and outcome for patients at increased risk of postoperative pulmonary complications: LAS VEGAS - an observational study in 29 countries. Eur J Anaesthesiol. 2017;34(8):492–507.CrossRef Investigators LV. Epidemiology, practice of ventilation and outcome for patients at increased risk of postoperative pulmonary complications: LAS VEGAS - an observational study in 29 countries. Eur J Anaesthesiol. 2017;34(8):492–507.CrossRef
4.
Zurück zum Zitat Senturk M. New concepts of the management of one-lung ventilation. Curr Opin Anaesthesiol. 2006;19(1):1–4.CrossRef Senturk M. New concepts of the management of one-lung ventilation. Curr Opin Anaesthesiol. 2006;19(1):1–4.CrossRef
5.
Zurück zum Zitat Severgnini P, Selmo G, Lanza C, Chiesa A, Frigerio A, Bacuzzi A, Dionigi G, Novario R, Gregoretti C, de Abreu MG, et al. Protective mechanical ventilation during general anesthesia for open abdominal surgery improves postoperative pulmonary function. Anesthesiology. 2013;118(6):1307–21.CrossRef Severgnini P, Selmo G, Lanza C, Chiesa A, Frigerio A, Bacuzzi A, Dionigi G, Novario R, Gregoretti C, de Abreu MG, et al. Protective mechanical ventilation during general anesthesia for open abdominal surgery improves postoperative pulmonary function. Anesthesiology. 2013;118(6):1307–21.CrossRef
6.
Zurück zum Zitat Hemmes SN, Serpa Neto A, Schultz MJ. Intraoperative ventilatory strategies to prevent postoperative pulmonary complications: a meta-analysis. Curr Opin Anaesthesiol. 2013;26(2):126–33.CrossRef Hemmes SN, Serpa Neto A, Schultz MJ. Intraoperative ventilatory strategies to prevent postoperative pulmonary complications: a meta-analysis. Curr Opin Anaesthesiol. 2013;26(2):126–33.CrossRef
7.
Zurück zum Zitat PROVE Network Investigators for the Clinical Trial Network of the European Society of Anaesthesiology; Sabrine N T Hemmes, Marcelo Gama de Abreu, Pelosi P, Schultz MJ. High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet. 2014;384(9942):495–503. PROVE Network Investigators for the Clinical Trial Network of the European Society of Anaesthesiology; Sabrine N T Hemmes, Marcelo Gama de Abreu, Pelosi P, Schultz MJ. High versus low positive end-expiratory pressure during general anaesthesia for open abdominal surgery (PROVHILO trial): a multicentre randomised controlled trial. Lancet. 2014;384(9942):495–503.
8.
Zurück zum Zitat Young CC, Harris EM, Vacchiano C, Bodnar S, Bukowy B, Elliott RRD, Migliarese J, Ragains C, Trethewey B, Woodward A, et al. Lung-protective ventilation for the surgical patient: international expert panel-based consensus recommendations. Br J Anaesth. 2019;123(6):898–913.CrossRef Young CC, Harris EM, Vacchiano C, Bodnar S, Bukowy B, Elliott RRD, Migliarese J, Ragains C, Trethewey B, Woodward A, et al. Lung-protective ventilation for the surgical patient: international expert panel-based consensus recommendations. Br J Anaesth. 2019;123(6):898–913.CrossRef
9.
Zurück zum Zitat Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ. ESA clinical trials network 2012: LAS VEGAS--local assessment of Ventilatory management during general Anaesthesia for surgery and its effects on postoperative pulmonary complications: a prospective, observational, international, multicentre cohort study. Eur J Anaesthesiol. 2013;30(5):205–7.CrossRef Hemmes SN, de Abreu MG, Pelosi P, Schultz MJ. ESA clinical trials network 2012: LAS VEGAS--local assessment of Ventilatory management during general Anaesthesia for surgery and its effects on postoperative pulmonary complications: a prospective, observational, international, multicentre cohort study. Eur J Anaesthesiol. 2013;30(5):205–7.CrossRef
10.
Zurück zum Zitat Arozullah AM, Daley J, Henderson WG, Khuri SF. Multifactorial risk index for predicting postoperative respiratory failure in men after major noncardiac surgery. The National Veterans Administration Surgical Quality Improvement Program. Ann Surg. 2000;232(2):242–53.CrossRef Arozullah AM, Daley J, Henderson WG, Khuri SF. Multifactorial risk index for predicting postoperative respiratory failure in men after major noncardiac surgery. The National Veterans Administration Surgical Quality Improvement Program. Ann Surg. 2000;232(2):242–53.CrossRef
11.
Zurück zum Zitat Arozullah AM, Khuri SF, Henderson WG, Daley J, Participants in the National Veterans Affairs Surgical Quality Improvement P. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Ann Intern Med. 2001;135(10):847–57.CrossRef Arozullah AM, Khuri SF, Henderson WG, Daley J, Participants in the National Veterans Affairs Surgical Quality Improvement P. Development and validation of a multifactorial risk index for predicting postoperative pneumonia after major noncardiac surgery. Ann Intern Med. 2001;135(10):847–57.CrossRef
12.
Zurück zum Zitat Canet J, Gallart L, Gomar C, Paluzie G, Valles J, Castillo J, Sabate S, Mazo V, Briones Z, Sanchis J, et al. Prediction of postoperative pulmonary complications in a population-based surgical cohort. Anesthesiology. 2010;113(6):1338–50.CrossRef Canet J, Gallart L, Gomar C, Paluzie G, Valles J, Castillo J, Sabate S, Mazo V, Briones Z, Sanchis J, et al. Prediction of postoperative pulmonary complications in a population-based surgical cohort. Anesthesiology. 2010;113(6):1338–50.CrossRef
13.
Zurück zum Zitat Smetana GW. Preoperative pulmonary evaluation: identifying and reducing risks for pulmonary complications. Cleve Clin J Med. 2006;73(Suppl 1):S36–41.CrossRef Smetana GW. Preoperative pulmonary evaluation: identifying and reducing risks for pulmonary complications. Cleve Clin J Med. 2006;73(Suppl 1):S36–41.CrossRef
14.
Zurück zum Zitat Guldner A, Kiss T, Serpa Neto A, Hemmes SN, Canet J, Spieth PM, Rocco PR, Schultz MJ, Pelosi P, Gama de Abreu M. Intraoperative protective mechanical ventilation for prevention of postoperative pulmonary complications: a comprehensive review of the role of tidal volume, positive end-expiratory pressure, and lung recruitment maneuvers. Anesthesiology. 2015;123(3):692–713.CrossRef Guldner A, Kiss T, Serpa Neto A, Hemmes SN, Canet J, Spieth PM, Rocco PR, Schultz MJ, Pelosi P, Gama de Abreu M. Intraoperative protective mechanical ventilation for prevention of postoperative pulmonary complications: a comprehensive review of the role of tidal volume, positive end-expiratory pressure, and lung recruitment maneuvers. Anesthesiology. 2015;123(3):692–713.CrossRef
15.
Zurück zum Zitat Serpa Neto A, Hemmes SN, Barbas CS, Beiderlinden M, Biehl M, Binnekade JM, Canet J, Fernandez-Bustamante A, Futier E, Gajic O, et al. Protective versus conventional ventilation for surgery: a systematic review and individual patient data Meta-analysis. Anesthesiology. 2015;123(1):66–78.CrossRef Serpa Neto A, Hemmes SN, Barbas CS, Beiderlinden M, Biehl M, Binnekade JM, Canet J, Fernandez-Bustamante A, Futier E, Gajic O, et al. Protective versus conventional ventilation for surgery: a systematic review and individual patient data Meta-analysis. Anesthesiology. 2015;123(1):66–78.CrossRef
16.
Zurück zum Zitat Futier E, Marret E, Jaber S. Perioperative positive pressure ventilation: an integrated approach to improve pulmonary care. Anesthesiology. 2014;121(2):400–8.CrossRef Futier E, Marret E, Jaber S. Perioperative positive pressure ventilation: an integrated approach to improve pulmonary care. Anesthesiology. 2014;121(2):400–8.CrossRef
17.
Zurück zum Zitat Slutsky AS, Ranieri VM. Ventilator-induced lung injury. N Engl J Med. 2013;369(22):2126–36.CrossRef Slutsky AS, Ranieri VM. Ventilator-induced lung injury. N Engl J Med. 2013;369(22):2126–36.CrossRef
18.
Zurück zum Zitat Kiss T, Wittenstein J, Becker C, Birr K, Cinnella G, Cohen E, El Tahan MR, Falcao LF, Gregoretti C, Granell M, et al. Protective ventilation with high versus low positive end-expiratory pressure during one-lung ventilation for thoracic surgery (PROTHOR): study protocol for a randomized controlled trial. Trials. 2019;20(1):213.CrossRef Kiss T, Wittenstein J, Becker C, Birr K, Cinnella G, Cohen E, El Tahan MR, Falcao LF, Gregoretti C, Granell M, et al. Protective ventilation with high versus low positive end-expiratory pressure during one-lung ventilation for thoracic surgery (PROTHOR): study protocol for a randomized controlled trial. Trials. 2019;20(1):213.CrossRef
19.
Zurück zum Zitat Marret E, Cinotti R, Berard L, Piriou V, Jobard J, Barrucand B, Radu D, Jaber S, Bonnet F, the PPVsg. Protective ventilation during anaesthesia reduces major postoperative complications after lung cancer surgery: a double-blind randomised controlled trial. Eur J Anaesthesiol. 2018;35(10):727–35.CrossRef Marret E, Cinotti R, Berard L, Piriou V, Jobard J, Barrucand B, Radu D, Jaber S, Bonnet F, the PPVsg. Protective ventilation during anaesthesia reduces major postoperative complications after lung cancer surgery: a double-blind randomised controlled trial. Eur J Anaesthesiol. 2018;35(10):727–35.CrossRef
20.
Zurück zum Zitat Lohser J, Slinger P. Lung injury after one-lung ventilation: a review of the pathophysiologic mechanisms affecting the ventilated and the collapsed lung. Anesth Analg. 2015;121(2):302–18.CrossRef Lohser J, Slinger P. Lung injury after one-lung ventilation: a review of the pathophysiologic mechanisms affecting the ventilated and the collapsed lung. Anesth Analg. 2015;121(2):302–18.CrossRef
21.
Zurück zum Zitat von Elm E, Altman DG, Egger M, Pocock SJ, Gotzsche PC, Vandenbroucke JP, Initiative S. The strengthening the reporting of observational studies in epidemiology (STROBE) statement: guidelines for reporting observational studies. Int J Surg. 2014;12(12):1495–9.CrossRef von Elm E, Altman DG, Egger M, Pocock SJ, Gotzsche PC, Vandenbroucke JP, Initiative S. The strengthening the reporting of observational studies in epidemiology (STROBE) statement: guidelines for reporting observational studies. Int J Surg. 2014;12(12):1495–9.CrossRef
22.
Zurück zum Zitat Bendixen HH, Hedley-Whyte J, Laver MB. Impaired oxygenation in surgical patients during general anesthesia with controlled ventilation. A concept of atelectasis. N Engl J Med. 1963;269:991–6.CrossRef Bendixen HH, Hedley-Whyte J, Laver MB. Impaired oxygenation in surgical patients during general anesthesia with controlled ventilation. A concept of atelectasis. N Engl J Med. 1963;269:991–6.CrossRef
23.
Zurück zum Zitat Benumof J. Conventional and differential lung management of one-lung ventilation. In: Anesthesia for thoracic surgery. Philadelphia: Saunders, W.B; 1995. p. 799. Benumof J. Conventional and differential lung management of one-lung ventilation. In: Anesthesia for thoracic surgery. Philadelphia: Saunders, W.B; 1995. p. 799.
24.
Zurück zum Zitat Slinger P. Perioperative lung injury. Best Pract Res Clin Anaesthesiol. 2008;22(1):177–91.CrossRef Slinger P. Perioperative lung injury. Best Pract Res Clin Anaesthesiol. 2008;22(1):177–91.CrossRef
25.
Zurück zum Zitat Futier E, Constantin JM, Paugam-Burtz C, Pascal J, Eurin M, Neuschwander A, Marret E, Beaussier M, Gutton C, Lefrant JY, et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. N Engl J Med. 2013;369(5):428–37.CrossRef Futier E, Constantin JM, Paugam-Burtz C, Pascal J, Eurin M, Neuschwander A, Marret E, Beaussier M, Gutton C, Lefrant JY, et al. A trial of intraoperative low-tidal-volume ventilation in abdominal surgery. N Engl J Med. 2013;369(5):428–37.CrossRef
26.
Zurück zum Zitat Blank RS, Colquhoun DA, Durieux ME, Kozower BD, McMurry TL, Bender SP, Naik BI. Management of one-lung Ventilation: impact of tidal volume on complications after thoracic surgery. Anesthesiology. 2016;124(6):1286–95.CrossRef Blank RS, Colquhoun DA, Durieux ME, Kozower BD, McMurry TL, Bender SP, Naik BI. Management of one-lung Ventilation: impact of tidal volume on complications after thoracic surgery. Anesthesiology. 2016;124(6):1286–95.CrossRef
27.
Zurück zum Zitat Colquhoun DA, Naik BI, Durieux ME, Shanks AM, Kheterpal S, Bender SP, Blank RS, Investigators M. Management of 1-lung ventilation-variation and trends in clinical practice: a report from the multicenter perioperative outcomes group. Anesth Analg. 2018;126(2):495–502.CrossRef Colquhoun DA, Naik BI, Durieux ME, Shanks AM, Kheterpal S, Bender SP, Blank RS, Investigators M. Management of 1-lung ventilation-variation and trends in clinical practice: a report from the multicenter perioperative outcomes group. Anesth Analg. 2018;126(2):495–502.CrossRef
28.
Zurück zum Zitat Blank RS, Lesh RE. Low tidal volume ventilation in the surgical patient: not particularly low and perhaps not particularly protective. Anesth Analg. 2019;128(4):831–3.CrossRef Blank RS, Lesh RE. Low tidal volume ventilation in the surgical patient: not particularly low and perhaps not particularly protective. Anesth Analg. 2019;128(4):831–3.CrossRef
29.
Zurück zum Zitat Ball L, Hemmes SNT, Serpa Neto A, Bluth T, Canet J, Hiesmayr M, Hollmann MW, Mills GH, Vidal Melo MF, Putensen C, et al. Intraoperative ventilation settings and their associations with postoperative pulmonary complications in obese patients. Br J Anaesth. 2018;121(4):899–908.CrossRef Ball L, Hemmes SNT, Serpa Neto A, Bluth T, Canet J, Hiesmayr M, Hollmann MW, Mills GH, Vidal Melo MF, Putensen C, et al. Intraoperative ventilation settings and their associations with postoperative pulmonary complications in obese patients. Br J Anaesth. 2018;121(4):899–908.CrossRef
30.
Zurück zum Zitat Baudouin SV. Lung injury after thoracotomy. Br J Anaesth. 2003;91(1):132–42.CrossRef Baudouin SV. Lung injury after thoracotomy. Br J Anaesth. 2003;91(1):132–42.CrossRef
31.
Zurück zum Zitat Cao C, Louie BE, Melfi F, Veronesi G, Razzak R, Romano G, Novellis P, Ranganath NK, Park BJ. Impact of pulmonary function on pulmonary complications after robotic-assisted thoracoscopic lobectomy. Eur J Cardiothorac Surg. 2020; 57(2):338–342. Cao C, Louie BE, Melfi F, Veronesi G, Razzak R, Romano G, Novellis P, Ranganath NK, Park BJ. Impact of pulmonary function on pulmonary complications after robotic-assisted thoracoscopic lobectomy. Eur J Cardiothorac Surg. 2020; 57(2):338–342.
32.
Zurück zum Zitat Im Y, Park HY, Shin S, Shin SH, Lee H, Ahn JH, Sohn I, Cho JH, Kim HK, Zo JI, et al. Prevalence of and risk factors for pulmonary complications after curative resection in otherwise healthy elderly patients with early stage lung cancer. Respir Res. 2019;20(1):136.CrossRef Im Y, Park HY, Shin S, Shin SH, Lee H, Ahn JH, Sohn I, Cho JH, Kim HK, Zo JI, et al. Prevalence of and risk factors for pulmonary complications after curative resection in otherwise healthy elderly patients with early stage lung cancer. Respir Res. 2019;20(1):136.CrossRef
33.
Zurück zum Zitat Serpa Neto A, Hemmes SN, Barbas CS, Beiderlinden M, Fernandez-Bustamante A, Futier E, Hollmann MW, Jaber S, Kozian A, Licker M, et al. Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and meta-analysis. Lancet Respir Med. 2014;2(12):1007–15.CrossRef Serpa Neto A, Hemmes SN, Barbas CS, Beiderlinden M, Fernandez-Bustamante A, Futier E, Hollmann MW, Jaber S, Kozian A, Licker M, et al. Incidence of mortality and morbidity related to postoperative lung injury in patients who have undergone abdominal or thoracic surgery: a systematic review and meta-analysis. Lancet Respir Med. 2014;2(12):1007–15.CrossRef
34.
Zurück zum Zitat Writing Committee for the PCGotPVNftCTNotESoA, Bluth T, Serpa Neto A, Schultz MJ, Pelosi P, Gama de Abreu M. Effect of Intraoperative High Positive End-Expiratory Pressure (PEEP) With Recruitment Maneuvers vs Low PEEP on Postoperative Pulmonary Complications in Obese Patients: A Randomized Clinical Trial. JAMA. 2019;321(23):2292–305.CrossRef Writing Committee for the PCGotPVNftCTNotESoA, Bluth T, Serpa Neto A, Schultz MJ, Pelosi P, Gama de Abreu M. Effect of Intraoperative High Positive End-Expiratory Pressure (PEEP) With Recruitment Maneuvers vs Low PEEP on Postoperative Pulmonary Complications in Obese Patients: A Randomized Clinical Trial. JAMA. 2019;321(23):2292–305.CrossRef
35.
Zurück zum Zitat Pelosi P, Rocco PRM, Gama de Abreu M. Close down the lungs and keep them resting to minimize ventilator-induced lung injury. Crit Care. 2018;22(1):72.CrossRef Pelosi P, Rocco PRM, Gama de Abreu M. Close down the lungs and keep them resting to minimize ventilator-induced lung injury. Crit Care. 2018;22(1):72.CrossRef
Metadaten
Titel
Intraoperative mechanical ventilation practice in thoracic surgery patients and its association with postoperative pulmonary complications: results of a multicenter prospective observational study
verfasst von
Christopher Uhlig
Ary Serpa Neto
Meta van der Woude
Thomas Kiss
Jakob Wittenstein
Benjamin Shelley
Helen Scholes
Michael Hiesmayr
Marcos Francisco Vidal Melo
Daniele Sances
Nesil Coskunfirat
Paolo Pelosi
Marcus Schultz
Marcelo Gama de Abreu
LAS VEGAS# investigators, Protective Ventilation Network (PROVEnet), Clinical Trial Network of the European Society of Anaesthesiology
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Anesthesiology / Ausgabe 1/2020
Elektronische ISSN: 1471-2253
DOI
https://doi.org/10.1186/s12871-020-01098-4

Weitere Artikel der Ausgabe 1/2020

BMC Anesthesiology 1/2020 Zur Ausgabe

Eingreifen von Umstehenden rettet vor Erstickungstod!

15.05.2024 Fremdkörperaspiration Nachrichten

Wer sich an einem Essensrest verschluckt und um Luft ringt, benötigt vor allem rasche Hilfe. Dass Umstehende nur in jedem zweiten Erstickungsnotfall bereit waren, diese zu leisten, ist das ernüchternde Ergebnis einer Beobachtungsstudie aus Japan. Doch es gibt auch eine gute Nachricht.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Häufigste Gründe für Brustschmerzen bei Kindern

06.05.2024 Pädiatrische Diagnostik Nachrichten

Akute Brustschmerzen sind ein Alarmsymptom par exellence, schließlich sind manche Auslöser lebensbedrohlich. Auch Kinder klagen oft über Schmerzen in der Brust. Ein Studienteam ist den Ursachen nachgegangen.

Update AINS

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