Skip to main content
Erschienen in: Journal of Cardiothoracic Surgery 1/2019

Open Access 01.12.2019 | Research article

Ten year follow-up of lung transplantations using initially rejected donor lungs after reconditioning using ex vivo lung perfusion

verfasst von: Haider Ghaidan, Mohammed Fakhro, Jesper Andreasson, Leif Pierre, Richard Ingemansson, Sandra Lindstedt

Erschienen in: Journal of Cardiothoracic Surgery | Ausgabe 1/2019

Abstract

Background

In 2006 and 2007 we performed double lung transplantation with marginal donor lungs assessed and reconditioned by Ex Vivo Lung Perfusion (EVLP), using a technique developed by Professor Stig Steen. Here we present a 10-year follow-up comparing the outcomes of lung transplantations performed at our clinic using EVLP lungs vs. conventional lungs.

Method

Between 2006 and 2007, 21 patients (6 EVLP, 15 conventional) underwent double lung transplantation (LTx) with follow-up on May 2017 at Lund University Hospital, Sweden. Pulmonary function was measured at 3/6/12 months, and annually thereafter for a period of 10 years in addition to survival and freedom from chronic lung allograft dysfunction (CLAD) being analyzed.

Results

Regarding Forced Expiratory Volume in 1 s (FEV1) and 6MWT at 3, 6, and 12 months and annually thereafter, no difference in median FEV1 nor 6MWT was found for EVLP-LTx vs. conventional-LTx (p > 0.05). No difference was shown in post-operative survival between EVLP-LTx vs. conventional LTx for patients with an overall survival up to 10-years (p > 0.05). The same pattern was shown in sub analyses for patients with a limited survival up to 1 and 5 years (p > 0.05).

Conclusion

No superiority was found in conventional-LTx over EVLP-LTx, neither in long-term survival nor pulmonary function. No difference in CLAD-free survival was seen between the two groups. We believe that EVLP is a safe and effective method to use in LTx, greatly increasing the donor pool by improving marginal lungs and providing an objective assessment of the viability of marginal donor lungs.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
6MWT
6 min walking test
CLAD
Chronic lung allograft dysfunction
DCD
Donation after circulatory death
DLTx
Double lung transplantation
EVLP
Ex vivo lung perfusion
FVC
Forces vital capacity
ISHLT
International society of heart and lung transplantation
LTx
Lung transplantation
Re-LTx
Redo lung transplantation
SLTx
Single lung transplantation

Introduction

Lung transplantation (LTx) is the golden standard for treating patients with irreversible end-stage pulmonary disease. A major challenge to LTx is the scarcity of donor organs resulting in deaths on the waiting list [1, 2]. Contrary to transplantation of other organs, only between 30% of potential donor organs are being utilized for LTx. This low proportion for acceptance of donor grafts is due to fact that the vast majority of grafts do not meet the criteria for transplantation [3, 4]. In 2006 we transplanted the first six patients with double LTX (DLTx) performed with donor lungs reconditioned ex vivo. The lungs were rejected for transplantation by the Scandia-transplant, Euro-transplant, and UK- transplant organizations due to arterial oxygen pressure less than 40 kPa. The donor lungs were reconditioned ex vivo in an extracorporeal membrane oxygenation circuit with STEEN solution mixed with erythrocytes. Ex vivo lung perfusion (EVLP) is today used at many lung transplant centers all over the world and is considered to be the golden standard in assessing and reconditioning marginal donor lungs for transplantation [59]. We have earlier reported the promising early outcome of these first six patients with a three-month survival at 100% while four of these patients showed no signs of Chronic Lung Allograft Dysfunction (CLAD) two years post-transplant [6] .
Long-term outcome including survival, pulmonary function, and occurrence of CLAD is to some extent unknown regarding EVLP as is whether there is any disadvantage compared to conventional LTx. This retrospective cohort study reviews the 10-year follow-up of the first six transplanted recipients. The study also compares EVLP DLTx to conventional DLTx performed at our center during the same time period.

Patients and methods

Data source

Between June 2006 and April 2007, 21 patients underwent LTx at Skåne University Hospital in Lund. Out of these, all were double-lung transplanted (DLTx). The median age for these patients was 52 years with a range of 22–66 years. In terms of gender, 9 were males and 12 females. The major indications for an LTx were chronic obstructive pulmonary disease (COPD) (n = 8), cystic fibrosis (CF) (n = 8), α1-antitrypsin deficiency (AAT1) (n = 1), pulmonary fibrosis (PF) (n = 2), pulmonary hypertension (PH) (n = 1), and lymphangioleiomyomatosis (LAM) (n = 1). With a total of 21 LTx, 6 were transplanted using EVLP, and 15 were transplanted using conventional LTx. This study has been performed in accordance with the Declaration of Helsinki and is approved by the Ethics Committee at Lund University with reference number 2016/638.

Marginal donors

The data from the marginal donors has been reported and described in detail from our previous publication [7]. Originally, nine donor lungs were investigated. To undergo reconditioning, the same donor criteria were met for ordinary donor lungs as for marginal lungs with the addition that we accepted lower partial pressure in arterial blood (PaO2). All lungs were turned down due to non-acceptable oxygenation capacity. The beforehand decided criteria to be accepted for transplantation after reconditioning in EVLP was that the PaO2 on fraction of inspired oxygen (FiO2) = 1 should be 50 kPa or higher. Median donor age with range was 59 (34–63) and median weight (kg) with range was 76 (55–94). All but one donor was CMV positive. Three of the donors had a known smoking history. All the six donor grafts had a median PaO2 (KPa) before harvesting (FIO2 = 1.0) of 21.1 with a range of 11.5 to 28.7.

EVLP setup

Our EVLP setup has been described in detail regarding our methodology in our previous studies [5, 7, 10].

Spirometry (FEV1)

Patients followed a planned clinical regime and were reviewed at regular intervals (3, 6, and 12 months, and annually thereafter for a period of 10 years). Spirometry were performed at each follow-up, assessing the patient’s FEV1 (liters).

6-min walking test

A 6MWT was performed at each follow-up with a regime similar to spirometry (3, 6, and 12 months, and annually thereafter). Assessing the patient’s expected work percentage determined on walking distance (meters), age (years), height (cm), and weight (kg).

Chronic lung allograft dysfunction (CLAD)

CLAD is a term that was introduced at first in 2010 [11, 12]. CLAD is principally caused from chronic rejection, usually resulting into one of following phenotypes: bronchiolitis obliterans (OB), neutrophilic reversible allograft dysfunction (NRAD), and restrictive allograft syndrome (RAS). Each of these conditions are usually presents with either airway obstruction or restriction and are mostly unresponsive to alterations in immunosuppression. According to the International Society for Heart and Lung Transplantation (ISHLT) guidelines, BOS, a major component of CLAD, is defined as more than 20% decline in FEV1 from the highest obtained baseline [12, 13], and is characterized by perivascular and interstitial mononuclear cell infiltrates or chronic rejection characterized by dense scarring and eosinophilic infiltrates. If rapid deterioration of pulmonary function was detected as a sign of CLAD, bronchoscopies with TBB were conducted and anti-rejection treatment was initiated with pulsed metylprednisolon often together with tacrolimus or everolimus as a replacement for cyclosporine.

Statistical methods

Data are presented as mean with standard deviation (SD), median with range, or frequency with percentage. Shapiro-Wilks test was used to determine which variables were normally distributed/parametric (mean, SD) vs. non-normally distributed/non-parametric (median, range). Independent (unpaired) student’s t-test was conducted for normally distributed continuous variables while Mann-Whitney U (Wilcoxon rank sum) test was used for non-normally distributed continuous data. Chi-square test or Fisher’s exact test were chosen for analysis of categorical variables. For survival analysis, the end-point used was death or Re-LTx. For freedom from CLAD analysis, the endpoint used was occurrence of CLAD until death/Re-LTx/follow-up. Cox regression in accordance with Cox proportional hazards model was performed for univariable survival analysis and freedom from CLAD analysis. Survival/freedom from CLAD-estimates were displayed in accordance with Kaplan-Meier with log-rank test to detect significance between survival/freedom from CLAD curves. A p-value < 0.05 was considered statistically significant. Statistical analyses were performed using SPSS Version 24.0 (IBM Corp., Armonk, NY, USA).

Results

Recipient characteristics

Baseline and clinical characteristics of recipients between EVLP-LTx and conventional-LTx are shown in Table 1. No significant difference between EVLP-LTx and conventional-LTx were shown regarding pulmonary function (FVC, FEV1, 6MWT), liver/kidney-status (AST, ALT, creatinine), and pre-operative life support (ECMO or mechanical ventilation) (p > 0.05).
Table 1
Recipient baseline and clinical characteristics of ex vivo lung perfusion (EVLP) lung transplantations (LTX) and conventional LTx
Variables
EVLP-LTx (n = 6)
Conventional-LTx (n = 15)
p-value
Weight (kg)
70.7 ± 19.3
59. 1 ± 7.9
0.060
Height (cm)
170.8 ± 11.8
169.9 ± 10.1
0.862
BMI
24.0 ± 5.3
20.5 ± 3.5
0.088
Male
3 (50%)
6 (40%)
0.523
Age (years)
54.1 ± 10.4
42.6 ± 14.8
0.100
Waiting list (days)
49.0 (7–174)
44 (4–389)
0.785
Pre-op Life support
 Mechanical ventilation
0 (0.00%)
1 (6.66%)
0.714
 ECMO
0 (0.00%)
1 (6.66%)
0.750
Major indication
  
0.407
 COPD
3 (50.00%)
5 (33.33%)
 
 AAT1
1 (16.66%)
0 (0.00%)
 
 PH
0 (0.00%)
1 (6.66%)
 
 CF
1 (16.66%)
7 (46.66%)
 
 PF
1 (16.66%)
1 (6.66%)
 
 LAM
0 (0.00%)
1 (6.66%)
 
Lab values
 FVC (liters)
2.0 ± 0.4
1.0 ± 0.6
0.540
 FEV1 (liters)
0.8 ± 10.4
54.1 ± 10.4
0.516
 6MWT (%)
39.6 ± 21.4
45.9 ± 25.1
0.600
 P-ALT (μkat/L)
0.41 ± 0.15
0.32 ± 10.4
0.181
 P-AST (μkat/L)
0.46 ± 0.12
0.41 ± 0.11
0.443
 P-creatinine (μmol/L)
64.4 ± 11.5
54.1 ± 15.8
0. 216
Tx-type
 SLTx
0 (0%)
0 (0%)
 
 DLTx
6 (100%)
15 (100%)
 
 HLTx
0 (0%)
0 (0%)
 
 Re-LTx
0 (0%)
0 (0%)
 
Data are mean (SD), number (%), or median (range). The numbers are based on patients with available data. COPD, chronic obstructive pulmonary disease; AAT1, Alpha 1-antitrypsin deficiency; PH, pulmonary hypertension; CF, cystic fibrosis; PF, pulmonary fibrosis; LAM, Lymphangioleiomyomatosis; BMI, body-mass index; FVC, forced volume vital capacity; FEV1, forced volume expiratory capacity 1 s; 6MWT, 6-min walking test; AST, aspartate transaminase; ALT, alanine transaminase; SLTx, single-lung transplantation; DLTx, double-lung transplantation; HLTx, heart-lung transplantation; Re-LTx, re-lungtransplantation; ECMO, extracorporeal membrane oxygenation

Mortality

Cause of death during follow-up stratified between EVLP-LTx and conventional-LTx is illustrated in Table 2. No difference was found in cause of death (rejection, infection, malignancy, or “miscellaneous”) between EVLP-LTx vs. conventional-LTx (p > 0.05).
Table 2
Cause of death after transplantation between ex vivo lung perfusion (EVLP) lung transplantations (LTX) and conventional LTx
 
EVLP-LTx (n = 6)
Conventional-LTx (n = 15)
p-value
Cause of death
  
0.406
Total number of deaths
3
6
 
Death from Organ Rejection
2 (66.66%)
2 (33.33%)
 
Death from Infection
0 (0.00%)
2 (33.33%)
 
Death from Malignancy
0 (0.00%)
1 (16.66%)
 
Death from Miscellaneous
1 (33.33%)
1 (16.66%)
 
The group “Death from Miscellaneous” includes patients with mortality caused by myocardial and cerebral ischaemia, multiple organ failure such as renal and liver failure, as well as other causes related to the patient’s age and individual health status

Survival/freedom from CLAD estimates

Cumulative survival rate estimates for EVLP-LTx and conventional-LTx at 1, 3, 5, and 10 years are illustrated in terms of percentage with an upper/lower 95% confidence interval (CI) (Fig. 1). For the entire cohort, EVLP-LTx showed 1-, 5-, and 7-year survival rates of 67% (CI 48–86), 67% (CI 48–86), and 50% (CI 30–70), respectively, compared to patients with conventional-LTx with 1-, 3-, 5-, and 7-year survival rates of 93% (CI 87–99), 73% (CI 62–85), 53% (CI 40–66), and 40% (CI 27–53), respectively (p > 0.05). In addition, survival at 1 year and 5 years showed no significant difference between EVLP-LTx and conventional-LTx (p > 0.05).
Freedom from CLAD estimates are shown in Fig. 2. Conventional-LTx showed freedom from CLAD rates at 1, 3, 5, and 7 years at 93% (CI 86–100), 70% (CI 45–94), 61% (CI 34–88), and 52% (CI 24–80), respectively, compared to patients with EVLP-LTx at 1- and 3-year rates of 100 and 75% (CI 53–97) (p > 0.05).

Cox regression

The Cox proportional hazards model (univariable) evaluating EVLP-LTx vs. conventional-LTx in addition to survival and freedom from CLAD is shown in Table 3. No significant difference was found between EVLP-LTx and conventional-LTx in overall survival of patients or in patients with a limited survival up to 1 and 5 years (p > 0.05). The same trend was found in the Cox regression analysis regarding freedom from CLAD regarding EVLP-LTx as opposed to conventional-LTx (p > 0.05).
Table 3
Cox proportional hazards model (univariable) ex vivo lung perfusion (EVLP) lung transplantations (LTX) and conventional LTx, evaluating survival and freedom from chronic allograft dysfunction (CLAD)
 
HR
95% CI
p-value
Overall survival
 EVLP
1. 245
0. 335–4. 633
0. 744
5-year limited survival
 EVLP
1. 286
0. 266–6. 206
0. 754
1-year limited survival
 EVLP
0. 197
0. 018–2. 175
0. 185
Freedom from CLAD
 EVLP
0. 470
0. 057–3. 917
0. 486
CI Confidence interval, HR Hazard ratio

Median FEV1 and 6MWT

Median pulmonary function with 95% CI over time regarding FEV1 in liters (L) and 6MWT in expected work percentage (%) is shown in Fig. 3. FEV1 for EVLP-LTx at 1, 5, and 7 years were 2.1 L (1.9–2.2), 2.2 L (2.1–2.5), and 2.1 (1.7–2.6) while conventional LTx showed at the same time intervals 2.6 L (1.0–3.3), 3.0 L (0.4–4.2), and 2.9 (0.5–3.1), respectively (p > 0.05). For the 6MWT, EVLP-LTx at 1, 5, and 7 years were 83% (57–87), 84% (70–112), and 79% (74–119), while conventional LTx was 71% (55–79), 88% (28–115), and 69% (10–123), respectively, at the same time intervals (p > 0.05).

Discussion

A new era is at hand in the field of LTx due to the immense potential of EVLP. The number of patients who are in need of a LTx surpasses the amount of available grafts. Using cellular and normothermic perfusion in combination with ventilation, EVLP improves marginal lungs and allows for an objective assessment of their viability. By increasing the availability of donor organs for transplantation, EVLP is being established as the new cornerstone in LTx. The very first LTx evaluated ex vivo was performed in 2001 by Steen et al. at our center in Lund, Sweden, from a non-heart beating donor [14]. A few years later we were again the first team to successfully perform a series of LTx by using grafts that did not meet standard transplantation criteria (i.e. marginal donor lungs) [7], with significant lower PaO2 levels. In the present study, we present our 10-year experience regarding the short- and long-term outcome of the first six patients that underwent LTx using marginal donor lungs evaluated using EVLP at our center. We also compare the outcome with conventional LTx performed at our clinic during the same time period. None of the marginal donor lungs would have normally been used for LTx if not for EVLP at our center. The marginal donor lungs used had been rejected by national and international transplantation centers due to low PaO2. The Lund EVLP protocol include priming the system with Steen solution and red blood cells with a target hematocrit at 14%, the pressure in the left atrium is kept at 0 mmHg by leaving the atrium open. The pulmonary artery pressure is kept under 20 mmHg to avoid pulmonary edema. During the evaluation the cardiac output target flow is 100%. Our EVLP protocols have been described in detail in our previous publications [7, 10].
Long term follow-up after EVLP has been limited by the recent introduction of the technique in clinical LTx. We have previously reported our short-term EVLP experience with a 100% survival rate at 30 days, with no significant differences shown between EVLP and standard LTx regarding time on mechanical ventilation, ICU stay, and overall hospital stay [6].
Up to 12 months after transplantation, Fildes et al. [15] reported no difference in mortality or number of infections between EVLP-LTx and conventional LTx. Wallinder et al. having one of the longest follow-ups with their 4-year experience, were able to demonstrate that conventional LTx lungs showed no superiority over EVLP-LTx in terms of survival and post-operative complications [16]. Our 10-year experience, the longest clinical EVLP follow-up to date, detected no significant difference between EVLP-LTx vs. conventional-LTx. The same pattern emerged in our sub analyses concerning patients with a limited survival up to 1 and 5 years. These findings, along with the fact that all EVLP donor lungs were initially rejected for LTx, are highly encouraging as it may allow for the expansion of the donor pool with grafts from marginal donor lungs. An important report that has studied EVLP for marginal donors is the NOVEL Lung trial, an FDA-mandated multicenter clinical trial that included 31 patients who received EVLP lungs [17]. Early outcomes in this trial regarding ICU/hospital stay, time on mechanical ventilation, primary graft dysfunction, and 30-day mortality showed great promise as they were similar to the control group consisting of 31 conventional LTx. It has been hypothesized in previous studies that EVLP provides its protective effect on the graft by interrupting possible cold storage injury and resuming the lungs to its proper physiological metabolic state. In addition, it decreases the microbial load from the donor, thereby protecting the immunosuppressed LTx-recipient from infection [18, 19].
In the present study no significant difference was found in freedom from CLAD between EVLP-LTx and conventional-LTx. This finding is in accordance with previous studies showing similar outcomes between the two groups concerning freedom from CLAD up to five years after LTx [16, 20]. Pulmonary function was followed up in our clinical program by FEV1 and 6MWT at 3, 6, and 12 months and annually thereafter for a period of ten years. Interestingly, no superiority was recognized regarding conventional-LTx over EVLP-LTx. FEV1 and 6MWT are well-known clinical tools that are non-invasive and provide excellent data on the clinical status of the recipient after transplantation [21]. To the authors knowledge, this is the first time that long-term outcome regarding pulmonary function has been investigated between the two groups. This finding is highly encouraging as it provides further evidence that EVLP is a reliable instrument in LTx for increasing the donor pool. EVLP has been suggested as a platform for administering medical agents and thus improving patient outcome. It has been shown that inhalation of CO prevents ischemia-reperfusion injury in animal models. It has also been shown that administration of nitroglycerin, selective adenosine 2A agonists, and dibutyryl cyclic adeonosine monophosphate in Steen solution lowers the rate of histological graft injury, lowers the levels of inflammatory cytokines, and keeps the microvascular structure of the graft unharmed compared to conventional LTx [22, 23]. It has also been reported that decreasing levels of thyroid stimulating hormone and desmopressin in brain-dead donors might have a role to play in LTx, where administration of such hormone replacements are made possible through EVLP and could improve graft function [24]. EVLP may also play a role in donation after circulatory death (DCD) especially in uncontrolled DCD donors when lung function is often unknown. Interestingly it has been reported that DCD-lungs that underwent EVLP showed better outcome regarding length of hospital stay and time on mechanical ventilation [25].
As EVLP allows for the extension of preservation time, it has the potential to open up possibilities for safer procedures by enabling more daytime surgery [26]. Extra preservation time could also reduce geographical limitations for recipients and donors, thus increasing both the safety and number of LTx [8].
EVLP might also serve as an important bridge in the future taking ex vivo lung bioengineering approaches to create functional, transplantable grafts into the clinic. Ex vivo lung bioengineering has the potential to overcome the challenges of organ donor shortages and prevent allograft immune rejection [27].

Limitations

This study is not without limitations. Only a small number of cases were followed up in our single center study. While a randomized clinical study of EVLP is ideal, it is not currently feasible due to ethical disagreement in the clinic about transplanting such grafts. It is difficult, therefore, to provide a true comparison between EVLP and conventional LTx. A larger dataset and equally large groups could give a more powerful analysis and limit the occurrence of type II errors.

Conclusions

On average 40% of brain dead donor lungs do not meet the criteria for LTx and are therefore not accepted for LTx. A considerable number of these organs may have been utilized in LTx through EVLP, which provides a method to evaluate and improve marginal donor lungs. According to the findings of our 10-year follow-up, the longest clinical follow-up to date, no differences were found between conventional LTx and EVLP regarding survival, pulmonary function, or incidence of CLAD.

Acknowledgements

Professor Stig Steen. All co-workers at the Department of Cardiothoracic Surgery, Pulmonary Medicine and Thoracic Intensive Care and Anesthesia, Skåne University Hospital, Lund University, Sweden.
This study has been performed in accordance with the Declaration of Helsinki and is approved by the Ethics Committee at Lund University with reference number 2016/638. All patients gave their written consent to participate.
All patients gave their written consent to participate.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Trulock EP, Edwards LB, Taylor DO, et al. The registry of the International Society for Heart and Lung Transplantation: twenty-first official adult lung and heart-lung transplant report--2004. J Heart Lung Transplant. 2004;23:804–15.CrossRef Trulock EP, Edwards LB, Taylor DO, et al. The registry of the International Society for Heart and Lung Transplantation: twenty-first official adult lung and heart-lung transplant report--2004. J Heart Lung Transplant. 2004;23:804–15.CrossRef
2.
Zurück zum Zitat Punch JD, Hayes DH, LaPorte FB, et al. Organ donation and utilization in the United States, 1996-2005. Am J Transplant. 2007;7:1327–38.CrossRef Punch JD, Hayes DH, LaPorte FB, et al. Organ donation and utilization in the United States, 1996-2005. Am J Transplant. 2007;7:1327–38.CrossRef
3.
Zurück zum Zitat Snell GI, Griffiths A, Levvey BJ, Oto T. Availability of lungs for transplantation: exploring the real potential of the donor pool. J Heart Lung Transplant. 2008;27:662–7.CrossRef Snell GI, Griffiths A, Levvey BJ, Oto T. Availability of lungs for transplantation: exploring the real potential of the donor pool. J Heart Lung Transplant. 2008;27:662–7.CrossRef
4.
Zurück zum Zitat Kawut SM, Reyentovich A, Wilt JS, et al. Outcomes of extended donor lung recipients after lung transplantation. Transplantation. 2005;79:310–6.CrossRef Kawut SM, Reyentovich A, Wilt JS, et al. Outcomes of extended donor lung recipients after lung transplantation. Transplantation. 2005;79:310–6.CrossRef
5.
Zurück zum Zitat Lindstedt S, Eyjolfsson A, Koul B, et al. How to recondition ex vivo initially rejected donor lungs for clinical transplantation: clinical experience from Lund university hospital. J Transp Secur. 2011;2011:754383. Lindstedt S, Eyjolfsson A, Koul B, et al. How to recondition ex vivo initially rejected donor lungs for clinical transplantation: clinical experience from Lund university hospital. J Transp Secur. 2011;2011:754383.
6.
Zurück zum Zitat Lindstedt S, Hlebowicz J, Koul B, et al. Comparative outcome of double lung transplantation using conventional donor lungs and non-acceptable donor lungs reconditioned ex vivo. Interact Cardiovasc Thorac Surg. 2011;12:162–5.CrossRef Lindstedt S, Hlebowicz J, Koul B, et al. Comparative outcome of double lung transplantation using conventional donor lungs and non-acceptable donor lungs reconditioned ex vivo. Interact Cardiovasc Thorac Surg. 2011;12:162–5.CrossRef
7.
Zurück zum Zitat Ingemansson R, Eyjolfsson A, Mared L, et al. Clinical transplantation of initially rejected donor lungs after reconditioning ex vivo. Ann Thorac Surg. 2009;87:255–60.CrossRef Ingemansson R, Eyjolfsson A, Mared L, et al. Clinical transplantation of initially rejected donor lungs after reconditioning ex vivo. Ann Thorac Surg. 2009;87:255–60.CrossRef
8.
Zurück zum Zitat Yeung JC, Krueger T, Yasufuku K, et al. Outcomes after transplantation of lungs preserved for more than 12 h: a retrospective study. Lancet Respir Med. 2017;5:119–24.CrossRef Yeung JC, Krueger T, Yasufuku K, et al. Outcomes after transplantation of lungs preserved for more than 12 h: a retrospective study. Lancet Respir Med. 2017;5:119–24.CrossRef
9.
Zurück zum Zitat Stanzi A, Neyrinck A, Somers J, et al. Do we need to cool the lung graft after ex vivo lung perfusion? A preliminary study. J Surg Res. 2014;192:647–55.CrossRef Stanzi A, Neyrinck A, Somers J, et al. Do we need to cool the lung graft after ex vivo lung perfusion? A preliminary study. J Surg Res. 2014;192:647–55.CrossRef
10.
Zurück zum Zitat Wierup P, Haraldsson A, Nilsson F, et al. Ex vivo evaluation of nonacceptable donor lungs. Ann Thorac Surg. 2006;81:460–6.CrossRef Wierup P, Haraldsson A, Nilsson F, et al. Ex vivo evaluation of nonacceptable donor lungs. Ann Thorac Surg. 2006;81:460–6.CrossRef
11.
Zurück zum Zitat Glanville AR. Bronchoscopic monitoring after lung transplantation. Semin Respir Crit Care Med. 2010;31:208–21.CrossRef Glanville AR. Bronchoscopic monitoring after lung transplantation. Semin Respir Crit Care Med. 2010;31:208–21.CrossRef
12.
Zurück zum Zitat Verleden GM, Raghu G, Meyer KC, et al. A new classification system for chronic lung allograft dysfunction. J Heart Lung Transplant. 2014;33:127–33.CrossRef Verleden GM, Raghu G, Meyer KC, et al. A new classification system for chronic lung allograft dysfunction. J Heart Lung Transplant. 2014;33:127–33.CrossRef
13.
Zurück zum Zitat Estenne M, Maurer JR, Boehler A, et al. Bronchiolitis obliterans syndrome 2001: an update of the diagnostic criteria. J Heart Lung Transplant. 2002;21:297–310.CrossRef Estenne M, Maurer JR, Boehler A, et al. Bronchiolitis obliterans syndrome 2001: an update of the diagnostic criteria. J Heart Lung Transplant. 2002;21:297–310.CrossRef
14.
Zurück zum Zitat Steen S, Sjoberg T, Pierre L, et al. Transplantation of lungs from a non-heart-beating donor. Lancet. 2001;357:825–9.CrossRef Steen S, Sjoberg T, Pierre L, et al. Transplantation of lungs from a non-heart-beating donor. Lancet. 2001;357:825–9.CrossRef
15.
Zurück zum Zitat Fildes JE, Archer LD, Blaikley J et al. Clinical outcome of patients transplanted with marginal donor lungs via ex vivo lung perfusion compared to standard lung transplantation. Transplantation 2015; 99: 1078–1083.CrossRef Fildes JE, Archer LD, Blaikley J et al. Clinical outcome of patients transplanted with marginal donor lungs via ex vivo lung perfusion compared to standard lung transplantation. Transplantation 2015; 99: 1078–1083.CrossRef
16.
Zurück zum Zitat Wallinder A, Riise GC, Ricksten SE, et al. Transplantation after ex vivo lung perfusion: a midterm follow-up. J Heart Lung Transplant. 2016;35:1303–10.CrossRef Wallinder A, Riise GC, Ricksten SE, et al. Transplantation after ex vivo lung perfusion: a midterm follow-up. J Heart Lung Transplant. 2016;35:1303–10.CrossRef
17.
Zurück zum Zitat Sanchez PG, Davis RD, D'ovidio F, et al. Normothermic ex vivo lung perfusion as an assessment of marginal donor lungs - the NOVEL lung trial. J Heart Lung Transplant. 2013;32:S16–7.CrossRef Sanchez PG, Davis RD, D'ovidio F, et al. Normothermic ex vivo lung perfusion as an assessment of marginal donor lungs - the NOVEL lung trial. J Heart Lung Transplant. 2013;32:S16–7.CrossRef
18.
Zurück zum Zitat Andreasson A, Karamanou DM, Perry JD, et al. The effect of ex vivo lung perfusion on microbial load in human donor lungs. J Heart Lung Transplant. 2014;33:910–6.CrossRef Andreasson A, Karamanou DM, Perry JD, et al. The effect of ex vivo lung perfusion on microbial load in human donor lungs. J Heart Lung Transplant. 2014;33:910–6.CrossRef
19.
Zurück zum Zitat Cypel M, Rubacha M, Yeung J, et al. Normothermic ex vivo perfusion prevents lung injury compared to extended cold preservation for transplantation. Am J Transplant. 2009;9:2262–9.CrossRef Cypel M, Rubacha M, Yeung J, et al. Normothermic ex vivo perfusion prevents lung injury compared to extended cold preservation for transplantation. Am J Transplant. 2009;9:2262–9.CrossRef
20.
Zurück zum Zitat Tikkanen JM, Cypel M, Machuca TN, et al. Functional outcomes and quality of life after normothermic ex vivo lung perfusion lung transplantation. J Heart Lung Transplant. 2015;34:547–56.CrossRef Tikkanen JM, Cypel M, Machuca TN, et al. Functional outcomes and quality of life after normothermic ex vivo lung perfusion lung transplantation. J Heart Lung Transplant. 2015;34:547–56.CrossRef
21.
Zurück zum Zitat Fakhro M, Ingemansson R, Algotsson L, Lindstedt S. Impact of forced expiratory volume in 1 second (FEV1) and 6-minute walking distance at 3, 6, and 12 months and annually on survival and occurrence of bronchiolitis obliterans syndrome (BOS) after lung transplantation. Ann Transplant. 2017;22:532–40.CrossRef Fakhro M, Ingemansson R, Algotsson L, Lindstedt S. Impact of forced expiratory volume in 1 second (FEV1) and 6-minute walking distance at 3, 6, and 12 months and annually on survival and occurrence of bronchiolitis obliterans syndrome (BOS) after lung transplantation. Ann Transplant. 2017;22:532–40.CrossRef
22.
Zurück zum Zitat Mulloy DP, Stone ML, Crosby IK, et al. Ex vivo rehabilitation of non-heart-beating donor lungs in preclinical porcine model: delayed perfusion results in superior lung function. J Thorac Cardiovasc Surg. 2012;144:1208–15.CrossRef Mulloy DP, Stone ML, Crosby IK, et al. Ex vivo rehabilitation of non-heart-beating donor lungs in preclinical porcine model: delayed perfusion results in superior lung function. J Thorac Cardiovasc Surg. 2012;144:1208–15.CrossRef
23.
Zurück zum Zitat Nakajima D, Chen F, Yamada T, et al. Reconditioning of lungs donated after circulatory death with normothermic ex vivo lung perfusion. J Heart Lung Transplant. 2012;31:187–93.CrossRef Nakajima D, Chen F, Yamada T, et al. Reconditioning of lungs donated after circulatory death with normothermic ex vivo lung perfusion. J Heart Lung Transplant. 2012;31:187–93.CrossRef
24.
Zurück zum Zitat Abdalla LG, Braga KA, Nepomuceno NA, et al. Ex vivo lung perfusion in Brazil. J Bras Pneumol. 2016;42:95–8.CrossRef Abdalla LG, Braga KA, Nepomuceno NA, et al. Ex vivo lung perfusion in Brazil. J Bras Pneumol. 2016;42:95–8.CrossRef
25.
Zurück zum Zitat Machuca TN, Mercier O, Collaud S, et al. Lung transplantation with donation after circulatory determination of death donors and the impact of ex vivo lung perfusion. Am J Transplant. 2015;15:993–1002.CrossRef Machuca TN, Mercier O, Collaud S, et al. Lung transplantation with donation after circulatory determination of death donors and the impact of ex vivo lung perfusion. Am J Transplant. 2015;15:993–1002.CrossRef
26.
Zurück zum Zitat Riddell P, Egan JJ. Will ex-vivo lung perfusion allow a move towards daytime lung transplantation surgery? Lancet Respir Med. 2017;5:88–9.CrossRef Riddell P, Egan JJ. Will ex-vivo lung perfusion allow a move towards daytime lung transplantation surgery? Lancet Respir Med. 2017;5:88–9.CrossRef
27.
Zurück zum Zitat Uhl FE, Wagner DE, Weiss DJ. Preparation of Decellularized lung matrices for cell culture and protein analysis. Methods Mol Biol. 2017;1627:253–83.CrossRef Uhl FE, Wagner DE, Weiss DJ. Preparation of Decellularized lung matrices for cell culture and protein analysis. Methods Mol Biol. 2017;1627:253–83.CrossRef
Metadaten
Titel
Ten year follow-up of lung transplantations using initially rejected donor lungs after reconditioning using ex vivo lung perfusion
verfasst von
Haider Ghaidan
Mohammed Fakhro
Jesper Andreasson
Leif Pierre
Richard Ingemansson
Sandra Lindstedt
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Cardiothoracic Surgery / Ausgabe 1/2019
Elektronische ISSN: 1749-8090
DOI
https://doi.org/10.1186/s13019-019-0948-1

Weitere Artikel der Ausgabe 1/2019

Journal of Cardiothoracic Surgery 1/2019 Zur Ausgabe

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.