[go: up one dir, main page]

Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Mar 28.
Published in final edited form as: Semin Respir Crit Care Med. 2013 Jul 2;34(3):305–319. doi: 10.1055/s-0033-1348474

Primary Graft Dysfunction

Yoshikazu Suzuki 1,#, Edward Cantu 1,#, Jason D Christie 2,3
PMCID: PMC3968688  NIHMSID: NIHMS559372  PMID: 23821506

Abstract

Primary graft dysfunction (PGD) is a syndrome encompassing a spectrum of mild to severe lung injury that occurs within the first 72 hours after lung transplantation. PGD is characterized by pulmonary edema with diffuse alveolar damage that manifests clinically as progressive hypoxemia with radiographic pulmonary infiltrates. In recent years, new knowledge has been generated on risks and mechanisms of PGD. Following ischemia and reperfusion, inflammatory and immunological injury-repair responses appear to be key controlling mechanisms. In addition, PGD has significant impact on short- and long-term outcomes; therefore, the choice of donor organ is impacted by this potential adverse consequence. Improved methods of reducing PGD risk and efforts to safely expand the pool are being developed. Ex-vivo lung perfusion is a strategy which may improve risk assessment and become a promising platform to implement treatment interventions to prevent PGD. This review will detail recent updates in the epidemiology, pathophysiology, molecular and genetic biomarkers and state-of-the-art technical developments affecting PGD. (158 words)

Keywords: primary graft dysfunction, lung transplantation, ischemia-reperfusion injury and repair, high-risk donor lung, ex-vivo lung perfusion

Definition and Clinical Presentation

Primary graft dysfunction (PGD) is a syndrome of acute lung injury that occurs within the first 72 hours after lung transplantation. PGD is characterized by pulmonary edema with diffuse alveolar damage that clinically manifests itself as progressive hypoxemia and radiographic pulmonary infiltrates without other identifiable causes.1-8. PGD is a form of the Acute Respiratory Distress Syndrome (ARDS), and shares characteristics with other factors predisposing to ARDS, such as trauma and sepsis.9 Historically, various labels were applied to PGD including ischemia-reperfusion injury, re-implantation response and edema, reperfusion edema, non-cardiogenic pulmonary edema, early graft dysfunction, primary graft failure, and post-transplant ARDS; however, the taxonomy was standardized in 2005 as PGD.1,8

PGD develops progressively and encompasses a spectrum of acute lung injury from milder dysfunction to more severe lung injury. The lack of standard defining criteria for PGD across studies resulted in historical variability in reported incidence rates, risk factors, clinical outcomes, and treatment effects, which led to inconsistencies in reproducibility and generalizability.1,6-17 Therefore, in 2005, the International Society of Heart and Lung Transplantation (ISHLT) Working Group on PGD proposed a standardized definition and grading system with the intent of establishing a reproducible and robust taxonomy.1 The proposed standardized definition of PGD was based on radiographic pulmonary infiltrates and a PaO2/FiO2 (P/F) ratio assessed at several time points after lung transplantation (Table 1, Figure 1). For example, the presence of radiographic infiltrates consistent with pulmonary edema and a P/F ratio of <200 after 72 hours of final lung perfusion are classified as “T72 Grade 3 PGD.”

Table 1.

2005 ISHLT Primary Graft Dysfunction Taxonomy

Grade at T0, T24, T48, T72 Radiographic infiltrates consistent with diffuse pulmonary edema PaO2/FiO2 Specific exceptions
0 - Any
1 + >300 On nasal canula or FiO2<0.3
2 + 200-300
3 + <200 Any patients on ECMO or on NO with FiO2>0.5 MV

Adapted from Reference1

T0 is defined as within 6 hours of final lung reperfusion or at the admission to the ICU. Patients are not considered having primary graft dysfunction without radiographic infiltrates.

PaO2/FiO2 should ideally be measured on PEEP of 5 cm H2O at FiO2 of 1.0 while patients are on MV.

ECMO: extracorporeal membrane oxygenation support, FiO2: fraction of inspired oxygen, ICU: intensive care unit, ISHLT: International Society of Heart Lung Transplantation, NO: nitric oxide, MV: mechanical ventilator, PaO2: partial arterial oxygen tension (mmHg), PEEP: positive end expiratory pressure

Fig. 1.

Fig. 1

Radiographic progression of severe grade 3 primary graft dysfunction (at T0, T24, T48, T72, T216 after extubation, and T3 months) after bilateral sequential lung transplantation with unexpected massive fat embolism of the lungs of a 22-year-old donor from motor vehicle accident. The recipient was a 62-year-old female with emphysema who required 4 days of venovenous extracorporeal membrane oxygenation, 8 days of ventilator support, and 22 days of hospital stay. She recovered with a good functional activity.

The diagnosis of PGD requires exclusion of mechanical, immune and infectious causes that can mimic, modify, and confound definition and grading (Table 2).1 Ideally, the P/F ratio is measured on a FiO2 of 1.0 and positive end expiratory pressure (PEEP) of 5 cm H2O. The ISHLT PGD definition and grading system was intended to be a first step to a continually refined definition (Table 2).1 Several proposed changes to the standardized definition have been suggested (Table 2)18-22 and further refinement efforts including validation and pathophysiological correlates are underway.

Table 2.

Exclusion Criteria, Limitations, and Refinements of 2005 ISHLT PGD Taxonomy

Exclusion criteria
Cardiogenic pulmonary edema
Pneumonia and aspiration
Hyperacute rejection
Pulmonary venous anastomotic obstruction
Limitations
Exclusion pathology can be missed clinically
Exclusion pathology can exist at the same time
Interobserver reliability of CXR reading
Variability in clinical settings
    example: single vs bilateral lung transplantation
        patients intubated vs extubated
        extubated on just oxygen mask vs on CPAP support
        arterial line removed, no P/F ratio measurements available
Variability in clinical conditions
    example: mode of ventilation and ventilator settings
        changes in cardiac output
Variability in logistics
    example: multiple blood gas results close to the time point
Proposed Refinements
T6 and T12 can be added (utility not yet validated)
Early trends in P/F ratio (the percentage change in P/F from T0 to T12) can be added (the utility not yet validated)
Type of the lung transplantation procedure separately designated (single vs bilateral)
Only include bilateral and diffuse infiltrates on CXR in bilateral lung transplantation
Extubated patients with non-invasive ventilation support included
Mode and conditions of ventilation appended
Use P/F ratio not worst, but closest to the time points among available multiple measurements

Adapted from References1,18-22

CPAP: continuous positive airway pressure, CXR: chest X-ray, ISHLT: International Society of Heart Lung Transplantation, P/F: PaO2: arterial oxygen tension (mmHg)/FiO2: fraction of inspired oxygen, PGD: primary graft dysfunction

Radiographic findings of PGD are nonspecific and include perihilar ground-glass opacities, peribronchial and perivascular thickening, and reticular interstitial and airspace opacities located in a dependent fashion predominantly in the middle and lower lung lobes.23-25 In the majority of cases, parenchymal opacities appear on postoperative day 1 and peak by day 3; however, different patterns of PGD resolution have been described. Clearance of these radiographic abnormalities usually begins on postoperative day 5-10; however, complete clearance is dependent on the severity of injury and can take between 10 days to a few months among survivors.23-26 Clinical improvement in the A-a gradient and P/F ratio often precedes radiographic resolution and poor correlation of radiographic finding has been reported.25 Recently, different patterns of PGD resolution have been reported, which may represent different injury resolution phenotypes.27

Epidemiology and Outcomes

In the era predating the ISHLT standardized definition, PGD had a reported incidence ranging between 11% and 57% in single-institution studies,7,9-17 with the more severe PGD (ISHLT grade 3) estimated to be between 15 to 35%.6 A retrospective analysis of 5262 lung recipients in the United Network for Organ Sharing (UNOS) /ISHLT registry between 1994 and 2000 showed that the overall incidence of T72 Grade 3 PGD was 10.2% without any significant changes by year (8-12% p=0.22); however, these findings were limited by the validity of the definition in this administrative dataset.28 A recent analysis of 1255 lung recipients enrolled between 2002 and 2010 in a prospective cohort study in 10 U.S. lung transplant centers (the Lung Transplant Outcomes Group: LTOG) showed that the incidence of T48-72 Grade 3 PGD was 16.8% with an overall incidence of Grade 3 PGD at any time point in the first 72 hours of 30.8%.29 The incidence of T48-72 Grade 3 PGD across the 10 centers varied significantly (2 to 27 %), reflecting different practice patterns and different risk factor distributions across centers. Since the ISHLT standardized definition and grading, the incidence of severe PGD (Grade 3) at T48-72 has been reported about 10-20% and the incidence of severe PGD (Grade 3) at any time point in the first 72 hours has been reported about 30% (Table 3).28-35

Table 3.

The Impact of PGD on Outcomes Using the 2005 ISHLT PGD Taxonomy

First Author Prekker30 Christie28 Christie31* Kreisel32 Diamond29 Daud33 Huang34 Whitson35
Center Minneapolis UNOS/ISHLT Registry Philadelphia St.Louis LTOG Registry St. Louis St. Louis Minneapolis
Published 2006 2005 2010 2011 2013 2006 2008 2007
Study dates 1992-2004 1994-2000 1991-2005 1988-2009 2002-2010 1998-2004 1998-2004 1992-2005
Number of subjects 402 5262
(UNOS/ISHLT registry)
446 1000
186 (19%) single vs 805 (81%) bilateral
1255 334 334
26 (7.8%) single vs 308 (92.2%) bilateral
374 (90 day survivors) out of 448
242 (65%) single vs 132 (35%) bilateral
Definition T0-48 Grade1-3 T72 Grade3 equivalent T24-72 Grade1-3 T0-48 Grade>3 (P/F<150) T48-72 Grade3 T0 Grade0-3 T24-72 Grade 0-3 T0-48 Grade0-3
Incidence 25% (T0G3)
15% (T48G3)
10.2% (T72G3) 28% (T24G3)
23% (T48G3)
18% (T72G3)
22.1% (T0-48G>3) 16.8% (T48-72G3)
30.8% (T0-72G3)
19% (T0G0)
39% (T0G1)
21% (T0G2)
21% (T0G3)
15.9% (T24G2)
7.9% (T48G2)
6.4% (T72G2)
16.2% (T24G3)
9.4% (T48G3)
7.9% (T72G3)
32% (T0-48G3)
Mortality (short-term) 90 day mortality
7% (T48G1)
13% (T48G2)
33% (T48G3)
(p=0.0001)
30 day mortality
42.1% (T72G3)
(RR=6.95 p<0.001)
43.6% attributable to 30 day mortality
30 day mortality
4.2% (T24G0)
3.5% (T24G1)
6.2% (T24G2)
24.5% (T24G3)
3.5% (T72G0)
5.0% (T72G1)
6.1% (T72G2)
36.4% (T72G3)
(p<0.001)
Hospital mortality
6.1% (overall)
14% (PGD)
4.0% (nonPGD) (p<0.001)
1 year survival
85% (overall)
73% (PGD)
87% (nonPGD) (p<0.001)
90 day mortality
23% (T48-72G3)
RR=4.8 ARI=18%
(p<0.001)
1 year mortality
34% (T48-72G3)
11% (the other)
RR=3.0 ARI=23%
(p<0.001)
90 day mortality
4.2% (overall)
0% (T0G0)
4% (T0G1)
1% (T0G1)
11%(T0G3) (p=0.004)
NS NS
Mortality (long-term) G3 worse
T0G3 best predicted long-term mortality
T72G3 worse
even conditional on survival at 1 year
(HR=1.35 p=0.011)
G3 worse
T72G3 more pronounced differences than T24G3
T72G1-2 worse than T72G0
Survival
58% at 5yrs, 33% at 10 yrs (overall)
44% at 5yrs, 19% at 10 yrs (PGD)
60% at 5yrs, 36% at 10 yrs (nonPGD)
(p<0.001)
NS T0G3 worse
even conditional on 90 day survival
RR=1.92 p=0.04 (T0G3)
T24-72G3 and T48-72G2 worse
even conditional on 90 day survival in univariate analysis
RR=2.43 p=0.03 (T48G2)
RR=3.27 p=0.003 (T72G2)
RR=2.57 p=0.007 (T24G3)
RR=4.09 p<0.0005 (T72G3)
compared to G0 at each T
G3 worse in bilateral
(no difference in single)
Survival
66% at 5yrs
38% at 10 yrs (G1)
64% at 5yrs
35% at 10 yrs (G2)
51% at 5yrs
11% at 10 yrs (G3)
(p=0.001)
BOS and Function (long-term) NS NS NS Median time to BOS 3.4 yrs
Freedom from BOS
84% at 1yr, 38% at 5yrs, 12% at 10 yrs (overall)
78% at 1yr, 28% at 5yrs, 8.5% at 10 yrs (PGD)
85% at 1yr, 41% at 5yrs, 13% at 10 yrs (nonPGD)
(p<0.01)
NS BOS stage1 incidence
50.3% (161/320)
even conditional on 90 day survival
Median time to BOS 3.89 yrs
PGD was independent risk factor of BOS stage1
RR=1.73 p=0.03 (T0G1)
RR=2.13 p=0.004 (T0G2)
RR=2.53 p=0.001 (T0G3)
compared to T0G0
T24-72G1-3
BOS stage1
RR=1.93 p=0.016 (T24G1)
RR=2.29 p=0.008 (T24G2)
RR=3.31 p<0.0005 (T24G3)
RR=2.07 p=0.003 (T72G1)
RR=2.81 p=0.003 (T72G2)
RR=4.25 p<0.0005 (T72G3)
BOS stage 2
RR=7.24 p<0.0005 (T72G3)
BOS stage 3
RR=7.99 p=0.0005 (T72G3)
compared to G0 at each T
G3 worse in BOS-free survival rate and mean FEV1 in bilateral
(no difference in single)

Adapted from References28-35

*

Severity grade of PGD was correlated to the level of established biomarkers (Protein C, PAI-1, and ICAM-1).

ARI: absolute risk increase, BOS: bronchiolitis obliterans syndrome, FEV1: forced expiratory volume in 1 second, G: grade, HR: hazard ratio, ICAM-1: intercellular adhesion molecule 1, ISHLT: International Society of Heart Lung Transplantation, LTOG: Lung Transplant Outcomes Group, NS: not stated, PAI-1: plasminogen activator inhibitor 1, P/F: PaO2: arterial oxygen tension (mmHg)/FiO2: fraction of inspired oxygen, PGD: primary graft dysfunction, RR: risk ratio, T: time point (hours), UNOS: United Network of Organ Sharing

PGD has a significant impact on early morbidity and mortality after lung transplantation, resulting in prolonged length of mechanical ventilation, prolonged intensive care unit (ICU) and hospital stay, increased cost, and increased short-term mortality.9 The impact of PGD on outcomes after the 2005 standardized definition was implemented is summarized in Table 3.28-35 Studies using the new definition have demonstrated better discrimination of PGD (Grade 3) to predict mortality in the short-term.30,31 Grade 3 PGD at T72 compared to Grade 0 demonstrated significantly higher risk of 30 day mortality and accounted for 50% of all-cause mortality within this period.29,31 When studied at 48-72 hours, Grade 3 PGD has been associated with increased 90 day mortality, with an absolute mortality increase of 18%.29 This risk persisted to 1 year post-transplant, with an absolute mortality increase of 23% at 1 year.29 Furthermore, grade 3 PGD is strongly associated with longer-term mortality,28,30-35 even after conditioning on 1 year survival.28

PGD has been associated with bronchiolitis obliterans syndrome (BOS), a significant limiting factor for long-term survival after lung transplantation.36 Initially, investigations evaluating linkage between PGD and BOS were conflicting before the ISHLT PGD standardized definition.9,12,13 However, subsequent studies showed this correlation between PGD and BOS to be independently significant, even for intermediate grades of PGD in dose-dependent fashion (Table 3).32-35 Recently, it was shown that markers of increased epithelial injury during PGD correlate with BOS, perhaps indicating a link between the degree of epithelial injury and aberrant repair or ongoing immune activation leading to BOS.37 PGD also has a negative impact on functional status. Survivors of T72 Grade 3 PGD at 12 months cover far less distance as measured by 6 minute walk distance than those without Grade 3 PGD.17 Because Grade 3 PGD has significant impact in short- and long term mortality and function, a thorough understanding of PGD risk may allow significant improvement in outcomes in lung transplant recipients.

Clinical risk factors

Many groups have studied risk factors for development of PGD.5,10,11,16,20,29,38-58 In the lung transplant process, there are several potentially injurious stages: 1) brain death of the donor, 2) explantation and preservation of donor organ, 3) cold ischemic storage and transport, 4) ischemia reperfusion injury (IRI) of the donor lung with implantation, and 5) postoperative recovery of the recipient.5 At each of these stages, a clear understanding of the pathophysiology of PGD and the contribution of each risk factor may allow for deliberate strategies and tailored interventions that can be implemented to improve outcomes related to PGD.

Studies examining clinical risk factors for PGD have been commonly limited by small sample sizes, single center designs, reliance on administrative data, and previously inconsistent definitions of PGD.29 Despite these limitations, several clinical risk factors have been consistently identified across studies, summarized in Table 4. 5,16,29,38-41 These risk factors can generally be categorized as donor, recipient, and operative related (Table 4).5,16 Donor-related risk factors include inherent (age, gender, race, smoking history) and acquired (primary and secondary injuries resulting from cause of death: pneumonia, aspiration, multiple blood transfusion, volume overload, hemodynamic instability, and ventilation associated injury) conditions which cannot be changed at the time of donor offer but may in the future be mitigated through ex vivo conditioning techniques or improved donor-recipient matching strategies.5,16,29,38-40,43 In particular, the donor smoking history has been a source of controversy, given inherent inaccuracy in determining donor smoking status29 as well as the fact that excluding donors with smoking history from the available pool of donor organs leads to more death for the whole transplant population.59 Studies of recipient-related factors have consistently reported significant association of diagnosis (idiopathic pulmonary fibrosis, sarcoidosis, and primary pulmonary hypertension), elevated pulmonary arterial pressures, and higher body mass index with PGD.11,16,29,38,39,41,42,44-46,48-50 These factors are the subjects of ongoing investigations aimed at understanding mechanistic underpinnings of increased PGD risk. Identified operative-related factors include single lung transplant, prolonged ischemic time, intracellular type preservation solutions, blood transfusion and use of cardiopulmonary bypass.10,20,29,39-41,44,45,47,50-58 Given the complexity of the interaction of multiple risk factors, efforts are currently underway on decision algorithms to aid donor recipient-matching aimed at reducing the incidence of PGD.

Table 4.

Clinical Risk Factors for PGD

Category Risk factors for PGD
Donor inherent variables Age>45yo, Age<21yo
African American race
Female gender
History of smoking >20py, >10py, current, any
Donor acquired variables Prolonged mechanical ventilation
Aspiration
Head trauma
Hemodynamic instability after brain death
Recipient variables Obesity Body mass index>25
Female gender
Diagnosis of idiopathic pulmonary hypertension
Diagnosis of secondary pulmonary hypertension
Diagnosis of idiopathic pulmonary fibrosis
Diagnosis of sarcoidosis
Elevated pulmonary arterial pressure at time of surgery
Operative variables Single lung transplantation
Prolonged ischemic time
Use of cardiopulmonary bypass
Blood products transfusion >1L
High FiO2 >=0.4 at reperfusion
Use of intra-cellular (hyperkalemic) type (Euro-Collins) preservation solution

Adapted from References5,16,29,39-41

Bold: independent risk factors for PGD in LTOG Multicentered Study29

FiO2: fraction of inspired oxygen, LTOG: Lung Transplant Outcomes Group, PGD: primary graft dysfunction, py: pack year, UNOS: United Network of Organ Sharing, yo: year old

Pathogenesis

PGD severity varies depending on (1) the baseline conditions of donor and recipient at the time of selection and matching, (2) conditions imposed on donor and recipient through the events of brain death, explantation, preservation, storage, and implantation, and (3) possibly genetic determinants that may modify injury and repair responses of donor lung in the recipient. The pathogenesis of PGD is complex and consequent to IRI resulting from direct damage of ischemia and preservation, generation of reactive oxygen species (ROS) at reperfusion, and activation of damage-amplifying proinflammatory cascade.

Experimental and clinical evidence suggests that PGD develops in a biphasic pattern with the early phase of PGD depending primarily on cells present in the donor lung and the later phase of PGD resulting from influx of recipient cells.41 Studies suggest that innate lung macrophages and lymphocytes modulate the early phases of PGD, while influx of recipient neutrophils and lymphocytes become more active in the subsequent later phases.60-62 Early free radical release from macrophages and pulmonary endothelium promotes the formation of inflammatory agents that recruit and activate recipient neutrophils and lymphocytes, which then escalate the process of injury.8,60,63-69 Further studies have implicated downstream effectors such as tumor necrosis factor (TNF)-alpha, interleukin (IL)-17 and other mediators of leukocyte, neutrophil, and innate immune components to be important in the recipient response of PGD. 61,70,71

Human studies using bronchoalveolar lavage or tissue biopsy samples have shown increased levels of IL-8 in the lungs of brain-dead donors that correlate in a dose-dependent manner with PGD severity and mortality.72 Immunolocalization measurements demonstrate diffuse IL-8 expression within donor lung tissue, with alveolar macrophages and epithelial cells being important sources. This abundant resident population of macrophages that act as a defense mechanism against environmental pathogens may act as a central mediator of the donor and recipient interplay leading to PGD in lung transplantation.

Lung transplantation is unique in that lungs continue to consume oxygen in the alveoli and to maintain aerobic metabolism during the ischemic preservation and storage period.73,74 This process is associated with higher oxygenation function and higher alveolar fluid clearance after reperfusion independent of the presence of aerobic metabolism.75,76 In theory, this partial alveolar oxygenation in the donor lung seems to be beneficial; however, multiple studies have demonstrated that it can have detrimental effects.73,77-80 High oxygen tension in the hypothermic lung is associated with excess ROS generation and lung injury, therefore, the practice of inflated lung procurement with room air is supported by the finding that hypothermic, hyperoxic, atelectatic lungs have worse gas exchange, edema, and peak inspiratory pressure.75,76,80,81 ROS generation is likely the direct result of tissue injury via oxidative damage to lipids, proteins, deoxyribonucleic acids, and biomolecules enhanced by the release of free iron Fe2+.82 Indirect effects have been demonstrated where ROS can serve as a cell signaling mediator responding to the loss of pulsatility by amplifying downstream events which result in endothelial activation.77-79

The long-term effects of PGD, particularly with respect to BOS, are also not fully understood. There is some recent evidence that recipients with pretransplant antibodies to self-antigens (k-alpha-1 tubulin, collagen type V, and collagen I) have increased risk of PGD , elevated proinflammatory mediators during the early post-transplant period, increased development of de novo anti-human leukocyte antigen (HLA) type II alloantibodies, and BOS.83-86 Likewise, there is speculation that PGD induces proinflammatory cascade to up-regulate HLA-II antigens and promoting donor specific alloimmunity, therefore mechanistically linking PGD and BOS,84-86 but this requires further study.

Molecular and genetic biomarkers

Investigations of clinical biomarkers, mediators, and genetic variants have largely been drawn from prior knowledge of basic pathophysiological mechanisms gained from basic cell and animal studies,5 and have included markers of alveolar epithelial and endothelial injury, cytokines and chemokines, adhesion molecules, indicators of hypercoagulability and impaired fibrinolysis, markers of vascular permeability and cell proliferation, markers of intracellular assembly for homeostasis and signaling, and markers of innate and acquired immunity. A number of biomarkers and genetic variants have been studied in clinical PGD (Tables 5 and 6),44,49,71,72,87-113 providing support that these mechanisms are contributing to clinical PGD, and that some may be amenable to targeted therapy. For example, it was recently shown that plasma long pentraxin 3 (PTX3), a readout of innate immune activation is elevated in PGD, and that functional variants in the encoding gene (PTX3) predispose to PGD.103,113 These results may define a population for intervention targeting this mechanistic axis. However, despite these studies, efforts at using markers to clinically predict the development, diagnosis, or prognosis of PGD are incomplete to date.107

Table 5.

Biomarkers for PGD in Human Lung Transplantation

First author, Publication year Biomarker Function
Pharm,87 1992 IL6 Proinflammatory cytokine
Mal,71 1998
Mathur,88 2006
IL6, IL8, IL1b, TNFa Proinflammatory cytokine
Fisher,72 2001
De Perrot,89 2002
IL8 Chemokine involved in neutrophil recruitment
Colombat,90 2004
Kawut,44 2009
P-Selectin positive platelet aggregate
sP-Selectin
Platelet adhesion molecule on the surface of activated endothelium
Abraham,91 2002
Krenn,92 2007
VEGF-A and –B
VAGF
Regulator of vascular permeability and angiogenesis
Christie,93 2007 Protein C
PAI1
Anticoagulant
Inhibitor of fibrinolysis
Covarrubias,94 2007 ICAM1 Adhesion molecule expressed by type I alveolar epithelial cell, alveolar endothelium, and neutrophil
Bobadilla,95 2008
Iwata,96 2008
Collagen type V
IgG Antibody to Collagen type V
Cryptic self-antigen and inducer for specific IL17 dependent cellular autoimmunity
Hoffman,97 2009 MCP1(CCL2)
IP10/CXCL10
IL13
IL2R
Chemokines involved in monocyte and lymphocyte recruitment
Anti-inflammatory and pleiotropic cytokine
Proinflammatory cytokines that activates lymphocyte
Moreno,98 2009 IL6, IL8
IL10
Proinflammatory cytokine
Antiinflammatory cytokine
Christie,99 2009
Pelaez,100 2010
sRAGE Marker of type I alveolar epithelial cell injury
Salama,101 2010 Endothelin1 Vasoconstrictive peptide, Regulator of vascular permeability
Lederer,49 2011 Leptin Proinflammatory cytokine produced by adipose tissue regulating adipose tissue mass and contributing to acute lung injury and fibroprolifration leading to lung fibrosis
Diamond,102 2011 CC16 Marker of lung epithelial injury produced by non-ciliated lung epithelium clara cell
Diamond,103 2011 PTX3 in IPF Phylogenetically conserved acute phase reactant involved in inflammation and innate immunity
Diamond,104 2012 Angiopoietin2 in IPF Inducer for increased endothelial permeability
Bastarache,105 2012 Estradiol in male Proinflammatory female sex hormone
Shah,106 2012 MCP1(CCL2) Chemokines recruiting monocyte, memory T cell, and natural killer cell
Shah,107 2012 sRAGE
PAI1
ICAM1
Marker of epithelial injury
Inhibitor of fibrinolysis
Adhesion molecule

Adapted from References44,49,71,72,87-107

a: alpha, b: beta, CC16: serum clara cell protein 16, CCL2: chemokine (C-C motif) ligand 2, CXCL10: chemokine (C-X-C motif) ligand 10, ICAM1: intercellular adhesion molecule 1, Ig: immunoglobulin, IL: interleukin, IP10: interferon-gamma induced protein 10, IPF: idiopathic pulmonary fibrosis, MCP1: monocyte chemotactic protein 1, PAI1: plasminogen activator inhibitor 1, PGD: primary graft dysfunction, PTX3: pentraxin 3, RAGE: receptor for advanced glycation end-products, s: soluble, TNF: tumor necrotic factor, VEGF: vascular endothelial growth factor

Table 6.

RNA and DNA Markers for PGD in Human Lung Transplantation

First author, Publication year Marker Function
Fisher,72 2001 IL-8 Chemokine involved in neutrophil recruitment
Taghavi,108 2002 Endothelin-1 Vasoconstrictive peptide, Regulator of vascular permeability
Kaneda,109 2006 IL6:IL10 ratio Proinflammatory: Anti-inflammatory and pleiotropic cytokine
Ray,110 2007 Metallothionein 3 (protective) Intra-cellular protein with antioxidant properties promoting cell proliferation and re-epithelialization
Anraku,111 2008 ATP11B
FGFR2
EGLN1
MCPH1
ATP-dependent ion transporter for oxidative stress
Receptor for HGF and KGF for alveolar epithelial repair
Hypoxia-inducible factor (sensor) for controlling hypoxia
DNA damage modulation for repairing damaged DNA
Hagedorn,112 2011 Autoantibody
(IgM) to 17 proteins
and donor lung gene expression
12 proteins out of 17 related to cell proliferation
Diamond,113 2012 PTX3 Phylogenetically conserved acute phase reactant involved in inflammation and innate immunity

Adapted from References72,108-113

ATP: adenosine triphosphate, DNA: deoxyribonucleic acid, HGF: hepatocyte growth factor, IL: interleukin, KGF: keratinocyte growth factor, Ig: immunoglobulin, PGD: primary graft dysfunction, PTX3: pentraxin 3, RNA: ribonucleic acid, SNP: single nucleotide polymorphism

Preventive Interventions

Strategies commonly used to prevent and minimize the development and severity of PGD include: 1) optimizing selection, matching, and management of donors and recipients preoperatively; 2) improving lung preservation and storage techniques; and 3) improving lung implantation and reperfusion techniques based on known risk factors of PGD.8,40,41,114,115

Several therapeutic agents have been investigated in an effort to reduce the incidence of PGD, including:5 1) agents replacing endogenous cytoprotective substances including prostaglandins, nitric oxide (NO), surfactant, endothelium-derived relaxing factor, and adenosine; 2) agents inhibiting proinflammatory mediators including Platelet Activating Factor 1 inhibitor and inhibitors of ROS; 3) agents inhibiting neutrophil and neutrophil derived mediators including inhibitors of ROS, cytokines (TNF-alpha, IL-1b), proteases, lipid mediators, adhesion molecules, and complement cascade.5,8,114,116 However, there have been only a few randomized trials, usually with limited sample sizes.5 These small trials have failed to definitively demonstrate efficacy and remain an area of intense study,98,117-122 even though some studies have shown modest improvements in early clinical outcomes with use of inhaled NO (iNO),98 surfactant,122 soluble complement receptor 1 inhibitor,119 and platelet activating factor antagonist.120

Treatment options after diagnosis of PGD

Therapy for established PGD after diagnosis remains generally supportive, and is influenced by those applied in patients with ARDS, including lung-protective ventilation strategies, avoidance of excess fluid administration, iNO or prostacycline, and extracorporeal membrane oxygenation support (ECMO).114 Although retransplantation can be considered in highly selected cases without other end-organ damage, predicted survival is poor;123 therefore, retransplantation for PGD is generally not recommended.114 Protocolized respiratory and hemodynamic management guidelines are feasible and may be effective in reducing the severity of PGD.124 However, there have been few studies systematically evaluating the application of strategies that have been specifically used in ARDS patients to patients with severe PGD and thus management is largely individualized by center.29

There are several animal studies and case series that have demonstrated improved outcomes with iNO administration to prevent and treat severe PGD and refractory hypoxemia in lung transplantation.125-128 However, there are also conflicting results in randomized controlled studies showing lack of efficacy for iNO use in the clinical setting.117,118 While use of iNO to prevent PGD has not been established, a randomized clinical trial showed lower incidence of Grade 3 PGD within 72 hours (17.2% vs 45% p<0.035) and decreased levels of inflammatory biomarkers with use of iNO beginning at time of transplantation through 48 hours postoperatively.98 Though iNO does not have an established role in either prevention of PGD development or treatment of established PGD, its use may be justified as salvage therapy in selected cases.

Veno-arterial (VA) ECMO has been used for salvage of refractory hypoxemia due to severe PGD following lung transplantation.129-132 Veno-venous (VV) ECMO has gained in popularity with potentially good survival if started early.133 In the past, patients receiving VV ECMO or VA ECMO had poor long-term survival with 30 day, 1 year, and 5 year survival of 55-58%, 39-42%, and 22-29%, respectively.132 However, the more recent development of high performance membranes and coated circuits has led to improved results. Early VV ECMO achieved significantly improved 30 day, 1 year, and 5 year survival of 82%, 64%, and 49%, respectively.133 Though survival has improved, allograft function is considerably worse than in transplant recipients not requiring ECMO (peak forced expiratory volume in 1 second: 58% in ECMO vs 83% in non-ECMO, p = 0.001), although these results are hampered by intrinsic differences in lung injury between comparator groups.

Extended criteria donor lungs

Procurement rates of lungs offered for transplantation remain between 14-17% of organ donors. Only 31% of patients on the waiting list receive lung transplantation, while 7% die on the list and an additional 8% are removed from the list due to clinical deterioration.134 Efforts to expand the donor pool by using extended criteria donor lungs have continued since the report of successful marginal donor use by Kron et al in 1993.135 The reported proportion of extended criteria donor lungs in single institution studies varies between 24-77%, although it is difficult to consistently define an extended criteria donor.115 Most reports demonstrate equivalent outcomes regarding incidence of PGD, early hospital morbidity and survival, and intermediate survival and freedom from BOS using extended criteria donors.115,135-144 However, two studies reported a significantly higher incidence of Grade 3 PGD and a significantly higher early organ-specific mortality with the use of extended criteria donor lungs.140,143 Caution and clinical judgment are needed when using marginal donor lungs with more than one extended criterion and when matching marginal donor lungs to high-risk recipients, especially recipients with pulmonary hypertension.145 Donor-recipient matching is complex in clinical practice and difficult to study because risk assessment is required on an individual case basis. It is unclear how far a given donor can safely be from “ideal criteria” because many factors (donor, recipient and operative) affect the risk and severity of PGD.

Emerging concepts

Normothermic ex vivo lung perfusion (EVLP) is a new evaluation technique for high-risk donor organs.146 EVLP is thought to reduce the risk of using high-risk donor lungs by allowing for an extended normothermic evaluation period. The EVLP system consists of a ventilator and an ex vivo circulation system with centrifugal pump, oxygenator, heat-exchanger, and leukocyte filter.147 Steen and colleagues in Lund, Sweden were the first to successfully apply ex vivo evaluation in 2001 on lungs from a donation after cardiac determination of death (DCDD).148 Following this success, additional study and modification has occurred. In a landmark trial from Toronto, Cypel and colleagues reported excellent clinical outcomes using high-risk donors ordinarily not transplantable with acellular perfusate and protective perfusion and ventilation strategies in 2011.146 Worldwide clinical experiences with EVLP are summarized in Table 7.146,149-154 Although most reports are small case series, the proportion of high-risk donor lungs transplanted after EVLP evaluation range between 46-87% resulting in equivalent short-term outcomes as compared with standard criteria donors. It is remarkable that the incidence of severe PGD is generally lower in the EVLP group in comparison to the conventional group, with equivalent survival (96%, 87%, 70% at 30 days, 1 year, and 3 years, respectively). These results demonstrate that EVLP not only enhances the ability to accept more organs for transplantation but also suggests the possibility of reducing the risk of PGD. EVLP might also offer a platform to use targeted therapeutics to enhance the quality of these organs in the future.155 Further clinical assessment of the impact on PGD and short-term outcomes is underway in a multicenter prospective trial in the United States.

Table 7. Clinical Experiences of EVLP.

Recipients of EVLP reconditioned donor lungs vs Recipients of conventional donor lungs

First Author Lindstedt149 Cypel146 Cypel150 Zych151 Aigner152 Valenza153 Wallinder154
Place Lund, Sweden Toronto, Canada Toronto, Canada Harefield, England Vienna, Austria Milano, Italy Gothenburg, Sweden
Publication Year 2011 2011 2012 2012 2012 2012 2012
Study Year (duration) 2006-2007 2008-2010 (17 months) 2008-2011 (39 months) 2009-2010 (24mo) 2010-2011 (16 mo) 2011 (6 mo) 2011 (10 mo)
Number of recipients transplanted 6 after 9 EVLP (67%) vs 15 20 after 23 EVLP (87%) vs 116 50 after 58 EVLP (86%) vs 253 6 after 13 EVLP (46%) vs 86 9 after 13 EVLP (69%) vs 119 2 after 2 EVLP (100%) vs 4 6 after 6 EVLP (2/6 double to single) vs 27
Duration of EVLP 1.5-2.0 (hrs) 4.0 (hrs) 4.0-6.0 (hrs) 2.4+-0.5 (hrs) 2-4 (hrs) 4.0 (hrs) 2.6-6.6 (hrs)
Proportion of DLTXP 100% vs 100% 70% vs 85% 76% vs 88% 100% vs NS 100% vs NS 100% vs 100% 67% (4/6) vs NS
Proportion of DCDD 0% vs 0% 45% (9/20) vs NS 44% (22/50) vs 5.1% (P=0.0001) 33% (2/6) vs 23% (3/13) 0% vs NS 0% vs 0% 0% vs NS
Time from Harvest to Implantation (min) NS 653 (267-1021) vs 370 (163-662) NS 493 (249-620) vs NS 577 (486-678) vs NS 1005+-21 vs 606+-88 864 (782-1049) vs NS
PaO2:FiO2 ratio NS 335 before EVLP, Among DCDD 275 (160-459) before EVLP 334 (143-532) before EVLP vs 452 (256-590) before conventional (P=0.0001) 317.7+-106.7 before 429.9+-68.3 after EVLP vs NS 216 (133-271) before 466 (434-525) after EVLP vs 447 (311-702) before conventional 184+-6 before EVLP vs 570+-30 before conventional 175.5 (77.4-313.5) before EVLP vs NS
PaO2:FiO2 in ICU NS 422 (85-538) at T0, 414 at T24, 443 at T72 372 (49-591) at T0 291+-56.3 vs NS 218+-32 vs 310+-97 at T0, 306 (282-331) vs 323 (270-396) at T72
T72 PGD2 or 3 NS 15% vs 30% (P=0.11) 2% vs 8.5% (P=0.14) NS 0% vs NS 0% (0/2) vs 25% (1/4) 17% (1/6) vs NS
T72 PGD3 NS 0% vs 9.4% (P=0.36) NS NS 11% (1/9) vs NS 0% vs 0% 0% vs NS
ECMO 17% (1/6) vs 27% (4/15) 0% vs 4% 2% vs 2.7% 33% (2/6) vs NS 11% (1/9) vs NS 0% vs 0% 0% vs NS
Ventilation time (day) NS 2 (1-101) vs 2 (1-43) 2 (1-101) vs 2.2 (1-43) 11+-12 vs NS 2 (1-7) vs 1 (1-47) NS 1 (1-24) days vs NS
ICU stay (day) 13(IQ 5, 24) vs 7 (IQ 5, 12) (P=0.44) 4 (1-101) vs 4 (1-103) 4 (1-100) vs 4.5 (1-257) 20.5 (IQ 4.0, 50.5) vs 7 (IQ 3.0, 25.5) 5.5 (3-18) vs 6 (2-51) 6 (3-9) vs 27 (5-50) 3.5 (2-27)
Hospital stay (day) median 52 (IQ 47, 60) vs 44 (IQ 37, 48) (P=0.9) 23 (7-101) vs 27 (9-156) 20 (7-156) vs 23 (1-299) 38 (IQ 25, 67.5) vs 37 (IQ 24, 59.5) 20 (14-59) vs 20.5 (12-90) 34+-20 vs 46+-33 26 (25-44) vs NS
Survival 100% at 30 days
100% at 3 months
83% at 6 months
67% at 1 year
90% (18/20) vs 94.8%(110/116) at 30 days (P=0.33)
80% vs 83.6% at 1 year
75% at 561 days (7-821) vs 81% at 542 days (9-828)
96% (48/50) vs 96.5% (244/253) at 30 days
87% vs 86% at 1 year
70% vs 72% at 3 year
100% vs 90% at 3 months
83% vs 88% at 6 months
100% vs 95.8% at 30 days
89% vs NS at 3 months
89% vs NS at 6 months
78% vs NS at 1 year
100% (2/2) vs 100% (4/4) at 30 days
50% (1/2) vs 75% (3/4) at 3 months
100% (6/6) vs NS at 30 days

Adapted from References146,149-154

<median (range minimum-maximum) or median+-standard deviation, IQ:interquartile range (IQ25, IQ75)>

DCDD: donation after cardiac determination of death, DLTXP: double lung transplantation, ECMO: extracorporeal membrane oxygenation support, EVLP: ex-vivo lung perfusion, hrs: hours, ICU: intensive care unit, min: minutes, mo: months, PGD: primary graft dysfunction, PaO2: arterial oxygen tension (mmHg), FiO2: fraction of inspired oxygen, T72PGD3: PGD grade 3 at 72 hours after lung transplantation

Alternatively, other approaches have been developed that challenge standard organ preservation methods. Warnecke et al, have reported results using a mobile normothermic ex vivo perfusion system designed to minimize cold ischemic injury.156 Among the 12 donors enrolled, five (42%) were designated extended criteria donors. Median mechanical ventilation time and ICU length of stay post-transplant was 21 hours (range 9.5-456) and 7 days (2-31 days), respectively. All were discharged and one died on day 140 due to cardiomyopathy. Based on these results, a prospective randomized multicenter trial is now underway to compare this method with standard static cold storage.156

Conclusions

PGD remains a key factor for both short- and long-term outcomes and thus limits the number of high-risk donor lungs utilized. Implementation of the ISHLT standard definition and grading criteria of PGD has enhanced our ability to better study and understand the incidence, related outcomes, risk factors, biomarkers and genetic factors influencing of PGD. Following ischemia-reperfusion, inflammatory and immunologic injury-repair responses appear to be key controlling mechanisms in PGD. Continued need to safely increase the donor lung pool has driven the development of EVLP as a deliberate strategy to assure the quality of donor lungs and potentially to implement treatment interventions. Initial clinical experience with EVLP looks promising. The impact on long-term outcomes warrants further study and the impact on incidence of PGD will need to be assessed in multicenter prospective trials.

Acknowledgments

Funding Sources: AI063589, HL087115, HL081619, HL096845, HL115354, HL116656, RWJ11642

Abbreviations

ARDS

acute respiratory distress syndrome

BOS

bronchiolitis obliterans syndrome

DCDD

donation after cardiac determination of death

ECMO

extracorporeal membrane oxygenation support

EVLP

ex vivo lung perfusion

HLA

human leukocyte antigen

ICU

intensive care unit

IL

interleukin

IRI

ischemia reperfusion injury

ISHLT

International Society of Heart and Lung Transplantation

LTOG

Lung Transplant Outcomes Group

NO

nitric oxide

PEEP

positive end expiratory pressure

P/F ratio

PaO2 (the partial pressure of oxygen in alveolar gas)/FiO2 (the fraction of inspired oxygen) ratio

PGD

primary graft dysfunction

PTX3

long pentraxin 3

ROS

reactive oxygen species

TNF

tumor necrosis factor

UNOS

United Network for Organ Sharing

VA

veno-arterial

VV

veno-venous

References

  • 1.Christie JD, Carby M, Bag R, et al. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction part II: definition. A consensus statement of the International Society for Heart and Lung Transplantation. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2005;24(10):1454–1459. doi: 10.1016/j.healun.2004.11.049. [DOI] [PubMed] [Google Scholar]
  • 2.Christie JD, Van Raemdonck D, de Perrot M, et al. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction part I: introduction and methods. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2005;24(10):1451–1453. doi: 10.1016/j.healun.2005.03.004. [DOI] [PubMed] [Google Scholar]
  • 3.Lee JC, Christie JD. Primary graft dysfunction. Proceedings of the American Thoracic Society. 2009;6(1):39–46. doi: 10.1513/pats.200808-082GO. [DOI] [PubMed] [Google Scholar]
  • 4.Lee JC, Christie JD, Keshavjee S. Primary graft dysfunction: definition, risk factors, short- and long-term outcomes. Seminars in respiratory and critical care medicine. 2010;31(2):161–171. doi: 10.1055/s-0030-1249111. [DOI] [PubMed] [Google Scholar]
  • 5.Lee JC, Christie JD. Primary graft dysfunction. Clinics in chest medicine. 2011;32(2):279–293. doi: 10.1016/j.ccm.2011.02.007. [DOI] [PubMed] [Google Scholar]
  • 6.Trulock EP. Lung transplantation. American journal of respiratory and critical care medicine. 1997;155(3):789–818. doi: 10.1164/ajrccm.155.3.9117010. [DOI] [PubMed] [Google Scholar]
  • 7.Christie JD, Bavaria JE, Palevsky HI, et al. Primary graft failure following lung transplantation. Chest. 1998;114(1):51–60. doi: 10.1378/chest.114.1.51. [DOI] [PubMed] [Google Scholar]
  • 8.de Perrot M, Liu M, Waddell TK, Keshavjee S. Ischemia-reperfusion-induced lung injury. American journal of respiratory and critical care medicine. 2003;167(4):490–511. doi: 10.1164/rccm.200207-670SO. [DOI] [PubMed] [Google Scholar]
  • 9.Arcasoy SM, Fisher A, Hachem RR, et al. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction part V: predictors and outcomes. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2005;24(10):1483–1488. doi: 10.1016/j.healun.2004.11.314. [DOI] [PubMed] [Google Scholar]
  • 10.Khan SU, Salloum J, O'Donovan PB, et al. Acute pulmonary edema after lung transplantation: the pulmonary reimplantation response. Chest. 1999;116(1):187–194. doi: 10.1378/chest.116.1.187. [DOI] [PubMed] [Google Scholar]
  • 11.King RC, Binns OA, Rodriguez F, et al. Reperfusion injury significantly impacts clinical outcome after pulmonary transplantation. The Annals of thoracic surgery. 2000;69(6):1681–1685. doi: 10.1016/s0003-4975(00)01425-9. [DOI] [PubMed] [Google Scholar]
  • 12.Fiser SM, Tribble CG, Long SM, et al. Ischemia-reperfusion injury after lung transplantation increases risk of late bronchiolitis obliterans syndrome. The Annals of thoracic surgery. 2002;73(4):1041–1047. doi: 10.1016/s0003-4975(01)03606-2. discussion 1047-1048. [DOI] [PubMed] [Google Scholar]
  • 13.Fisher AJ, Wardle J, Dark JH, Corris PA. Non-immune acute graft injury after lung transplantation and the risk of subsequent bronchiolitis obliterans syndrome (BOS). The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2002;21(11):1206–1212. doi: 10.1016/s1053-2498(02)00450-3. [DOI] [PubMed] [Google Scholar]
  • 14.Thabut G, Vinatier I, Stern JB, et al. Primary graft failure following lung transplantation: predictive factors of mortality. Chest. 2002;121(6):1876–1882. doi: 10.1378/chest.121.6.1876. [DOI] [PubMed] [Google Scholar]
  • 15.Chatila WM, Furukawa S, Gaughan JP, Criner GJ. Respiratory failure after lung transplantation. Chest. 2003;123(1):165–173. doi: 10.1378/chest.123.1.165. [DOI] [PubMed] [Google Scholar]
  • 16.Christie JD, Kotloff RM, Pochettino A, et al. Clinical risk factors for primary graft failure following lung transplantation. Chest. 2003;124(4):1232–1241. doi: 10.1378/chest.124.4.1232. [DOI] [PubMed] [Google Scholar]
  • 17.Christie JD, Sager JS, Kimmel SE, et al. Impact of primary graft failure on outcomes following lung transplantation. Chest. 2005;127(1):161–165. doi: 10.1378/chest.127.1.161. [DOI] [PubMed] [Google Scholar]
  • 18.Prekker ME, Herrington CS, Hertz MI, Radosevich DM, Dahlberg PS. Early Trends in PaO(2)/fraction of inspired oxygen ratio predict outcome in lung transplant recipients with severe primary graft dysfunction. Chest. 2007;132(3):991–997. doi: 10.1378/chest.06-2752. [DOI] [PubMed] [Google Scholar]
  • 19.Oto T, Griffiths AP, Levvey BJ, Williams TJ, Snell GI. Unilateral radiographic abnormalities after bilateral lung transplantation: exclusion from the definition of primary graft dysfunction? The Journal of thoracic and cardiovascular surgery. 2006;132(6):1441–1446. doi: 10.1016/j.jtcvs.2006.08.003. [DOI] [PubMed] [Google Scholar]
  • 20.Oto T, Griffiths AP, Levvey BJ, et al. Definitions of primary graft dysfunction after lung transplantation: differences between bilateral and single lung transplantation. The Journal of thoracic and cardiovascular surgery. 2006;132(1):140–147. doi: 10.1016/j.jtcvs.2006.03.029. [DOI] [PubMed] [Google Scholar]
  • 21.Oto T, Levvey BJ, Snell GI. Potential refinements of the International Society for Heart and Lung Transplantation primary graft dysfunction grading system. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2007;26(5):431–436. doi: 10.1016/j.healun.2007.01.029. [DOI] [PubMed] [Google Scholar]
  • 22.Christie J, Keshavjee S, Orens J, et al. Potential refinements of the International Society for Heart and Lung Transplantation primary graft dysfunction grading system. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2008;27(1):138. doi: 10.1016/j.healun.2007.09.030. [DOI] [PubMed] [Google Scholar]
  • 23.Collins J. Imaging of the chest after lung transplantation. J Thorac Imag. 2002;17(2):102–112. doi: 10.1097/00005382-200204000-00002. [DOI] [PubMed] [Google Scholar]
  • 24.Anderson DC, Glazer HS, Semenkovich JW, et al. Lung transplant edema: chest radiography after lung transplantation--the first 10 days. Radiology. 1995;195(1):275–281. doi: 10.1148/radiology.195.1.7892485. [DOI] [PubMed] [Google Scholar]
  • 25.Kundu S, Herman SJ, Winton TL. Reperfusion edema after lung transplantation: radiographic manifestations. Radiology. 1998;206(1):75–80. doi: 10.1148/radiology.206.1.9423654. [DOI] [PubMed] [Google Scholar]
  • 26.Herman SJ, Rappaport DC, Weisbrod GL, et al. Single-lung transplantation: imaging features. Radiology. 1989;170(1 Pt 1):89–93. doi: 10.1148/radiology.170.1.2642351. [DOI] [PubMed] [Google Scholar]
  • 27.Shah RJ, Diamond JM, Kawut SM, et al. A Panel of Lung Injury Biomarkers Enhances the Definition of Primary Graft Dysfunction (PGD) after Lung Transplantation for Early Clinical Studies. J Heart Lung Transpl. 2012;31(4):S103–S103. doi: 10.1016/j.healun.2012.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Christie JD, Kotloff RM, Ahya VN, et al. The effect of primary graft dysfunction on survival after lung transplantation. American journal of respiratory and critical care medicine. 2005;171(11):1312–1316. doi: 10.1164/rccm.200409-1243OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Diamond JM, Lee JC, Kawut SM, et al. Clinical Risk Factors for Primary Graft Dysfunction after Lung Transplantation. American journal of respiratory and critical care medicine. 2013 doi: 10.1164/rccm.201210-1865OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Prekker ME, Nath DS, Walker AR, et al. Validation of the proposed International Society for Heart and Lung Transplantation grading system for primary graft dysfunction after lung transplantation. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2006;25(4):371–378. doi: 10.1016/j.healun.2005.11.436. [DOI] [PubMed] [Google Scholar]
  • 31.Christie JD, Bellamy S, Ware LB, et al. Construct validity of the definition of primary graft dysfunction after lung transplantation. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2010;29(11):1231–1239. doi: 10.1016/j.healun.2010.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kreisel D, Krupnick AS, Puri V, et al. Short- and long-term outcomes of 1000 adult lung transplant recipients at a single center. The Journal of thoracic and cardiovascular surgery. 2011;141(1):215–222. doi: 10.1016/j.jtcvs.2010.09.009. [DOI] [PubMed] [Google Scholar]
  • 33.Daud SA, Yusen RD, Meyers BF, et al. Impact of immediate primary lung allograft dysfunction on bronchiolitis obliterans syndrome. American journal of respiratory and critical care medicine. 2007;175(5):507–513. doi: 10.1164/rccm.200608-1079OC. [DOI] [PubMed] [Google Scholar]
  • 34.Huang HJ, Yusen RD, Meyers BF, et al. Late primary graft dysfunction after lung transplantation and bronchiolitis obliterans syndrome. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2008;8(11):2454–2462. doi: 10.1111/j.1600-6143.2008.02389.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Whitson BA, Prekker ME, Herrington CS, et al. Primary graft dysfunction and long-term pulmonary function after lung transplantation. J Heart Lung Transpl. 2007;26(10):1004–1011. doi: 10.1016/j.healun.2007.07.018. [DOI] [PubMed] [Google Scholar]
  • 36.Christie JD, Edwards LB, Kucheryavaya AY, et al. The Registry of the International Society for Heart and Lung Transplantation: 29th adult lung and heart-lung transplant report-2012. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2012;31(10):1073–1086. doi: 10.1016/j.healun.2012.08.004. [DOI] [PubMed] [Google Scholar]
  • 37.Shah RJ, Diamond JM, Kawut S, et al. Plasma RAGE Levels Measured 24 Hours after Lung Transplantation Are Associated with Bronchiolitis Obliterans Syndrome (BOS). J Heart Lung Transpl. 2012;31(4):S77–S78. [Google Scholar]
  • 38.Whitson BA, Nath DS, Johnson AC, et al. Risk factors for primary graft dysfunction after lung transplantation. The Journal of thoracic and cardiovascular surgery. 2006;131(1):73–80. doi: 10.1016/j.jtcvs.2005.08.039. [DOI] [PubMed] [Google Scholar]
  • 39.Kuntz CL, Hadjiliadis D, Ahya VN, et al. Risk factors for early primary graft dysfunction after lung transplantation: a registry study. Clinical transplantation. 2009;23(6):819–830. doi: 10.1111/j.1399-0012.2008.00951.x. [DOI] [PubMed] [Google Scholar]
  • 40.de Perrot M, Bonser RS, Dark J, et al. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction part III: donor-related risk factors and markers. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2005;24(10):1460–1467. doi: 10.1016/j.healun.2005.02.017. [DOI] [PubMed] [Google Scholar]
  • 41.Barr ML, Kawut SM, Whelan TP, et al. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction part IV: recipient-related risk factors and markers. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2005;24(10):1468–1482. doi: 10.1016/j.healun.2005.02.019. [DOI] [PubMed] [Google Scholar]
  • 42.Fang A, Studer S, Kawut SM, et al. Elevated pulmonary artery pressure is a risk factor for primary graft dysfunction following lung transplantation for idiopathic pulmonary fibrosis. Chest. 2011;139(4):782–787. doi: 10.1378/chest.09-2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Oto T, Griffiths AP, Levvey B, et al. A donor history of smoking affects early but not late outcome in lung transplantation. Transplantation. 2004;78(4):599–606. doi: 10.1097/01.tp.0000131975.98323.13. [DOI] [PubMed] [Google Scholar]
  • 44.Kawut SM, Okun J, Shimbo D, et al. Soluble p-selectin and the risk of primary graft dysfunction after lung transplantation. Chest. 2009;136(1):237–244. doi: 10.1378/chest.08-2697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sommers KE, Griffith BP, Hardesty RL, Keenan RJ. Early lung allograft function in twin recipients from the same donor: risk factor analysis. The Annals of thoracic surgery. 1996;62(3):784–790. doi: 10.1016/s0003-4975(96)00371-2. [DOI] [PubMed] [Google Scholar]
  • 46.Whelan TPM, Dunitz JM, Kelly RF, et al. Effect of preoperative pulmonary artery pressure on early survival after lung transplantation for idiopathic pulmonary fibrosis. J Heart Lung Transpl. 2005;24(9):1269–1274. doi: 10.1016/j.healun.2004.10.014. [DOI] [PubMed] [Google Scholar]
  • 47.Cassivi SD, Meyers BF, Battafarano RJ, et al. Thirteen-year experience in lung transplantation for emphysema. The Annals of thoracic surgery. 2002;74(5):1663–1669. doi: 10.1016/s0003-4975(02)04064-x. discussion 1669-1670. [DOI] [PubMed] [Google Scholar]
  • 48.Fiser SM, Kron IL, McLendon Long S, et al. Early intervention after severe oxygenation index elevation improves survival following lung transplantation. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2001;20(6):631–636. doi: 10.1016/s1053-2498(01)00249-2. [DOI] [PubMed] [Google Scholar]
  • 49.Lederer DJ, Kawut SM, Wickersham N, et al. Obesity and primary graft dysfunction after lung transplantation: the Lung Transplant Outcomes Group Obesity Study. American journal of respiratory and critical care medicine. 2011;184(9):1055–1061. doi: 10.1164/rccm.201104-0728OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.De Oliveira NC, Osaki S, Maloney J, Cornwell RD, Meyer KC. Lung transplant for interstitial lung disease: outcomes for single versus bilateral lung transplantation. Interactive cardiovascular and thoracic surgery. 2012;14(3):263–267. doi: 10.1093/icvts/ivr085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nagendran M, Maruthappu M, Sugand K. Should double lung transplant be performed with or without cardiopulmonary bypass? Interactive cardiovascular and thoracic surgery. 2011;12(5):799–804. doi: 10.1510/icvts.2010.263624. [DOI] [PubMed] [Google Scholar]
  • 52.Dalibon N, Geffroy A, Moutafis M, et al. Use of cardiopulmonary bypass for lung transplantation: a 10-year experience. Journal of cardiothoracic and vascular anesthesia. 2006;20(5):668–672. doi: 10.1053/j.jvca.2006.01.004. [DOI] [PubMed] [Google Scholar]
  • 53.Szeto WY, Kreisel D, Karakousis GC, et al. Cardiopulmonary bypass for bilateral sequential lung transplantation in patients with chronic obstructive pulmonary disease without adverse effect on lung function or clinical outcome. The Journal of thoracic and cardiovascular surgery. 2002;124(2):241–249. doi: 10.1067/mtc.2002.121303. [DOI] [PubMed] [Google Scholar]
  • 54.Aigner C. eComment: To pump or not to pump in lung transplantation--question solved? Interactive cardiovascular and thoracic surgery. 2011;12(5):805. doi: 10.1510/icvts.2010.263624A. [DOI] [PubMed] [Google Scholar]
  • 55.Webert KE, Blajchman MA. Transfusion-related acute lung injury. Current opinion in hematology. 2005;12(6):480–487. doi: 10.1097/01.moh.0000177829.85904.39. [DOI] [PubMed] [Google Scholar]
  • 56.Silliman CC, Boshkov LK, Mehdizadehkashi Z, et al. Transfusion-related acute lung injury: epidemiology and a prospective analysis of etiologic factors. Blood. 2003;101(2):454–462. doi: 10.1182/blood-2002-03-0958. [DOI] [PubMed] [Google Scholar]
  • 57.Silliman CC, Fung YL, Ball JB, Khan SY. Transfusion-related acute lung injury (TRALI): current concepts and misconceptions. Blood reviews. 2009;23(6):245–255. doi: 10.1016/j.blre.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mangalmurti NS, Xiong Z, Hulver M, et al. Loss of red cell chemokine scavenging promotes transfusion-related lung inflammation. Blood. 2009;113(5):1158–1166. doi: 10.1182/blood-2008-07-166264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Bonser RS, Taylor R, Collett D, et al. Effect of donor smoking on survival after lung transplantation: a cohort study of a prospective registry. Lancet. 2012;380(9843):747–755. doi: 10.1016/S0140-6736(12)60160-3. [DOI] [PubMed] [Google Scholar]
  • 60.Eppinger MJ, Jones ML, Deeb GM, Bolling SF, Ward PA. Pattern of injury and the role of neutrophils in reperfusion injury of rat lung. The Journal of surgical research. 1995;58(6):713–718. doi: 10.1006/jsre.1995.1112. [DOI] [PubMed] [Google Scholar]
  • 61.Eppinger MJ, Deeb GM, Bolling SF, Ward PA. Mediators of ischemia-reperfusion injury of rat lung. The American journal of pathology. 1997;150(5):1773–1784. [PMC free article] [PubMed] [Google Scholar]
  • 62.Fiser SM, Tribble CG, Long SM, et al. Pulmonary macrophages are involved in reperfusion injury after lung transplantation. The Annals of thoracic surgery. 2001;71(4):1134–1138. doi: 10.1016/s0003-4975(01)02407-9. discussion 1138-1139. [DOI] [PubMed] [Google Scholar]
  • 63.Naidu BV, Krishnadasan B, Farivar AS, et al. Early activation of the alveolar macrophage is critical to the development of lung ischemia-reperfusion injury. The Journal of thoracic and cardiovascular surgery. 2003;126(1):200–207. doi: 10.1016/s0022-5223(03)00390-8. [DOI] [PubMed] [Google Scholar]
  • 64.Yang ZQ, Sharma AK, Linden J, Kron IL, Laubach VE. CD4(+) T lymphocytes mediate acute pulmonary ischemia-reperfusion injury. J Thorac Cardiov Sur. 2009;137(3):695–702. doi: 10.1016/j.jtcvs.2008.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.van der Kaaij NP, Kluin J, Haitsma JJ, et al. Ischemia of the lung causes extensive long-term pulmonary injury: an experimental study. Respiratory research. 2008;9:28. doi: 10.1186/1465-9921-9-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Sharma AK, Lapar DJ, Zhao YG, et al. Natural Killer T Cell-derived IL-17 Mediates Lung Ischemia-Reperfusion Injury. American journal of respiratory and critical care medicine. 2011;183(11):1539–1549. doi: 10.1164/rccm.201007-1173OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Johnston LK, Rims CR, Gill SE, McGuire JK, Manicone AM. Pulmonary macrophage subpopulations in the induction and resolution of acute lung injury. American journal of respiratory cell and molecular biology. 2012;47(4):417–426. doi: 10.1165/rcmb.2012-0090OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.McCourtie AS, Merry HE, Farivar AS, Goss CH, Mulligan MS. Alveolar macrophage secretory products augment the response of rat pulmonary artery endothelial cells to hypoxia and reoxygenation. The Annals of thoracic surgery. 2008;85(3):1056–1060. doi: 10.1016/j.athoracsur.2007.10.058. [DOI] [PubMed] [Google Scholar]
  • 69.McCourtie AS, Farivar AS, Woolley SM, et al. Alveolar macrophage secretory products effect type 2 pneumocytes undergoing hypoxia-reoxygenation. The Annals of thoracic surgery. 2008;86(6):1774–1779. doi: 10.1016/j.athoracsur.2008.07.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Serrick C, Adoumie R, Giaid A, Shennib H. The early release of interleukin-2, tumor necrosis factor-alpha and interferon-gamma after ischemia reperfusion injury in the lung allograft. Transplantation. 1994;58(11):1158–1162. [PubMed] [Google Scholar]
  • 71.Mal H, Dehoux M, Sleiman C, et al. Early release of proinflammatory cytokines after lung transplantation. Chest. 1998;113(3):645–651. doi: 10.1378/chest.113.3.645. [DOI] [PubMed] [Google Scholar]
  • 72.Fisher AJ, Donnelly SC, Hirani N, et al. Elevated levels of interleukin-8 in donor lungs is associated with early graft failure after lung transplantation. American journal of respiratory and critical care medicine. 2001;163(1):259–265. doi: 10.1164/ajrccm.163.1.2005093. [DOI] [PubMed] [Google Scholar]
  • 73.Akashi A, Nakahara K, Kamiike W, et al. Attenuation of Warm Ischemic-Injury of Rat Lung by Inflation with Room Air - Assessment of Cellular-Components and the Surfactant in the Bronchoalveolar Lavage Fluid in Relation to Changes in Cellular Adenosine-Triphosphate. Transplantation. 1993;55(1):24–30. doi: 10.1097/00007890-199301000-00006. [DOI] [PubMed] [Google Scholar]
  • 74.Date H, Matsumura A, Manchester JK, et al. Changes in alveolar oxygen and carbon dioxide concentration and oxygen consumption during lung preservation. The maintenance of aerobic metabolism during lung preservation. The Journal of thoracic and cardiovascular surgery. 1993;105(3):492–501. [PubMed] [Google Scholar]
  • 75.Sakuma T, Takahashi K, Ohya N, et al. Ischemia-reperfusion lung injury in rabbits: mechanisms of injury and protection. The American journal of physiology. 1999;276(1 Pt 1):L137–145. doi: 10.1152/ajplung.1999.276.1.L137. [DOI] [PubMed] [Google Scholar]
  • 76.Fukuse T, Hirata T, Liu CJ, Hitomi S, Wada H. Energy metabolism and reperfusion injury in warm and cold ischemia of inflated and deflated lungs. Transplantation proceedings. 2000;32(7):2424–2425. doi: 10.1016/s0041-1345(00)01726-7. [DOI] [PubMed] [Google Scholar]
  • 77.Chatterjee S, Chapman K, Fisher A. Lung Ischemia: A Model for Endothelial Mechanotransduction. Cell biochemistry and biophysics. 2008;52(3):125–138. doi: 10.1007/s12013-008-9030-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Browning EA, Chatterjee S, Fisher AB. Stop the flow: a paradigm for cell signaling mediated by reactive oxygen species in the pulmonary endothelium. Annual review of physiology. 2012;74:403–424. doi: 10.1146/annurev-physiol-020911-153324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Fisher AB, Al-Mehdi AB, Manevich Y. Shear stress and endothelial cell activation. Critical care medicine. 2002;30(5 Suppl):S192–197. doi: 10.1097/00003246-200205001-00004. [DOI] [PubMed] [Google Scholar]
  • 80.Fukuse T, Hirata T, Ishikawa S, et al. Optimal alveolar oxygen concentration for cold storage of the lung. Transplantation. 2001;72(2):300–304. doi: 10.1097/00007890-200107270-00024. [DOI] [PubMed] [Google Scholar]
  • 81.Van Raemdonck DE, Jannis NC, De Leyn PR, Flameng WJ, Lerut TE. Alveolar expansion itself but not continuous oxygen supply enhances postmortem preservation of pulmonary grafts. European journal of cardio-thoracic surgery : official journal of the European Association for Cardio-thoracic Surgery. 1998;13(4):431–440. doi: 10.1016/s1010-7940(98)00046-3. discussion 440-431. [DOI] [PubMed] [Google Scholar]
  • 82.Rauen U, de Groot H. Mammalian cell injury induced by hypothermia - the emerging role for reactive oxygen species. Biol Chem. 2002;383(3-4):477–488. doi: 10.1515/BC.2002.050. [DOI] [PubMed] [Google Scholar]
  • 83.Burlingham WJ, Love RB, Jankowska-Gan E, et al. IL-17-dependent cellular immunity to collagen type V predisposes to obliterative bronchiolitis in human lung transplants. Journal of Clinical Investigation. 2007;117(11):3498–3506. doi: 10.1172/JCI28031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bharat A, Narayanan K, Street T, et al. Early posttransplant inflammation promotes the development of alloimmunity and chronic human lung allograft rejection. Transplantation. 2007;83(2):150–158. doi: 10.1097/01.tp.0000250579.08042.b6. [DOI] [PubMed] [Google Scholar]
  • 85.Bharat A, Kuo E, Steward N, et al. Immunological link between primary graft dysfunction and chronic lung allograft rejection. The Annals of thoracic surgery. 2008;86(1):189–195. doi: 10.1016/j.athoracsur.2008.03.073. discussion 196-187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Bharat A, Saini D, Steward N, et al. Antibodies to self-antigens predispose to primary lung allograft dysfunction and chronic rejection. The Annals of thoracic surgery. 2010;90(4):1094–1101. doi: 10.1016/j.athoracsur.2010.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pham SM, Yoshida Y, Aeba R, et al. Interleukin-6, a marker of preservation injury in clinical lung transplantation. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 1992;11(6):1017–1024. [PubMed] [Google Scholar]
  • 88.Mathur A, Baz M, Staples ED, et al. Cytokine profile after lung transplantation: correlation with allograft injury. The Annals of thoracic surgery. 2006;81(5):1844–1849. doi: 10.1016/j.athoracsur.2005.11.053. discussion 1849-1850. [DOI] [PubMed] [Google Scholar]
  • 89.De Perrot M, Sekine Y, Fischer S, et al. Interleukin-8 release during early reperfusion predicts graft function in human lung transplantation. American journal of respiratory and critical care medicine. 2002;165(2):211–215. doi: 10.1164/ajrccm.165.2.2011151. [DOI] [PubMed] [Google Scholar]
  • 90.Colombat M, Castier Y, Leseche G, et al. Early expression of adhesion molecules after lung transplantation: evidence for a role of aggregated P-selectin-positive platelets in human primary graft failure. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2004;23(9):1087–1092. doi: 10.1016/j.healun.2003.08.020. [DOI] [PubMed] [Google Scholar]
  • 91.Abraham D, Taghavi S, Riml P, et al. VEGF-A and -C but not -B mediate increased vascular, permeability in preserved lung grafts. Transplantation. 2002;73(11):1703–1706. doi: 10.1097/00007890-200206150-00003. [DOI] [PubMed] [Google Scholar]
  • 92.Krenn K, Klepetko W, Taghavi S, et al. Recipient vascular endothelial growth factor serum levels predict primary lung graft dysfunction. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2007;7(3):700–706. doi: 10.1111/j.1600-6143.2006.01673.x. [DOI] [PubMed] [Google Scholar]
  • 93.Christie JD, Robinson N, Ware LB, et al. Association of protein C and type 1 plasminogen activator inhibitor with primary graft dysfunction. American journal of respiratory and critical care medicine. 2007;175(1):69–74. doi: 10.1164/rccm.200606-827OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Covarrubias M, Ware LB, Kawut SM, et al. Plasma intercellular adhesion molecule-1 and von Willebrand factor in primary graft dysfunction after lung transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2007;7(11):2573–2578. doi: 10.1111/j.1600-6143.2007.01981.x. [DOI] [PubMed] [Google Scholar]
  • 95.Bobadilla JL, Love RB, Jankowska-Gan E, et al. Th-17, monokines, collagen type V, and primary graft dysfunction in lung transplantation. American journal of respiratory and critical care medicine. 2008;177(6):660–668. doi: 10.1164/rccm.200612-1901OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Iwata T, Philipovskiy A, Fisher AJ, et al. Anti-type V collagen humoral immunity in lung transplant primary graft dysfunction. Journal of immunology. 2008;181(8):5738–5747. doi: 10.4049/jimmunol.181.8.5738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hoffman SA, Wang L, Shah CV, et al. Plasma cytokines and chemokines in primary graft dysfunction post-lung transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2009;9(2):389–396. doi: 10.1111/j.1600-6143.2008.02497.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Moreno I, Vicente R, Mir A, et al. Effects of inhaled nitric oxide on primary graft dysfunction in lung transplantation. Transplantation proceedings. 2009;41(6):2210–2212. doi: 10.1016/j.transproceed.2009.05.019. [DOI] [PubMed] [Google Scholar]
  • 99.Christie JD, Shah CV, Kawut SM, et al. Plasma levels of receptor for advanced glycation end products, blood transfusion, and risk of primary graft dysfunction. American journal of respiratory and critical care medicine. 2009;180(10):1010–1015. doi: 10.1164/rccm.200901-0118OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pelaez A, Force SD, Gal AA, et al. Receptor for advanced glycation end products in donor lungs is associated with primary graft dysfunction after lung transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2010;10(4):900–907. doi: 10.1111/j.1600-6143.2009.02995.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Salama M, Andrukhova O, Hoda MA, et al. Concomitant endothelin-1 overexpression in lung transplant donors and recipients predicts primary graft dysfunction. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2010;10(3):628–636. doi: 10.1111/j.1600-6143.2009.02957.x. [DOI] [PubMed] [Google Scholar]
  • 102.Diamond JM, Kawut SM, Lederer DJ, et al. Elevated plasma clara cell secretory protein concentration is associated with high-grade primary graft dysfunction. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2011;11(3):561–567. doi: 10.1111/j.1600-6143.2010.03431.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Diamond JM, Lederer DJ, Kawut SM, et al. Elevated plasma long pentraxin-3 levels and primary graft dysfunction after lung transplantation for idiopathic pulmonary fibrosis. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2011;11(11):2517–2522. doi: 10.1111/j.1600-6143.2011.03702.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Diamond JM, Porteous MK, Cantu E, et al. Elevated plasma angiopoietin-2 levels and primary graft dysfunction after lung transplantation. PloS one. 2012;7(12):e51932. doi: 10.1371/journal.pone.0051932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Bastarache JA, Diamond JM, Kawut SM, et al. Postoperative estradiol levels associate with development of primary graft dysfunction in lung transplantation patients. Gender medicine. 2012;9(3):154–165. doi: 10.1016/j.genm.2012.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Shah RJ, Diamond JM, Lederer DJ, et al. Plasma monocyte chemotactic protein-1 levels at 24 hours are a biomarker of primary graft dysfunction after lung transplantation. Translational research : the journal of laboratory and clinical medicine. 2012;160(6):435–442. doi: 10.1016/j.trsl.2012.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Shah RJ, Bellamy SL, Localio AR, et al. A panel of lung injury biomarkers enhances the definition of primary graft dysfunction (PGD) after lung transplantation. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2012;31(9):942–949. doi: 10.1016/j.healun.2012.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Taghavi S, Abraham D, Riml P, et al. Co-expression of endothelin-1 and vascular endothelial growth factor mediates increased vascular permeability in lung grafts before reperfusion. J Heart Lung Transpl. 2002;21(5):600–603. doi: 10.1016/s1053-2498(01)00346-1. [DOI] [PubMed] [Google Scholar]
  • 109.Kaneda H, Waddell TK, de Perrot M, et al. Pre-implantation multiple cytokine mRNA expression analysis of donor lung grafts predicts survival after lung transplantation in humans. American Journal of Transplantation. 2006;6(3):544–551. doi: 10.1111/j.1600-6143.2005.01204.x. [DOI] [PubMed] [Google Scholar]
  • 110.Ray M, Dharmarajan S, Freudenberg J, Zhang W, Patterson GA. Expression profiling of human donor lungs to understand primary graft dysfunction after lung transplantation. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2007;7(10):2396–2405. doi: 10.1111/j.1600-6143.2007.01918.x. [DOI] [PubMed] [Google Scholar]
  • 111.Anraku M, Cameron MJ, Waddell TK, et al. Impact of human donor lung gene expression profiles on survival after lung transplantation: a case-control study. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2008;8(10):2140–2148. doi: 10.1111/j.1600-6143.2008.02354.x. [DOI] [PubMed] [Google Scholar]
  • 112.Hagedorn PH, Burton CM, Sahar E, et al. Integrative analysis correlates donor transcripts to recipient autoantibodies in primary graft dysfunction after lung transplantation. Immunology. 2011;132(3):394–400. doi: 10.1111/j.1365-2567.2010.03373.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Diamond JM, Meyer NJ, Feng R, et al. Variation in PTX3 is associated with primary graft dysfunction after lung transplantation. American journal of respiratory and critical care medicine. 2012;186(6):546–552. doi: 10.1164/rccm.201204-0692OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Shargall Y, Guenther G, Ahya VN, et al. Report of the ISHLT Working Group on Primary Lung Graft Dysfunction part VI: treatment. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2005;24(10):1489–1500. doi: 10.1016/j.healun.2005.03.011. [DOI] [PubMed] [Google Scholar]
  • 115.Van Raemdonck D, Neyrinck A, Verleden GM, et al. Lung donor selection and management. Proceedings of the American Thoracic Society. 2009;6(1):28–38. doi: 10.1513/pats.200808-098GO. [DOI] [PubMed] [Google Scholar]
  • 116.de Perrot M, Keshavjee S. Lung preservation. Seminars in thoracic and cardiovascular surgery. 2004;16(4):300–308. doi: 10.1053/j.semtcvs.2004.09.012. [DOI] [PubMed] [Google Scholar]
  • 117.Meade MO, Granton JT, Matte-Martyn A, et al. A randomized trial of inhaled nitric oxide to prevent ischemia-reperfusion injury after lung transplantation. American journal of respiratory and critical care medicine. 2003;167(11):1483–1489. doi: 10.1164/rccm.2203034. [DOI] [PubMed] [Google Scholar]
  • 118.Botha P, Jeyakanthan M, Rao JN, et al. Inhaled nitric oxide for modulation of ischemia-reperfusion injury in lung transplantation. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2007;26(11):1199–1205. doi: 10.1016/j.healun.2007.08.008. [DOI] [PubMed] [Google Scholar]
  • 119.Keshavjee S, Davis RD, Zamora MR, de Perrot M, Patterson GA. A randomized, placebo-controlled trial of complement inhibition in ischemia-reperfusion injury after lung transplantation in human beings. J Thorac Cardiov Sur. 2005;129(2):423–428. doi: 10.1016/j.jtcvs.2004.06.048. [DOI] [PubMed] [Google Scholar]
  • 120.Wittwer T, Grote M, Oppelt P, et al. Impact of PAF antagonist BN 52021 (Ginkolide B) on post-ischemic graft function in clinical lung transplantation. J Heart Lung Transpl. 2001;20(3):358–363. doi: 10.1016/s1053-2498(00)00226-6. [DOI] [PubMed] [Google Scholar]
  • 121.Herrington CS, Prekker ME, Arrington AK, et al. A randomized, placebo-controlled trial of aprotinin to reduce primary graft dysfunction following lung transplantation. Clinical transplantation. 2011;25(1):90–96. doi: 10.1111/j.1399-0012.2010.01319.x. [DOI] [PubMed] [Google Scholar]
  • 122.Struber M, Fischer S, Niedermeyer J, et al. Effects of exogenous surfactant instillation in clinical lung transplantation: A prospective, randomized trial. J Thorac Cardiov Sur. 2007;133(6):1620–1625. doi: 10.1016/j.jtcvs.2006.12.057. [DOI] [PubMed] [Google Scholar]
  • 123.Novick RJ, Stitt LW, Al-Kattan K, et al. Pulmonary retransplantation: Predictors of graft function and survival in 230 patients. Annals of Thoracic Surgery. 1998;65(1):227–234. doi: 10.1016/s0003-4975(97)01191-0. [DOI] [PubMed] [Google Scholar]
  • 124.Currey J, Pilcher DV, Davies A, et al. Implementation of a management guideline aimed at minimizing the severity of primary graft dysfunction after lung transplant. The Journal of thoracic and cardiovascular surgery. 2010;139(1):154–161. doi: 10.1016/j.jtcvs.2009.08.031. [DOI] [PubMed] [Google Scholar]
  • 125.Struber M, Harringer W, Ernst M, et al. Inhaled nitric oxide as a prophylactic treatment against reperfusion injury of the lung. Thorac Cardiov Surg. 1999;47(3):179–182. doi: 10.1055/s-2007-1013137. [DOI] [PubMed] [Google Scholar]
  • 126.Date H, Triantafillou AN, Trulock EP, et al. Inhaled nitric oxide reduces human lung allograft dysfunction. J Thorac Cardiov Sur. 1996;111(5):913–919. doi: 10.1016/s0022-5223(96)70364-1. [DOI] [PubMed] [Google Scholar]
  • 127.Adatia I, Lillehei C, Arnold JH, et al. Inhaled nitric oxide in the treatment of postoperative graft dysfunction after lung transplantation. The Annals of thoracic surgery. 1994;57(5):1311–1318. doi: 10.1016/0003-4975(94)91382-x. [DOI] [PubMed] [Google Scholar]
  • 128.Macdonald P, Mundy J, Rogers P, et al. Successful treatment of life-threatening acute reperfusion injury after lung transplantation with inhaled nitric oxide. The Journal of thoracic and cardiovascular surgery. 1995;110(3):861–863. doi: 10.1016/S0022-5223(95)70125-7. [DOI] [PubMed] [Google Scholar]
  • 129.Meyers BF, Sundt TM, Henry S, et al. Selective use of extracorporeal membrane oxygenation is warranted after lung transplantation. J Thorac Cardiov Sur. 2000;120(1):20–28. doi: 10.1067/mtc.2000.105639. [DOI] [PubMed] [Google Scholar]
  • 130.Glassman LR, Keenan RJ, Fabrizio MC, et al. Extracorporeal membrane oxygenation as an adjunct treatment for primary graft failure in adult lung transplant recipients. The Journal of thoracic and cardiovascular surgery. 1995;110(3):723–726. doi: 10.1016/S0022-5223(95)70104-4. discussion 726-727. [DOI] [PubMed] [Google Scholar]
  • 131.Hartwig MG, Appel JZ, Cantu E, et al. Improved results treating lung allograft failure with venovenous extracorporeal membrane oxygenation. Annals of Thoracic Surgery. 2005;80(5):1872–1880. doi: 10.1016/j.athoracsur.2005.04.063. [DOI] [PubMed] [Google Scholar]
  • 132.Bermudez CA, Adusumilli PS, McCurry KR, et al. Extracorporeal membrane oxygenation for primary graft dysfunction after lung transplantation: long-term survival. The Annals of thoracic surgery. 2009;87(3):854–860. doi: 10.1016/j.athoracsur.2008.11.036. [DOI] [PubMed] [Google Scholar]
  • 133.Hartwig MG, Walczak R, Lin SS, Davis RD. Improved survival but marginal allograft function in patients treated with extracorporeal membrane oxygenation after lung transplantation. The Annals of thoracic surgery. 2012;93(2):366–371. doi: 10.1016/j.athoracsur.2011.05.017. [DOI] [PubMed] [Google Scholar]
  • 134.Naik PM, Angel LF. Special issues in the management and selection of the donor for lung transplantation. Seminars in immunopathology. 2011;33(2):201–210. doi: 10.1007/s00281-011-0256-x. [DOI] [PubMed] [Google Scholar]
  • 135.Kron IL, Tribble CG, Kern JA, et al. Successful transplantation of marginally acceptable thoracic organs. Annals of surgery. 1993;217(5):518–522. doi: 10.1097/00000658-199305010-00012. discussion 522-514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Sundaresan S, Semenkovich J, Ochoa L, et al. Successful outcome of lung transplantation is not compromised by the use of marginal donor lungs. The Journal of thoracic and cardiovascular surgery. 1995;109(6):1075–1079. doi: 10.1016/S0022-5223(95)70190-7. discussion 1079-1080. [DOI] [PubMed] [Google Scholar]
  • 137.Gabbay E, Williams TJ, Griffiths AP, et al. Maximizing the utilization of donor organs offered for lung transplantation. American journal of respiratory and critical care medicine. 1999;160(1):265–271. doi: 10.1164/ajrccm.160.1.9811017. [DOI] [PubMed] [Google Scholar]
  • 138.Bhorade SM, Vigneswaran W, McCabe MA, Garrity ER. Liberalization of donor criteria may expand the donor pool without adverse consequence in lung transplantation. J Heart Lung Transpl. 2000;19(12):1199–1204. doi: 10.1016/s1053-2498(00)00215-1. [DOI] [PubMed] [Google Scholar]
  • 139.Straznicka M, Follette DM, Eisner MD, et al. Aggressive management of lung donors classified as unacceptable: excellent recipient survival one year after transplantation. The Journal of thoracic and cardiovascular surgery. 2002;124(2):250–258. doi: 10.1067/mtc.2002.123813. [DOI] [PubMed] [Google Scholar]
  • 140.Pierre AF, Sekine Y, Hutcheon MA, Waddell TK, Keshavjee SH. Marginal donor lungs: a reassessment. The Journal of thoracic and cardiovascular surgery. 2002;123(3):421–427. doi: 10.1067/mtc.2002.120345. discussion, 427-428. [DOI] [PubMed] [Google Scholar]
  • 141.Lardinois D, Banysch M, Korom S, et al. Extended donor lungs: eleven years experience in a consecutive series. Eur J Cardio-Thorac. 2005;27(5):762–767. doi: 10.1016/j.ejcts.2005.01.019. [DOI] [PubMed] [Google Scholar]
  • 142.Aigner C, Winkler G, Jaksch P, et al. Extended donor criteria for lung transplantation--a clinical reality. European journal of cardio-thoracic surgery : official journal of the European Association for Cardio-thoracic Surgery. 2005;27(5):757–761. doi: 10.1016/j.ejcts.2005.01.024. [DOI] [PubMed] [Google Scholar]
  • 143.Botha P, Trivedi D, Weir CJ, et al. Extended donor criteria in lung transplantation: impact on organ allocation. The Journal of thoracic and cardiovascular surgery. 2006;131(5):1154–1160. doi: 10.1016/j.jtcvs.2005.12.037. [DOI] [PubMed] [Google Scholar]
  • 144.Meers C, Van Raemdonck D, Verleden GM, et al. The number of lung transplants can be safely doubled using extended criteria donors; a single-center review. Transplant international : official journal of the European Society for Organ Transplantation. 2010;23(6):628–635. doi: 10.1111/j.1432-2277.2009.01033.x. [DOI] [PubMed] [Google Scholar]
  • 145.de Perrot M, Weder W, Patterson GA, Keshavjee S. Strategies to increase limited donor resources. The European respiratory journal : official journal of the European Society for Clinical Respiratory Physiology. 2004;23(3):477–482. doi: 10.1183/09031936.04.00094504. [DOI] [PubMed] [Google Scholar]
  • 146.Cypel M, Yeung JC, Liu MY, et al. Normothermic Ex Vivo Lung Perfusion in Clinical Lung Transplantation. New Engl J Med. 2011;364(15):1431–1440. doi: 10.1056/NEJMoa1014597. [DOI] [PubMed] [Google Scholar]
  • 147.Cypel M, Yeung JC, Hirayama S, et al. Technique for prolonged normothermic ex vivo lung perfusion. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2008;27(12):1319–1325. doi: 10.1016/j.healun.2008.09.003. [DOI] [PubMed] [Google Scholar]
  • 148.Steen S, Sjoberg T, Pierre L, et al. Transplantation of lungs from a non-heartbeating donor. Lancet. 2001;357(9259):825–829. doi: 10.1016/S0140-6736(00)04195-7. [DOI] [PubMed] [Google Scholar]
  • 149.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. Interactive cardiovascular and thoracic surgery. 2011;12(2):162–165. doi: 10.1510/icvts.2010.244830. [DOI] [PubMed] [Google Scholar]
  • 150.Cypel M, Yeung JC, Machuca T, et al. Experience with the first 50 ex vivo lung perfusions in clinical transplantation. The Journal of thoracic and cardiovascular surgery. 2012;144(5):1200–1206. doi: 10.1016/j.jtcvs.2012.08.009. [DOI] [PubMed] [Google Scholar]
  • 151.Zych B, Popov AF, Stavri G, et al. Early outcomes of bilateral sequential single lung transplantation after ex-vivo lung evaluation and reconditioning. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2012;31(3):274–281. doi: 10.1016/j.healun.2011.10.008. [DOI] [PubMed] [Google Scholar]
  • 152.Aigner C, Slama A, Hotzenecker K, et al. Clinical ex vivo lung perfusion--pushing the limits. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2012;12(7):1839–1847. doi: 10.1111/j.1600-6143.2012.04027.x. [DOI] [PubMed] [Google Scholar]
  • 153.Valenza F, Rosso L, Gatti S, et al. Extracorporeal lung perfusion and ventilation to improve donor lung function and increase the number of organs available for transplantation. Transplantation proceedings. 2012;44(7):1826–1829. doi: 10.1016/j.transproceed.2012.06.023. [DOI] [PubMed] [Google Scholar]
  • 154.Wallinder A, Ricksten SE, Hansson C, et al. Transplantation of initially rejected donor lungs after ex vivo lung perfusion. The Journal of thoracic and cardiovascular surgery. 2012;144(5):1222–1228. doi: 10.1016/j.jtcvs.2012.08.011. [DOI] [PubMed] [Google Scholar]
  • 155.Cypel M, Liu M, Rubacha M, et al. Functional repair of human donor lungs by IL-10 gene therapy. Sci Transl Med. 2009;1(4):4ra9. doi: 10.1126/scitranslmed.3000266. [DOI] [PubMed] [Google Scholar]
  • 156.Warnecke G, Moradiellos J, Tudorache I, et al. Normothermic perfusion of donor lungs for preservation and assessment with the Organ Care System Lung before bilateral transplantation: a pilot study of 12 patients. Lancet. 2012;380(9856):1851–1858. doi: 10.1016/S0140-6736(12)61344-0. [DOI] [PubMed] [Google Scholar]

RESOURCES