Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
Next Article in Journal
Prospective Study to Evaluate Rectus Femoris Muscle Ultrasound for Body Composition Analysis in Patients Undergoing Bariatric Surgery
Previous Article in Journal
The Fate of Mitral Valve Surgery in the Pediatric Age: A 25-Year Single-Center Experience
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancements in Predictive Tools for Primary Graft Dysfunction in Liver Transplantation: A Comprehensive Review

Department of General Transplant and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(13), 3762; https://doi.org/10.3390/jcm13133762
Submission received: 29 May 2024 / Revised: 20 June 2024 / Accepted: 24 June 2024 / Published: 27 June 2024
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)

Abstract

:

Highlights

What are the main findings?
  • Primary graft dysfunction (PGD) involves early allograft dysfunction (EAD) and more severe primary nonfunction (PNF), both stemming from ischemia–reperfusion injury (IRI).
  • Accurate and early diagnosis of PGD is crucial to the retransplantation decision-making process and the implementation of future mitigation strategies.
  • Novel tools for predicting PNF utilize serum markers, marker-derived models, tissue biopsy analysis, metabolomics, evaluations of organ perfusion and liver metabolism, and assessments of graft perfusate during machine perfusion.
What is the implication of the main finding?
  • The current implementation of extended criteria donors (ECD) exacerbates the limitations of the binary EAD criteria and has prompted a refreshed approach to the graft dysfunction assessment protocol.
  • Recently, new serum parameters have been associated with EAD occurrence, and their incorporation into graft dysfunction evaluations may improve accuracy.

Abstract

Orthotopic liver transplantation stands as the sole curative solution for end-stage liver disease. Nevertheless, the discrepancy between the demand and supply of grafts in transplant medicine greatly limits the success of this treatment. The increasing global shortage of organs necessitates the utilization of extended criteria donors (ECD) for liver transplantation, thereby increasing the risk of primary graft dysfunction (PGD). Primary graft dysfunction (PGD) encompasses early allograft dysfunction (EAD) and the more severe primary nonfunction (PNF), both of which stem from ischemia–reperfusion injury (IRI) and mitochondrial damage. Currently, the only effective treatment for PNF is secondary transplantation within the initial post-transplant week, and the occurrence of EAD suggests an elevated, albeit still uncertain, likelihood of retransplantation urgency. Nonetheless, the ongoing exploration of novel IRI mitigation strategies offers hope for future improvements in PGD outcomes. Establishing an intuitive and reliable tool to predict upcoming graft dysfunction is vital for early identification of high-risk patients and for making informed retransplantation decisions. Accurate diagnostics for PNF and EAD constitute essential initial steps in implementing future mitigation strategies. Recently, novel methods for PNF prediction have been developed, and several models for EAD assessments have been introduced. Here, we provide an overview of the currently scrutinized predictive tools for PNF and EAD evaluation strategies, accompanied by recommendations for future studies.

1. Introduction

The incongruity between the demand and supply of organs in transplant medicine has resulted in a mortality rate of approximately 20–25% among patients on the waiting list for liver transplantation, thus necessitating the use of extended criteria donors (ECD) [1]. To address the challenges of organ scarcity and to improve outcomes for patients awaiting hepatic transplantation, donors with features, such as macrovesicular steatosis, viral infections, or post circulatory death (DCD), have been incorporated into the donor pool. However, graft quality has emerged as a critical determinant of postoperative liver performance, and the likelihood of primary graft dysfunction (PGD) has been assessed as higher among recipients of ECD-derived livers [2].
Primary graft dysfunction (PGD) comprises early allograft dysfunction (EAD) and more severe primary nonfunction (PNF). Although there is no consensus regarding the definition of EAD, a widely accepted description was proposed by Olthoff et al. in 2010 to encompass at least one of the following during the first 7 days after transplantation: bilirubin concentration ≥ 10 mg/dL on day 7, INR ≥ 1.6 on day 7, AST or ALT activity > 2000 U/L during the initial 7 days [3]. The occurrence of EAD in post-transplant patients is associated with higher mortality rates and diminished graft survival. Primary nonfunction is typically defined as death or retransplantation within the first week after surgery, without other discernible causes (e.g., infection or graft rejection).
Although EAD and PNF describe different clinical outcomes, the underlying cause is believed to be similar and is attributed to both ischemia–reperfusion injury (IRI) and mitochondrial damage [4]. Retransplantation can often be avoided in the case of EAD, but it remains the only effective treatment for PNF. However, alternative mitigation strategies, such as inhaled nitric oxide or the administration of prostaglandins, are being actively explored [5]. Furthermore, the development of hypothermic oxygenated perfusion (HOPE) and normothermic regional perfusion (NRP) has improved graft quality in ECD and introduced a novel opportunity for organ assessment and the prediction of postoperative complications [6].
Establishing an intuitive and reliable tool to foresee upcoming graft dysfunction is crucial for early high-risk patient identification and an appropriate retransplantation decision-making process. Accurate diagnostics for PNF and EAD are essential initial steps for implementing future mitigation strategies. Recently, novel methods for PNF prediction have been developed, and several models for EAD assessment have been introduced. Here, we provide an overview of the currently scrutinized predictive tools for PNF and EAD evaluation strategies, accompanied by recommendations for future studies.
Several of the tools described in this article were evaluated based on their c-statistic performance. The c-statistic, also referred to as the concordance statistic or the area under the receiver operating characteristic curve (AUC-ROC), gauges the discriminatory power of a predictive model. In essence, it measures how effectively a model distinguishes between patients with a specific condition and those without a specific condition. A c-statistic value of 0.5 indicates no discriminatory power, while a value of 1.0 signifies perfect discrimination.

2. Ischemia–Reperfusion Injury (IRI) on the Cellular Level

Primary graft dysfunction (PGD) is associated with ischemia–reperfusion injury (IRI), which is caused by the disruption and subsequent restoration of blood supply during organ procurement and liver transplantation. A comprehensive grasp of the underlying mechanisms responsible for IRI is essential for the intentional development of diagnostic tools. Its complex cellular and molecular processes not only impact the liver itself but also result in systemic injury. Numerous participants, including hepatocytes, neutrophils, Kupffer cells, platelets, hepatic stellate cells (HSCs), and liver sinusoidal endothelial cells (LSECs), are involved in the occurrence of IRI [7].
Termination of the initial blood supply decreases the amount of oxygen received by hepatocytes and liver sinusoidal endothelial cells (LSECs), leading to mitochondrial damage, ATP depletion, and increased lactate accumulation [8]. This results in IRI-induced hepatocyte and LSEC death accompanied by the release of inflammatory cytokines (IL-1β, IL-6, TNFα, TGFβ), damage-associated molecular patterns (DAMPs), and DNA fragments. Resident liver macrophages (Kupffer cells) stimulated by these factors increase toll-like receptor (TLR) expression and initiate chemokine 8 (CXCL8) secretion, which binds to complement proteins (C3a and C5a) derived from damaged LSECs and hepatocytes, and induces the migration of neutrophils to the liver as the organ reperfuses [9,10]. Kupffer cells also release IL-1β and TNFα, which promote adhesion between neutrophils (CD11b/CD18a) and LSECs (ICAM-1) [11]. The further neutrophil degranulation and reactive oxygen species (ROS) secretion, stimulated mainly by DAMPs and DNA from the damaged cells, exacerbate the destruction of liver tissue [12]. At the same time, increased P-selectin expression on the surface of the epithelium activates platelets, which release thromboxane 2A (TXA2), plasminogen activator inhibitor-1 (PAI-1), and TGFβ, leading to thrombosis and HSC-dependent graft fibrosis [13,14]. Figure 1 depicts the cellular mechanism behind the liver IRI.

3. Prediction of Primary Nonfunction

3.1. Serum Lactate Concentration

Ischemia and the impaired oxygen supplementation of hepatocytes result in insufficient ATP synthesis in mitochondria [7]. The lack of ATP, which is the main energy currency of the cell, prevents the metabolism of lactate within the hepatocyte. This inhibits the lactic acid cycle (also known as the Cori cycle) in which lactate, produced mainly in muscles, is converted to pyruvate and used for gluconeogenesis (Figure 2). This leads to hyperlactatemia, which is further increased by reperfusion injury, damaging the hepatocyte lactate transporters (MCT1, MCT2) and disrupting the uptake of lactates in the liver [15]. Therefore, the serum lactate concentration may correlate with the severity of IRI and indicate upcoming PGD.
A practical application of this theory was recently described by Golse et al. [16], who observed that the intraoperative arterial lactate concentration shortly after liver reperfusion could serve as an early predictor of PGD. In their study on a group of 296 patients, a lactate concentration ≥ 5 mmol/L at the end of liver transplantation predicted PNF with a sensitivity of 83.3% and a specificity of 74.3%. Furthermore, the addition of lactate measurements to the BAR-score improved its predictive power regarding 3-month patient survival from an increase in the c-statistic of 0.74 to 0.87.
On the other hand, Galli et al. [17], in a larger cohort of 1137 patients, obtained a sensitivity of 61% and specificity of 68% for PNF prediction when a cutoff of 5 mmol/L was applied and a sensitivity of 44% and specificity of 96% for arterial blood lactate ≥ 9.5 mmol/L.
In another study, conducted in a pediatric liver transplantation group, higher lactate levels were also reported among children experiencing PNF [18]. Moreover, a postoperative serum lactate level > 3 mmol/L correlated with biliary complications and mortality, with an AUROC of 0.73 and 0.72, respectively, in a group of 145 patients.
The lactate concentration is a promising prognostic tool for PNF prediction; however, further research and validation in large cohort studies are required. Its main advantages are simplicity, accessibility, and the possibility of early calculation. Furthermore, similar tools that rely on serum lactate levels, such as the lactate-to-platelet ratio and the lactate-to-venous-arterial CO2 and O2 ratio, have not yet been comprehensively assessed in terms of PNF diagnostics [19,20].

3.2. Scoring Models and Alternative Serum Markers

Inflammation and graft fibrosis resulting from IRI exert an omnidirectional systemic effect. PNF cases, such as those with severe IRI, should mirror these changes in laboratory findings associated with liver function. Al-Freah et al. [21] developed a PNF-predicting model based on a combination of the albumin concentration at the time of transplant surgery, lactate level and AST activity on posttransplant day 1, INR value and bilirubin concentration on day 3, and AST activity on day 7. They introduced the King’s College Hospital PNF score (King-PNF score) for a group of 1286 patients undergoing liver transplantation, of whom 3.7% were diagnosed with postoperative PNF. The proposed model achieved a c-statistic of 0.912 for the training cohort and 0.831 for the validation cohort, outperforming previously developed systems. However, PNF has been defined as death or retransplantation within the first two weeks following surgery, which deviates from the commonly proposed timeframe of one week. In an external validation conducted by Nie et al. [22], the King-PNF score predicted PNF occurrence with an AUROC of 0.891, superseding both the MEAF and lactate-adjusted BAR score (Table 1).
Although the King-PNF score remains superior to other models in terms of accuracy, its calculation still requires 7 postoperative days (3 days in the modified version), which is a major obstacle to early PNF diagnostics and is at a disadvantage when compared to arterial blood lactate measurements. Several recent studies have suggested that other parameters that may improve PNF prediction models include C-reactive protein (CRP), serum urea, and procalcitonin [23,24]. In addition, a study by Fukazawa et al. [25] showed that the predictive power of serum markers could be improved when adjusted for donor-recipient size mismatch. A novel target was proposed by Li et al. [26], who observed that levels of extracellular histones (released during IRI-induced cell death along with DNA and DAMPs) were significantly higher in liver recipients experiencing PGD during the first 72 h after surgery.

3.3. Assessment during Machine Perfusion

The development of hypothermic oxygenated perfusion (HOPE) and normothermic machine perfusion (NMP) has not only improved graft quality and reduced the risk of severe graft-related complications but also introduced novel opportunities for liver assessments prior to transplantation [27,28]. The graft perfusate can provide crucial information regarding organ viability, potentially identifying patients at a high risk for the occurrence of PNF. Various markers, including AST, ALT, CRP, lactate dehydrogenase (LDH), and hyaluronic acid (HA), have been proposed as prospective targets; however, none of them have been validated or introduced into routine clinical practice [29]. Nevertheless, they still represent a promising topic of interest for future analyses.
Recent articles have advocated for routine liver assessments during ex situ hypothermic oxygenated perfusion to improve the decision-making process on whether to transplant the organ or not. Eden et al. [4] proposed flavin mononucleotide (FMN), nicotinamide adenine dinucleotide + hydrogen (NADH), and mitochondrial CO2 production during machine perfusion for screening mitochondrial functions prior to transplantation. Patrono et al. [30] emphasized that lactate clearance during NMP is a key prerequisite for graft acceptance, especially in low-quality organs. Verhelst et al. reported a very promising analysis of glycome patterns in the perfusate derived from hepatic venous flushing prior to transplantation. A single glycan, agalacto core-alpha-1,6-fucosylated biantennary glycan (NGA2F), was significantly elevated among patients suffering from graft dysfunction and was able to predict PNF with 100% accuracy in a study group of 122 patients [31].
A further investigation of injury markers during the organ-preservation phase and the development of a comprehensive model could enable effective PGD prediction during the pre-transplantation period. Therefore, highly damaged organs can be identified and rejected early, while moderate-risk graft recipients can be carefully monitored in order to implement mitigation strategies or secondary transplantation as early as possible.

3.4. Role of the Liver Biopsy

Another alternative strategy that can identify damaged organs is liver biopsy, thus potentially facilitating early PGD prediction. Among the various methods of tissue analysis, a growing interest has been observed in the use of metabolomic biosignatures for preoperative graft biomarker assessments. Cortes et al. [32] used mass spectrometry coupled with liquid chromatography to measure levels of phospholipids, lysophospholipids, sphingomyelins, bile acids, and histidine metabolism products, revealing impaired lipid and histidine pathways in dysfunctional grafts. This model can predict EAD preoperatively with 91% sensitivity and 82% specificity while accurately classifying all PNF cases into the graft dysfunction group. Recently, Zhang et al. [33] applied metabolomics to create the graft metabolite- and clinical parameter-based PNF (GMCP-PNF) predictive model. This innovative system achieved a c-statistic of 0.965 (100% sensitivity, 85% specificity) for preoperative PNF prediction by incorporating both clinical features (graft weight, warm and cold ischemia time, donor total bilirubin, AST, ALT, anhepatic time, and steatosis) and the metabolomic biosignature of eight key metabolites. Furthermore, they identified 59 disparately expressed metabolic features between EAD and PNF cases, suggesting potential differences in disease etiology between these conditions. In addition to metabolomics, electron microscopy can provide preoperative information about donor liver function by displaying ultrastructural changes indicative of hepatocyte injury [34].
Liver biopsy analysis can also provide crucial information regarding the probability of graft dysfunction when performed directly after organ reperfusion. Severe IRI observed on histopathological examination, including necrosis, steatosis, and monomorphonuclear infiltrates, indicates an elevated likelihood of PGD [35,36,37]. Bruns et al. [38] emphasized that microarray analysis and quantitative real-time PCR can be used for post-reperfusion tissue biopsy evaluation, reporting 20 markers that may be able to predict postoperative graft dysfunction. Khorsandi et al. [39] showed that the microRNA expression profile could shed more light on the prediction of PNF, with miR-22 being of utmost importance. Nevertheless, due to the small size of the study groups in both trials, further research is needed.

3.5. Measurement of Graft Metabolism and Perfusion

IRI-induced mitochondrial damage impairs the metabolic performance of the graft, while platelet aggregation disrupts blood flow and further inhibits metabolic pathways by reducing the oxygen supply. Therefore, measurement of the liver function, as well as graft perfusion, can assess the IRI and the increased risk of PNF occurrence.
This chain of events, primarily metabolic activity, can be evaluated using the maximal enzymatic liver function capacity (LiMAx) test [40]. It measures the level of metabolites in exhaled air after 13C-methacetin administration, which is metabolized by hepatic cytochrome P-450. Lock et al. [41] applied this method to predict critical complications after liver transplantation in a group of 99 patients. Cutoff values of 64 µg/kg/h directly after surgery and 42 µg/kg/h on the first postoperative day (POD) achieved a sensitivity of 1.0 (0.31–1.0) and a specificity of 1.0 (0.94–1.0) for PNF prediction, outperforming the AST activity measurement. Nevertheless, due to the small size of the study group, this encouraging result requires further validation.
Another approach that focuses particularly on the perfusion aspect of graft dysfunction is the indocyanine green fluorescence imaging (ICGFI) technique. Upon intravenous administration, indocyanine green (ICG) passes through the circulatory system bound to plasma proteins until it reaches the liver, where ICG is taken up by hepatocytes and secreted directly into the bile. A near-infrared camera detects fluorescence signals emitted by ICG; thus, the ICG fluorescence pattern can indicate hepatocyte disfunction, as well as impaired blood flow or bile secretion.
Levesque et al. [42] measured the plasma disappearance rate (PDR) of ICG during the first 5 days following liver transplantation and found that a PDR of ICG lower than 12.85%/min was predictive of hepatic artery thrombosis, sepsis, and PNF. They also suggested that a sequential decline in PDR before POD5 might indicate graft acute rejection. A few years later, Olmedilla et al. [43] introduced a scoring system that relied on postoperative ICG clearance. Patients were divided into four categories ranging from 0 to 3 based on the PDR of ICG. The highest scoring group had a 50% risk of early death (within 30 days) or retransplantation (within 7 days), compared to a risk of 4.4%, 6.5%, and 12% for categories 0, 1, and 2, respectively. Recently, Figueroa et al. [44] created an intraoperative classification encompassing three types of ICG fluorescence, which incorporated ICG parenchymal uptake and the homogeneity of perfused areas. Their research showed that abnormal fluorescence patterns recorded immediately after organ reperfusion could identify patients at risk of PGD development.
As an alternative to the ICG clearance, Narasaki et al. [45] recommended measuring the fluorescence intensity (FI) of the liver surface following ICG administration to evaluate its function. Although their study examined patients undergoing hepatopancreatobiliary surgery due to various types of carcinoma, they developed a mathematical model to describe the measured FI curves and calculated the parameters that define the temporal course of FI. Their findings provided the basis for a follow-up study by Dousse et al. [46], who expanded the previous model and introduced a novel parameter called “a150” in liver transplantation settings. This parameter reflects the speed at which the fluorescent signal reaches the plateau phase, and its value is negatively correlated with the time required for the signal to reach the plateau phase (the higher the a150 value, the shorter the time required, and the faster the plateau is achieved). Furthermore, its measurement was restricted to a predefined time period of 150 s. In their study, including 76 liver transplantations, an a150 value ≥ 0.0155 s−1 predicted 3-month graft survival with a sensitivity of 83.3% and a specificity of 78.6%, whereas, an a150 value ≥ 0.0178 s−1 diagnosed PNF with a sensitivity of 75% sensitivity and a specificity of 81.9%. Moreover, the authors reported that an arterial lactate concentration ≥ 5 mmol/L at the end of liver transplantation was a risk factor for the occurrence of PNF (OR = 13.15, p = 0.02) and suggested that the combination of a150 and lactate levels could be used for PNF prediction.
Overall, PDR and FI following ICG administration emerged as a promising tool for assessing postoperative liver function and the early prediction of PGD. However, the ICG pathway strongly depends on graft perfusion and bile production; therefore, its accuracy in PNF diagnostics may be significantly reduced by other conditions, such as hepatic artery thrombosis or biliary complications. In theory, this could limit the specificity of such tests as a predictive tool for PNF, as different outcomes could obtain results above the cutoff threshold.
Nevertheless, disturbed blood flow stands as an important factor in graft failure development. A study by Matsushima et al. [47] conducted on a large study group of 1001 individuals showed that intraoperative portal flow below 65 mL/min/100 g (liver weight) predicted poorer 1-year graft survival and demonstrated an elevated likelihood of severe reperfusion injury, EAD, and PNF. On the other hand, patients with high portal flow (≥155 mL/min/100 g) were more often complicated by hepatic artery thrombosis and biliary disturbances; however, intraoperative hepatic flow was lower in this group. Recently, Zheng et al. [48] proposed that a hemodynamic graft assessment prior to the transplantation procedure relies on Doppler ultrasonography (DUS) and contrast-enhanced ultrasonography (CEUS) performed on donors prior to surgical procurement. They observed that reduced enhancement of donor livers on CEUS is a risk factor for PGD development; therefore, it can provide important information even before organ procurement.
Examinations to assess graft perfusion expand the pool of potential predictive tools for PNF; however, they are unlikely to be able to diagnose PNF alone because of the many factors that influence their outcome, so combinations with some alternative methods may be beneficial. Perhaps the combination of a metabolic measurement and a graft perfusion assessment could provide a universal tool in the future.

4. Assessment of Early Allograft Dysfunction

The primary objective of introducing early allograft dysfunction (EAD) was to develop a tool capable of defining the poor initial function of the transplanted organ, thereby providing an endpoint other than graft loss or the patient’s death. In addition, such a tool would be able to accurately assess which patients are at a high risk of postoperative graft failure, and thus, they require comprehensive postoperative monitoring and early intervention. Despite the unanimous agreement on the need for such parameters, there is no consensus regarding their definition. Currently, the most widely accepted EAD description is that proposed by Olthoff et al. [3] more than a decade ago. They defined EAD as the fulfilment of at least one of the following conditions within the first 7 days after transplantation: bilirubin concentration ≥ 10 mg/dL on day 7, INR ≥ 1.6 on day 7, and AST or ALT activity > 2000 U/L during the initial 7 days. Subsequent research has validated that EAD occurrence (as defined by the criteria of Olthoff et al.) in the posttransplant period is associated with inferior graft outcomes and worse survival rates [49].
Recently, due to global organ shortages, grafts obtained from extended criteria donors (ECD) have been incorporated into clinical practice, resulting in an elevated incidence of EAD [50,51]. However, despite the higher likelihood of EAD in ECD-derived graft recipients, they are not associated with poorer graft or patient survival [50,51,52]. However, this information is inconclusive, as other researchers have observed increased complication rates among ECD recipients [53]. This may be due to the nonhomogeneous definitions of ECD across different articles.
Nonetheless, this issue highlights the major shortcomings of the current EAD definition. While patients undergoing EAD experience elevated rates of mortality and morbidity, the rigid and binary nature of EAD complicates the prediction of postoperative courses. Many patients who meet the EAD criteria achieve long-term survival without significant complications, whereas those who do not fall into the EAD category may encounter postoperative sequelae. Consequently, some authors have advocated for reconsidering the definition of early graft dysfunction by proposing a more flexible and progressive formulation. The overarching goal of this revised approach is to accurately evaluate the risk of graft failure, particularly with regard to early graft failure (EGF), which is usually described as graft loss or the patient’s death within the first 3 months following transplantation. Table 2 provides an overview of alternative models for the assessment of early graft dysfunction that have been developed over the last 10 years. They are compared in terms of their c-statistic for the prediction of EGF. A brief description of the models discussed in this article is shown in the Supplementary Table S1.
The intricacies of predicting postoperative graft failure were already highlighted in 2013 by Wagener et al. [54], who emphasized the limited accuracy and predictive power of the EAD construct. Consequently, they explored alternative solutions, revealing that the Model for End-Stage Liver Disease (MELD), which is widely used in pre-transplant graft allocation, can be used as a predictive tool for early graft loss and mortality when applied on the 5th postoperative day. A follow-up study by Benko [55] supported these findings by achieving an even higher c-statistic by using a MELD score cutoff of 16, compared to 18.9 in the previous article. However, both studies were conducted on relatively small groups of patients (572 and 116, respectively), necessitating further validation. Meanwhile, Angelico et al. [56] introduced the Donor-Recipient Allocation Model (DReAM) in a much larger study group, surpassing some previously developed scoring systems. The c-statistic obtained by DReAM was comparatively modest; however, in the subsequent validation group of 448 patients, this value had improved to 0.76.
In 2015, Pareja et al. [57] described the Model for Early Allograft Function scoring (MEAF) and its potential to predict graft loss or a patient’s death within 90 days of liver transplantation. It is also noteworthy that the likelihood of PNF was significantly elevated with an MEAF score higher than 7. A further study by Jochmans et al. [58] demonstrated that MEAF predicted EGF with a c-statistic of 0.727 (rising to 0.782 in a multivariable model) and outperformed the binary EAD model (AUROC of 0.644). Moreover, Richards et al. [59] highlighted the superior sensitivity of MEAF, as it can be calculated earlier than EAD. Furthermore, a recent external validation by Nie et al. [22] showed that MEAF predicted PNF and EGF with a c-statistic of 0.872 and 0.802, respectively, in a group of 720 patients.
Another model introduced in 2018 by Agopian et al. [60] is the Liver Graft Assessment Following Transplantation (L-GrAFT). In a study group of 2008 patients, there was a better prediction of 90-day graft loss or mortality by AUROC than by the MEAF and EAD models. Further multicenter validation, conducted by the same research team, showed c-statistics of 0.78, 0.72, and 0.68 for L-GrAFT, MEAF, and EAD, respectively, in a study group comprising 3201 patients, as well as 0.81, 0.57, and 0.64 in a study group of 171 participants [61].
One of the most recent models for the assessment of postoperative graft dysfunction was developed by Avolio et al. [62] for the Early Allograft Failure Simplified Estimation (EASE) score. The c-index of this model was 0.87 and 0.78 in two study groups, 1609 and 538, respectively, outperforming both L-GrAFT and MEAF scores.
Other systems for assessing graft dysfunction developed in the last decade include the ABC model introduced by Rhu et al. [63], as well as the system proposed by Diaz-Nieto et al. [64]. The Diaz-Nieto model relies on postoperative serum transaminase activity and stratifies patients into four groups with a different risk of 30-day graft failure. The strengths of this system include the possibility of early evaluation and a relatively straightforward calculation process. Nevertheless, both models either lack external validation or behave similarly to the binary EAD model.
Overall, most newly developed systems are more accurate than binary EAD, even when adjusted for the graft-to-recipient weight ratio [65]. The most impressive c-statistic among the graft dysfunction assessment models was achieved with the Comprehensive Complication Index (CCI), predicting 90-day graft loss with an AUROC of 0.94; however, the c-statistic in the validation group was significantly lower, yielding 0.77 [66]. Nevertheless, contemporary literature posits that the L-GrAFT and EASE scores are emerging as the most auspicious indicators for EGF prognostication [67]. The dynamic MEAF model outperformed the binary EAD definition in terms of EGF prediction accuracy and took less time to calculate; however, MEAF achieved a lower c-statistic in almost every study compared to the EASE and L-GrAFT models. Interestingly, the King-PNF score also performed excellently in EGF prediction [22]. Moreover, MELD calculated based on POD5 is a promising tool as it obtains a high c-statistic; however, an external validation with a large study group is required. Another advantage of the postoperative MELD calculation is that it has been widely used and recognized worldwide.
Table 2. Overview of models developed over the past 10 years to assess postoperative graft dysfunction. These systems were compared in terms of their predictive power for early graft failure (EGF), which is understood to be patient death or graft loss within the first 90 days following surgery.
Table 2. Overview of models developed over the past 10 years to assess postoperative graft dysfunction. These systems were compared in terms of their predictive power for early graft failure (EGF), which is understood to be patient death or graft loss within the first 90 days following surgery.
YearStudy [Ref.]SizeOutcomeIncidenceModelc-Statistic
2013Wagener et al. [54]572EGF7%MELD0.81
2014Angelico et al. [56]2864EGF7%BAR0.57
D-MELD0.58
SOFT0.59
DReAM0.66
2015Pareja et al. [57]829EGF10%MEAFno data
2017Benko et al. [55]116EGF10%MELD0.84
2017Jochmans et al. [58]660EGF12%MEAF0.73
EAD0.64
2018Agopian et al. [60]2008EGF11%L-GrAFT0.83
MEAF0.70
EAD0.68
2019Diaz-Nieto et al. [64]1194Graft failure within 30 days9%Diaz-Nieto scoreno data
2020Richards et al. [59]183Graft failure within 28 days8%MEAF0.74
2020Avolio et al. [62]1609EGF7%EASE0.87
DRIno data
EAD0.72
D-MELD0.70
ET-DRI0.63
MEAFno data
L-GrAFT0.85
5388%EASE0.78
DRI0.57
EAD0.63
D-MELD0.72
ET-DRI0.58
MEAF0.73
L-GrAFT0.71
2021Lai et al. [66]1262EGF15%CCI0.94
MELD0.60
D-MELD0.60
BAR0.60
EAD0.58
5203%CCI0.77
MELD0.57
D-MELD0.57
BAR0.56
EAD0.47
2021Agopian et al. [61]3201EGF7%L-GrAFT0.78
MEAF0.72
EAD0.68
1714%L-GrAFT0.81
MEAF0.57
EAD0.64
2021Rhu et al. [63]1153Graft failure within 2 months7%ABC Model0.73
MEAF0.69
EAD0.66
35913%ABC Model0.74
MEAF0.71
EAD0.66
2022Manzia et al. [65]331EGF16%mEAD0.74
EAD0.64
1235%mEAD0.68
EAD0.52
2023Moosburner [67]906EGFno dataDRI0.50
ET-DRI0.54
D-MELD0.59
MEAF0.72
L-GrAFT0.80
EASE0.80
ABC Model0.68
EAD0.69
BAR0.60
2023Nie et al. [22]720EGF9%MEAF0.80
King-PNF score0.87
BAR-Lac0.76

Alternative Assessment of Allograft Dysfunction

On the other hand, most of the currently available EAD assessment models rely on overlapping parameters; however, they vary in terms of formula calculations. Meanwhile, an ongoing study has revealed novel laboratory markers associated with EAD occurrence. Implementing some of these findings into the EAD assessment model may improve its accuracy. Therefore, in Table 3, we provide an overview of serum parameters that have been reported to be associated with EAD development over the past 10 years. Furthermore, if any of these markers were validated in a large external study while maintaining a high discriminatory power, it would supersede pre-existing EAD assessment models due to the lower complexity and earlier calculation. These parameters also represent a promising spectrum of targets for further PNF prediction research. The majority of these markers are derived from current knowledge regarding the IRI mechanism at the cellular level.
Nunez et al. [68] observed that an elevated serum level of the complement anaphylatoxins C3a and C5a correlated with graft steatosis and EAD occurrence; however, it did not achieve a significant effect. They also noted that interleukin-33 (IL-33) is one of the damage-associated molecular patterns (DAMPs) released by IRI-injured hepatic cells, and its elevation corresponds to graft dysfunction [69]. A further study by Barbier et al. [70] demonstrated an increase in the concentration of IL-33 in patients undergoing EAD. In a group of 40 patients, IL-33 possessed a c-statistic value of 0.76 for predicting graft dysfunction. The role of IL-33 in the IRI mechanism is currently under scrutiny. In addition to serving as a DAMP, recent studies have shown that IL-33, which is mainly released from LSECs, stimulates inflammation by increasing neutrophil extracellular trap formation while possessing a protective effect on hepatocytes [71,72].
In addition, the interleukin-6 (IL-6) concentration may possess a predictive value as it is also released from damaged liver cells and plays a significant role in inflammation signaling. Faitot et al. [73] reported that the IL-6 level at the time of reperfusion correlates with graft survival. However, some older articles describe its limited potential to predict postoperative outcomes; therefore, further research is needed [74,75]. Interestingly, Chae et al. [76] observed an elevated EAD incidence among patients with increased preoperative IL-6.
Karakhanova et al. [77] noted that the preoperative concentration of interferon-gamma (IFNɣ) correlated with postoperative life expectancy and predicted EAD with an AUROC of 0.76. They also observed that EAD cases expressed higher levels of interleukin-10 (IL-10) and chemokine-10 (CXCL10) within the initial postoperative days and that the c-statistic for IL-10 on EAD prognostication amounted to 0.84. These findings are particularly interesting because it is currently believed that inflammatory cytokines and DAMPs, released by IRI-affected apoptotic cells, activate Kupffer cells to produce an inflammatory phenotype and inhibit the production of immunosuppressive IL-10 [7]. Perhaps its increased concentration is derived from regulatory T cells, which secrete IL-10 in response to IFNɣ released by Kupffer cells [7,77,78]. Furthermore, Karakhanova et al. [76], in contrast to Chae et al. [77], did not observe a preoperative change in IL-6 between the EAD and non-EAD groups.
A recent study by Faria et al. [79] showed that elevated high-mobility group box 1 protein (HMGB1) and nucleosomes belonging to DAMPs correlated with EAD development and were significantly higher in PNF cases; however, due to the small size of the study group, this could be a random effect [79]. This corresponds to a study by Sosa et al. [80], which showed that the HMGB1 concentration correlated with the severity of IRI.
In addition, the increase in macrophage and neutrophil activity after reperfusion may reveal more clearly the severity of IRI. In a study group of 1960 participants, Kwon et al. [81] observed that EAD patients had increased neutrophil levels compared to the lymphocyte number. A neutrophil-to-lymphocyte ratio (NLR) ≥ 2.85 had a sensitivity of 64% and a specificity of 70% for EAD diagnostics. Thomsen et al. [82] noted that macrophage involvement can be measured based on soluble macrophage activation marker (sCD163) levels, which are elevated in EAD patients. Park et al. [83] proposed an increased C-reactive-protein-to-albumin ratio (CRP/ALB) as an EAD marker, as it should reflect ongoing inflammatory changes.
As an alternative to the immune response observation, the analysis of lipid metabolism is a very promising approach to assess graft dysfunction. Ceglarek et al. [84] investigated circulating liver metabolites of the cholesterol pathway, amino acids, and acylcarnitines in the plasma of graft recipients. By applying liquid chromatography/tandem mass spectrometry (LC-MS/MS), they observed that EAD cases were associated with lower circulating esterified sterol concentrations. The usefulness of metabolomics in liver transplantation was subsequently supported by Tsai et al. [85], who achieved an excellent c-statistic of 0.95 for EAD prediction by combining levels of cholesterol oleate, phosphatidylcholine (PC), and lysophosphatidylcholine (LysoPC). A few years later, the same research team found that the combination of betaine, palmitic acid, PC, and LysoPC could differentiate between EAD and non-EAD cases with an AUROC of 0.82, which was further increased to 0.85 by incorporating total bilirubin [86]. Furthermore, Yang et al. [87] observed that serum total cholesterol (sTC) below 1.42 mmol/L on POD3 correlated with a higher EAD incidence. These results indicate the potential of metabolomics in graft dysfunction evaluations; however, due to the small size of the study groups, these findings require further validation.
A study by Pollara et al. [88] showed that preoperative EAD prediction could be predicted by deceased donor plasma mitochondrial DNA (mtDNA) level measurements. The elevated mtDNA serum concentration is suspected to correlate with posttransplant complications because it may serve as a DAMP and amplify the immune response in graft recipients. Thus, it may simultaneously play a causative role and may be released from cells damaged by IRI. Further research by Yoshino et al. [89] noted elevated circulating mtDNA levels in graft recipients undergoing EAD.
Other parameters recently associated with EAD occurrence include phosphorus, B-type natriuretic peptide (BNP), and myoglobin (Mb), which are increased in EAD cases [90,91,92]. In a study by Chae et al. [91], a serum BNP level ≥ 100 pg/mL predicted EAD with an AUROC of 0.75, whereas Mb obtained a c-statistic of 0.66 in a study by Zhang et al. [92]. Gorgen et al. [93] observed a decrease in serum factor V on POD1 in EAD cases, whereas Nedel et al. [94] reported lower thrombin-activatable fibrinolysis inhibitor (TAFI) among patients suffering from graft dysfunction.
Overall, recent studies have expanded the spectrum of parameters associated with EAD occurrence and may improve the graft dysfunction assessment protocol. Some of them (e.g., serum analysis using metabolomics) have achieved excellent accuracy in EAD prediction, whereas others (e.g., mtDNA measurement) can provide crucial information even before organ procurement. However, the majority of these studies have been conducted in small study groups, so further research is needed. Nevertheless, if some of these markers, such as lipid pathway metabolites, confirm their accuracy in large-scale studies, they will significantly improve graft viability assessments. Moreover, some of the parameters may enhance the currently used EAD assessment models, of which the performance is still very limited.
Table 3. Overview of serum markers reported to be associated with EAD development over the past 10 years. ↑—increased concentration, ↓—decreased concentration.
Table 3. Overview of serum markers reported to be associated with EAD development over the past 10 years. ↑—increased concentration, ↓—decreased concentration.
YearStudy [Ref.]SizeOutcomeIncidenceMarkers
2013Hong et al. [90]304EAD16%↑ phosphorus
2015Nedel et al. [94]21EAD10%↓ TAFI
2016Karakhanova et al. [77]41EAD49%↓ IFNɣ, ↑ IL-10, ↑ CXCL10
2016Chae et al. [91]104EAD30%↑ BNP
2016Ceglarek et al. [84]40MEAF ≥ 6.1no data↓ SIE%/↓ CHE%
2017Yang et al. [87]231EAD17%↓ sTC
2018Chae et al. [76]226EAD12%↑ IL-6, ↑ IL-17
2018Faitot et al. [73]274EAD29%↑ IL-6
2018Pollara et al. [88]65EADno data↑ mtDNA
2019Gorgen et al. [93]227EAD27%↓ factor V
2018Tsai et al. [85]51EAD24%↓ cholesterol oleate, ↓ LysoPC, ↑ PC
2019Kwon et al. [81]1960EAD11%↑ NLR
2019Thomsen et al. [82]27EAD59%↑ sCD163
2019Park et al. [83]588EAD14%↑ CRP/ALB
2020Faria et al. [79]22EAD50%↑ HMGB1, ↑ nucleosome
2020Nunez et al. [68]99EADno data↑ IL-33, ↑ C3a, C5a
2021Tsai et al. [86]74EAD30%↑ betaine, ↓ LysoPC, ↓ PC, ↑ palmitic acid
2021Yoshino et al. [89]21EAD33%↑ cmtDNA
2021Barbier et al. [70]40EADno data↑ IL-33
2022Zhang et al. [92]150EAD35%↑ Mb
Abbreviations: thrombin-activatable fibrinolysis inhibitor (TAFI), interferon-gamma (IFNɣ), interleukin (IL), chemokine (CXCL), B-type natriuretic peptide (BNP), esterified β-sitosterol (SIE), esterified cholesterol (CHE), serum total cholesterol (sTC), mitochondrial DNA (mtDNA), lysophosphatidylcholine (LysoPC), phosphatidylcholine (PC), neutrophil-to-lymphocyte ratio (NLR), soluble macrophage activation marker (sCD163), C-reactive protein to albumin ratio (CRP/ALB), high-mobility group box 1 protein (HMGB1), complement anaphylatoxins (C3a, C5a), circulation mitochondrial DNA (cmtDNA), myoglobin (Mb).

5. Conclusions and Recommendations

Despite the significant advances that have been made in the last decade, there is still no universal tool for primary nonfunction (PNF) prediction. Among the predictive models, the KING-PNF score seems to be the most accurate; however, the time required for its calculation limits its usefulness. The measurement of arterial blood lactate concentrations shortly after reperfusion offers a promising alternative due to its accessibility and the possibility of early evaluation, although current knowledge of its predictive power is inconclusive. In terms of accuracy, metabolomics and liver function capacity tests are the most encouraging solutions; nevertheless, further validation in large study groups is needed. Moreover, in the context of the increased utilization of machine perfusion, preoperative graft viability assessment emerges as a promising diagnostic window. Organ function evaluation could be improved by incorporating novel markers that have recently been associated with early allograft dysfunction (EAD) but have not yet been described in terms of PNF prediction. Furthermore, these parameters could improve currently available EAD-assessment models. Cytokine profiling has emerged as a prospective addition, and serum metabolomics also provide an excellent c-statistic for EAD prediction. Despite growing evidence of the need for more accurate and dynamic EAD assessment models, there is no unanimously accepted grading system. The inconclusiveness between outcome-defining studies hampers the evaluation of introduced models and markers, thus making it impossible to perform a reliable meta-analysis. Another factor exacerbating the analysis is the ongoing development of new systems without a thorough examination of some previous ones. Nonetheless, contemporary literature recognizes the EASE and L-GrAFT scores as the most accurate methods for early graft dysfunction assessments, and the MEAF remains a more efficient tool than the binary EAD definition. Furthermore, the incorporation of some novel serum markers may significantly improve their outcomes. This review summarizes current knowledge regarding primary graft dysfunction (PGD) prediction and lays the foundation for future studies on PNF prediction and EAD assessment improvements.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm13133762/s1, Table S1: A brief description of the graft dysfunction assessment models discussed in this article.

Author Contributions

Conceptualization, P.G. and M.R.; methodology, P.G.; software, M.R.; validation, W.F. and M.G.; formal analysis, P.G.; resources, M.R.; data curation, P.G.; writing—original draft preparation, P.G. and M.R.; writing—review and editing, W.F. and M.G.; visualization, M.R.; supervision, M.G. All authors have read and agreed to the published version of the manuscript.

Funding

We confirm that no financial support was received for this article.

Conflicts of Interest

The authors have no conflicts of interest to disclose.

References

  1. Kwong, A.; Kim, W.R.; Lake, J.R.; Smith, J.M.; Schladt, D.P.; Skeans, M.A.; Noreen, S.M.; Foutz, J.; Miller, E.; Snyder, J.J.; et al. OPTN/SRTR 2018 annual data report: Liver. Am. J. Transplant. 2020, 20, 193–299. [Google Scholar] [CrossRef] [PubMed]
  2. Foley, D.P.; Fernandez, L.A.; Leverson, G.; Chin, L.T.; Krieger, N.; Cooper, J.T.; Shames, B.D.; Becker, Y.T.; Odorico, J.S.; Knechtle, S.J.; et al. Donation after cardiac death: The University of Wisconsin experience with liver transplantation. Ann. Surg. 2005, 242, 724–731. [Google Scholar] [CrossRef]
  3. Olthoff, K.M.; Kulik, L.; Samstein, B.; Kaminski, M.; Abecassis, M.; Emond, J.; Shaked, A.; Christie, J.D. Validation of a current definition of early allograft dysfunction in liver transplant recipients and analysis of risk factors. Liver Transpl. 2010, 16, 943–949. [Google Scholar] [CrossRef] [PubMed]
  4. Eden, J.; Breuer, E.; Birrer, D.; Müller, M.; Pfister, M.; Mayr, H.; Sun, K.; Widmer, J.; Huwyler, F.; Ungethüm, U.; et al. Screening for mitochondrial function before use-routine liver assessment during hypothermic oxygenated perfusion impacts liver utilization. EBioMedicine 2023, 98, 104857. [Google Scholar] [CrossRef] [PubMed]
  5. Masior, Ł.; Grąt, M. Primary Nonfunction and Early Allograft Dysfunction after Liver Transplantation. Dig. Dis. 2022, 40, 766–776. [Google Scholar] [CrossRef] [PubMed]
  6. Tang, G.; Zhang, L.; Xia, L.; Zhang, J.; Wei, Z.; Zhou, R. Hypothermic oxygenated perfusion in liver transplantation: A meta-analysis of randomized controlled trials and matched studies. Int. J. Surg. 2023, 110, 464–477. [Google Scholar] [CrossRef] [PubMed]
  7. Dar, W.A.; Sullivan, E.; Bynon, J.S.; Eltzschig, H.; Ju, C. Ischaemia reperfusion injury in liver transplantation: Cellular and molecular mechanisms. Liver Int. 2019, 39, 788–801. [Google Scholar] [CrossRef] [PubMed]
  8. Gonzalez-Flecha, B.; Cutrin, J.C.; Boveris, A. Time course and mechanism of oxidative stress and tissue damage in rat liver subjected to in vivo ischemia-reperfusion. J. Clin. Investig. 1993, 91, 456–464. [Google Scholar] [CrossRef]
  9. Tsung, A.; Klune, J.R.; Zhang, X.; Jeyabalan, G.; Cao, Z.; Peng, X.; Stolz, D.B.; Geller, D.A.; Rosengart, M.R.; Billiar, T.R.; et al. HMGB1 release induced by liver ischemia involves Toll-like receptor 4 dependent reactive oxygen species production and calcium-mediated signaling. J. Exp. Med. 2007, 204, 2913–2923. [Google Scholar] [CrossRef]
  10. Jaeschke, H.; Farhood, A.; Bautista, A.P.; Spolarics, Z.; Spitzer, J.J. Complement activates Kupffer cells and neutrophils during reperfusion after hepatic ischemia. Am. J. Physiol. 1993, 264, G801–G809. [Google Scholar] [CrossRef]
  11. Heymann, F.; Peusquens, J.; Ludwig-Portugall, I.; Kohlhepp, M.; Ergen, C.; Niemietz, P.; Martin, C.; van Rooijen, N.; Ochando, J.C.; Randolph, G.J.; et al. Liver inflammation abrogates immunological tolerance induced by Kupffer cells. Hepatology 2015, 62, 279–291. [Google Scholar] [CrossRef] [PubMed]
  12. Roh, Y.S.; Zhang, B.; Loomba, R.; Seki, E. TLR2 and TLR9 contribute to alcohol-mediated liver injury through induction of CXCL1 and neutrophil infiltration. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G30–G41. [Google Scholar] [CrossRef] [PubMed]
  13. Sindram, D.; Porte, R.J.; Hoffman, M.R.; Bentley, R.C.; Clavien, P.A. Platelets induce sinusoidal endothelial cell apoptosis upon reperfusion of the cold ischemic rat liver. Gastroenterology 2000, 118, 183–191. [Google Scholar] [CrossRef]
  14. Cheng, F.; Li, Y.; Feng, L.; Li, S. Hepatic stellate cell activation and hepatic fibrosis induced by ischemia/reperfusion injury. Transpl. Proc. 2008, 40, 2167–2170. [Google Scholar] [CrossRef] [PubMed]
  15. Kulik, U.; Moesta, C.; Spanel, R.; Borlak, J. Dysfunctional Cori and Krebs cycle and inhibition of lactate transporters constitute a mechanism of primary nonfunction of fatty liver allografts. Transl. Res. 2023, 264, 33–65. [Google Scholar] [CrossRef] [PubMed]
  16. Golse, N.; Guglielmo, N.; El Metni, A.; Frosio, F.; Cosse, C.; Naili, S.; Ichai, P.; Ciacio, O.; Pittau, G.; Allard, M.A.; et al. Arterial Lactate Concentration at the End of Liver Transplantation Is an Early Predictor of Primary Graft Dysfunction. Ann. Surg. 2019, 270, 131–138. [Google Scholar] [CrossRef] [PubMed]
  17. Galli, A.M.; Kothari, R.; Adelmann, D.; Holm, Z.; Bokoch, M.P.; De Gasperi, A.; Niemann, C.U.; Kolodzie, K. Lactate concentration at the end of liver transplant: Early predictor of graft function or just one piece of the puzzle? Clin. Transplant. 2023, 37, e15057. [Google Scholar] [CrossRef] [PubMed]
  18. Fernández-Sarmiento, J.; Wilches-Cuadros, M.A.; Hernandez-Sarmiento, R.; Mulett, H.; Moreno-Medina, K.; Molano, N.; Dominguez, J.A.P.; Acevedo, L.; Salinas, C.; Rivera, J.; et al. Association Between Serum Lactate and Unsatisfactory Outcomes in Critically Ill Children in the Immediate Post-operative Period of Liver Transplantation. Front. Pediatr. 2022, 9, 796504. [Google Scholar] [CrossRef] [PubMed]
  19. Sáez de la Fuente, I.; Sáez de la Fuente, J.; Molina Collado, Z.; Chacon Alves, S.; Sanchez-Bayton Griffith, M.; Gonzalez de Aledo, A.L.; Barea Mendoza, J.; Sanchez-Izquierdo Riera, J.A.; Garcia de Lorenzo, A.; Montejo Gonzalez, J.C.; et al. Combination of arterial lactate levels and Cv-aCO2/Da-vO2 ratio to predict early allograft dysfunction after liver transplantation. Clin. Transplant. 2021, 35, e14482. [Google Scholar] [CrossRef]
  20. Takahashi, K.; Nagai, S.; Gosho, M.; Kitajima, T.; Kim, J.; Oda, T.; Abouljoud, M. The Lactate-to-Platelet Ratio: A Novel Predictor for Short-Term Early Allograft Failure After Liver Transplantation. Transpl. Proc. 2021, 53, 2993–2999. [Google Scholar] [CrossRef]
  21. Al-Freah, M.A.B.; McPhail, M.J.W.; Dionigi, E.; Foxton, M.R.; Auzinger, G.; Rela, M.; Wendon, J.A.; O’Grady, J.G.; Heneghan, M.A.; Heaton, N.D.; et al. Improving the Diagnostic Criteria for Primary Liver Graft Nonfunction in Adults Utilizing Standard and Transportable Laboratory Parameters: An Outcome-Based Analysis. Am. J. Transplant. 2017, 17, 1255–1266. [Google Scholar] [CrossRef] [PubMed]
  22. Nie, Y.; Huang, J.B.; He, S.J.; Chen, H.D.; Jia, J.J.; Li, J.J.; He, X.S.; Zhao, Q. Validation and performance of three scoring systems for predicting primary non-function and early allograft failure after liver transplantation. Hepatobiliary Pancreat. Dis. Int. 2023. [Google Scholar] [CrossRef] [PubMed]
  23. Halle-Smith, J.M.; Hall, L.; Hann, A.; Arshad, A.; Armstrong, M.J.; Bangash, M.N.; Murphy, N.; Cuell, J.; Isaac, J.L.; Ferguson, J.; et al. Low C-reactive Protein and Urea Distinguish Primary Nonfunction from Early Allograft Dysfunction within 48 Hours of Liver Transplantation. Transplant. Direct 2023, 9, e1484. [Google Scholar] [CrossRef] [PubMed]
  24. Frick, K.; Beller, E.A.; Kalisvaart, M.; Dutkowski, P.; Schüpbach, R.A.; Klinzing, S. Procalcitonin in early allograft dysfunction after orthotopic liver transplantation: A retrospective single centre study. BMC Gastroenterol. 2022, 22, 404. [Google Scholar] [CrossRef]
  25. Fukazawa, K.; Nishida, S.; Pretto, E.A., Jr. Peak Serum AST Is a Better Predictor of Acute Liver Graft Injury after Liver Transplantation When Adjusted for Donor/Recipient BSA Size Mismatch (ASTi). J. Transplant. 2014, 2014, 351984. [Google Scholar] [CrossRef] [PubMed]
  26. Li, X.; Gou, C.; Pang, Y.; Wang, Y.; Liu, Y.; Wen, T. Extracellular histones are clinically associated with primary graft dysfunction in human liver transplantation. RSC Adv. 2019, 9, 10264–10271, Erratum in RSC Adv. 2019, 9, 26435–26435. [Google Scholar] [CrossRef]
  27. Schlegel, A.; Mueller, M.; Muller, X.; Eden, J.; Panconesi, R.; von Felten, S.; Steigmiller, K.; Sousa Da Silva, R.X.; de Rougemont, O.; Mabrut, J.-Y.; et al. A multicenter randomized-controlled trial of hypothermic oxygenated perfusion (HOPE) for human liver grafts before transplantation. J. Hepatol. 2023, 78, 783–793. [Google Scholar] [CrossRef]
  28. Mugaanyi, J.; Dai, L.; Lu, C.; Mao, S.; Huang, J.; Lu, C. A Meta-Analysis and Systematic Review of Normothermic and Hypothermic Machine Perfusion in Liver Transplantation. J. Clin. Med. 2022, 12, 235. [Google Scholar] [CrossRef]
  29. Verhoeven, C.J.; Farid, W.R.; de Jonge, J.; Metselaar, H.J.; Kazemier, G.; van der Laan, L.J. Biomarkers to assess graft quality during conventional and machine preservation in liver transplantation. J. Hepatol. 2014, 61, 672–684. [Google Scholar] [CrossRef]
  30. Patrono, D.; De Carlis, R.; Gambella, A.; Farnesi, F.; Podestà, A.; Lauterio, A.; Tandoi, F.; De Carlis, L.; Romagnoli, R. Viability assessment and transplantation of fatty liver grafts using end-ischemic normothermic machine perfusion. Liver Transplant. 2023, 29, 508–520. [Google Scholar] [CrossRef]
  31. Verhelst, X.; Geerts, A.; Jochmans, I.; Vanderschaeghe, D.; Paradissis, A.; Vanlander, A.; Berrevoet, F.; Dahlqvist, G.; Nevens, F.; Pirenne, J.; et al. Glycome Patterns of Perfusate in Livers Before Transplantation Associate With Primary Nonfunction. Gastroenterology 2018, 154, 1361–1368. [Google Scholar] [CrossRef]
  32. Cortes, M.; Pareja, E.; García-Cañaveras, J.C.; Donato, M.T.; Montero, S.; Mir, J.; Castell, J.V.; Lahoz, A. Metabolomics discloses donor liver biomarkers associated with early allograft dysfunction. J. Hepatol. 2014, 61, 564–574. [Google Scholar] [CrossRef]
  33. Zhang, X.; Zhang, C.; Huang, H.; Chen, R.; Lin, Y.; Chen, L.; Shao, L.; Liu, J.; Ling, Q. Primary nonfunction following liver transplantation: Learning of graft metabolites and building a predictive model. Clin. Transl. Med. 2021, 11, e483. [Google Scholar] [CrossRef] [PubMed]
  34. Chen, Z.; Lin, X.; Chen, C.; Liao, Y.; Han, M.; He, X.; Ju, W.; Chen, M. Ultrastructural changes of donor livers in liver transplantation indicate hepatocytes injury. Microsc. Res. Tech. 2022, 85, 2251–2258. [Google Scholar] [CrossRef] [PubMed]
  35. Fuentes-Valenzuela, E.; Tejedor-Tejada, J.; García-Pajares, F.; Rubiales, B.M.; Nájera-Muñoz, R.; Maroto-Martín, C.; Sánchez-Delgado, L.; Alonso-Martín, C.; Álvarez, C.A.; Sánchez-Antolín, G. Postreperfusion Liver Biopsy as Predictor of Early Graft Dysfunction and Survival After Orthotopic Liver Transplantation. J. Clin. Exp. Hepatol. 2022, 12, 1133–1141. [Google Scholar] [CrossRef]
  36. Ali, J.M.; Davies, S.E.; Brais, R.J.; Randle, L.V.; Klinck, J.R.; Allison, M.E.; Chen, Y.; Pasea, L.; Harper, S.F.; Pettigrew, G.J. Analysis of ischemia/reperfusion injury in time-zero biopsies predicts liver allograft outcomes. Liver Transpl. 2015, 21, 487–499. [Google Scholar] [CrossRef] [PubMed]
  37. Zanchet, M.V.; Silva, L.L.; Matias, J.E.; Coelho, J.C. Post-Reperfusion Liver Biopsy and Its Value in Predicting Mortality and Graft Dysfunction after Liver Transplantation. Arq. Bras. Cir. Dig. 2016, 29, 189–193. [Google Scholar] [CrossRef] [PubMed]
  38. Bruns, H.; Heil, J.; Schultze, D.; Al Saeedi, M.; Schemmer, P. Early markers of reperfusion injury after liver transplantation: Association with primary dysfunction. Hepatobiliary Pancreat. Dis. Int. 2015, 14, 246–252. [Google Scholar] [CrossRef] [PubMed]
  39. Khorsandi, S.E.; Quaglia, A.; Salehi, S.; Jassem, W.; Vilca-Melendez, H.; Prachalias, A.; Srinivasan, P.; Heaton, N. The microRNA Expression Profile in Donation after Cardiac Death (DCD) Livers and Its Ability to Identify Primary Non Function. PLoS ONE 2015, 10, e0127073. [Google Scholar] [CrossRef]
  40. Stockmann, M.; Lock, J.F.; Riecke, B.; Heyne, K.; Martus, P.; Fricke, M.; Lehmann, S.; Niehues, S.M.; Schwabe, M.; Lemke, A.J.; et al. Prediction of postoperative outcome after hepatectomy with a new bedside test for maximal liver function capacity. Ann. Surg. 2009, 250, 119–125. [Google Scholar] [CrossRef]
  41. Lock, J.F.; Schwabauer, E.; Martus, P.; Videv, N.; Pratschke, J.; Malinowski, M.; Neuhaus, P.; Stockmann, M. Early diagnosis of primary nonfunction and indication for reoperation after liver transplantation. Liver Transpl. 2010, 16, 172–180. [Google Scholar] [CrossRef] [PubMed]
  42. Levesque, E.; Saliba, F.; Benhamida, S.; Ichaï, P.; Azoulay, D.; Adam, R.; Castaing, D.; Samuel, D. Plasma disappearance rate of indocyanine green: A tool to evaluate early graft outcome after liver transplantation. Liver Transpl. 2009, 15, 1358–1364. [Google Scholar] [CrossRef] [PubMed]
  43. Olmedilla, L.; Lisbona, C.J.; Pérez-Peña, J.M.; López-Baena, J.A.; Garutti, I.; Salcedo, M.; Sanz, J.; Tisner, M.; Asencio, J.M.; Fernández-Quero, L.; et al. Early Measurement of Indocyanine Green Clearance Accurately Predicts Short-Term Outcomes after Liver Transplantation. Transplantation 2016, 100, 613–620. [Google Scholar] [CrossRef] [PubMed]
  44. Figueroa, R.; Golse, N.; Alvarez, F.A.; Ciacio, O.; Pittau, G.; Sa Cunha, A.; Cherqui, D.; Adam, R.; Vibert, E. Indocyanine green fluorescence imaging to evaluate graft perfusion during liver transplantation. HPB 2019, 21, 387–392. [Google Scholar] [CrossRef] [PubMed]
  45. Narasaki, H.; Noji, T.; Wada, H.; Ebihara, Y.; Tsuchikawa, T.; Okamura, K.; Tanaka, E.; Shichinohe, T.; Hirano, S. Intraoperative Real-Time Assessment of Liver Function with Near-Infrared Fluorescence Imaging. Eur. Surg. Res. 2017, 58, 235–245. [Google Scholar] [CrossRef] [PubMed]
  46. Dousse, D.; Vibert, E.; Nicolas, Q.; Terasawa, M.; Cano, L.; Allard, M.A.; Salloum, C.; Ciacio, O.; Pittau, G.; Sa Cunha, A.; et al. Indocyanine Green Fluorescence Imaging to Predict Graft Survival after Orthotopic Liver Transplantation: A Pilot Study. Liver Transpl. 2020, 26, 1263–1274. [Google Scholar] [CrossRef] [PubMed]
  47. Matsushima, H.; Sasaki, K.; Fujiki, M.; Uso, T.D.; Aucejo, F.; Kwon CH, D.; Eghtesad, B.; Miller, C.; Quintini, C.; Hashimoto, K. Too Much, Too Little, or Just Right? The Importance of Allograft Portal Flow in Deceased Donor Liver Transplantation. Transplantation 2020, 104, 770–778. [Google Scholar] [CrossRef] [PubMed]
  48. Zheng, B.W.; Zhang, H.J.; Gu, S.J.; Wu, T.; Wu, L.L.; Lian, Y.F.; Tong, G.; Yi, S.H.; Ren, J. Contrast-enhanced ultrasonography to evaluate risk factors for short-term and long-term outcomes after liver transplantation: A pilot prospective study. Eur. J. Radiol. 2021, 135, 109475. [Google Scholar] [CrossRef] [PubMed]
  49. Lee, D.D.; Croome, K.P.; Shalev, J.A.; Musto, K.R.; Sharma, M.; Keaveny, A.P.; Taner, C.B. Early allograft dysfunction after liver transplantation: An intermediate outcome measure for targeted improvements. Ann. Hepatol. 2016, 15, 53–60. [Google Scholar] [CrossRef]
  50. Pandya, K.; Sastry, V.; Panlilio, M.T.; Yip TC, F.; Salimi, S.; West, C.; Virtue, S.; Wells, M.; Crawford, M.; Pulitano, C.; et al. Differential Impact of Extended Criteria Donors After Brain Death or Circulatory Death in Adult Liver Transplantation. Liver Transpl. 2020, 26, 1603–1617. [Google Scholar] [CrossRef]
  51. Gong, J.L.; Yu, J.; Wang, T.L.; He, X.S.; Tang, Y.H.; Zhu, X.F. Application of extended criteria donor grafts in liver transplantation for acute-on-chronic liver failure: A retrospective cohort study. World J. Gastroenterol. 2023, 29, 5630–5640. [Google Scholar] [CrossRef] [PubMed]
  52. Guorgui, J.; Ito, T.; Younan, S.; Agopian, V.G.; Dinorcia, J., 3rd; Farmer, D.G.; Busuttil, R.W.; Kaldas, F.M. The Utility of Extended Criteria Donor Livers in High Acuity Liver Transplant Recipients. Am. Surg. 2021, 87, 1684–1689. [Google Scholar] [CrossRef]
  53. Park, P.J.; Yu, Y.D.; Yoon, Y.I.; Kim, S.R.; Kim, D.S. Single-Center Experience Using Marginal Liver Grafts in Korea. Transpl. Proc. 2018, 50, 1147–1152. [Google Scholar] [CrossRef]
  54. Wagener, G.; Raffel, B.; Young, A.T.; Minhaz, M.; Emond, J. Predicting early allograft failure and mortality after liver transplantation: The role of the postoperative model for end-stage liver disease score. Liver Transpl. 2013, 19, 534–542. [Google Scholar] [CrossRef] [PubMed]
  55. Benko, T.; Gallinat, A.; Minor, T.; Saner, F.H.; Sotiropoulos, G.C.; Paul, A.; Hoyer, D.P. The postoperative Model for End stage Liver Disease score as a predictor of short-term outcome after transplantation of extended criteria donor livers. Eur. J. Gastroenterol. Hepatol. 2017, 29, 716–722. [Google Scholar] [CrossRef] [PubMed]
  56. Angelico, M.; Nardi, A.; Romagnoli, R.; Marianelli, T.; Corradini, S.G.; Tandoi, F.; Gavrila, C.; Salizzoni, M.; Pinna, A.D.; Cillo, U.; et al. A Bayesian methodology to improve prediction of early graft loss after liver transplantation derived from the liver match study. Dig. Liver Dis. 2014, 46, 340–347. [Google Scholar] [CrossRef] [PubMed]
  57. Pareja, E.; Cortes, M.; Hervás, D.; Mir, J.; Valdivieso, A.; Castell, J.V.; Lahoz, A. A score model for the continuous grading of early allograft dysfunction severity. Liver Transplant. 2015, 21, 38–46. [Google Scholar] [CrossRef] [PubMed]
  58. Jochmans, I.; Fieuws, S.; Monbaliu, D.; Pirenne, J. “Model for Early Allograft Function” Outperforms “Early Allograft Dysfunction” as a Predictor of Transplant Survival. Transplantation 2017, 101, e258–e264. [Google Scholar] [CrossRef] [PubMed]
  59. Richards, J.A.; Sherif, A.E.; Butler, A.J.; Hunt, F.; Allison, M.; Oniscu, G.C.; Watson, C.J.E. Model for early allograft function is predictive of early graft loss in donation after circulatory death liver transplantation. Clin. Transplant. 2020, 34, e13982. [Google Scholar] [CrossRef]
  60. Agopian, V.G.; Harlander-Locke, M.P.; Markovic, D.; Dumronggittigule, W.; Xia, V.; Kaldas, F.M.; Zarrinpar, A.; Yersiz, H.; Farmer, D.G.; Hiatt, J.R.; et al. Evaluation of Early Allograft Function Using the Liver Graft Assessment Following Transplantation Risk Score Model. JAMA Surg. 2018, 153, 436–444. [Google Scholar] [CrossRef]
  61. Agopian, V.G.; Markovic, D.; Klintmalm, G.B.; Saracino, G.; Chapman, W.C.; Vachharajani, N.; Florman, S.S.; Tabrizian, P.; Haydel, B.; Nasralla, D.; et al. Multicenter validation of the liver graft assessment following transplantation (L-GrAFT) score for assessment of early allograft dysfunction. J. Hepatol. 2021, 74, 881–892. [Google Scholar] [CrossRef] [PubMed]
  62. Avolio, A.W.; Franco, A.; Schlegel, A.; Lai, Q.; Meli, S.; Burra, P.; Patrono, D.; Ravaioli, M.; Bassi, D.; Ferla, F.; et al. Development and Validation of a Comprehensive Model to Estimate Early Allograft Failure Among Patients Requiring Early Liver Retransplant. JAMA Surg. 2020, 155, e204095. [Google Scholar] [CrossRef]
  63. Rhu, J.; Kim, J.M.; Kim, K.; Yoo, H.; Choi, G.S.; Joh, J.W. Prediction model for early graft failure after liver transplantation using aspartate aminotransferase, total bilirubin and coagulation factor. Sci. Rep. 2021, 11, 12909. [Google Scholar] [CrossRef] [PubMed]
  64. Diaz-Nieto, R.; Lykoudis, P.; Robertson, F.; Sharma, D.; Moore, K.; Malago, M.; Davidson, B.R. A simple scoring model for predicting early graft failure and postoperative mortality after liver transplantation. Ann. Hepatol. 2019, 18, 902–912. [Google Scholar] [CrossRef] [PubMed]
  65. Manzia, T.M.; Lai, Q.; Hartog, H.; Aijtink, V.; Pellicciaro, M.; Angelico, R.; Gazia, C.; Polak, W.G.; Rossi, M.; Tisone, G. Graft weight integration in the early allograft dysfunction formula improves the prediction of early graft loss after liver transplantation. Updates Surg. 2022, 74, 1307–1316. [Google Scholar] [CrossRef]
  66. Lai, Q.; Melandro, F.; Nowak, G.; Nicolini, D.; Iesari, S.; Fasolo, E.; Mennini, G.; Romano, A.; Mocchegiani, F.; Ackenine, K.; et al. The role of the comprehensive complication index for the prediction of survival after liver transplantation. Updates Surg. 2021, 73, 209–221. [Google Scholar] [CrossRef] [PubMed]
  67. Moosburner, S.; Wiering, L.; Roschke, N.N.; Winter, A.; Demir, M.; Gaßner JM, G.V.; Zimmer, M.; Ritschl, P.; Globke, B.; Lurje, G.; et al. Validation of risk scores for allograft failure after liver transplantation in Germany: A retrospective cohort analysis. Hepatol. Commun. 2023, 7, e0012. [Google Scholar] [CrossRef] [PubMed]
  68. Núñez, K.; Hamed, M.; Fort, D.; Bruce, D.; Thevenot, P.; Cohen, A. Links between donor macrosteatosis, interleukin-33 and complement after liver transplantation. World J. Transplant. 2020, 10, 117–128. [Google Scholar] [CrossRef] [PubMed]
  69. Núñez, K.G.; Frank, A.; Gonzalez-Rosario, J.; Galliano, G.; Bridle, K.; Crawford, D.; Seal, J.; Abbruscato, F.; Vashistha, H.; Thevenot, P.T.; et al. Interleukin-33/ Cyclin D1 imbalance in severe liver steatosis predicts susceptibility to ischemia reperfusion injury. PLoS ONE 2019, 14, e0216242. [Google Scholar] [CrossRef]
  70. Barbier, L.; Robin, A.; Sindayigaya, R.; Ducousso, H.; Dujardin, F.; Thierry, A.; Hauet, T.; Girard, J.P.; Pellerin, L.; Gombert, J.M.; et al. Endogenous Interleukin-33 Acts as an Alarmin in Liver Ischemia-Reperfusion and Is Associated With Injury After Human Liver Transplantation. Front. Immunol. 2021, 12, 744927. [Google Scholar] [CrossRef]
  71. Yazdani, H.O.; Chen, H.W.; Tohme, S.; Tai, S.; van der Windt, D.J.; Loughran, P.; Rosborough, B.R.; Sud, V.; Beer-Stolz, D.; Turnquist, H.R.; et al. IL-33 exacerbates liver sterile inflammation by amplifying neutrophil extracellular trap formation. J Hepatol. 2017, 68, 130–139. [Google Scholar] [CrossRef] [PubMed]
  72. Sakai, N.; Van Sweringen, H.L.; Quillin, R.C.; Schuster, R.; Blanchard, J.; Burns, J.M.; Tevar, A.D.; Edwards, M.J.; Lentsch, A.B. Interleukin-33 Is Hepatoprotective During Liver Ischemia/Reperfusion in Mice. Hepatology 2012, 56, 1468–1478. [Google Scholar] [CrossRef] [PubMed]
  73. Faitot, F.; Besch, C.; Lebas, B.; Addeo, P.; Ellero, B.; Woehl-Jaegle, M.L.; Namer, I.J.; Bachellier, P.; Freys, G. Interleukin 6 at reperfusion: A potent predictor of hepatic and extrahepatic early complications after liver transplantation. Clin. Transplant. 2018, 32, e13357. [Google Scholar] [CrossRef] [PubMed]
  74. Maring, J.K.; Klompmaker, I.J.; Zwaveling, J.H.; van Der Meer, J.; Limburg, P.C.; Slooff, M.J. Endotoxins and cytokines during liver transplantation: Changes in plasma levels and effects on clinical outcome. Liver Transplant. 2000, 6, 480–488. [Google Scholar] [CrossRef] [PubMed]
  75. Brenner, T.; Rosenhagen, C.; Brandt, H.; Schmitt, F.C.; Jung, G.E.; Schemmer, P.; Schmidt, J.; Mieth, M.; Bruckner, T.; Lichtenstern, C.; et al. Cell death biomarkers as early predictors for hepatic dysfunction in patients after orthotopic liver transplantation. Transplantation 2012, 94, 185–191. [Google Scholar] [CrossRef]
  76. Chae, M.S.; Kim, J.W.; Chung, H.S.; Park, C.S.; Lee, J.; Choi, J.H.; Hong, S.H. The impact of serum cytokines in the development of early allograft dysfunction in living donor liver transplantation. Medicine 2018, 97, e0400. [Google Scholar] [CrossRef]
  77. Karakhanova, S.; Oweira, H.; Steinmeyer, B.; Sachsenmaier, M.; Jung, G.; Elhadedy, H.; Schmidt, J.; Hartwig, W.; Bazhin, A.V.; Werner, J. Interferon-γ, interleukin-10 and interferon-inducible protein 10 (CXCL10) as serum biomarkers for the early allograft dysfunction after liver transplantation. Transpl. Immunol. 2016, 34, 14–24. [Google Scholar] [CrossRef]
  78. Ng, T.H.; Britton, G.J.; Hill, E.V.; Verhagen, J.; Burton, B.R.; Wraith, D.C. Regulation of adaptive immunity; the role of interleukin-10. Front. Immunol. 2013, 4, 129. [Google Scholar] [CrossRef]
  79. Faria, L.C.; Andrade AM, F.; Trant CG, M.C.; Lima, A.S.; Machado PA, B.; Porto, R.D.; Andrade MV, M. Circulating levels of High-mobility group box 1 protein and nucleosomes are associated with outcomes after liver transplant. Clin. Transplant. 2020, 34, e13869. [Google Scholar] [CrossRef]
  80. Sosa, R.A.; Terry, A.Q.; Kaldas, F.M.; Jin, Y.P.; Rossetti, M.; Ito, T.; Li, F.; Ahn, R.S.; Naini, B.V.; Groysberg, V.M.; et al. Disulfide High-Mobility Group Box 1 Drives Ischemia-Reperfusion Injury in Human Liver Transplantation. Hepatology 2021, 73, 1158–1175. [Google Scholar] [CrossRef]
  81. Kwon, H.M.; Moon, Y.J.; Jung, K.W.; Park, Y.S.; Jun, I.G.; Kim, S.O.; Song, J.G.; Hwang, G.S. Neutrophil-to-lymphocyte ratio is a predictor of early graft dysfunction following living donor liver transplantation. Liver Int. 2019, 39, 1545–1556. [Google Scholar] [CrossRef] [PubMed]
  82. Thomsen, K.L.; Robertson, F.P.; Holland-Fischer, P.; Davidson, B.R.; Mookerjee, R.P.; Møller, H.J.; Jalan, R.; Grønbæk, H. The Macrophage Activation Marker Soluble CD163 is Associated With Early Allograft Dysfunction After Liver Transplantation. J. Clin. Exp. Hepatol. 2019, 9, 302–311. [Google Scholar] [CrossRef] [PubMed]
  83. Park, J.; Lim, S.J.; Choi, H.J.; Hong, S.H.; Park, C.S.; Choi, J.H.; Chae, M.S. Predictive utility of the C-reactive protein to albumin ratio in early allograft dysfunction in living donor liver transplantation: A retrospective observational cohort study. PLoS ONE 2019, 14, e0226369. [Google Scholar] [CrossRef]
  84. Ceglarek, U.; Kresse, K.; Becker, S.; Fiedler, G.M.; Thiery, J.; Quante, M.; Wieland, R.; Bartels, M.; Aust, G. Circulating sterols as predictors of early allograft dysfunction and clinical outcome in patients undergoing liver transplantation. Metabolomics 2016, 12, 182. [Google Scholar] [CrossRef] [PubMed]
  85. Tsai, H.I.; Lo, C.J.; Zheng, C.W.; Lee, C.W.; Lee, W.C.; Lin, J.R.; Shiao, M.S.; Cheng, M.L.; Yu, H.P. A Lipidomics Study Reveals Lipid Signatures Associated with Early Allograft Dysfunction in Living Donor Liver Transplantation. J. Clin. Med. 2018, 8, 30. [Google Scholar] [CrossRef]
  86. Tsai, H.I.; Lo, C.J.; Lee, C.W.; Lin, J.R.; Lee, W.C.; Ho, H.Y.; Tsai, C.Y.; Cheng, M.L.; Yu, H.P. A panel of biomarkers in the prediction for early allograft dysfunction and mortality after living donor liver transplantation. Am. J. Transl. Res. 2021, 13, 372–382. [Google Scholar]
  87. Yang, J.; Wang, H.Q.; Yang, J.Y.; Wen, T.F.; Li, B.; Wang, W.T.; Yan, L.N. Role of the postoperative cholesterol in early allograft dysfunction and survival after living donor liver transplantation. Hepatobiliary Pancreat. Dis. Int. 2017, 16, 610–616. [Google Scholar] [CrossRef] [PubMed]
  88. Pollara, J.; Edwards, R.W.; Lin, L.; Bendersky, V.A.; Brennan, T.V. Circulating mitochondria in deceased organ donors are associated with immune activation and early allograft dysfunction. JCI Insight. 2018, 3, e121622. [Google Scholar] [CrossRef] [PubMed]
  89. Yoshino, O.; Wong, B.K.L.; Cox, D.R.A.; Lee, E.; Hepworth, G.; Christophi, C.; Jones, R.; Dobrovic, A.; Muralidharan, V.; Perini, M.V. Elevated levels of circulating mitochondrial DNA predict early allograft dysfunction in patients following liver transplantation. J. Gastroenterol. Hepatol. 2021, 36, 3500–3507. [Google Scholar] [CrossRef]
  90. Hong, S.H.; Kwak, J.A.; Chon, J.Y.; Park, C.S. Prediction of early allograft dysfunction using serum phosphorus level in living donor liver transplantation. Transpl. Int. 2013, 26, 402–410. [Google Scholar] [CrossRef]
  91. Chae, M.S.; Koo, J.M.; Park, C.S. Predictive Role of Intraoperative Serum Brain Natriuretic Peptide for Early Allograft Dysfunction in Living Donor Liver Transplantation. Ann. Transplant. 2016, 21, 538–549. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, J.; Han, Y.; Ke, S.; Gao, R.; Shi, X.; Zhao, S.; You, P.; Jia, H.; Ding, Q.; Zheng, Y.; et al. Postoperative serum myoglobin as a predictor of early allograft dysfunction after liver transplantation. Front. Surg. 2022, 9, 1026586. [Google Scholar] [CrossRef] [PubMed]
  93. Gorgen, A.; Prediger, C.; Prediger, J.E.; Chedid, M.F.; Backes, A.N.; de Araujo, A.; Grezzana-Filho TJ, M.; Leipnitz, I.; Chedid, A.D.; Alvares-da-Silva, M.R.; et al. Serum Factor V Is a Continuous Biomarker of Graft Dysfunction and a Predictor of Graft Loss After Liver Transplantation. Transplantation 2019, 103, 944–951. [Google Scholar] [CrossRef]
  94. Nedel, W.L.; Rodrigues Filho, E.M.; Pasqualotto, A.C. Thrombin-activatable Fibrinolysis Inhibitor (TAFI) as a Novel Prognostic Factor after Orthotropic Liver Transplantation: A Pilot Study. Transplant. Proc. 2015, 47, 1912–1914. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Illustrating the lumen of a liver sinusoid. Ischemia–reperfusion injury (IRI) damages hepatocytes and liver sinusoidal endothelial cells (LSECs). Ischemia and the lack of an oxygen supply disrupt mitochondria and adenosine triphosphate (ATP) production, resulting in impaired gluconeogenesis and lactate accumulation. Inflammatory cytokines released from apoptotic cells stimulate Kupffer cell (KC) inflammatory activation and trigger neutrophil (N) and macrophage (M) migration to the liver. Cytokines produced by Kupffer cells (KCs) increase the chemotaxis of neutrophils (N) and stimulate their adhesion to endothelial cells. Damage-associated molecular patterns (DAMPs) and DNA fragments derived from IRI-induced cell death trigger neutrophil (N) degranulation and reactive oxygen species (ROS) secretion, further damaging the liver tissue. At the same time, inflammatory cytokines increase P-selectin expression on the membrane of liver sinusoidal endothelial cells (LSECs), causing platelet aggregation. Activated platelets not only induce thrombosis in the transplanted graft but also release cytokines that stimulate hepatic stellate cells (HSCs) and initiate fibrotic changes. Abbreviations: ischemia–reperfusion injury (IRI), liver sinusoidal endothelial cell (LSEC), adenosine triphosphate (ATP), Kupffer cell (KC), neutrophil (N), macrophage (M), damage-associated molecular patterns (DAMPs), reactive oxygen species (ROS), hepatic stellate cell (HSC), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), tumor necrosis factor alpha (TNFα), transforming growth factor beta (TGFβ), toll-like receptor (TLR), complement anaphylatoxins (C3a, C5a), intracellular adhesion molecule-1 (ICAM-1), integrin receptor (CD11b/CD18a), thromboxane 2A (TXA2), and plasminogen activator inhibitor-1 (PAI-1). Created with BioRender.com (accessed on 26 May 2024).
Figure 1. Illustrating the lumen of a liver sinusoid. Ischemia–reperfusion injury (IRI) damages hepatocytes and liver sinusoidal endothelial cells (LSECs). Ischemia and the lack of an oxygen supply disrupt mitochondria and adenosine triphosphate (ATP) production, resulting in impaired gluconeogenesis and lactate accumulation. Inflammatory cytokines released from apoptotic cells stimulate Kupffer cell (KC) inflammatory activation and trigger neutrophil (N) and macrophage (M) migration to the liver. Cytokines produced by Kupffer cells (KCs) increase the chemotaxis of neutrophils (N) and stimulate their adhesion to endothelial cells. Damage-associated molecular patterns (DAMPs) and DNA fragments derived from IRI-induced cell death trigger neutrophil (N) degranulation and reactive oxygen species (ROS) secretion, further damaging the liver tissue. At the same time, inflammatory cytokines increase P-selectin expression on the membrane of liver sinusoidal endothelial cells (LSECs), causing platelet aggregation. Activated platelets not only induce thrombosis in the transplanted graft but also release cytokines that stimulate hepatic stellate cells (HSCs) and initiate fibrotic changes. Abbreviations: ischemia–reperfusion injury (IRI), liver sinusoidal endothelial cell (LSEC), adenosine triphosphate (ATP), Kupffer cell (KC), neutrophil (N), macrophage (M), damage-associated molecular patterns (DAMPs), reactive oxygen species (ROS), hepatic stellate cell (HSC), interleukin 1 beta (IL-1β), interleukin 6 (IL-6), tumor necrosis factor alpha (TNFα), transforming growth factor beta (TGFβ), toll-like receptor (TLR), complement anaphylatoxins (C3a, C5a), intracellular adhesion molecule-1 (ICAM-1), integrin receptor (CD11b/CD18a), thromboxane 2A (TXA2), and plasminogen activator inhibitor-1 (PAI-1). Created with BioRender.com (accessed on 26 May 2024).
Jcm 13 03762 g001
Figure 2. Illustration of the lactic acid cycle (Cori cycle). Lactates, produced mainly in the muscles, cannot be efficiently metabolized in the liver due to ischemia–reperfusion injury (IRI) and the lack of ATP in hepatocyte mitochondria. Hyperlactatemia is exacerbated by the failure of MCT1 and MCT2 membrane transporters, leading to dysfunctional lactate uptake by the hepatocyte. Abbreviations: adenosine triphosphate (ATP), lactate dehydrogenase (LDH), monocarboxylate transporter (MCT1, MCT2). Created with BioRender.com (accessed on 26 May 2024).
Figure 2. Illustration of the lactic acid cycle (Cori cycle). Lactates, produced mainly in the muscles, cannot be efficiently metabolized in the liver due to ischemia–reperfusion injury (IRI) and the lack of ATP in hepatocyte mitochondria. Hyperlactatemia is exacerbated by the failure of MCT1 and MCT2 membrane transporters, leading to dysfunctional lactate uptake by the hepatocyte. Abbreviations: adenosine triphosphate (ATP), lactate dehydrogenase (LDH), monocarboxylate transporter (MCT1, MCT2). Created with BioRender.com (accessed on 26 May 2024).
Jcm 13 03762 g002
Table 1. Comparison of the King’s College Hospital PNF score (King-PNF score) with other available models for predicting PNF.
Table 1. Comparison of the King’s College Hospital PNF score (King-PNF score) with other available models for predicting PNF.
YearStudy [Ref.]SizeOutcomeIncidenceModelc-Statistic
2017Al-Freah et al. [21] 1125PNF3.70%King-PNF score0.91
UK EGD Criteria0.67
US PNF Criteria0.78
2023Nie et al. [22]720PNF3.90%MEAF0.87
King-PNF score0.89
BAR-Lac0.83
EAD39%MEAF0.84
King-PNF score0.81
BAR-Lac0.63
early graft failure9.30%MEAF0.80
King-PNF score0.87
BAR-Lac0.76
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gierej, P.; Radziszewski, M.; Figiel, W.; Grąt, M. Advancements in Predictive Tools for Primary Graft Dysfunction in Liver Transplantation: A Comprehensive Review. J. Clin. Med. 2024, 13, 3762. https://doi.org/10.3390/jcm13133762

AMA Style

Gierej P, Radziszewski M, Figiel W, Grąt M. Advancements in Predictive Tools for Primary Graft Dysfunction in Liver Transplantation: A Comprehensive Review. Journal of Clinical Medicine. 2024; 13(13):3762. https://doi.org/10.3390/jcm13133762

Chicago/Turabian Style

Gierej, Piotr, Marcin Radziszewski, Wojciech Figiel, and Michał Grąt. 2024. "Advancements in Predictive Tools for Primary Graft Dysfunction in Liver Transplantation: A Comprehensive Review" Journal of Clinical Medicine 13, no. 13: 3762. https://doi.org/10.3390/jcm13133762

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

Article Metrics

Back to TopTop