Open access peer-reviewed chapter

Machine Perfusion Strategies in Liver and Renal Transplantation

Written By

Stella Vasileiadou, Nikolaos Antoniadis, Athanasios Kofinas, Konstantina-Eleni Karakasi, Georgios Katsanos and Georgios Tsoulfas

Submitted: 30 September 2022 Reviewed: 24 March 2023 Published: 10 April 2023

DOI: 10.5772/intechopen.111436

From the Edited Volume

Current Challenges and Advances in Organ Donation and Transplantation

Edited by Georgios Tsoulfas

Chapter metrics overview

109 Chapter Downloads

View Full Metrics

Abstract

Transplantation is the only treatment for patients with end-stage renal and hepatic disease but unfortunately, it is limited worldwide due to the organ shortage. The need to expand the donor pool with the use of donors after cardiac death (DCD) and extended criteria donors (ECD) has led to major improvements in organ preservation. From cold static storage and preservation solutions to different types of machine perfusion, the possibility of successfully transplanting a marginal organ becomes reality. In this chapter, we examine the machine perfusion methods and the advantages of new technology in minimizing ischemic injury and improving the transplant outcome. Τhe establishment of protocols with the use of biomarkers in order to assess the transplant suitability of the graft will eventually provide the ideal opportunity to intervene and improve the quality of the organ.

Keywords

  • machine perfusion
  • liver transplantation
  • kidney transplantation
  • organ preservation
  • hypothermic machine perfusion
  • normothermic machine perfusion

1. Introduction

In this chapter, after a brief introductory reference to the definition and history of machine perfusion in transplantation, their contemporary necessity is discussed. For this purpose, two important topics such as ischemia–reperfusion injury and marginal grafts are analyzed. Subsequently, the machine perfusion strategies in liver and kidney transplantation are mentioned separately.

The review of the literature was conducted using the combination of various keywords (liver transplantation, kidney/renal transplantation, machine perfusion, hypothermic/normothermic/subnormothermic machine perfusion, ischemia–reperfusion injury, marginal grafts, extended criteria donors, donors after cardiac death, cold static storage, and organ preservation) in PubMed research database. The inclusion criteria of the articles were the English language and their relevance to the topic, which was judged by the authors after studying the abstracts.

Transplantation is the only treatment for patients with end-stage renal and hepatic disease. Despite the recent U.S. milestone of 1 million transplants until 2022, the number of patients on waiting lists remains high [1]. The great need for increment of the donor pool leads to the utilization of allografts from extended criteria donors (ECD) and donors after circulatory death (DCD). The vulnerability of these marginal grafts in ischemia–reperfusion injury (IRI) [2] revealed all the weak points of standard preservation methods and resulted in poor transplant outcomes. Moreover, the ECD and DCD kidney grafts have increased immunogenicity and as a result higher rates of rejection [3].

The combination of nonideal donors with limitations in graft evaluation results in discarded organs that further exacerbate the transplantation gap. In an attempt to find solutions, the interest of scientific community turned to preservation strategies, particularly the application of machine perfusion methods to these organs, in order to assess and make them suitable for transplantation [4, 5].

The concept of machine perfusion (MP) in the field of transplantation is not new. In 1885, the initial idea of extracorporeal circulation with the construction of a closed circulatory system [6] and 52 years later the transition from blood to chemical perfusion solutions, in order to keep organs “alive” [7], laid the foundations for the development of organ preservation. Machine perfusion in a human kidney was performed for the first time in 1967 by Belzer. The first liver transplantation in humans was performed by Starzl, with the application of a hypothermic low-flow perfusion method that used diluted blood [8, 9]. Thus, although the original concept involved room temperature and dynamic perfusion, in 1969 Collins GM changed completely the perspective, with the 12-hour preservation of canine kidneys in an iced solution, which later evolved into Collins solution with a longer preservation time [10]. Nowadays, preserving the organ in an ice box at 0–4°C, after flushing it with a cold preservation solution (usually the University of Wisconsin or Histidine-Tryptophan-Ketoglutarate solution), is the most common method worldwide. Static cold storage (SCS) has been established as the gold standard due to its simplicity, efficacy, and low cost [11]. The idea of machine perfusion was abandoned but, in the beginning of this century, gained interest again in clinical practice.

Machine perfusion is an ex-vivo platform where organs are connected to a pump and perfused with the solution in a controlled flow, constantly, until implantation. The main purpose is to provide essential substances and remove waste products in order to maintain cellular metabolism in a predetermined way. There is variation in machine perfusion methods in terms of temperature, oxygen and nutrient supply, preservation solutions, delivery method, duration, and time-point of perfusion. Depending on the temperature, there are three types of machine perfusion: normothermic (35.5–37.5°C), hypothermic (1–8°C), and subnormothermic (20–35.5°C) temperature with the latter sought in fewer studies [12]. Also, the variation of temperatures differs slightly in the literature.

Hypothermia reduces cellular metabolism while at the same time, the preservation solution enhances ATP production [13]. Also, the addition of oxygen restores mitochondrial oxidative activity by electron transport [14]. Hypothermic machine perfusion (HMP) has the significant advantage of safety, as in case of machine failure, the graft can easily be preserved in SCS.

The aim of reducing cold ischemia time (CIT), which is an independent risk factor for delayed graft function (DGF), has led to the development of normothermic machine perfusion (NMP). During this method, the reperfusion phase after ischemia time takes place in an artificial environment as close as possible to physiological conditions (temperature and oxygen), in order to avoid the inflammatory process and the destructive injury, it causes to the graft. Normothermia has the important advantage of assessing the graft in real-time, as bile and urine are produced by liver and kidney grafts, respectively [15]. On the other hand, it is a more complicated and costly procedure as it requires a sufficient amount of oxygen, oxygen carriers, and necessary substances for the graft function [16]. In an effort to limit the use of human blood products with red blood cells or plasma, the development of extracellular oxygen carriers began [17].

Subnormothermic machine perfusion (SNMP) is an intermediate state between hypothermic and normothermic conditions, maintaining low cellular energy demands and mitochondrial activity, while the organ is still “alive.” This method is simpler than NMP as red blood cells are not necessary [18]. Although SNMP started as a promising technique, disappointing results in a porcine kidney model prevented its further clinical application. The SNMP group had increased rates of renal and tubular injury than the NMP group [19]. Currently, it is still at an experimental stage in both liver and kidney transplantation with important clinical prospects.

Controlled oxygenated rewarming (COR) is an alternative perfusion method that focuses on transitioning the graft from hypothermia to normothermia by gradually increasing the temperature (from 8–20°C). Mitochondrial function can be affected by rapid temperature change when the graft is reperfused after cold ischemic preservation. In particular, the opening of the mitochondrial permeability transition pore contributes in a destructive way to the occurrence of IRI. COR allows smooth recovery of mitochondrial function and studies have shown its feasibility in clinical application in liver and kidney transplantation [20, 21].

In the last decade, many studies have been published on different models of machine perfusion and large clinical trials in humans have been performed, changing the concept of preservation in transplantation. From procurement to transplantation, the reasons for this development, in addition to the increasing use of ECD or DCD grafts, are the comprehension of the complicated biological pathways and phenomena such as IRI.

1.1 Ischemia: Reperfusion injury

During organ procurement, the blood supply to the organs is interrupted, and after implantation or ex-situ normothermic machine perfusion of the grafts, it is restored. This process, defined as ischemia–reperfusion injury, leads to cell death in an oxidative environment and production of reactive oxygen species (ROS) [2].

IRI consists of three phases and each of them causes a different effect on the organ. First, it is the ischemic phase, when the blood and oxygen supply of the organ is inhibited. Dramatic depletion of ATP unregulates sodium-potassium ATPases in cell membranes, resulting in increased calcium concentration and acidic pH [2223]. It is of notice that the type of donor (donation after brain death or after circulatory death) plays an important role in ischemia injury to the graft [24]. Second, it is the reperfusion phase, when ischemic tissues are reoxygenated and as a result release ROS, that is mainly formed in complex 1 - mitochondrial respiratory chain [25]. Furthermore, in liver grafts, disorders in the mitochondrial permeability transition pore (MPTP) allow the release of danger-associated molecular patterns (DAMPs) from the nucleus [26], which interact with Toll-like receptors (TLRs) and activate Kupffer cells [27]. Finally, a prolonged period of inflammation is established where important liver microstructures are deranged. For example, damage to the endothelial glycocalyx or the combination of increased vasoconstrictive agents and decreased nitric oxide levels deteriorates the microcirculation and raises the risk of early allograft dysfunction (EAD) [28, 29]. Third is the late injury phase, which follows the release of ROS and continues the cascade of cytotoxic events with the activation of resident and recruited immune cells [30].

IRI is globally independent risk factor in transplantation and is related to acute rejection, postreperfusion syndrome, DGF, and primary non-function (PNF) [31]. The standard static cold storage was an inferior preservation method for kidneys [32] and failed to protect marginal livers from the effects of ischemia–reperfusion injury [33], so the need for a different strategy was imperative. Continuous machine perfusion circulation of metabolic substances and removal of inflammatory products, such as toxins and cytokines [34], reduce the degree of graft injury, for example, to the liver’s deep peribiliary glands and stroma [35].

1.2 Marginal grafts

As already mentioned, the use of suboptimal grafts is constantly increasing due to the transplant gap between the donor pool and the recipient waiting list. Marginal liver grafts are organs with an increased chance of failure or PNF after transplantation. Also, the definition includes cases where diseases can be transmitted from the donor to the recipient [36]. Usually, high risk liver grafts come from donors with factors such as: liver macrosteatosis, cardiac death, advanced age, long stay in the intensive care unit, infections and malignancies.

Macrosteatosis is not a prohibitive factor for transplantation if the percentage is <30% but the fatty liver is considered to have worse postoperative outcomes than grafts without steatosis [37]. DCD livers due to prolonged warm ischemia time are susceptible to ischemic-type biliary lesions. Microcirculation disorders, during IRI, cause damage to the arterial plexus of the biliary tree, so there is high risk of biliary strictures after the transplantation [38]. Advanced age has negative effects on liver reserves, so injury after reperfusion is more serious and increases the risk of posttransplant complications [39].

Machine perfusion techniques appear to benefit these borderline livers most, with characteristics such as macrosteatosis >30%, DCD donors, donors >60 years old, cold ischemia time > 8-10 h, warm ischemia time > 30 min, insufficient in situ perfusion, prolonged organ procurement, and generally discarded grafts due to eliminated transaminases or any nonvascular problem [40, 41].

Different scoring systems have been developed to predict graft survival: Survival outcomes following liver transplantation (SOFT), donor risk index (DRI), Eurotransplant-DRI, etc. Clinical parameters (cardiac arrest >15 min, age > 55 years, BMI >30), laboratory results (AST, ALT, HBV, and HCV), ICU conditions (days, drugs, and infections) and of course histology after biopsy are included and considered in the transplant decision. Finally, nonheart beating and DCD donors are ECDs by definition [42].

The definition of borderline kidneys, which actually includes donor risk factors for kidney transplants, is derived from living donors, as in the early stages were mainly associated with risks to the recipient [43]. The Kidney donor risk index (KDRI) is a practical tool for predicting the posttransplant outcome. Factors such as weight, height, race, history of diabetes, hepatitis C status, and DCD are included [44]. The OPTN-approved criteria for ECD kidneys consider the following donor parameters: age, cerebrovascular accident as the cause of death, history of hypertension, and creatinine level [45].

Also, acute kidney injury or extracorporeal membrane oxygenation (ECMO) prior to organ retrieval significantly affects the graft quality [46, 47].

Advertisement

2. Machine perfusion of the liver

2.1 Hypothermic machine perfusion

During hypothermic machine perfusion, the liver graft is perfused with a preservation solution, mainly University of Wisconsin (UW) or Vasosol (modified UW), usually at 4–11°C. In this way, the graft takes essential substances and eliminates waste products [40, 48]. The pO2 level is 20 kPa, as no additional oxygen is provided. HMP clinical series was first published in 2010, as a safe and effective perfusion system by James Guarrera [49].

In hypothermic conditions, deceleration of cellular metabolism and cryoprotection, prepare the liver graft for the time of reperfusion. HMP provides protection to the graft in a complicated and not completely clarified way, as far as ischemia–reperfusion injury is concerned. Studies have shown that it has positive effects on mitochondrial complexes and increases cellular energy, leading to metabolism of molecules such as succinate [50, 51].

Also, livers perfused under hypothermic conditions had less of the ultrastructural effects seen in SCS. The number of CD68+ macrophages and, in general, inflammatory injury agents were significantly decreased, as shown by measurements at the end of preservation and after graft reperfusion [51, 52].

During hypothermic oxygenated machine perfusion (HOPE), the pO2 level is 60–100 kPa, and in this technique additional perfusate oxygen is provided to the graft. Oxygen is diluted in the perfusion fluid so there is no need for oxygen carriers [53]. Oxygenation in hypothermic conditions restores mitochondrial function and promotes metabolic activity. As a result, the reduction of ROS and succinate does not lead to further damage in mitochondrial function [54, 55].

HOPE was initially used in a clinical setting in 2014, where Maastricht Type III-DCD liver grafts were treated and subsequently assessed [56]. A matched case analysis of DCD livers compared HOPE treatment with standard SCS. 25 HOPE livers had fewer biliary complications and better 1-year graft survival than 50 SCS livers, but with longer cold ischemic time in the last group [57]. HOPE-DCD livers showed a reduced risk of acute rejection and improved 5-year graft survival in a recent study with the same comparators (94% in HOPE DCD vs. 78% in SCS DCD). They also have better laboratory results (INR and lactates on the first day) and fewer postoperative complications [58]. Moreover, HOPE treatment proved beneficial in macrosteatotic livers and has a positive impact on graft function and survival compared to the unperfused group [59]. Similarly, grafts from donors with advanced age seem to have good results with HOPE treatment. In two transplantations with octogenarian grafts, this perfusion technique promoted the recovery process after IRI and reduced hospitalization [60].

The hypothermic machine perfusion uses either the portal vein alone or the portal vein and hepatic artery together. The dual perfusion with oxygen is called dual flow hypothermic oxygenated perfusion (D-HOPE) and attempted to optimize the oxygenation of the biliary system. D-HOPE DCD livers had low peak ALT and bilirubin levels compared with SCS, but no difference in patient and 1-year graft survival. Perfusion pressure is also a technical detail of great importance during machine perfusion. High pressure leads to additional damage at endothelial cells, so the desired levels are 20–30 mmHg in the hepatic artery and 3–5 mmHg in the portal vein during HMP [61].

A meta-analysis of 12 studies comparing HMP and SCS in liver grafts showed the superiority of HMP in reducing posttransplant AST and ALT serum levels, early allograft dysfunction, total biliary complications, and ischemic cholangiopathy. No statistical significance was observed in outcomes for primary nonfunction, hepatic artery thrombosis, postreperfusion syndrome, patient survival, and liver graft survival (1 month, 6 months, and 1 year) [62]. An additional advantage of HMP is that in case of pump failure, the organ can be stored in cold static conditions, so it is a safe perfusion method.

The viability assessment during HMP is challenging because of metabolic deceleration and absence of bile production. However, there is a growing interest in the literature for potential markers during HMP and HOPE. Peak AST after transplantation was found to be associated with AST and LDH levels after 2 h of perfusion [49]. Also, cellular ATP concentration during D-HOPE appears to be related to liver posttransplant function at a biochemical level [61]. A recent study by Muller et al. discovered a new mitochondrial biomarker that can be counted by spectroscope after 30–45 min of HOPE. Flavin mitochondrial mononucleotide (FMN) is easily detected in HOPE perfusate and can predict graft function and possible complications before implantation [63].

2.2 Normothermic machine perfusion

Normothermic machine perfusion uses temperatures between 35 and 38°C and components from blood products such as erythrocytes. Oxygenators are necessary and dual perfusion (portal and arterial) is performed. The aim is to mimic the physiologic environment so that the metabolism and function of the liver graft being maintained. The first clinical introduction was in 2016 when a phase-1 clinical trial was published [64]. Minimizing clod ischemia time enables long preservation of the liver graft without injury. In fact, a recent protocol described a metabolic, active, and functional human liver after 7 days of NMP [65].

The combination of HOPE and NMP versus NMP in nontransplanted livers was studied by the Birmingham group. The HOPE/NMP livers showed lower indicators of inflammation and better recovery of their metabolic rate [66]. According to the first prospective multicenter randomized controlled trial, the use of NMP (121 livers) instead of SCS (101 livers) increases the utilization of grafts by 50%. NMP despite the longer preservation time (11.9 vs. 7.7 h) reduced the grade of graft injury as seen by the low levels of AST and EAD, in both DCD and DBD livers. However, there was no superiority of NMP in PNF, biliary complications, ischemic cholangiopathy, graft, and patient survival [67].

The great advantage of NMP is the ability to evaluate the graft during preservation time. Observation of liver parameters such as bile production with laboratory test results, such as transaminases, lactate, glucose, and pH, have created scoring systems to aid in the transplant decision. Perfusate lactate, bile production, vascular flows, and liver appearance of five rejected livers were evaluated during NMP (after SCS), and finally transplanted with good outcomes [68]. In a study in 23 human livers, biochemical parameters of bile were examined after 6 hours of NMP and compared with the grade of bile duct injury. Biliary bicarbonate, pH, glucose, and bile/perfusate glucose ratio were identified as potential biomarkers of posttransplant cholangiopathy [69].

2.3 Subnormothermic machine perfusion

Subnormothermic machine perfusion is a method that uses temperatures between HMP and NMP, usually 21–25°C, and an oxygenated preservation solution such as HOPE. This subphysiologic temperature, on one hand, reduces the metabolic rate but, on the other hand, the graft is warm enough to allow viability assessment [70].

Three hours of SNMP in seven discarded human livers showed satisfactory bile production and liver function [71] but the study did not include posttransplant outcomes in order to determine the favorable effect of SNMP.

2.4 Controlled oxygenated rewarming

In an experimental animal study, a gradual raise in temperature (up to 20°C) with simultaneous oxygenated perfusion was performed for 90 min after a period of end-ischemia HMP. Posttransplant peak transaminase level was lower, and 6 months of patient survival was higher in the COR group compared with the control group [72]. Combined protocols, such as DHOPE-COR-NMP used in discarded livers, have significantly increased the number of transplantable grafts [73].

2.5 Therapeutics in machine perfusion methods

During machine perfusion, in addition to the standard enrichment of the preservation solution (glucose, heparine, insulin, antibiotics, maybe oxygen, and nutrients), agents targeting graft recondition can be added. Machine Perfusion therapeutic agents aim to improve the marginal graft to follow a successful transplantation. The unique advantage of this ex vivo procedure is that the agents are washed out of the graft and cannot interact with the recipient’s immune system. Defatting cocktails that increase lipolysis, vasodilators with vascular protective effects, and gene therapies with specific targets and without side effects are just several possible treatments for a marginal liver graft.

2.5.1 Defatting agents

In a porcine model, 48 h of NMP in steatotic livers resulted in higher metabolism (triglyceride, glucose, and urea in the perfusate) and less lipid detection in the tissue (histologically demonstrated) [74]. In another rodent model with steatotic grafts, 3 hours of NMP with the addition of a defatting cocktail showed a reduction of lipids in the cells with a raise in lipid mobility at the gene level [75]. The defatting cocktail was tested on discarded human grafts with good outcomes compared to the control group. Triglycerides accumulation and the grade of macrovesicular steatosis were significantly reduced [76].

2.5.2 Vasodilators

Prostaglandin E1 is a vasodilator with fibrin clot dissolution activity, which has been shown to meliorate microcirculation injury. Rodent studies where PGE1 is used in NMP verified the beneficial effect on treated livers. The levels of aspartate and alanine aminotransferase were decreased and the bile production was higher [77, 78]. Nitroglycerin and prostaglandin E1 were contained in the Vasosol solution used in the first clinical trial of HMP in human livers [79]. Apart from PGE1, prostacyclin had the same advantages in treated porcine livers [80]. Another porcine transplant study researched the effect of BQ123 and verapamil as vasodilators and noticed that treated livers had better flow in the hepatic artery [81].

2.5.3 Gene modulation agents

Antisense oligonucleotides (ASOs) and siRNA are auspicious gene agents that can have specific targets without the need for viral convection. Silencing of miRNA-122 by ASO resulted in reduced HCV activity in a porcine study [82]. Despite the fact that antiviral medication is widespread and efficient, this finding is important in the evolution of MP therapeutics. siRNAs can have multiple targets such as Fas pathway, p53, RelB, TNF-a, and proapoptotic caspases. All these targets are associated with different aspects of IRI and can potentially protect livers, as shown in animal models under normothermic or hypothermic conditions [83, 84, 85].

2.5.4 Others

Anti-inflammatory agents (alprostadil, n- acetylcysteine, carbon monoxide, and sevoflurane) used under subnormothermic or hypothermic conditions, especially the NLRP3 inflammasome inhibitor mcc950 [86], enkephalin, which is a Δ-opioid agonist treated rat livers under NMP [87], and human liver stem cells in an extracellular form [88], significantly reduced IRI markers.

Advertisement

3. Machine perfusion of the kidney

3.1 Hypothermic machine perfusion

HMP has been established as a safe and effective preservation method with benefits in kidney transplantation. In one of the first RCTs with 336 deceased donors, nonoxygenated HMP was superior to CSC in reducing DGF and increasing 1-year graft survival, regardless of donor type [32]. This publication integrated HMP into daily clinical practice. Afterward, the good short-term results (DGF and 1-month kidney function) of HMP were also confirmed in DCD kidneys. Regarding long-term outcomes, the 164 kidney transplants (82 HMP vs. 82 SCS) had no significant difference in 1-year graft survival [89].

The addition of oxygen and the development of HOPE showed promising results. Application of HOPE in a rodent study significantly decreased markers of reperfusion injury (IL-6, ENO, and TNF-alpha) after kidney transplantation [90]. In a recent meta-analysis, PNF and DGF were significantly reduced in HMP kidneys compared with SCS, but no difference in acute rejection was observed. Also, one-year graft survival was longer in the HMP group [91].

Although the benefit of HMP over SCS has been proven, a large randomized clinical trial COPE-POMP, which evaluated ECD kidneys stored on ice and then perfused in oxygenated hypothermic conditions for 2 hours before implantation, showed no advantages in graft survival (3 months and 1 year) and function (PNF and DGF). These results may be associated with the use of HMP in different centers [92]. Another recent RCT, COPE-COMPARE, compared oxygenated HMP with nonoxygenated HMP in Maastricht III DCD kidneys (50 years and older). The first group improved renal function while decreasing posttransplant complications and acute rejections [93].

The ideal duration and timepoint of HMP are under research, with most studies using end-ischemia time. This phase is practical as the graft is transported on ice and assessed prior to transplantation. In a clinical study of 66 ECD kidneys subjected to end-ischemic perfusion for 369 minutes, the percentage of DGF was 0 versus 9.3% in the contralateral kidneys preserved in SCS [94].

Several potential biomarkers for graft assessment have been studied. The levels of glutathione S-transferase (GST), N-acetyl-beta-D- glucosaminidase (NAG), heart-type fatty acid-binding protein (H-FABP), or LDH measured in the perfusate during MP and are related to DGF [95, 96, 97]. Parameters in HMP, such as resistance and flow, are used in scoring systems to assess kidney quality and risk of DGF [98]. It is noteworthy that CIT, as an independent risk factor in graft function after transplantation, increases the percentage of DGF as hours of cold ischemia pass. As the study of the data from the “Machine Perfusion Trial” showed, if CIT was above 10 hours, DGF was not reduced by HMP [99].

3.2 Normothermic machine perfusion

The first clinical application of NMP (35 min after 11 hours of SCS) in human kidney transplantation was in 2011 with good postoperative results [100]. A larger clinical study compared 18 ECD kidneys that underwent NMP (for approximately 63 min) with matched ECD kidneys that were preserved on ice. DGF was lower in the NMP group (5.6 vs. 36.2%) but 1-year graft survival was comparable in both groups [101].

The advantages of preserving a kidney in normothermic conditions are many. Cold ischemic injury is minimized as aerobic metabolism and renal functions are restarted [102]. The preservation time is investigated in the literature. In a recent study, discarded human kidneys underwent 24 h of NMP using urine recirculation with stable or slightly better tubular function after histopathological assessment [103].

The observation of the graft is feasible and provides the opportunity for assessment.

Parameters, such as total urine output, renal blood flow, and macroscopic perfusion during NMP, are used in score systems to evaluate the graft quality and “transplantability” [104]. Of course, therapies and agents that improve the condition of the graft prior to transplantation are feasible in normothermic preservation. A functional organ in NMP is more easily modified than in hypothermic conditions where cellular mechanisms are under-functioning.

3.3 Therapeutics in machine perfusion

3.3.1 Hypothermic conditions

Mesenchymal stem cells (MSC) and their extracellular vesicles, which have been used in a rat model where the kidneys underwent HMP, have a potential protective role in the perfusion process. The results showed increased activation of enzymes related to cell metabolism and membrane permeability [105]. Carbon monoxide-releasing molecule 401 (CORM-401) decreased IRI markers and improved graft function [106]. Anticoagulative agents, such as thrombalexin and heparin conjugate, have been tested to improve microcirculation, with positive outcomes in the kidneys [107, 108].

3.3.2 Normothermic conditions

Application of MSCs to NMP had a suppressive role in the inflammatory process, in human kidneys [109]. Erythropoietin and its derivative, cyclic helix B peptide (CHBP), can reduce inflammatory agents, such as caspase-3 and IL-1β, providing renoprotection against IRI and improving urine production of the kidney. Recently, a mouse kidney model defined the role of endoplasmic reticulum stress (ERS) in IRI and pointed out the therapeutic potential of CHBP with CHOP (an indicator of ERS) as a biomarker [110, 111, 112]. Also, nanoparticles can be used as drug-delivered agents in blocking or preventing events of inflammation at a molecular level. For example, a possible target is the vascular cells of the graft’s endothelium that are mainly damaged by IRI and performed antidonor antibodies. Nanoparticles, with the presence of anti-CD 31 antibodies, can target endothelial cells and deliver therapeutic agents during NMP [113].

Advertisement

4. Conclusion

Banking organs on a worldwide scale after quality tests would be the ideal method of organ preservation in order to select the perfect-match recipient and then proceed to scheduled surgery [114]. This goal has been achieved in stem cell transplantation and important achievements are taking place in solid organ transplantation, through the development of organ preservation with machine perfusion. Starting with animal experiments and, subsequently, with human clinical studies and randomized controlled trials, machine perfusion is an alternative that still has a lot to offer in solid organ transplantation.

The advantages of MP in short-term outcomes of transplantation are dependent on the preservation method but also the beneficial effects in long-term organ function are continuously demonstrated. It is a platform where, beyond quality, the immunogenicity of the graft could be studied and modified according to the profile of the recipient [103]. The specific targets and the lower dosages, which can be achieved using MP therapeutics agents compared to conventional systemic pharmacological agents, open a new chapter in marginal graft utilization. This may in the future help change the transplant process to a more personalized medicine approach.

New MP technology reveals new perspectives in transplant logistics. Graft evaluation leads to more transplantable organs perhaps in more complex recipients. The prolonged time of preservation provides the opportunity for preoperative planning and elective surgery in day time.

Which perfusion technique enables the highest utilization rate of otherwise discarded livers with the best available outcomes, regarding complications, graft- and patient survival” [115]. This question, which could concern the kidney as well, remains to be answered. So far, there is no evidence of the superiority of a preservation strategy. Randomized clinical trials with combined perfusion protocols may provide the answer to the ideal approach in solid organ transplantation.

References

  1. 1. Available from: https://unos.org
  2. 2. Chouchani ET, Pell VR, Gaude E, Aksentijević D, Sundier SY, Robb EL, et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 2014;515:431-435
  3. 3. Rao PS, Ojo A. The alphabet soup of kidney transplantation: SCD, DCD, ECD–fundamentals for the practicing nephrologist. Clinical Journal of the American Society of Nephrology. 2009;4:1827-1831. DOI: 10.2215/CJN.02270409
  4. 4. Meister FA, Czigany Z, Bednarsch J, Bocker J, Amygdalos I, Morales Santana DA, et al. Hypothermic oxygenated machine perfusion of extended criteria kidney allografts from brain dead donors: Protocol for a prospective pilot study. JMIR Research Protocols. 2019;8:e14622
  5. 5. Czigany Z, Schoning W, Ulmer TF, Bednarsch J, Amygdalos I, Cramer T, et al. Hypothermic oxygenated machine perfusion (HOPE) for orthotopic liver transplantation of human liver allografts from extended criteria donors (ECD) in donation after brain death (DBD): A prospective multicentre randomised controlled trial (HOPE ECD-DBD). BMJ Open. 2017;7:e017558
  6. 6. Okiljević B, ŠuŠak S, Redžek A, Rosić M, Velicki L. Development of cardiopulmonary bypass–a historical review. Srpski Arhiv za Celokupno Lekarstvo. 2016;144:670-675
  7. 7. Carrel A. The culture of whole organs: I. technique of the culture of the thyroid gland. The Journal of Experimental Medicine. 1937;65:515-526
  8. 8. Brettschneider L, Daloze PM, Huguet C, et al. Successful orthotopic transplantation of liver homografts after eight to twenty-five hours preservation. Surgical Forum. 1967;18:376-378
  9. 9. Starzl TE, Groth CG, Brettschneider L, et al. Extended survival in 3 cases of orthotopic homotransplantation of the human liver. Surgery. 1968;63:549-563
  10. 10. Jing L, Yao L, Zhao M, Peng LP, Liu M. Organ preservation: From the past to the future. Acta Pharmacologica Sinica. 2018;39:845-857. DOI: 10.1038/aps.2017.182
  11. 11. Liu WP, Humphries AL, Russell R, Stoddard LD, Moretz WH, Moretz WH. 48-hour storage of canine kidneys after brief perfusion with Collins’ solution. Annals of Surgery. 1971;173:748-757
  12. 12. Rojas-Pena A, Bartlett RH. Ex situ organ preservation: The temperature paradigm. Transplantation. 2018;102:554-556. DOI: 10.1097/TP.0000000000002081
  13. 13. Karangwa SA, Dutkowski P, Fontes P, Friend PJ, Guarrera JV, Markmann JF, et al. Machine perfusion of donor livers for transplantation: A proposal for standard- ized nomenclature and reporting guide- lines. American Journal of Transplantation. 2016;16(10):2932-2942
  14. 14. Stegemann J, Minor T. Energy charge restoration, mitochondrial protection and reversal of preservation induced liver injury by hypothermic oxygenation prior to reperfusion. Cryobiology. 2009;58(3):331-336
  15. 15. Minor T, von Horn C, Paul A. Role of tem- perature in reconditioning and evaluation of cold preserved kidney and liver grafts. Current Opinion in Organ Transplantation. 2017;22(3):267-267
  16. 16. Hessheimer AJ, Fondevila C. Liver perfusion devices: How close are we to widespread application? Current Opinion in Organ Transplantation. 2017;22(2):105-111
  17. 17. Fontes P, Lopez R, van der Plaats A, Vodovotz Y, Minervini M, Scott V, et al. Liver preservation with machine perfusion and a newly developed cell-free oxygen carrier solution under subnormothermic conditions. American Journal of Transplantation. 2015;15(2):381-394
  18. 18. Selten J, Schlegel A, de Jonge J, Dutkowski P. Hypo- and normothermic perfusion of the liver: Which way to go? Best Practice & Research. Clinical Gastroenterology. 2017;31(2):171-179
  19. 19. Adams TD, Patel M, Hosgood SA, Nicholson ML. Lowering perfusate temperature from 37 to 32°C diminishes function in a porcine model of ex vivo kidney perfusion. Transplantation direct. 2017;3:e140
  20. 20. Schopp I, Reissberg E, Lüer B, Efferz P, Minor T. Controlled rewarming after hypothermia: Adding a new principle to renal preservation. Clinical and Translational Science. 2015;8:475-478
  21. 21. Hoyer DP, Mathé Z, Gallinat A, Canbay AC, Treckmann JW, Rauen U, et al. Controlled oxygenated rewarming of cold stored livers prior to transplantation. Transplantation. 2016;100:147-152
  22. 22. Carini R, Bellomo G, Benedetti A, Fulceri R, Gamberucci A, Parola M, et al. Alteration of Na+ homeostasis as a critical step in the development of irreversible hepatocyte injury after adenosine triphosphate depletion. Hepatology. 1995;21:1089-1098
  23. 23. Zaouali MA, Ben Abdennebi H, Padrissa-Altes S, Mahfoudh-Boussaid A, Rosello-Catafau J. Pharmacological strategies against cold ischemia reperfusion injury. Expert Opinion on Pharmacotherapy. 2010;11:537-555
  24. 24. Hessheimer AJ, Garcia-Valdecasas JC, Fondevila C. Abdominal regional in-situ perfusion in donation after circulatory determination of death donors. Current Opinion in Organ Transplantation. 2016;21:322-328
  25. 25. Niatsetskaya ZV, Sosunov SA, Matsiukevich D, Utkina-Sosunova IV, Ratner VI, Starkov AA, et al. The oxygen free radicals originating from mitochondrial complex I contribute to oxidative brain injury following hypoxia-ischemia in neonatal mice. The Journal of Neuroscience. 2012;32:3235-3244
  26. 26. Hoyer DP, Mathe Z, Gallinat A, Canbay AC, Treckmann JW, Rauen U, et al. Controlled oxygenated rewarming of cold stored livers prior to transplantation: First clinical application of a new concept. Transplantation. 2016;100:147-152
  27. 27. van Golen RF, Reiniers MJ, Olthof PB, van Gulik TM, Heger M. Sterile inflammation in hepatic ischemia/reperfusion injury: Present concepts and potential therapeutics. Journal of Gastroenterology and Hepatology. 2013;28:394-400
  28. 28. Koo A, Komatsu H, Tao G, Inoue M, Guth PH, Kaplowitz N. Contribution of no-reflow phenomenon to hepatic injury after ischemia-reperfusion: Evidence for a role for superoxide anion. Hepatology. 1992;15:507-514
  29. 29. Schiefer J, Peter F, Stefan K, Bianca T, Dagmar K, Lorenz K, et al. Glycocalyx damage within human liver grafts correlates with graft injury and postoperative graft function after Orthotopic liver transplantation. Transplantation. 2020;104(1):72-78. DOI: 10.1097/TP.0000000000002838
  30. 30. Stewart RK, Dangi A, Huang C, Murase N, Kimura S, Stolz DB, et al. A novel mouse model of depletion of stellate cells clarifies their role in ischemia/reperfusion- and endotoxin-induced acute liver injury. Journal of Hepatology. 2014;60:298-305
  31. 31. Land WG. The role of postischemic reperfusion injury and other nonantigen-dependent inflammatory pathways in transplantation. Transplantation. 2005;79:505-514
  32. 32. Moers C, Smits JM, Maathuis M-HJ, et al. Machine perfusion or cold storage in deceased-donor kidney transplantation. The New England Journal of Medicine. 2009;360:7
  33. 33. Quillin RC 3rd, Guarrera JV. Hypothermic machine perfusion in liver transplantation. Liver Transplantation. 2018;24:276-281. DOI: 10.1002/lt.25004
  34. 34. Marecki H, Bozorgzadeh A, Porte RJ, Leuvenink HG, Uygun K, Martins NP. Liver ex situ machine perfusion preservation: A review of the methodology and results of large animal studies and clinical trials. Liver Transplantation. 2017;23:679-695. DOI: 10.1002/lt.24751
  35. 35. van Rijn R, van Leeuwen OB, Matton APM, Burlage LC, Wiersema-Buist JMC, van den Heuvel RHJ, et al. Porte: Hypothermic oxygenated machine perfusion reduces bile duct reperfusion injury after transplantation of donation after circulatory death livers. Liver Transplantation. 2018;24:655-664. DOI: 10.1002/lt.25023
  36. 36. Attia M, Silva MA, Mirza DF. The marginal liver donor—An update. Transplant International. 2008;21:713-724
  37. 37. European association for the study of the liver. EASL clinical practice guidelines: Liver transplantation. Journal of Hepatology. 2016;64(2):433-485
  38. 38. Jay CL, Lyuksemburg V, Ladner DP, Wang E, Caicedo JC, Holl JL, et al. Ischemic cholangiopathy after controlled donation after cardiac death liver transplantation: A meta-analysis. Annals of Surgery. 2011;253:259-264
  39. 39. Detelich D, Markmann JF. The dawn of liver perfusion machines. Current Opinion in Organ Transplantation. 2018;23:151-161
  40. 40. Czigany Z, Lurje I, Tolba RH, et al. Machine perfusion for liver transplantation in the era of marginal organs-new kids on the block. Liver International. 2019;39:228-249
  41. 41. Czigany Z, Tacke F, Lurje G. Evolving trends in machine liver perfusion: Comments on clinical end points and selection criteria. Gastroenterology. 2019;157:1166-1167
  42. 42. Jimenez-Romero C, Caso Maestro O, Cambra Molero F, Justo Alonso I, Alegre Torrado C, Manrique Municio A, et al. Using old liver grafts for liver transplantation: Where are the limits? World Journal of Gastroenterology. 2014;20:10691-10702. DOI: 10.3748/wjg.v20.i31.10691
  43. 43. Reese PP, Caplan AL, Kesselheim AS, Bloom RD. Creating a medical, ethical, and legal framework for complex living kidney donors. Clinical Journal of the American Society of Nephrology. 2006;1:1148-1153. DOI: 10.2215/CJN.02180606
  44. 44. Schamberger B, Lohmann D, Sollinger D, Stein R, Lutz J. Association of kidney donor risk index with the outcome after kidney transplantation in the eurotransplant senior program. Annals of Transplantation. 2018;23:775-781. DOI: 10.12659/AOT.909622
  45. 45. Metzger RA, Delmonico FL, Feng S, Port FK, Wynn JJ, Merion RM. Expanded criteria donors for kidney transplantation. American Journal of Transplantation. 2003;3(Suppl. 4):114-125. DOI: 10.1034/j.1600-6143.3.s4.11.x
  46. 46. Domagala P, Gorski L, Wszola M, Kieszek R, Diuwe P, Goralski P, et al. Successful transplantation of kidneys from deceased donors with terminal acute kidney injury. Renal Failure. 2019;41:167-174. DOI: 10.1080/0886022X.2019.1590209
  47. 47. Bronchard R, Durand L, Legeai C, Cohen J, Guerrini P, Bastien O. Brain-dead donors on extracorporeal membrane oxygenation. Critical Care Medicine. 2017;45:1734-1741. DOI: 10.1097/CCM.0000000000002564
  48. 48. Schlegel A, Kron P, Dutkowski P. Hypothermicoxygenatedliverperfusion: Basic mechanisms and clinical application. Current Transplantation Reports. 2015;2:52-62
  49. 49. Guarrera JV, Henry SD, Samstein B, Reznik E, Musat C, Lukose TI, et al. Hypothermic machine preservation facilitates successful transplantation of “orphan” extended criteria donor livers. American Journal of Transplantation. 2015;15:161-169
  50. 50. Nachber E, Tulipan J, Stone J, Bae C, Reznik L, Kato T, et al. Hypothermic machine preservation reduces molecular markers of ischemia/reperfusion injury in human liver transplantation. American Journal of Transplantation. 2012;12:2477-2486
  51. 51. Schlegel A, de Rougemont O, Graf R, Clavien PA, Dutkowski P. Protective mechanisms of end-ischemic cold machine perfusion in DCD liver grafts. Journal of Hepatology. 2013;58:278-286
  52. 52. Henry SD, Nachber E, Tulipan J, Stone J, Bae C, Reznik L, et al. Hypothermic machine preservation reduces molecular markers of ischemia/reperfusion injury in human liver transplantation. American Journal of Transplantation. 2012;12:2477-2486
  53. 53. de Rougemont O, Breitenstein S, Leskosek B, Weber A, Graf R, Clavien PA, et al. One hour hypothermic oxygenated perfusion (HOPE) protects nonviable liver allografts donated after cardiac death. Annals of Surgery. 2009;250:674-683
  54. 54. Yoshikawa R, Matsuno N, Morito N, et al. Evaluation using an isolated reperfusion model for porcine liver donated after cardiac death preserved with oxygenated hypothermic machine perfusion. Annals of Transplantation. 2018;23:822-827
  55. 55. Kron P, Schlegel A, Mancina L, Clavien PA, Dutkowski P. Hypothermic oxygenated perfusion (HOPE) for fatty liver grafts in rats and humans. Journal of Hepatology. 2018;68(1):82-91
  56. 56. Dutkowski P, Schlegel A, de Oliveira M, Mullhaupt B, Neff F, Clavien PA. HOPE for human liver grafts obtained from donors after cardiac death. Journal of Hepatology. 2014;60:765-772
  57. 57. Dutkowski P, Polak WG, Muiesan P, et al. First comparison of hypo- thermic oxygenated perfusion versus static cold storage of human donation after cardiac death liver transplants: An international- matched case analysis. Annals of Surgery. 2015;262:764-770
  58. 58. Schlegel A, Muller X, Kalisvaart M, et al. Outcomes of DCD liver transplantation using organs treated by hypothermic oxygenated perfusion before implantation. Journal of Hepatology. 2019;70(1):50-57
  59. 59. Kron P, Schlegel A, Mancina L, Clavien PA, Dutkowski P. Hypothermic oxygenated perfusion (HOPE) for fatty liver grafts in rats and humans.Journal of Hepatology. 21 Sep 2017:S0168-8278(17):32268-7
  60. 60. Rayar M, Maillot B, Bergeat D, et al. A preliminary clinical experience using hypo- thermic oxygenated machine perfusion for rapid recovery of octogenarian liver grafts. Progress in Transplantation. 2018;29(1):97-98. DOI: 10.1177/1526924818817072
  61. 61. van Rijn R, Karimian N, Matton APM, Burlage LC, Westerkamp AC, van den Berg AP, et al. Dual hypothermic oxygenated machine perfusion in liver transplants donated after circulatory death. The British Journal of Surgery. 2017;104:907-917. DOI: 10.1002/bjs.10515
  62. 62. Jia J, Nie Y, Li J, Xie H, Zhou L, Yu J, et al. Systematic Review and Meta-Analysis of Machine Perfusion vs Static Cold Storage of Liver Allografts on Liver Transplantation Outcomes: The Future Direction of Graft Preservation. Front Med (Lausanne). 12 May 2020;7:135
  63. 63. Muller X, Schlegel A, Kron P, Eshmuminov D, Wurdinger M, Meierhofer D, et al. Novel real-time prediction of liver graft function during hypothermic oxygenated machine perfusion before liver transplantation. Annals of Surgery. 2019;270:783-790
  64. 64. Ravikumar R, Jassem W, Mergental H, Heaton N, Mirza D, Perera MTPR, et al. Liver transplantation after ex vivo normothermic machine preservation: A phase 1 (first-in-man) clinical trial. American Journal of Transplantation. 2016;16(6):1779-1787
  65. 65. Eshmuminov D, Becker D, Bautista Borrego L, Hefti M, Schuler MJ, Hagedorn C, et al. An integrated perfusion machine preserves injured human livers for 1 week. Nature Biotechnology. Feb 2020;38(2):189-198
  66. 66. Boteon YL, Laing RW, Schlegel A, Wallace L, Smith A, Attard J, et al. Combined hypothermic and normothermic machine perfusion improves functional recovery of extended criteria donor livers. Liver Transplantation. 2018;24:1699-1715. DOI: 10.1002/lt.25315
  67. 67. Nasralla D, Coussios CC, Mergental H, Akhtar MZ, Butler AJ, Ceresa CDL, et al. A randomized trial of normothermic preservation in liver transplantation. Nature. 2018;557:50-56
  68. 68. Mergental H, Perera MT, Laing RW, Muiesan P, Isaac JR, Smith A, et al. Transplantation of declined liver allografts following normothermic ex-situ evaluation. American Journal of Transplantation. 2016;16:3235-3245. DOI: 10.1111/ajt.13875
  69. 69. Matton APM, de Vries Y, Burlage LC, et al. Biliary bicarbonate, pH and glucose are suitable biomarkers of biliary viability during ex situ normothermic machine perfusion of human donor livers. Transplantation. Jul 2019;103(7):1405-1413
  70. 70. Goldaracena N, Barbas AS, Selzner M. Normothermic and Subnoromothermic ex vivo liver perfusion in liver transplantation. Current Opinion in Organ Transplantation. 2016;21:315-321
  71. 71. Bruinsma BG, Yeh H, Ozer S, et al. Subnormothermic machine perfusion for ex vivo preservation and recovery of the human liver for transplantation. American Journal of Transplantation. 2014;14:1400-1409
  72. 72. Minor T, Efferz P, Fox M, et al. Controlled oxygenated rewarming of cold stored liver grafts by thermally graduated machine perfusion prior to reperfusion. American Journal of Transplantation. 2013;13:1450-1460
  73. 73. van Leeuwen OB, de Vries Y, Fujiyoshi M, Nijsten MWN, Ubbink R, Pelgrim GJ, et al. Transplantation of high-risk donor livers after ex situ resuscitation and assessment using combined hypo- and Normothermic machine perfusion: A prospective clinical trial. Annals of Surgery. Nov 2019;270(5):906-914
  74. 74. Jamieson RW, Zilvetti M, Roy D, et al. Hepatic steatosis and normothermic perfusion-preliminary experiments in a por- cine model. Transplantation. 2011;92:289-295. DOI: 10.1097/TP.0b013e318223d817
  75. 75. Nagrath D, Xu H, Tanimura Y, et al. Metabolic preconditioning of donor organs: Defatting fatty livers by normothermic perfusion ex vivo. Metabolic Engineering. 2009;11:274-283. DOI: 10.1016/j. ymben.2009.05.005
  76. 76. Boteon YL, Attard J, Boteon APCS, et al. Manipulation of lipid metabolism during normothermic machine perfusion: Effect of defatting therapies on donor liver functional recovery. Liver Transplantation. 2019;25:1007-1022
  77. 77. Hara Y, Akamatsu Y, Maida K, et al. A new liver graft preparation method for uncontrolled non-heart-beating donors, combining short oxygenated warm perfusion and prostaglandin E1. The Journal of Surgical Research. 2013;184:1134-1142. DOI: 10.1016/j.jss.2013.04.030
  78. 78. Maida K, Akamatsu Y, Hara Y, et al. Short oxygenated warm perfusion with prostaglandin E1 administration before cold preservation as a novel resuscitation method for liver grafts from donors after car- diac death in a rat in vivo model. Transplantation. 2016;100:1052-1058. DOI: 10.1097/TP.0000000000001127
  79. 79. Guarrera JV, Karim NA. Liver preservation: Is there anything new yet? Current Opinion in Organ Transplantation. 2008;13:148-154. DOI: 10.1097/MOT.0b013e3282f63930]
  80. 80. Nassar A, Liu Q, Farias K, et al. Role of vasodilation during normo- thermic machine perfusion of DCD porcine livers. The International Journal of Artificial Organs. 2014;37:165-172. DOI: 10.5301/ijao.5000297
  81. 81. Echeverri J, Goldaracena N, Kaths JM, et al. Comparison of BQ123, epoprostenol, and verapamil as vasodilators during normothermic ex vivo liver machine perfusion. Transplantation. 2018;102:601-608. DOI: 10.1097/TP.0000000000002021
  82. 82. Goldaracena N, Spetzler VN, Echeverri J, et al. Inducing hepatitis C virus resistance after pig liver transplantation-a proof of con- cept of liver graft modification using warm ex vivo perfusion. American Journal of Transplantation. 2017;17:970-978. DOI: 10.1111/ajt.14100
  83. 83. Gillooly AR, Perry J, Martins PN. First report of siRNA uptake (for RNA interference) during ex vivo hypothermic and normother- mic liver machine perfusion. Transplantation. 2019;103:e56-e57. DOI: 10.1097/TP.0000000000002515
  84. 84. Thijssen MF, Brüggenwirth IMA, Gillooly A, et al. Gene silencing with siRNA (RNA interference): A new therapeutic option during ex vivo machine liver perfusion preservation. Liver Transplantation. 2019;25:140-151. DOI: 10.1002/lt.25383
  85. 85. Brüggenwirth IMA, Martins PN. RNA interference therapeutics in organ transplantation: The dawn of a new era. American Journal of Transplantation. 2020;20:931-941. DOI: 10.1111/ajt.15689
  86. 86. Yu Y, Cheng Y, Pan Q, et al. Effect of the selective NLRP3 inflammasome inhibitor mcc950 on transplantation outcome in a pig liver transplantation model with organs from donors after circulatory death preserved by hypothermic machine perfusion. Transplantation. 2019;103:353-362. DOI: 10.1097/TP.0000000000002461
  87. 87. Beal EW, Kim JL, Reader BF, et al. [D-Ala2, D-Leu5] Enkephalin improves liver preservation during normothermic ex vivo perfusion. The Journal of Surgical Research. 2019;241:323-335. DOI: 10.1016/j.jss.2019.04.010
  88. 88. Rigo F, De Stefano N, Navarro-Tableros V, et al. Extracellular vesicles from human liver stem cells reduce injury in an ex vivo normothermic hypoxic rat liver perfusion model. Transplantation. 2018;102:e205-e210. DOI: 10.1097/TP.0000000000002123
  89. 89. Jochmans I, Moers C, Smits JM, et al. Machine perfusion versus cold storage for the preservation of kidneys donated after cardiac death: A multicenter, randomized, controlled trial. Annals of Surgery. 2010;252:756
  90. 90. Kron P, Schlegel A, Muller X, Gaspert A, Clavien P-A, Dutkowski P. Hypothermic oxygenated perfusion (HOPE) – A simple and effective method to modulate the immune response in kidney transplantation. Transplantation. May 2019;103(5):e128-e136
  91. 91. Bellini MI, Nozdrin M, Yiu J, Papalois V. Machine perfusion for abdominal organ preservation: A systematic review of kidney and liver human grafts. Journal of Clinical Medicine. 2019;8(8):1221. DOI: 10.3390/jcm8081221
  92. 92. Husen P, Boffa C, Jochmans I, Krikke C, Davies L, Mazilescu L, et al. Oxygenated end-hypothermic machine perfusion in expanded criteria donor kidney transplant: A randomized clinical trial. JAMA Surgery. 1 Jun 2021;156(6):517-525
  93. 93. Jochmans I, Brat A, Davies L, Hofker HS, van de Leemkolk FEM, Leuvenink HGD, et al. Compare trial collaboration and consortium for organ preservation in Europe (COPE). Oxygenated versus standard cold perfusion preservation in kidney transplantation (COMPARE): A randomised, double-blind, paired, phase 3 trial. Lancet. 2020;396(10263):1653-1662
  94. 94. Gallinat A, Amrillaeva V, Hoyer DP, Kocabayoglu P, Benko T, Treckmann JW, et al. Reconditioning by end-ischemic hypothermic in-house machine perfusion: A promising strategy to improve outcome in expanded criteria donors kidney transplantation. Clinical Transplantation. Mar 2017;31(3)
  95. 95. Jochmans I, Moers C, Smits JM, Leuvenink HG, Treckmann J, Paul A, et al. The prognostic value of renal resistance during hypothermic machine perfusion of deceased donor kidneys. American Journal of Transplantation. 2011;11:2214-2220. DOI: 10.1111/j.1600-6143.2011.03685.x
  96. 96. Jochmans I, Pirenne J. Graft quality assessment in kidney transplantation: Not an exact science yet! Current Opinion in Organ Transplantation. 2011;16:174-179. DOI: 10.1097/MOT.0b013e3283446b31
  97. 97. Dare AJ, Pettigrew GJ, Saeb-Parsy K. Preoperative assessment of the deceased-donor kidney: From macroscopic appearance to molecular biomarkers. Transplantation. 2014;97:797-807. DOI: 10.1097/01.TP.0000441361.34103.53
  98. 98. Ding CG, Li Y, Tian XH, et al. Predictive score model for delayed graft function based on hypothermic machine perfusion variables in kidney transplantation. Chinese Medical Journal. 2018;131:2651-2657
  99. 99. Kox J, Moers C, Monbaliu D, Strelniece A, Treckmann J, Jochmans I, et al. The benefits of hypothermic machine preservation and short cold ischemia times in deceased donor kidneys. Transplantation. 2018;102:1344-1350. DOI: 10.1097/TP.0000000000002188
  100. 100. Hosgood SA, Nicholson ML. First in man renal transplantation after ex vivo normothermic perfusion. Transplantation. 2011;92:735-738. DOI: 10.1097/TP.0b013e31822d4e04
  101. 101. Nicholson ML, Hosgood SA. Renal transplantation after ex vivo normothermic perfusion: The first clinical study. American Journal of Transplantation. 2013;13:1246-1252. DOI: 10.1111/ajt.12179
  102. 102. Hosgood SA, van Heurn E, Nicholson ML. Normothermic machine perfusion of the kidney: Better conditioning and repair? Transplant International. 2015;28:657-664
  103. 103. Weissenbacher A, Lo Faro L, Boubriak O, et al. Twenty-four-hour normothermic perfusion of discarded human kidneys with urine recirculation. American Journal of Transplantation. 2019;19:178-192
  104. 104. Hosgood SA, Barlow AD, Hunter JP, Nicholson ML. Ex vivo normothermic perfusion for quality assessment of marginal donor kidney transplants. The British Journal of Surgery. 2015;102:1433-1440. DOI: 10.1002/bjs.9894
  105. 105. Gregorini M, Corradetti V, Pattonieri EF, et al. Perfusion of isolated rat kidney with mesenchymal stromal cells/extracellular vesicles prevents ischaemic injury. Journal of Cellular and Molecular Medicine. 2017;21:3381-3393. DOI: 10.1111/jcmm.13249
  106. 106. Bhattacharjee RN, Richard-Mohamed M, Sun Q, et al. CORM-401 reduces ischemia reperfusion injury in an ex vivo renal porcine model of the donation after circulatory death. Transplantation. 2018;102:1066-1074. DOI: 10.1097/TP.0000000000002201
  107. 107. Hamaoui K, Gowers S, Boutelle M, et al. Organ pretreatment with cytotopic endothelial localizing peptides to ameliorate microvascular thrombosis and perfusion deficits in ex vivo renal hemoreperfusion models. Transplantation. 2016;100:e128-e139. DOI: 10.1097/TP.0000000000001437
  108. 108. Sedigh A, Nordling S, Carlsson F, et al. Perfusion of porcine kidneys with macromolecular heparin reduces early ischemia reperfusion injury. Transplantation. 2019;103:420-427. DOI: 10.1097/TP.0000000000002469
  109. 109. Brasile L, Henry N, Orlando G, et al. Potentiating renal regeneration using mesenchymal stem cells. Transplantation. 2019;103:307-313. DOI: 10.1097/TP.0000000000002455
  110. 110. Yang B, Hosgood SA, Bagul A, et al. Erythropoietin regulates apop- tosis, inflammation and tissue remodelling via caspase-3 and IL-1β in isolated hemoperfused kidneys. European Journal of Pharmacology. 2011;660:420-430. DOI: 10.1016/j.ejphar.2011.03.044
  111. 111. Yang C, Hosgood SA, Meeta P, et al. Cyclic helix B peptide in preservation solution and autologous blood perfusate ameliorates ischemia-reperfusion injury in isolated porcine kidneys. Transplantation direct. 2015;1:e6. DOI: 10.1097/TXD.0000000000000515
  112. 112. Zhang Y, Wang Q, Liu A, Wu Y, Liu F, Wang H, et al. Erythropoietin derived peptide improved endoplasmic reticulum stress and ischemia-reperfusion related cellular and renal injury. Frontiers in Medicine (Lausanne). 2020;7:5. DOI: 10.3389/fmed.2020.00005
  113. 113. Tietjen GT, Hosgood SA, DiRito J, et al. Nanoparticle targeting to the endothelium during normothermic machine perfusion of human kidneys. Science Translational Medicine. 2017;9:eaam6764. DOI: 10.1126/scitranslmed.aam6764
  114. 114. Giwa S, Lewis JK, Alvarez L, et al. The promise of organ and tissue preservation to transform medicine. Nature Biotechnology. 2017;35(6):530-542
  115. 115. Schlegel A, Muller X, Dutkowski P. Machine perfusion strategies in liver transplantation. Hepatobiliary Surgery and Nutrition. 2019;8(5):490-501. DOI: 10.21037/hbsn.2019.04.04

Written By

Stella Vasileiadou, Nikolaos Antoniadis, Athanasios Kofinas, Konstantina-Eleni Karakasi, Georgios Katsanos and Georgios Tsoulfas

Submitted: 30 September 2022 Reviewed: 24 March 2023 Published: 10 April 2023